PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 29989304-1 2019 The magnitude of interaction between the CYP3A4 substrate tacrolimus and various CYP3A4 inhibitors is highly unpredictable. Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-47 29989304-1 2019 The magnitude of interaction between the CYP3A4 substrate tacrolimus and various CYP3A4 inhibitors is highly unpredictable. Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-87 29989304-2 2019 We investigated whether an individual"s baseline in vivo CYP3A4 activity, assessed using the oral midazolam (MDZ) probe, could be used to predict the magnitude of drug-drug interaction between tacrolimus and the potent CYP3A4 inhibitor itraconazole. Tacrolimus 193-203 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 57-63 29989304-2 2019 We investigated whether an individual"s baseline in vivo CYP3A4 activity, assessed using the oral midazolam (MDZ) probe, could be used to predict the magnitude of drug-drug interaction between tacrolimus and the potent CYP3A4 inhibitor itraconazole. Tacrolimus 193-203 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 219-225 30551354-7 2019 Meanwhile TMP could regulate the Interleukin 10 (IL-10) and FK506 could restrain the Interleukin 2 (IL-2). Tacrolimus 60-65 interleukin 2 Homo sapiens 85-98 30551354-7 2019 Meanwhile TMP could regulate the Interleukin 10 (IL-10) and FK506 could restrain the Interleukin 2 (IL-2). Tacrolimus 60-65 interleukin 2 Homo sapiens 100-104 30713247-8 2019 3) Rheumatoid arthritis patients with a genetic mutation of ATP-binding cassette subfamily B member 1 (ABCB1) had high plasma concentrations of tacrolimus and its 13-O-demethylate. Tacrolimus 144-154 ATP binding cassette subfamily B member 1 Homo sapiens 60-101 30713247-8 2019 3) Rheumatoid arthritis patients with a genetic mutation of ATP-binding cassette subfamily B member 1 (ABCB1) had high plasma concentrations of tacrolimus and its 13-O-demethylate. Tacrolimus 144-154 ATP binding cassette subfamily B member 1 Homo sapiens 103-108 30713247-9 2019 The ABCB1 genetic mutation and associated high plasma concentration of tacrolimus decreased kidney function. Tacrolimus 71-81 ATP binding cassette subfamily B member 1 Homo sapiens 4-9 31061321-0 2019 Effect of Serum Parathyroid Hormone on Tacrolimus Therapy in Kidney Transplant Patients: A Possible Biomarker for a Tacrolimus Dosage Schedule. Tacrolimus 39-49 parathyroid hormone Homo sapiens 16-35 31061321-0 2019 Effect of Serum Parathyroid Hormone on Tacrolimus Therapy in Kidney Transplant Patients: A Possible Biomarker for a Tacrolimus Dosage Schedule. Tacrolimus 116-126 parathyroid hormone Homo sapiens 16-35 31061321-3 2019 This retrospective observational study assessed the relationship between serum iPTH levels and the blood concentration or dosage of tacrolimus, a CYP3A substrate, after oral administration in kidney transplant patients. Tacrolimus 132-142 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 146-151 30476735-0 2019 Downregulation of TRPC6 expression is a critical molecular event during FK506 treatment for overactive bladder. Tacrolimus 72-77 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 18-23 30476735-9 2019 TRPC6 elevation in OAB rats was inhibited by FK506, and this inhibition coincided with improvements in urodynamic indices. Tacrolimus 45-50 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 0-5 30476735-11 2019 FK506 inhibited NFAT translocation to the nucleus and disrupted the interaction of TRPC6 with FKBP12. Tacrolimus 0-5 nuclear factor of activated T-cells 5 Rattus norvegicus 16-20 30476735-11 2019 FK506 inhibited NFAT translocation to the nucleus and disrupted the interaction of TRPC6 with FKBP12. Tacrolimus 0-5 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 83-88 30476735-11 2019 FK506 inhibited NFAT translocation to the nucleus and disrupted the interaction of TRPC6 with FKBP12. Tacrolimus 0-5 FKBP prolyl isomerase 1A Rattus norvegicus 94-100 30415182-1 2019 Calcineurin-inhibitor induced pain syndrome (CIPS) is a condition characterized by lower extremity pain in patients receiving tacrolimus or cyclosporine therapy following organ transplantation. Tacrolimus 126-136 calcineurin binding protein 1 Homo sapiens 0-21 31244435-0 2019 Association of Genetic Variants in CYP3A4, CYP3A5, CYP2C8, and CYP2C19 with Tacrolimus Pharmacokinetics in Renal Transplant Recipients. Tacrolimus 76-86 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-41 31244435-0 2019 Association of Genetic Variants in CYP3A4, CYP3A5, CYP2C8, and CYP2C19 with Tacrolimus Pharmacokinetics in Renal Transplant Recipients. Tacrolimus 76-86 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 43-49 31244435-0 2019 Association of Genetic Variants in CYP3A4, CYP3A5, CYP2C8, and CYP2C19 with Tacrolimus Pharmacokinetics in Renal Transplant Recipients. Tacrolimus 76-86 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 51-57 31244435-0 2019 Association of Genetic Variants in CYP3A4, CYP3A5, CYP2C8, and CYP2C19 with Tacrolimus Pharmacokinetics in Renal Transplant Recipients. Tacrolimus 76-86 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 63-70 29479631-1 2019 The calcineurin inhibitor tacrolimus, cornerstone of most immunosuppressive regimens, is a drug with a narrow therapeutic window: underexposure can lead to allograft rejection and overexposure can result in an increased incidence of infections, toxicity and malignancies. Tacrolimus 26-36 calcineurin binding protein 1 Homo sapiens 4-25 30251062-3 2019 The CYP3A5 (rs776746; CYP3A5*3) genotype was determined after monitoring and analysing tacrolimus blood concentrations. Tacrolimus 87-97 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 30251062-3 2019 The CYP3A5 (rs776746; CYP3A5*3) genotype was determined after monitoring and analysing tacrolimus blood concentrations. Tacrolimus 87-97 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 22-28 30251062-7 2019 Significant correlations were observed between C0 and area under the concentration-time curve (AUC0-12) of tacrolimus after the discontinuation of clotrimazole in the CYP3A5 expresser and non-expresser groups, respectively (R2 = 0.49 and 0.42, all P < 0.05), but not before the discontinuation of clotrimazole. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 167-173 30251062-8 2019 CONCLUSION: The effects of clotrimazole on tacrolimus pharmacokinetics in the CYP3A5 expresser patients were significantly greater than those in the CYP3A5 non-expresser patients. Tacrolimus 43-53 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 78-84 30251062-10 2019 Careful dose adjustment of tacrolimus based on CYP3A5 genotypes may be beneficial for the patients with heart transplants who are concomitantly treated with clotrimazole. Tacrolimus 27-37 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 47-53 30296431-9 2019 CONCLUSIONS: Younger age at the time of diagnosis with PBC or at liver transplantation, tacrolimus use, and biochemical markers of cholestasis after liver transplantation are associated with PBC recurrence. Tacrolimus 88-98 dihydrolipoamide S-acetyltransferase Homo sapiens 191-194 31497276-2 2019 Objective: To evaluate M235T and T174M polymorphisms of angiotensinogen gene along with some demographic and clinical factors including age; sex; body mass index (BMI); model for end-stage liver disease (MELD) score; prednisolone, mycophenolate mofetil and tacrolimus dose; and serum level in NODAT among liver recipients. Tacrolimus 257-267 angiotensinogen Homo sapiens 56-71 31483188-8 2019 Taken together, we have shown that JAK3 inhibitor could be used in replacement of tacrolimus in a highly translatable NHP islet transplantation model and these results suggest that JAK3 inhibitor will be potentially incorporated in human allogeneic islet transplantation. Tacrolimus 82-92 Janus kinase 3 Homo sapiens 35-39 30648591-11 2019 KBrO3 increased the protein levels of these signals and the specific inhibitor, such as cyclosporine A and tacrolimus, attenuated the signaling in H9c2 cells at concentrations sufficient to inhibit calcineurin in addition to the reduction of mRNA levels of UBE3A, similar to ANP or BNP. Tacrolimus 107-117 ubiquitin protein ligase E3A Rattus norvegicus 257-262 30648591-11 2019 KBrO3 increased the protein levels of these signals and the specific inhibitor, such as cyclosporine A and tacrolimus, attenuated the signaling in H9c2 cells at concentrations sufficient to inhibit calcineurin in addition to the reduction of mRNA levels of UBE3A, similar to ANP or BNP. Tacrolimus 107-117 natriuretic peptide B Rattus norvegicus 282-285 30489455-1 2019 BACKGROUND: CYP3A4/5 and P-glycoprotein (P-gp, ABCB1) affect tacrolimus (TAC) exposure in T cells and kidney cells. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 12-20 30489455-1 2019 BACKGROUND: CYP3A4/5 and P-glycoprotein (P-gp, ABCB1) affect tacrolimus (TAC) exposure in T cells and kidney cells. Tacrolimus 61-71 ATP binding cassette subfamily B member 1 Homo sapiens 25-39 30489455-1 2019 BACKGROUND: CYP3A4/5 and P-glycoprotein (P-gp, ABCB1) affect tacrolimus (TAC) exposure in T cells and kidney cells. Tacrolimus 61-71 ATP binding cassette subfamily B member 1 Homo sapiens 41-45 29479631-2 2019 Tacrolimus is metabolised in the liver and intestine by the cytochrome P450 3A (CYP3A) isoforms CYP3A4 and CYP3A5. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 60-78 30489455-1 2019 BACKGROUND: CYP3A4/5 and P-glycoprotein (P-gp, ABCB1) affect tacrolimus (TAC) exposure in T cells and kidney cells. Tacrolimus 61-71 ATP binding cassette subfamily B member 1 Homo sapiens 47-52 30489455-1 2019 BACKGROUND: CYP3A4/5 and P-glycoprotein (P-gp, ABCB1) affect tacrolimus (TAC) exposure in T cells and kidney cells. Tacrolimus 73-76 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 12-20 29479631-2 2019 Tacrolimus is metabolised in the liver and intestine by the cytochrome P450 3A (CYP3A) isoforms CYP3A4 and CYP3A5. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 80-85 30489455-1 2019 BACKGROUND: CYP3A4/5 and P-glycoprotein (P-gp, ABCB1) affect tacrolimus (TAC) exposure in T cells and kidney cells. Tacrolimus 73-76 ATP binding cassette subfamily B member 1 Homo sapiens 25-39 30489455-1 2019 BACKGROUND: CYP3A4/5 and P-glycoprotein (P-gp, ABCB1) affect tacrolimus (TAC) exposure in T cells and kidney cells. Tacrolimus 73-76 ATP binding cassette subfamily B member 1 Homo sapiens 41-45 29479631-2 2019 Tacrolimus is metabolised in the liver and intestine by the cytochrome P450 3A (CYP3A) isoforms CYP3A4 and CYP3A5. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 96-102 30489455-1 2019 BACKGROUND: CYP3A4/5 and P-glycoprotein (P-gp, ABCB1) affect tacrolimus (TAC) exposure in T cells and kidney cells. Tacrolimus 73-76 ATP binding cassette subfamily B member 1 Homo sapiens 47-52 29479631-2 2019 Tacrolimus is metabolised in the liver and intestine by the cytochrome P450 3A (CYP3A) isoforms CYP3A4 and CYP3A5. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 107-113 30730287-7 2019 Tacrolimus apparent clearance was significantly reduced in CYP3A5 nonexpressers (CYP3A5*3/*3), concomitant with azole antifungal drugs and Wuzhi capsule (WZ). Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 59-65 30730287-7 2019 Tacrolimus apparent clearance was significantly reduced in CYP3A5 nonexpressers (CYP3A5*3/*3), concomitant with azole antifungal drugs and Wuzhi capsule (WZ). Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 81-87 30293884-9 2019 Interferon (IFN)-gamma concentrations in peripheral blood also diminished significantly with tacrolimus (P<0.01). Tacrolimus 93-103 interferon gamma Homo sapiens 0-22 30537010-0 2019 TLR9 rs352139 Genetic Variant Promotes Tacrolimus Elimination in Chinese Liver Transplant Patients During the Early Posttransplantation Period. Tacrolimus 39-49 toll like receptor 9 Homo sapiens 0-4 30537010-9 2019 RESULTS: Tacrolimus dose-adjusted trough blood concentrations (C/D) ratios were significantly lower for donor TLR9 rs352139 AG/GG carriers than AA carriers at weeks 1, 2, and 3 after LT. Tacrolimus 9-19 toll like receptor 9 Homo sapiens 110-114 30537010-10 2019 In multivariate analysis, donor and recipient CYP3A5 rs776746 and donor TLR9 rs352139 were independent predictors of tacrolimus C/D ratios in the early period after transplantation in both cohorts. Tacrolimus 117-127 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 30537010-10 2019 In multivariate analysis, donor and recipient CYP3A5 rs776746 and donor TLR9 rs352139 were independent predictors of tacrolimus C/D ratios in the early period after transplantation in both cohorts. Tacrolimus 117-127 toll like receptor 9 Homo sapiens 72-76 30537010-13 2019 In addition, we demonstrated that the TLR9 rs352139 genetic variant promotes tacrolimus metabolism of liver cells via upregulation of CYP3A5, which is dependent on the repression of NF-kappaB/pregnane X receptor (PXR) signaling. Tacrolimus 77-87 toll like receptor 9 Homo sapiens 38-42 30537010-13 2019 In addition, we demonstrated that the TLR9 rs352139 genetic variant promotes tacrolimus metabolism of liver cells via upregulation of CYP3A5, which is dependent on the repression of NF-kappaB/pregnane X receptor (PXR) signaling. Tacrolimus 77-87 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 134-140 30537010-13 2019 In addition, we demonstrated that the TLR9 rs352139 genetic variant promotes tacrolimus metabolism of liver cells via upregulation of CYP3A5, which is dependent on the repression of NF-kappaB/pregnane X receptor (PXR) signaling. Tacrolimus 77-87 nuclear receptor subfamily 1 group I member 2 Homo sapiens 182-211 30537010-13 2019 In addition, we demonstrated that the TLR9 rs352139 genetic variant promotes tacrolimus metabolism of liver cells via upregulation of CYP3A5, which is dependent on the repression of NF-kappaB/pregnane X receptor (PXR) signaling. Tacrolimus 77-87 nuclear receptor subfamily 1 group I member 2 Homo sapiens 213-216 30537010-14 2019 CONCLUSIONS: Donor TLR9 rs352139 genetic variant facilitated tacrolimus elimination during the early stage after LT in Chinese patients, which might be related to the upregulation of CYP3A5 enzyme via the NF-kB/PXR signaling pathway. Tacrolimus 61-71 toll like receptor 9 Homo sapiens 19-23 30537010-14 2019 CONCLUSIONS: Donor TLR9 rs352139 genetic variant facilitated tacrolimus elimination during the early stage after LT in Chinese patients, which might be related to the upregulation of CYP3A5 enzyme via the NF-kB/PXR signaling pathway. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 183-189 30537010-14 2019 CONCLUSIONS: Donor TLR9 rs352139 genetic variant facilitated tacrolimus elimination during the early stage after LT in Chinese patients, which might be related to the upregulation of CYP3A5 enzyme via the NF-kB/PXR signaling pathway. Tacrolimus 61-71 nuclear receptor subfamily 1 group I member 2 Homo sapiens 211-214 30293884-7 2019 At week 24, peripheral blood CD4+CD25+high T cells with tacrolimus decreased significantly (P<0.01), but increased significantly without tacrolimus (P<0.01). Tacrolimus 56-66 CD4 molecule Homo sapiens 29-32 30293884-7 2019 At week 24, peripheral blood CD4+CD25+high T cells with tacrolimus decreased significantly (P<0.01), but increased significantly without tacrolimus (P<0.01). Tacrolimus 56-66 interferon stimulated exonuclease gene 20 Homo sapiens 33-37 30584253-0 2018 Whole exome sequencing for the identification of CYP3A7 variants associated with tacrolimus concentrations in kidney transplant patients. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 49-55 31255354-0 2019 Impact of CYP3A5 Genetic Polymorphism on Intrapatient Variability of Tacrolimus Exposure in Chinese Kidney Transplant Recipients. Tacrolimus 69-79 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 31582605-9 2019 A physiologically based pharmacokinetic model adapted to tacrolimus pharmacokinetic data in patients who underwent living-donor liver transplantation was constructed, and clarified that oral clearance of this drug was affected by CYP3A5 genotypes in both the liver and intestine to the same extent. Tacrolimus 57-67 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 230-236 30348662-0 2019 FK506 Resistance of Saccharomyces cerevisiae Pdr5 and Candida albicans Cdr1 Involves Mutations in the Transmembrane Domains and Extracellular Loops. Tacrolimus 0-5 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 45-49 30584253-10 2018 The combined set LMM showed that only CYP3A7 rs2257401 was associated with tacrolimus C0/D after adjusting for patient age, albumin, and creatinine. Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 38-44 30348662-3 2019 Thus, to obtain further insights we searched for FK506-resistant mutants of Saccharomyces cerevisiae cells overexpressing either the endogenous multidrug efflux pump Pdr5 or its Candida albicans orthologue, Cdr1. Tacrolimus 49-54 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 166-170 30348662-4 2019 A simple but powerful screen gave 69 FK506-resistant mutants with, between them, 72 mutations in either Pdr5 or Cdr1. Tacrolimus 37-42 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 104-108 30348662-7 2019 We propose that FK506 inhibits Pdr5 and Cdr1 drug efflux by slowing transporter opening and/or substrate release, and that FK506 resistance of Pdr5/Cdr1 drug efflux is achieved by modifying critical intramolecular contact points that, when mutated, enable the cotransport of FK506 with other pump substrates. Tacrolimus 16-21 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 31-35 30584253-11 2018 The CYP3A7 rs2257401 genotype variant showed a significant difference on the tacrolimus C0/D in those expressing CYP3A5, showing its own effect. Tacrolimus 77-87 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 4-10 30348662-7 2019 We propose that FK506 inhibits Pdr5 and Cdr1 drug efflux by slowing transporter opening and/or substrate release, and that FK506 resistance of Pdr5/Cdr1 drug efflux is achieved by modifying critical intramolecular contact points that, when mutated, enable the cotransport of FK506 with other pump substrates. Tacrolimus 123-128 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 143-147 30348662-7 2019 We propose that FK506 inhibits Pdr5 and Cdr1 drug efflux by slowing transporter opening and/or substrate release, and that FK506 resistance of Pdr5/Cdr1 drug efflux is achieved by modifying critical intramolecular contact points that, when mutated, enable the cotransport of FK506 with other pump substrates. Tacrolimus 123-128 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 143-147 30584253-11 2018 The CYP3A7 rs2257401 genotype variant showed a significant difference on the tacrolimus C0/D in those expressing CYP3A5, showing its own effect. Tacrolimus 77-87 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 113-119 30584253-12 2018 The results suggest that CYP3A7 rs2257401 may serve as a significant genetic marker for tacrolimus pharmacokinetics in kidney transplantation. Tacrolimus 88-98 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 25-31 30318624-0 2018 Once-daily vs twice-daily tacrolimus for de novo living kidney transplantation patients including ABO/HLA compatible and incompatible: A randomized trial. Tacrolimus 26-36 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 98-101 30576367-0 2018 Alteration of urinary neutrophil gelatinase-associated lipocalin as a predictor of tacrolimus-induced chronic renal allograft fibrosis in tacrolimus dose adjustments following kidney transplantation. Tacrolimus 83-93 lipocalin 2 Homo sapiens 22-64 29689130-5 2018 Genotyping the donor for the ABCB1 c.1199 G>A (exon 11, rs2229109) allele may be of interest before prescribing tacrolimus to the recipient, although this polymorphism is rather rare and its effect may be limited to certain mechanisms of graft loss. Tacrolimus 115-125 ATP binding cassette subfamily B member 1 Homo sapiens 29-34 30524720-2 2018 The aim of this study is to compare the carotid intima-media thickness (cIMT) between de novo tacrolimus/mycophenolate and tacrolimus/sirolimus at low doses. Tacrolimus 94-104 CIMT Homo sapiens 72-76 30576367-0 2018 Alteration of urinary neutrophil gelatinase-associated lipocalin as a predictor of tacrolimus-induced chronic renal allograft fibrosis in tacrolimus dose adjustments following kidney transplantation. Tacrolimus 138-148 lipocalin 2 Homo sapiens 22-64 30459239-9 2018 Further analysis showed that addition of LY294002 and FK-506 partly attenuated these protective effects of OP-1 against NP cell apoptosis and activation of the PI3K/Akt/mTOR pathway in a hyperosmotic culture. Tacrolimus 54-60 AKT serine/threonine kinase 1 Rattus norvegicus 165-168 30459239-9 2018 Further analysis showed that addition of LY294002 and FK-506 partly attenuated these protective effects of OP-1 against NP cell apoptosis and activation of the PI3K/Akt/mTOR pathway in a hyperosmotic culture. Tacrolimus 54-60 mechanistic target of rapamycin kinase Rattus norvegicus 169-173 30524720-2 2018 The aim of this study is to compare the carotid intima-media thickness (cIMT) between de novo tacrolimus/mycophenolate and tacrolimus/sirolimus at low doses. Tacrolimus 123-133 CIMT Homo sapiens 72-76 30524720-9 2018 Conclusions: The use of sirolimus plus tacrolimus de novo in kidney transplantation is associated with a reduction in cIMT after 12 months, a decrease more significant than seen with the combination of mycophenolate plus tacrolimus. Tacrolimus 39-49 CIMT Homo sapiens 118-122 30318624-1 2018 Tacrolimus (TAC) is available as a twice-daily capsule (TAC-BID), once-daily capsule (TAC-QD), and once-daily tablet. Tacrolimus 0-10 BH3 interacting domain death agonist Homo sapiens 60-63 30288799-10 2018 CONCLUSION: Topical calcineurin inhibitors are a suitable therapy for AD, and selection of the specific TCI should be based on factors which differentiate tacrolimus from pimecrolimus. Tacrolimus 155-165 latexin Homo sapiens 104-107 30388516-10 2018 Moreover, knockdown of PPP3R1 in Jurkat T cell line enhanced the immunosuppressive effect of tacrolimus. Tacrolimus 93-103 protein phosphatase 3 regulatory subunit B, alpha Homo sapiens 23-29 30388516-12 2018 The functional polymorphism might alter PPP3R1 expression via modulating the interaction of miR-582-5p with PPP3R1, which further affected the immunosuppressive effect of tacrolimus. Tacrolimus 171-181 protein phosphatase 3 regulatory subunit B, alpha Homo sapiens 40-46 30388516-12 2018 The functional polymorphism might alter PPP3R1 expression via modulating the interaction of miR-582-5p with PPP3R1, which further affected the immunosuppressive effect of tacrolimus. Tacrolimus 171-181 microRNA 582 Homo sapiens 92-99 30388516-12 2018 The functional polymorphism might alter PPP3R1 expression via modulating the interaction of miR-582-5p with PPP3R1, which further affected the immunosuppressive effect of tacrolimus. Tacrolimus 171-181 protein phosphatase 3 regulatory subunit B, alpha Homo sapiens 108-114 29691732-0 2018 Impact of the CYP3A5*1 Allele on the Pharmacokinetics of Tacrolimus in Japanese Heart Transplant Patients. Tacrolimus 57-67 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 29691732-2 2018 A common variant of the cytochrome P450 (CYP) 3A5 gene, CYP3A5*3, affects blood concentrations of tacrolimus. Tacrolimus 98-108 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-49 29691732-2 2018 A common variant of the cytochrome P450 (CYP) 3A5 gene, CYP3A5*3, affects blood concentrations of tacrolimus. Tacrolimus 98-108 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 56-62 29691732-4 2018 We retrospectively examined the impact of the CYP3A5 genotype on tacrolimus pharmacokinetics at the early stage of heart transplantation. Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 29691732-11 2018 These results demonstrate the importance of the CYP3A5 genotype in tacrolimus dose optimization based on therapeutic drug monitoring after heart transplantation. Tacrolimus 67-77 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 48-54 30442353-8 2018 The association of T/C and C/C genotypes in the SNP rs11706052 (IMPDH2) with the occurrence of rejection episodes considering only patients who received immunosuppressive drug MMF associated with cyclosporine or tacrolimus and corticoids, demonstrated association with a protection against rejection 15.6-fold. Tacrolimus 212-222 inosine monophosphate dehydrogenase 2 Homo sapiens 64-70 30222497-9 2018 The proportion of terminal deoxynucleotidyl transferase-mediated nick end labeling-positive cells and mRNA levels of inflammatory cytokines were higher in tacrolimus-treated corneas, compared with controls. Tacrolimus 155-165 deoxynucleotidyltransferase, terminal Mus musculus 18-55 30483129-11 2018 Moreover, the present study reveals that combination of radicicol, VER-155008, cyclosporine A, and FK506, which are specific pharmacological inhibitors of Hsp90, Hsp70, Cyps, and FKBPs, respectively, resulted in a stronger inhibition of intoxication of cells with C2 toxin compared to application of the single inhibitors. Tacrolimus 99-104 heat shock protein 90 alpha family class A member 1 Homo sapiens 155-160 30577195-1 2018 BACKGROUND: The purpose of this study is to explore the association of CYP3A5, ABCB1, and CYP2C8 polymorphisms with the risk of developing early kidney impairment in Chinese liver transplant recipients receiving tacrolimus. Tacrolimus 212-222 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 71-77 30577195-11 2018 CONCLUSIONS: CYP2C8*3 and CYP3A5*3 appear to be predictive of risk of tacrolimus-induced early renal impairment. Tacrolimus 70-80 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 13-19 30577195-11 2018 CONCLUSIONS: CYP2C8*3 and CYP3A5*3 appear to be predictive of risk of tacrolimus-induced early renal impairment. Tacrolimus 70-80 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 26-32 30341186-9 2018 Analysis of the effects of FK506 and methylprednisolone, common immunosuppressive agents following organ transplantation, suggested a link between these immunosuppressive drugs and impaired macrophage migration via the Cx43/iNOS/Src/FAK pathway. Tacrolimus 27-32 gap junction protein, alpha 1 Mus musculus 219-223 30341186-9 2018 Analysis of the effects of FK506 and methylprednisolone, common immunosuppressive agents following organ transplantation, suggested a link between these immunosuppressive drugs and impaired macrophage migration via the Cx43/iNOS/Src/FAK pathway. Tacrolimus 27-32 nitric oxide synthase 2, inducible Mus musculus 224-228 30341186-9 2018 Analysis of the effects of FK506 and methylprednisolone, common immunosuppressive agents following organ transplantation, suggested a link between these immunosuppressive drugs and impaired macrophage migration via the Cx43/iNOS/Src/FAK pathway. Tacrolimus 27-32 Rous sarcoma oncogene Mus musculus 229-232 30341186-9 2018 Analysis of the effects of FK506 and methylprednisolone, common immunosuppressive agents following organ transplantation, suggested a link between these immunosuppressive drugs and impaired macrophage migration via the Cx43/iNOS/Src/FAK pathway. Tacrolimus 27-32 PTK2 protein tyrosine kinase 2 Mus musculus 233-236 30483129-11 2018 Moreover, the present study reveals that combination of radicicol, VER-155008, cyclosporine A, and FK506, which are specific pharmacological inhibitors of Hsp90, Hsp70, Cyps, and FKBPs, respectively, resulted in a stronger inhibition of intoxication of cells with C2 toxin compared to application of the single inhibitors. Tacrolimus 99-104 heat shock protein family A (Hsp70) member 4 Homo sapiens 162-167 30019212-0 2018 Evaluating tacrolimus pharmacokinetic models in adult renal transplant recipients with different CYP3A5 genotypes. Tacrolimus 11-21 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 97-103 30166405-1 2018 CYP3A5 genotype is a major determinant of tacrolimus clearance, and has been shown to affect systemic tacrolimus metabolite/parent ratios in healthy volunteers, which may have implications for efficacy and toxicity. Tacrolimus 42-52 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 30166405-1 2018 CYP3A5 genotype is a major determinant of tacrolimus clearance, and has been shown to affect systemic tacrolimus metabolite/parent ratios in healthy volunteers, which may have implications for efficacy and toxicity. Tacrolimus 102-112 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 30166405-2 2018 In a cohort of 50 renal transplant recipients who underwent quantification of CYP3A4 activity using the oral midazolam drug probe, we confirmed that CYP3A5 genotype is the single most important determinant of tacrolimus metabolite/parent ratio [CYP3A5 expressors displayed 2.7- and 2-fold higher relative exposure to 13-desmethyltacrolimus (DMT) and 31-DMT, respectively; P < 0.001]. Tacrolimus 209-219 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 78-84 30166405-2 2018 In a cohort of 50 renal transplant recipients who underwent quantification of CYP3A4 activity using the oral midazolam drug probe, we confirmed that CYP3A5 genotype is the single most important determinant of tacrolimus metabolite/parent ratio [CYP3A5 expressors displayed 2.7- and 2-fold higher relative exposure to 13-desmethyltacrolimus (DMT) and 31-DMT, respectively; P < 0.001]. Tacrolimus 209-219 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 149-155 30166405-2 2018 In a cohort of 50 renal transplant recipients who underwent quantification of CYP3A4 activity using the oral midazolam drug probe, we confirmed that CYP3A5 genotype is the single most important determinant of tacrolimus metabolite/parent ratio [CYP3A5 expressors displayed 2.7- and 2-fold higher relative exposure to 13-desmethyltacrolimus (DMT) and 31-DMT, respectively; P < 0.001]. Tacrolimus 209-219 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 245-251 30166405-4 2018 Additional analyses in 16 healthy volunteers showed that dual pharmacological inhibition of CYP3A4 and P-glycoprotein using itraconazole resulted in increased tacrolimus metabolite/parent ratios (+65%, +112%, and 25% for 13-, 15-, and 31-DMT, respectively; P < 0.01). Tacrolimus 159-169 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 92-98 30166405-4 2018 Additional analyses in 16 healthy volunteers showed that dual pharmacological inhibition of CYP3A4 and P-glycoprotein using itraconazole resulted in increased tacrolimus metabolite/parent ratios (+65%, +112%, and 25% for 13-, 15-, and 31-DMT, respectively; P < 0.01). Tacrolimus 159-169 ATP binding cassette subfamily B member 1 Homo sapiens 103-117 30401377-2 2018 METHODS: From October 2005 to November 2016, 61 LDLT patients administered tacrolimus, who could be genotyped for CYP3A5*3 and *1, were chosen from the electronic record database. Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 114-120 30178515-1 2018 BACKGROUND: Tacrolimus pharmacokinetics are influenced by age and CYP3A5 genotype with CYP3A5 expressors (CYP3A5*1/*1 or *1/*3) being fast metabolizers. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 66-72 30178515-1 2018 BACKGROUND: Tacrolimus pharmacokinetics are influenced by age and CYP3A5 genotype with CYP3A5 expressors (CYP3A5*1/*1 or *1/*3) being fast metabolizers. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 87-93 30178515-1 2018 BACKGROUND: Tacrolimus pharmacokinetics are influenced by age and CYP3A5 genotype with CYP3A5 expressors (CYP3A5*1/*1 or *1/*3) being fast metabolizers. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 87-93 30178515-3 2018 OBJECTIVE: To determine whether age and CYP3A5 genotype-guided starting dose of tacrolimus result in earlier attainment of therapeutic drug concentrations. Tacrolimus 80-90 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 40-46 30178515-11 2018 CONCLUSIONS: CYP3A5 genotype-guided dosing stratified by age resulted in earlier attainment of therapeutic tacrolimus concentrations and fewer out-of-range concentrations. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 29989311-1 2018 Tacrolimus, the major immunosuppressant after heart transplant (HTx) therapy, is a narrow therapeutic index drug. Tacrolimus 0-10 Zic family member 3 Homo sapiens 64-67 30181374-2 2018 In mammals, FK506 inhibits the calcineurin-nuclear factor of activated T cells (NFAT) pathway to prevent T-cell proliferation by forming a ternary complex with its binding protein, FKBP12, and calcineurin. Tacrolimus 12-17 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 181-187 30498355-0 2018 CYP3A5 genotype-based model to predict tacrolimus dosage in the early postoperative period after living donor liver transplantation. Tacrolimus 39-49 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 29989311-9 2018 Multivariate analysis showed that high tacrolimus trough level variability was associated with >8-fold increased risk for any rejection beyond the first year post-HTx (P = .011). Tacrolimus 39-49 Zic family member 3 Homo sapiens 166-169 30132043-6 2018 Pharmacologic inhibition of mTOR signaling using rapamycin (20 nM), FK506 (5 nM), or 4EGI-1 (1 microM), and siRNA knockdown of mTOR, or the mTOR complex binding proteins, raptor or rictor, blocked PCB 95-induced dendritic growth. Tacrolimus 68-73 mechanistic target of rapamycin kinase Rattus norvegicus 28-32 30498175-1 2018 AIM: The current study will attempt to throw light on the role of desmoglein 1 and desmoglein 3 in the pathogenesis of erosive lichen planus and their response to topical application of tacrolimus. Tacrolimus 186-196 desmoglein 1 Homo sapiens 66-78 29708622-0 2018 Evaluation of tacrolimus-related CYP3A5 genotyping in China: Results from the First External Quality Assessment Exercise. Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 29708622-2 2018 The cytochrome P450 3A5 (CYP3A5) has been proved to be associated with tacrolimus dose requirement. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-23 29708622-2 2018 The cytochrome P450 3A5 (CYP3A5) has been proved to be associated with tacrolimus dose requirement. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 25-31 29708622-3 2018 Molecular detection for CYP3A5 genotyping is demanded for the optimization of treatments of tacrolimus. Tacrolimus 92-102 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-30 30498175-1 2018 AIM: The current study will attempt to throw light on the role of desmoglein 1 and desmoglein 3 in the pathogenesis of erosive lichen planus and their response to topical application of tacrolimus. Tacrolimus 186-196 desmoglein 3 Homo sapiens 83-95 30498175-11 2018 Also, there is a significant decrease in the level of anti-Dsgl and anti-Dsg3 autoantibodies with topical tacrolimus 0.1% ointment. Tacrolimus 106-116 desmoglein 3 Homo sapiens 73-77 30498175-12 2018 CLINICAL SIGNIFICANCE: Monitoring the serum level of antibodies against keratinocyte cadherins Dsg 1 and Dsg 3 can be used to evaluate the effect of topical application of tacrolimus on Erosive Oral lichen planus. Tacrolimus 172-182 desmoglein 1 Homo sapiens 95-100 30498175-12 2018 CLINICAL SIGNIFICANCE: Monitoring the serum level of antibodies against keratinocyte cadherins Dsg 1 and Dsg 3 can be used to evaluate the effect of topical application of tacrolimus on Erosive Oral lichen planus. Tacrolimus 172-182 desmoglein 3 Homo sapiens 105-110 30316389-1 2018 BACKGROUND: Inhibition of calcineurin inhibitor (CNI) metabolism with diltiazem reduces the dose of tacrolimus required to achieve its therapeutic blood concentration in kidney transplant recipients (KTRs). Tacrolimus 100-110 calcineurin binding protein 1 Homo sapiens 26-47 29878980-0 2018 Impact of POR and CYP3A5 Polymorphisms on Trough Concentration to Dose Ratio of Tacrolimus in the Early Post-operative Period Following Kidney Transplantation. Tacrolimus 80-90 cytochrome p450 oxidoreductase Homo sapiens 10-13 29878980-0 2018 Impact of POR and CYP3A5 Polymorphisms on Trough Concentration to Dose Ratio of Tacrolimus in the Early Post-operative Period Following Kidney Transplantation. Tacrolimus 80-90 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 18-24 29878980-3 2018 The significant impact of CYP3A5*3 polymorphisms on tacrolimus exposure has been reported. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 26-32 29878980-4 2018 Conflicting results of the additional influence of POR*28 polymorphisms on tacrolimus pharmacokinetic interindividual variability have been observed among different populations. Tacrolimus 75-85 cytochrome p450 oxidoreductase Homo sapiens 51-54 29878980-5 2018 The objective of this study was to explore the interaction between POR*28 and CYP3A5*3 polymorphisms and their main effects on tacrolimus trough concentration to dose ratios on day 7 after kidney transplantation. Tacrolimus 127-137 cytochrome p450 oxidoreductase Homo sapiens 67-70 29878980-5 2018 The objective of this study was to explore the interaction between POR*28 and CYP3A5*3 polymorphisms and their main effects on tacrolimus trough concentration to dose ratios on day 7 after kidney transplantation. Tacrolimus 127-137 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 78-84 29878980-20 2018 CONCLUSIONS: After adjusting for the influences of hemoglobin, age, and prednisolone dose, significant impacts of the CYP3A5 and POR polymorphisms on tacrolimus exposure were found. Tacrolimus 150-160 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 118-124 29878980-20 2018 CONCLUSIONS: After adjusting for the influences of hemoglobin, age, and prednisolone dose, significant impacts of the CYP3A5 and POR polymorphisms on tacrolimus exposure were found. Tacrolimus 150-160 cytochrome p450 oxidoreductase Homo sapiens 129-132 30254425-6 2018 Furthermore, both tacrolimus and anti-VEGF significantly decreased the VEGF-A expression on Days 7 and 14, with no significant difference between the two groups. Tacrolimus 18-28 vascular endothelial growth factor A Rattus norvegicus 71-77 30254425-8 2018 Additionally, the expression of IL-1beta, IL-6, monocyte chemotactic protein-1, macrophage inflammatory protein-1alpha and TGF-beta were significantly decreased by tacrolimus. Tacrolimus 164-174 interleukin 1 beta Rattus norvegicus 32-40 30254425-8 2018 Additionally, the expression of IL-1beta, IL-6, monocyte chemotactic protein-1, macrophage inflammatory protein-1alpha and TGF-beta were significantly decreased by tacrolimus. Tacrolimus 164-174 interleukin 6 Rattus norvegicus 42-46 30254425-8 2018 Additionally, the expression of IL-1beta, IL-6, monocyte chemotactic protein-1, macrophage inflammatory protein-1alpha and TGF-beta were significantly decreased by tacrolimus. Tacrolimus 164-174 C-C motif chemokine ligand 2 Rattus norvegicus 48-78 30254425-8 2018 Additionally, the expression of IL-1beta, IL-6, monocyte chemotactic protein-1, macrophage inflammatory protein-1alpha and TGF-beta were significantly decreased by tacrolimus. Tacrolimus 164-174 C-C motif chemokine ligand 3 Rattus norvegicus 80-118 30254425-8 2018 Additionally, the expression of IL-1beta, IL-6, monocyte chemotactic protein-1, macrophage inflammatory protein-1alpha and TGF-beta were significantly decreased by tacrolimus. Tacrolimus 164-174 transforming growth factor, beta 1 Rattus norvegicus 123-131 30031280-12 2018 TQ and tacrolimus also significantly attenuated mRNA expression levels of IL-4, IL-5 and IFN-gamma (p < 0.001). Tacrolimus 7-17 interleukin 4 Mus musculus 74-78 30208561-0 2018 FK506 Attenuates the MRP1-Mediated Chemoresistant Phenotype in Glioblastoma Stem-Like Cells. Tacrolimus 0-5 ATP binding cassette subfamily C member 1 Homo sapiens 21-25 30208561-3 2018 Tacrolimus (FK506) has been identified as an MRP1 regulator in differentiated glioblastoma (GBM) cells (non-GSCs); however, the effect of FK506 on GSCs is currently unknown. Tacrolimus 0-10 ATP binding cassette subfamily C member 1 Homo sapiens 45-49 30208561-3 2018 Tacrolimus (FK506) has been identified as an MRP1 regulator in differentiated glioblastoma (GBM) cells (non-GSCs); however, the effect of FK506 on GSCs is currently unknown. Tacrolimus 12-17 ATP binding cassette subfamily C member 1 Homo sapiens 45-49 30208561-4 2018 The objective of the following research is to evaluate the effect of FK506 on the MRP1-related chemo-resistant phenotype of GSCs. Tacrolimus 69-74 ATP binding cassette subfamily C member 1 Homo sapiens 82-86 30208561-12 2018 In vivo, the FK506/Vincristine treatment decreased the tumor size as well as ki67, Glial Fibrillary Acidic Protein (GFAP), and nestin expression. Tacrolimus 13-18 glial fibrillary acidic protein Homo sapiens 83-114 30208561-12 2018 In vivo, the FK506/Vincristine treatment decreased the tumor size as well as ki67, Glial Fibrillary Acidic Protein (GFAP), and nestin expression. Tacrolimus 13-18 glial fibrillary acidic protein Homo sapiens 116-120 30208561-13 2018 We conclude that FK506 confers a chemo-sensitive phenotype to MRP1-drug substrate in GSCs. Tacrolimus 17-22 ATP binding cassette subfamily C member 1 Homo sapiens 62-66 29775201-1 2018 Tacrolimus exhibits inter-patient pharmacokinetic variability attributed to CYP3A5 isoenzymes and the efflux transporter, P-glycoprotein. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 76-82 29775201-1 2018 Tacrolimus exhibits inter-patient pharmacokinetic variability attributed to CYP3A5 isoenzymes and the efflux transporter, P-glycoprotein. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 122-136 29775201-3 2018 An established guideline provides tacrolimus genotype dosing recommendations based on CYP3A5*1(W/T) and loss of protein function variants: CYP3A5*3 (rs776746), CYP3A5*6 (rs10264272), CYP3A5*7 (rs41303343) and may provide more comprehensive race-adjusted dosing recommendations. Tacrolimus 34-44 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 86-92 29775201-3 2018 An established guideline provides tacrolimus genotype dosing recommendations based on CYP3A5*1(W/T) and loss of protein function variants: CYP3A5*3 (rs776746), CYP3A5*6 (rs10264272), CYP3A5*7 (rs41303343) and may provide more comprehensive race-adjusted dosing recommendations. Tacrolimus 34-44 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 139-145 29775201-3 2018 An established guideline provides tacrolimus genotype dosing recommendations based on CYP3A5*1(W/T) and loss of protein function variants: CYP3A5*3 (rs776746), CYP3A5*6 (rs10264272), CYP3A5*7 (rs41303343) and may provide more comprehensive race-adjusted dosing recommendations. Tacrolimus 34-44 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 139-145 29775201-3 2018 An established guideline provides tacrolimus genotype dosing recommendations based on CYP3A5*1(W/T) and loss of protein function variants: CYP3A5*3 (rs776746), CYP3A5*6 (rs10264272), CYP3A5*7 (rs41303343) and may provide more comprehensive race-adjusted dosing recommendations. Tacrolimus 34-44 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 139-145 29775201-11 2018 The CYP3A5*3*6*7 metabolic composite was significantly associated with tacrolimus clearance (P value < .05), which was faster in extensive (mean: 45.0 L/hr) and intermediate (29.5 L/hr) metabolizers than poor metabolizers (19.8 L/hr). Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 29775201-12 2018 Simulations support CYP3A5*3*6*7 genotype-based tacrolimus dosing to enhance general race-adjusted regimens, with dose increases of 1.5-fold and 2-fold, respectively, in intermediate and extensive metabolizers for comparable exposures to poor metabolizers. Tacrolimus 48-58 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 29775201-13 2018 This model offers a novel approach to determine tacrolimus dosing adjustments that maintain comparable therapeutic exposure between black and white recipients with different CYP3A5 genotypes. Tacrolimus 48-58 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 174-180 29790290-0 2018 Tacrolimus prevents von Willebrand factor secretion by allostimulated human glomerular endothelium. Tacrolimus 0-10 von Willebrand factor Homo sapiens 20-41 29790290-5 2018 Therefore, we hypothesized that tacrolimus would prevent alloantibody-induced glomerular lesions, in part via inhibition of vWF secretion from endothelial cells. Tacrolimus 32-42 von Willebrand factor Homo sapiens 124-127 29790290-7 2018 We observed that tacrolimus treatment decreased vWF secretion after stimulation with both classes of anti-HLA antibodies and decreased platelet adhesion on allostimulated endothelial cells in a microfluidic chamber. Tacrolimus 17-27 von Willebrand factor Homo sapiens 48-51 29790290-8 2018 In kidney recipients, tacrolimus trough levels were negatively associated with vWF blood levels. Tacrolimus 22-32 von Willebrand factor Homo sapiens 79-82 29603629-0 2018 CYP3A5*3 and ABCB1 61A>G Significantly Influence Dose-adjusted Trough Blood Tacrolimus Concentrations in the First Three Months Post-Kidney Transplantation. Tacrolimus 79-89 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 29603629-0 2018 CYP3A5*3 and ABCB1 61A>G Significantly Influence Dose-adjusted Trough Blood Tacrolimus Concentrations in the First Three Months Post-Kidney Transplantation. Tacrolimus 79-89 ATP binding cassette subfamily B member 1 Homo sapiens 13-18 29330784-12 2018 A multivariate analysis of variance revealed that the tacrolimus concentration in oral fluids is related to the tacrolimus concentration in whole blood and tacrolimus plasma-binding proteins including albumin and cholesterol. Tacrolimus 54-64 albumin Homo sapiens 201-208 29330784-12 2018 A multivariate analysis of variance revealed that the tacrolimus concentration in oral fluids is related to the tacrolimus concentration in whole blood and tacrolimus plasma-binding proteins including albumin and cholesterol. Tacrolimus 112-122 albumin Homo sapiens 201-208 29330784-12 2018 A multivariate analysis of variance revealed that the tacrolimus concentration in oral fluids is related to the tacrolimus concentration in whole blood and tacrolimus plasma-binding proteins including albumin and cholesterol. Tacrolimus 112-122 albumin Homo sapiens 201-208 30031280-12 2018 TQ and tacrolimus also significantly attenuated mRNA expression levels of IL-4, IL-5 and IFN-gamma (p < 0.001). Tacrolimus 7-17 interleukin 5 Mus musculus 80-84 30031280-12 2018 TQ and tacrolimus also significantly attenuated mRNA expression levels of IL-4, IL-5 and IFN-gamma (p < 0.001). Tacrolimus 7-17 interferon gamma Mus musculus 89-98 29733390-11 2018 Independently, the PXR G7635G SNP is related to this increase, proving the role of PXR in tacrolimus metabolism. Tacrolimus 90-100 nuclear receptor subfamily 1 group I member 2 Homo sapiens 19-22 29733390-11 2018 Independently, the PXR G7635G SNP is related to this increase, proving the role of PXR in tacrolimus metabolism. Tacrolimus 90-100 nuclear receptor subfamily 1 group I member 2 Homo sapiens 83-86 29733390-0 2018 Increase in tacrolimus exposure after steroid tapering is influenced by CYP3A5 and pregnane X receptor genetic polymorphisms in renal transplant recipients. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 72-78 29733390-0 2018 Increase in tacrolimus exposure after steroid tapering is influenced by CYP3A5 and pregnane X receptor genetic polymorphisms in renal transplant recipients. Tacrolimus 12-22 nuclear receptor subfamily 1 group I member 2 Homo sapiens 83-102 29733390-1 2018 Background: Tacrolimus, a drug for prevention of rejection after kidney transplantation, has a narrow therapeutic window and is metabolized by the cytochrome P540 3A (CYP3A) system. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 147-165 30092051-3 2018 Tacrolimus (0.2 mg/kg daily) was administered from day 1 to day 30 (TAC1) or from day 7 to day 30 (TAC7), respectively. Tacrolimus 0-10 tachykinin, precursor 1 Rattus norvegicus 68-72 29733390-1 2018 Background: Tacrolimus, a drug for prevention of rejection after kidney transplantation, has a narrow therapeutic window and is metabolized by the cytochrome P540 3A (CYP3A) system. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 167-172 29733390-5 2018 Methods: In a cohort of renal transplant recipients, the influence of CYP3A5 and PXR SNPs (A7635G, C8055T and C25385T) on the dose-normalized Tacrolimus trough concentration (DnC0) and their potential interaction with each other after steroid taper were analysed by linear regression. Tacrolimus 142-152 nuclear receptor subfamily 1 group I member 2 Homo sapiens 81-84 29535109-14 2018 CYP3A5 polymorphism had a significant impact on tacrolimus concentration. Tacrolimus 48-58 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 30116308-0 2018 Analysis of FK506-mediated functional recovery and neuroprotection in a rat model of spinal cord injury indicates that EGF is modulated in astrocytes. Tacrolimus 12-17 epidermal growth factor like 1 Rattus norvegicus 119-122 30116308-9 2018 Furthermore, FK506 upregulated EGF expression of astrocytes both in vivo and in vitro. Tacrolimus 13-18 epidermal growth factor like 1 Rattus norvegicus 31-34 30116308-11 2018 Furthermore, addition of anti-EGF neutralizing antibodies could interrupt the promotion of neurite outgrowth by FK506-CM. Tacrolimus 112-117 epidermal growth factor like 1 Rattus norvegicus 30-33 30116308-12 2018 The present study indicates that astrocytes have an important role as mediators of FK506-improved spinal cord function recovery, and this partially clarifies the role of cell-cell interaction through modulating EGF in this process. Tacrolimus 83-88 epidermal growth factor like 1 Rattus norvegicus 211-214 29380240-2 2018 Previously, we have shown that the calcineurin inhibiting drugs tacrolimus and cyclosporin A reduce adipocyte and myocyte glucose uptakes by reducing the amount of glucose transporter type 4 (GLUT4) at the cell surface, due to an increased internalization rate. Tacrolimus 64-74 solute carrier family 2 member 4 Homo sapiens 164-190 29855074-0 2018 Effect of CYP3A5*1 expression on tacrolimus required dose after liver transplantation: A systematic review and meta-analysis. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 29855074-1 2018 We systematically collected eligible data to measure the effect of CYP3A5*1 expression on personalized tacrolimus therapy. Tacrolimus 103-113 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-73 29855074-2 2018 Six databases were searched for studies on adult liver transplant recipients and donors of liver graft which reported tacrolimus dose requirement, trough blood concentration, and/or concentration/dose (C/D) ratio in expressers and nonexpressers of CYP3A5*1. Tacrolimus 118-128 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 248-254 29855074-8 2018 CYP3A5*1 expression in recipients increased tacrolimus required dose by 0.023 at first, 0.022 at third, and 0.012 mg/kg/day at sixth month. Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 29855074-10 2018 Considering CYP3A5*1 polymorphism can be helpful in individualization of tacrolimus efficient dose prior to administration, and it can remove initial high-risk lag time (over/underdose period before reaching target blood level) at first few days post-transplantation. Tacrolimus 73-83 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 29920787-2 2018 Single nucleotide polymorphisms (SNPs) in genes important to tacrolimus bioavailability and clearance (ABCB1, CYP3A4, and CYP3A5) are associated with differences in tacrolimus pharmacokinetics. Tacrolimus 61-71 ATP binding cassette subfamily B member 1 Homo sapiens 103-108 29920787-2 2018 Single nucleotide polymorphisms (SNPs) in genes important to tacrolimus bioavailability and clearance (ABCB1, CYP3A4, and CYP3A5) are associated with differences in tacrolimus pharmacokinetics. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 110-116 29920787-2 2018 Single nucleotide polymorphisms (SNPs) in genes important to tacrolimus bioavailability and clearance (ABCB1, CYP3A4, and CYP3A5) are associated with differences in tacrolimus pharmacokinetics. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 122-128 29920787-2 2018 Single nucleotide polymorphisms (SNPs) in genes important to tacrolimus bioavailability and clearance (ABCB1, CYP3A4, and CYP3A5) are associated with differences in tacrolimus pharmacokinetics. Tacrolimus 165-175 ATP binding cassette subfamily B member 1 Homo sapiens 103-108 29920787-2 2018 Single nucleotide polymorphisms (SNPs) in genes important to tacrolimus bioavailability and clearance (ABCB1, CYP3A4, and CYP3A5) are associated with differences in tacrolimus pharmacokinetics. Tacrolimus 165-175 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 110-116 29920787-2 2018 Single nucleotide polymorphisms (SNPs) in genes important to tacrolimus bioavailability and clearance (ABCB1, CYP3A4, and CYP3A5) are associated with differences in tacrolimus pharmacokinetics. Tacrolimus 165-175 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 122-128 29920787-6 2018 Compared with CYP3A poor metabolizers (PM), time to therapeutic tacrolimus trough was increased by 5.1 +- 1.6 days for CYP3A extensive metabolizers (EM, P < 0.001). Tacrolimus 64-74 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 119-124 29380240-2 2018 Previously, we have shown that the calcineurin inhibiting drugs tacrolimus and cyclosporin A reduce adipocyte and myocyte glucose uptakes by reducing the amount of glucose transporter type 4 (GLUT4) at the cell surface, due to an increased internalization rate. Tacrolimus 64-74 solute carrier family 2 member 4 Homo sapiens 192-197 29380240-5 2018 Short- and long-term treatments with tacrolimus, cyclosporin A or another CNI deltamethrin (herbicide) decreased basal and insulin-dependent glucose uptake in adipocytes, without any additive effects observed when added together. Tacrolimus 37-47 insulin Homo sapiens 123-130 30228949-7 2018 While both rapamycin and the calcineurin inhibitor tacrolimus elongated the survival of OVA-expressing skin grafts, and inhibited short-term antigen-specific CD8+ T cell responses, rapamycin but not tacrolimus permitted the statistically significant infiltration of CD8+ effector memory T cells into UV-induced SCC lesions. Tacrolimus 51-61 CD8a molecule Homo sapiens 158-161 29621122-11 2018 CONCLUSIONS: Tacrolimus C0 values, its variability, and CYP3A5 polymorphisms were identified as risk factors of AR and tacrolimus ADR. Tacrolimus 119-129 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 56-62 30040022-0 2018 Tacrolimus population pharmacokinetic models according to CYP3A5/CYP3A4/POR genotypes in Chinese Han renal transplant patients. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 58-64 30040022-0 2018 Tacrolimus population pharmacokinetic models according to CYP3A5/CYP3A4/POR genotypes in Chinese Han renal transplant patients. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-71 30040022-0 2018 Tacrolimus population pharmacokinetic models according to CYP3A5/CYP3A4/POR genotypes in Chinese Han renal transplant patients. Tacrolimus 0-10 cytochrome p450 oxidoreductase Homo sapiens 72-75 30040022-7 2018 CONCLUSION: Our final model confirmed that CYP3A5*3 plays a more significant role in tacrolimus PK and could affect the blood concentrations and CL/F (clearance rate/bioavailbility). Tacrolimus 85-95 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 43-49 30228949-7 2018 While both rapamycin and the calcineurin inhibitor tacrolimus elongated the survival of OVA-expressing skin grafts, and inhibited short-term antigen-specific CD8+ T cell responses, rapamycin but not tacrolimus permitted the statistically significant infiltration of CD8+ effector memory T cells into UV-induced SCC lesions. Tacrolimus 51-61 CD8a molecule Homo sapiens 266-269 29804290-0 2018 Tacrolimus Elimination in Four Patients With a CYP3A5*3/*3 CYP3A4*22/*22 Genotype Combination. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 47-53 30036394-7 2018 In vitro experiments revealed that 50 ng/ml tacrolimus affected NFATc1 amplification by 58% (mean; p = 0.02). Tacrolimus 44-54 nuclear factor of activated T cells 1 Homo sapiens 64-70 29664738-4 2018 We further report that ARNT expression itself is controlled by the FKBP12/YY1 transcriptional repressor complex and that disruption of such FKBP12/YY1 complexes by picomolar FK506 at subimmunosuppressive doses increases ARNT expression, subsequently leading to homodimeric ARNT-induced ALK3 transcription. Tacrolimus 174-179 aryl hydrocarbon receptor nuclear translocator Homo sapiens 23-27 29664738-4 2018 We further report that ARNT expression itself is controlled by the FKBP12/YY1 transcriptional repressor complex and that disruption of such FKBP12/YY1 complexes by picomolar FK506 at subimmunosuppressive doses increases ARNT expression, subsequently leading to homodimeric ARNT-induced ALK3 transcription. Tacrolimus 174-179 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 140-146 29664738-4 2018 We further report that ARNT expression itself is controlled by the FKBP12/YY1 transcriptional repressor complex and that disruption of such FKBP12/YY1 complexes by picomolar FK506 at subimmunosuppressive doses increases ARNT expression, subsequently leading to homodimeric ARNT-induced ALK3 transcription. Tacrolimus 174-179 YY1 transcription factor Homo sapiens 147-150 29664738-4 2018 We further report that ARNT expression itself is controlled by the FKBP12/YY1 transcriptional repressor complex and that disruption of such FKBP12/YY1 complexes by picomolar FK506 at subimmunosuppressive doses increases ARNT expression, subsequently leading to homodimeric ARNT-induced ALK3 transcription. Tacrolimus 174-179 aryl hydrocarbon receptor nuclear translocator Homo sapiens 220-224 29664738-4 2018 We further report that ARNT expression itself is controlled by the FKBP12/YY1 transcriptional repressor complex and that disruption of such FKBP12/YY1 complexes by picomolar FK506 at subimmunosuppressive doses increases ARNT expression, subsequently leading to homodimeric ARNT-induced ALK3 transcription. Tacrolimus 174-179 aryl hydrocarbon receptor nuclear translocator Homo sapiens 220-224 29266762-1 2018 Calcineurin inhibitor toxicity (CNT) is a frequent occurrence in transplanted renal grafts and autochthone kidneys from patients undergoing long-term treatment with calcineurin inhibitors, notably cyclosporin A (CsA) and tacrolimus. Tacrolimus 221-231 calcineurin binding protein 1 Mus musculus 0-21 29502069-4 2018 RESULTS: Patients with AC and OVA-sensitised WT mice exhibited increased levels of Gal-3 in the conjunctiva compared with control, an effect reverted by the action of Dex and TC therapy. Tacrolimus 175-177 lectin, galactose binding, soluble 3 Mus musculus 83-88 30036394-0 2018 Analysis of NFATc1 amplification in T cells for pharmacodynamic monitoring of tacrolimus in kidney transplant recipients. Tacrolimus 78-88 nuclear factor of activated T cells 1 Homo sapiens 12-18 30036394-2 2018 Here, we tested whether measuring NFATc1 amplification, a member of the calcineurin pathway, is suitable for TDM of tacrolimus. Tacrolimus 116-126 nuclear factor of activated T cells 1 Homo sapiens 34-40 30036394-3 2018 MATERIALS AND METHODS: NFATc1 amplification was monitored in T cells of kidney transplant recipients who received either tacrolimus- (n = 11) or belatacept-based (n = 10) therapy. Tacrolimus 121-131 nuclear factor of activated T cells 1 Homo sapiens 23-29 30036394-5 2018 RESULTS: At day 30 after transplantation, in tacrolimus-treated patients, NFATc1 amplification was inhibited in CD4+ T cells expressing the co-stimulation receptor CD28 (mean inhibition 37%; p = 0.01) and in CD8+CD28+ T cells (29% inhibition; p = 0.02), while this was not observed in CD8+CD28- T cells or belatacept-treated patients. Tacrolimus 45-55 nuclear factor of activated T cells 1 Homo sapiens 74-80 30036394-5 2018 RESULTS: At day 30 after transplantation, in tacrolimus-treated patients, NFATc1 amplification was inhibited in CD4+ T cells expressing the co-stimulation receptor CD28 (mean inhibition 37%; p = 0.01) and in CD8+CD28+ T cells (29% inhibition; p = 0.02), while this was not observed in CD8+CD28- T cells or belatacept-treated patients. Tacrolimus 45-55 CD28 molecule Homo sapiens 164-168 30036394-5 2018 RESULTS: At day 30 after transplantation, in tacrolimus-treated patients, NFATc1 amplification was inhibited in CD4+ T cells expressing the co-stimulation receptor CD28 (mean inhibition 37%; p = 0.01) and in CD8+CD28+ T cells (29% inhibition; p = 0.02), while this was not observed in CD8+CD28- T cells or belatacept-treated patients. Tacrolimus 45-55 CD28 molecule Homo sapiens 212-216 30036394-5 2018 RESULTS: At day 30 after transplantation, in tacrolimus-treated patients, NFATc1 amplification was inhibited in CD4+ T cells expressing the co-stimulation receptor CD28 (mean inhibition 37%; p = 0.01) and in CD8+CD28+ T cells (29% inhibition; p = 0.02), while this was not observed in CD8+CD28- T cells or belatacept-treated patients. Tacrolimus 45-55 CD28 molecule Homo sapiens 212-216 30036394-6 2018 Tacrolimus pre-dose concentrations of these patients correlated inversely with NFATc1 amplification in CD28+ T cells (rs = -0.46; p < 0.01). Tacrolimus 0-10 nuclear factor of activated T cells 1 Homo sapiens 79-85 30036394-6 2018 Tacrolimus pre-dose concentrations of these patients correlated inversely with NFATc1 amplification in CD28+ T cells (rs = -0.46; p < 0.01). Tacrolimus 0-10 CD28 molecule Homo sapiens 103-107 30036394-8 2018 CONCLUSION: In conclusion, measuring NFATc1 amplification is a direct tool for monitoring biological effects of tacrolimus on T cells in whole blood samples of kidney transplant recipients. Tacrolimus 112-122 nuclear factor of activated T cells 1 Homo sapiens 37-43 29664738-4 2018 We further report that ARNT expression itself is controlled by the FKBP12/YY1 transcriptional repressor complex and that disruption of such FKBP12/YY1 complexes by picomolar FK506 at subimmunosuppressive doses increases ARNT expression, subsequently leading to homodimeric ARNT-induced ALK3 transcription. Tacrolimus 174-179 bone morphogenetic protein receptor type 1A Homo sapiens 286-290 29316256-5 2018 Tacrolimus treatment significantly altered the relative abundance of Allobaculum, Bacteroides, and Lactobacillus and CD4+ CD25hi FoxP3+ regulatory T cells in the colonic mucosa and the circulation. Tacrolimus 0-10 CD4 antigen Mus musculus 117-120 29316256-5 2018 Tacrolimus treatment significantly altered the relative abundance of Allobaculum, Bacteroides, and Lactobacillus and CD4+ CD25hi FoxP3+ regulatory T cells in the colonic mucosa and the circulation. Tacrolimus 0-10 interleukin 2 receptor, alpha chain Mus musculus 122-126 29316256-5 2018 Tacrolimus treatment significantly altered the relative abundance of Allobaculum, Bacteroides, and Lactobacillus and CD4+ CD25hi FoxP3+ regulatory T cells in the colonic mucosa and the circulation. Tacrolimus 0-10 forkhead box P3 Mus musculus 129-134 29804850-7 2018 RESULTS: The histologically proven rejection grade in group AT was significantly lower than that in group T. The serum levels of hepatocyte growth factor and the expression of cMet in group AT accompanied by low CD40 expression were also significantly higher than those of the lung grafts of group T. CONCLUSIONS: These results suggest that co-administration of ADMSCs with tacrolimus is a beneficial therapeutic approach in lung transplantation. Tacrolimus 374-384 MET proto-oncogene, receptor tyrosine kinase Rattus norvegicus 176-180 29991328-1 2018 AIM: To investigate the association between donor CYP3A5 and ABCB1 polymorphisms and tacrolimus (Tac)-induced nephrotoxicity and renal function in kidney transplant recipients. Tacrolimus 85-95 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 50-56 29991328-1 2018 AIM: To investigate the association between donor CYP3A5 and ABCB1 polymorphisms and tacrolimus (Tac)-induced nephrotoxicity and renal function in kidney transplant recipients. Tacrolimus 85-95 ATP binding cassette subfamily B member 1 Homo sapiens 61-66 29804290-0 2018 Tacrolimus Elimination in Four Patients With a CYP3A5*3/*3 CYP3A4*22/*22 Genotype Combination. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 59-65 29804290-1 2018 Cytochrome P450 3A5 (CYP3A5) and cytochrome P450 3A4 (CYP3A4) are the predominate enzymes responsible for tacrolimus metabolism. Tacrolimus 106-116 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-19 29804290-1 2018 Cytochrome P450 3A5 (CYP3A5) and cytochrome P450 3A4 (CYP3A4) are the predominate enzymes responsible for tacrolimus metabolism. Tacrolimus 106-116 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 21-27 29804290-1 2018 Cytochrome P450 3A5 (CYP3A5) and cytochrome P450 3A4 (CYP3A4) are the predominate enzymes responsible for tacrolimus metabolism. Tacrolimus 106-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 33-52 29804290-1 2018 Cytochrome P450 3A5 (CYP3A5) and cytochrome P450 3A4 (CYP3A4) are the predominate enzymes responsible for tacrolimus metabolism. Tacrolimus 106-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 54-60 29804290-2 2018 The presence of CYP3A4 and CYP3A5 genetic variants significantly affects tacrolimus clearance and dose requirements. Tacrolimus 73-83 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 16-22 29804290-2 2018 The presence of CYP3A4 and CYP3A5 genetic variants significantly affects tacrolimus clearance and dose requirements. Tacrolimus 73-83 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 27-33 29920880-0 2018 Effect of CYP3A5*1 expression on tacrolimus required dose for transplant pediatrics: A systematic review and meta-analysis. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 30264877-12 2018 CONCLUSION: Costimulatory blockade-based and anti-CD20 antibody/tacrolimus-based immunosuppressive therapies seem to be comparably safe with steroid therapy in nonhuman primates receiving corneal xenotransplantation, as they did not reactivate Rhesus Cytomegalovirus and maintained manageable systemic status. Tacrolimus 64-74 keratin 20 Homo sapiens 50-54 29757603-5 2018 Pin1 activity, and synapse and dendritic spine numbers are rescued by FK506, a highly specific and United States Food and Drug Administration approved CN inhibitor. Tacrolimus 70-75 peptidylprolyl cis/trans isomerase, NIMA-interacting 1 Homo sapiens 0-4 29757603-7 2018 As such, we suggest prospective clinical trials to determine if systemic FK506 can normalize CN activity in the brain, preserve Pin1 function and support synaptic health in early AD. Tacrolimus 73-78 peptidylprolyl cis/trans isomerase, NIMA-interacting 1 Homo sapiens 128-132 29920880-1 2018 This systematic review was designed to find out optimal tacrolimus dose in pediatrics according to their CYP3A5*1 genotype by performing meta-analysis. Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 105-111 29920880-5 2018 Analysis of tacrolimus required dose, blood concentration, and C/D ratio in 14 time points post-transplantation resulted in significant differences between expressers and non-expressers of CYP3A5*1. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 189-195 29894515-8 2018 The administration of the beta2-GPI DNA vaccine mixed with FK506 reduced the blood IgG anti-beta2-GPI antibody titers and suppressed APS manifestations in mice. Tacrolimus 59-64 apolipoprotein H Mus musculus 26-35 29894515-8 2018 The administration of the beta2-GPI DNA vaccine mixed with FK506 reduced the blood IgG anti-beta2-GPI antibody titers and suppressed APS manifestations in mice. Tacrolimus 59-64 apolipoprotein H Mus musculus 92-101 29666890-0 2018 Manipulating the expression of SARP family regulator BulZ and its target gene product to increase tacrolimus production. Tacrolimus 98-108 ankyrin repeat domain 42 Homo sapiens 31-35 30003047-0 2018 Immunosuppressant drug tacrolimus induced mitochondrial nephrotoxicity, modified PCNA and Bcl-2 expression attenuated by Ocimum basilicum L. in CD1 mice. Tacrolimus 23-33 proliferating cell nuclear antigen Mus musculus 81-85 30003047-0 2018 Immunosuppressant drug tacrolimus induced mitochondrial nephrotoxicity, modified PCNA and Bcl-2 expression attenuated by Ocimum basilicum L. in CD1 mice. Tacrolimus 23-33 CD1 antigen complex Mus musculus 144-147 29666890-2 2018 In this study, we identified that BulZ, a Streptomyces antibiotic regulatory protein (SARP) family regulator, acted as a positive regulator for spore differentiation and tacrolimus production. Tacrolimus 170-180 ankyrin repeat domain 42 Homo sapiens 42-84 29594315-2 2018 We previously reported that MITA correctly reflected the change in mRNA of human whole-blood cells treated with dexamethasone, cyclosporine, FK506, or several other immunosuppressive drugs. Tacrolimus 141-146 stimulator of interferon response cGAMP interactor 1 Mus musculus 28-32 29666890-2 2018 In this study, we identified that BulZ, a Streptomyces antibiotic regulatory protein (SARP) family regulator, acted as a positive regulator for spore differentiation and tacrolimus production. Tacrolimus 170-180 ankyrin repeat domain 42 Homo sapiens 86-90 30078783-6 2018 IL-18 was a potential CYP3A expression modulator and was capable of affecting tacrolimus pharmacokinetics. Tacrolimus 78-88 interleukin 18 Homo sapiens 0-5 29676018-0 2018 Conversion from tacrolimus-mycophenolate mofetil to tacrolimus-mTOR immunosuppression after kidney-pancreas transplantation reduces the incidence of both BK and CMV viremia. Tacrolimus 16-26 mechanistic target of rapamycin kinase Homo sapiens 63-67 29676018-0 2018 Conversion from tacrolimus-mycophenolate mofetil to tacrolimus-mTOR immunosuppression after kidney-pancreas transplantation reduces the incidence of both BK and CMV viremia. Tacrolimus 52-62 mechanistic target of rapamycin kinase Homo sapiens 63-67 29676018-1 2018 BACKGROUND: We sought to determine whether conversion from tacrolimus/mycophenolate mofetil (TAC-MMF) into tacrolimus/mTOR inhibitor (TAC-mTOR) immunosuppression would reduce the incidences of BK and CMV viremia after kidney/pancreas (KP) transplantation. Tacrolimus 107-117 mechanistic target of rapamycin kinase Homo sapiens 138-142 29676018-2 2018 METHODS: In this single-center review, the TAC-mTOR cohort (n = 39) was converted at 1 month post-transplant to an mTOR inhibitor and reduced-dose tacrolimus. Tacrolimus 147-157 mechanistic target of rapamycin kinase Homo sapiens 47-51 29546446-0 2018 CYP3A4 and GCK genetic polymorphisms are the risk factors of tacrolimus-induced new-onset diabetes after transplantation in renal transplant recipients. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 29546446-0 2018 CYP3A4 and GCK genetic polymorphisms are the risk factors of tacrolimus-induced new-onset diabetes after transplantation in renal transplant recipients. Tacrolimus 61-71 glucokinase Homo sapiens 11-14 29546446-1 2018 PURPOSE: We intend to investigate the association between tacrolimus-induced new-onset diabetes after transplantation (NODAT) and polymorphisms of CYP3A4, CYP3A5, ATP-binding cassette transporter sub-family C member 8 (ABCC8), and glucokinase (GCK) in renal transplant recipients. Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 147-153 29879072-14 2018 Since previous studies indicate a role of Th1 inflammation in TAFRO syndrome pathogenesis, tacrolimus may, therefore, be effective in treating TAFRO syndrome. Tacrolimus 91-101 negative elongation factor complex member C/D Homo sapiens 42-45 29546446-1 2018 PURPOSE: We intend to investigate the association between tacrolimus-induced new-onset diabetes after transplantation (NODAT) and polymorphisms of CYP3A4, CYP3A5, ATP-binding cassette transporter sub-family C member 8 (ABCC8), and glucokinase (GCK) in renal transplant recipients. Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 155-161 29546446-1 2018 PURPOSE: We intend to investigate the association between tacrolimus-induced new-onset diabetes after transplantation (NODAT) and polymorphisms of CYP3A4, CYP3A5, ATP-binding cassette transporter sub-family C member 8 (ABCC8), and glucokinase (GCK) in renal transplant recipients. Tacrolimus 58-68 ATP binding cassette subfamily C member 8 Homo sapiens 163-217 29546446-1 2018 PURPOSE: We intend to investigate the association between tacrolimus-induced new-onset diabetes after transplantation (NODAT) and polymorphisms of CYP3A4, CYP3A5, ATP-binding cassette transporter sub-family C member 8 (ABCC8), and glucokinase (GCK) in renal transplant recipients. Tacrolimus 58-68 ATP binding cassette subfamily C member 8 Homo sapiens 219-224 29546446-1 2018 PURPOSE: We intend to investigate the association between tacrolimus-induced new-onset diabetes after transplantation (NODAT) and polymorphisms of CYP3A4, CYP3A5, ATP-binding cassette transporter sub-family C member 8 (ABCC8), and glucokinase (GCK) in renal transplant recipients. Tacrolimus 58-68 glucokinase Homo sapiens 231-242 29546446-1 2018 PURPOSE: We intend to investigate the association between tacrolimus-induced new-onset diabetes after transplantation (NODAT) and polymorphisms of CYP3A4, CYP3A5, ATP-binding cassette transporter sub-family C member 8 (ABCC8), and glucokinase (GCK) in renal transplant recipients. Tacrolimus 58-68 glucokinase Homo sapiens 244-247 29546446-10 2018 CONCLUSIONS: The polymorphisms of CYP3A4 *18B and GCK G-30A were related to NODAT induced by tacrolimus. Tacrolimus 93-103 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 34-40 29546446-10 2018 CONCLUSIONS: The polymorphisms of CYP3A4 *18B and GCK G-30A were related to NODAT induced by tacrolimus. Tacrolimus 93-103 glucokinase Homo sapiens 50-53 29757021-1 2018 INTRODUCTION: Cornerstone immunosuppressive therapy currently relies on immediate-release tacrolimus, a calcineurin inhibitor (CNI) that is potentially nephrotoxic and is more diabetogenic than cyclosporine A. Tacrolimus 90-100 calcineurin binding protein 1 Homo sapiens 104-125 29757021-7 2018 Other studies report that Advagraf -treated patients receiving a mTOR-inhibitor agent (sirolimus or everolimus) instead of MMF: this was associated with good allograft outcome, and might also prevent late-onset cytomegalovirus infection. Tacrolimus 26-34 mechanistic target of rapamycin kinase Homo sapiens 65-69 29160300-0 2018 Genome-wide association study identifies the common variants in CYP3A4 and CYP3A5 responsible for variation in tacrolimus trough concentration in Caucasian kidney transplant recipients. Tacrolimus 111-121 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 64-70 29904381-0 2018 The Calcineurin Inhibitor Tacrolimus Specifically Suppresses Human T Follicular Helper Cells. Tacrolimus 26-36 calcineurin binding protein 1 Homo sapiens 4-25 29904381-8 2018 Results: The calcineurin inhibitor (CNIs) tacrolimus specifically suppresses both LN Tfh cells and circulating Tfh cells, but not their regulatory counterparts or other CD4 T cell subsets. Tacrolimus 42-52 calcineurin binding protein 1 Homo sapiens 13-34 29942927-5 2018 NCX1 inhibition by small interfering RNA or small molecules activates the calcineurin/nuclear factor of activated T cells (NFAT) pathway and inhibits apoptosis induced by the immunosuppressors cyclosporine A (CsA) and tacrolimus in insulin-producing cell. Tacrolimus 218-228 solute carrier family 8 (sodium/calcium exchanger), member 1 Mus musculus 0-4 29454984-6 2018 Interferon (IFN)-gamma and IL-2 content in the serum were lower after operation (P < .05) in the AdOX40Ig and FK506 groups. Tacrolimus 113-118 interferon gamma Rattus norvegicus 0-22 29454984-6 2018 Interferon (IFN)-gamma and IL-2 content in the serum were lower after operation (P < .05) in the AdOX40Ig and FK506 groups. Tacrolimus 113-118 interleukin 2 Rattus norvegicus 27-31 29454984-7 2018 On the contrary, IL-4 and IL-10 content in the serum was higher after operation (P < 0.05) in the AdOX40Ig and FK506 groups. Tacrolimus 114-119 interleukin 4 Rattus norvegicus 17-21 29454984-7 2018 On the contrary, IL-4 and IL-10 content in the serum was higher after operation (P < 0.05) in the AdOX40Ig and FK506 groups. Tacrolimus 114-119 interleukin 10 Rattus norvegicus 26-31 29851248-3 2018 Endothelial dysfunction induced by tacrolimus is related to both increased nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX2) and uncoupled endothelial nitric oxide synthase (eNOS)-driven superoxide production and Rho-kinase-mediated eNOS inhibition. Tacrolimus 35-45 cytochrome b-245, beta polypeptide Mus musculus 136-140 29851248-3 2018 Endothelial dysfunction induced by tacrolimus is related to both increased nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX2) and uncoupled endothelial nitric oxide synthase (eNOS)-driven superoxide production and Rho-kinase-mediated eNOS inhibition. Tacrolimus 35-45 nitric oxide synthase 3, endothelial cell Mus musculus 156-189 29851248-3 2018 Endothelial dysfunction induced by tacrolimus is related to both increased nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX2) and uncoupled endothelial nitric oxide synthase (eNOS)-driven superoxide production and Rho-kinase-mediated eNOS inhibition. Tacrolimus 35-45 Rho-associated coiled-coil containing protein kinase 2 Mus musculus 230-240 29160300-0 2018 Genome-wide association study identifies the common variants in CYP3A4 and CYP3A5 responsible for variation in tacrolimus trough concentration in Caucasian kidney transplant recipients. Tacrolimus 111-121 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 75-81 29160300-1 2018 The immunosuppressant tacrolimus (TAC) is metabolized by both cytochrome P450 3A4 (CYP3A4) and CYP3A5 enzymes. Tacrolimus 22-32 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-81 29160300-1 2018 The immunosuppressant tacrolimus (TAC) is metabolized by both cytochrome P450 3A4 (CYP3A4) and CYP3A5 enzymes. Tacrolimus 22-32 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-89 29160300-1 2018 The immunosuppressant tacrolimus (TAC) is metabolized by both cytochrome P450 3A4 (CYP3A4) and CYP3A5 enzymes. Tacrolimus 22-32 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 95-101 29863266-9 2018 The detection of SOD and ROS showed that ox-LDL could induce the cell oxidative stress injury, whereas tacrolimus could inhibit such an effect. Tacrolimus 103-113 superoxide dismutase 1 Homo sapiens 17-20 29550576-10 2018 The clinical impact of CYP3A5 genetic differences may be small under the current immunosuppressive regimen consisting of mycophenolate mofetil, steroids, basiliximab, and lower target trough levels of tacrolimus with suitable TDM in a low immunological risk population. Tacrolimus 201-211 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 23-29 29488613-0 2018 Association of the PPARgamma/PI3K/Akt pathway with the cardioprotective effects of tacrolimus in myocardial ischemic/reperfusion injury. Tacrolimus 83-93 peroxisome proliferator-activated receptor gamma Rattus norvegicus 19-28 29446493-9 2018 The results indicate that allo-MSC are inherently immunogenic when delivered intramuscularly to healthy and ischemic mouse hind limb, but induce an IgG1-skewed humoral response that is suppressed by tacrolimus. Tacrolimus 199-209 immunoglobulin heavy constant gamma 1 (G1m marker) Mus musculus 148-152 29718866-2 2018 The current study aimed to assess the efficacy and safety of tacrolimus in the treatment of cases of refractory IgAN.In this retrospective observational study, 34 primary IgAN patients with refractory proteinuria received tacrolimus for at least 12 months. Tacrolimus 61-71 IGAN1 Homo sapiens 112-116 29718866-9 2018 Crescent formation in biopsy specimens was seen more often in nonresponder patients.Tacrolimus was safe and effective at lowering proteinuria in refractory IgAN patients. Tacrolimus 84-94 IGAN1 Homo sapiens 156-160 29550576-8 2018 Despite therapeutic drug monitoring (TDM), trough levels of tacrolimus were lower in carriers with the CYP3A5*1 allele (expressers) than in those with the CTP3A5*3/*3 genotype (non-expressers) throughout the 1-year post-transplantation period. Tacrolimus 60-70 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 103-109 29488613-0 2018 Association of the PPARgamma/PI3K/Akt pathway with the cardioprotective effects of tacrolimus in myocardial ischemic/reperfusion injury. Tacrolimus 83-93 AKT serine/threonine kinase 1 Rattus norvegicus 34-37 29488613-8 2018 The phosphorylation of PPARgamma and Akt was significantly activated by tacrolimus, whereas inhibitors of PPARgamma/PI3K/Akt significantly abolished the effects of tacrolimus (P<0.05). Tacrolimus 72-82 peroxisome proliferator-activated receptor gamma Rattus norvegicus 23-32 29488613-8 2018 The phosphorylation of PPARgamma and Akt was significantly activated by tacrolimus, whereas inhibitors of PPARgamma/PI3K/Akt significantly abolished the effects of tacrolimus (P<0.05). Tacrolimus 72-82 AKT serine/threonine kinase 1 Rattus norvegicus 37-40 29488613-8 2018 The phosphorylation of PPARgamma and Akt was significantly activated by tacrolimus, whereas inhibitors of PPARgamma/PI3K/Akt significantly abolished the effects of tacrolimus (P<0.05). Tacrolimus 164-174 peroxisome proliferator-activated receptor gamma Rattus norvegicus 23-32 29488613-8 2018 The phosphorylation of PPARgamma and Akt was significantly activated by tacrolimus, whereas inhibitors of PPARgamma/PI3K/Akt significantly abolished the effects of tacrolimus (P<0.05). Tacrolimus 164-174 peroxisome proliferator-activated receptor gamma Rattus norvegicus 106-115 29488613-8 2018 The phosphorylation of PPARgamma and Akt was significantly activated by tacrolimus, whereas inhibitors of PPARgamma/PI3K/Akt significantly abolished the effects of tacrolimus (P<0.05). Tacrolimus 164-174 AKT serine/threonine kinase 1 Rattus norvegicus 121-124 29488613-9 2018 Together, these results suggest that tacrolimus may protect rats from MIRI through activation of the PPARgamma/PI3K/Akt pathway. Tacrolimus 37-47 peroxisome proliferator-activated receptor gamma Rattus norvegicus 101-110 29488613-9 2018 Together, these results suggest that tacrolimus may protect rats from MIRI through activation of the PPARgamma/PI3K/Akt pathway. Tacrolimus 37-47 AKT serine/threonine kinase 1 Rattus norvegicus 116-119 29731088-2 2018 Tacrolimus (Tac)-induced apoptosis was characterized by nuclear fragmentation and caspase-3 activation. Tacrolimus 0-10 caspase 3 Homo sapiens 82-91 29731088-2 2018 Tacrolimus (Tac)-induced apoptosis was characterized by nuclear fragmentation and caspase-3 activation. Tacrolimus 0-3 caspase 3 Homo sapiens 82-91 29731062-4 2018 OBJECTIVE: The aim of this study was to identify the proportion of CYP3A5 gene polymorphism in Myanmar kidney transplant recipients and to determine the impact of CYP3A5 gene polymorphisms on tacrolimus level in CYP3A5 expressors and nonexpressors. Tacrolimus 192-202 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 163-169 29731088-8 2018 Interestingly, we found that 1 nmol/L Tac treatment induced activation of caspase-12 protease as well as the catalytic activity of caspase-3 but not catalytic activation of caspase-6, -8, and -9 proteases in Jurkat cells. Tacrolimus 38-41 caspase 3 Homo sapiens 131-140 29731062-4 2018 OBJECTIVE: The aim of this study was to identify the proportion of CYP3A5 gene polymorphism in Myanmar kidney transplant recipients and to determine the impact of CYP3A5 gene polymorphisms on tacrolimus level in CYP3A5 expressors and nonexpressors. Tacrolimus 192-202 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 163-169 29362864-3 2018 Treatment of CD-1 mice with Tacrolimus (1 mg/kg/d for 28 days) resulted in kidney injury and was associated with alteration of a gene expression signature associated with cellular stress, fibrosis and inflammation. Tacrolimus 28-38 CD1 antigen complex Mus musculus 13-17 29731062-8 2018 The tacrolimus concentration/dose ratio in the CYP3A5 expressor group was lower than in the CYP3A5 nonexpressor group (1.49 +- 0.69 vs 3.49 +- 3.08 [P = .003] at 1 month; and 1.54 +- 0.9 vs 7.88 +- 8.25 [P = .0001] at 3 months). Tacrolimus 4-14 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 47-53 29731062-10 2018 CYP3A5 genetic polymorphism is one of the important factors in determining daily requirements for tacrolimus and in adjusting tacrolimus trough concentrations. Tacrolimus 98-108 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 29731062-10 2018 CYP3A5 genetic polymorphism is one of the important factors in determining daily requirements for tacrolimus and in adjusting tacrolimus trough concentrations. Tacrolimus 126-136 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 29615122-0 2018 Effect of CYP3 A4, CYP3 A5 and ABCB1 gene polymorphisms on the clinical efficacy of tacrolimus in the treatment of nephrotic syndrome. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 10-17 29615122-0 2018 Effect of CYP3 A4, CYP3 A5 and ABCB1 gene polymorphisms on the clinical efficacy of tacrolimus in the treatment of nephrotic syndrome. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 19-26 29615122-0 2018 Effect of CYP3 A4, CYP3 A5 and ABCB1 gene polymorphisms on the clinical efficacy of tacrolimus in the treatment of nephrotic syndrome. Tacrolimus 84-94 ATP binding cassette subfamily B member 1 Homo sapiens 31-36 29621269-1 2018 The objective of the current study was to explore the role of ABCB1 and CYP3A5 genetic polymorphisms in predicting the bioavailability of tacrolimus and the risk for post-transplant diabetes. Tacrolimus 138-148 ATP binding cassette subfamily B member 1 Homo sapiens 62-67 29621269-1 2018 The objective of the current study was to explore the role of ABCB1 and CYP3A5 genetic polymorphisms in predicting the bioavailability of tacrolimus and the risk for post-transplant diabetes. Tacrolimus 138-148 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 72-78 29621269-5 2018 ABCB1 1236 C>T and 2677G>T/A showed inverse association while CYP3A5*3 showed a positive association with the bioavailability of tacrolimus. Tacrolimus 135-145 ATP binding cassette subfamily B member 1 Homo sapiens 0-5 29621269-5 2018 ABCB1 1236 C>T and 2677G>T/A showed inverse association while CYP3A5*3 showed a positive association with the bioavailability of tacrolimus. Tacrolimus 135-145 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 68-74 29362864-4 2018 Tacrolimus also affected renal miRNA expression, including miRNAs previously involved in fibrotic and inflammatory processes as "fibromirs" such as miR-21-5p, miR-199a-5p and miR-214-3p. Tacrolimus 0-10 microRNA 215 Mus musculus 148-157 29362864-5 2018 In agreement with in vivo data, Renal Proximal Tubular Epithelial cells exposed to Tacrolimus (25 and 50 microM) showed upregulation of miR-21-5p and the concomitant induction of epithelial phenotypic changes, inflammation and oxidative stress. Tacrolimus 83-93 microRNA 215 Mus musculus 136-145 29629825-0 2018 CYP3A pharmacogenetic association with tacrolimus pharmacokinetics differs based on route of drug administration. Tacrolimus 39-49 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-5 28681225-10 2018 Cytochrome P450 3A5 expressers and recipients who received a kidney from a deceased donor had a significantly higher tacrolimus clearance. Tacrolimus 117-127 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-19 28681225-15 2018 CONCLUSION: During the first 6 weeks after transplantation, the tacrolimus weight-normalized starting dose should be higher in pediatric kidney transplant recipients with a lower bodyweight, those who express the cytochrome P450 3A5 genotype, and those who receive a kidney from a deceased donor. Tacrolimus 64-74 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 213-232 28941036-0 2018 Association of donor small ubiquitin-like modifier 4 rs237025 genetic variant with tacrolimus elimination in the early period after liver transplantation. Tacrolimus 83-93 small ubiquitin like modifier 4 Homo sapiens 21-52 28941036-2 2018 In this study, we aimed to investigate the association of donor and recipient small ubiquitin-like modifier 4 (SUMO4) rs237025 polymorphisms with tacrolimus elimination and the potential mechanism. Tacrolimus 146-156 small ubiquitin like modifier 4 Homo sapiens 78-109 28941036-2 2018 In this study, we aimed to investigate the association of donor and recipient small ubiquitin-like modifier 4 (SUMO4) rs237025 polymorphisms with tacrolimus elimination and the potential mechanism. Tacrolimus 146-156 small ubiquitin like modifier 4 Homo sapiens 111-116 28941036-7 2018 RESULTS: Tacrolimus C/D ratios was significantly lower for donor SUMO4 rs237025 AA carriers than AG/GG carriers at weeks 1, 2, 3. Tacrolimus 9-19 small ubiquitin like modifier 4 Homo sapiens 65-70 28941036-8 2018 In multivariate analysis, donor and recipient CYP3A5 rs776747, donor SUMO4 rs237025 and total bilirubin were independent predictors of tacrolimus C/D ratios in the early post-transplantation period both in Cohort A and Cohort B. Tacrolimus 135-145 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 28941036-8 2018 In multivariate analysis, donor and recipient CYP3A5 rs776747, donor SUMO4 rs237025 and total bilirubin were independent predictors of tacrolimus C/D ratios in the early post-transplantation period both in Cohort A and Cohort B. Tacrolimus 135-145 small ubiquitin like modifier 4 Homo sapiens 69-74 28941036-9 2018 When combined donor CYP3A5 rs776746 and donor SUMO4 rs237025 genotypes, tacrolimus C/D ratios was highly significant at all investigated time points within the four groups. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 28941036-9 2018 When combined donor CYP3A5 rs776746 and donor SUMO4 rs237025 genotypes, tacrolimus C/D ratios was highly significant at all investigated time points within the four groups. Tacrolimus 72-82 small ubiquitin like modifier 4 Homo sapiens 46-51 28833755-7 2018 In vitro, tacrolimus inhibited receptor activator of nuclear factor-kappaB ligand-mediated osteoclast formation augmented by interleukin-1beta, thapsigargin, or both. Tacrolimus 10-20 interleukin 1 beta Mus musculus 125-142 28833755-8 2018 Furthermore, tacrolimus inhibited glucose-regulated protein (GRP78), protein kinase R-like endoplasmic reticulum kinase, inositol-requiring enzyme 1 (IRE 1), and activating transcription factor 6 (ATF6) augmented by interleukin-1beta, thapsigargin, or both. Tacrolimus 13-23 heat shock protein 5 Mus musculus 61-66 28833755-8 2018 Furthermore, tacrolimus inhibited glucose-regulated protein (GRP78), protein kinase R-like endoplasmic reticulum kinase, inositol-requiring enzyme 1 (IRE 1), and activating transcription factor 6 (ATF6) augmented by interleukin-1beta, thapsigargin, or both. Tacrolimus 13-23 endoplasmic reticulum (ER) to nucleus signalling 2 Mus musculus 121-148 28833755-8 2018 Furthermore, tacrolimus inhibited glucose-regulated protein (GRP78), protein kinase R-like endoplasmic reticulum kinase, inositol-requiring enzyme 1 (IRE 1), and activating transcription factor 6 (ATF6) augmented by interleukin-1beta, thapsigargin, or both. Tacrolimus 13-23 endoplasmic reticulum (ER) to nucleus signalling 2 Mus musculus 150-155 28833755-8 2018 Furthermore, tacrolimus inhibited glucose-regulated protein (GRP78), protein kinase R-like endoplasmic reticulum kinase, inositol-requiring enzyme 1 (IRE 1), and activating transcription factor 6 (ATF6) augmented by interleukin-1beta, thapsigargin, or both. Tacrolimus 13-23 activating transcription factor 6 Mus musculus 162-195 28833755-8 2018 Furthermore, tacrolimus inhibited glucose-regulated protein (GRP78), protein kinase R-like endoplasmic reticulum kinase, inositol-requiring enzyme 1 (IRE 1), and activating transcription factor 6 (ATF6) augmented by interleukin-1beta, thapsigargin, or both. Tacrolimus 13-23 activating transcription factor 6 Mus musculus 197-201 28833755-8 2018 Furthermore, tacrolimus inhibited glucose-regulated protein (GRP78), protein kinase R-like endoplasmic reticulum kinase, inositol-requiring enzyme 1 (IRE 1), and activating transcription factor 6 (ATF6) augmented by interleukin-1beta, thapsigargin, or both. Tacrolimus 13-23 interleukin 1 beta Mus musculus 216-233 29454235-0 2018 Donor and recipient P450 gene polymorphisms influence individual pharmacological effects of tacrolimus in Chinese liver transplantation patients. Tacrolimus 92-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 29454235-1 2018 The immunosuppressant drug tacrolimus (Tac) used for the prevention of immunological rejection is a metabolic substrate of cytochrome P450 enzymes. Tacrolimus 27-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-138 28891077-0 2018 A comparison of the effects of CYP3A5 polymorphism on tacrolimus blood concentrations measured by 4 immunoassay methods in renal transplant patients. Tacrolimus 54-64 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 31-37 28891077-2 2018 The aim of this study was to investigate differences in the effects of CYP3A5 polymorphism on tacrolimus concentrations obtained by four immunoassay methods in renal transplant patients. Tacrolimus 94-104 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 71-77 28891077-11 2018 Because patients with the CYP3A5*1 allele identified using LTIA may show higher blood concentrations of tacrolimus at lower target concentrations, for example 3.0 ng/mL, compared with other immunoassay methods, there is a need for sufficient consideration of the interpretation of values measured by LTIA. Tacrolimus 104-114 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 26-32 29629825-2 2018 CYP3A5 genotype is an established predictor of oral tacrolimus dose requirements, and clinical guideline recommendations exist for CYP3A5-guided dose selection. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 29629825-3 2018 However, the association between CYP3A5 and nonoral tacrolimus administration is currently poorly understood, and differs from the oral tacrolimus relationship. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 29629825-4 2018 In addition to CYP3A5, other pharmacogenes associated with CYP3A activity, including CYP3A4, CYP3A7 and POR have also been identified as predictors of tacrolimus exposure. Tacrolimus 151-161 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 29629825-4 2018 In addition to CYP3A5, other pharmacogenes associated with CYP3A activity, including CYP3A4, CYP3A7 and POR have also been identified as predictors of tacrolimus exposure. Tacrolimus 151-161 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 15-20 29629825-4 2018 In addition to CYP3A5, other pharmacogenes associated with CYP3A activity, including CYP3A4, CYP3A7 and POR have also been identified as predictors of tacrolimus exposure. Tacrolimus 151-161 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 85-91 29629825-4 2018 In addition to CYP3A5, other pharmacogenes associated with CYP3A activity, including CYP3A4, CYP3A7 and POR have also been identified as predictors of tacrolimus exposure. Tacrolimus 151-161 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 93-99 29629825-4 2018 In addition to CYP3A5, other pharmacogenes associated with CYP3A activity, including CYP3A4, CYP3A7 and POR have also been identified as predictors of tacrolimus exposure. Tacrolimus 151-161 cytochrome p450 oxidoreductase Homo sapiens 104-107 29547545-0 2018 Prediction of Tacrolimus Exposure by CYP3A5 Genotype and Exposure of Co-Administered Everolimus in Japanese Renal Transplant Recipients. Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 29547545-1 2018 While tacrolimus and everolimus have common metabolic pathways through CYP3A4/5, tacrolimus is metabolized solely by CYP3A4 in recipients with the CYP3A5*3/*3. Tacrolimus 6-16 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 71-77 29547545-1 2018 While tacrolimus and everolimus have common metabolic pathways through CYP3A4/5, tacrolimus is metabolized solely by CYP3A4 in recipients with the CYP3A5*3/*3. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 117-123 29547545-1 2018 While tacrolimus and everolimus have common metabolic pathways through CYP3A4/5, tacrolimus is metabolized solely by CYP3A4 in recipients with the CYP3A5*3/*3. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 147-153 29547545-2 2018 The purpose of this study was to evaluate how the area under the blood concentration-time curves (AUC) of tacrolimus could be predicted based on CYP3A5 genotype and the AUC of everolimus in renal transplant patients taking both drugs. Tacrolimus 106-116 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 145-151 29547545-4 2018 Significant correlations between the AUC/D of tacrolimus and everolimus were found for patients with the CYP3A5*1 allele or CYP3A5*3/*3 at both one month and one year. Tacrolimus 46-56 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 105-111 29547545-4 2018 Significant correlations between the AUC/D of tacrolimus and everolimus were found for patients with the CYP3A5*1 allele or CYP3A5*3/*3 at both one month and one year. Tacrolimus 46-56 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 124-130 29547545-6 2018 A good correlation between single doses of tacrolimus and everolimus was found for CYP3A5*3/*3 patients at 1 year after transplantation (r = 0.794, p < 0.001). Tacrolimus 43-53 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 83-89 29547545-7 2018 The variability of the AUC0-24/D of tacrolimus for each CYP3A5 genotype could be predicted based on the AUC0-12/D of everolimus. Tacrolimus 36-46 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 56-62 29547545-8 2018 Clinicians may be able to comprehensively carry out the dose adjustments of tacrolimus and everolimus based on relationship with AUCs of both drugs in each CYP3A5 genotype. Tacrolimus 76-86 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 156-162 29274751-6 2018 NCX2 or NCX3 heterozygotes displayed impaired maintenance of hippocampal LTP, a phenotype that in NCX2+/- mice was correlated with elevated calcineurin activity and rescued by treatment with the calcineurin (CaN) inhibitor FK506. Tacrolimus 223-228 solute carrier family 8 (sodium/calcium exchanger), member 2 Mus musculus 0-4 29553465-18 2018 Myeloperoxidase and malondialdehyde levels were significantly correlated between the EGb761 and FK506 groups, even at lower levels in the EGb761 group (p<0.001). Tacrolimus 96-101 myeloperoxidase Rattus norvegicus 0-15 29274751-6 2018 NCX2 or NCX3 heterozygotes displayed impaired maintenance of hippocampal LTP, a phenotype that in NCX2+/- mice was correlated with elevated calcineurin activity and rescued by treatment with the calcineurin (CaN) inhibitor FK506. Tacrolimus 223-228 solute carrier family 8 (sodium/calcium exchanger), member 3 Mus musculus 8-12 29162334-12 2018 With IR-Tac, tacrolimus Cmax was 33% higher in CYP3A5 expressers compared with nonexpressers (P=0.04): With LCPT, this difference was 11% (P=0.4). Tacrolimus 13-23 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 47-53 29559812-7 2018 The IRR (95% confidence interval [CI]) for lymphoma, tacrolimus versus TCSs, was 3.74 (1.00-14.06) in children and 1.27 (0.94-1.71) in adults. Tacrolimus 53-63 insulin receptor related receptor Homo sapiens 4-7 29563827-0 2018 CYP3A5 polymorphisms in renal transplant recipients: influence on tacrolimus treatment. Tacrolimus 66-76 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 29563827-3 2018 Genetic polymorphism in CYP3A5 enzyme expression contributes to differences in tacrolimus bioavailability between individuals. Tacrolimus 79-89 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-30 29563827-4 2018 Individuals carrying one or more copies of the wild-type allele *1 express CYP3A5, which increases tacrolimus clearance. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 75-81 29563827-5 2018 CYP3A5 expressers require 1.5 to 2-fold higher tacrolimus doses compared to usual dosing to achieve therapeutic blood concentrations. Tacrolimus 47-57 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 29563827-8 2018 Differences between CYP3A5 genotypes in tacrolimus disposition have not translated into differences in clinical outcomes, such as acute rejection and graft survival. Tacrolimus 40-50 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 29563827-10 2018 CYP3A5 genotype may predict differences in absorption of extended-release and immediate-release oral formulations of tacrolimus. Tacrolimus 117-127 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 29563827-11 2018 Two studies found that CYP3A5 expressers require higher doses of tacrolimus in the extended-release formulation compared to immediate release. Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 23-29 29563827-12 2018 CYP3A5 genotype plays a role in determining the impact of interacting drugs, such as fluconazole, on tacrolimus pharmacokinetics. Tacrolimus 101-111 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 29563827-13 2018 Evidence conflicts regarding the impact of CYP3A5 genotype on risk of nephrotoxicity associated with tacrolimus. Tacrolimus 101-111 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 43-49 28945011-1 2018 Schisantherin A and schisandrin A, the most abundant active ingredients of Wuzhi capsule, are known to inhibit tacrolimus metabolism by inhibiting CYP3A4/5. Tacrolimus 111-121 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 147-153 29412644-0 2018 Conformational Entropy of FK506 Binding to FKBP12 Determined by Nuclear Magnetic Resonance Relaxation and Molecular Dynamics Simulations. Tacrolimus 26-31 FKBP prolyl isomerase 1A Homo sapiens 43-49 29412644-3 2018 Here we report backbone and methyl-axis order parameters of the apo and FK506-bound forms of FKBP12, based on 15N and 2H NMR relaxation. Tacrolimus 72-77 FKBP prolyl isomerase 1A Homo sapiens 93-99 29412644-4 2018 Binding of FK506 to FKBP12 results in localized changes in order parameters, notably for the backbone of residues E54 and I56 and the side chains of I56, I90, and I91, all positioned in the binding site. Tacrolimus 11-16 FKBP prolyl isomerase 1A Homo sapiens 20-26 29314738-9 2018 CONCLUSION: Adding tacrolimus onto anti-TNF therapy is a promising therapeutic option with sustained benefit for refractory RA patients despite treatment with anti-TNF therapy combined with methotrexate. Tacrolimus 19-29 tumor necrosis factor Homo sapiens 164-167 29218605-10 2018 But FK506 increased the mRNA levels of IL12 and inhibited the expression of ICAM-1 mRNAs and had no effects on the IFN-gammaRalpha, MDC, and SOCS1 mRNA in HaCaT cells stimulated with TNF-alpha and IFN-gamma. Tacrolimus 4-9 intercellular adhesion molecule 1 Homo sapiens 76-82 29162334-1 2018 BACKGROUND: Differences in tacrolimus dosing across ancestries is partly attributable to polymorphisms in CYP3A5 genes that encode tacrolimus-metabolizing cytochrome P450 3A5 enzymes. Tacrolimus 27-37 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 106-112 29162334-1 2018 BACKGROUND: Differences in tacrolimus dosing across ancestries is partly attributable to polymorphisms in CYP3A5 genes that encode tacrolimus-metabolizing cytochrome P450 3A5 enzymes. Tacrolimus 131-141 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 106-112 29162334-2 2018 The CYP3A5*1 allele, preponderant in African Americans, is associated with rapid metabolism, subtherapeutic concentrations, and higher dose requirements for tacrolimus, all contributing to worse outcomes. Tacrolimus 157-167 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 29422055-7 2018 However, Fk506-treated mice displayed a decrease in GFAP levels in the ARC. Tacrolimus 9-14 glial fibrillary acidic protein Mus musculus 52-56 29131377-1 2018 FKBP12, known as FK506 binding protein, binds to immunosuppressive drug FK506, which must be taken by patients who received organ transplant. Tacrolimus 17-22 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 0-6 29131377-5 2018 Inhibiting FKBP12 by FK506 significantly increased the rate of 1-cell and fragmented embryos, greatly reduced the rate of 2-cell embryos during in vitro fertilization. Tacrolimus 21-26 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 11-17 29131377-7 2018 QPCR demonstrated that Fbxo43 and P27kip, which are related to the release of MII oocyte arrest, and calcium channel partner protein Orai1 were downregulated, while Cdc2 and Ca2+ sensor at ER, stromal interaction molecule 1 (Stim1) were upregulated for a short time after adding FK506. Tacrolimus 279-284 ORAI calcium release-activated calcium modulator 1 Homo sapiens 133-138 29495430-4 2018 We assessed the amount of tacrolimus and its metabolites, M-1 and M-3, that would be ingested by the breastfed neonates. Tacrolimus 26-36 myoregulin Homo sapiens 58-69 29495430-10 2018 Low concentrations of tacrolimus and its metabolites, M-1 and M-3, in colostrum show that neonates will ingest trace amounts of the drug. Tacrolimus 22-32 myoregulin Homo sapiens 54-65 29520229-0 2018 Tacrolimus Reverses UVB Irradiation-Induced Epidermal Langerhans Cell Reduction by Inhibiting TNF-alpha Secretion in Keratinocytes via Regulation of NF-kappaB/p65. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 94-103 29520229-0 2018 Tacrolimus Reverses UVB Irradiation-Induced Epidermal Langerhans Cell Reduction by Inhibiting TNF-alpha Secretion in Keratinocytes via Regulation of NF-kappaB/p65. Tacrolimus 0-10 nuclear factor kappa B subunit 1 Homo sapiens 149-158 29520229-0 2018 Tacrolimus Reverses UVB Irradiation-Induced Epidermal Langerhans Cell Reduction by Inhibiting TNF-alpha Secretion in Keratinocytes via Regulation of NF-kappaB/p65. Tacrolimus 0-10 RELA proto-oncogene, NF-kB subunit Homo sapiens 159-162 29520229-10 2018 Results: Topical tacrolimus significantly reversed high-dose UVB irradiation-induced epidermal LC reduction and CD1a+ cell increment in culture medium. Tacrolimus 17-27 CD1a molecule Homo sapiens 112-116 29520229-11 2018 Tacrolimus significantly inhibited UVB irradiation-induced tumor necrosis factor-alpha (TNF-alpha) and nuclear factor kappa B (NF-kappaB)/p65 mRNA and protein expression in HaCaT cells. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 59-86 29520229-11 2018 Tacrolimus significantly inhibited UVB irradiation-induced tumor necrosis factor-alpha (TNF-alpha) and nuclear factor kappa B (NF-kappaB)/p65 mRNA and protein expression in HaCaT cells. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 88-97 29520229-11 2018 Tacrolimus significantly inhibited UVB irradiation-induced tumor necrosis factor-alpha (TNF-alpha) and nuclear factor kappa B (NF-kappaB)/p65 mRNA and protein expression in HaCaT cells. Tacrolimus 0-10 nuclear factor kappa B subunit 1 Homo sapiens 103-125 29520229-11 2018 Tacrolimus significantly inhibited UVB irradiation-induced tumor necrosis factor-alpha (TNF-alpha) and nuclear factor kappa B (NF-kappaB)/p65 mRNA and protein expression in HaCaT cells. Tacrolimus 0-10 nuclear factor kappa B subunit 1 Homo sapiens 127-136 29520229-11 2018 Tacrolimus significantly inhibited UVB irradiation-induced tumor necrosis factor-alpha (TNF-alpha) and nuclear factor kappa B (NF-kappaB)/p65 mRNA and protein expression in HaCaT cells. Tacrolimus 0-10 RELA proto-oncogene, NF-kB subunit Homo sapiens 138-141 29520229-12 2018 Tacrolimus also significantly inhibited high-dose UVB irradiation-induced TNF-alpha expression in cultured tissues. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 74-83 29520229-14 2018 Conclusion: Topical tacrolimus 0.03% could reverse UVB irradiation-induced epidermal LC reduction by inhibiting TNF-alpha secretion in keratinocytes via regulation of NF-kappaB/p65. Tacrolimus 20-30 tumor necrosis factor Homo sapiens 112-121 29520229-14 2018 Conclusion: Topical tacrolimus 0.03% could reverse UVB irradiation-induced epidermal LC reduction by inhibiting TNF-alpha secretion in keratinocytes via regulation of NF-kappaB/p65. Tacrolimus 20-30 nuclear factor kappa B subunit 1 Homo sapiens 167-176 29520229-14 2018 Conclusion: Topical tacrolimus 0.03% could reverse UVB irradiation-induced epidermal LC reduction by inhibiting TNF-alpha secretion in keratinocytes via regulation of NF-kappaB/p65. Tacrolimus 20-30 RELA proto-oncogene, NF-kB subunit Homo sapiens 177-180 29469606-0 2018 Detection of a rare CYP3A4 variant in a transplant patient characterized by a tacrolimus poor metabolizer phenotype. Tacrolimus 78-88 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-26 29527316-2 2018 We aimed to describe the utility of tacrolimus to prevent colectomy during second-line vedolizumab initiation after corticosteroid and anti-TNF treatment failure in paediatric severe colitis. Tacrolimus 36-46 tumor necrosis factor Homo sapiens 140-143 29527316-10 2018 Conclusion: We report real-world data on the outcome of tacrolimus around vedolizumab initiation in paediatric UC or CD after corticosteroid and anti-TNF therapy treatment failure. Tacrolimus 56-66 tumor necrosis factor Homo sapiens 150-153 29283464-6 2018 Changes in release of TGF-beta were sensitive to the calcineurin (CaN) inhibitor FK506. Tacrolimus 81-86 transforming growth factor alpha Mus musculus 22-30 29102373-6 2018 Specifically, combination therapy inhibited TLR7 expression in the kidneys of mice with lupus nephritis; combination of tacrolimus and mycophenolate mofetil led to better stabilization of the podocyte actin cytoskeleton through the reciprocal regulation of RhoA and Rac1 activities. Tacrolimus 120-130 ras homolog family member A Mus musculus 257-261 29128286-0 2018 TRPA1 channel participates in tacrolimus-induced pruritus in a chronic contact hypersensitivity murine model. Tacrolimus 30-40 transient receptor potential cation channel, subfamily A, member 1 Mus musculus 0-5 29102373-6 2018 Specifically, combination therapy inhibited TLR7 expression in the kidneys of mice with lupus nephritis; combination of tacrolimus and mycophenolate mofetil led to better stabilization of the podocyte actin cytoskeleton through the reciprocal regulation of RhoA and Rac1 activities. Tacrolimus 120-130 Rac family small GTPase 1 Mus musculus 266-270 29288897-9 2018 The NFAT signaling inhibitor, FK-506, eliminated the effect of TRPC6 on HK-2 cells. Tacrolimus 30-36 transient receptor potential cation channel subfamily C member 6 Homo sapiens 63-68 29594146-0 2018 Early Conversion from Tacrolimus to Belatacept in a Highly Sensitized Renal Allograft Recipient with Calcineurin Inhibitor-Induced de novo Post-Transplant Hemolytic Uremic Syndrome. Tacrolimus 22-32 calcineurin binding protein 1 Homo sapiens 101-122 29256966-0 2018 Influence of donor liver CYP3A4*20 loss-of-function genotype on tacrolimus pharmacokinetics in transplanted patients. Tacrolimus 64-74 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-31 29256966-2 2018 Here, we report the effect of the CYP3A4*20 frameshift allele in two Spanish liver transplant patients treated with tacrolimus. Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 34-40 29256966-10 2018 CONCLUSION: This first description of CYP3A4*20 null genotype in liver-transplanted patients, supports the relevance of CYP3A genotyping in tacrolimus therapy. Tacrolimus 140-150 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 38-44 29256966-10 2018 CONCLUSION: This first description of CYP3A4*20 null genotype in liver-transplanted patients, supports the relevance of CYP3A genotyping in tacrolimus therapy. Tacrolimus 140-150 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 38-43 28974433-11 2018 The higher level of circulating Tfh cells in tacrolimus group might be related to STAT3 signaling. Tacrolimus 45-55 signal transducer and activator of transcription 3 Sus scrofa 82-87 29178680-8 2018 The accumulation of eosinophils and Th2 cells in the skin was suppressed by both dexamethasone and FK506, indicating an essential role of Th2 cells in eosinophil recruitment. Tacrolimus 99-104 heart and neural crest derivatives expressed 2 Mus musculus 36-39 28689771-0 2018 Immunosuppression with tacrolimus improved implantation and rescued expression of uterine progesterone receptor and its co-regulators FKBP52 and PIASy at nidation in the obese and diabetic mice: Comparative studies with metformin. Tacrolimus 23-33 progesterone receptor Mus musculus 90-111 28689771-0 2018 Immunosuppression with tacrolimus improved implantation and rescued expression of uterine progesterone receptor and its co-regulators FKBP52 and PIASy at nidation in the obese and diabetic mice: Comparative studies with metformin. Tacrolimus 23-33 FK506 binding protein 4 Mus musculus 134-140 28689771-0 2018 Immunosuppression with tacrolimus improved implantation and rescued expression of uterine progesterone receptor and its co-regulators FKBP52 and PIASy at nidation in the obese and diabetic mice: Comparative studies with metformin. Tacrolimus 23-33 protein inhibitor of activated STAT 4 Mus musculus 145-150 28689771-3 2018 Immunosuppression with tacrolimus improved pregnancy outcomes in obese and diabetic mice and repeated IF in women with elevated Th1/Th2 blood cell ratios. Tacrolimus 23-33 negative elongation factor complex member C/D Homo sapiens 128-131 28689771-8 2018 Therapeutic interventions with tacrolimus or metformin normalized the expression of decidual IFNgamma, PGR and FKBP52, increased co-localization of protein inhibitor of activated STATy (PIASy) to PGR and resulted in the upregulation of uterine IL11and LIF. Tacrolimus 31-41 interferon gamma Mus musculus 93-101 28689771-8 2018 Therapeutic interventions with tacrolimus or metformin normalized the expression of decidual IFNgamma, PGR and FKBP52, increased co-localization of protein inhibitor of activated STATy (PIASy) to PGR and resulted in the upregulation of uterine IL11and LIF. Tacrolimus 31-41 progesterone receptor Mus musculus 103-106 28689771-8 2018 Therapeutic interventions with tacrolimus or metformin normalized the expression of decidual IFNgamma, PGR and FKBP52, increased co-localization of protein inhibitor of activated STATy (PIASy) to PGR and resulted in the upregulation of uterine IL11and LIF. Tacrolimus 31-41 FK506 binding protein 4 Mus musculus 111-117 28689771-8 2018 Therapeutic interventions with tacrolimus or metformin normalized the expression of decidual IFNgamma, PGR and FKBP52, increased co-localization of protein inhibitor of activated STATy (PIASy) to PGR and resulted in the upregulation of uterine IL11and LIF. Tacrolimus 31-41 protein inhibitor of activated STAT 4 Mus musculus 186-191 28689771-8 2018 Therapeutic interventions with tacrolimus or metformin normalized the expression of decidual IFNgamma, PGR and FKBP52, increased co-localization of protein inhibitor of activated STATy (PIASy) to PGR and resulted in the upregulation of uterine IL11and LIF. Tacrolimus 31-41 progesterone receptor Mus musculus 196-199 28689771-8 2018 Therapeutic interventions with tacrolimus or metformin normalized the expression of decidual IFNgamma, PGR and FKBP52, increased co-localization of protein inhibitor of activated STATy (PIASy) to PGR and resulted in the upregulation of uterine IL11and LIF. Tacrolimus 31-41 interleukin 11 Mus musculus 244-248 28689771-8 2018 Therapeutic interventions with tacrolimus or metformin normalized the expression of decidual IFNgamma, PGR and FKBP52, increased co-localization of protein inhibitor of activated STATy (PIASy) to PGR and resulted in the upregulation of uterine IL11and LIF. Tacrolimus 31-41 leukemia inhibitory factor Mus musculus 252-255 29375315-8 2017 In addition, FK506 treatment down-regulated the expression level of GFAP, a specific marker of astrocytes. Tacrolimus 13-18 glial fibrillary acidic protein Rattus norvegicus 68-72 29801578-4 2018 For calcineurin inhibitors like cyclosporine and in particular tacrolimus however, cytochrome P450 3A4 and 3A5 variants were found to significantly affect the pharmacokinetics. Tacrolimus 63-73 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-102 29178680-8 2018 The accumulation of eosinophils and Th2 cells in the skin was suppressed by both dexamethasone and FK506, indicating an essential role of Th2 cells in eosinophil recruitment. Tacrolimus 99-104 heart and neural crest derivatives expressed 2 Mus musculus 138-141 29539600-1 2018 BACKGROUND: Tacrolimus is mainly metabolized by cytochrome P450 3A5 (CYP3A5), which is expressed in the liver. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 48-67 29055041-3 2018 It was hypothesized that cytochrome P450 (CYP)3A inhibition of the small intestine by voriconazole and P-glycoprotein (P-gp) inhibition of the small intestine by risperidone exerted a synergistic effect on the bioavailability of tacrolimus. Tacrolimus 229-239 ATP binding cassette subfamily B member 1 Homo sapiens 103-117 29539600-1 2018 BACKGROUND: Tacrolimus is mainly metabolized by cytochrome P450 3A5 (CYP3A5), which is expressed in the liver. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 69-75 29539600-2 2018 However, CYP3A5 is also expressed in the kidney tissue and may contribute to local tacrolimus clearance in the kidney allograft. Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 9-15 29055041-0 2018 The possible clinical impact of risperidone on P-glycoprotein-mediated transport of tacrolimus: A case report and in vitro study. Tacrolimus 84-94 ATP binding cassette subfamily B member 1 Homo sapiens 47-61 29055041-3 2018 It was hypothesized that cytochrome P450 (CYP)3A inhibition of the small intestine by voriconazole and P-glycoprotein (P-gp) inhibition of the small intestine by risperidone exerted a synergistic effect on the bioavailability of tacrolimus. Tacrolimus 229-239 ATP binding cassette subfamily B member 1 Homo sapiens 119-123 29055041-4 2018 The aim of the present study was to evaluate the effect of risperidone on the P-gp-mediated transport of tacrolimus. Tacrolimus 105-115 ATP binding cassette subfamily B member 1 Homo sapiens 78-82 30251601-0 2018 Influence of CYP3A and ABCB1 Single Nucleotide Polymorphisms on the Pharmacokinetics/Pharmacodynamics of Tacrolimus in Pediatric Patients. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 13-18 29298646-1 2018 BACKGROUND: The calcineurin inhibitor tacrolimus (Tac) is an integral part of the standard immunosuppressive regimen after renal transplantation (RTx). Tacrolimus 38-48 calcineurin binding protein 1 Homo sapiens 16-37 29161757-6 2018 Tacrolimus total daily dose at first therapeutic level was significantly higher in CYP3A5*1 expressers (12 mg/day) compared to nonexpressers (8 mg/day; P < .001). Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 83-89 29161757-9 2018 The findings from this retrospective study suggest that AAs with CYP3A5*1 expression require 50% more tacrolimus and have an increased incidence of DGF and acute rejection. Tacrolimus 102-112 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 65-71 30251601-0 2018 Influence of CYP3A and ABCB1 Single Nucleotide Polymorphisms on the Pharmacokinetics/Pharmacodynamics of Tacrolimus in Pediatric Patients. Tacrolimus 105-115 ATP binding cassette subfamily B member 1 Homo sapiens 23-28 30251601-4 2018 This article focuses on the effects of ABCB1 and CYP3A SNPs on tacrolimus in children who are undergoing organ transplantations. Tacrolimus 63-73 ATP binding cassette subfamily B member 1 Homo sapiens 39-44 30251601-4 2018 This article focuses on the effects of ABCB1 and CYP3A SNPs on tacrolimus in children who are undergoing organ transplantations. Tacrolimus 63-73 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 49-54 30251601-9 2018 However, although the amount of literature is limited, it does show a link between ABCB1 SNPs and tacrolimus pharmacodynamics. Tacrolimus 98-108 ATP binding cassette subfamily B member 1 Homo sapiens 83-88 30251601-10 2018 In addition, the literature shows a strong link between CYP3A5 SNP and pharmacokinetics of tacrolimus, but there is no direct evidence that CYP3A5 SNP has the same effect on the pharmacodynamics of tacrolimus. Tacrolimus 91-101 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 56-62 30251601-11 2018 CONCLUSION: More standardized clinical trials are needed to assess the relationship between CYP3A5 SNP and tacrolimus pharmacodynamics in children, particularly in terms of acute rejection and nephrotoxicity. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 92-98 30531101-6 2018 By contrast, NCX2+/- mice was correlated with elevated calcineurin (CaN) activity and rescued by treatment with the calcineurin inhibitor FK506. Tacrolimus 138-143 solute carrier family 8 (sodium/calcium exchanger), member 2 Mus musculus 13-17 30156148-9 2018 Notably, the CYP3A7-CYP3A4 switch taking place during the very early life will affect tacrolimus metabolism. Tacrolimus 86-96 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 13-26 30156148-11 2018 The guideline has recommended that the tacrolimus dosage should be adjusted according to the CYP3A5 genotype. Tacrolimus 39-49 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 93-99 30156148-12 2018 Additionally, genetic CYP3A4 variation (e.g., CYP3A4*22) is also associated with interindividual variability of exposure level to tacrolimus. Tacrolimus 130-140 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 22-28 30156148-12 2018 Additionally, genetic CYP3A4 variation (e.g., CYP3A4*22) is also associated with interindividual variability of exposure level to tacrolimus. Tacrolimus 130-140 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 46-52 29393157-0 2018 Tacrolimus Dose Optimization Strategy for Refractory Ulcerative Colitis Based on the Cytochrome P450 3A5 Polymorphism Prediction Using Trough Concentration after 24 Hours. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 85-104 29393157-2 2018 Cytochrome P450 (CYP) 3A5 polymorphism affects tacrolimus blood concentrations. Tacrolimus 47-57 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-25 29393157-7 2018 In the CYP3A5 non-expressers, the dose of tacrolimus (mg/kg) was lower and dose-adjusted trough levels (ng/mL per mg/kg) were higher compared with those in expressers. Tacrolimus 42-52 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 7-13 29393157-12 2018 Key Message: The trough concentration 24 h after the first tacrolimus administration appears to be a useful predictor of -CYP3A5 polymorphism. Tacrolimus 59-69 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 122-128 29375701-8 2018 In conclusion, the clearance of tacrolimus in patients with autoimmune diseases was affected by the CYP3A5 genotype, as previously reported in organ transplant patients. Tacrolimus 32-42 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 100-106 28617417-2 2018 FKBP14 is an ER-resident protein belonging to the family of FK506-binding peptidyl-prolyl cis-trans isomerases (PPIases); it catalyzes the folding of type III collagen and interacts with type III, type VI, and type X collagens. Tacrolimus 60-65 FKBP prolyl isomerase 14 Homo sapiens 0-6 29348725-0 2018 Protective effect of ginsenoside Rb1 against tacrolimus-induced apoptosis in renal proximal tubular LLC-PK1 cells. Tacrolimus 45-55 RB transcriptional corepressor 1 Sus scrofa 33-36 29348725-1 2018 Background: The aim of the present study was to evaluate the potential protective effects of six ginsenosides (Rb1, Rb2, Rc, Rd, Rg1, and Rg3) isolated from Panax ginseng against tacrolimus (FK506)-induced apoptosis in renal proximal tubular LLC-PK1 cells. Tacrolimus 179-189 RB transcriptional corepressor 1 Sus scrofa 111-114 29348725-1 2018 Background: The aim of the present study was to evaluate the potential protective effects of six ginsenosides (Rb1, Rb2, Rc, Rd, Rg1, and Rg3) isolated from Panax ginseng against tacrolimus (FK506)-induced apoptosis in renal proximal tubular LLC-PK1 cells. Tacrolimus 191-196 RB transcriptional corepressor 1 Sus scrofa 111-114 29348725-5 2018 Results: Reduction in cell viability by 60muM FK506 was ameliorated significantly by cotreatment with ginsenosides Rg1 and Rb1. Tacrolimus 46-51 RB transcriptional corepressor 1 Sus scrofa 123-126 29348725-6 2018 The phosphorylation of p38, extracellular signal-regulated kinases, and KIM-1, and cleavage of caspase-3, increased markedly in LLC-PK1 cells treated with FK506 and significantly decreased after cotreatment with ginsenoside Rb1. Tacrolimus 155-160 caspase 3 Sus scrofa 95-104 29348725-6 2018 The phosphorylation of p38, extracellular signal-regulated kinases, and KIM-1, and cleavage of caspase-3, increased markedly in LLC-PK1 cells treated with FK506 and significantly decreased after cotreatment with ginsenoside Rb1. Tacrolimus 155-160 RB transcriptional corepressor 1 Sus scrofa 224-227 29348725-8 2018 Conclusion: The antiapoptotic effects of ginsenoside Rb1 on FK506-induced apoptosis were mediated by the inhibition of mitogen-activated protein kinases and caspase activation. Tacrolimus 60-65 RB transcriptional corepressor 1 Sus scrofa 53-56 29199543-4 2018 NR1I2 and NR1I3 genetic polymorphisms can affect the pharmacokinetics and therapeutic response to many drugs, such as irinotecan, tacrolimus and atazanavir. Tacrolimus 130-140 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-5 28094348-0 2018 Weight of ABCB1 and POR genes on oral tacrolimus exposure in CYP3A5 nonexpressor pediatric patients with stable kidney transplant. Tacrolimus 38-48 ATP binding cassette subfamily B member 1 Homo sapiens 10-15 28094348-0 2018 Weight of ABCB1 and POR genes on oral tacrolimus exposure in CYP3A5 nonexpressor pediatric patients with stable kidney transplant. Tacrolimus 38-48 cytochrome p450 oxidoreductase Homo sapiens 20-23 28094348-0 2018 Weight of ABCB1 and POR genes on oral tacrolimus exposure in CYP3A5 nonexpressor pediatric patients with stable kidney transplant. Tacrolimus 38-48 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-67 29199543-4 2018 NR1I2 and NR1I3 genetic polymorphisms can affect the pharmacokinetics and therapeutic response to many drugs, such as irinotecan, tacrolimus and atazanavir. Tacrolimus 130-140 nuclear receptor subfamily 1 group I member 3 Homo sapiens 10-15 29023988-3 2018 However, the structural conservation of the FK1 domains between FKBP51 and FKBP52 make it difficult to obtain satisfactory selectivity in FK506-based drug design. Tacrolimus 138-143 FKBP prolyl isomerase 4 Homo sapiens 75-81 28522399-7 2017 Sacran solutions and 0.1%Tacrolimus reduced disease severity, suppressed histological changes and decreased the serum Th-1 (IFN-gamma, TNF-alpha, IL-2) and Th-2 (IL-4, IL-6, IL-10) cytokines in allergic mice (vs. controls). Tacrolimus 25-35 negative elongation factor complex member C/D, Th1l Mus musculus 118-122 29229832-3 2017 Calcineurin can be inhibited with Tacrolimus through the recruitment and inhibition of the 12-kDa cis-trans proline isomerase FK506-binding protein (FKBP12). Tacrolimus 34-44 FKBP prolyl isomerase 1A Rattus norvegicus 149-155 29229832-7 2017 Using a rat model of PD, partial elimination of the functional interaction between FKBP12 and calcineurin, with low doses of the Food and Drug Administration (FDA)-approved compound Tacrolimus, blocks calcineurin"s activity toward those proteins and protects against the toxic hallmarks of alpha-syn pathology. Tacrolimus 182-192 FKBP prolyl isomerase 1A Rattus norvegicus 83-89 28973643-0 2017 Tacrolimus rescues the signaling and gene expression signature of endothelial ALK1 loss-of-function and improves HHT vascular pathology. Tacrolimus 0-10 activin A receptor, type II-like 1 Mus musculus 78-82 28973643-4 2017 By screening the NIH clinical collections of FDA-approved drugs, we identified tacrolimus (FK-506) as the most potent activator of ALK1 signaling in BMP9-challenged C2C12 reporter cells. Tacrolimus 79-89 activin A receptor, type II-like 1 Mus musculus 131-135 28973643-4 2017 By screening the NIH clinical collections of FDA-approved drugs, we identified tacrolimus (FK-506) as the most potent activator of ALK1 signaling in BMP9-challenged C2C12 reporter cells. Tacrolimus 79-89 growth differentiation factor 2 Mus musculus 149-153 28973643-4 2017 By screening the NIH clinical collections of FDA-approved drugs, we identified tacrolimus (FK-506) as the most potent activator of ALK1 signaling in BMP9-challenged C2C12 reporter cells. Tacrolimus 91-97 activin A receptor, type II-like 1 Mus musculus 131-135 28973643-4 2017 By screening the NIH clinical collections of FDA-approved drugs, we identified tacrolimus (FK-506) as the most potent activator of ALK1 signaling in BMP9-challenged C2C12 reporter cells. Tacrolimus 91-97 growth differentiation factor 2 Mus musculus 149-153 28973643-5 2017 In HUVECs, tacrolimus activated Smad1/5/8 and opposed the pro-angiogenic gene expression signature associated with ALK1 loss-of-function, by notably reducing Dll4 expression. Tacrolimus 11-21 SMAD family member 1 Mus musculus 32-39 28973643-5 2017 In HUVECs, tacrolimus activated Smad1/5/8 and opposed the pro-angiogenic gene expression signature associated with ALK1 loss-of-function, by notably reducing Dll4 expression. Tacrolimus 11-21 activin A receptor, type II-like 1 Mus musculus 115-119 28973643-5 2017 In HUVECs, tacrolimus activated Smad1/5/8 and opposed the pro-angiogenic gene expression signature associated with ALK1 loss-of-function, by notably reducing Dll4 expression. Tacrolimus 11-21 delta like canonical Notch ligand 4 Mus musculus 158-162 28973643-6 2017 In these cells, tacrolimus also inhibited Akt and p38 stimulation by vascular endothelial growth factor, a major driver of angiogenesis. Tacrolimus 16-26 mitogen-activated protein kinase 14 Mus musculus 50-53 28973643-7 2017 In the BMP9/10-immunodepleted postnatal retina-a mouse model of HHT vascular pathology-tacrolimus activated endothelial Smad1/5/8 and prevented the Dll4 overexpression and hypervascularization associated with this model. Tacrolimus 87-97 growth differentiation factor 2 Mus musculus 7-11 28973643-7 2017 In the BMP9/10-immunodepleted postnatal retina-a mouse model of HHT vascular pathology-tacrolimus activated endothelial Smad1/5/8 and prevented the Dll4 overexpression and hypervascularization associated with this model. Tacrolimus 87-97 SMAD family member 1 Mus musculus 120-127 28973643-7 2017 In the BMP9/10-immunodepleted postnatal retina-a mouse model of HHT vascular pathology-tacrolimus activated endothelial Smad1/5/8 and prevented the Dll4 overexpression and hypervascularization associated with this model. Tacrolimus 87-97 delta like canonical Notch ligand 4 Mus musculus 148-152 28973643-8 2017 Finally, tacrolimus stimulated Smad1/5/8 signaling in C2C12 cells expressing BMP9-unresponsive ALK1 HHT mutants and in HHT patient blood outgrowth ECs. Tacrolimus 9-19 SMAD family member 1 Mus musculus 31-38 28973643-8 2017 Finally, tacrolimus stimulated Smad1/5/8 signaling in C2C12 cells expressing BMP9-unresponsive ALK1 HHT mutants and in HHT patient blood outgrowth ECs. Tacrolimus 9-19 growth differentiation factor 2 Mus musculus 77-81 28973643-8 2017 Finally, tacrolimus stimulated Smad1/5/8 signaling in C2C12 cells expressing BMP9-unresponsive ALK1 HHT mutants and in HHT patient blood outgrowth ECs. Tacrolimus 9-19 activin A receptor, type II-like 1 Mus musculus 95-99 28437851-5 2017 Predictors of tacrolimus clearance were CYP3A5 genotype, midazolam clearance, hematocrit, weight, and age (R2 = 0.61). Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 40-46 28522399-7 2017 Sacran solutions and 0.1%Tacrolimus reduced disease severity, suppressed histological changes and decreased the serum Th-1 (IFN-gamma, TNF-alpha, IL-2) and Th-2 (IL-4, IL-6, IL-10) cytokines in allergic mice (vs. controls). Tacrolimus 25-35 interferon gamma Mus musculus 124-133 28522399-7 2017 Sacran solutions and 0.1%Tacrolimus reduced disease severity, suppressed histological changes and decreased the serum Th-1 (IFN-gamma, TNF-alpha, IL-2) and Th-2 (IL-4, IL-6, IL-10) cytokines in allergic mice (vs. controls). Tacrolimus 25-35 tumor necrosis factor Mus musculus 135-144 28522399-7 2017 Sacran solutions and 0.1%Tacrolimus reduced disease severity, suppressed histological changes and decreased the serum Th-1 (IFN-gamma, TNF-alpha, IL-2) and Th-2 (IL-4, IL-6, IL-10) cytokines in allergic mice (vs. controls). Tacrolimus 25-35 interleukin 2 Mus musculus 146-150 28522399-7 2017 Sacran solutions and 0.1%Tacrolimus reduced disease severity, suppressed histological changes and decreased the serum Th-1 (IFN-gamma, TNF-alpha, IL-2) and Th-2 (IL-4, IL-6, IL-10) cytokines in allergic mice (vs. controls). Tacrolimus 25-35 heart and neural crest derivatives expressed 2 Mus musculus 156-160 28522399-7 2017 Sacran solutions and 0.1%Tacrolimus reduced disease severity, suppressed histological changes and decreased the serum Th-1 (IFN-gamma, TNF-alpha, IL-2) and Th-2 (IL-4, IL-6, IL-10) cytokines in allergic mice (vs. controls). Tacrolimus 25-35 interleukin 4 Mus musculus 162-166 28522399-7 2017 Sacran solutions and 0.1%Tacrolimus reduced disease severity, suppressed histological changes and decreased the serum Th-1 (IFN-gamma, TNF-alpha, IL-2) and Th-2 (IL-4, IL-6, IL-10) cytokines in allergic mice (vs. controls). Tacrolimus 25-35 interleukin 6 Mus musculus 168-172 28522399-7 2017 Sacran solutions and 0.1%Tacrolimus reduced disease severity, suppressed histological changes and decreased the serum Th-1 (IFN-gamma, TNF-alpha, IL-2) and Th-2 (IL-4, IL-6, IL-10) cytokines in allergic mice (vs. controls). Tacrolimus 25-35 interleukin 10 Mus musculus 174-179 28603840-1 2017 AIMS: The CYP3A metric 4beta-hydroxycholesterol (4betaOHC) has been shown to correlate with tacrolimus steady-state apparent oral clearance (CL/F). Tacrolimus 92-102 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 10-15 27378609-0 2017 Effect of ABCB1 diplotype on tacrolimus disposition in renal recipients depends on CYP3A5 and CYP3A4 genotype. Tacrolimus 29-39 ATP binding cassette subfamily B member 1 Homo sapiens 10-15 27378609-0 2017 Effect of ABCB1 diplotype on tacrolimus disposition in renal recipients depends on CYP3A5 and CYP3A4 genotype. Tacrolimus 29-39 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 83-89 27378609-0 2017 Effect of ABCB1 diplotype on tacrolimus disposition in renal recipients depends on CYP3A5 and CYP3A4 genotype. Tacrolimus 29-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 94-100 28885495-0 2017 Isoglycyrrhizinate Magnesium Enhances Hepatoprotective Effect of FK506 on Ischemia-Reperfusion Injury Through HMGB1 Inhibition in a Rat Model of Liver Transplantation. Tacrolimus 65-70 high mobility group box 1 Rattus norvegicus 110-115 28885495-10 2017 However, autophagy induced by FK506 also lead to high-mobility group box (HMGB) 1 release from nuclei, resulting in hepatocyte injury through triggering of p38 phosphorylation and chemokine release. Tacrolimus 30-35 mitogen activated protein kinase 14 Rattus norvegicus 156-159 28885495-12 2017 CONCLUSIONS: Iso could inhibit release of HMGB1 by FK506 and enhance the hepatoprotective effect of FK506 in rat LT. Tacrolimus 51-56 high mobility group box 1 Rattus norvegicus 42-47 28965948-9 2017 To further study the mechanism of FK506 protection effect, we discovered it could increase autophagy via inhibit mTOR pathway. Tacrolimus 34-39 mechanistic target of rapamycin kinase Mus musculus 113-117 28965948-10 2017 CONCLUSION: FK506 protect heart function after MI as it improved myocardial cells autophagy process via inhibiting mTOR pathway. Tacrolimus 12-17 mechanistic target of rapamycin kinase Mus musculus 115-119 29155873-1 2017 BACKGROUND: The immunosuppressive drug tacrolimus has the short-term effect of reducing proteinuria in patients with immunoglobulin A nephropathy (IgAN). Tacrolimus 39-49 IGAN1 Homo sapiens 147-151 29155873-17 2017 The use of tacrolimus for a short period of time for patients with IgAN temporarily reduces proteinuria, but the data showed no long-term efficacy regarding proteinuria reduction and improvement of renal function. Tacrolimus 11-21 IGAN1 Homo sapiens 67-71 28864813-4 2017 Sequestration of FKBP12 by rapamycin or tacrolimus activates hepcidin both in vitro and in murine hepatocytes. Tacrolimus 40-50 hepcidin antimicrobial peptide Mus musculus 61-69 28864813-5 2017 Acute tacrolimus treatment transiently increases hepcidin in wild-type mice. Tacrolimus 6-16 hepcidin antimicrobial peptide Mus musculus 49-57 28346662-3 2017 Here, we report three cases of anti-MDA5 antibody-associated DM with RP-ILD in which the patients were treated with combined-modality therapy, including high-dose prednisolone, tacrolimus, intravenous cyclophosphamide and intravenous immunoglobulin (IVIG). Tacrolimus 177-187 interferon induced with helicase C domain 1 Homo sapiens 36-40 28603840-8 2017 In a linear mixed model for the 14-day period, determinants of tacrolimus C0 were CYP3A5 genotype, haematocrit, age and weight (overall R2 = 0.179). Tacrolimus 63-73 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 82-88 28864749-5 2017 Subsequently, 32 patients were enrolled in a prospective, randomized, controlled study and randomly assigned to receive tacrolimus by CYP3A5 genotype plus Wuzhi tablet co-administration guided dosing (study group) or standard dosing (control group). Tacrolimus 120-130 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 134-140 28864749-8 2017 Our results show that CYP3A5 genotype plus Wuzhi tablet co-administration guided tacrolimus dosing is a promising therapy for CYP3A5 expressers in the early post-transplant stage, while further study with a larger sample size is required to prove these findings. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 126-132 28761977-9 2017 Calcineurin inhibitors ciclosporin A (CsA) and tacrolimus (FK-506) decreased Fgf23 gene expression and FGF23 protein production. Tacrolimus 47-57 fibroblast growth factor 23 Rattus norvegicus 77-82 28945481-0 2017 Long-Term Influence of CYP3A5, CYP3A4, ABCB1, and NR1I2 Polymorphisms on Tacrolimus Concentration in Chinese Renal Transplant Recipients. Tacrolimus 73-83 nuclear receptor subfamily 1 group I member 2 Homo sapiens 50-55 28945481-7 2017 RESULTS: The alleles CYP3A5*3 and CYP3A4*18B were significantly associated with dose-adjusted tacrolimus blood trough concentrations and had a strong time-genotype interaction with tacrolimus pharmacokinetics. Tacrolimus 94-104 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 21-27 28945481-7 2017 RESULTS: The alleles CYP3A5*3 and CYP3A4*18B were significantly associated with dose-adjusted tacrolimus blood trough concentrations and had a strong time-genotype interaction with tacrolimus pharmacokinetics. Tacrolimus 94-104 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 34-40 28945481-7 2017 RESULTS: The alleles CYP3A5*3 and CYP3A4*18B were significantly associated with dose-adjusted tacrolimus blood trough concentrations and had a strong time-genotype interaction with tacrolimus pharmacokinetics. Tacrolimus 181-191 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 21-27 28945481-7 2017 RESULTS: The alleles CYP3A5*3 and CYP3A4*18B were significantly associated with dose-adjusted tacrolimus blood trough concentrations and had a strong time-genotype interaction with tacrolimus pharmacokinetics. Tacrolimus 181-191 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 34-40 28945481-10 2017 Thus, genotyping of the CYP3A4 and CYP3A5 genes should be considered with respect to determining tacrolimus dose regimens during the post-transplantation period. Tacrolimus 97-107 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-30 28945481-10 2017 Thus, genotyping of the CYP3A4 and CYP3A5 genes should be considered with respect to determining tacrolimus dose regimens during the post-transplantation period. Tacrolimus 97-107 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 35-41 28901449-0 2017 FK506 suppresses hypoxia-induced inflammation and protects tight junction function via the CaN-NFATc1 signaling pathway in retinal microvascular epithelial cells. Tacrolimus 0-5 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 95-101 28901449-1 2017 The present study aimed to identify whether FK506 suppresses hypoxia-induced inflammation and protects tight junction function via the calcineurin-nuclear factor of activated T-cells 1 (CaN-NFATc1) signaling pathway in mouse retinal microvascular endothelial cells (mRMECs). Tacrolimus 44-49 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 190-196 28901449-6 2017 The TEER value was decreased following hypoxia, but increased following treatment with FK506 (1 and 10 microM) for 24 and 48 h. The protein expression of ZO-1 was also increased following FK506 treatment for 24 h at 1 and 10 microM. Tacrolimus 87-92 tight junction protein 1 Mus musculus 154-158 28901449-6 2017 The TEER value was decreased following hypoxia, but increased following treatment with FK506 (1 and 10 microM) for 24 and 48 h. The protein expression of ZO-1 was also increased following FK506 treatment for 24 h at 1 and 10 microM. Tacrolimus 188-193 tight junction protein 1 Mus musculus 154-158 28901449-9 2017 Following treatment with FK506, the level of total NFATc1 was downregulated and the level of phosphorylated NFATc1 was upregulated. Tacrolimus 25-30 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 51-57 28901449-9 2017 Following treatment with FK506, the level of total NFATc1 was downregulated and the level of phosphorylated NFATc1 was upregulated. Tacrolimus 25-30 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 108-114 28901449-10 2017 Taken together, FK506 suppressed injury to the tight junctions and downregulated the expression of inflammatory cytokines in hypoxia-induced mRMECs via the CaN-NFATc1 signaling pathway. Tacrolimus 16-21 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 160-166 29149986-2 2017 The aim of this work was to study the effects of cyclosporine (CsA), tacrolimus (FK-506), and rapamycin (RAPA) on the release of three local factors directly implicated in bone-remodeling regulation and apoptosis of human osteoblasts: interleukin (IL)-6, osteoprotegerin, and receptor activator of nuclear factor kappabeta (RANKL). Tacrolimus 69-79 TNF receptor superfamily member 11b Homo sapiens 255-270 29149986-2 2017 The aim of this work was to study the effects of cyclosporine (CsA), tacrolimus (FK-506), and rapamycin (RAPA) on the release of three local factors directly implicated in bone-remodeling regulation and apoptosis of human osteoblasts: interleukin (IL)-6, osteoprotegerin, and receptor activator of nuclear factor kappabeta (RANKL). Tacrolimus 69-79 TNF superfamily member 11 Homo sapiens 324-329 29149986-2 2017 The aim of this work was to study the effects of cyclosporine (CsA), tacrolimus (FK-506), and rapamycin (RAPA) on the release of three local factors directly implicated in bone-remodeling regulation and apoptosis of human osteoblasts: interleukin (IL)-6, osteoprotegerin, and receptor activator of nuclear factor kappabeta (RANKL). Tacrolimus 81-87 TNF receptor superfamily member 11b Homo sapiens 255-270 29149986-2 2017 The aim of this work was to study the effects of cyclosporine (CsA), tacrolimus (FK-506), and rapamycin (RAPA) on the release of three local factors directly implicated in bone-remodeling regulation and apoptosis of human osteoblasts: interleukin (IL)-6, osteoprotegerin, and receptor activator of nuclear factor kappabeta (RANKL). Tacrolimus 81-87 TNF superfamily member 11 Homo sapiens 324-329 29149986-8 2017 MAIN FINDINGS: A significant increase in IL-6 (mRNA and released protein) was observed in the presence of FK-506 and RAPA. Tacrolimus 106-112 interleukin 6 Homo sapiens 41-45 29151414-12 2017 The fewer tacrolimus daily dosage, the lower APACHE II score and levels of PCT and BNP, the more effective promotion of PaO2/FiO2 after NIV treatment, and the better curative effect is suggested. Tacrolimus 10-20 natriuretic peptide B Homo sapiens 83-86 28761977-9 2017 Calcineurin inhibitors ciclosporin A (CsA) and tacrolimus (FK-506) decreased Fgf23 gene expression and FGF23 protein production. Tacrolimus 47-57 fibroblast growth factor 23 Rattus norvegicus 103-108 28761977-9 2017 Calcineurin inhibitors ciclosporin A (CsA) and tacrolimus (FK-506) decreased Fgf23 gene expression and FGF23 protein production. Tacrolimus 59-65 fibroblast growth factor 23 Rattus norvegicus 77-82 28761977-9 2017 Calcineurin inhibitors ciclosporin A (CsA) and tacrolimus (FK-506) decreased Fgf23 gene expression and FGF23 protein production. Tacrolimus 59-65 fibroblast growth factor 23 Rattus norvegicus 103-108 29073235-10 2017 Finally, the calcineurin inhibitor FK506 and Cyclosporin A reversed the effect of SFKs inhibition on NFAT1. Tacrolimus 35-40 nuclear factor of activated T cells 2 Homo sapiens 101-106 29079741-1 2017 FK506 binding protein of 51 kDa (FKBP51) is a heat shock protein 90 (Hsp90) co-chaperone involved in the regulation of steroid hormone receptors activity. Tacrolimus 0-5 heat shock protein 90 alpha family class A member 1 Homo sapiens 46-67 29079741-1 2017 FK506 binding protein of 51 kDa (FKBP51) is a heat shock protein 90 (Hsp90) co-chaperone involved in the regulation of steroid hormone receptors activity. Tacrolimus 0-5 heat shock protein 90 alpha family class A member 1 Homo sapiens 69-74 29090089-8 2017 Moreover, we identified synergistic effects of sirtinol and FK506 on prolonging allograft survival, and sirtinol synergizes with FK506 to promote Foxp3 expression. Tacrolimus 129-134 forkhead box P3 Mus musculus 146-151 29090089-9 2017 CONCLUSION: Sirtinol, a Sirt1 inhibitor, may be a promising immunosuppressive drug to prevent the rejection reaction in combination with FK506. Tacrolimus 137-142 sirtuin 1 Mus musculus 24-29 29083842-0 2017 Retraction: Inhibition of Arachidonic Acid Release by Cytosolic Phospholipase A2 Is Involved in the Antiapoptotic Effect of FK506 and Cyclosporin A on Astrocytes Exposed to Simulated Ischemia In Vitro Tacrolimus 124-129 phospholipase A2 group IVA Homo sapiens 54-80 28271256-0 2017 Impact of the CYP3A5 genotype on the distributions of dose-adjusted trough concentrations and incidence of rejection in Japanese renal transplant recipients receiving different tacrolimus formulations. Tacrolimus 177-187 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 28851804-0 2017 Association of FK506 binding proteins with RyR channels - effect of CLIC2 binding on sub-conductance opening and FKBP binding. Tacrolimus 15-20 ryanodine receptor 2 Homo sapiens 43-46 28851804-0 2017 Association of FK506 binding proteins with RyR channels - effect of CLIC2 binding on sub-conductance opening and FKBP binding. Tacrolimus 15-20 chloride intracellular channel 2 Homo sapiens 68-73 28851804-2 2017 Beneficially low RyR activity and maximum conductance opening may be stabilised when RyRs bind to FK506 binding proteins (FKBPs) and destabilised by FKBP dissociation, with submaximal opening during RyR hyperactivity associated with myopathies and neurological disorders. Tacrolimus 98-103 ryanodine receptor 2 Homo sapiens 85-88 28851804-8 2017 There are significant FKBP and RyR isoform-specific actions of CLIC2, rapamycin and FK506 on FKBP-RyR association. Tacrolimus 84-89 ryanodine receptor 2 Homo sapiens 31-34 28851804-8 2017 There are significant FKBP and RyR isoform-specific actions of CLIC2, rapamycin and FK506 on FKBP-RyR association. Tacrolimus 84-89 ryanodine receptor 2 Homo sapiens 98-101 28982713-0 2017 Publisher"s Note: Association of FK506 binding proteins with RyR channels - effect of CLIC2 binding on sub-conductance opening and FKBP binding. Tacrolimus 33-38 ryanodine receptor 2 Homo sapiens 61-64 28982713-0 2017 Publisher"s Note: Association of FK506 binding proteins with RyR channels - effect of CLIC2 binding on sub-conductance opening and FKBP binding. Tacrolimus 33-38 chloride intracellular channel 2 Homo sapiens 86-91 28640392-5 2017 A histopathological analysis showed that both cyclosporin A and tacrolimus exacerbated the NOD1-mediated coronary arteritis in a dose-dependent manner. Tacrolimus 64-74 nucleotide-binding oligomerization domain containing 1 Mus musculus 91-95 29045960-1 2017 OBJECTIVE: Tacrolimus prolonged-release(PR) formulation is a new once-daily formulation of the calcineurin inhibitor tacrolimus, which is currently used in adult liver or kidney transplant patients,and is also gradually widely used in children with nephrotic syndrome.The present study was undertaken to preliminarily investigate the pharmacokinetic characteristics of tacrolimus PR in pediatric nephrotic syndrome recipients. Tacrolimus 11-21 calcineurin binding protein 1 Homo sapiens 95-116 29045960-1 2017 OBJECTIVE: Tacrolimus prolonged-release(PR) formulation is a new once-daily formulation of the calcineurin inhibitor tacrolimus, which is currently used in adult liver or kidney transplant patients,and is also gradually widely used in children with nephrotic syndrome.The present study was undertaken to preliminarily investigate the pharmacokinetic characteristics of tacrolimus PR in pediatric nephrotic syndrome recipients. Tacrolimus 117-127 calcineurin binding protein 1 Homo sapiens 95-116 29018183-7 2017 The calcineurin inhibitor, tacrolimus, was introduced on the fifth postoperative day. Tacrolimus 27-37 calcineurin binding protein 1 Homo sapiens 4-25 28271256-4 2017 RESULTS: Recipients with at least one CYP3A5*1 wild-type allele (EMs) and recipients with homozygous expression of the variant allele CYP3A5*3 (PMs) were significantly identified using the tacrolimus C 0h/D cut-off values of 2.77 and 0.85 ng/mL/mg, respectively, and discrimination rates of 75.3 and 85.4%, respectively, for Tac-BID and Tac-QD groups. Tacrolimus 189-199 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 134-140 28271256-5 2017 The %CV of the tacrolimus C 0h/D in CYP3A5 EMs taking Tac-QD was significantly lower than that in those taking Tac-BID (20.4 versus 23.3%, P = 0.003). Tacrolimus 15-25 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 28271256-7 2017 CONCLUSIONS: The tacrolimus C 0h/D values with definite cut-offs for CYP3A5 genotypes were specifically identified in Japanese renal transplant recipients taking Tac-QD. Tacrolimus 17-27 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 69-75 28710230-7 2017 FK506 and GSK3 inhibition showed the opposite effect regarding the NFATc3 localization of iCSCs. Tacrolimus 0-5 nuclear factor of activated T cells 3 Homo sapiens 67-73 28849081-6 2017 The results of the present study demonstrated that lercanidipine and FK506 inhibited Ang II-induced cardiomyocyte hypertrophy, as evidenced by decreases in fetal gene (atrial natriuretic peptide and brain natriuretic peptide) expression levels and cell surface area. Tacrolimus 69-74 angiotensinogen Rattus norvegicus 85-91 28849081-6 2017 The results of the present study demonstrated that lercanidipine and FK506 inhibited Ang II-induced cardiomyocyte hypertrophy, as evidenced by decreases in fetal gene (atrial natriuretic peptide and brain natriuretic peptide) expression levels and cell surface area. Tacrolimus 69-74 natriuretic peptide B Rattus norvegicus 199-224 28640063-0 2017 Pharmacodynamic Monitoring of Tacrolimus-Based Immunosuppression in CD14+ Monocytes After Kidney Transplantation. Tacrolimus 30-40 CD14 molecule Homo sapiens 68-72 28777242-1 2017 OBJECTIVES: Several genetic factors were identified to be responsible for interidividual variability in tacrolimus (TAC) pharmacokinetics, with the predominant role of CYP3A5 and CYP3A4 polymorphisms. Tacrolimus 104-114 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 168-174 28777242-1 2017 OBJECTIVES: Several genetic factors were identified to be responsible for interidividual variability in tacrolimus (TAC) pharmacokinetics, with the predominant role of CYP3A5 and CYP3A4 polymorphisms. Tacrolimus 104-114 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 179-185 28466977-4 2017 The study patients received daily dose of tacrolimus 1-3 mg based on initial Th1/Th2 cell ratio. Tacrolimus 42-52 negative elongation factor complex member C/D Homo sapiens 77-80 28669722-0 2017 Tacrolimus induces fibroblasts apoptosis and reduces epidural fibrosis by regulating miR-429 and its target of RhoE. Tacrolimus 0-10 microRNA 429 Rattus norvegicus 85-92 28669722-0 2017 Tacrolimus induces fibroblasts apoptosis and reduces epidural fibrosis by regulating miR-429 and its target of RhoE. Tacrolimus 0-10 mitogen-activated protein kinase kinase kinase 11 Rattus norvegicus 111-115 28669722-6 2017 The expression of miR-429 in fibroblasts treated with FK506 was determined by RT-qPCR. Tacrolimus 54-59 microRNA 429 Rattus norvegicus 18-25 28669722-11 2017 The results revealed FK506 induces fibroblast apoptosis and significantly downregulates miR-429 expression in fibroblasts. Tacrolimus 21-26 microRNA 429 Rattus norvegicus 88-95 28669722-14 2017 The rat model demonstrated miR-429 inhibition promotes fibroblast apoptosis and epidural fibrosis, which is consistent with the results of FK506 treatment. Tacrolimus 139-144 microRNA 429 Rattus norvegicus 27-34 28669722-15 2017 Our study demonstrates that FK506 induces fibroblast apoptosis and reduces epidural fibrosis by regulating miR-429 expression and its target of RhoE. Tacrolimus 28-33 microRNA 429 Rattus norvegicus 107-114 28669722-15 2017 Our study demonstrates that FK506 induces fibroblast apoptosis and reduces epidural fibrosis by regulating miR-429 expression and its target of RhoE. Tacrolimus 28-33 mitogen-activated protein kinase kinase kinase 11 Rattus norvegicus 144-148 28722255-3 2017 All of these agents are inhibitors of cytochrome P450 3A4, which plays a key role in metabolizing immunosuppressant drugs such as cyclosporine, tacrolimus, and sirolimus. Tacrolimus 144-154 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 38-57 28540692-0 2017 Effects of CYP3A5 polymorphisms on tacrolimus pharmacokinetics in pediatric kidney transplantation: a systematic review and meta-analysis of observational studies. Tacrolimus 35-45 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 11-17 28540692-1 2017 BACKGROUND: CYP3A5 genetic polymorphisms have been reported to be strongly associated with the tacrolimus pharmacokinetics in adult kidney transplantation. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 28540692-2 2017 However, there is no published meta-analysis in the influence of CYP3A5 variants on the requirements of the tacrolimus dose in pediatric renal-transplant recipients (RTRs). Tacrolimus 108-118 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 65-71 28540692-3 2017 We wished to determine the effects of CYP3A5 polymorphisms on tacrolimus pharmacokinetics in pediatric RTRs. Tacrolimus 62-72 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 38-44 28540692-7 2017 A significant difference was observed in the mean trough concentration/dose of tacrolimus between recipients carrying CYP3A5* 3/*3 variants (referred to as "non-expressers") and those carrying CYP3A5*1 (referred to as "expressers") [standard mean difference (SMD)=-1.09, 95% confidence interval (CI): -1.92 to -0.25, P=0.011]. Tacrolimus 79-89 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 118-124 28540692-9 2017 CONCLUSION: Our meta-analysis identified a positive correlation between CYP3A5 genotypes and tacrolimus pharmacokinetics in pediatric RTRs. Tacrolimus 93-103 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 72-78 28893866-2 2017 Activation of bone morphogenetic protein receptor 2 (BMPR2) signalling by FK506 (tacrolimus) reverses occlusive vasculopathy in rodent PAH models. Tacrolimus 74-79 bone morphogenetic protein receptor type 2 Homo sapiens 14-51 28893866-2 2017 Activation of bone morphogenetic protein receptor 2 (BMPR2) signalling by FK506 (tacrolimus) reverses occlusive vasculopathy in rodent PAH models. Tacrolimus 74-79 bone morphogenetic protein receptor type 2 Homo sapiens 53-58 28893866-2 2017 Activation of bone morphogenetic protein receptor 2 (BMPR2) signalling by FK506 (tacrolimus) reverses occlusive vasculopathy in rodent PAH models. Tacrolimus 81-91 bone morphogenetic protein receptor type 2 Homo sapiens 14-51 28893866-2 2017 Activation of bone morphogenetic protein receptor 2 (BMPR2) signalling by FK506 (tacrolimus) reverses occlusive vasculopathy in rodent PAH models. Tacrolimus 81-91 bone morphogenetic protein receptor type 2 Homo sapiens 53-58 28893866-5 2017 PAH patients had significantly lower BMPR2 expression in peripheral blood mononuclear cells versus healthy controls (n=13; p=0.005), which improved after FK506 treatment. Tacrolimus 154-159 bone morphogenetic protein receptor type 2 Homo sapiens 37-42 28893866-6 2017 While we observed that some patients responded with a pronounced increase in BMPR2 expression as well as improvement in 6-min walk distance, and serological and echocardiographic parameters of heart failure, these changes were not significant.Low-level FK506 is well tolerated and increases BMPR2 in subsets of PAH patients. Tacrolimus 253-258 bone morphogenetic protein receptor type 2 Homo sapiens 77-82 28893866-6 2017 While we observed that some patients responded with a pronounced increase in BMPR2 expression as well as improvement in 6-min walk distance, and serological and echocardiographic parameters of heart failure, these changes were not significant.Low-level FK506 is well tolerated and increases BMPR2 in subsets of PAH patients. Tacrolimus 253-258 bone morphogenetic protein receptor type 2 Homo sapiens 291-296 28224698-9 2017 In conclusion, CYP3A5 genotype and several clinical variables were identified as modulators of the tacrolimus-azole interaction, but these did not permit accurate predictions in individual patients. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 28466977-8 2017 CONCLUSION: We confirm our previous report that Th1/Th2 ratio can predict ART outcomes in patients with RIF and immunosuppressant treatment with tacrolimus, and peripheral blood Th1 cell levels were negatively correlated with pregnancy outcome. Tacrolimus 145-155 negative elongation factor complex member C/D Homo sapiens 48-51 28137823-14 2017 CONCLUSION: Topical tacrolimus is effective in reducing the photophobia in patients with APS-1-associated keratitis, but showed no effects on the severity of keratitis. Tacrolimus 20-30 autoimmune regulator Homo sapiens 89-94 28633129-4 2017 On the concentration level of 2-5mug/ml tacrolimus plus 10ng/ml PDGF-BB, combination of drugs could effectively promote ECs proliferation and migration, and meanwhile inhibit VSMCs proliferation and migration, and the inhibition of p-mTOR"s expression within VSMCs played an important role in this differentiated effect. Tacrolimus 40-50 mechanistic target of rapamycin kinase Homo sapiens 234-238 28508247-2 2017 Based mainly on the results of short-term studies, the calcineurin inhibitor tacrolimus prevails over the mammalian target of rapamycin (mTOR) inhibitors. Tacrolimus 77-87 calcineurin binding protein 1 Homo sapiens 55-76 28624888-1 2017 PURPOSE: The study aims to evaluate the impact of recipients" and donors" polymorphisms in multidrug resistance-associated protein 2 (MRP2) gene ABCC2 -24C>T and 1249G>A on disposition of mycophenolic acid (MPA) and their interaction with cyclosporine (CsA) (compared to tacrolimus, TAC) in stable de novo adult renal transplant patients of Croatian origin. Tacrolimus 277-287 ATP binding cassette subfamily C member 2 Homo sapiens 91-132 28624888-1 2017 PURPOSE: The study aims to evaluate the impact of recipients" and donors" polymorphisms in multidrug resistance-associated protein 2 (MRP2) gene ABCC2 -24C>T and 1249G>A on disposition of mycophenolic acid (MPA) and their interaction with cyclosporine (CsA) (compared to tacrolimus, TAC) in stable de novo adult renal transplant patients of Croatian origin. Tacrolimus 277-287 ATP binding cassette subfamily C member 2 Homo sapiens 134-138 28624888-1 2017 PURPOSE: The study aims to evaluate the impact of recipients" and donors" polymorphisms in multidrug resistance-associated protein 2 (MRP2) gene ABCC2 -24C>T and 1249G>A on disposition of mycophenolic acid (MPA) and their interaction with cyclosporine (CsA) (compared to tacrolimus, TAC) in stable de novo adult renal transplant patients of Croatian origin. Tacrolimus 277-287 ATP binding cassette subfamily C member 2 Homo sapiens 145-150 28135009-0 2017 ATP-binding cassette subfamily B member 1 1236C/T polymorphism significantly affects the therapeutic outcome of tacrolimus in patients with refractory ulcerative colitis. Tacrolimus 112-122 ATP binding cassette subfamily B member 1 Homo sapiens 0-41 28135009-2 2017 Both cytochrome P-450 3A5 (CYP3A5) and ATP-binding cassette subfamily B member 1 (ABCB1) associated with tacrolimus metabolism are known to have several genetic polymorphisms. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 5-25 28135009-2 2017 Both cytochrome P-450 3A5 (CYP3A5) and ATP-binding cassette subfamily B member 1 (ABCB1) associated with tacrolimus metabolism are known to have several genetic polymorphisms. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 27-33 28135009-4 2017 We aimed to investigate the influence of both CYP3A5 and ABCB1 polymorphisms on the efficacy of tacrolimus in ulcerative colitis treatment under the tight dose-adjusting strategy. Tacrolimus 96-106 ATP binding cassette subfamily B member 1 Homo sapiens 57-62 28135009-2 2017 Both cytochrome P-450 3A5 (CYP3A5) and ATP-binding cassette subfamily B member 1 (ABCB1) associated with tacrolimus metabolism are known to have several genetic polymorphisms. Tacrolimus 105-115 ATP binding cassette subfamily B member 1 Homo sapiens 39-80 28135009-2 2017 Both cytochrome P-450 3A5 (CYP3A5) and ATP-binding cassette subfamily B member 1 (ABCB1) associated with tacrolimus metabolism are known to have several genetic polymorphisms. Tacrolimus 105-115 ATP binding cassette subfamily B member 1 Homo sapiens 82-87 28135009-4 2017 We aimed to investigate the influence of both CYP3A5 and ABCB1 polymorphisms on the efficacy of tacrolimus in ulcerative colitis treatment under the tight dose-adjusting strategy. Tacrolimus 96-106 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 28720321-1 2017 AIMS: Tacrolimus (Tac) inhibits insulin secretion in a Tac-trough blood level dependent way early post-transplant in renal transplant recipients (Rtx). Tacrolimus 6-16 insulin Homo sapiens 32-39 28771227-0 2017 Klotho enhances FoxO3-mediated manganese superoxide dismutase expression by negatively regulating PI3K/AKT pathway during tacrolimus-induced oxidative stress. Tacrolimus 122-132 klotho Homo sapiens 0-6 28605053-0 2017 Prevalence of CYP3A5 Genomic Variances and Their Impact on Tacrolimus Dosing Requirements among Kidney Transplant Recipients in Eastern North Carolina. Tacrolimus 59-69 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 28605053-1 2017 To assess the prevalence of CYP3A5 genomic variances and their impact on tacrolimus (TAC) dosing requirements among kidney transplant recipients in eastern North Carolina, we conducted a single-center retrospective cohort study at a large tertiary care medical center. Tacrolimus 73-83 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 28-34 28214069-4 2017 Immunosuppressive agents, such as calcineurin inhibitors (tacrolimus, cyclosporine) are substrates of cytochromes P450 3A4 and P-gp. Tacrolimus 58-68 ATP binding cassette subfamily B member 1 Homo sapiens 127-131 28704257-0 2017 The combination of CYP3A4*22 and CYP3A5*3 single-nucleotide polymorphisms determines tacrolimus dose requirement after kidney transplantation. Tacrolimus 85-95 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-25 28704257-0 2017 The combination of CYP3A4*22 and CYP3A5*3 single-nucleotide polymorphisms determines tacrolimus dose requirement after kidney transplantation. Tacrolimus 85-95 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 28771227-0 2017 Klotho enhances FoxO3-mediated manganese superoxide dismutase expression by negatively regulating PI3K/AKT pathway during tacrolimus-induced oxidative stress. Tacrolimus 122-132 forkhead box O3 Homo sapiens 16-21 28771227-0 2017 Klotho enhances FoxO3-mediated manganese superoxide dismutase expression by negatively regulating PI3K/AKT pathway during tacrolimus-induced oxidative stress. Tacrolimus 122-132 AKT serine/threonine kinase 1 Homo sapiens 103-106 28050888-0 2017 A New CYP3A5*3 and CYP3A4*22 Cluster Influencing Tacrolimus Target Concentrations: A Population Approach. Tacrolimus 49-59 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 6-12 28901186-10 2017 The percentage of CD4+/CD25+/Foxp3+ regulatory T cells were significantly increased in the rADSC-IR-FK506 group as compared to controls. Tacrolimus 100-105 forkhead box P3 Homo sapiens 29-34 28901186-11 2017 Analysis of recipient peripheral blood revealed that transforming growth factor beta1 (TGFbeta1) was significantly increased in the rADSC-IR-FK506 group. Tacrolimus 141-146 transforming growth factor beta 1 Homo sapiens 53-85 28901186-11 2017 Analysis of recipient peripheral blood revealed that transforming growth factor beta1 (TGFbeta1) was significantly increased in the rADSC-IR-FK506 group. Tacrolimus 141-146 transforming growth factor beta 1 Homo sapiens 87-95 28050888-1 2017 BACKGROUND: Single nucleotide polymorphisms (SNPs) in the CYP3A5 and CYP3A4 genes have been reported to be an important cause of variability in the pharmacokinetics of tacrolimus in renal transplant patients. Tacrolimus 168-178 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 58-64 28050888-1 2017 BACKGROUND: Single nucleotide polymorphisms (SNPs) in the CYP3A5 and CYP3A4 genes have been reported to be an important cause of variability in the pharmacokinetics of tacrolimus in renal transplant patients. Tacrolimus 168-178 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-75 28050888-8 2017 CONCLUSIONS: Tacrolimus disposition in renal transplant recipients was described using a new population pharmacokinetic model that included the CYP3A5*3 and CYP3A4*22 genotype, age, and hematocrit. Tacrolimus 13-23 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 144-150 28050888-8 2017 CONCLUSIONS: Tacrolimus disposition in renal transplant recipients was described using a new population pharmacokinetic model that included the CYP3A5*3 and CYP3A4*22 genotype, age, and hematocrit. Tacrolimus 13-23 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 157-163 28663197-6 2017 Treatment of neurons with the calcineurin inhibitors FK506 or cyclosporin A resulted in nuclear accumulation of phospho-HDAC1 and was neuroprotective. Tacrolimus 53-58 histone deacetylase 1 Mus musculus 120-125 28050888-0 2017 A New CYP3A5*3 and CYP3A4*22 Cluster Influencing Tacrolimus Target Concentrations: A Population Approach. Tacrolimus 49-59 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-25 28401703-0 2017 Tacrolimus population pharmacokinetics according to CYP3A5 genotype and clinical factors in Chinese adult kidney transplant recipients. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 52-58 28621555-0 2017 SLC28A3 rs7853758 as a new biomarker of tacrolimus elimination and new-onset hypertension in Chinese liver transplantation patients. Tacrolimus 40-50 solute carrier family 28 member 3 Homo sapiens 0-7 28621555-4 2017 The model of donors" CYP3A5 rs776746 and recipients" CYP3A4 rs2242480 could predict tacrolimus metabolism at week 1 and the model of donors" CYP3A5 rs776746, recipients" CYP3A4 rs2242480, recipients" SLC28A3 rs7853758 and hemoglobin could predict tacrolimus disposition at weeks 2, 3 and 4. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 21-27 28621555-4 2017 The model of donors" CYP3A5 rs776746 and recipients" CYP3A4 rs2242480 could predict tacrolimus metabolism at week 1 and the model of donors" CYP3A5 rs776746, recipients" CYP3A4 rs2242480, recipients" SLC28A3 rs7853758 and hemoglobin could predict tacrolimus disposition at weeks 2, 3 and 4. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 53-59 28621555-4 2017 The model of donors" CYP3A5 rs776746 and recipients" CYP3A4 rs2242480 could predict tacrolimus metabolism at week 1 and the model of donors" CYP3A5 rs776746, recipients" CYP3A4 rs2242480, recipients" SLC28A3 rs7853758 and hemoglobin could predict tacrolimus disposition at weeks 2, 3 and 4. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 141-147 28621555-4 2017 The model of donors" CYP3A5 rs776746 and recipients" CYP3A4 rs2242480 could predict tacrolimus metabolism at week 1 and the model of donors" CYP3A5 rs776746, recipients" CYP3A4 rs2242480, recipients" SLC28A3 rs7853758 and hemoglobin could predict tacrolimus disposition at weeks 2, 3 and 4. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 170-176 28621555-4 2017 The model of donors" CYP3A5 rs776746 and recipients" CYP3A4 rs2242480 could predict tacrolimus metabolism at week 1 and the model of donors" CYP3A5 rs776746, recipients" CYP3A4 rs2242480, recipients" SLC28A3 rs7853758 and hemoglobin could predict tacrolimus disposition at weeks 2, 3 and 4. Tacrolimus 84-94 solute carrier family 28 member 3 Homo sapiens 200-207 28621555-6 2017 CONCLUSION: Rs7853758 in recipients" SLC28A3 has a correlation with tacrolimus pharmacokinetics and the risk of NOHP in Chinese LT patients. Tacrolimus 68-78 solute carrier family 28 member 3 Homo sapiens 37-44 28533324-0 2017 CRISPR/Cas9 Genetic Modification of CYP3A5 *3 in HuH-7 Human Hepatocyte Cell Line Leads to Cell Lines with Increased Midazolam and Tacrolimus Metabolism. Tacrolimus 131-141 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 28533324-0 2017 CRISPR/Cas9 Genetic Modification of CYP3A5 *3 in HuH-7 Human Hepatocyte Cell Line Leads to Cell Lines with Increased Midazolam and Tacrolimus Metabolism. Tacrolimus 131-141 MIR7-3 host gene Homo sapiens 49-54 28533324-3 2017 In metabolism assays, HuH-7 had less tacrolimus (all P < 0.05) or midazolam (MDZ) (all P < 0.005) disappearance than all engineered cell lines. Tacrolimus 37-47 MIR7-3 host gene Homo sapiens 22-27 28401703-7 2017 In this study, we observed that POD, HCT and CYP3A5*3 genotype were determinant factors in CL/F and POD related with V/F of tacrolimus significantly. Tacrolimus 124-134 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 28401703-10 2017 WHAT IS NEW AND CONCLUSION: The HCT, CYP3A5*3 genetic polymorphism and POD contributed to the interindividual variability of oral tacrolimus in Chinese adult renal transplant patients. Tacrolimus 130-140 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 29050276-0 2017 A new donors" CYP3A5 and recipients" CYP3A4 cluster predicting tacrolimus disposition, and new-onset hypertension in Chinese liver transplant patients. Tacrolimus 63-73 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 28299629-0 2017 Inhibition of Calcineurin A by FK506 Suppresses Seizures and Reduces the Expression of GluN2B in Membrane Fraction. Tacrolimus 31-36 glutamate ionotropic receptor NMDA type subunit 2B Rattus norvegicus 87-93 28299629-4 2017 Thus, we hypothesized that one of the potential anti-epileptic mechanisms of FK506 is mediated by its ability to promote the phosphorylation of GluN2B and reduce the expression of GluN2B in membrane fraction by down-regulating CaNA. Tacrolimus 77-82 glutamate ionotropic receptor NMDA type subunit 2B Rattus norvegicus 144-150 28299629-4 2017 Thus, we hypothesized that one of the potential anti-epileptic mechanisms of FK506 is mediated by its ability to promote the phosphorylation of GluN2B and reduce the expression of GluN2B in membrane fraction by down-regulating CaNA. Tacrolimus 77-82 glutamate ionotropic receptor NMDA type subunit 2B Rattus norvegicus 180-186 28299629-8 2017 The levels of phosphorylated GluN2B were decreased in epileptic rats but increased after the FK506 treatment. Tacrolimus 93-98 glutamate ionotropic receptor NMDA type subunit 2B Rattus norvegicus 29-35 28299629-10 2017 However, the expression of GluN2B in membrane fraction was suppressed after FK506 treatment. Tacrolimus 76-81 glutamate ionotropic receptor NMDA type subunit 2B Rattus norvegicus 27-33 28299629-11 2017 Based on these results, FK506 may reduce the severity and frequency of seizures by reducing the expression of GluN2B in membrane fraction. Tacrolimus 24-29 glutamate ionotropic receptor NMDA type subunit 2B Rattus norvegicus 110-116 29050276-0 2017 A new donors" CYP3A5 and recipients" CYP3A4 cluster predicting tacrolimus disposition, and new-onset hypertension in Chinese liver transplant patients. Tacrolimus 63-73 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-43 28738959-10 2017 Moreover, FK506 increased the expression of P27 and RB1, and reduced the expression of CDK4, CDK6 and MMP9. Tacrolimus 10-15 dynactin subunit 6 Homo sapiens 44-47 28738959-10 2017 Moreover, FK506 increased the expression of P27 and RB1, and reduced the expression of CDK4, CDK6 and MMP9. Tacrolimus 10-15 RB transcriptional corepressor 1 Homo sapiens 52-55 28738959-10 2017 Moreover, FK506 increased the expression of P27 and RB1, and reduced the expression of CDK4, CDK6 and MMP9. Tacrolimus 10-15 cyclin dependent kinase 4 Homo sapiens 87-91 28738959-10 2017 Moreover, FK506 increased the expression of P27 and RB1, and reduced the expression of CDK4, CDK6 and MMP9. Tacrolimus 10-15 cyclin dependent kinase 6 Homo sapiens 93-97 28738959-10 2017 Moreover, FK506 increased the expression of P27 and RB1, and reduced the expression of CDK4, CDK6 and MMP9. Tacrolimus 10-15 matrix metallopeptidase 9 Homo sapiens 102-106 28146606-2 2017 Tacrolimus dosing is challenged by considerable pharmacokinetic variability, both between patients and over time after transplantation, partly due to variability in cytochrome P450 3A (CYP3A) activity. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 165-183 28673995-3 2017 Abrogation of host IDO expression by deletion of the IDO gene or the IFN-gamma gene in donor T cells or by FK506 treatment resulted in acute lethal pulmonary inflammation known as idiopathic pneumonia syndrome (IPS). Tacrolimus 107-112 indoleamine 2,3-dioxygenase 1 Homo sapiens 19-22 28673995-5 2017 Accordingly, a histone deacetylase inhibitor (HDACi) could reduce IPS in the state where IFN-gamma expression was suppressed by FK506. Tacrolimus 128-133 interferon gamma Homo sapiens 89-98 28747916-4 2017 Here, we determined the effect of tacrolimus and MPA on DNA methylation of the gene promoter region of interferon gamma (IFNgamma), a pro-inflammatory cytokine. Tacrolimus 34-44 interferon gamma Homo sapiens 103-130 28747916-12 2017 IFNgamma protein production was suppressed by tacrolimus. Tacrolimus 46-56 interferon gamma Homo sapiens 0-8 28146606-2 2017 Tacrolimus dosing is challenged by considerable pharmacokinetic variability, both between patients and over time after transplantation, partly due to variability in cytochrome P450 3A (CYP3A) activity. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 185-190 28146606-3 2017 The aim of this study was to assess the value of the endogenous CYP3A marker 4beta-hydroxycholesterol (4betaOHC) for tacrolimus dose individualization early after kidney transplantation. Tacrolimus 117-127 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 64-69 29270498-12 2017 Proteinuria (urinary protein-to-creatinine ratio) was significantly reduced in both patients with MGN and those with FSGS after 6 months of ACTHar gel alone and was further reduced among the patients with MGN with the addition of tacrolimus. Tacrolimus 230-240 actinin alpha 4 Homo sapiens 117-121 27572744-9 2017 Moreover, the results in hypoxia-treated primary spinal cord microglia confirmed the effect of FK-506 on TNF-a, IL-1b, and IL-6 expression and NF-kappaB activation. Tacrolimus 95-101 interleukin 1 beta Rattus norvegicus 112-117 27572744-9 2017 Moreover, the results in hypoxia-treated primary spinal cord microglia confirmed the effect of FK-506 on TNF-a, IL-1b, and IL-6 expression and NF-kappaB activation. Tacrolimus 95-101 interleukin 6 Rattus norvegicus 123-127 28584011-2 2017 We and others have reported that cyclosporine A and tacrolimus decrease anti-inflammatory regulatory T cells and increase proinflammatory interleukin-17-producing T cells; therefore, we hypothesized that inhibition of these effects using noncellular therapies would prevent the hypertension, endothelial dysfunction, and renal glomerular injury induced by calcineurin inhibitor therapy. Tacrolimus 52-62 calcineurin binding protein 1 Mus musculus 356-377 28584011-3 2017 Daily treatment of mice with cyclosporine A or tacrolimus for 1 week significantly decreased CD4+/FoxP3+ regulatory T cells in the spleen and lymph nodes, as well as induced hypertension, vascular injury and dysfunction, and glomerular mesangial expansion in mice. Tacrolimus 47-57 forkhead box P3 Mus musculus 98-103 28706520-7 2017 Notably, NFAT antagonism by cyclosporin A or FK506 impaired IL-22 upregulation at normothermia and entirely prevented its enhanced expression upon hypothermic culture conditions. Tacrolimus 45-50 interleukin 22 Mus musculus 60-65 28642710-0 2017 Tacrolimus Updated Guidelines through popPK Modeling: How to Benefit More from CYP3A Pre-emptive Genotyping Prior to Kidney Transplantation. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 79-84 28642710-4 2017 The integration of CYP3A4 and CY3A5 genotype in tacrolimus population-based PK (PopPK) modeling approaches has been proven to accurately predict the dose requirement to reach the therapeutic window. Tacrolimus 48-58 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-25 28642710-10 2017 Our model estimated that tacrolimus concentrations were 33% IC95%[20-26%], 41% IC95%[36-45%] lower in CYP3A IM and EM when compared to PM, respectively. Tacrolimus 25-35 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 102-107 28257599-7 2017 Treatment with tacrolimus and cyclosporin render CD4+ CD25- cells more susceptible to Treg control. Tacrolimus 15-25 CD4 molecule Homo sapiens 49-52 28257599-7 2017 Treatment with tacrolimus and cyclosporin render CD4+ CD25- cells more susceptible to Treg control. Tacrolimus 15-25 interleukin 2 receptor subunit alpha Homo sapiens 54-58 28672892-10 2017 On day 30 following intervention, the expression of IL-17 and CCL2 in the prostate of rats in the tacrolimus and celecoxib groups was significantly downregulated compared with the NS group (P<0.05). Tacrolimus 98-108 interleukin 17A Rattus norvegicus 52-57 28672892-10 2017 On day 30 following intervention, the expression of IL-17 and CCL2 in the prostate of rats in the tacrolimus and celecoxib groups was significantly downregulated compared with the NS group (P<0.05). Tacrolimus 98-108 C-C motif chemokine ligand 2 Rattus norvegicus 62-66 28560399-8 2017 Verification of the miRNA-target genes revealed that Smad5, Jagged 1 and MAPK9 were significantly upregulated, whereas Smad7, BMP and activin membrane-bound inhibitor, and dual-specificity phosphatase 2 were significantly downregulated during FK506-induced osteodifferentiation. Tacrolimus 243-248 dual specificity phosphatase 2 Rattus norvegicus 134-202 28672869-4 2017 Here, we showed that mutations of tax-6 and cnb-1, which respectively encode the catalytic subunit and the regulatory subunit of calcineurin, together with tacrolimus treatment, consistently led to decreased fat accumulation and delayed growth in the nematode Caenorhabditis elegans. Tacrolimus 156-166 Serine/threonine-protein phosphatase 2B catalytic subunit Caenorhabditis elegans 34-39 29259481-8 2017 Conclusion: For recurrent miscarriage cases that show an elevated Th1/Th2 cell ratio after achieving pregnancy, immunosuppressive treatment with tacrolimus could be effective. Tacrolimus 145-155 negative elongation factor complex member C/D Homo sapiens 66-69 28669959-0 2017 [Correlation of blood concentration of tacrolimus with serum cystatin C in renal transplant recipients and effect of tacrolimus on glucose and lipid metabolism]. Tacrolimus 39-49 cystatin C Homo sapiens 61-71 28669959-1 2017 OBJECTIVE: To investigate the correlation between blood concentrations of tacrolimus (FK506) and cystatin C (Cys C) and the effect of FK506 on glycolipid metabolism in renal transplant recipients. Tacrolimus 74-84 cystatin C Homo sapiens 97-107 28669959-1 2017 OBJECTIVE: To investigate the correlation between blood concentrations of tacrolimus (FK506) and cystatin C (Cys C) and the effect of FK506 on glycolipid metabolism in renal transplant recipients. Tacrolimus 74-84 cystatin C Homo sapiens 109-114 28669959-1 2017 OBJECTIVE: To investigate the correlation between blood concentrations of tacrolimus (FK506) and cystatin C (Cys C) and the effect of FK506 on glycolipid metabolism in renal transplant recipients. Tacrolimus 86-91 cystatin C Homo sapiens 97-107 28669959-4 2017 Results Plasma FK506 concentration decreased with age in the recipients and showed a positive correlation with Cys C (r=0.985, P=0.015) but no obvious correlation with Scr (r=0.259, P=0.741). Tacrolimus 15-20 cystatin C Homo sapiens 111-116 28669959-7 2017 Cys C is positively related to blood concentration of FK506 in the renal transplantation recipients. Tacrolimus 54-59 cystatin C Homo sapiens 0-5 28611384-7 2017 Compared to the high tacrolimus group, expression levels of six cytochrome P450 enzymes, CYP1A1, CYP2B6, CYP3A5, CYP4A11, CYP19A1, and CYP17A1 were significantly higher in the low tacrolimus group. Tacrolimus 180-190 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 89-95 28611384-7 2017 Compared to the high tacrolimus group, expression levels of six cytochrome P450 enzymes, CYP1A1, CYP2B6, CYP3A5, CYP4A11, CYP19A1, and CYP17A1 were significantly higher in the low tacrolimus group. Tacrolimus 180-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-103 28611384-7 2017 Compared to the high tacrolimus group, expression levels of six cytochrome P450 enzymes, CYP1A1, CYP2B6, CYP3A5, CYP4A11, CYP19A1, and CYP17A1 were significantly higher in the low tacrolimus group. Tacrolimus 180-190 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 105-111 28611384-7 2017 Compared to the high tacrolimus group, expression levels of six cytochrome P450 enzymes, CYP1A1, CYP2B6, CYP3A5, CYP4A11, CYP19A1, and CYP17A1 were significantly higher in the low tacrolimus group. Tacrolimus 180-190 cytochrome P450 family 4 subfamily A member 11 Homo sapiens 113-120 28611384-7 2017 Compared to the high tacrolimus group, expression levels of six cytochrome P450 enzymes, CYP1A1, CYP2B6, CYP3A5, CYP4A11, CYP19A1, and CYP17A1 were significantly higher in the low tacrolimus group. Tacrolimus 180-190 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-129 28611384-7 2017 Compared to the high tacrolimus group, expression levels of six cytochrome P450 enzymes, CYP1A1, CYP2B6, CYP3A5, CYP4A11, CYP19A1, and CYP17A1 were significantly higher in the low tacrolimus group. Tacrolimus 180-190 cytochrome P450 family 17 subfamily A member 1 Homo sapiens 135-142 28611384-9 2017 Enzyme assays showed that CYP3A5 and CYP17A1 exerted direct metabolic effects on tacrolimus and mycophenolate mofetil, respectively. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 26-32 28611384-9 2017 Enzyme assays showed that CYP3A5 and CYP17A1 exerted direct metabolic effects on tacrolimus and mycophenolate mofetil, respectively. Tacrolimus 81-91 cytochrome P450 family 17 subfamily A member 1 Homo sapiens 37-44 29270498-14 2017 Discussion: Combination therapy with ACTHar gel and tacrolimus was well tolerated by patients with treatment-resistant MGN and FSGS and significantly reduced proteinuria and improved clinical response rates compared with ACTHar gel alone. Tacrolimus 52-62 actinin alpha 4 Homo sapiens 127-131 28008657-0 2017 Bodyweight-adjustments introduce significant correlations between CYP3A metrics and tacrolimus clearance. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 66-71 28324194-0 2017 Sex Differences in the Blood Concentration of Tacrolimus in Systemic Lupus Erythematosus and Rheumatoid Arthritis Patients with CYP3A5*3/*3. Tacrolimus 46-56 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 128-134 28324194-1 2017 The purpose of this study was to describe the impact of sex and cytochrome P450 3A5 (CYP3A5) variant on the blood concentration of tacrolimus in patients with systemic lupus erythematosus or rheumatoid arthritis. Tacrolimus 131-141 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 64-83 28324194-1 2017 The purpose of this study was to describe the impact of sex and cytochrome P450 3A5 (CYP3A5) variant on the blood concentration of tacrolimus in patients with systemic lupus erythematosus or rheumatoid arthritis. Tacrolimus 131-141 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 85-91 28324194-5 2017 In the CYP3A5*3/*3 group, the concentration of tacrolimus was significantly higher in men than in women (p < 0.05, effect size: d = 1.78). Tacrolimus 47-57 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 7-13 28324194-6 2017 Furthermore, in the CYP3A5*3/*3 group, the concentration of tacrolimus was significantly higher in women aged over 50 years than in women aged under 50 years (p < 0.05, effect size: d = 1.18). Tacrolimus 60-70 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 28324194-8 2017 Since the blood concentration of tacrolimus in patients with CYP3A5*3/*3 varies depending on sex and age, these factors should be considered when studying the difference of sex in CYP3A. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-67 28324194-8 2017 Since the blood concentration of tacrolimus in patients with CYP3A5*3/*3 varies depending on sex and age, these factors should be considered when studying the difference of sex in CYP3A. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 61-66 28374426-0 2017 Response to: "Response to: Bodyweight-adjustments introduce significant correlations between CYP3A metrics and tacrolimus clearance". Tacrolimus 111-121 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-98 28044353-0 2017 Donor CYP3A5 genotype influences tacrolimus disposition on the first day after paediatric liver transplantation. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 6-12 28044353-2 2017 METHODS: The contribution of the CYP3A4/5 genotype in paediatric liver transplant recipients and donors to the tacrolimus blood trough concentrations (C0 ) and the tacrolimus concentration/weight-adjusted dose ratio on day 1 was evaluated in 67 liver-transplanted children: 33 boys and 34 girls, mean age 4.5 years. Tacrolimus 111-121 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 33-39 28044353-3 2017 RESULTS: Donor CYP3A5 genotype appears to be significantly associated with tacrolimus disposition on the first day after liver transplantation (P < 0.0002). Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 28044353-7 2017 CONCLUSIONS: Donor CYP3A5 genotype, recipient age and, to a lesser extent, donor gender appear to be associated with tacrolimus disposition on day 1 after transplant. Tacrolimus 117-127 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 19-25 28168728-0 2017 Response to: "Bodyweight-adjustments introduce significant correlations between CYP3A metrics and tacrolimus clearance". Tacrolimus 98-108 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 80-85 28342282-10 2017 Patients expressing CYP3A5*1 genotypes had lower tacrolimus AUC0-24 values vs those with CYP3A5*3/*3 (IR-TACP<.001; ER-TACP=.008). Tacrolimus 49-59 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 28611128-6 2017 Knockdown of KCNE2 increased intracellular Ca2+ transient, calcineurin activity, and nuclear NFAT (nuclear factor of activated T cells) protein levels, and pretreatment with inhibitor of L-type Ca2+ channel (nifedipine) or calcineurin (FK506) attenuated the activation of calcineurin-NFAT pathway and cardiomyocyte hypertrophy. Tacrolimus 236-241 potassium voltage-gated channel subfamily E regulatory subunit 2 Rattus norvegicus 13-18 28339015-7 2017 It was hypothesized that FK506 may inhibit the activity of DAPK1 by inhibiting calcineurin activity, which may be primarily involved in anti-apoptosis following DAI induction. Tacrolimus 25-30 death associated protein kinase 1 Homo sapiens 59-64 28229376-1 2017 INTRODUCTION: Tacrolimus is a calcineurin inhibitor used as an immunosuppressant drug in solid organ transplantation, and is mainly metabolized by cytochrome P450 (CYP) 3A4 and CYP3A5. Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 147-172 28229376-1 2017 INTRODUCTION: Tacrolimus is a calcineurin inhibitor used as an immunosuppressant drug in solid organ transplantation, and is mainly metabolized by cytochrome P450 (CYP) 3A4 and CYP3A5. Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 177-183 28229376-2 2017 Studies have shown an association between the CYP3A5 genotype and tacrolimus dose-adjusted trough concentrations. Tacrolimus 66-76 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 28229376-3 2017 Variants in the genes PPARA, POR and CYP3A4 have recently been shown to influence tacrolimus metabolism. Tacrolimus 82-92 peroxisome proliferator activated receptor alpha Homo sapiens 22-27 28229376-3 2017 Variants in the genes PPARA, POR and CYP3A4 have recently been shown to influence tacrolimus metabolism. Tacrolimus 82-92 cytochrome p450 oxidoreductase Homo sapiens 29-32 28229376-3 2017 Variants in the genes PPARA, POR and CYP3A4 have recently been shown to influence tacrolimus metabolism. Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-43 28229376-5 2017 In the present study, we investigated a potential association between CYP3A5*3, PPARA c.209-1003G>A, POR*28 and CYP3A4*22 and dose-adjusted tacrolimus trough concentrations in a primarily corticosteroid-free (>85%) population of Danish pediatric and adult kidney transplant recipients. Tacrolimus 143-153 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 70-76 28229376-5 2017 In the present study, we investigated a potential association between CYP3A5*3, PPARA c.209-1003G>A, POR*28 and CYP3A4*22 and dose-adjusted tacrolimus trough concentrations in a primarily corticosteroid-free (>85%) population of Danish pediatric and adult kidney transplant recipients. Tacrolimus 143-153 peroxisome proliferator activated receptor alpha Homo sapiens 80-85 28229376-5 2017 In the present study, we investigated a potential association between CYP3A5*3, PPARA c.209-1003G>A, POR*28 and CYP3A4*22 and dose-adjusted tacrolimus trough concentrations in a primarily corticosteroid-free (>85%) population of Danish pediatric and adult kidney transplant recipients. Tacrolimus 143-153 cytochrome p450 oxidoreductase Homo sapiens 104-107 28229376-5 2017 In the present study, we investigated a potential association between CYP3A5*3, PPARA c.209-1003G>A, POR*28 and CYP3A4*22 and dose-adjusted tacrolimus trough concentrations in a primarily corticosteroid-free (>85%) population of Danish pediatric and adult kidney transplant recipients. Tacrolimus 143-153 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 115-121 28229376-8 2017 RESULTS: It was confirmed that CYP3A5*1 wild-type carriers had lower median dose-adjusted tacrolimus trough concentrations compared with noncarriers. Tacrolimus 90-100 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 31-37 28229376-13 2017 CONCLUSION: This study shows that the known association of the CYP3A5 genotype with tacrolimus dose-adjusted trough concentrations has the same impact in a corticosteroid-sparse population. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 63-69 28620390-7 2017 The proportion of IL-21+-activated Tfh-cells was partially decreased by in vitro addition of belatacept or tacrolimus (by ~60%). Tacrolimus 107-117 interleukin 21 Homo sapiens 18-23 29113387-0 2017 Tacrolimus dose requirement based on the CYP3A5 genotype in renal transplant patients. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 41-47 29113387-2 2017 The aim of the present study is to determine whether the single nucleotide polymorphism of CYP3A5 is a predictive index of FK506 dose requirement, and also the selection yardstick of FK506 or CsA treatment.We tested archival peripheral blood of 218 kidney recipients for CYP3A5 genotyping with PCR-SSP. Tacrolimus 123-128 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 91-97 29113387-3 2017 Meanwhile, the dose of FK506 and CsA was recorded, blood concentration of the drugs was measured, and graft outcome was monitored.These results indicate that CYP3A5*AA/AG carriers need higher FK506 dose than CYP3A5*GG homozygote to achieve the target blood concentration. Tacrolimus 23-28 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 158-164 29123971-4 2017 Genome wide association study data for the immunosuppressant tacrolimus have identified multiple variants in the CYP3A5 gene associated with trough concentrations. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 113-119 29113387-3 2017 Meanwhile, the dose of FK506 and CsA was recorded, blood concentration of the drugs was measured, and graft outcome was monitored.These results indicate that CYP3A5*AA/AG carriers need higher FK506 dose than CYP3A5*GG homozygote to achieve the target blood concentration. Tacrolimus 192-197 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 158-164 29113387-4 2017 For CYP3A5*GG carriers, taking FK506 or CsA are both advisable. Tacrolimus 31-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 29113387-6 2017 CYP3A5 genotyping is a new approach to detecting FK506 dose requirement and a predictive index for the FK506 or CsA treatment selection in kidney recipients. Tacrolimus 49-54 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 29113387-6 2017 CYP3A5 genotyping is a new approach to detecting FK506 dose requirement and a predictive index for the FK506 or CsA treatment selection in kidney recipients. Tacrolimus 103-108 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 27754593-2 2017 Here, we assessed age-specific effects of the calcineurin inhibitor tacrolimus (TAC) in a murine transplant model and assessed its clinical relevance on human T cells. Tacrolimus 68-78 calcineurin binding protein 1 Mus musculus 46-67 28339015-8 2017 Through researching the expression of nerve regeneration associated proteins (NF-H and GAP-43) following DAI, the present study provides novel data to suggest that FK506 promotes axon formation and nerve regeneration following experimental DAI. Tacrolimus 164-169 neurofilament heavy chain Homo sapiens 78-82 28339015-8 2017 Through researching the expression of nerve regeneration associated proteins (NF-H and GAP-43) following DAI, the present study provides novel data to suggest that FK506 promotes axon formation and nerve regeneration following experimental DAI. Tacrolimus 164-169 growth associated protein 43 Homo sapiens 87-93 28339051-15 2017 Tacrolimus reduced urinary protein and slowed the progression of DN, partially by recovering the protein expression of nephrin in the renal tissue of diabetic rats, and maintaining the integrity of the structure and function of podocytes. Tacrolimus 0-10 NPHS1 adhesion molecule, nephrin Rattus norvegicus 119-126 28316087-11 2017 Further, CYP2C8*3 and CYP2J2 c.-76G>T SNPs influenced the renal function of these patients and the occurrence of adverse events during treatment with tacrolimus and mycophenolate sodium. Tacrolimus 153-163 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 9-15 28316087-11 2017 Further, CYP2C8*3 and CYP2J2 c.-76G>T SNPs influenced the renal function of these patients and the occurrence of adverse events during treatment with tacrolimus and mycophenolate sodium. Tacrolimus 153-163 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 22-28 28316087-1 2017 STUDY OBJECTIVE: To investigate the influence of single nucleotide polymorphisms (SNPs) in genes encoding metabolizing enzymes (CYP2C8, CYP2J2, and UGT2B7) and transporters (ABCC2 and ABCG2) on dose and dose-adjusted trough blood concentrations (C:D ratio), clinical outcomes, and occurrence of adverse events of tacrolimus and mycophenolate sodium in Brazilian kidney transplant recipients. Tacrolimus 313-323 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 128-134 28316087-6 2017 ABCC2 c.3972T allele carriers showed higher tacrolimus C:D values than did carriers of the c.3972CC genotype. Tacrolimus 44-54 ATP binding cassette subfamily C member 2 Homo sapiens 0-5 28316087-7 2017 The CYP2C8*3 variant was also associated with slightly higher tacrolimus C:D values and higher estimated glomerular filtration rate but only in CYP3A5-nonexpressing patients (CYP3A5*3C/*3C carriers). Tacrolimus 62-72 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 4-10 28316087-10 2017 CONCLUSION: The ABCC2 c.3972C >T polymorphism affected tacrolimus C:D in Brazilian kidney transplant recipients. Tacrolimus 58-68 ATP binding cassette subfamily C member 2 Homo sapiens 16-21 28414795-4 2017 Cadm1 expression was upregulated in BMMs by receptor activator of nuclear factor kappa B ligand (RANKL), and blocked by a calcineurin/NFATc1 inhibitor, FK506. Tacrolimus 152-157 cell adhesion molecule 1 Mus musculus 0-5 28457398-10 2017 On the subgroup with GFR <45 mL/min/1.73 m2, those on tacrolimus had lower MgS than those on cyclosporine, but those same patients presented with significantly different GFR, higher in the tacrolimus subgroup. Tacrolimus 57-67 CD59 molecule (CD59 blood group) Homo sapiens 35-40 28457398-13 2017 A significant difference on MgS levels between patients on tacrolimus and cyclosporine was found only when considering GFR <45 mL/min/1.73 m2, in which patients on tacrolimus had significantly higher GFR than patients on cyclosporine, which may explain these results. Tacrolimus 167-177 CD59 molecule (CD59 blood group) Homo sapiens 133-138 28414795-4 2017 Cadm1 expression was upregulated in BMMs by receptor activator of nuclear factor kappa B ligand (RANKL), and blocked by a calcineurin/NFATc1 inhibitor, FK506. Tacrolimus 152-157 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 134-140 28378777-1 2017 Bruck Syndrome is a connective tissue disease associated with inactivating mutations in lysyl hydroxylase 2 (LH2/PLOD2) or FK506 binding protein 65 (FKBP65/FKBP10). Tacrolimus 123-128 FK506 binding protein 10 Mus musculus 149-155 28400553-5 2017 Relapses in tacrolimus treatment were associated with serum titers of aquaporin 4 antibody (AQP4-IgG) (P = 0.028). Tacrolimus 12-22 aquaporin 4 Homo sapiens 70-81 28400553-5 2017 Relapses in tacrolimus treatment were associated with serum titers of aquaporin 4 antibody (AQP4-IgG) (P = 0.028). Tacrolimus 12-22 aquaporin 4 Homo sapiens 92-96 28378777-1 2017 Bruck Syndrome is a connective tissue disease associated with inactivating mutations in lysyl hydroxylase 2 (LH2/PLOD2) or FK506 binding protein 65 (FKBP65/FKBP10). Tacrolimus 123-128 FK506 binding protein 10 Mus musculus 156-162 27779452-9 2017 A beneficial angiogenic response to potential therapies, FK506 and Elafin, was related to reduced slit guidance ligand 3 (SLIT3), an antimigratory factor. Tacrolimus 57-62 slit guidance ligand 3 Homo sapiens 122-127 28435308-0 2017 Association of the PPP3CA c.249G>A variant with clinical outcomes of tacrolimus-based therapy in kidney transplant recipients. Tacrolimus 72-82 protein phosphatase 3 catalytic subunit alpha Homo sapiens 19-25 28157649-13 2017 CONCLUSION: Tacrolimus of different formulations had different impact on patients with different CYP3A5 genotypes after kidney transplantation. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 97-103 28157649-1 2017 OBJECTIVE: This study is to analyze concentration changes of the prolonged-release and shorter-acting formulation of tacrolimus in patients with different CYP3A5 genotypes after kidney transplantation. Tacrolimus 117-127 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 155-161 27503321-7 2017 CONCLUSIONS: These results strongly suggest that the interaction of FK506 with GDNF and NGF mediates distinct enhancement of neurite growth. Tacrolimus 68-73 glial cell derived neurotrophic factor Gallus gallus 79-83 27503321-7 2017 CONCLUSIONS: These results strongly suggest that the interaction of FK506 with GDNF and NGF mediates distinct enhancement of neurite growth. Tacrolimus 68-73 nerve growth factor Gallus gallus 88-91 28362060-17 2017 In the direct comparison, based on a single trial including 222 participants, tacrolimus plus sirolimus had increased mortality (HR 2.76, 95% CrI 1.30 to 6.69) and graft loss (HR 2.34, 95% CrI 1.28 to 4.61) at maximal follow-up compared with tacrolimus. Tacrolimus 78-88 EP300 interacting inhibitor of differentiation 1 Homo sapiens 142-147 28362060-17 2017 In the direct comparison, based on a single trial including 222 participants, tacrolimus plus sirolimus had increased mortality (HR 2.76, 95% CrI 1.30 to 6.69) and graft loss (HR 2.34, 95% CrI 1.28 to 4.61) at maximal follow-up compared with tacrolimus. Tacrolimus 78-88 EP300 interacting inhibitor of differentiation 1 Homo sapiens 189-194 27598231-8 2017 Moreover, higher frequencies of FoxP3-positive regulatory T cells in renal allografts were observed in ASP2409- and belatacept-based regimens compared with tacrolimus-based regimens. Tacrolimus 156-166 forkhead box protein P3 Macaca fascicularis 32-37 28405602-12 2017 After drug conversion of a twice-daily CNI formulation to a once-daily tacrolimus formulation, CSA-treated patients needed longer to improve their cognitive functioning. Tacrolimus 71-81 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 95-98 28405602-14 2017 CONCLUSIONS: Patients with once-daily tacrolimus formulation had a better psychomotor speed than CSA-treated patients. Tacrolimus 38-48 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 97-100 28112181-0 2017 IL-3 and CTLA4 gene polymorphisms may influence the tacrolimus dose requirement in Chinese kidney transplant recipients. Tacrolimus 52-62 interleukin 3 Homo sapiens 0-4 28112181-0 2017 IL-3 and CTLA4 gene polymorphisms may influence the tacrolimus dose requirement in Chinese kidney transplant recipients. Tacrolimus 52-62 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 9-14 27822915-4 2017 Multiple regression analysis showed that concomitant use of oral itraconazole or voriconazole significantly increased the (C/Dpo)/(C/Div) of tacrolimus (p = 0.002), probably owing to the inhibition of enterohepatic cytochrome P450 3A4. Tacrolimus 141-151 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 215-234 28243539-7 2017 As expected, FK506 could induce a significant elevation of GVBD rate and increase the MPF level of denuded oocytes (DOs). Tacrolimus 13-18 mesothelin Mus musculus 86-89 26882121-3 2017 Aside from CYP3A5*3, the rs3814055 polymorphism in the NR1I2 gene was associated with the tacrolimus pharmacokinetics based on false discovery rate-corrected multiple tests and the least absolute shrinkage and selection operator analysis. Tacrolimus 90-100 nuclear receptor subfamily 1 group I member 2 Homo sapiens 55-60 26882121-6 2017 An in vitro luciferase reporter assay indicated that downregulation of PXR expression is the likely molecular mechanism responsible for the increased exposure to tacrolimus in subjects carrying the rs3814055 C>T variant. Tacrolimus 162-172 nuclear receptor subfamily 1 group I member 2 Homo sapiens 71-74 28219593-0 2017 Betel Nut Chewing Is Associated With Reduced Tacrolimus Concentration in Taiwanese Liver Transplant Recipients. Tacrolimus 45-55 NUT midline carcinoma family member 1 Homo sapiens 6-9 28219593-2 2017 Tacrolimus, an immunosuppressant that protects against organ rejection in transplant recipients, not only is mainly metabolized by CYP3A enzymes but also has a narrow therapeutic range. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 131-136 28219593-6 2017 RESULTS: Dose-adjusted blood trough levels of tacrolimus tended to be significantly (P = .04) lower in betel nut chewers (mean = 0.81, medium = 0.7, 95% confidence interval [CI] = 0.73 to 0.90) than in nonchewers (mean = 1.12, medium = 0.88, 95% CI = 1.03 to 1.22) during the 18-month study period. Tacrolimus 46-56 NUT midline carcinoma family member 1 Homo sapiens 109-112 28280692-1 2017 The calcineurin inhibitor (CNI) tacrolimus (TAC) is an integral part of the immunosuppressive regimen after solid organ transplantation. Tacrolimus 32-42 calcineurin binding protein 1 Homo sapiens 4-25 28280692-1 2017 The calcineurin inhibitor (CNI) tacrolimus (TAC) is an integral part of the immunosuppressive regimen after solid organ transplantation. Tacrolimus 32-42 calcineurin binding protein 1 Homo sapiens 27-30 28193233-8 2017 Placental production of TNFalphaand IL16 in the tacrolimus-treated HFD-dNONcNZO dams were restored to non-diabetic levels and the treatment resulted in the inhibition of aberrant monocyte/macrophage activation during pregnancy in the HFD-dNONcNZO dams. Tacrolimus 48-58 interleukin 16 Mus musculus 36-40 28108248-10 2017 We herein demonstrated that the new FK506-immobilized beads specifically isolated more FKBP12 than the original beads, thereby proving our method to be applicable to target identification experiments. Tacrolimus 36-41 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 87-93 28196514-9 2017 Interestingly, FK506 injections significantly attenuated the MPTP-induced effects in the humanized CD34+ mice compared the C57BL/6 mice. Tacrolimus 15-20 CD34 antigen Mus musculus 99-103 29371918-8 2017 TC improved Bcl2/Bax ratio, decreased apoptosis, CYP2E1 protein expression and liver fibrosis levels, however, EV offered no such protection. Tacrolimus 0-2 B cell leukemia/lymphoma 2 Mus musculus 12-16 29371918-8 2017 TC improved Bcl2/Bax ratio, decreased apoptosis, CYP2E1 protein expression and liver fibrosis levels, however, EV offered no such protection. Tacrolimus 0-2 BCL2-associated X protein Mus musculus 17-20 29371918-8 2017 TC improved Bcl2/Bax ratio, decreased apoptosis, CYP2E1 protein expression and liver fibrosis levels, however, EV offered no such protection. Tacrolimus 0-2 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 49-55 29762990-0 2017 [Tacrolimus down-regulates the mRNA levels of IL-17 and IL-23 in the muscle tissues of mice with experimental autoimmune myositis]. Tacrolimus 1-11 interleukin 17A Mus musculus 46-51 27885697-0 2017 Dynamic effects of CYP3A5 polymorphism on dose requirement and trough concentration of tacrolimus in renal transplant recipients. Tacrolimus 87-97 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 19-25 27885697-1 2017 WHAT IS KNOWN AND OBJECTIVE: Tacrolimus is a widely used immunosuppressive drug with marked pharmacokinetic variability partly due to CYP3A5 polymorphism. Tacrolimus 29-39 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 134-140 27885697-2 2017 Our study aimed to investigate the dynamic effects of CYP3A5 genotypes on dose requirement and trough concentration (C0 ) of tacrolimus in renal transplant recipients. Tacrolimus 125-135 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 54-60 28084894-0 2017 Donors FMO3 polymorphisms affect tacrolimus elimination in Chinese liver transplant patients. Tacrolimus 33-43 flavin containing dimethylaniline monoxygenase 3 Homo sapiens 7-11 28084894-4 2017 RESULTS: Donor FMO3 rs1800822 allele T and rs909530 allele T were associated with fast tacrolimus elimination. Tacrolimus 87-97 flavin containing dimethylaniline monoxygenase 3 Homo sapiens 15-19 28084894-5 2017 Combination of polymorphisms of donor FMO3 rs1800822 and rs909530 genotype impacted on tacrolimus elimination (p = 0.0221). Tacrolimus 87-97 flavin containing dimethylaniline monoxygenase 3 Homo sapiens 38-42 28084894-7 2017 CONCLUSION: Donor"s FMO3 polymorphisms might affect tacrolimus elimination. Tacrolimus 52-62 flavin containing dimethylaniline monoxygenase 3 Homo sapiens 20-24 28118317-0 2017 CD45RA Distinguishes CD4+CD25+CD127-/low TSDR Demethylated Regulatory T Cell Subpopulations With Differential Stability and Susceptibility to Tacrolimus-Mediated Inhibition of Suppression. Tacrolimus 142-152 protein tyrosine phosphatase receptor type C Homo sapiens 0-4 28118317-0 2017 CD45RA Distinguishes CD4+CD25+CD127-/low TSDR Demethylated Regulatory T Cell Subpopulations With Differential Stability and Susceptibility to Tacrolimus-Mediated Inhibition of Suppression. Tacrolimus 142-152 interferon stimulated exonuclease gene 20 Homo sapiens 25-29 28118317-0 2017 CD45RA Distinguishes CD4+CD25+CD127-/low TSDR Demethylated Regulatory T Cell Subpopulations With Differential Stability and Susceptibility to Tacrolimus-Mediated Inhibition of Suppression. Tacrolimus 142-152 interleukin 7 receptor Homo sapiens 30-35 29260042-6 2017 We hypothesize that use of tacrolimus may have contributed to the lack of response to topical interferon-alpha2b. Tacrolimus 27-37 interferon alpha 2 Homo sapiens 94-112 27885697-16 2017 CYP3A5 expressers have decreased dose-adjusted tacrolimus C0 when compared to non-expressers. Tacrolimus 47-57 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 28030534-0 2017 Single-Nucleotide Polymorphism of CYP3A5 Impacts the Exposure to Tacrolimus in Pediatric Renal Transplant Recipients: A Pharmacogenetic Substudy of the TWIST Trial. Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 29762990-0 2017 [Tacrolimus down-regulates the mRNA levels of IL-17 and IL-23 in the muscle tissues of mice with experimental autoimmune myositis]. Tacrolimus 1-11 interleukin 23, alpha subunit p19 Mus musculus 56-61 28122021-0 2017 The Effect of Tacrolimus and Mycophenolic Acid on CD14+ Monocyte Activation and Function. Tacrolimus 14-24 CD14 molecule Homo sapiens 50-54 28122021-7 2017 Tacrolimus (200 ng/ml) inhibited phosphorylation of p38MAPK by 30% (mean) in CD14+ monocytes which was significantly less than in activated CD3+ T cells (max 60%; p < 0.05). Tacrolimus 0-10 CD14 molecule Homo sapiens 77-81 28122021-10 2017 p-ERK was inhibited with a maximum of 15% after spiking with either tacrolimus or MPA. Tacrolimus 68-78 mitogen-activated protein kinase 1 Homo sapiens 2-5 28060893-8 2017 Molecular features involved in inflammation and immune response contributing to DN progression were significantly downregulated by tacrolimus (e.g. the tumor necrosis factor alpha (TNF), interleukin 4, or interleukin 10). Tacrolimus 131-141 tumor necrosis factor Homo sapiens 152-179 27890698-7 2017 Thus, inhibition of CYP3A4 is presumably the underlying reason for the observed increase in the concentrations of the CYP3A4 substrate tacrolimus in the patient. Tacrolimus 135-145 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-26 27890698-7 2017 Thus, inhibition of CYP3A4 is presumably the underlying reason for the observed increase in the concentrations of the CYP3A4 substrate tacrolimus in the patient. Tacrolimus 135-145 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 118-124 28060893-8 2017 Molecular features involved in inflammation and immune response contributing to DN progression were significantly downregulated by tacrolimus (e.g. the tumor necrosis factor alpha (TNF), interleukin 4, or interleukin 10). Tacrolimus 131-141 tumor necrosis factor Homo sapiens 181-184 28056508-4 2017 Using DARTS and LC-MS/MS analyses in combination with reference studies, V-ATPase catalytic subunit A (ATP6V1A) was identified as a new target protein of FK506. Tacrolimus 154-159 ATPase H+ transporting V1 subunit A Homo sapiens 103-110 28056508-5 2017 The biological relevance of ATP6V1A in mediating the neuroprotective effects of FK506 was validated by analyzing FK506 activity with respect to autophagy via acridine orange staining and transcription factor EB (TFEB) translocation assay. Tacrolimus 80-85 ATPase H+ transporting V1 subunit A Homo sapiens 28-35 28056508-5 2017 The biological relevance of ATP6V1A in mediating the neuroprotective effects of FK506 was validated by analyzing FK506 activity with respect to autophagy via acridine orange staining and transcription factor EB (TFEB) translocation assay. Tacrolimus 80-85 transcription factor EB Homo sapiens 187-210 28056508-5 2017 The biological relevance of ATP6V1A in mediating the neuroprotective effects of FK506 was validated by analyzing FK506 activity with respect to autophagy via acridine orange staining and transcription factor EB (TFEB) translocation assay. Tacrolimus 80-85 transcription factor EB Homo sapiens 212-216 28060893-8 2017 Molecular features involved in inflammation and immune response contributing to DN progression were significantly downregulated by tacrolimus (e.g. the tumor necrosis factor alpha (TNF), interleukin 4, or interleukin 10). Tacrolimus 131-141 interleukin 4 Homo sapiens 187-200 28060893-8 2017 Molecular features involved in inflammation and immune response contributing to DN progression were significantly downregulated by tacrolimus (e.g. the tumor necrosis factor alpha (TNF), interleukin 4, or interleukin 10). Tacrolimus 131-141 interleukin 10 Homo sapiens 205-219 28060893-9 2017 On the other hand, pro-fibrotic stimuli being detrimental to renal function were induced by tacrolimus like the transforming growth factor beta 1 (TGFB1), endothelin 1 (EDN1), or type IV collagen alpha 1 (COL4A1). Tacrolimus 92-102 transforming growth factor beta 1 Homo sapiens 112-145 28056508-5 2017 The biological relevance of ATP6V1A in mediating the neuroprotective effects of FK506 was validated by analyzing FK506 activity with respect to autophagy via acridine orange staining and transcription factor EB (TFEB) translocation assay. Tacrolimus 113-118 ATPase H+ transporting V1 subunit A Homo sapiens 28-35 28060893-9 2017 On the other hand, pro-fibrotic stimuli being detrimental to renal function were induced by tacrolimus like the transforming growth factor beta 1 (TGFB1), endothelin 1 (EDN1), or type IV collagen alpha 1 (COL4A1). Tacrolimus 92-102 transforming growth factor beta 1 Homo sapiens 147-152 28056508-6 2017 These analyses demonstrated that the binding of FK506 with ATP6V1A induces autophagy by activating the translocation of TFEB from the cytosol into the nucleus. Tacrolimus 48-53 ATPase H+ transporting V1 subunit A Homo sapiens 59-66 28056508-6 2017 These analyses demonstrated that the binding of FK506 with ATP6V1A induces autophagy by activating the translocation of TFEB from the cytosol into the nucleus. Tacrolimus 48-53 transcription factor EB Homo sapiens 120-124 28060893-9 2017 On the other hand, pro-fibrotic stimuli being detrimental to renal function were induced by tacrolimus like the transforming growth factor beta 1 (TGFB1), endothelin 1 (EDN1), or type IV collagen alpha 1 (COL4A1). Tacrolimus 92-102 endothelin 1 Homo sapiens 155-167 28060893-9 2017 On the other hand, pro-fibrotic stimuli being detrimental to renal function were induced by tacrolimus like the transforming growth factor beta 1 (TGFB1), endothelin 1 (EDN1), or type IV collagen alpha 1 (COL4A1). Tacrolimus 92-102 endothelin 1 Homo sapiens 169-173 28060893-9 2017 On the other hand, pro-fibrotic stimuli being detrimental to renal function were induced by tacrolimus like the transforming growth factor beta 1 (TGFB1), endothelin 1 (EDN1), or type IV collagen alpha 1 (COL4A1). Tacrolimus 92-102 collagen type IV alpha 1 chain Homo sapiens 205-211 28060893-10 2017 CONCLUSION: Patients with DN and elevated TNF levels might benefit from tacrolimus treatment regarding maintaining GFR and reducing inflammation. Tacrolimus 72-82 tumor necrosis factor Homo sapiens 42-45 28056860-8 2017 A decline in eGFR was the most frequent adverse event during tacrolimus treatment. Tacrolimus 61-71 epidermal growth factor receptor Homo sapiens 13-17 28655393-0 2017 The role of single nucleotide polymorphisms of CYP3A and ABCB1 on tacrolimus predose concentration in kidney transplant recipients. Tacrolimus 66-76 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-52 28056860-9 2017 During tacrolimus treatment, a >=40% decrease in eGFR was observed in 43 (10.5%) patients. Tacrolimus 7-17 epidermal growth factor receptor Homo sapiens 52-56 27458792-8 2017 We also observed dysregulation of miRNAs (e.g. let-7, miR-26b, miR-145, and miR-183) in cultured human hepatocytes treated with tacrolimus or high glucose, the two independent risk factors of NODALT identified in this cohort. Tacrolimus 128-138 microRNA 26b Homo sapiens 54-61 27458792-8 2017 We also observed dysregulation of miRNAs (e.g. let-7, miR-26b, miR-145, and miR-183) in cultured human hepatocytes treated with tacrolimus or high glucose, the two independent risk factors of NODALT identified in this cohort. Tacrolimus 128-138 microRNA 145 Homo sapiens 63-70 27458792-8 2017 We also observed dysregulation of miRNAs (e.g. let-7, miR-26b, miR-145, and miR-183) in cultured human hepatocytes treated with tacrolimus or high glucose, the two independent risk factors of NODALT identified in this cohort. Tacrolimus 128-138 microRNA 183 Homo sapiens 76-83 27458792-9 2017 The hepatic miRNA profiles altered by tacrolimus or hyperglycemia were associated with insulin resistance and glucose homeostatic imbalance as revealed by enrichment analysis. Tacrolimus 38-48 insulin Homo sapiens 87-94 28655393-0 2017 The role of single nucleotide polymorphisms of CYP3A and ABCB1 on tacrolimus predose concentration in kidney transplant recipients. Tacrolimus 66-76 ATP binding cassette subfamily B member 1 Homo sapiens 57-62 28655393-8 2017 On the other hand, less than 1% of our transplant recipients possess the <italic>CYP3A</italic> genotype, which requires high daily tacrolimus dose. Tacrolimus 144-154 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 87-92 28246425-2 2017 The influence of interleukin-10 (IL-10) and interleukin-18 (IL-18) polymorphisms on tacrolimus pharmacokinetics had been described in liver and kidney transplantation. Tacrolimus 84-94 interleukin 18 Homo sapiens 44-58 27717793-0 2017 The effect of CYP3A5 genetic polymorphisms on adverse events in patients with ulcerative colitis treated with tacrolimus. Tacrolimus 110-120 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 28246425-2 2017 The influence of interleukin-10 (IL-10) and interleukin-18 (IL-18) polymorphisms on tacrolimus pharmacokinetics had been described in liver and kidney transplantation. Tacrolimus 84-94 interleukin 18 Homo sapiens 60-65 28246425-0 2017 Influence of IL-18 and IL-10 Polymorphisms on Tacrolimus Elimination in Chinese Lung Transplant Patients. Tacrolimus 46-56 interleukin 10 Homo sapiens 23-28 28246425-10 2017 IL-18 rs5744247 allele C and rs1946518 allele A were associated with fast tacrolimus metabolism. Tacrolimus 74-84 interleukin 18 Homo sapiens 0-5 28246425-11 2017 Combined analysis showed that the numbers of low IL-18 mRNA expression alleles had positive correlation with tacrolimus C/D ratios in lung transplant recipients. Tacrolimus 109-119 interleukin 18 Homo sapiens 49-54 28246425-12 2017 The influence of IL-18 polymorphisms on tacrolimus C/D ratios was observed in CYP3A5 expresser recipients, but not in CYP3A5 nonexpresser recipients. Tacrolimus 40-50 interleukin 18 Homo sapiens 17-22 28246425-12 2017 The influence of IL-18 polymorphisms on tacrolimus C/D ratios was observed in CYP3A5 expresser recipients, but not in CYP3A5 nonexpresser recipients. Tacrolimus 40-50 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 78-84 28246425-15 2017 IL-18 polymorphisms may influence tacrolimus elimination in lung transplantation patients. Tacrolimus 34-44 interleukin 18 Homo sapiens 0-5 29179182-11 2017 FK506 could significantly decrease TRPCs, CaN, phosphorylation of ERK1/2 and alpha-SMA expression. Tacrolimus 0-5 mitogen activated protein kinase 3 Rattus norvegicus 66-72 29279557-11 2017 It is notable that genotypes in patients where tacrolimus was not detected in the blood were wild types: 2677G/G and 3435C/C in MDR1. Tacrolimus 47-57 ATP binding cassette subfamily B member 1 Homo sapiens 128-132 27747372-0 2017 FOXP3 rs3761548 polymorphism is associated with tacrolimus-induced acute nephrotoxicity in renal transplant patients. Tacrolimus 48-58 forkhead box P3 Homo sapiens 0-5 27747372-1 2017 PURPOSE: The purpose of this study was to investigate the potential impact of FOXP3 and CCDC22 gene polymorphisms on efficacy and safety of tacrolimus (TAC) in renal transplant patients. Tacrolimus 140-150 forkhead box P3 Homo sapiens 78-83 27747372-1 2017 PURPOSE: The purpose of this study was to investigate the potential impact of FOXP3 and CCDC22 gene polymorphisms on efficacy and safety of tacrolimus (TAC) in renal transplant patients. Tacrolimus 140-150 coiled-coil domain containing 22 Homo sapiens 88-94 28805561-4 2017 The aim of this study was to examine the association between KCNJ11 and KCNQ1 gene polymorphisms and posttransplant diabetes mellitus in kidney allograft recipients treated with tacrolimus. Tacrolimus 178-188 potassium inwardly rectifying channel subfamily J member 11 Homo sapiens 61-67 28805561-4 2017 The aim of this study was to examine the association between KCNJ11 and KCNQ1 gene polymorphisms and posttransplant diabetes mellitus in kidney allograft recipients treated with tacrolimus. Tacrolimus 178-188 potassium voltage-gated channel subfamily Q member 1 Homo sapiens 72-77 29179182-12 2017 CONCLUSION: Taken together, these results suggest that the therapeutic effect of FK506 on IgAN might be partially associated with the down-regulated expression of TRPC channels, CaN and phosphorylation of ERK1/2. Tacrolimus 81-86 mitogen activated protein kinase 3 Rattus norvegicus 205-211 26667830-1 2017 Tacrolimus is dependent on CYP3A5 enzyme for metabolism. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 27-33 28092916-9 2017 However, the levels of CD4+Foxp3+ and CD3-CD56+ T cells were higher in the brand-name tacrolimus group than in the generic tacrolimus group 8 weeks after transplantation (p < 0.05). Tacrolimus 86-96 CD4 molecule Homo sapiens 23-26 28092916-10 2017 CONCLUSIONS: The level of CD4+Foxp3+ T cells was higher in the brand-name tacrolimus group than in the generic tacrolimus group after transplantation. Tacrolimus 74-84 CD4 molecule Homo sapiens 26-29 28092916-10 2017 CONCLUSIONS: The level of CD4+Foxp3+ T cells was higher in the brand-name tacrolimus group than in the generic tacrolimus group after transplantation. Tacrolimus 111-121 CD4 molecule Homo sapiens 26-29 28092916-11 2017 This finding showed that brand-name tacrolimus could have more potential immunosuppressive activity than generic tacrolimus regarding the contribution of CD4+Foxp3+ T cells to graft tolerance in liver transplant recipients. Tacrolimus 36-46 CD4 molecule Homo sapiens 154-157 27181115-2 2017 Tacrolimus (TAC) has proved to show efficacy against inadequate response to tumor necrosis factor alpha inhibitors, yet its add-on effects on TCZ remain unknown. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 76-103 28253495-0 2017 Tacrolimus Triggers Transient Receptor Potential Vanilloid-1-Dependent Relapse of Pancreatitis-Related Pain in Mice. Tacrolimus 0-10 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 20-60 27977332-1 2017 AIM: Meta-analyses and large cohort studies provide confusing results on the association of the CYP3A5 6986A>G allelic variant and adverse outcomes in kidney transplant recipients under tacrolimus-based immunosuppressive regimen. Tacrolimus 189-199 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 96-102 27977332-2 2017 A residual effect of CYP3A5 recipient genotype is unexpected if kidney transplant recipients have similar exposure of tacrolimus. Tacrolimus 118-128 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 21-27 27977332-5 2017 We found a significant association of CYP3A5 genotypes with tacrolimus daily dose as well as with tacrolimus dose-adjusted concentrations. Tacrolimus 60-70 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 38-44 27977332-5 2017 We found a significant association of CYP3A5 genotypes with tacrolimus daily dose as well as with tacrolimus dose-adjusted concentrations. Tacrolimus 98-108 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 38-44 28253495-3 2017 Calcineurin, a phosphatase, negatively regulates various channel functions including TRPV1, and calcineurin inhibitor-induced pain syndrome by tacrolimus, a calcineurin inhibitor, used as an immunosuppressant, has been a clinical problem. Tacrolimus 143-153 calcineurin binding protein 1 Mus musculus 96-117 28253495-3 2017 Calcineurin, a phosphatase, negatively regulates various channel functions including TRPV1, and calcineurin inhibitor-induced pain syndrome by tacrolimus, a calcineurin inhibitor, used as an immunosuppressant, has been a clinical problem. Tacrolimus 143-153 calcineurin binding protein 1 Mus musculus 157-178 28253495-6 2017 Pancreatitis-related symptoms disappeared in 24 h, whereas the referred hyperalgesia recurred following the administration of tacrolimus, which was abolished by the blockers of TRPV1 but not T-type Ca2+ channels. Tacrolimus 126-136 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 177-182 28253495-7 2017 Thus, tacrolimus appears to cause the TRPV1-dependent relapse of pancreatitis-related pain, suggesting the involvement of calcineurin in the termination of pancreatic pain. Tacrolimus 6-16 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 38-43 28031829-1 2016 A 59-year-old woman receiving methotrexate and tacrolimus for rheumatoid arthritis (RA) was referred to our hospital following bilateral ground-glass opacity observed in her chest X-ray and elevated serum KL-6. Tacrolimus 47-57 mucin 1, cell surface associated Homo sapiens 205-209 27488997-9 2016 Tacrolimus also did not change total AQP2 abundance but significantly increased the abundances of pS261-AQP2 and pS264-AQP2. Tacrolimus 0-10 aquaporin 2 Rattus norvegicus 104-108 27488997-9 2016 Tacrolimus also did not change total AQP2 abundance but significantly increased the abundances of pS261-AQP2 and pS264-AQP2. Tacrolimus 0-10 aquaporin 2 Rattus norvegicus 104-108 27488997-11 2016 Tacrolimus did increase the expression of pS264-AQP2 in the apical plasma membrane (by immunohistochemistry). Tacrolimus 0-10 aquaporin 2 Rattus norvegicus 48-52 28101175-10 2016 FK506 treatment altered the expression levels of miR-146a and miR-155, indicating that they may have an important role in regulating the immune response to the rejection effect. Tacrolimus 0-5 microRNA 146a Rattus norvegicus 49-57 28101175-10 2016 FK506 treatment altered the expression levels of miR-146a and miR-155, indicating that they may have an important role in regulating the immune response to the rejection effect. Tacrolimus 0-5 microRNA 155 Rattus norvegicus 62-69 27658575-4 2016 Yak FKBP6 encodes a polypeptide of 295 amino acid residues with an FK506-binding domain (FKBP_C) and three tetratricopeptide repeat domains. Tacrolimus 67-72 FKBP prolyl isomerase family member 6 (inactive) Bos taurus 4-9 27152719-12 2016 In OLP, tacrolimus treatment reduced the expression of Cat K in the epithelium but increased it in the stroma. Tacrolimus 8-18 cathepsin K Homo sapiens 55-60 27633115-0 2016 Mechanism of tacrolimus-induced chronic renal fibrosis following transplantation is regulated by ox-LDL and its receptor, LOX-1. Tacrolimus 13-23 oxidized low density lipoprotein receptor 1 Rattus norvegicus 122-127 28736028-0 2017 Association Between Tacrolimus Pharmacokinetics and Cytochrome P450 3A5 and Multidrug Resistance Protein 1 Exon 21 Polymorphisms. Tacrolimus 20-30 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 52-71 28736028-0 2017 Association Between Tacrolimus Pharmacokinetics and Cytochrome P450 3A5 and Multidrug Resistance Protein 1 Exon 21 Polymorphisms. Tacrolimus 20-30 ATP binding cassette subfamily B member 1 Homo sapiens 76-106 28736028-1 2017 BACKGROUND: Individual differences in the pharmacokinetics (PK) of tacrolimus (TAC), an immunosuppressive drug, are reportedly associated with single-nucleotide polymorphisms (SNPs) of cytochrome P450 (CYP) 3A5 and multidrug resistance protein 1 (MDR1). Tacrolimus 67-77 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 185-210 28736028-1 2017 BACKGROUND: Individual differences in the pharmacokinetics (PK) of tacrolimus (TAC), an immunosuppressive drug, are reportedly associated with single-nucleotide polymorphisms (SNPs) of cytochrome P450 (CYP) 3A5 and multidrug resistance protein 1 (MDR1). Tacrolimus 67-77 ATP binding cassette subfamily B member 1 Homo sapiens 215-245 28736028-1 2017 BACKGROUND: Individual differences in the pharmacokinetics (PK) of tacrolimus (TAC), an immunosuppressive drug, are reportedly associated with single-nucleotide polymorphisms (SNPs) of cytochrome P450 (CYP) 3A5 and multidrug resistance protein 1 (MDR1). Tacrolimus 67-77 ATP binding cassette subfamily B member 1 Homo sapiens 247-251 27930655-8 2016 According to median regression analysis, AUC of prednisone t0-6 was significantly associated with the decrease of Gd-IgA1 t0-6 (P = 0.01) and IgA1 t0-6 (p = 0.002), whereas AUC of tacrolimus t0-6 was associated with the decrease of IgA1 t0-6 (p = 0.02). Tacrolimus 180-190 immunoglobulin heavy constant alpha 1 Homo sapiens 117-121 27930655-8 2016 According to median regression analysis, AUC of prednisone t0-6 was significantly associated with the decrease of Gd-IgA1 t0-6 (P = 0.01) and IgA1 t0-6 (p = 0.002), whereas AUC of tacrolimus t0-6 was associated with the decrease of IgA1 t0-6 (p = 0.02). Tacrolimus 180-190 immunoglobulin heavy constant alpha 1 Homo sapiens 142-146 27930655-8 2016 According to median regression analysis, AUC of prednisone t0-6 was significantly associated with the decrease of Gd-IgA1 t0-6 (P = 0.01) and IgA1 t0-6 (p = 0.002), whereas AUC of tacrolimus t0-6 was associated with the decrease of IgA1 t0-6 (p = 0.02). Tacrolimus 180-190 immunoglobulin heavy constant alpha 1 Homo sapiens 142-146 27030163-2 2016 PURPOSE: Herein, we prepared ovalbumin (OVA)-modified liposomes encapsulating the immunosuppressive drug FK506 (OVA-LipFK) and aimed to demonstrate the delivery selectivity of the liposomes to splenic B cells, and its antiallergic effect in an OVA-sensitized allergic model mouse. Tacrolimus 105-110 serine (or cysteine) peptidase inhibitor, clade B, member 1, pseudogene Mus musculus 29-38 28164520-0 2016 The Effect of ABCB1 Polymorphisms on Serial Tacrolimus Concentrations in Stable Austrian Long-Term Kidney Transplant Recipients. Tacrolimus 44-54 ATP binding cassette subfamily B member 1 Homo sapiens 14-19 27473149-7 2016 In cultured human cells, treatment with FK506 (tacrolimus), a calcineurin inhibitor, results in accumulation of pTDP species. Tacrolimus 40-45 calcineurin binding protein 1 Homo sapiens 62-83 27473149-7 2016 In cultured human cells, treatment with FK506 (tacrolimus), a calcineurin inhibitor, results in accumulation of pTDP species. Tacrolimus 47-57 calcineurin binding protein 1 Homo sapiens 62-83 28164520-1 2016 BACKGROUND: The multidrug resistance 1 gene (ABCB1) encodes P-glycoprotein (PGP), mainly expressed in the liver and engaged in metabolism of drugs including the immunosuppressant tacrolimus (TAC). Tacrolimus 179-189 ATP binding cassette subfamily B member 1 Homo sapiens 16-38 28164520-1 2016 BACKGROUND: The multidrug resistance 1 gene (ABCB1) encodes P-glycoprotein (PGP), mainly expressed in the liver and engaged in metabolism of drugs including the immunosuppressant tacrolimus (TAC). Tacrolimus 179-189 ATP binding cassette subfamily B member 1 Homo sapiens 45-50 28164520-1 2016 BACKGROUND: The multidrug resistance 1 gene (ABCB1) encodes P-glycoprotein (PGP), mainly expressed in the liver and engaged in metabolism of drugs including the immunosuppressant tacrolimus (TAC). Tacrolimus 179-189 ATP binding cassette subfamily B member 1 Homo sapiens 60-74 28164520-1 2016 BACKGROUND: The multidrug resistance 1 gene (ABCB1) encodes P-glycoprotein (PGP), mainly expressed in the liver and engaged in metabolism of drugs including the immunosuppressant tacrolimus (TAC). Tacrolimus 179-189 ATP binding cassette subfamily B member 1 Homo sapiens 76-79 27399255-10 2016 Everolimus and tacrolimus are two immunosuppressive drugs metabolized by CYP3A4. Tacrolimus 15-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 73-79 27338269-0 2016 Pure red cell aplasia induced by anti-erythropoietin antibodies, well-controlled with tacrolimus. Tacrolimus 86-96 erythropoietin Homo sapiens 38-52 27338269-3 2016 The efficacy of tacrolimus, another calcineurin inhibitor, in patients with anti-EPO PRCA has not been determined. Tacrolimus 16-26 erythropoietin Homo sapiens 81-84 27338269-4 2016 The present report is the first our knowledge to describe the successful treatment of a patient with anti-EPO PRCA using tacrolimus. Tacrolimus 121-131 erythropoietin Homo sapiens 106-109 27372923-3 2016 Therefore, the aim of this study was to investigate the expression profile of TGFbeta isoforms, their receptors, and TGFbeta-related genes in human retinal pigment epithelial cells (RPE) after tacrolimus (FK-506) treatment in the presence or absence of lipopolysaccharide (LPS)-induced inflammation. Tacrolimus 193-203 transforming growth factor beta 1 Homo sapiens 78-85 27386875-0 2016 Repair of Neurological Function in Response to FK506 Through CaN/NFATc1 Pathway Following Traumatic Brain Injury in Rats. Tacrolimus 47-52 nuclear factor of activated T-cells 1 Rattus norvegicus 65-71 27386875-2 2016 The nuclear factor of activated T cells (NFATc1) pathway plays an important role in regenerating neurological function following traumatic brain injury (TBI), but the precise mechanism underlying FK506-induced repair of neurological functions remains unclear. Tacrolimus 196-201 nuclear factor of activated T-cells 1 Rattus norvegicus 41-47 27386875-8 2016 Moreover, the cortical mRNA and serum protein expression levels of interleukin-2 (IL-2) and interferon-gamma (IFN-gamma) were decreased in FK506 group, especially at 6 h and at 1 day after TBI. Tacrolimus 139-144 interleukin 2 Rattus norvegicus 67-80 27386875-8 2016 Moreover, the cortical mRNA and serum protein expression levels of interleukin-2 (IL-2) and interferon-gamma (IFN-gamma) were decreased in FK506 group, especially at 6 h and at 1 day after TBI. Tacrolimus 139-144 interleukin 2 Rattus norvegicus 82-86 27386875-8 2016 Moreover, the cortical mRNA and serum protein expression levels of interleukin-2 (IL-2) and interferon-gamma (IFN-gamma) were decreased in FK506 group, especially at 6 h and at 1 day after TBI. Tacrolimus 139-144 interferon gamma Rattus norvegicus 92-108 27386875-8 2016 Moreover, the cortical mRNA and serum protein expression levels of interleukin-2 (IL-2) and interferon-gamma (IFN-gamma) were decreased in FK506 group, especially at 6 h and at 1 day after TBI. Tacrolimus 139-144 interferon gamma Rattus norvegicus 110-119 27386875-10 2016 Mechanistically, FK506 significantly down-regulated the mRNA and protein levels of calcium-regulated phosphatase (calcineurin, CaN) and inhibited the activation of NFATc1. Tacrolimus 17-22 nuclear factor of activated T-cells 1 Rattus norvegicus 164-170 27386875-11 2016 These results demonstrate that FK506 relieved inflammatory responses by regulating the NFATc1 signaling pathway and promoting the synaptic reconstruction of neurons and glial cells by regulating cell apoptosis, thereby facilitated improvements in neurological function. Tacrolimus 31-36 nuclear factor of activated T-cells 1 Rattus norvegicus 87-93 29441922-0 2016 The impact of CYP3A5 on the metabolism of cyclosporine A and tacrolimus in the evaluation of efficiency and safety of immunosuppressive treatment in patients after kidney transplantation. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 29441922-1 2016 The aim of the study was to determine the impact of CYP3A5 mutation on the serum levels of immunosuppressive drugs (tacrolimus and cyclosporine A), and on the occurrence of acute rejection episodes among patients after kidney transplantation. Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 52-58 27372923-0 2016 Transforming growth factor beta-related genes in human retinal pigment epithelial cells after tacrolimus treatment. Tacrolimus 94-104 transforming growth factor beta 1 Homo sapiens 0-31 27372923-3 2016 Therefore, the aim of this study was to investigate the expression profile of TGFbeta isoforms, their receptors, and TGFbeta-related genes in human retinal pigment epithelial cells (RPE) after tacrolimus (FK-506) treatment in the presence or absence of lipopolysaccharide (LPS)-induced inflammation. Tacrolimus 205-211 transforming growth factor beta 1 Homo sapiens 117-124 27372923-5 2016 RESULTS: Analysis using oligonucleotide microarrays revealed 20 statistically significant differentially expressed TGFbeta-related genes after LPS treatment in relation to control cells, and after tacrolimus and LPS treatment in relation to LPS-treated cells. Tacrolimus 197-207 transforming growth factor beta 1 Homo sapiens 115-122 27372923-6 2016 Moreover, our results showed that mRNA levels for TGFbeta2 and TGFbetaR3 after tacrolimus treatment, and for TGFbetaR3 after tacrolimus and LPS treatment in RPE cells were decreased. Tacrolimus 79-89 transforming growth factor beta 2 Homo sapiens 50-58 27310200-8 2016 Cyclosporine predominantly inhibited CYP3A4 (half maximal inhibitory concentration = 0.71 microM) rather than CYP3A5 (>5 microM), whereas tacrolimus showed similar inhibition for CYP3A4 (0.29 microM) and CYP3A5 (0.41 microM). Tacrolimus 141-151 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 182-188 27706725-0 2016 Effect of pregnane X receptor polymorphisms on tacrolimus blood concentrations and the resulting adverse reactions in kidney transplantation recipients. Tacrolimus 47-57 nuclear receptor subfamily 1 group I member 2 Homo sapiens 10-29 27653228-8 2016 CONCLUSIONS: This study confirms the effect of CYP3A5*3 on tacrolimus dose requirement in liver transplantation and shows unexpected associations between the type of, and exposure to, CNI and either chronic rejection or graft loss. Tacrolimus 59-69 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 47-53 27746739-11 2016 ERK1/2 phosphorylation was inhibited by FK506 and C3. Tacrolimus 40-45 mitogen activated protein kinase 3 Rattus norvegicus 0-6 27706725-1 2016 We investigated the effect of pregnane X receptor (PXR) polymorphisms on tacrolimus (FK506) blood trough concentrations and the associated adverse reactions in kidney transplantation recipients (KTRs). Tacrolimus 73-83 nuclear receptor subfamily 1 group I member 2 Homo sapiens 30-49 27706725-1 2016 We investigated the effect of pregnane X receptor (PXR) polymorphisms on tacrolimus (FK506) blood trough concentrations and the associated adverse reactions in kidney transplantation recipients (KTRs). Tacrolimus 73-83 nuclear receptor subfamily 1 group I member 2 Homo sapiens 51-54 27706725-1 2016 We investigated the effect of pregnane X receptor (PXR) polymorphisms on tacrolimus (FK506) blood trough concentrations and the associated adverse reactions in kidney transplantation recipients (KTRs). Tacrolimus 85-90 nuclear receptor subfamily 1 group I member 2 Homo sapiens 30-49 27498776-0 2016 The POR rs1057868-rs2868177 GC-GT diplotype is associated with high tacrolimus concentrations in early post-renal transplant recipients. Tacrolimus 68-78 cytochrome p450 oxidoreductase Homo sapiens 4-7 27641617-7 2016 Calcineurin antagonist FK506 could weaken the neuroprotection and the dephosphorylation of NMDAR induced by TRPC1/4 overexpression. Tacrolimus 23-28 transient receptor potential cation channel, subfamily C, member 1 Rattus norvegicus 108-113 27498776-2 2016 In this study we examined the association between 6 POR SNPs and tacrolimus concentrations in Chinese renal transplant recipients. Tacrolimus 65-75 cytochrome p450 oxidoreductase Homo sapiens 52-55 27498776-9 2016 The CYP3A5*3 and POR rs1057868-rs2868177 GC-GT diplotype explained 31.7% and 5.7%, respectively, of the inter-individual variability of tacrolimus C0D7/D, whereas the POR rs1057868-rs2868177 GC-GT diplotype could explain 10.9% of the inter-individual variability of tacrolimus C0D7/D in CYP3A5 non-expressers. Tacrolimus 136-146 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 27498776-9 2016 The CYP3A5*3 and POR rs1057868-rs2868177 GC-GT diplotype explained 31.7% and 5.7%, respectively, of the inter-individual variability of tacrolimus C0D7/D, whereas the POR rs1057868-rs2868177 GC-GT diplotype could explain 10.9% of the inter-individual variability of tacrolimus C0D7/D in CYP3A5 non-expressers. Tacrolimus 136-146 cytochrome p450 oxidoreductase Homo sapiens 17-20 27498776-9 2016 The CYP3A5*3 and POR rs1057868-rs2868177 GC-GT diplotype explained 31.7% and 5.7%, respectively, of the inter-individual variability of tacrolimus C0D7/D, whereas the POR rs1057868-rs2868177 GC-GT diplotype could explain 10.9% of the inter-individual variability of tacrolimus C0D7/D in CYP3A5 non-expressers. Tacrolimus 136-146 cytochrome p450 oxidoreductase Homo sapiens 167-170 27498776-9 2016 The CYP3A5*3 and POR rs1057868-rs2868177 GC-GT diplotype explained 31.7% and 5.7%, respectively, of the inter-individual variability of tacrolimus C0D7/D, whereas the POR rs1057868-rs2868177 GC-GT diplotype could explain 10.9% of the inter-individual variability of tacrolimus C0D7/D in CYP3A5 non-expressers. Tacrolimus 136-146 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 287-293 27498776-9 2016 The CYP3A5*3 and POR rs1057868-rs2868177 GC-GT diplotype explained 31.7% and 5.7%, respectively, of the inter-individual variability of tacrolimus C0D7/D, whereas the POR rs1057868-rs2868177 GC-GT diplotype could explain 10.9% of the inter-individual variability of tacrolimus C0D7/D in CYP3A5 non-expressers. Tacrolimus 266-276 cytochrome p450 oxidoreductase Homo sapiens 17-20 27498776-10 2016 CONCLUSION: The CYP3A5*3 and POR rs1057868-rs2868177 GC-GT diplotype accounted for the inter-individual variation of tacrolimus C0D7/D. Tacrolimus 117-127 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 16-22 27498776-10 2016 CONCLUSION: The CYP3A5*3 and POR rs1057868-rs2868177 GC-GT diplotype accounted for the inter-individual variation of tacrolimus C0D7/D. Tacrolimus 117-127 cytochrome p450 oxidoreductase Homo sapiens 29-32 27498776-11 2016 Genotyping of POR rs1057868-rs2868177 diplotypes would help to differentiate initial tacrolimus dose requirements and to achieve early target C0 ranges in Chinese renal transplant recipients. Tacrolimus 85-95 cytochrome p450 oxidoreductase Homo sapiens 14-17 27318696-9 2016 Moreover, calcineurin inhibitors, cyclosporine A and FK506, partially reversed TRPV2 activation-induced inhibition of brown adipocyte differentiation. Tacrolimus 53-58 transient receptor potential cation channel, subfamily V, member 2 Mus musculus 79-84 26990694-0 2016 Long-Term Clinical Impact of Adaptation of Initial Tacrolimus Dosing to CYP3A5 Genotype. Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 72-78 26990694-1 2016 Pretransplantation adaptation of the daily dose of tacrolimus to CYP3A5 genotype is associated with improved achievement of target trough concentration (C0 ), but whether this improvement affects clinical outcomes is unknown. Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 65-71 27138785-0 2016 The Effect of Weight and CYP3A5 Genotype on the Population Pharmacokinetics of Tacrolimus in Stable Paediatric Renal Transplant Recipients. Tacrolimus 79-89 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 25-31 27138785-7 2016 Children with lower weight and CYP3A5 expressers required higher weight-normalised tacrolimus doses. Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 31-37 27138785-10 2016 Dose simulations show that a daily tacrolimus dose of 0.2 mg/kg generates a pre-dose concentration (C 0) ranging from 5 to 10 microg/L depending on the weight and CYP3A5 polymorphism. Tacrolimus 35-45 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 163-169 27138785-12 2016 CONCLUSIONS: In patients beyond the first year after transplantation, there is a cumulative effect of CYP3A5*1 polymorphism and weight on the tacrolimus C 0. Tacrolimus 142-152 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 102-108 27138785-13 2016 Children with lower weight and carriers of the CYP3A5*1 allele have higher weight-normalised tacrolimus dose requirements. Tacrolimus 93-103 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 47-53 27602099-1 2016 Tacrolimus (TAC) has been shown to improve remission from proteinuria in patients with refractory IgA nephropathy (IgAN); however, the efficacy and safety of TAC in such patients have not been fully explored. Tacrolimus 0-10 IGAN1 Homo sapiens 115-119 27314545-2 2016 We sought to determine the singular and combined impact of CYP3A4*22 and CYP3A5*3 variants on tacrolimus drug disposition in adult heart transplant recipients. Tacrolimus 94-104 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 59-65 27314545-2 2016 We sought to determine the singular and combined impact of CYP3A4*22 and CYP3A5*3 variants on tacrolimus drug disposition in adult heart transplant recipients. Tacrolimus 94-104 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 73-79 27314545-7 2016 However, tacrolimus C0 /D was 1.8-fold lower (P<.001) in CYP3A5 expressers vs non-expressers. Tacrolimus 9-19 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 60-66 27314545-8 2016 When combined CYP3A genotypes were evaluated, tacrolimus C0 /D was 1.8-fold lower in EMs vs IMs (P<.001) and EMs vs PMs (P=.001). Tacrolimus 46-56 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-19 27314545-10 2016 CONCLUSION: Combined CYP3A genotype was associated with tacrolimus drug disposition in adult heart transplant recipients, but the effect was largely driven by CYP3A5*3. Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-26 27314545-10 2016 CONCLUSION: Combined CYP3A genotype was associated with tacrolimus drug disposition in adult heart transplant recipients, but the effect was largely driven by CYP3A5*3. Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 159-165 27580845-4 2016 Our results showed that CsA and FK506 treatment decreased proteinuria via a mechanism associated to a reduction in the foot-process fusion and desmin, and a recovery of synaptopodin and podocin. Tacrolimus 32-37 nephrosis 2, podocin Mus musculus 186-193 27604685-0 2016 Previous exposure to biologics and C-reactive protein are associated with the response to tacrolimus in inflammatory bowel disease. Tacrolimus 90-100 C-reactive protein Homo sapiens 35-53 27580845-5 2016 In PAN-treated mouse podocytes, pre-incubation with CsA and FK506 restored the distribution of the actin cytoskeleton, increased the expression of synaptopodin and podocin, improved podocyte viability, and reduced the migrating activities of podocytes. Tacrolimus 60-65 nephrosis 2, podocin Mus musculus 164-171 27580845-6 2016 Treatment with CsA and FK506 also inhibited PAN-induced podocytes apoptosis, which was associated with the induction of Bcl-xL and inhibition of Bax, cleaved caspase 3, and cleaved PARP expression. Tacrolimus 23-28 BCL2-like 1 Mus musculus 120-126 27580845-6 2016 Treatment with CsA and FK506 also inhibited PAN-induced podocytes apoptosis, which was associated with the induction of Bcl-xL and inhibition of Bax, cleaved caspase 3, and cleaved PARP expression. Tacrolimus 23-28 BCL2-associated X protein Mus musculus 145-148 27580845-6 2016 Treatment with CsA and FK506 also inhibited PAN-induced podocytes apoptosis, which was associated with the induction of Bcl-xL and inhibition of Bax, cleaved caspase 3, and cleaved PARP expression. Tacrolimus 23-28 poly (ADP-ribose) polymerase family, member 1 Mus musculus 181-185 27580845-7 2016 Further studies revealed that CsA and FK506 inhibited PAN-induced p38 and JNK signaling, thereby protecting podocytes from PAN-induced injury. Tacrolimus 38-43 mitogen-activated protein kinase 14 Mus musculus 66-69 27580845-7 2016 Further studies revealed that CsA and FK506 inhibited PAN-induced p38 and JNK signaling, thereby protecting podocytes from PAN-induced injury. Tacrolimus 38-43 mitogen-activated protein kinase 8 Mus musculus 74-77 27501378-9 2016 The administration of liposomal FK506 markedly suppressed the expression of cytokines, such as interferon-gamma and tumor necrosis factor-alpha, and reduced inflammation and fibrosis in the myocardium on day 21 compared to free FK506. Tacrolimus 32-37 interferon gamma Rattus norvegicus 95-143 27648296-11 2016 In all patients treated with tacrolimus, tacrolimus could be discontinued with SASP. Tacrolimus 29-39 aspartic peptidase retroviral like 1 Homo sapiens 79-83 27648296-11 2016 In all patients treated with tacrolimus, tacrolimus could be discontinued with SASP. Tacrolimus 41-51 aspartic peptidase retroviral like 1 Homo sapiens 79-83 27518575-4 2016 Tacrolimus and Everolimus exerted potent antiproliferative properties in Huh 7 and Hep3B in which cells Sirolimus was inactive. Tacrolimus 0-10 MIR7-3 host gene Homo sapiens 73-78 27503662-1 2016 BACKGROUND The aim of the present study was to investigate the pharmacokinetics of the once-daily tacrolimus formulation (QD form) in relation to polymorphisms of the donor cytochrome P450 family 3 sub-family A polypeptide 5 (CYP3A5) gene and recipient adenosine triphosphate-binding cassette sub-family B member 1 (ABCB1) gene. Tacrolimus 98-108 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 173-224 27503662-1 2016 BACKGROUND The aim of the present study was to investigate the pharmacokinetics of the once-daily tacrolimus formulation (QD form) in relation to polymorphisms of the donor cytochrome P450 family 3 sub-family A polypeptide 5 (CYP3A5) gene and recipient adenosine triphosphate-binding cassette sub-family B member 1 (ABCB1) gene. Tacrolimus 98-108 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 226-232 27503662-1 2016 BACKGROUND The aim of the present study was to investigate the pharmacokinetics of the once-daily tacrolimus formulation (QD form) in relation to polymorphisms of the donor cytochrome P450 family 3 sub-family A polypeptide 5 (CYP3A5) gene and recipient adenosine triphosphate-binding cassette sub-family B member 1 (ABCB1) gene. Tacrolimus 98-108 ATP binding cassette subfamily B member 1 Homo sapiens 253-314 27503662-1 2016 BACKGROUND The aim of the present study was to investigate the pharmacokinetics of the once-daily tacrolimus formulation (QD form) in relation to polymorphisms of the donor cytochrome P450 family 3 sub-family A polypeptide 5 (CYP3A5) gene and recipient adenosine triphosphate-binding cassette sub-family B member 1 (ABCB1) gene. Tacrolimus 98-108 ATP binding cassette subfamily B member 1 Homo sapiens 316-321 26964692-8 2016 Furthermore, after topical tacrolimus treatment, tear periostin tended to decrease in most patients with AKC along with their clinical improvement. Tacrolimus 27-37 periostin Homo sapiens 54-63 27191765-1 2016 OBJECTIVE: Genetic polymorphisms of the P450 2C9 enzyme (CYP2C9), CYP2C19 and CYP3A5 gene are known to affect the metabolism of many drugs applied in liver transplant recipients, such as warfarin, voriconazole, and tacrolimus. Tacrolimus 215-225 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 27191765-1 2016 OBJECTIVE: Genetic polymorphisms of the P450 2C9 enzyme (CYP2C9), CYP2C19 and CYP3A5 gene are known to affect the metabolism of many drugs applied in liver transplant recipients, such as warfarin, voriconazole, and tacrolimus. Tacrolimus 215-225 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 66-73 27191765-1 2016 OBJECTIVE: Genetic polymorphisms of the P450 2C9 enzyme (CYP2C9), CYP2C19 and CYP3A5 gene are known to affect the metabolism of many drugs applied in liver transplant recipients, such as warfarin, voriconazole, and tacrolimus. Tacrolimus 215-225 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 78-84 26969240-8 2016 Furthermore, the SRIF effect was eliminated when the activity of calmodulin (CaM), calcineurin and protein phosphatase 1 (PP1) was blocked with W-7, FK-506 and okadaic acid, respectively. Tacrolimus 149-155 calmodulin 1 Rattus norvegicus 65-75 26969240-8 2016 Furthermore, the SRIF effect was eliminated when the activity of calmodulin (CaM), calcineurin and protein phosphatase 1 (PP1) was blocked with W-7, FK-506 and okadaic acid, respectively. Tacrolimus 149-155 calmodulin 1 Rattus norvegicus 77-80 26373896-1 2016 The efficacy and safety of tacrolimus twice-a-day (BID) and once-a-day (QD) formulations are similar. Tacrolimus 27-37 BH3 interacting domain death agonist Homo sapiens 51-54 29897725-0 2016 [Associations of SLCO1B1 polymorphisms with tacrolimus concentrations in Chinese renal transplant recipients]. Tacrolimus 44-54 solute carrier organic anion transporter family member 1B1 Homo sapiens 17-24 29897725-1 2016 The study aims to investigate the associations of SLCO1B1 polymorphisms with tacrolimus concentrations in Chinese renal transplant recipients. Tacrolimus 77-87 solute carrier organic anion transporter family member 1B1 Homo sapiens 50-57 29897725-6 2016 In CYP3A5 nonexpressers, the dose-adjusted concentration of tacrolimus in SLCO1B1 rs2306283 CC carriers was considerably higher than that in CT and TT carriers. Tacrolimus 60-70 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 3-9 29897725-6 2016 In CYP3A5 nonexpressers, the dose-adjusted concentration of tacrolimus in SLCO1B1 rs2306283 CC carriers was considerably higher than that in CT and TT carriers. Tacrolimus 60-70 solute carrier organic anion transporter family member 1B1 Homo sapiens 74-81 29897725-7 2016 The results illustrated that SLCO1B1 rs2306283 polymorphisms were associated with tacrolimus concentrations, and genotyping for this SNP may be useful for individualized medicine of tacrolimus. Tacrolimus 82-92 solute carrier organic anion transporter family member 1B1 Homo sapiens 29-36 29897725-7 2016 The results illustrated that SLCO1B1 rs2306283 polymorphisms were associated with tacrolimus concentrations, and genotyping for this SNP may be useful for individualized medicine of tacrolimus. Tacrolimus 182-192 solute carrier organic anion transporter family member 1B1 Homo sapiens 29-36 26714287-1 2016 Patients expressing the cytochrome P450 (CYP) 3A5 gene require a higher tacrolimus dose to achieve therapeutic exposure compared with nonexpressers. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-49 27462157-0 2016 Decreased calcineurin immunoreactivity in the postmortem brain of a patient with schizophrenia who had been prescribed the calcineurin inhibitor, tacrolimus, for leukemia. Tacrolimus 146-156 calcineurin binding protein 1 Homo sapiens 123-144 27462157-13 2016 Compared with the healthy control group and schizophrenia group, the percentages of CaN-immunoreactive neurons in layers III-VI of the BA46 and the putamen tended to be lower in the tacrolimus case. Tacrolimus 182-192 milk fat globule EGF and factor V/VIII domain containing Homo sapiens 135-139 26714287-2 2016 This randomized-controlled study investigated whether adaptation of the tacrolimus starting dose according to CYP3A5 genotype increases the proportion of kidney transplant recipients being within the target tacrolimus predose concentration range (10-15 ng/mL) at first steady-state. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 110-116 27217047-0 2016 Effects of CYP3A5 polymorphism on the pharmacokinetics of a once-daily modified-release tacrolimus formulation and acute kidney injury in hematopoietic stem cell transplantation. Tacrolimus 88-98 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 11-17 27044907-8 2016 Graft-versus-host disease prophylaxis with tacrolimus/sirolimus was associated with a low NRM of 11.5% in the alloHCT recipients in CR1. Tacrolimus 43-53 complement C3b/C4b receptor 1 (Knops blood group) Homo sapiens 132-135 27217047-9 2016 However, in patients who were co-administered AZ, the C0 values of tacrolimus were higher in patients with the CYP3A5*3/*3 allele than with the CYP3A5*1 allele (P = 0.034), although daily doses of Tac-QD in patients with CYP3A5*3/*3 were significantly lower than those with the CYP3A5*1 allele (P = 0.041). Tacrolimus 67-77 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 111-117 27217047-9 2016 However, in patients who were co-administered AZ, the C0 values of tacrolimus were higher in patients with the CYP3A5*3/*3 allele than with the CYP3A5*1 allele (P = 0.034), although daily doses of Tac-QD in patients with CYP3A5*3/*3 were significantly lower than those with the CYP3A5*1 allele (P = 0.041). Tacrolimus 67-77 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 144-150 27217047-9 2016 However, in patients who were co-administered AZ, the C0 values of tacrolimus were higher in patients with the CYP3A5*3/*3 allele than with the CYP3A5*1 allele (P = 0.034), although daily doses of Tac-QD in patients with CYP3A5*3/*3 were significantly lower than those with the CYP3A5*1 allele (P = 0.041). Tacrolimus 67-77 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 144-150 27217047-9 2016 However, in patients who were co-administered AZ, the C0 values of tacrolimus were higher in patients with the CYP3A5*3/*3 allele than with the CYP3A5*1 allele (P = 0.034), although daily doses of Tac-QD in patients with CYP3A5*3/*3 were significantly lower than those with the CYP3A5*1 allele (P = 0.041). Tacrolimus 67-77 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 144-150 27221654-2 2016 Donor interleukin-6 (IL-6) rs1800796 single nucleotide polymorphisms (SNPs) affect the metabolism of tacrolimus following liver transplantation-related hepatic H/R. Tacrolimus 101-111 interleukin 6 Homo sapiens 6-19 27221654-2 2016 Donor interleukin-6 (IL-6) rs1800796 single nucleotide polymorphisms (SNPs) affect the metabolism of tacrolimus following liver transplantation-related hepatic H/R. Tacrolimus 101-111 interleukin 6 Homo sapiens 21-25 27569929-3 2016 This study aimed to evaluate the correlation between the polymorphism of FOXP3 and the blood level of tacrolimus in renal transplant recipients. Tacrolimus 102-112 forkhead box P3 Homo sapiens 73-78 27086024-14 2016 Expression of ICAM-1 by ocular surface epithelium decreased significantly in both groups (tacrolimus, P = 0.003; methylprednisolone, P = 0.008), whereas HLA-DR expression decreased significantly only in the tacrolimus group (P = 0.03). Tacrolimus 90-100 intercellular adhesion molecule 1 Homo sapiens 14-20 27086024-14 2016 Expression of ICAM-1 by ocular surface epithelium decreased significantly in both groups (tacrolimus, P = 0.003; methylprednisolone, P = 0.008), whereas HLA-DR expression decreased significantly only in the tacrolimus group (P = 0.03). Tacrolimus 207-217 intercellular adhesion molecule 1 Homo sapiens 14-20 27060922-0 2016 FK506 positively regulates the migratory potential of melanocyte-derived cells by enhancing syndecan-2 expression. Tacrolimus 0-5 syndecan 2 Homo sapiens 92-102 27060922-3 2016 Interestingly, FK506 also increased the expression of syndecan-2, a transmembrane heparan sulfate proteoglycan through c-jun terminal kinase activation. Tacrolimus 15-20 syndecan 2 Homo sapiens 54-64 27060922-3 2016 Interestingly, FK506 also increased the expression of syndecan-2, a transmembrane heparan sulfate proteoglycan through c-jun terminal kinase activation. Tacrolimus 15-20 Jun proto-oncogene, AP-1 transcription factor subunit Homo sapiens 119-124 27060922-4 2016 Moreover, siRNA-mediated reduction of syndecan-2 expression decreased FK506-mediated cell spreading and migration in melanoma cells and decreased focal adhesion kinase phosphorylation in both melanocytes and melanoma cells. Tacrolimus 70-75 syndecan 2 Homo sapiens 38-48 27060922-5 2016 Consistent with these effects on syndecan-2 expression, FK506 enhanced the membrane and melanosome localizations of PKCbetaII, a regulator of tyrosinase activity. Tacrolimus 56-61 syndecan 2 Homo sapiens 33-43 27060922-8 2016 Taken together, these data indicate that FK506 regulates cell migration by enhancing syndecan-2 expression, further suggesting that syndecan-2 could be a potential target for the treatment of patients with vitiligo. Tacrolimus 41-46 syndecan 2 Homo sapiens 85-95 27060922-8 2016 Taken together, these data indicate that FK506 regulates cell migration by enhancing syndecan-2 expression, further suggesting that syndecan-2 could be a potential target for the treatment of patients with vitiligo. Tacrolimus 41-46 syndecan 2 Homo sapiens 132-142 27569929-0 2016 Impact of FOXP3 Polymorphisms on the Blood Level of Tacrolimus in Renal Transplant Recipients. Tacrolimus 52-62 forkhead box P3 Homo sapiens 10-15 27569929-12 2016 CONCLUSIONS: Our study identified a new relationship between FOXP3 rs3761548 and the blood level of tacrolimus. Tacrolimus 100-110 forkhead box P3 Homo sapiens 61-66 27569929-13 2016 These results suggest that the polymorphism of FOXP3 may affect tacrolimus pharmacokinetics. Tacrolimus 64-74 forkhead box P3 Homo sapiens 47-52 27569951-5 2016 CD4(+) T-cell iATP level in the FK506 group and FK506 + prednisone (Pred) groups was higher than in the FK506 + mycophenolate mofetil (MMF), FK506 + MMF + Pred, and rapamycin (Rapa) groups. Tacrolimus 32-37 CD4 molecule Homo sapiens 0-3 26953159-6 2016 Induction of beta-cell proliferation by either 5-IT or harmine, another natural product DYRK1A inhibitor, was suppressed by coincubation with the calcineurin inhibitor FK506, suggesting involvement of DYRK1A and nuclear factor of activated T cells signaling. Tacrolimus 168-173 dual specificity tyrosine phosphorylation regulated kinase 1A Homo sapiens 88-94 27295076-0 2016 Calcineurin inhibitors cyclosporine A and tacrolimus induce vascular inflammation and endothelial activation through TLR4 signaling. Tacrolimus 42-52 toll-like receptor 4 Mus musculus 117-121 26953159-6 2016 Induction of beta-cell proliferation by either 5-IT or harmine, another natural product DYRK1A inhibitor, was suppressed by coincubation with the calcineurin inhibitor FK506, suggesting involvement of DYRK1A and nuclear factor of activated T cells signaling. Tacrolimus 168-173 dual specificity tyrosine phosphorylation regulated kinase 1A Homo sapiens 201-207 27102446-7 2016 At day 12, a new subcutaneous injection of tacrolimus-loaded microspheres was performed and two days after injection, tacrolimus plasma levels were again increased and both IL-2 plasma levels and PP2B activity decreased. Tacrolimus 43-53 interleukin 2 Rattus norvegicus 173-177 27193181-8 2016 The effect of such mutations on the binding affinity of FK506-related ligands is relevant for assessing the thermodynamic forces regulating molecular recognition in FKBP12 inhibition. Tacrolimus 56-61 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 165-171 26432904-1 2016 Tacrolimus is a widely used immunosuppressive drug that inhibits the phosphatase calcineurin when bound to the 12 kDa FK506-binding protein (FKBP12). Tacrolimus 0-10 FK506 binding protein 1a Mus musculus 141-147 26432904-8 2016 When treated with tacrolimus, however, BP and the renal abundance of phosphorylated NCC were lower in mice lacking FKBP12 along the nephron than in control mice. Tacrolimus 18-28 FK506 binding protein 1a Mus musculus 115-121 26432904-9 2016 Mice lacking FKBP12 along the nephron also maintained a normal relationship between plasma potassium levels and the abundance of phosphorylated NCC with tacrolimus treatment. Tacrolimus 153-163 FK506 binding protein 1a Mus musculus 13-19 27320564-0 2016 Long-Term Influence of CYP3A5 Gene Polymorphism on Pharmacokinetics of Tacrolimus and Patient Outcome After Living Donor Liver Transplantation. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 23-29 26990259-5 2016 RESULTS: The dose-adjusted blood trough concentration of tacrolimus on day 28 was affected by CYP3A5*3/*3 and hemoglobin level (P < 0.001 and P = 0.007), but not by everolimus (P = 0.171). Tacrolimus 57-67 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 94-100 25315214-10 2016 RESULTS: Tacrolimus suppressed the expression of MDC, IP-10, I-309, GRO-alpha, and TNF-alpha in LPS-stimulated THP-1 cells in a dose- and time-dependent manner. Tacrolimus 9-19 C-C motif chemokine ligand 22 Homo sapiens 49-52 25315214-10 2016 RESULTS: Tacrolimus suppressed the expression of MDC, IP-10, I-309, GRO-alpha, and TNF-alpha in LPS-stimulated THP-1 cells in a dose- and time-dependent manner. Tacrolimus 9-19 C-X-C motif chemokine ligand 10 Homo sapiens 54-59 25315214-10 2016 RESULTS: Tacrolimus suppressed the expression of MDC, IP-10, I-309, GRO-alpha, and TNF-alpha in LPS-stimulated THP-1 cells in a dose- and time-dependent manner. Tacrolimus 9-19 C-C motif chemokine ligand 1 Homo sapiens 61-66 25315214-10 2016 RESULTS: Tacrolimus suppressed the expression of MDC, IP-10, I-309, GRO-alpha, and TNF-alpha in LPS-stimulated THP-1 cells in a dose- and time-dependent manner. Tacrolimus 9-19 C-X-C motif chemokine ligand 1 Homo sapiens 68-77 25315214-10 2016 RESULTS: Tacrolimus suppressed the expression of MDC, IP-10, I-309, GRO-alpha, and TNF-alpha in LPS-stimulated THP-1 cells in a dose- and time-dependent manner. Tacrolimus 9-19 tumor necrosis factor Homo sapiens 83-92 25315214-14 2016 CONCLUSION: Tacrolimus suppressed LPS-induced MDC, I-309, IP-10, GRO-alpha, and TNF-alpha expressions in monocytes through the MAPK-ERK pathway; thus, tacrolimus may yield therapeutic efficacy by modulating AD-associated cytokines and chemokines. Tacrolimus 12-22 C-C motif chemokine ligand 22 Homo sapiens 46-49 25315214-14 2016 CONCLUSION: Tacrolimus suppressed LPS-induced MDC, I-309, IP-10, GRO-alpha, and TNF-alpha expressions in monocytes through the MAPK-ERK pathway; thus, tacrolimus may yield therapeutic efficacy by modulating AD-associated cytokines and chemokines. Tacrolimus 12-22 C-C motif chemokine ligand 1 Homo sapiens 51-56 25315214-14 2016 CONCLUSION: Tacrolimus suppressed LPS-induced MDC, I-309, IP-10, GRO-alpha, and TNF-alpha expressions in monocytes through the MAPK-ERK pathway; thus, tacrolimus may yield therapeutic efficacy by modulating AD-associated cytokines and chemokines. Tacrolimus 12-22 C-X-C motif chemokine ligand 10 Homo sapiens 58-63 25315214-14 2016 CONCLUSION: Tacrolimus suppressed LPS-induced MDC, I-309, IP-10, GRO-alpha, and TNF-alpha expressions in monocytes through the MAPK-ERK pathway; thus, tacrolimus may yield therapeutic efficacy by modulating AD-associated cytokines and chemokines. Tacrolimus 12-22 C-X-C motif chemokine ligand 1 Homo sapiens 65-74 25315214-14 2016 CONCLUSION: Tacrolimus suppressed LPS-induced MDC, I-309, IP-10, GRO-alpha, and TNF-alpha expressions in monocytes through the MAPK-ERK pathway; thus, tacrolimus may yield therapeutic efficacy by modulating AD-associated cytokines and chemokines. Tacrolimus 12-22 tumor necrosis factor Homo sapiens 80-89 25315214-14 2016 CONCLUSION: Tacrolimus suppressed LPS-induced MDC, I-309, IP-10, GRO-alpha, and TNF-alpha expressions in monocytes through the MAPK-ERK pathway; thus, tacrolimus may yield therapeutic efficacy by modulating AD-associated cytokines and chemokines. Tacrolimus 151-161 C-X-C motif chemokine ligand 10 Homo sapiens 58-63 25315214-14 2016 CONCLUSION: Tacrolimus suppressed LPS-induced MDC, I-309, IP-10, GRO-alpha, and TNF-alpha expressions in monocytes through the MAPK-ERK pathway; thus, tacrolimus may yield therapeutic efficacy by modulating AD-associated cytokines and chemokines. Tacrolimus 151-161 C-X-C motif chemokine ligand 1 Homo sapiens 65-74 25315214-14 2016 CONCLUSION: Tacrolimus suppressed LPS-induced MDC, I-309, IP-10, GRO-alpha, and TNF-alpha expressions in monocytes through the MAPK-ERK pathway; thus, tacrolimus may yield therapeutic efficacy by modulating AD-associated cytokines and chemokines. Tacrolimus 151-161 tumor necrosis factor Homo sapiens 80-89 27193181-7 2016 Here we apply the technique to a real drug-receptor system, by satisfactorily reproducing the experimental dissociation free energies of FK506-related bulky ligands towards the native FKBP12 enzyme and by predicting the dissociation constants for the same ligands towards the mutant I56D. Tacrolimus 137-142 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 184-190 26473523-1 2016 BACKGROUND: Tacrolimus (Prograf, Advagraf, and FK-506) is the most commonly prescribed calcineurin inhibitor after kidney and liver transplantation. Tacrolimus 12-22 calcineurin binding protein 1 Homo sapiens 87-108 26473523-1 2016 BACKGROUND: Tacrolimus (Prograf, Advagraf, and FK-506) is the most commonly prescribed calcineurin inhibitor after kidney and liver transplantation. Tacrolimus 24-31 calcineurin binding protein 1 Homo sapiens 87-108 26473523-1 2016 BACKGROUND: Tacrolimus (Prograf, Advagraf, and FK-506) is the most commonly prescribed calcineurin inhibitor after kidney and liver transplantation. Tacrolimus 47-53 calcineurin binding protein 1 Homo sapiens 87-108 27220536-5 2016 METHODS: GFAP-gp120 tg mice were treated with FK506 and analyzed for neuropathology, behavior, mitochondrial markers, and calcium flux by two-photon microscopy. Tacrolimus 46-51 glial fibrillary acidic protein Mus musculus 9-13 27220536-6 2016 RESULTS: We found that FK506 reduced the neuronal cell loss and neuro-inflammation in the gp120 tg mice. Tacrolimus 23-28 Envelope surface glycoprotein gp160, precursor Human immunodeficiency virus 1 90-95 27220536-7 2016 Moreover, while vehicle-treated gp120 tg mice displayed damaged mitochondria and increased neuro-inflammatory markers, FK506 rescued the morphological mitochondrial alterations and neuro-inflammation while increasing levels of optic atrophy 1 and mitofusin 1. Tacrolimus 119-124 OPA1, mitochondrial dynamin like GTPase Mus musculus 227-242 27220536-7 2016 Moreover, while vehicle-treated gp120 tg mice displayed damaged mitochondria and increased neuro-inflammatory markers, FK506 rescued the morphological mitochondrial alterations and neuro-inflammation while increasing levels of optic atrophy 1 and mitofusin 1. Tacrolimus 119-124 mitofusin 1 Mus musculus 247-258 27220536-9 2016 However, at a functional level, FK506 ameliorated the gp120 tg mice hyperactivity in the open field. Tacrolimus 32-37 Envelope surface glycoprotein gp160, precursor Human immunodeficiency virus 1 54-59 27500266-2 2016 Experimental studies indicate that an organ rinse with the calcineurin inhibitor tacrolimus before implantation protects against IRI. Tacrolimus 81-91 calcineurin binding protein 1 Homo sapiens 59-80 27500266-10 2016 AST was higher in patients treated with tacrolimus (P = 0.011). Tacrolimus 40-50 solute carrier family 17 member 5 Homo sapiens 0-3 27225724-0 2016 Effects of Genetic Polymorphism in CYP3A4 and CYP3A5 Genes on Tacrolimus Dose Among Kidney Transplant Recipients. Tacrolimus 62-72 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-41 27225724-0 2016 Effects of Genetic Polymorphism in CYP3A4 and CYP3A5 Genes on Tacrolimus Dose Among Kidney Transplant Recipients. Tacrolimus 62-72 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 27225724-1 2016 INTRODUCTION: This study aimed to evaluate the effects of single nucleotide polymorphisms CYP3A4*1B and CYP3A5*3 on tacrolimus dose requirement among kidney transplant recipients. Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-96 27225724-1 2016 INTRODUCTION: This study aimed to evaluate the effects of single nucleotide polymorphisms CYP3A4*1B and CYP3A5*3 on tacrolimus dose requirement among kidney transplant recipients. Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 104-110 27225724-4 2016 RESULTS: The median tacrolimus dose was significantly lower in the CYP3A4*1/*1 carriers (0.06 mg/kg/d; range, 0.007 mg/kg/d to 0.17 mg/kg/d) as compared to the CYP3A4*1B/*1B carriers (0.1 mg/kg/d; range, 0.03 mg/kg/d to 0.22 mg/kg/d; P = .001). Tacrolimus 20-30 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 67-73 27225724-4 2016 RESULTS: The median tacrolimus dose was significantly lower in the CYP3A4*1/*1 carriers (0.06 mg/kg/d; range, 0.007 mg/kg/d to 0.17 mg/kg/d) as compared to the CYP3A4*1B/*1B carriers (0.1 mg/kg/d; range, 0.03 mg/kg/d to 0.22 mg/kg/d; P = .001). Tacrolimus 20-30 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 160-166 27225724-5 2016 Patients with at least 1 CYP3A5*1 wild-type allele required higher median doses of tacrolimus (median, 0.08 mg/kg/d; range, 0.03 mg/kg/d to 0.22 mg/kg/d) as compared to the CYP3A5*3 carriers (median, 0.05 mg/kg/d; range, 0.007 mg/kg/d to 0.17 mg/kg/d; P = .002). Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 25-31 27225724-5 2016 Patients with at least 1 CYP3A5*1 wild-type allele required higher median doses of tacrolimus (median, 0.08 mg/kg/d; range, 0.03 mg/kg/d to 0.22 mg/kg/d) as compared to the CYP3A5*3 carriers (median, 0.05 mg/kg/d; range, 0.007 mg/kg/d to 0.17 mg/kg/d; P = .002). Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 173-179 27225724-7 2016 Moreover, we found a correlation between genetic variations in CYP3A4 and CYP3A5 enzymes and tacrolimus blood levels among our kidney transplant recipients. Tacrolimus 93-103 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 63-69 27225724-7 2016 Moreover, we found a correlation between genetic variations in CYP3A4 and CYP3A5 enzymes and tacrolimus blood levels among our kidney transplant recipients. Tacrolimus 93-103 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 74-80 26856676-9 2016 In the presence of tacrolimus immunosuppression, loss of HIF-1alpha activity in myeloid cells of the recipient by HIF-1alpha gene deletion accelerated OAD development in mouse tracheal allografts. Tacrolimus 19-29 hypoxia inducible factor 1, alpha subunit Mus musculus 57-67 26953629-3 2016 A novel once-daily formulation of tacrolimus (LCPT) has demonstrated noninferiority, similar safety, improved bioavailability, a consistent concentration time profile, and less peak and peak-trough fluctuations vs. tacrolimus twice-daily (Tac BID). Tacrolimus 34-44 BH3 interacting domain death agonist Homo sapiens 243-246 27320607-7 2016 RESULTS: Grp78/BiP and Grp94 expressions were increased 36 hours after FK506 treatment. Tacrolimus 71-76 heat shock protein family A (Hsp70) member 5 Homo sapiens 9-14 27320607-7 2016 RESULTS: Grp78/BiP and Grp94 expressions were increased 36 hours after FK506 treatment. Tacrolimus 71-76 heat shock protein family A (Hsp70) member 5 Homo sapiens 15-18 27320607-7 2016 RESULTS: Grp78/BiP and Grp94 expressions were increased 36 hours after FK506 treatment. Tacrolimus 71-76 heat shock protein 90 beta family member 1 Homo sapiens 23-28 27320607-8 2016 Increased phospho-PERK expression was observed 6 hours after FK506 treatment and peak activation of phospho-PERK was observed at 36 hours. Tacrolimus 61-66 eukaryotic translation initiation factor 2 alpha kinase 3 Homo sapiens 18-22 27320564-7 2016 CONCLUSIONS: Immune suppressive therapy with the use of tacrolimus should be tailored on the basis of CYP3A5 genotype, which may reduce the adverse effects and improve graft outcome. Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 102-108 27320607-9 2016 CHOP/GADD153 expression was increased 48 hours after FK506 treatment. Tacrolimus 53-58 DNA damage inducible transcript 3 Homo sapiens 0-4 27699258-6 2016 Cotreatment with HDAC inhibitors and FK506 led to an enhanced antimyeloma effect with a greater PPP3CA reduction compared with HDAC inhibitors alone both in vitro and in vivo. Tacrolimus 37-42 protein phosphatase 3 catalytic subunit alpha Homo sapiens 96-102 27320607-9 2016 CHOP/GADD153 expression was increased 48 hours after FK506 treatment. Tacrolimus 53-58 DNA damage inducible transcript 3 Homo sapiens 5-12 27320607-10 2016 Expression of iNOS after FK506 treatment began to increase at 12 hours, peaked at 24 hours, and decreased after 36 hours. Tacrolimus 25-30 nitric oxide synthase 2 Homo sapiens 14-18 27128949-0 2016 Tacrolimus Modulates TGF-beta Signaling to Induce Epithelial-Mesenchymal Transition in Human Renal Proximal Tubule Epithelial Cells. Tacrolimus 0-10 transforming growth factor beta 1 Homo sapiens 21-29 27128949-4 2016 FK506 effects on TGF-beta mRNA were assessed by RT PCR and TGF-beta secretion was measured by ELISA. Tacrolimus 0-5 transforming growth factor beta 1 Homo sapiens 17-25 27128949-8 2016 Additionally, the study demonstrates that FK506 activation of the TGF-beta/ SMAD pathways is an essential step in the EMT process. Tacrolimus 42-47 transforming growth factor beta 1 Homo sapiens 66-74 26844388-10 2016 At 48 h after SE, FK506 treatment blocked CRTC1 nuclear localization and dephosphorylation of Ser151. Tacrolimus 18-23 CREB regulated transcription coactivator 1 Rattus norvegicus 42-47 27699258-6 2016 Cotreatment with HDAC inhibitors and FK506 led to an enhanced antimyeloma effect with a greater PPP3CA reduction compared with HDAC inhibitors alone both in vitro and in vivo. Tacrolimus 37-42 histone deacetylase 9 Homo sapiens 127-131 27045291-7 2016 Similarily, ~4% of B-cells in HS and even fewer in CsA or tacrolimus treated patients produced IL-10 (0.5% and 1.5%, p<0.05) and this was confirmed both in non-transplanted CsA-treated healthy subjects and in in vitro co-culture experiments. Tacrolimus 58-68 interleukin 10 Homo sapiens 95-100 26762975-4 2016 The FKBP25 structure revealed that the N-terminal helix-loop-helix (HLH) domain and C-terminal FK506-binding domain (FKBD) interact with each other and that both of the domains are involved in DNA binding. Tacrolimus 95-100 FKBP prolyl isomerase 3 Homo sapiens 4-10 26764098-6 2016 In mouse cells, depletion of ER luminal FKBP10 was almost as potent as FK506 in attenuating expression of PrP(C). Tacrolimus 71-76 prion protein Mus musculus 106-111 26924289-0 2016 Prediction of tacrolimus metabolism and dosage requirements based on CYP3A4 phenotype and CYP3A5(*)3 genotype in Chinese renal transplant recipients. Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-75 26924289-0 2016 Prediction of tacrolimus metabolism and dosage requirements based on CYP3A4 phenotype and CYP3A5(*)3 genotype in Chinese renal transplant recipients. Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 90-96 26924289-8 2016 CONCLUSION: This study provides the equations to predict tacrolimus metabolism and dosage requirements based on the endogenous CYP3A4 phenotype, CYP3A5(*)3 genotype and other non-genetic variables. Tacrolimus 57-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 127-133 26924289-8 2016 CONCLUSION: This study provides the equations to predict tacrolimus metabolism and dosage requirements based on the endogenous CYP3A4 phenotype, CYP3A5(*)3 genotype and other non-genetic variables. Tacrolimus 57-67 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 145-151 26433738-4 2016 Dose-adjusted tacrolimus concentration was used as a surrogate marker of CYP3A4 activity. Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 73-79 26433738-9 2016 Liver transplant recipients with impaired renal function or a low dose-adjusted tacrolimus concentration suggesting a high CYP3A4 are at risk of low 1,25(OH)2D3 concentrations. Tacrolimus 80-90 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 123-129 26829596-3 2016 The objective of this study was to test whether the allelic variant of POR, which is associated with an increased metabolic activity of CYP3A, impacts tacrolimus (Tac) pharmacokinetics. Tacrolimus 151-161 cytochrome p450 oxidoreductase Homo sapiens 71-74 27234753-9 2016 Two of the 11 (18.1%) SNPs were significantly associated with NODAT development after adjusting for age, sex, and tacrolimus usage: MMP-2 (rs1132896) and MMP-2 (rs243849). Tacrolimus 114-124 matrix metallopeptidase 2 Homo sapiens 132-137 27234753-9 2016 Two of the 11 (18.1%) SNPs were significantly associated with NODAT development after adjusting for age, sex, and tacrolimus usage: MMP-2 (rs1132896) and MMP-2 (rs243849). Tacrolimus 114-124 matrix metallopeptidase 2 Homo sapiens 154-159 27003330-1 2016 BACKGROUND: The calcineurin inhibitor (CNI) tacrolimus (Tac) is an effective immunosuppressant used after liver transplantation (LTx), but is often associated with CNI nephrotoxicity. Tacrolimus 44-54 calcineurin binding protein 1 Homo sapiens 16-37 25784181-8 2016 FK506 could inhibit apoptosis of facial motor neurons after facial nerve transection, possibly via up-regulation of bcl-2 expression and down-regulation of bax expression. Tacrolimus 0-5 BCL2, apoptosis regulator Rattus norvegicus 116-121 25784181-8 2016 FK506 could inhibit apoptosis of facial motor neurons after facial nerve transection, possibly via up-regulation of bcl-2 expression and down-regulation of bax expression. Tacrolimus 0-5 BCL2 associated X, apoptosis regulator Rattus norvegicus 156-159 26802601-9 2016 The results of our study suggest an association between the CCL2 gene rs1024611 G allele and PTDM in patients treated with tacrolimus or cyclosporine. Tacrolimus 123-133 C-C motif chemokine ligand 2 Homo sapiens 60-64 26764098-4 2016 We show that FK506 treatment results in a profound reduction in PrP(C) expression due to a defect in the translocation of PrP(C) into the endoplasmic reticulum with subsequent degradation by the proteasome. Tacrolimus 13-18 prion protein Mus musculus 64-69 26764098-8 2016 Both FK506 treatment and FKBP10 depletion were effective in reducing PrP(Sc) propagation in cell models. Tacrolimus 5-10 prion protein Mus musculus 69-72 26239045-0 2016 Impact of cytochrome P450 2C19 polymorphisms on the pharmacokinetics of tacrolimus when coadministered with voriconazole. Tacrolimus 72-82 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 10-30 26239045-1 2016 This study evaluated the effects of cytochrome P450 (CYP) 2C19 polymorphisms on tacrolimus pharmacokinetics when coadministered with voriconazole. Tacrolimus 80-90 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 36-62 26239045-6 2016 These results demonstrate that CYP2C19 genotypes influenced the exposure of tacrolimus when coadministered with voriconazole, although tacrolimus is mainly metabolized by CYP3A. Tacrolimus 76-86 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 31-38 26239045-6 2016 These results demonstrate that CYP2C19 genotypes influenced the exposure of tacrolimus when coadministered with voriconazole, although tacrolimus is mainly metabolized by CYP3A. Tacrolimus 135-145 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 171-176 26749369-0 2016 Crystal structure of the FK506 binding domain of human FKBP25 in complex with FK506. Tacrolimus 25-30 FKBP prolyl isomerase 3 Homo sapiens 55-61 26749369-0 2016 Crystal structure of the FK506 binding domain of human FKBP25 in complex with FK506. Tacrolimus 78-83 FKBP prolyl isomerase 3 Homo sapiens 55-61 26749369-2 2016 Similar to other FKBPs, FK506 binding domain (FKBD) of hFKBP25 also binds to immunosuppressive drugs such as rapamycin and FK506, albeit with a lower affinity for the latter. Tacrolimus 24-29 FKBP prolyl isomerase 3 Homo sapiens 55-62 26749369-2 2016 Similar to other FKBPs, FK506 binding domain (FKBD) of hFKBP25 also binds to immunosuppressive drugs such as rapamycin and FK506, albeit with a lower affinity for the latter. Tacrolimus 123-128 FKBP prolyl isomerase 3 Homo sapiens 55-62 26903089-2 2016 The FK506 binding protein FKBP52 is able to induce oligomers in the pathogenic Tau P301L mutant and in a truncated form of the wild-type human Tau protein. Tacrolimus 4-9 FKBP prolyl isomerase 4 Homo sapiens 26-32 26903089-4 2016 We find that FKBP52 indeed can isomerize selected prolyl bonds in the different Tau proteins, and that this activity is carried solely by its first FK506 binding domain. Tacrolimus 148-153 FKBP prolyl isomerase 4 Homo sapiens 13-19 26903089-6 2016 In addition, we identified a novel molecular interaction implying the PHF6 peptide of Tau and the FK1/FK2 domains of FKBP52 independent of FK506 binding; these data point toward a non-catalytic molecular interaction that might govern the effect of FKBP52 on Tau. Tacrolimus 139-144 FKBP prolyl isomerase 4 Homo sapiens 117-123 26936590-11 2016 The use of tacrolimus was associated with increased concentrations of MCP-1 in plasma and rapamycin was associated with decreased concentrations of MCP-1 in plasma. Tacrolimus 11-21 mast cell protease 1-like 1 Rattus norvegicus 70-75 27627555-0 2016 The Effect of P-Glycoprotein Inhibition and Activation on the Absorption and Serum Levels of Cyclosporine and Tacrolimus in Rats. Tacrolimus 110-120 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 14-28 27627555-1 2016 BACKGROUND: Permeability glycoprotein (P-glycoprotein or P-gp) plays an important role in the intestinal absorption of the immunosuppressive agents: cyclosporine and tacrolimus. Tacrolimus 166-176 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 39-53 27627555-1 2016 BACKGROUND: Permeability glycoprotein (P-glycoprotein or P-gp) plays an important role in the intestinal absorption of the immunosuppressive agents: cyclosporine and tacrolimus. Tacrolimus 166-176 phosphoglycolate phosphatase Rattus norvegicus 57-61 27627555-2 2016 OBJECTIVES: The aim of this study was to determine how the intestinal absorption of cyclosporine and tacrolimus is affected when they are used with P-gp activating or inhibiting agents. Tacrolimus 101-111 phosphoglycolate phosphatase Rattus norvegicus 148-152 26659252-8 2016 Similarly, treatment with chemical inhibitors of calcineurin, such as FK506 and cyclosporin A, or knockdown of calcineurin expression by RNA interference (RNAi), exacerbated the Abeta42-induced rough-eye phenotype. Tacrolimus 70-75 Calcineurin A1 Drosophila melanogaster 49-60 26635230-0 2016 Impact of CYP3A5 polymorphism on trough concentrations and outcomes of tacrolimus minimization during the early period after kidney transplantation. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 26635230-1 2016 PURPOSE: The purpose of this study is to determine the impacts of CYP3A5 polymorphism on tacrolimus concentration and the proportion of patients within a target therapeutic range during the first week after transplantation together with the 3-month acute rejection rate in kidney transplant patients receiving a minimized tacrolimus regimen. Tacrolimus 89-99 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 66-72 26635230-1 2016 PURPOSE: The purpose of this study is to determine the impacts of CYP3A5 polymorphism on tacrolimus concentration and the proportion of patients within a target therapeutic range during the first week after transplantation together with the 3-month acute rejection rate in kidney transplant patients receiving a minimized tacrolimus regimen. Tacrolimus 322-332 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 66-72 26635230-11 2016 CONCLUSIONS: Although CYP3A5 polymorphism significantly influenced the tacrolimus dose required to achieve the target concentration, the impact of CYP3A5 polymorphism on BPAR was not observed in this study. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 22-28 26581186-3 2016 This treatment did not affect the transcription levels of tyrosinase, suggesting that FK506 increases melanin synthesis by regulating cellular levels of tyrosinase. Tacrolimus 86-91 tyrosinase Homo sapiens 153-163 26626486-4 2016 Following prolonged A1R activation, GluA2 internalization was selectively blocked by PP2A inhibitors (okadaic acid and fostriecin), whereas inhibitors of PP2A, PP1 (tautomycetin), and PP2B (FK506) all prevented GluA1 internalization. Tacrolimus 190-195 glutamate ionotropic receptor AMPA type subunit 2 Rattus norvegicus 36-41 26750745-10 2016 LDL-cholesterol and apolipoprotein B levels were significantly lower in patients treated by tacrolimus in comparison with CsA (p < 0.05). Tacrolimus 92-102 apolipoprotein B Homo sapiens 20-36 26581186-4 2016 Interestingly, FK506 promoted melanosome maturation by increasing melanosomal pH (a marker of melanosome maturation), thereby enhancing the stability of melanosome-localized tyrosinase. Tacrolimus 15-20 tyrosinase Homo sapiens 174-184 27109987-6 2016 Reduced NHE3 and DRA expression was associated with high tacrolimus trough levels. Tacrolimus 57-67 solute carrier family 9 member A3 Homo sapiens 8-12 27109969-4 2016 Tacrolimus (TAC), calcineurin inhibitor, is one of immunosuppression pillar in organ transplantation and its action is strongly correlated with blood concentration and therefore the therapeutic drug monitoring is recommended in the guidelines. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 18-39 27109987-6 2016 Reduced NHE3 and DRA expression was associated with high tacrolimus trough levels. Tacrolimus 57-67 solute carrier family 26 member 3 Homo sapiens 17-20 27109969-4 2016 Tacrolimus (TAC), calcineurin inhibitor, is one of immunosuppression pillar in organ transplantation and its action is strongly correlated with blood concentration and therefore the therapeutic drug monitoring is recommended in the guidelines. Tacrolimus 12-15 calcineurin binding protein 1 Homo sapiens 18-39 27109987-7 2016 Titration of tacrolimus to low levels by year 2 was paralleled by partially restored NHE3 and DRA expression. Tacrolimus 13-23 solute carrier family 9 member A3 Homo sapiens 85-89 27109987-7 2016 Titration of tacrolimus to low levels by year 2 was paralleled by partially restored NHE3 and DRA expression. Tacrolimus 13-23 solute carrier family 26 member 3 Homo sapiens 94-97 27110018-1 2016 BACKGROUND: CYP3A5 gene polymorphism rs776746 has been associated with lower tacrolimus dose requirements and bioavailability in both adults and children. Tacrolimus 77-87 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 27110018-10 2016 CONCLUSIONS: Tacrolimus dose was significant higher in functional CYP3A5 expressers. Tacrolimus 13-23 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 66-72 26663079-0 2016 Npas4 Transcription Factor Expression Is Regulated by Calcium Signaling Pathways and Prevents Tacrolimus-induced Cytotoxicity in Pancreatic Beta Cells. Tacrolimus 94-104 neuronal PAS domain protein 4 Homo sapiens 0-5 26705892-0 2016 The importance of MDR1 gene polymorphisms for tacrolimus dosage. Tacrolimus 46-56 ATP binding cassette subfamily B member 1 Homo sapiens 18-22 26705892-1 2016 Polymorphisms of the multi drug resistance (MDR1) gene cause variability in P-glycoprotein mediated metabolism of tacrolimus. Tacrolimus 114-124 ATP binding cassette subfamily B member 1 Homo sapiens 44-48 26705892-2 2016 The aim of this study was to examine the relationship between MDR1 gene single nucleotide polymorphisms (SNPs) and their haplotypes with dosage of tacrolimus in kidney transplant recipients who were cytochrome (CYP) 3A5*3 homozygotes. Tacrolimus 147-157 ATP binding cassette subfamily B member 1 Homo sapiens 62-66 26792730-4 2016 Utilizing human Jurkat T cells, we demonstrate that CrkII-SH3N binding of C3G is inhibited by cyclosporin A (CsA) plus FK506 that inhibit the cyclophilin A (CypA) and FK506 binding protein (FKBP) peptidyl-prolyl cis-trans isomerases (PPIases; also termed immunophilins), respectively. Tacrolimus 119-124 CRK proto-oncogene, adaptor protein Homo sapiens 52-57 26792730-4 2016 Utilizing human Jurkat T cells, we demonstrate that CrkII-SH3N binding of C3G is inhibited by cyclosporin A (CsA) plus FK506 that inhibit the cyclophilin A (CypA) and FK506 binding protein (FKBP) peptidyl-prolyl cis-trans isomerases (PPIases; also termed immunophilins), respectively. Tacrolimus 119-124 Rap guanine nucleotide exchange factor 1 Homo sapiens 74-77 26792730-4 2016 Utilizing human Jurkat T cells, we demonstrate that CrkII-SH3N binding of C3G is inhibited by cyclosporin A (CsA) plus FK506 that inhibit the cyclophilin A (CypA) and FK506 binding protein (FKBP) peptidyl-prolyl cis-trans isomerases (PPIases; also termed immunophilins), respectively. Tacrolimus 119-124 peptidylprolyl isomerase A Homo sapiens 142-155 26792730-4 2016 Utilizing human Jurkat T cells, we demonstrate that CrkII-SH3N binding of C3G is inhibited by cyclosporin A (CsA) plus FK506 that inhibit the cyclophilin A (CypA) and FK506 binding protein (FKBP) peptidyl-prolyl cis-trans isomerases (PPIases; also termed immunophilins), respectively. Tacrolimus 119-124 peptidylprolyl isomerase A Homo sapiens 157-161 26792730-6 2016 Treatment of Jurkat T cells with CsA plus FK506 led to a time-dependent conformational change in overexpressed human CrkII1-236 protein-containing FRET-based biosensor, supporting the accumulation of cis conformers of human CrkII1-236 in the presence of PPIase inhibitors. Tacrolimus 42-47 FKBP prolyl isomerase 3 Homo sapiens 254-260 27009276-8 2016 We also observed that the treatment with tacrolimus, aCNI, in this nephrotic model suppressed the redistribution ofCN, nephrin, and otherSDcomponents and ameliorated proteinuria. Tacrolimus 41-51 NPHS1 adhesion molecule, nephrin Homo sapiens 119-126 26792730-7 2016 Our data suggest that the Gly(219)-Pro-Tyr motif in the human CrkII linker region serves as the recognition and isomerization site of PPIases, and raise the possibility that CsA and FK506 might interfere with selected effector T cell functions via a CrkII-, but not CrkI-dependent mechanisms. Tacrolimus 182-187 CRK proto-oncogene, adaptor protein Homo sapiens 62-67 26792730-7 2016 Our data suggest that the Gly(219)-Pro-Tyr motif in the human CrkII linker region serves as the recognition and isomerization site of PPIases, and raise the possibility that CsA and FK506 might interfere with selected effector T cell functions via a CrkII-, but not CrkI-dependent mechanisms. Tacrolimus 182-187 CRK proto-oncogene, adaptor protein Homo sapiens 250-255 26663079-7 2016 Finally, beta cell cytotoxicity of the calcineurin inhibitor and immunosuppressant tacrolimus (FK-506) was prevented by Npas4 overexpression. Tacrolimus 83-93 neuronal PAS domain protein 4 Homo sapiens 120-125 26663079-7 2016 Finally, beta cell cytotoxicity of the calcineurin inhibitor and immunosuppressant tacrolimus (FK-506) was prevented by Npas4 overexpression. Tacrolimus 95-101 calcineurin binding protein 1 Homo sapiens 39-60 26663079-7 2016 Finally, beta cell cytotoxicity of the calcineurin inhibitor and immunosuppressant tacrolimus (FK-506) was prevented by Npas4 overexpression. Tacrolimus 95-101 neuronal PAS domain protein 4 Homo sapiens 120-125 26325438-8 2016 For tacrolimus, rs776746 CYP3A5*3/*3 and CYP3A5*3/*1 were associated with higher blood levels than CYP3A5*1/*1 (P = .002). Tacrolimus 4-14 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 25-31 26824445-8 2016 This inverse association between MPA and SCSC persisted among OTRs with no history of azathioprine use, even after adjustment for simultaneous use of the calcineurin inhibitor tacrolimus (OR = 0.52, 95% CI 0.32-0.84). Tacrolimus 176-186 calcineurin binding protein 1 Homo sapiens 154-175 26809267-4 2016 The PTX- and FK506-coloaded MPEG-PCL micelles (P-F/M) were prepared by a one-step solid dispersion method without any surfactants, toxic organic solvent, or severe experimental conditions. Tacrolimus 13-18 msh homeobox 2 Homo sapiens 47-52 26485092-0 2016 Genomewide Association Study of Tacrolimus Concentrations in African American Kidney Transplant Recipients Identifies Multiple CYP3A5 Alleles. Tacrolimus 32-42 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 127-133 26325438-8 2016 For tacrolimus, rs776746 CYP3A5*3/*3 and CYP3A5*3/*1 were associated with higher blood levels than CYP3A5*1/*1 (P = .002). Tacrolimus 4-14 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 41-47 26325438-8 2016 For tacrolimus, rs776746 CYP3A5*3/*3 and CYP3A5*3/*1 were associated with higher blood levels than CYP3A5*1/*1 (P = .002). Tacrolimus 4-14 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 41-47 26892112-7 2016 After reducing tacrolimus levels from 3-4 ng/mL to <1.5-1.8 ng/mL, the fever dropped immediately and C10/C11 disappeared after 2 months. Tacrolimus 15-25 homeobox C10 Homo sapiens 104-107 26892112-7 2016 After reducing tacrolimus levels from 3-4 ng/mL to <1.5-1.8 ng/mL, the fever dropped immediately and C10/C11 disappeared after 2 months. Tacrolimus 15-25 aldo-keto reductase family 1 member C4 Homo sapiens 108-111 26521259-14 2016 CONCLUSIONS: Both donor and recipient CYP3A5 genotype significantly influences tacrolimus once daily pharmacokinetics. Tacrolimus 79-89 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 38-44 26749212-8 2016 The stimulation with PMA and IM greatly augmented Eomes binding to CNS-54, CNS-34, CNS+19 and CNS+30, which was inhibited by FK506. Tacrolimus 125-130 eomesodermin Mus musculus 50-55 26521259-4 2016 The primary objective of this study was to investigate the effect of CYP3A4*22 and CYP3A5*3 of both donor and recipient on once daily tacrolimus pharmacokinetics. Tacrolimus 134-144 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-75 26521259-4 2016 The primary objective of this study was to investigate the effect of CYP3A4*22 and CYP3A5*3 of both donor and recipient on once daily tacrolimus pharmacokinetics. Tacrolimus 134-144 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 83-89 26566632-0 2016 FK506 reduces albuminuria through improving podocyte nephrin and podocin expression in diabetic rats. Tacrolimus 0-5 NPHS1 adhesion molecule, nephrin Rattus norvegicus 53-60 26566632-0 2016 FK506 reduces albuminuria through improving podocyte nephrin and podocin expression in diabetic rats. Tacrolimus 0-5 NPHS2 stomatin family member, podocin Rattus norvegicus 65-72 26566632-7 2016 In diabetic model rats, FK506 treatment at a dose of 0.5 or 1.0 mg/kg significantly increased the expression of nephrin and podocin when compared to control. Tacrolimus 24-29 NPHS1 adhesion molecule, nephrin Rattus norvegicus 112-119 26566632-7 2016 In diabetic model rats, FK506 treatment at a dose of 0.5 or 1.0 mg/kg significantly increased the expression of nephrin and podocin when compared to control. Tacrolimus 24-29 NPHS2 stomatin family member, podocin Rattus norvegicus 124-131 26566632-15 2016 CONCLUSION: Our results show that FK506 not only upregulates expression of nephrin and podocin but also inhibits macrophage activation to protect against podocyte injury. Tacrolimus 34-39 NPHS1 adhesion molecule, nephrin Rattus norvegicus 75-82 26566632-15 2016 CONCLUSION: Our results show that FK506 not only upregulates expression of nephrin and podocin but also inhibits macrophage activation to protect against podocyte injury. Tacrolimus 34-39 NPHS2 stomatin family member, podocin Rattus norvegicus 87-94 25594762-4 2016 In an immortalized normal urothelial cell line SVHUC, CsA and FK506 reduced NFATc1 expression, NFAT transcriptional activity, and the expression of c-myc, a downstream target of NFATc1 signals. Tacrolimus 62-67 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 76-82 26838733-3 2016 Electrophysiological experiments revealed that tacrolimus (10 micromol/L) increased the large-conductance Ca(2+)-activated K(+) currents (BKCa) in rat aortic vascular smooth muscle cells (AVSMCs), which could be blocked by ryanodine (10 micromol/L). Tacrolimus 47-57 potassium calcium-activated channel subfamily M alpha 1 Rattus norvegicus 138-142 26838733-5 2016 In conclusion, tacrolimus could indirectly activate BKCa currents by increasing Ca(2+) sparks released from ryanodine receptors, which inhibited the NE- or KCl-induced contraction in rat aorta. Tacrolimus 15-25 potassium calcium-activated channel subfamily M alpha 1 Rattus norvegicus 52-56 25594762-4 2016 In an immortalized normal urothelial cell line SVHUC, CsA and FK506 reduced NFATc1 expression, NFAT transcriptional activity, and the expression of c-myc, a downstream target of NFATc1 signals. Tacrolimus 62-67 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 178-184 25594762-6 2016 CsA and FK506 were additionally found to up-regulate the expression of several molecules that play a protective role in bladder tumorigenesis, including p53, p21, and p27, and down-regulate that of oncogenic genes, such as cyclin D1, cyclin D3, and cyclin E, in SVHUC cells with the carcinogen challenge. Tacrolimus 8-13 transformation related protein 53, pseudogene Mus musculus 153-156 25594762-6 2016 CsA and FK506 were additionally found to up-regulate the expression of several molecules that play a protective role in bladder tumorigenesis, including p53, p21, and p27, and down-regulate that of oncogenic genes, such as cyclin D1, cyclin D3, and cyclin E, in SVHUC cells with the carcinogen challenge. Tacrolimus 8-13 cyclin-dependent kinase inhibitor 1A (P21) Mus musculus 158-161 25594762-6 2016 CsA and FK506 were additionally found to up-regulate the expression of several molecules that play a protective role in bladder tumorigenesis, including p53, p21, and p27, and down-regulate that of oncogenic genes, such as cyclin D1, cyclin D3, and cyclin E, in SVHUC cells with the carcinogen challenge. Tacrolimus 8-13 cyclin-dependent kinase inhibitor 1B Mus musculus 167-170 25594762-6 2016 CsA and FK506 were additionally found to up-regulate the expression of several molecules that play a protective role in bladder tumorigenesis, including p53, p21, and p27, and down-regulate that of oncogenic genes, such as cyclin D1, cyclin D3, and cyclin E, in SVHUC cells with the carcinogen challenge. Tacrolimus 8-13 cyclin D1 Mus musculus 223-232 25594762-6 2016 CsA and FK506 were additionally found to up-regulate the expression of several molecules that play a protective role in bladder tumorigenesis, including p53, p21, and p27, and down-regulate that of oncogenic genes, such as cyclin D1, cyclin D3, and cyclin E, in SVHUC cells with the carcinogen challenge. Tacrolimus 8-13 cyclin D3 Mus musculus 234-243 26763872-5 2016 In contrast, temporarily immunosuppressed allogeneic mice, following cessation of tacrolimus injection displayed diminished progression of the teratocarcinoma, accompanied by an accumulation of CD4/CD8-positive T cells, and finally achieved complete elimination of the teratocarcinoma. Tacrolimus 82-92 CD4 antigen Mus musculus 194-197 26768967-6 2016 The bound tacrolimus was released gradually from Tac Alb-NPs for ~ 24 h, which was sufficient time for pulmonary delivery. Tacrolimus 10-20 albumin Mus musculus 53-56 26823971-3 2016 Tacrolimus is primarily metabolized by cytochrome P450 3A5 (CYP3A5). Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 39-58 26823971-3 2016 Tacrolimus is primarily metabolized by cytochrome P450 3A5 (CYP3A5). Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 60-66 26796537-2 2016 In the current study, we show that obesity results in perilymphatic accumulation of inflammatory cells and that local inhibition of this response with topical tacrolimus, an inhibitor of T cell differentiation, increases lymphatic vessel density, decreases perilymphatic iNOS expression, increases lymphatic vessel pumping frequency, and restores lymphatic clearance of interstitial fluid to normal levels. Tacrolimus 159-169 nitric oxide synthase 2, inducible Mus musculus 271-275 26784512-0 2016 Donor ABCB1 3435 C>T genetic polymorphisms influence early renal function in kidney transplant recipients treated with tacrolimus. Tacrolimus 122-132 ATP binding cassette subfamily B member 1 Homo sapiens 6-11 26784512-4 2016 RESULTS: The genotype of CYP3A5 (6986A>G) in recipients showed strong influence on tacrolimus pharmacokinetics. Tacrolimus 86-96 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 25-31 26784512-6 2016 CONCLUSIONS: ABCB1 3435 CC genotype in donor influences early renal function and creatinine recovery in tacrolimus-treated kidney transplant recipients. Tacrolimus 104-114 ATP binding cassette subfamily B member 1 Homo sapiens 13-18 26735686-11 2016 CONCLUSIONS: HCV-positive renal transplant recipients show significantly improved glucose-stimulated insulin sensitivity and overall glucose tolerance after conversion from tacrolimus to cyclosporine A. Tacrolimus 173-183 insulin Homo sapiens 101-108 27374298-2 2016 In this study, we investigated the effects of tacrolimus and betamethasone, each used as topical applications in atopic dermatitis (AD), on Th2 cell development mediated by LCs. Tacrolimus 46-56 heart and neural crest derivatives expressed 2 Mus musculus 140-143 26788461-9 2016 Cyclosporine A and tacrolimus suppress immune reactions by inhibiting the phosphatase calcineurin-dependent activation of NFAT1. Tacrolimus 19-29 nuclear factor of activated T cells 2 Homo sapiens 122-127 27374298-4 2016 Mice were primed with ovalbumin (OVA) peptide-pulsed LDCs, which had been treated with tacrolimus or betamethasone, via the hind footpad. Tacrolimus 87-97 serine (or cysteine) peptidase inhibitor, clade B, member 1, pseudogene Mus musculus 22-31 27374298-4 2016 Mice were primed with ovalbumin (OVA) peptide-pulsed LDCs, which had been treated with tacrolimus or betamethasone, via the hind footpad. Tacrolimus 87-97 serine (or cysteine) peptidase inhibitor, clade B, member 1, pseudogene Mus musculus 33-36 27546370-0 2016 Assessment of global ischemic/reperfusion and Tacrolimus administration on CA1 region of hippocampus: gene expression profiles of BAX and BCL2 genes. Tacrolimus 46-56 carbonic anhydrase 1 Rattus norvegicus 75-78 26856709-0 2016 Impact of CYP3A5 and MDR-1 gene polymorphisms on the dose and level of tacrolimus among living-donor liver transplanted patients: single center experience. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 26856709-0 2016 Impact of CYP3A5 and MDR-1 gene polymorphisms on the dose and level of tacrolimus among living-donor liver transplanted patients: single center experience. Tacrolimus 71-81 ATP binding cassette subfamily B member 1 Homo sapiens 21-26 26856709-1 2016 AIM OF WORK: To assess the impact of Cytochrome P450 3A5 (CYP3A5) and multidrug resistance-1 gene (MDR-1) single nucleotide polymorphisms on the dose and blood level of tacrolimus among liver transplanted patients. Tacrolimus 169-179 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-56 26856709-1 2016 AIM OF WORK: To assess the impact of Cytochrome P450 3A5 (CYP3A5) and multidrug resistance-1 gene (MDR-1) single nucleotide polymorphisms on the dose and blood level of tacrolimus among liver transplanted patients. Tacrolimus 169-179 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 58-64 26856709-1 2016 AIM OF WORK: To assess the impact of Cytochrome P450 3A5 (CYP3A5) and multidrug resistance-1 gene (MDR-1) single nucleotide polymorphisms on the dose and blood level of tacrolimus among liver transplanted patients. Tacrolimus 169-179 ATP binding cassette subfamily B member 1 Homo sapiens 70-92 26856709-1 2016 AIM OF WORK: To assess the impact of Cytochrome P450 3A5 (CYP3A5) and multidrug resistance-1 gene (MDR-1) single nucleotide polymorphisms on the dose and blood level of tacrolimus among liver transplanted patients. Tacrolimus 169-179 ATP binding cassette subfamily B member 1 Homo sapiens 99-104 26856709-6 2016 CONCLUSION: CYP3A5 (6986A > G) genotype, rather than MDR-1 (2677G > A/T) variant, has an impact on tacrolimus pharmacokinetics. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 27546370-4 2016 OBJECTIVE: In this investigation, we evaluated the molecular mechanism of FK-506 in apoptosis using gene expression quantification of BAX and BCL-2 genes in hippocampus following global ischemic/reperfusion. Tacrolimus 74-80 BCL2, apoptosis regulator Rattus norvegicus 142-147 27546370-14 2016 CONCLUSION: The results showed that global cerebral ischemia/reperfusion induced BAX as pro-apoptotic gene and tacrolimus a neuroprotective drug inhibited BAX gene expression and induced BCL-2 gene expression as anti-apoptotic gene (Tab. Tacrolimus 111-121 BCL2 associated X, apoptosis regulator Rattus norvegicus 155-158 27546370-14 2016 CONCLUSION: The results showed that global cerebral ischemia/reperfusion induced BAX as pro-apoptotic gene and tacrolimus a neuroprotective drug inhibited BAX gene expression and induced BCL-2 gene expression as anti-apoptotic gene (Tab. Tacrolimus 111-121 BCL2, apoptosis regulator Rattus norvegicus 187-192 27546370-0 2016 Assessment of global ischemic/reperfusion and Tacrolimus administration on CA1 region of hippocampus: gene expression profiles of BAX and BCL2 genes. Tacrolimus 46-56 BCL2 associated X, apoptosis regulator Rattus norvegicus 130-133 27546370-0 2016 Assessment of global ischemic/reperfusion and Tacrolimus administration on CA1 region of hippocampus: gene expression profiles of BAX and BCL2 genes. Tacrolimus 46-56 BCL2, apoptosis regulator Rattus norvegicus 138-142 27546370-4 2016 OBJECTIVE: In this investigation, we evaluated the molecular mechanism of FK-506 in apoptosis using gene expression quantification of BAX and BCL-2 genes in hippocampus following global ischemic/reperfusion. Tacrolimus 74-80 BCL2 associated X, apoptosis regulator Rattus norvegicus 134-137 27771715-0 2016 ER Stress via CHOP Pathway is Involved in FK506-Induced Apoptosis in Rat Fibroblasts. Tacrolimus 42-47 DNA-damage inducible transcript 3 Rattus norvegicus 14-18 26849622-7 2016 The insulin-induced elevation of TRPC6 transcripts was blocked in the presence of tacrolimus, cyclosporine A, and NFAT-inhibitor (each p < 0.01 by ANOVA and Bonferroni"s multiple comparison test). Tacrolimus 82-92 insulin Homo sapiens 4-11 26849622-7 2016 The insulin-induced elevation of TRPC6 transcripts was blocked in the presence of tacrolimus, cyclosporine A, and NFAT-inhibitor (each p < 0.01 by ANOVA and Bonferroni"s multiple comparison test). Tacrolimus 82-92 transient receptor potential cation channel subfamily C member 6 Homo sapiens 33-38 26849622-10 2016 Insulin increased the activity of NFATc1 in nuclear extracts (p < 0.001) whereas tacrolimus, cyclosporine A, and NFAT-inhibitor blocked that insulin effect (p < 0.001; two way ANOVA). Tacrolimus 84-94 insulin Homo sapiens 144-151 26849622-13 2016 Insulin-stimulated surface expression of TRPC6 as well as transplasmamembrane cation influx could be reduced by pretreatment with tacrolimus. Tacrolimus 130-140 insulin Homo sapiens 0-7 26849622-13 2016 Insulin-stimulated surface expression of TRPC6 as well as transplasmamembrane cation influx could be reduced by pretreatment with tacrolimus. Tacrolimus 130-140 transient receptor potential cation channel subfamily C member 6 Homo sapiens 41-46 27771715-13 2016 We further demonstrated that FK506-induced apoptosis was mediated by ER stress via activating CHOP, evidenced by decreased apoptosis after inhibition of ER stress using TUDCA or silencing expression of CHOP. Tacrolimus 29-34 DNA-damage inducible transcript 3 Rattus norvegicus 94-98 27771715-13 2016 We further demonstrated that FK506-induced apoptosis was mediated by ER stress via activating CHOP, evidenced by decreased apoptosis after inhibition of ER stress using TUDCA or silencing expression of CHOP. Tacrolimus 29-34 DNA-damage inducible transcript 3 Rattus norvegicus 202-206 27771715-14 2016 Furthermore, Co-immunoprecipitation results indicated that treatment of FK506 induced disassociation of FKBP12.6 from RyR2 and its translocation from ER membrane to cytosol, consequently promoting ER stress-mediated apoptosis. Tacrolimus 72-77 FKBP prolyl isomerase 1B Rattus norvegicus 104-112 27771715-14 2016 Furthermore, Co-immunoprecipitation results indicated that treatment of FK506 induced disassociation of FKBP12.6 from RyR2 and its translocation from ER membrane to cytosol, consequently promoting ER stress-mediated apoptosis. Tacrolimus 72-77 ryanodine receptor 2 Rattus norvegicus 118-122 27771715-15 2016 CONCLUSION: FK506-induced fibroblasts apoptosis was mediated by ER stress via CHOP signaling pathway. Tacrolimus 12-17 DNA-damage inducible transcript 3 Rattus norvegicus 78-82 27623226-6 2016 Although tacrolimus causes an elevation of total cholesterol and LDL-c, the significant alterations of the children lipid profile suggest the existence of a high cardiovascular risk. Tacrolimus 9-19 component of oligomeric golgi complex 2 Homo sapiens 65-70 27644556-6 2016 Secretion of IL-13 and IL-17A in CD4(+) T cells was lower in DHA/EPA- and FK506-treated mice than in mice treated with FK506 alone. Tacrolimus 74-79 interleukin 13 Mus musculus 13-18 26651976-1 2016 BACKGROUND: The contribution of the CYP3A5 enzyme to the metabolism of clinically used drugs has been established only for a few CYP3A substrates, such as the immunosuppressant tacrolimus, while for drugs used in the field of psychiatry its role is still vague. Tacrolimus 177-187 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 26651976-1 2016 BACKGROUND: The contribution of the CYP3A5 enzyme to the metabolism of clinically used drugs has been established only for a few CYP3A substrates, such as the immunosuppressant tacrolimus, while for drugs used in the field of psychiatry its role is still vague. Tacrolimus 177-187 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 36-41 26471483-8 2016 The in vitro results showed that these drugs had no effect on phenotype, function and stability of Tregs, although tacrolimus affected the expression of chemokine receptors and IL-10 production. Tacrolimus 115-125 interleukin 10 Homo sapiens 177-182 26613512-9 2016 Moreover, we showed a significant decrease in activation of mTOR Complex 1 downstream targets after treatment with everolimus that was attenuated when combined with tacrolimus, but not with cyclosporin A. Tacrolimus 165-175 mechanistic target of rapamycin kinase Homo sapiens 60-64 27644556-6 2016 Secretion of IL-13 and IL-17A in CD4(+) T cells was lower in DHA/EPA- and FK506-treated mice than in mice treated with FK506 alone. Tacrolimus 74-79 interleukin 17A Mus musculus 23-29 27644556-6 2016 Secretion of IL-13 and IL-17A in CD4(+) T cells was lower in DHA/EPA- and FK506-treated mice than in mice treated with FK506 alone. Tacrolimus 74-79 CD4 antigen Mus musculus 33-36 27644556-6 2016 Secretion of IL-13 and IL-17A in CD4(+) T cells was lower in DHA/EPA- and FK506-treated mice than in mice treated with FK506 alone. Tacrolimus 119-124 interleukin 13 Mus musculus 13-18 27644556-6 2016 Secretion of IL-13 and IL-17A in CD4(+) T cells was lower in DHA/EPA- and FK506-treated mice than in mice treated with FK506 alone. Tacrolimus 119-124 interleukin 17A Mus musculus 23-29 27644556-6 2016 Secretion of IL-13 and IL-17A in CD4(+) T cells was lower in DHA/EPA- and FK506-treated mice than in mice treated with FK506 alone. Tacrolimus 119-124 CD4 antigen Mus musculus 33-36 27644569-13 2016 Stress gastric ulcer was controlled and myeloperoxidase activity in the gastric mucosa was suppressed by FK506. Tacrolimus 105-110 myeloperoxidase Sus scrofa 40-55 26905810-0 2016 Tacrolimus restores podocyte injury and stabilizes the expression of Cabin1 in 5/6 nephrectomized rats. Tacrolimus 0-10 calcineurin binding protein 1 Rattus norvegicus 69-75 27486667-10 2016 Of the 6 SNPs, rs1927914 in the TLR4 gene and rs1039559 in the TLR6 gene were significantly associated with the development of PTDM after adjustment for age, gender, and tacrolimus usage. Tacrolimus 170-180 toll like receptor 4 Homo sapiens 32-36 27486667-10 2016 Of the 6 SNPs, rs1927914 in the TLR4 gene and rs1039559 in the TLR6 gene were significantly associated with the development of PTDM after adjustment for age, gender, and tacrolimus usage. Tacrolimus 170-180 toll like receptor 6 Homo sapiens 63-67 27190501-3 2016 Rapamycin and FK506 bind to FK506-binding proteins (FKBPs), such as FKBP12, which causes suppression of the immune system and inhibition of mTOR. Tacrolimus 14-19 FK506 binding protein 1a Mus musculus 68-74 27190501-3 2016 Rapamycin and FK506 bind to FK506-binding proteins (FKBPs), such as FKBP12, which causes suppression of the immune system and inhibition of mTOR. Tacrolimus 14-19 mechanistic target of rapamycin kinase Mus musculus 140-144 26561008-1 2015 The tight complexes FKBP12 forms with immunosuppressive drugs, such as FK506 and rapamycin, are frequently used as models for developing approaches to structure-based drug design. Tacrolimus 71-76 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 20-26 26341908-9 2015 We tested our classifier with FK506, which regulates mitochondrial calcium and found that this drug modulated the WT alpha-syn calcium transients to such an extent that the classifier easily identified the calcium transients as belonging to Non-tg mice. Tacrolimus 30-35 synuclein, alpha Mus musculus 117-126 26271661-2 2015 CYP3A5*3, CYP3A4*1B and CYP3A4*22 alleles of liver grafts may explain about one third of the inter-individual differences in pharmacokinetics of ciclosporin and tacrolimus in recipients. Tacrolimus 161-171 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 26915843-8 2016 Compared to those who were being administered triple ISx, recipients receiving tacrolimus-based dual and monotherapies had lower use of statins, angiotensin-converting enzyme inhibitors/angiotensin II receptor blockers (ACEi/ARBs), and antibacterial agents. Tacrolimus 79-89 intestine specific homeobox Homo sapiens 53-56 26915847-0 2016 Effects of CYP3A5 Genetic Polymorphism on the Pharmacokinetics of Tacrolimus in Renal Transplant Recipients. Tacrolimus 66-76 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 11-17 26915847-8 2016 The mean maintenance dose of tacrolimus required for those with CYP3A5*1/*1 (high-expressive) was significantly higher than those with CYP3A5*1/*3 (intermediate-expressive) and CYP3A5*3/*3 (low-expressive) (P < .01) for both formulations. Tacrolimus 29-39 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 64-70 26915847-10 2016 CONCLUSIONS: The requirement for daily maintenance dose was higher in those with CYP3A5*1/*1 variants in both tacrolimus formulations in the Malaysian patients. Tacrolimus 110-120 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 81-87 26756013-0 2015 Gene Expression Profiles of BAD and Bcl-xL in the CA1 Region of the Hippocampus Following Global Ischemic/Reperfusion and FK-506 Administration. Tacrolimus 122-128 Bcl2-like 1 Rattus norvegicus 36-42 26756013-0 2015 Gene Expression Profiles of BAD and Bcl-xL in the CA1 Region of the Hippocampus Following Global Ischemic/Reperfusion and FK-506 Administration. Tacrolimus 122-128 carbonic anhydrase 1 Rattus norvegicus 50-53 26756013-6 2015 OBJECTIVES: In this study, we assessed the neurotrophic properties of FK-506 on expression of the BAD and Bcl-xL genes in the hippocampus following global ischemia and reperfusion. Tacrolimus 70-76 Bcl2-like 1 Rattus norvegicus 106-112 26756013-14 2015 RESULTS: The quantitative results of real-time PCR show that the mRNA expression ratio of Bcl-xL down-regulated was 0.75 +- 0.06 in the ischemia/reperfusion group versus 1.57 +- 0.09 in the control group (P value < 0.001), whereas Bcl-xL gene expression was greater in the ischemia/reperfusion +FK506 group (1.93 +- 0.15) than in the ischemia/reperfusion group. Tacrolimus 298-303 Bcl2-like 1 Rattus norvegicus 90-96 26271661-2 2015 CYP3A5*3, CYP3A4*1B and CYP3A4*22 alleles of liver grafts may explain about one third of the inter-individual differences in pharmacokinetics of ciclosporin and tacrolimus in recipients. Tacrolimus 161-171 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 10-16 26271661-2 2015 CYP3A5*3, CYP3A4*1B and CYP3A4*22 alleles of liver grafts may explain about one third of the inter-individual differences in pharmacokinetics of ciclosporin and tacrolimus in recipients. Tacrolimus 161-171 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-30 26271661-6 2015 RESULTS: The recipients with grafts from normal CYP3A4 expresser donors carrying CYP3A5*3/*3 required CNI maintenance doses more or less similar to the bodyweight-controlled starting doses (9.1 mg kg(-1) of ciclosporin and 0.1 mg kg(-1) of tacrolimus). Tacrolimus 240-250 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 81-87 26331794-8 2015 The serum erythropoietin level was significantly higher in transplant recipients taking tacrolimus. Tacrolimus 88-98 erythropoietin Homo sapiens 10-24 26615671-1 2015 AIM: The present study investigated in Tunisian renal transplant patients, genetic polymorphisms of CYP3A4 -392A>G and CYP3A5 6986A>G and their influence on tacrolimus (Tac) pharmacokinetics during early and late post-transplant (PT) phases and established customized ranges of Tac doses matching the C0 target levels according to CYP3A4 and CYP3A5 genotype combination and the PT phase. Tacrolimus 163-173 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 100-106 26615671-1 2015 AIM: The present study investigated in Tunisian renal transplant patients, genetic polymorphisms of CYP3A4 -392A>G and CYP3A5 6986A>G and their influence on tacrolimus (Tac) pharmacokinetics during early and late post-transplant (PT) phases and established customized ranges of Tac doses matching the C0 target levels according to CYP3A4 and CYP3A5 genotype combination and the PT phase. Tacrolimus 163-173 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 122-128 26615671-1 2015 AIM: The present study investigated in Tunisian renal transplant patients, genetic polymorphisms of CYP3A4 -392A>G and CYP3A5 6986A>G and their influence on tacrolimus (Tac) pharmacokinetics during early and late post-transplant (PT) phases and established customized ranges of Tac doses matching the C0 target levels according to CYP3A4 and CYP3A5 genotype combination and the PT phase. Tacrolimus 163-173 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 337-343 26615671-1 2015 AIM: The present study investigated in Tunisian renal transplant patients, genetic polymorphisms of CYP3A4 -392A>G and CYP3A5 6986A>G and their influence on tacrolimus (Tac) pharmacokinetics during early and late post-transplant (PT) phases and established customized ranges of Tac doses matching the C0 target levels according to CYP3A4 and CYP3A5 genotype combination and the PT phase. Tacrolimus 163-173 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 348-354 26322864-5 2015 Inspired by the natural product FK506, we designed molecules that have affinity for both FKBP12 and HIV protease. Tacrolimus 32-37 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 89-95 26567527-3 2015 FKBP6 is a peptidyl prolyl cis-trans isomerase with three domains of the tetratricopeptide repeat (TPR), but lacks FK-506 binding ability. Tacrolimus 115-121 FKBP prolyl isomerase family member 6 (inactive) Homo sapiens 0-5 26318285-8 2015 FK506 and cyclosporine A, inhibitors of calcineurin activation, determined that calcineurin was associated with the iron-induced changes in mitochondrial morphology and the phosphorylation levels of Drp1. Tacrolimus 0-5 collapsin response mediator protein 1 Mus musculus 199-203 25431882-2 2015 To investigate the possible effect of tacrolimus on the development of impaired glucose tolerance in transplant recipients, this study focused on early and second phase insulin secretion, which may be affected by reactive oxygen species under tacrolimus therapy. Tacrolimus 38-48 insulin Homo sapiens 169-176 25431882-2 2015 To investigate the possible effect of tacrolimus on the development of impaired glucose tolerance in transplant recipients, this study focused on early and second phase insulin secretion, which may be affected by reactive oxygen species under tacrolimus therapy. Tacrolimus 243-253 insulin Homo sapiens 169-176 25925687-10 2015 Analyses showed that >16 HLA-DQ epitope mismatches and pretransplant, peripheral blood, donor-reactive IFN-gamma ELISPOT assay results correlated with development of DSAs and/or AR on tacrolimus withdrawal. Tacrolimus 187-197 interferon gamma Homo sapiens 106-115 24737642-10 2015 CONCLUSION: These results suggest that aliskiren has protective effects against tacrolimus-induced nephrotoxicity; implying that renin inhibitor may counteract nephrotic syndrome associated with immunosuppressant use. Tacrolimus 80-90 renin Rattus norvegicus 129-134 26634478-0 2015 Effect of CYP3A5 gene polymorphisms on tacrolimus concentration/dosage ratio in adult liver transplant patients. Tacrolimus 39-49 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 26634478-1 2015 We examined the influence of the cytochrome P450 3A5 (CYP3A5) genes in both donors and recipients on the concentration-dosage ratio (C/D) of tacrolimus in Chinese liver transplant patients. Tacrolimus 141-151 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-52 26634478-1 2015 We examined the influence of the cytochrome P450 3A5 (CYP3A5) genes in both donors and recipients on the concentration-dosage ratio (C/D) of tacrolimus in Chinese liver transplant patients. Tacrolimus 141-151 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 54-60 26634478-4 2015 The relationship between the C/D of tacrolimus for 3 months after surgery and the CYP3A5 genotype was analyzed. Tacrolimus 36-46 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 82-88 26634478-7 2015 The C/D of tacrolimus in patients with the CYP3A5*1 allele or carrying CYP3A5*1 allele in the liver was lower than that in CYP3A5*3/*3 patients with the CYP3A5*3/*3 genotype in the liver (P < 0.01). Tacrolimus 11-21 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 43-49 26634478-7 2015 The C/D of tacrolimus in patients with the CYP3A5*1 allele or carrying CYP3A5*1 allele in the liver was lower than that in CYP3A5*3/*3 patients with the CYP3A5*3/*3 genotype in the liver (P < 0.01). Tacrolimus 11-21 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 71-77 26634478-7 2015 The C/D of tacrolimus in patients with the CYP3A5*1 allele or carrying CYP3A5*1 allele in the liver was lower than that in CYP3A5*3/*3 patients with the CYP3A5*3/*3 genotype in the liver (P < 0.01). Tacrolimus 11-21 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 71-77 26634478-7 2015 The C/D of tacrolimus in patients with the CYP3A5*1 allele or carrying CYP3A5*1 allele in the liver was lower than that in CYP3A5*3/*3 patients with the CYP3A5*3/*3 genotype in the liver (P < 0.01). Tacrolimus 11-21 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 71-77 26634478-8 2015 The CYP3A5*1 genotype in donors as well as in patients both contributes to interindividual variation in the C/D of tacrolimus in adult liver transplantation. Tacrolimus 115-125 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 26575262-1 2015 The calcineurin inhibitor tacrolimus is the cornerstone of most immunosuppressive treatment protocols after solid organ transplantation in the United States. Tacrolimus 26-36 calcineurin binding protein 1 Homo sapiens 4-25 26436650-8 2015 Moreover, NFATC3 nuclear translocation, synaptopodin degradation, integrin beta3 (ITGB3) activation, and actin fiber loss, which are downstream of calcium/calcineurin signaling, were induced by miR-30 reduction but blocked by the calcineurin inhibitor FK506. Tacrolimus 252-257 nuclear factor of activated T cells 3 Homo sapiens 10-16 26436650-8 2015 Moreover, NFATC3 nuclear translocation, synaptopodin degradation, integrin beta3 (ITGB3) activation, and actin fiber loss, which are downstream of calcium/calcineurin signaling, were induced by miR-30 reduction but blocked by the calcineurin inhibitor FK506. Tacrolimus 252-257 membrane associated ring-CH-type finger 8 Homo sapiens 194-197 26362749-6 2015 Olopatadine also significantly inhibited increases in vascular permeability and nerve growth factor production in the skin induced by topical tacrolimus treatment. Tacrolimus 142-152 nerve growth factor Rattus norvegicus 80-99 26823720-0 2015 FK506 ameliorates podocyte injury in type 2 diabetic nephropathy by down-regulating TRPC6 and NFAT expression. Tacrolimus 0-5 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 84-89 26823720-0 2015 FK506 ameliorates podocyte injury in type 2 diabetic nephropathy by down-regulating TRPC6 and NFAT expression. Tacrolimus 0-5 nuclear factor of activated T-cells 5 Rattus norvegicus 94-98 26823720-6 2015 In the rat model of DN, the expressions of TRPC6 and NFAT were significantly elevated compared with the normal rat group; however, the treatment with FK506 normalized the increased expression of TRPC6 and NFAT and attenuated podocyte ultrastructure injury. Tacrolimus 150-155 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 43-48 26823720-6 2015 In the rat model of DN, the expressions of TRPC6 and NFAT were significantly elevated compared with the normal rat group; however, the treatment with FK506 normalized the increased expression of TRPC6 and NFAT and attenuated podocyte ultrastructure injury. Tacrolimus 150-155 nuclear factor of activated T-cells 5 Rattus norvegicus 53-57 26823720-6 2015 In the rat model of DN, the expressions of TRPC6 and NFAT were significantly elevated compared with the normal rat group; however, the treatment with FK506 normalized the increased expression of TRPC6 and NFAT and attenuated podocyte ultrastructure injury. Tacrolimus 150-155 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 195-200 26823720-6 2015 In the rat model of DN, the expressions of TRPC6 and NFAT were significantly elevated compared with the normal rat group; however, the treatment with FK506 normalized the increased expression of TRPC6 and NFAT and attenuated podocyte ultrastructure injury. Tacrolimus 150-155 nuclear factor of activated T-cells 5 Rattus norvegicus 205-209 26823720-8 2015 In cell experiments, FK506 improved the decreased expression of nephrin and suppressed the elevated expression of both TRPC6 and NFAT caused by high glucose in accordance with TRPC6 blocker U73122. Tacrolimus 21-26 NPHS1 adhesion molecule, nephrin Rattus norvegicus 64-71 26823720-8 2015 In cell experiments, FK506 improved the decreased expression of nephrin and suppressed the elevated expression of both TRPC6 and NFAT caused by high glucose in accordance with TRPC6 blocker U73122. Tacrolimus 21-26 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 119-124 26823720-8 2015 In cell experiments, FK506 improved the decreased expression of nephrin and suppressed the elevated expression of both TRPC6 and NFAT caused by high glucose in accordance with TRPC6 blocker U73122. Tacrolimus 21-26 nuclear factor of activated T-cells 5 Rattus norvegicus 129-133 26823720-8 2015 In cell experiments, FK506 improved the decreased expression of nephrin and suppressed the elevated expression of both TRPC6 and NFAT caused by high glucose in accordance with TRPC6 blocker U73122. Tacrolimus 21-26 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 176-181 26823720-9 2015 Our results demonstrated that FK506 could ameliorate podocyte injury in T2DM, which may be related to suppressed expressions of TRPC6 and NFAT. Tacrolimus 30-35 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 128-133 26823720-9 2015 Our results demonstrated that FK506 could ameliorate podocyte injury in T2DM, which may be related to suppressed expressions of TRPC6 and NFAT. Tacrolimus 30-35 nuclear factor of activated T-cells 5 Rattus norvegicus 138-142 26232156-10 2015 In the second case, the patient was revealed to have an uncommon genotype for CYP3A5, causing higher metabolism and lower serum tacrolimus concentrations than the general population. Tacrolimus 128-138 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 78-84 26362749-5 2015 These results suggest that H1R antagonistic activity-independent mechanism contribute to the inhibitory effect of olopatadine on tacrolimus-induced skin temperature elevation. Tacrolimus 129-139 histamine receptor H 1 Rattus norvegicus 27-30 26875279-0 2015 [EPIDERMAI GROWTH FACTOR SECRETED BY ASTROCYTES STIMULATED WITH TACROLIMUS PROMOTING NEURITE OUTGROWTH]. Tacrolimus 64-74 myotrophin Rattus norvegicus 11-24 26875279-1 2015 OBJECTIVE: To illustrate the role of epidermal growth factor (EGF) secreted by astrocytes in the process of tacrolimus (FK506) in promoting neurite outgrowth. Tacrolimus 108-118 epidermal growth factor like 1 Rattus norvegicus 37-60 26875279-1 2015 OBJECTIVE: To illustrate the role of epidermal growth factor (EGF) secreted by astrocytes in the process of tacrolimus (FK506) in promoting neurite outgrowth. Tacrolimus 108-118 epidermal growth factor like 1 Rattus norvegicus 62-65 26875279-1 2015 OBJECTIVE: To illustrate the role of epidermal growth factor (EGF) secreted by astrocytes in the process of tacrolimus (FK506) in promoting neurite outgrowth. Tacrolimus 120-125 epidermal growth factor like 1 Rattus norvegicus 37-60 26875279-1 2015 OBJECTIVE: To illustrate the role of epidermal growth factor (EGF) secreted by astrocytes in the process of tacrolimus (FK506) in promoting neurite outgrowth. Tacrolimus 120-125 epidermal growth factor like 1 Rattus norvegicus 62-65 26543771-0 2015 CYP3A5 and ABCB1 genotype influence tacrolimus and sirolimus pharmacokinetics in renal transplant recipients. Tacrolimus 36-46 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 26543771-0 2015 CYP3A5 and ABCB1 genotype influence tacrolimus and sirolimus pharmacokinetics in renal transplant recipients. Tacrolimus 36-46 ATP binding cassette subfamily B member 1 Homo sapiens 11-16 26543771-1 2015 CYP3A5 and ABCB1 polymorphisms have been shown to influence tacrolimus blood concentrations and dose requirements, but the conclusion in the current reports were inconformity. Tacrolimus 60-70 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 26543771-1 2015 CYP3A5 and ABCB1 polymorphisms have been shown to influence tacrolimus blood concentrations and dose requirements, but the conclusion in the current reports were inconformity. Tacrolimus 60-70 ATP binding cassette subfamily B member 1 Homo sapiens 11-16 26133248-1 2015 PURPOSE: To evaluate topical tacrolimus ointment for treating Thygeson"s superficial punctate keratitis (Thygeson SPK). Tacrolimus 29-39 symplekin scaffold protein Homo sapiens 114-117 26770526-2 2015 The genetic polymorphisms of CYP3A4, CYP3A5 and MDR1 in 216 RT patients were detected by PCR-RFLP, the genetic and clinical factors and blood concentration/dose x body weight (C/D) values of tacrolimus were performed the single factor correlation analysis, and established the dose prediction algorithm of tacrolimus by stepwise multiple regression analysis. Tacrolimus 191-201 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-35 26770526-2 2015 The genetic polymorphisms of CYP3A4, CYP3A5 and MDR1 in 216 RT patients were detected by PCR-RFLP, the genetic and clinical factors and blood concentration/dose x body weight (C/D) values of tacrolimus were performed the single factor correlation analysis, and established the dose prediction algorithm of tacrolimus by stepwise multiple regression analysis. Tacrolimus 191-201 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 26770526-2 2015 The genetic polymorphisms of CYP3A4, CYP3A5 and MDR1 in 216 RT patients were detected by PCR-RFLP, the genetic and clinical factors and blood concentration/dose x body weight (C/D) values of tacrolimus were performed the single factor correlation analysis, and established the dose prediction algorithm of tacrolimus by stepwise multiple regression analysis. Tacrolimus 191-201 ATP binding cassette subfamily B member 1 Homo sapiens 48-52 26770526-2 2015 The genetic polymorphisms of CYP3A4, CYP3A5 and MDR1 in 216 RT patients were detected by PCR-RFLP, the genetic and clinical factors and blood concentration/dose x body weight (C/D) values of tacrolimus were performed the single factor correlation analysis, and established the dose prediction algorithm of tacrolimus by stepwise multiple regression analysis. Tacrolimus 306-316 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-35 26770526-2 2015 The genetic polymorphisms of CYP3A4, CYP3A5 and MDR1 in 216 RT patients were detected by PCR-RFLP, the genetic and clinical factors and blood concentration/dose x body weight (C/D) values of tacrolimus were performed the single factor correlation analysis, and established the dose prediction algorithm of tacrolimus by stepwise multiple regression analysis. Tacrolimus 306-316 ATP binding cassette subfamily B member 1 Homo sapiens 48-52 26770526-3 2015 CYP3A5*3, hematocrit and albumin were correlated with the C/D values of tacrolimus, the best regression model could explain 28.3% reason of individual dose differences of tacrolimus, among which CYP3A5*3 polymorphism could explain 23.5%. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 26770526-3 2015 CYP3A5*3, hematocrit and albumin were correlated with the C/D values of tacrolimus, the best regression model could explain 28.3% reason of individual dose differences of tacrolimus, among which CYP3A5*3 polymorphism could explain 23.5%. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 195-201 26770526-3 2015 CYP3A5*3, hematocrit and albumin were correlated with the C/D values of tacrolimus, the best regression model could explain 28.3% reason of individual dose differences of tacrolimus, among which CYP3A5*3 polymorphism could explain 23.5%. Tacrolimus 171-181 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 26770526-3 2015 CYP3A5*3, hematocrit and albumin were correlated with the C/D values of tacrolimus, the best regression model could explain 28.3% reason of individual dose differences of tacrolimus, among which CYP3A5*3 polymorphism could explain 23.5%. Tacrolimus 171-181 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 195-201 26770526-4 2015 The genetic factors played an important role in the dose differences of tacrolimus, the patients should be checked CYP3A5*3 genotype before administration of tacrolimus to predict the tacrolimus doses, thus helping to improve the safety and effectiveness of tacrolimus application. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 115-121 26133248-15 2015 CONCLUSION: Tacrolimus eye ointment 0.03% was effective for controlling Thygeson SPK for a long period with good patient tolerance and no noticeable local or systemic side effects. Tacrolimus 12-22 symplekin scaffold protein Homo sapiens 81-84 26237274-7 2015 These data suggest that DPP IV inhibition has an important role in the renoprotection against tacrolimus-induced nephrotoxicity via antioxidative and antiapoptotic effects and preservation of the GLP-1 system. Tacrolimus 94-104 dipeptidylpeptidase 4 Rattus norvegicus 24-30 26224283-7 2015 CXCL10 is a cytokine that recruits CXCR3(+) cells such as activated T cells, and we found that FK506 treatment specifically decreased CSF CXCL10 from among 27 cytokines tested. Tacrolimus 95-100 C-X-C motif chemokine ligand 10 Homo sapiens 0-6 26224283-7 2015 CXCL10 is a cytokine that recruits CXCR3(+) cells such as activated T cells, and we found that FK506 treatment specifically decreased CSF CXCL10 from among 27 cytokines tested. Tacrolimus 95-100 C-X-C motif chemokine receptor 3 Homo sapiens 35-40 26224283-7 2015 CXCL10 is a cytokine that recruits CXCR3(+) cells such as activated T cells, and we found that FK506 treatment specifically decreased CSF CXCL10 from among 27 cytokines tested. Tacrolimus 95-100 C-X-C motif chemokine ligand 10 Homo sapiens 138-144 25529296-1 2015 BACKGROUND: FK506-binding proteins (FKBPs) contain a domain with peptidyl-prolyl-cis/trans-isomerase (PPIase) activity and bind the immunosuppressive drugs FK506 and rapamycin. Tacrolimus 12-17 FKBP prolyl isomerase like Homo sapiens 65-100 25529296-1 2015 BACKGROUND: FK506-binding proteins (FKBPs) contain a domain with peptidyl-prolyl-cis/trans-isomerase (PPIase) activity and bind the immunosuppressive drugs FK506 and rapamycin. Tacrolimus 12-17 FKBP prolyl isomerase like Homo sapiens 102-108 26517911-5 2015 Both physically interact with orthologues of the FK506-binding proteins that chaperon both transporters to the plasma membrane in an action that seems to involve heat shock protein (Hsp)90. Tacrolimus 49-54 heat shock protein 90 alpha family class A member 1 Homo sapiens 182-188 26450467-0 2015 Combinational Effect of CYP3A5 and MDR-1 Polymorphisms on Tacrolimus Pharmacokinetics in Liver Transplant Patients. Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-30 26450467-0 2015 Combinational Effect of CYP3A5 and MDR-1 Polymorphisms on Tacrolimus Pharmacokinetics in Liver Transplant Patients. Tacrolimus 58-68 ATP binding cassette subfamily B member 1 Homo sapiens 35-40 26450467-1 2015 OBJECTIVES: Previous studies have reported reduced tacrolimus dose-adjusted exposure in individuals expressing the CYP3A5*1 allele (reference single nucleotide polymorphism identification number 776746). Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 115-121 26450467-3 2015 The objective of this study was to investigate the influence of CYP3A5 and MDR1 allelic variants and their correlation on the pharmacokinetics of tacrolimus and a modified release formulation of tacrolimus in stable patients with liver transplant. Tacrolimus 146-156 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 64-70 26450467-3 2015 The objective of this study was to investigate the influence of CYP3A5 and MDR1 allelic variants and their correlation on the pharmacokinetics of tacrolimus and a modified release formulation of tacrolimus in stable patients with liver transplant. Tacrolimus 146-156 ATP binding cassette subfamily B member 1 Homo sapiens 75-79 26450467-3 2015 The objective of this study was to investigate the influence of CYP3A5 and MDR1 allelic variants and their correlation on the pharmacokinetics of tacrolimus and a modified release formulation of tacrolimus in stable patients with liver transplant. Tacrolimus 195-205 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 64-70 26450467-3 2015 The objective of this study was to investigate the influence of CYP3A5 and MDR1 allelic variants and their correlation on the pharmacokinetics of tacrolimus and a modified release formulation of tacrolimus in stable patients with liver transplant. Tacrolimus 195-205 ATP binding cassette subfamily B member 1 Homo sapiens 75-79 26450467-9 2015 Dose levels of tacrolimus and the modified formulation were significantly higher in donors and recipients who expressed CYP3A5 versus donors and recipients who did not express this allele. Tacrolimus 15-25 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 120-126 26450467-10 2015 In addition, patients who received a liver from a donor expressing CYP3A5 had significantly lower trough concentrations of tacrolimus and the modified formulation. Tacrolimus 123-133 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-73 26450467-11 2015 CYP3A5 expression in the donor liver affected tacrolimus (40.46%, P = .001) and modified formulation (37.56%, P = .001) variability. Tacrolimus 46-56 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 26450467-13 2015 CONCLUSIONS: Our data suggest that CYP3A5*1 in either the donor or recipient resulted in higher mean daily doses of tacrolimus or its modified formulation to achieve target drug exposure in liver transplant patients. Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 35-41 26198442-5 2015 OBJECTIVE: To investigate whether the glucocorticosteroid dexamethasone and the calcineurin inhibitor tacrolimus could affect TSLP expression induced by TNF-alpha in lesional keratinocytes of AD. Tacrolimus 102-112 calcineurin binding protein 1 Mus musculus 80-101 26198442-5 2015 OBJECTIVE: To investigate whether the glucocorticosteroid dexamethasone and the calcineurin inhibitor tacrolimus could affect TSLP expression induced by TNF-alpha in lesional keratinocytes of AD. Tacrolimus 102-112 thymic stromal lymphopoietin Mus musculus 126-130 26198442-5 2015 OBJECTIVE: To investigate whether the glucocorticosteroid dexamethasone and the calcineurin inhibitor tacrolimus could affect TSLP expression induced by TNF-alpha in lesional keratinocytes of AD. Tacrolimus 102-112 tumor necrosis factor Mus musculus 153-162 26198442-6 2015 METHODS: The effects of topical dexamethasone and tacrolimus on TSLP expression were evaluated in an AD mouse model induced by repeated 2,4,6-trinitro-1-chlorobenzene application. Tacrolimus 50-60 thymic stromal lymphopoietin Mus musculus 64-68 26150050-6 2015 The combined clearance of 6beta-hydroxycortisol and 6beta-hydroxycortisone correlated significantly with cyclosporine pharmacokinetics (p < 0.001) after oral dose of a BCS 1 formulation, whereas no relationships were seen after administration of tacrolimus and sirolimus formulations, both of which belonged to BCS 2. Tacrolimus 249-259 BCS1 homolog, ubiquinol-cytochrome c reductase complex chaperone Homo sapiens 171-176 26237274-0 2015 Inhibition of dipeptidyl peptidase IV protects tacrolimus-induced kidney injury. Tacrolimus 47-57 dipeptidylpeptidase 4 Rattus norvegicus 14-37 26237274-2 2015 In this study, we investigated whether a dipeptidyl peptidase (DPP) IV inhibitor has a protective effect against tacrolimus-induced renal injury. Tacrolimus 113-123 dipeptidylpeptidase 4 Rattus norvegicus 41-70 26239116-7 2015 In HEK-293 cells overexpressing TRPC6, currents through TRPC6 were altered in the presence of FK506. Tacrolimus 94-99 transient receptor potential cation channel subfamily C member 6 Homo sapiens 32-37 26239116-7 2015 In HEK-293 cells overexpressing TRPC6, currents through TRPC6 were altered in the presence of FK506. Tacrolimus 94-99 transient receptor potential cation channel subfamily C member 6 Homo sapiens 56-61 26239116-9 2015 FK506 modified the pattern of association between FKBP25 and TRPCs as well as impaired OAG-evoked TRPC3 and TRPC6 coupling in both human and mouse platelets. Tacrolimus 0-5 FKBP prolyl isomerase 3 Homo sapiens 50-56 28356851-1 2015 BACKGROUND: The primary goal of this study was to evaluate the influence of cytochrome P450 (CYP) 3A5 (6986A>G) and ABCB1 (3435C>T) polymorphisms on tacrolimus (TAC) dosage regimen and exposure. Tacrolimus 155-165 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 76-101 28356851-1 2015 BACKGROUND: The primary goal of this study was to evaluate the influence of cytochrome P450 (CYP) 3A5 (6986A>G) and ABCB1 (3435C>T) polymorphisms on tacrolimus (TAC) dosage regimen and exposure. Tacrolimus 155-165 ATP binding cassette subfamily B member 1 Homo sapiens 119-124 25565672-1 2015 BACKGROUND: Blood tacrolimus (TAC) concentration delivered via intravenous administration is known to be influenced by genetic polymorphism of CYP3A5 and interaction with triazole antifungal agents. Tacrolimus 18-28 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 143-149 26184414-0 2015 Effect of CYP3A5 and ABCB1 polymorphisms on the interaction between tacrolimus and itraconazole in patients with connective tissue disease. Tacrolimus 68-78 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 26352670-0 2015 Angiopoietin-Like-4, a Potential Target of Tacrolimus, Predicts Earlier Podocyte Injury in Minimal Change Disease. Tacrolimus 43-53 angiopoietin like 4 Homo sapiens 0-19 26352670-9 2015 Moreover, we found that tacrolimus treatment significantly promoted podocyte repair and reduced glomerular and urinary Angptl4 expression at an earlier stage with a significant serum Angptl4 upregulation. Tacrolimus 24-34 angiopoietin like 4 Homo sapiens 119-126 26352670-9 2015 Moreover, we found that tacrolimus treatment significantly promoted podocyte repair and reduced glomerular and urinary Angptl4 expression at an earlier stage with a significant serum Angptl4 upregulation. Tacrolimus 24-34 angiopoietin like 4 Homo sapiens 183-190 26160968-2 2015 Because sirolimus (SIR) and calcineurin inhibitor-either cyclosporine (CsA) or tacrolimus-have become more common as graft-versus-host disease (GVHD) prophylaxis, we are witnessing a higher frequency of this complication. Tacrolimus 79-89 calcineurin binding protein 1 Homo sapiens 28-49 25781547-0 2015 ABCB1 (MDR-1) pharmacogenetics of tacrolimus in renal transplanted patients: a Next Generation Sequencing approach. Tacrolimus 34-44 ATP binding cassette subfamily B member 1 Homo sapiens 0-5 25781547-0 2015 ABCB1 (MDR-1) pharmacogenetics of tacrolimus in renal transplanted patients: a Next Generation Sequencing approach. Tacrolimus 34-44 ATP binding cassette subfamily B member 1 Homo sapiens 7-12 25781547-1 2015 BACKGROUND: A CYP3A5 gene polymorphism is the main determinant of Tacrolimus (Tac) dose requirements among renal transplanted patients. Tacrolimus 66-76 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 26518936-0 2015 Liver Transplant Patient Carriers of Polymorphism Cyp3a5*1 Donors May Need More Doses of Tacrolimus From the First Month After Transplantation. Tacrolimus 89-99 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 50-56 26518936-1 2015 BACKGROUND: The aim of this work was to evaluate the CYP3A5:CYP3A5*1/CYP3A5*3 (6986A>G) polymorphism related to the pharmacokinetic characteristics of tacrolimus during the first 3 months after transplantation, analyzing both donor and recipient genotype, in liver transplant patients. Tacrolimus 154-164 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 53-59 26518936-1 2015 BACKGROUND: The aim of this work was to evaluate the CYP3A5:CYP3A5*1/CYP3A5*3 (6986A>G) polymorphism related to the pharmacokinetic characteristics of tacrolimus during the first 3 months after transplantation, analyzing both donor and recipient genotype, in liver transplant patients. Tacrolimus 154-164 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 60-66 26518936-1 2015 BACKGROUND: The aim of this work was to evaluate the CYP3A5:CYP3A5*1/CYP3A5*3 (6986A>G) polymorphism related to the pharmacokinetic characteristics of tacrolimus during the first 3 months after transplantation, analyzing both donor and recipient genotype, in liver transplant patients. Tacrolimus 154-164 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 60-66 26518936-8 2015 CONCLUSIONS: The presence of the CYP3A5-6986A>G-polymorphism in the donor affects tacrolimus pharmacokinetics in the recipient, although the difference was statistically significant only for the first month after transplantation. Tacrolimus 85-95 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 26518936-9 2015 This means that in liver transplant patients receiving grafts from donors carrying the CYP3A5*1-polymorphism, a larger dose of tacrolimus from the first month after transplantation would be needed. Tacrolimus 127-137 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 87-93 26518946-1 2015 BACKGROUND: Tacrolimus pharmacokinetics prediction by CYP3A5 genotyping is not available in many Asian resource-limited settings. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 54-60 26518946-12 2015 The CYP3A5 genotypes were significantly correlated with the target tacrolimus dose at day 7 (r(2) = 0.307) and the stable dose at months 1 to 3 (r(2) = 0.337). Tacrolimus 67-77 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 26518962-7 2015 The expression of interleukin (IL)-2, tumor necrosis factor-alpha, and interferon-gamma in the FK506+TP8L2 group was significantly lower and the expression of IL-4 and IL-10 was significantly higher than those in other groups; the differences in cytokine levels were significant (P < .05). Tacrolimus 95-100 interferon gamma Rattus norvegicus 71-87 26260319-7 2015 Inhibition of NFAT transcriptional activity by FK506 attenuated calbindin-D28k expression. Tacrolimus 47-52 calbindin 1 Mus musculus 64-78 26366742-3 2015 FK506-binding proteins (FKBPs) form a complex with calcineurin in the presence of FK506 (FKBP12-FK506) and inhibit calcineurin activity. Tacrolimus 0-5 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 89-95 26366742-3 2015 FK506-binding proteins (FKBPs) form a complex with calcineurin in the presence of FK506 (FKBP12-FK506) and inhibit calcineurin activity. Tacrolimus 82-87 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 89-95 26366742-6 2015 Deletional analysis of the four genes revealed that the Deltafkbp12-1 strain was resistant to FK506, indicating FKBP12-1 as the key mediator of FK506-binding to calcineurin. Tacrolimus 144-149 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 112-118 26366742-7 2015 The endogenously expressed FKBP12-1-EGFP fusion protein localized to the cytoplasm and nuclei under normal growth conditions but also to the hyphal septa following FK506 treatment, revealing its interaction with calcineurin. Tacrolimus 164-169 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 27-33 26366742-11 2015 Together, these results indicate that while only FKBP12-1 is the bona fide binding partner of FK506, leading to the inhibition of calcineurin in A. fumigatus, FKBP12-4 may play a role in basal growth and the caspofungin-mediated paradoxical growth response. Tacrolimus 94-99 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 49-55 26366742-12 2015 Exploitation of differences between A. fumigatus FKBP12-1 and human FKBP12 will be critical for the generation of fungal-specific FK506 analogs to inhibit fungal calcineurin and treat invasive fungal disease. Tacrolimus 130-135 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 49-55 26366742-12 2015 Exploitation of differences between A. fumigatus FKBP12-1 and human FKBP12 will be critical for the generation of fungal-specific FK506 analogs to inhibit fungal calcineurin and treat invasive fungal disease. Tacrolimus 130-135 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 68-74 26114223-0 2015 Progressive decline in tacrolimus clearance after renal transplantation is partially explained by decreasing CYP3A4 activity and increasing haematocrit. Tacrolimus 23-33 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 109-115 26114223-4 2015 RESULTS: Tacrolimus clearance decreased gradually throughout the entire first year but only in CYP3A5*3/*3 homozygous recipients (25.6 +- 11.1 l h(-1) at day 7; 17 +- 9.1 l h(-1) at month 12; P < 0.001). Tacrolimus 9-19 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 95-101 26114223-5 2015 In mixed model analysis, decreasing CYP3A4 activity, measured by apparent oral MDZ clearance (924 +- 443 ml min(-1) at day 7 vs. 730 +- 344 ml min(-1) at month 1; P < 0.001), explained 55.4% of the decline in tacrolimus clearance in the first month. Tacrolimus 212-222 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 36-42 26114223-9 2015 CONCLUSIONS: The maturation of tacrolimus disposition in the first year after renal transplantation observed in CYP3A5*3/*3 homozygous patients can partly be explained by a (steroid tapering-related) decline in CYP3A4 activity and a progressive increase in haematocrit. Tacrolimus 31-41 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 112-118 26114223-9 2015 CONCLUSIONS: The maturation of tacrolimus disposition in the first year after renal transplantation observed in CYP3A5*3/*3 homozygous patients can partly be explained by a (steroid tapering-related) decline in CYP3A4 activity and a progressive increase in haematocrit. Tacrolimus 31-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 211-217 26184414-0 2015 Effect of CYP3A5 and ABCB1 polymorphisms on the interaction between tacrolimus and itraconazole in patients with connective tissue disease. Tacrolimus 68-78 ATP binding cassette subfamily B member 1 Homo sapiens 21-26 26184414-7 2015 CONCLUSIONS: In CYP3A5*3/*3 patients, because the metabolic pathway for tacrolimus occurs only through CYP3A4, the combination with ITCZ seems to lead to a higher risk of acute renal dysfunction. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 16-22 26184414-7 2015 CONCLUSIONS: In CYP3A5*3/*3 patients, because the metabolic pathway for tacrolimus occurs only through CYP3A4, the combination with ITCZ seems to lead to a higher risk of acute renal dysfunction. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 103-109 26184414-8 2015 Therefore, we suggest that the target blood tacrolimus concentration be set as low as possible through dose-adjustment for patients with the CYP3A5*3/*3 allele. Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 141-147 26049029-5 2015 Pretreatment with the protease inhibitor, leupeptin, PAR2 antibody, and tacrolimus significantly reduced the number of degranulated mast cells and tryptase content, and consequently alleviated scratching behavior. Tacrolimus 72-82 tryptase alpha/beta 1 Mus musculus 147-155 26104917-9 2015 The CaN inhibitor FK506 reduced these IL-6 neuroprotective properties. Tacrolimus 18-23 interleukin 6 Homo sapiens 38-42 26622455-0 2015 Investigation of CYP 3A5 and ABCB1 gene polymorphisms in the long-term following renal transplantation: Effects on tacrolimus exposure and kidney function. Tacrolimus 115-125 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 17-24 26622455-0 2015 Investigation of CYP 3A5 and ABCB1 gene polymorphisms in the long-term following renal transplantation: Effects on tacrolimus exposure and kidney function. Tacrolimus 115-125 ATP binding cassette subfamily B member 1 Homo sapiens 29-34 26253719-2 2015 Mip exhibits peptidyl prolyl cis-trans isomerase (PPIase) activity, which can be inhibited by rapamycin and FK506. Tacrolimus 108-113 major intrinsic protein of lens fiber Homo sapiens 0-3 26227094-6 2015 Subgroup analysis showed that POR*28 polymorphisms significantly decreased tacrolimus C0/D by 1.50 - 1.84-fold (p < 0.05) in patients who were CYP3A5 expressers (CYP3A5*1 carriers, n = 46), while similar results could not be obtained from CYP3A5 non-expressers (CYP3A5*3/*3 carriers, n = 37). Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 165-171 26227094-7 2015 Additionally, population pharmacokinetic analysis identified that the combined genotype of CYP3A5-POR was the only covariant for the apparent clearance of tacrolimus (CL/F). Tacrolimus 155-165 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 91-97 26227094-7 2015 Additionally, population pharmacokinetic analysis identified that the combined genotype of CYP3A5-POR was the only covariant for the apparent clearance of tacrolimus (CL/F). Tacrolimus 155-165 cytochrome p450 oxidoreductase Homo sapiens 98-101 26227094-8 2015 CONCLUSIONS: The study demonstrated that the POR*28 C>T mutation could decrease the C0/D of tacrolimus in renal recipients who were CYP3A5 expressers. Tacrolimus 95-105 cytochrome p450 oxidoreductase Homo sapiens 45-48 26227094-8 2015 CONCLUSIONS: The study demonstrated that the POR*28 C>T mutation could decrease the C0/D of tacrolimus in renal recipients who were CYP3A5 expressers. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 135-141 26227094-9 2015 The population pharmacokinetic model showed that the combined genotype of CYP3A5-POR was associated with the CL/F of tacrolimus which might provide references for personalized use of tacrolimus in clinic. Tacrolimus 117-127 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 74-80 26227094-9 2015 The population pharmacokinetic model showed that the combined genotype of CYP3A5-POR was associated with the CL/F of tacrolimus which might provide references for personalized use of tacrolimus in clinic. Tacrolimus 117-127 cytochrome p450 oxidoreductase Homo sapiens 81-84 26227094-9 2015 The population pharmacokinetic model showed that the combined genotype of CYP3A5-POR was associated with the CL/F of tacrolimus which might provide references for personalized use of tacrolimus in clinic. Tacrolimus 183-193 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 74-80 26227094-9 2015 The population pharmacokinetic model showed that the combined genotype of CYP3A5-POR was associated with the CL/F of tacrolimus which might provide references for personalized use of tacrolimus in clinic. Tacrolimus 183-193 cytochrome p450 oxidoreductase Homo sapiens 81-84 26227094-0 2015 The genetic polymorphisms of POR*28 and CYP3A5*3 significantly influence the pharmacokinetics of tacrolimus in Chinese renal transplant recipients. Tacrolimus 97-107 cytochrome p450 oxidoreductase Homo sapiens 29-32 26227094-0 2015 The genetic polymorphisms of POR*28 and CYP3A5*3 significantly influence the pharmacokinetics of tacrolimus in Chinese renal transplant recipients. Tacrolimus 97-107 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 40-46 26227094-5 2015 RESULTS: The significant influences of CYP3A5*3, CYP3A4*1G, and POR*28 polymorphisms on tacrolimus dose-adjusted trough concentrations (C0/D) were observed in 83 renal recipients. Tacrolimus 88-98 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 39-45 26227094-5 2015 RESULTS: The significant influences of CYP3A5*3, CYP3A4*1G, and POR*28 polymorphisms on tacrolimus dose-adjusted trough concentrations (C0/D) were observed in 83 renal recipients. Tacrolimus 88-98 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 49-55 26227094-5 2015 RESULTS: The significant influences of CYP3A5*3, CYP3A4*1G, and POR*28 polymorphisms on tacrolimus dose-adjusted trough concentrations (C0/D) were observed in 83 renal recipients. Tacrolimus 88-98 cytochrome p450 oxidoreductase Homo sapiens 64-67 26227094-6 2015 Subgroup analysis showed that POR*28 polymorphisms significantly decreased tacrolimus C0/D by 1.50 - 1.84-fold (p < 0.05) in patients who were CYP3A5 expressers (CYP3A5*1 carriers, n = 46), while similar results could not be obtained from CYP3A5 non-expressers (CYP3A5*3/*3 carriers, n = 37). Tacrolimus 75-85 cytochrome p450 oxidoreductase Homo sapiens 30-33 26227094-6 2015 Subgroup analysis showed that POR*28 polymorphisms significantly decreased tacrolimus C0/D by 1.50 - 1.84-fold (p < 0.05) in patients who were CYP3A5 expressers (CYP3A5*1 carriers, n = 46), while similar results could not be obtained from CYP3A5 non-expressers (CYP3A5*3/*3 carriers, n = 37). Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 146-152 26227094-6 2015 Subgroup analysis showed that POR*28 polymorphisms significantly decreased tacrolimus C0/D by 1.50 - 1.84-fold (p < 0.05) in patients who were CYP3A5 expressers (CYP3A5*1 carriers, n = 46), while similar results could not be obtained from CYP3A5 non-expressers (CYP3A5*3/*3 carriers, n = 37). Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 165-171 26227094-6 2015 Subgroup analysis showed that POR*28 polymorphisms significantly decreased tacrolimus C0/D by 1.50 - 1.84-fold (p < 0.05) in patients who were CYP3A5 expressers (CYP3A5*1 carriers, n = 46), while similar results could not be obtained from CYP3A5 non-expressers (CYP3A5*3/*3 carriers, n = 37). Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 165-171 25862606-9 2015 However, in the presence of tacrolimus, P-p65(s529) expression was inhibited without affecting nuclear localization of total p65. Tacrolimus 28-38 RELA proto-oncogene, NF-kB subunit Homo sapiens 42-45 25905982-5 2015 RESULTS: Tacrolimus significantly and SRL modestly inhibited interferon (IFN)-gamma (Th1) and IL-17 (Th17)-producing cells. Tacrolimus 9-19 interferon gamma Homo sapiens 61-83 25905982-5 2015 RESULTS: Tacrolimus significantly and SRL modestly inhibited interferon (IFN)-gamma (Th1) and IL-17 (Th17)-producing cells. Tacrolimus 9-19 interleukin 17A Homo sapiens 94-99 26334901-6 2015 When compared with the wild-type genotype, the variant genotypes rs5767743 and rs5767700 correlated with significantly increased PPARalpha and CYP3A4 mRNA expression and lower tacrolimus trough concentration/dose ratios (P < 0.05 for all).Donor PPARalpha gene polymorphisms influence the susceptibility to metabolic disorders following LT and may also be associated with a fasten tacrolimus metabolism because of elevated CYP3A4 expression. Tacrolimus 176-186 peroxisome proliferator activated receptor alpha Homo sapiens 129-138 26334901-6 2015 When compared with the wild-type genotype, the variant genotypes rs5767743 and rs5767700 correlated with significantly increased PPARalpha and CYP3A4 mRNA expression and lower tacrolimus trough concentration/dose ratios (P < 0.05 for all).Donor PPARalpha gene polymorphisms influence the susceptibility to metabolic disorders following LT and may also be associated with a fasten tacrolimus metabolism because of elevated CYP3A4 expression. Tacrolimus 176-186 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 143-149 26334901-6 2015 When compared with the wild-type genotype, the variant genotypes rs5767743 and rs5767700 correlated with significantly increased PPARalpha and CYP3A4 mRNA expression and lower tacrolimus trough concentration/dose ratios (P < 0.05 for all).Donor PPARalpha gene polymorphisms influence the susceptibility to metabolic disorders following LT and may also be associated with a fasten tacrolimus metabolism because of elevated CYP3A4 expression. Tacrolimus 176-186 peroxisome proliferator activated receptor alpha Homo sapiens 248-257 26334901-6 2015 When compared with the wild-type genotype, the variant genotypes rs5767743 and rs5767700 correlated with significantly increased PPARalpha and CYP3A4 mRNA expression and lower tacrolimus trough concentration/dose ratios (P < 0.05 for all).Donor PPARalpha gene polymorphisms influence the susceptibility to metabolic disorders following LT and may also be associated with a fasten tacrolimus metabolism because of elevated CYP3A4 expression. Tacrolimus 176-186 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 425-431 26334901-6 2015 When compared with the wild-type genotype, the variant genotypes rs5767743 and rs5767700 correlated with significantly increased PPARalpha and CYP3A4 mRNA expression and lower tacrolimus trough concentration/dose ratios (P < 0.05 for all).Donor PPARalpha gene polymorphisms influence the susceptibility to metabolic disorders following LT and may also be associated with a fasten tacrolimus metabolism because of elevated CYP3A4 expression. Tacrolimus 383-393 peroxisome proliferator activated receptor alpha Homo sapiens 129-138 26334901-6 2015 When compared with the wild-type genotype, the variant genotypes rs5767743 and rs5767700 correlated with significantly increased PPARalpha and CYP3A4 mRNA expression and lower tacrolimus trough concentration/dose ratios (P < 0.05 for all).Donor PPARalpha gene polymorphisms influence the susceptibility to metabolic disorders following LT and may also be associated with a fasten tacrolimus metabolism because of elevated CYP3A4 expression. Tacrolimus 383-393 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 143-149 26334901-6 2015 When compared with the wild-type genotype, the variant genotypes rs5767743 and rs5767700 correlated with significantly increased PPARalpha and CYP3A4 mRNA expression and lower tacrolimus trough concentration/dose ratios (P < 0.05 for all).Donor PPARalpha gene polymorphisms influence the susceptibility to metabolic disorders following LT and may also be associated with a fasten tacrolimus metabolism because of elevated CYP3A4 expression. Tacrolimus 383-393 peroxisome proliferator activated receptor alpha Homo sapiens 248-257 26334901-6 2015 When compared with the wild-type genotype, the variant genotypes rs5767743 and rs5767700 correlated with significantly increased PPARalpha and CYP3A4 mRNA expression and lower tacrolimus trough concentration/dose ratios (P < 0.05 for all).Donor PPARalpha gene polymorphisms influence the susceptibility to metabolic disorders following LT and may also be associated with a fasten tacrolimus metabolism because of elevated CYP3A4 expression. Tacrolimus 383-393 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 425-431 26370456-0 2015 The effects of tacrolimus on the activity and expression of tissue factor in the rat ovary with ischemia-reperfusion induced injury. Tacrolimus 15-25 coagulation factor III, tissue factor Rattus norvegicus 60-73 26370456-1 2015 In the present study, the effects of tacrolimus on the activity and expression of tissue factor (TF) were investigated in the ovarian ischemia-reperfusion induced injury in rats. Tacrolimus 37-47 coagulation factor III, tissue factor Rattus norvegicus 82-95 26370456-1 2015 In the present study, the effects of tacrolimus on the activity and expression of tissue factor (TF) were investigated in the ovarian ischemia-reperfusion induced injury in rats. Tacrolimus 37-47 coagulation factor III, tissue factor Rattus norvegicus 97-99 26370456-6 2015 Moreover, tacrolimus decreased TF activity in the TBI group compared with the IR group. Tacrolimus 10-20 coagulation factor III, tissue factor Rattus norvegicus 31-33 26307985-3 2015 We evaluated SNPs in genes that have previously shown correlations in other kinds of solid organ transplantation, namely ABCB1 and CYP3A5 genes with tacrolimus (Tac) and ABCC2, UGT1A9 and SLCO1B1 genes with mycophenolic acid (MPA), during the first six months after lung transplantation (51 patients). Tacrolimus 149-159 ATP binding cassette subfamily B member 1 Homo sapiens 121-126 26307985-3 2015 We evaluated SNPs in genes that have previously shown correlations in other kinds of solid organ transplantation, namely ABCB1 and CYP3A5 genes with tacrolimus (Tac) and ABCC2, UGT1A9 and SLCO1B1 genes with mycophenolic acid (MPA), during the first six months after lung transplantation (51 patients). Tacrolimus 149-159 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 131-137 25888602-0 2015 The FK506-binding protein FKBP52 in vitro induces aggregation of truncated Tau forms with prion-like behavior. Tacrolimus 4-9 FKBP prolyl isomerase 4 Homo sapiens 26-32 25888602-0 2015 The FK506-binding protein FKBP52 in vitro induces aggregation of truncated Tau forms with prion-like behavior. Tacrolimus 4-9 microtubule associated protein tau Homo sapiens 75-78 25888602-3 2015 We showed previously that the immunophilin FK506-binding protein of MW ~52 kDa (FKBP52) interferes with this function of full-length Tau and provokes aggregation of a disease-related mutant of Tau. Tacrolimus 43-48 FKBP prolyl isomerase 4 Homo sapiens 80-86 25888602-3 2015 We showed previously that the immunophilin FK506-binding protein of MW ~52 kDa (FKBP52) interferes with this function of full-length Tau and provokes aggregation of a disease-related mutant of Tau. Tacrolimus 43-48 microtubule associated protein tau Homo sapiens 133-136 25888602-3 2015 We showed previously that the immunophilin FK506-binding protein of MW ~52 kDa (FKBP52) interferes with this function of full-length Tau and provokes aggregation of a disease-related mutant of Tau. Tacrolimus 43-48 microtubule associated protein tau Homo sapiens 193-196 25862606-10 2015 The data demonstrate the application of IFC to simultaneously assess phosphorylation of p65 and its cellular localization and the results obtained by this analysis corroborate current insights regarding the specific effect of tacrolimus on serine 529 phosphorylation. Tacrolimus 226-236 RELA proto-oncogene, NF-kB subunit Homo sapiens 88-91 26092277-5 2015 We used fluorescent CaM, FKBP12.6, and domain-peptide biosensor (F-DPc10) to measure, directly in cardiac myocytes, (1) RyR2 activation by hydrogen peroxide (H2O2)-induced oxidation, (2) RyR2 conformation change caused by oxidation, (3) CaM-RyR2 and FK506-binding protein (FKBP12.6)-RyR2 interaction upon oxidation, and (4) whether dantrolene affects 1-3. Tacrolimus 250-255 ryanodine receptor 2 Homo sapiens 120-124 25936769-6 2015 The results showed that expression levels of HO-1 were significantly upregulated in response to treatment with CsA, FK506, RAPA and MMF, whereas the expression levels of IL-7 were markedly downregulated by treatment with the above immunosuppressants. Tacrolimus 116-121 heme oxygenase 1 Rattus norvegicus 45-49 25936769-7 2015 CsA, FK506 and MMF significantly enhanced the expression levels of IDO, whereas RAPA exhibited no apparent effect on IDO. Tacrolimus 5-10 indoleamine 2,3-dioxygenase 1 Rattus norvegicus 67-70 26073814-4 2015 The AFM biosensor presented herein utilises the endogen drug receptor, FKBP12, to quantify Tacrolimus levels. Tacrolimus 91-101 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 71-77 25769064-10 2015 Treatment with FK506 produced a synergistic effect in the grafts from AID(-/-) recipients with further reduction of intimal hyperplasia and fibrosis scores. Tacrolimus 15-20 activation-induced cytidine deaminase Mus musculus 70-73 25707487-7 2015 Tacrolimus trough levels were readily achieved posttransplant; initially lower in Arm 2 versus 1 with delayed initiation in Arm 3. Tacrolimus 0-10 Jupiter microtubule associated homolog 1 Homo sapiens 82-87 26024817-1 2015 In order to improve oral bioavailability of tacrolimus (FK506), a novel poly(methyl vinyl ether-co-maleic anhydride)-graft-hydroxypropyl-beta-cyclodextrin amphiphilic copolymer (CD-PVM/MA) is developed, combining the bioadhesiveness of PVM/MA, P-glycoprotein (P-gp), and cytochrome P450-inhibitory effect of CD into one. Tacrolimus 44-54 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 244-258 26024817-1 2015 In order to improve oral bioavailability of tacrolimus (FK506), a novel poly(methyl vinyl ether-co-maleic anhydride)-graft-hydroxypropyl-beta-cyclodextrin amphiphilic copolymer (CD-PVM/MA) is developed, combining the bioadhesiveness of PVM/MA, P-glycoprotein (P-gp), and cytochrome P450-inhibitory effect of CD into one. Tacrolimus 44-54 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 260-264 26024817-1 2015 In order to improve oral bioavailability of tacrolimus (FK506), a novel poly(methyl vinyl ether-co-maleic anhydride)-graft-hydroxypropyl-beta-cyclodextrin amphiphilic copolymer (CD-PVM/MA) is developed, combining the bioadhesiveness of PVM/MA, P-glycoprotein (P-gp), and cytochrome P450-inhibitory effect of CD into one. Tacrolimus 56-61 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 244-258 26024817-1 2015 In order to improve oral bioavailability of tacrolimus (FK506), a novel poly(methyl vinyl ether-co-maleic anhydride)-graft-hydroxypropyl-beta-cyclodextrin amphiphilic copolymer (CD-PVM/MA) is developed, combining the bioadhesiveness of PVM/MA, P-glycoprotein (P-gp), and cytochrome P450-inhibitory effect of CD into one. Tacrolimus 56-61 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 260-264 26024817-6 2015 The permeability of FK506 was improved dramatically by CD-PVM/MA-NPs compared to its solution, probably due to the synergistic inhibition effect of P-gp and cytochrome P450 3A (CYP3A). Tacrolimus 20-25 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 148-152 26024817-6 2015 The permeability of FK506 was improved dramatically by CD-PVM/MA-NPs compared to its solution, probably due to the synergistic inhibition effect of P-gp and cytochrome P450 3A (CYP3A). Tacrolimus 20-25 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 157-175 26024817-6 2015 The permeability of FK506 was improved dramatically by CD-PVM/MA-NPs compared to its solution, probably due to the synergistic inhibition effect of P-gp and cytochrome P450 3A (CYP3A). Tacrolimus 20-25 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 177-182 26024817-11 2015 The present study suggested that the novel multifunctional CD-PVM/MA is a promising efficient oral delivery carrier for FK506, due to its ability in solubilization, inhibitory effects on both P-gp and CYP 3A, high bioadhesion, and sustained release capability. Tacrolimus 120-125 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 192-196 26024817-11 2015 The present study suggested that the novel multifunctional CD-PVM/MA is a promising efficient oral delivery carrier for FK506, due to its ability in solubilization, inhibitory effects on both P-gp and CYP 3A, high bioadhesion, and sustained release capability. Tacrolimus 120-125 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 201-207 25979971-2 2015 However, use of IL2-RA remains controversial in LRT with tacrolimus (TAC)/mycophenolic acid (MPA) with or without steroids. Tacrolimus 57-67 interleukin 2 receptor subunit alpha Homo sapiens 16-22 25600613-6 2015 The relationship of the IC50 values of 11 inhibitors between tacrolimus and typical CYP3A substrates (midazolam and testosterone) was also analysed. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 84-89 25600613-7 2015 A strong correlation was found between the IC50 values of tacrolimus and typical CYP3A substrates (r(2) >= 0.85). Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-86 25600613-14 2015 within an approximately 2-fold range of observed values) the effect of CYP3A inhibitors on the tacrolimus AUCp.o. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 71-76 26361868-10 2015 Inhibition of the calcineurin/NFATc1 pathway by cyclosporin A and FK506, attenuated Tbeta4-induced osteoblastic differentiation and activation of Wnt-related genes, as well as nuclear beta-catenin in hPDLCs. Tacrolimus 66-71 nuclear factor of activated T cells 1 Homo sapiens 30-36 26361868-10 2015 Inhibition of the calcineurin/NFATc1 pathway by cyclosporin A and FK506, attenuated Tbeta4-induced osteoblastic differentiation and activation of Wnt-related genes, as well as nuclear beta-catenin in hPDLCs. Tacrolimus 66-71 thymosin beta 4 X-linked Homo sapiens 84-90 25881619-1 2015 OBJECTIVES: Emerging data suggest that the combination of tacrolimus and the CCR5 antagonist maraviroc, both cytochrome P450-3A4 substrates, may be effective in preventing graft-versus-host disease in patients undergoing allogeneic HSCT. Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 109-128 26293093-0 2015 Antiproteinuric Effects of Green Tea Extract on Tacrolimus-Induced Nephrotoxicity in Mice. Tacrolimus 48-58 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 Mus musculus 33-36 26125566-4 2015 The aim of this study was to investigate the role of NGAL as an early and accurate indicator of DGF and Tacrolimus (Tac) toxicity and as a mediator of tissue regeneration in KT from ECD. Tacrolimus 104-114 lipocalin 2 Homo sapiens 53-57 26125566-4 2015 The aim of this study was to investigate the role of NGAL as an early and accurate indicator of DGF and Tacrolimus (Tac) toxicity and as a mediator of tissue regeneration in KT from ECD. Tacrolimus 104-107 lipocalin 2 Homo sapiens 53-57 26087255-1 2015 The direct effect of immunosuppressive drugs calcineurin inhibitor (Tacrolimus, TAC) and mTOR inhibitor (Sirolimus, SRL) on B cell activation, differentiation and proliferation is not well documented. Tacrolimus 68-78 calcineurin binding protein 1 Homo sapiens 45-66 25801146-2 2015 Individuals who express CYP3A5 (extensive and intermediate metabolizers) generally have decreased dose-adjusted trough concentrations of tacrolimus as compared with those who are CYP3A5 nonexpressers (poor metabolizers), possibly delaying achievement of target blood concentrations. Tacrolimus 137-147 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-30 25801146-3 2015 We summarize evidence from the published literature supporting this association and provide dosing recommendations for tacrolimus based on CYP3A5 genotype when known (updates at www.pharmgkb.org). Tacrolimus 119-129 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 139-145 25840504-10 2015 African American recipients demonstrated a unique decrease in expression of the FLT3 gene in response to higher tacrolimus levels. Tacrolimus 112-122 fms related receptor tyrosine kinase 3 Homo sapiens 80-84 26031588-0 2015 Tacrolimus reduction with everolimus addition for calcineurin inhibitor-induced arteriolopathy in kidney allografts. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 50-71 26031588-1 2015 AIM: The aim of this study was to evaluate the effect of tacrolimus (TAC) reduction with everolimus (EVR) addition on the maintenance immunosuppression for the recipients with calcineurin inhibitor arteriolopathy (CNIA). Tacrolimus 57-67 calcineurin binding protein 1 Homo sapiens 176-197 26067485-1 2015 AIM: Determine the effect of the genetic variants beyond CYP3A5*3 on tacrolimus disposition. Tacrolimus 69-79 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 57-63 26067485-2 2015 PATIENTS & METHODS: We studied genetic correlates of tacrolimus trough concentrations with POR*28, CYP3A4*22 and ABCC2 haplotypes in a large, ethnically diverse kidney transplant cohort (n = 2008). Tacrolimus 57-67 cytochrome p450 oxidoreductase Homo sapiens 95-98 26067485-2 2015 PATIENTS & METHODS: We studied genetic correlates of tacrolimus trough concentrations with POR*28, CYP3A4*22 and ABCC2 haplotypes in a large, ethnically diverse kidney transplant cohort (n = 2008). Tacrolimus 57-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 103-109 26067485-2 2015 PATIENTS & METHODS: We studied genetic correlates of tacrolimus trough concentrations with POR*28, CYP3A4*22 and ABCC2 haplotypes in a large, ethnically diverse kidney transplant cohort (n = 2008). Tacrolimus 57-67 ATP binding cassette subfamily C member 2 Homo sapiens 117-122 26067485-3 2015 RESULTS: Subjects carrying one or more CYP3A5*1 alleles had lower tacrolimus trough concentrations (p = 9.2 x 10(-75)). Tacrolimus 66-76 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 39-45 26067485-4 2015 The presence of one or two POR*28 alleles was associated with a 4.63% reduction in tacrolimus trough concentrations after adjusting for CYP3A5*1 and clinical factors (p = 0.037). Tacrolimus 83-93 cytochrome p450 oxidoreductase Homo sapiens 27-30 26067485-4 2015 The presence of one or two POR*28 alleles was associated with a 4.63% reduction in tacrolimus trough concentrations after adjusting for CYP3A5*1 and clinical factors (p = 0.037). Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 136-142 26067485-7 2015 CONCLUSION: This study confirmed that CYP3A5*1 was associated with lower tacrolimus trough concentrations. Tacrolimus 73-83 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 38-44 26067485-8 2015 POR*28 was associated with decreased tacrolimus trough concentrations although the effect was small possibly through enhanced CYP3A4 enzyme activity. Tacrolimus 37-47 cytochrome p450 oxidoreductase Homo sapiens 0-3 25953903-1 2015 Interchanging Leu-119 for Pro-119 at the tip of the beta4-beta5 loop in the first FK506 binding domain (FK1) of the FKBP51 and FKBP52 proteins, respectively, has been reported to largely reverse the inhibitory (FKBP51) or stimulatory (FKBP52) effects of these co-chaperones on the transcriptional activity of glucocorticoid and androgen receptor-protein complexes. Tacrolimus 82-87 FKBP prolyl isomerase 4 Homo sapiens 127-133 25953903-1 2015 Interchanging Leu-119 for Pro-119 at the tip of the beta4-beta5 loop in the first FK506 binding domain (FK1) of the FKBP51 and FKBP52 proteins, respectively, has been reported to largely reverse the inhibitory (FKBP51) or stimulatory (FKBP52) effects of these co-chaperones on the transcriptional activity of glucocorticoid and androgen receptor-protein complexes. Tacrolimus 82-87 FKBP prolyl isomerase 4 Homo sapiens 235-241 25491283-0 2015 The calcineurin inhibitor tacrolimus as a new therapy in severe cherubism. Tacrolimus 26-36 calcineurin binding protein 1 Homo sapiens 4-25 25943142-7 2015 CONCLUSION: Distal RTA and urine concentrating defect were highly prevalent after kidney transplantation both in the sirolimus and tacrolimus treated patients. Tacrolimus 131-141 MAS related GPR family member F Homo sapiens 19-22 25817300-12 2015 The tacrolimus blood concentrations were significantly higher in patients with the genotype GG of IL-2-330 (P = 0.012) at the third month. Tacrolimus 4-14 interleukin 2 Homo sapiens 98-102 25883698-6 2015 Calcineurin inhibitor, which includes cyclosporine, pimecrolimus and tacrolimus, impairs calcineurin-induced up-regulation of IL-2 expression, resulting in increased susceptibility to invasive fungal diseases. Tacrolimus 69-79 calcineurin binding protein 1 Homo sapiens 0-21 25883698-6 2015 Calcineurin inhibitor, which includes cyclosporine, pimecrolimus and tacrolimus, impairs calcineurin-induced up-regulation of IL-2 expression, resulting in increased susceptibility to invasive fungal diseases. Tacrolimus 69-79 interleukin 2 Homo sapiens 126-130 25758355-7 2015 Ang II-induced hypertrophy and OPN protein expression were regressed in the presence of an NHE1 inhibitor, EMD 87580, or a calcineurin inhibitor, FK506. Tacrolimus 146-151 angiogenin, ribonuclease, RNase A family, 5 Mus musculus 0-3 25758355-7 2015 Ang II-induced hypertrophy and OPN protein expression were regressed in the presence of an NHE1 inhibitor, EMD 87580, or a calcineurin inhibitor, FK506. Tacrolimus 146-151 secreted phosphoprotein 1 Mus musculus 31-34 25758355-9 2015 NHE1-induced activation of Gata4 was inhibited by FK506. Tacrolimus 50-55 solute carrier family 9 (sodium/hydrogen exchanger), member 1 Mus musculus 0-4 25758355-9 2015 NHE1-induced activation of Gata4 was inhibited by FK506. Tacrolimus 50-55 GATA binding protein 4 Mus musculus 27-32 25850753-1 2015 Tacrolimus is prescribed to prevent allograft rejection in pediatric liver transplant recipients; however, its metabolism through the cytochrome P-450 enzyme system presents a multitude of challenges in regard to drug interactions. Tacrolimus 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 25770929-4 2015 It was observed that topical administration of tacrolimus significantly accelerated the tumor promotion events in dimethylbenz(a)anthracene (DMBA)-initiated and 12-O-tetradecanoylphorbol-13-acetate (TPA) promoted two-stage mouse skin carcinogenesis, which were accompanied by reduced CD4(+)/CD8(+) ratio of lymph nodes and serum IL-2 level. Tacrolimus 47-57 CD4 antigen Mus musculus 284-287 25770929-4 2015 It was observed that topical administration of tacrolimus significantly accelerated the tumor promotion events in dimethylbenz(a)anthracene (DMBA)-initiated and 12-O-tetradecanoylphorbol-13-acetate (TPA) promoted two-stage mouse skin carcinogenesis, which were accompanied by reduced CD4(+)/CD8(+) ratio of lymph nodes and serum IL-2 level. Tacrolimus 47-57 interleukin 2 Mus musculus 329-333 26176181-0 2015 Influence of CYP3A5 genotypes on tacrolimus dose requirement: age and its pharmacological interaction with ABCB1 genetics in the Chinese paediatric liver transplantation. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 26177012-0 2015 Effect of CYP3A5 polymorphism on the pharmacokinetics of tacrolimus and acute rejection in renal transplant recipients: experience at a single centre. Tacrolimus 57-67 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 26177012-1 2015 AIM: As a substrate of cytochrome P450 (CYP) 3A5, tacrolimus is characterised by a narrow therapeutic index and large inter-individual variability. Tacrolimus 50-60 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 23-48 26177012-2 2015 Our objective was to determine the influence of CYP3A5 genetic polymorphisms on tacrolimus pharmacokinetics and acute rejection in Chinese renal transplant recipients. Tacrolimus 80-90 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 48-54 26177012-10 2015 CONCLUSION: CYP3A5 genetic polymorphism is associated with the concentration/dose ratio of tacrolimus in Chinese renal recipients. Tacrolimus 91-101 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 26177012-11 2015 Patients with CYP3A5*1*1 genotypes require higher doses of tacrolimus to achieve the target concentration and may be at risk of acute rejection soon after transplant because of inadequate immunosuppression. Tacrolimus 59-69 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 26039043-0 2015 Effects of the CYP3A4*1B Genetic Polymorphism on the Pharmacokinetics of Tacrolimus in Adult Renal Transplant Recipients: A Meta-Analysis. Tacrolimus 73-83 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 15-21 26039043-1 2015 BACKGROUND AND OBJECTIVE: The association between the CYP3A4*1B single nucleotide polymorphism (SNP) and tacrolimus pharmacokinetics in different studies is controversial. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 54-60 26039043-2 2015 Therefore, a meta-analysis was employed to evaluate the correlation between the CYP3A4*1B genetic polymorphism and tacrolimus pharmacokinetics at different post-transplantation times in adult renal transplant recipients. Tacrolimus 115-125 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 80-86 26039043-7 2015 In light of the heterogeneity, the analysis was repeated after removing the only study in an Indian population, and CYP3A4*1/*1 European recipients (mostly Caucasian) required a lower weight-adjusted tacrolimus daily dose within the first year post-transplantation. Tacrolimus 200-210 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-122 26039043-8 2015 The tacrolimus trough concentration/weight-adjusted tacrolimus daily dose ratio (C0/Dose ratio) was significantly higher in CYP3A4*1/*1 recipients than in CYP3A4*1B carriers at 6 months (WMD 52.588; 95% CI 22.387 ~ 82.789) and 12 months (WMD 62.219; 95% CI 14.218 ~ 110.221) post-transplantation. Tacrolimus 4-14 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 124-130 26039043-8 2015 The tacrolimus trough concentration/weight-adjusted tacrolimus daily dose ratio (C0/Dose ratio) was significantly higher in CYP3A4*1/*1 recipients than in CYP3A4*1B carriers at 6 months (WMD 52.588; 95% CI 22.387 ~ 82.789) and 12 months (WMD 62.219; 95% CI 14.218 ~ 110.221) post-transplantation. Tacrolimus 4-14 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 155-161 26039043-8 2015 The tacrolimus trough concentration/weight-adjusted tacrolimus daily dose ratio (C0/Dose ratio) was significantly higher in CYP3A4*1/*1 recipients than in CYP3A4*1B carriers at 6 months (WMD 52.588; 95% CI 22.387 ~ 82.789) and 12 months (WMD 62.219; 95% CI 14.218 ~ 110.221) post-transplantation. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 124-130 26039043-10 2015 CONCLUSION: Based on our meta-analysis, the CYP3A4*1B genetic polymorphism affects tacrolimus dose requirements and tacrolimus trough concentration/weight-adjusted tacrolimus daily dose ratio within the first year post-transplantation in adult renal transplant recipients, especially in European recipients (mostly Caucasian). Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 44-50 26039043-10 2015 CONCLUSION: Based on our meta-analysis, the CYP3A4*1B genetic polymorphism affects tacrolimus dose requirements and tacrolimus trough concentration/weight-adjusted tacrolimus daily dose ratio within the first year post-transplantation in adult renal transplant recipients, especially in European recipients (mostly Caucasian). Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 44-50 26039043-10 2015 CONCLUSION: Based on our meta-analysis, the CYP3A4*1B genetic polymorphism affects tacrolimus dose requirements and tacrolimus trough concentration/weight-adjusted tacrolimus daily dose ratio within the first year post-transplantation in adult renal transplant recipients, especially in European recipients (mostly Caucasian). Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 44-50 25650384-0 2015 Influence of tacrolimus on podocyte injury inducted by angiotensin II. Tacrolimus 13-23 angiotensinogen Homo sapiens 55-69 25650384-4 2015 RESULTS: The activities of podocytes significantly increased after FK506 intervention for 24 hours and 48 hours when compared with the Ang II stimulation group, and the apoptosis rate was markedly lower than that of the Ang II stimulation group, while in contrast to TRPC6 mRNA and protein expression. Tacrolimus 67-72 angiogenin Homo sapiens 135-138 25650384-4 2015 RESULTS: The activities of podocytes significantly increased after FK506 intervention for 24 hours and 48 hours when compared with the Ang II stimulation group, and the apoptosis rate was markedly lower than that of the Ang II stimulation group, while in contrast to TRPC6 mRNA and protein expression. Tacrolimus 67-72 angiogenin Homo sapiens 220-223 25650384-4 2015 RESULTS: The activities of podocytes significantly increased after FK506 intervention for 24 hours and 48 hours when compared with the Ang II stimulation group, and the apoptosis rate was markedly lower than that of the Ang II stimulation group, while in contrast to TRPC6 mRNA and protein expression. Tacrolimus 67-72 transient receptor potential cation channel subfamily C member 6 Homo sapiens 267-272 25650384-5 2015 CONCLUSION: FK506 can directly act on the podocytes to inhibit Ang II-induced damage on podocyte structures and reduce the apoptosis rate of podocytes, which may be related to stabilizing TRPC6 expression and distribution in podocytes by FK506, thus maintaining the structure and function integrity of the slit diaphragm and playing a role in protecting podocytes and antiproteinuria. Tacrolimus 12-17 angiogenin Homo sapiens 63-66 25650384-5 2015 CONCLUSION: FK506 can directly act on the podocytes to inhibit Ang II-induced damage on podocyte structures and reduce the apoptosis rate of podocytes, which may be related to stabilizing TRPC6 expression and distribution in podocytes by FK506, thus maintaining the structure and function integrity of the slit diaphragm and playing a role in protecting podocytes and antiproteinuria. Tacrolimus 12-17 transient receptor potential cation channel subfamily C member 6 Homo sapiens 188-193 25650384-5 2015 CONCLUSION: FK506 can directly act on the podocytes to inhibit Ang II-induced damage on podocyte structures and reduce the apoptosis rate of podocytes, which may be related to stabilizing TRPC6 expression and distribution in podocytes by FK506, thus maintaining the structure and function integrity of the slit diaphragm and playing a role in protecting podocytes and antiproteinuria. Tacrolimus 238-243 transient receptor potential cation channel subfamily C member 6 Homo sapiens 188-193 25647147-11 2015 Immunohistochemistry showed that, after tacrolimus treatment, the expressions of nuclear factor of activated T cell and interleukin 2 were downregulated in submucosal fibroblasts, neovascular cells, and glandular cells. Tacrolimus 40-50 interleukin 2 Rattus norvegicus 120-133 25647147-12 2015 CONCLUSIONS: This study suggests that low-dose systemic tacrolimus has a preventive effect on laryngotracheal stenosis by inhibiting the activation of immune cells in the injured airway mucosa via the calcineurin/nuclear factor of activated T cell/interleukin 2 pathway. Tacrolimus 56-66 interleukin 2 Rattus norvegicus 248-261 25650527-0 2015 Association between endothelial and platelet function markers and adiponectin in renal transplanted recipients on cyclosporine and tacrolimus immunosuppression based therapy. Tacrolimus 131-141 adiponectin, C1Q and collagen domain containing Homo sapiens 66-77 25271728-3 2015 Polymorphism of the cytochrome P450 3A5 (CYP3A5) gene contributes significantly to tacrolimus dose requirements. Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-39 25271728-3 2015 Polymorphism of the cytochrome P450 3A5 (CYP3A5) gene contributes significantly to tacrolimus dose requirements. Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 41-47 25271728-4 2015 Recently, CYP3A4*22 was reported to additionally affect tacrolimus pharmacokinetics (PK). Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 10-16 25964218-6 2015 CONCLUSION: Low serum TGF-beta and high IL-10 levels at post-transplantation month 6 might have a positive effect on graft survival in living donor kidney recipients on tacrolimus-based immunosuppressive treatment. Tacrolimus 169-179 transforming growth factor beta 1 Homo sapiens 22-30 25964218-6 2015 CONCLUSION: Low serum TGF-beta and high IL-10 levels at post-transplantation month 6 might have a positive effect on graft survival in living donor kidney recipients on tacrolimus-based immunosuppressive treatment. Tacrolimus 169-179 interleukin 10 Homo sapiens 40-45 25817604-0 2015 ABCB1 genetic variant and its associated tacrolimus pharmacokinetics affect renal function in patients with rheumatoid arthritis. Tacrolimus 41-51 ATP binding cassette subfamily B member 1 Homo sapiens 0-5 25817604-1 2015 BACKGROUND: This study aimed to evaluate the blood exposure of and clinical responses to tacrolimus based on genetic variants of CYP3A5 and ABCB1 in patients with rheumatoid arthritis. Tacrolimus 89-99 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 129-135 25817604-1 2015 BACKGROUND: This study aimed to evaluate the blood exposure of and clinical responses to tacrolimus based on genetic variants of CYP3A5 and ABCB1 in patients with rheumatoid arthritis. Tacrolimus 89-99 ATP binding cassette subfamily B member 1 Homo sapiens 140-145 25817604-5 2015 RESULTS: Dose-normalized blood concentration of tacrolimus was significantly higher in the CYP3A5*3/*3 group than in the *1 allele carrier group. Tacrolimus 48-58 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 91-97 25817604-6 2015 A lower metabolic ratio of 13-O-demethylate to tacrolimus was observed in the CYP3A5*3/*3 group. Tacrolimus 47-57 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 78-84 25817604-7 2015 The ABCB1 3435TT group had higher dose-normalized blood concentrations of tacrolimus and 13-O-demethylate. Tacrolimus 74-84 ATP binding cassette subfamily B member 1 Homo sapiens 4-9 25817604-10 2015 Patients lacking the CYP3A5*3 allele had a higher incidence of tacrolimus withdrawal. Tacrolimus 63-73 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 21-27 25817604-11 2015 CONCLUSION: CYP3A5*3 increased the blood exposure of tacrolimus through its metabolic reduction. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 25817604-12 2015 ABCB1 C3435T led to a higher blood exposure of tacrolimus and its major metabolite. Tacrolimus 47-57 ATP binding cassette subfamily B member 1 Homo sapiens 0-5 25817604-13 2015 The ABCB1 genetic variant and its associated tacrolimus pharmacokinetics affected renal function in rheumatoid arthritis patients. Tacrolimus 45-55 ATP binding cassette subfamily B member 1 Homo sapiens 4-9 26177348-0 2015 Comparison of tacrolimus and cyclosporin A in CYP3A5 expressing Chinese de novo kidney transplant recipients: a 2-year prospective study. Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 25752645-9 2015 Importantly, we showed that calcineurin-dependent NFAT3 nuclear translocation was essential to AngII-mediated IL-6 transcription, QL mimicked the effect of FK506, the calcineurin inhibitor, on suppression of IL-6 expression and stress fibres formation. Tacrolimus 156-161 angiotensinogen Rattus norvegicus 95-100 25752645-9 2015 Importantly, we showed that calcineurin-dependent NFAT3 nuclear translocation was essential to AngII-mediated IL-6 transcription, QL mimicked the effect of FK506, the calcineurin inhibitor, on suppression of IL-6 expression and stress fibres formation. Tacrolimus 156-161 interleukin 6 Rattus norvegicus 110-114 25752645-9 2015 Importantly, we showed that calcineurin-dependent NFAT3 nuclear translocation was essential to AngII-mediated IL-6 transcription, QL mimicked the effect of FK506, the calcineurin inhibitor, on suppression of IL-6 expression and stress fibres formation. Tacrolimus 156-161 calcineurin binding protein 1 Rattus norvegicus 167-188 25752645-9 2015 Importantly, we showed that calcineurin-dependent NFAT3 nuclear translocation was essential to AngII-mediated IL-6 transcription, QL mimicked the effect of FK506, the calcineurin inhibitor, on suppression of IL-6 expression and stress fibres formation. Tacrolimus 156-161 interleukin 6 Rattus norvegicus 208-212 25491283-8 2015 Immunohistologic analyses on granuloma showed that tacrolimus caused a significant reduction in the number of TRAP-positive osteoclasts and NFATc1 nuclear staining in multinucleated giant cells. Tacrolimus 51-61 nuclear factor of activated T cells 1 Homo sapiens 140-146 25491283-9 2015 Molecular analysis showed that tacrolimus treatment also resulted in increased OPG expression. Tacrolimus 31-41 basic transcription factor 3 pseudogene 11 Homo sapiens 79-82 25673014-0 2015 The donor ABCB1 (MDR-1) C3435T polymorphism is a determinant of the graft glomerular filtration rate among tacrolimus treated kidney transplanted patients. Tacrolimus 107-117 ATP binding cassette subfamily B member 1 Homo sapiens 10-15 25631176-0 2015 The role of NFATc1 in prostate cancer progression: cyclosporine A and tacrolimus inhibit cell proliferation, migration, and invasion. Tacrolimus 70-80 nuclear factor of activated T cells 1 Homo sapiens 12-18 25673014-0 2015 The donor ABCB1 (MDR-1) C3435T polymorphism is a determinant of the graft glomerular filtration rate among tacrolimus treated kidney transplanted patients. Tacrolimus 107-117 ATP binding cassette subfamily B member 1 Homo sapiens 17-22 25673014-2 2015 The donor ABCB1 polymorphisms have been related with chronic histological damage and long-term renal function among kidney transplanted patients who received cyclosporine A and tacrolimus (Tac). Tacrolimus 177-187 ATP binding cassette subfamily B member 1 Homo sapiens 10-15 25673014-2 2015 The donor ABCB1 polymorphisms have been related with chronic histological damage and long-term renal function among kidney transplanted patients who received cyclosporine A and tacrolimus (Tac). Tacrolimus 189-192 ATP binding cassette subfamily B member 1 Homo sapiens 10-15 25661468-7 2015 Here, we describe a very unusual neuropsychiatric side effect of tacrolimus and its resolution with another calcineurin inhibitor, cyclosporine, in an adolescent renal transplant recipient. Tacrolimus 65-75 calcineurin binding protein 1 Homo sapiens 108-129 25644968-7 2015 It was found that the concentrations of fasting plasma insulin and C-peptide in NODAT and non-NODAT patients treated with tacrolimus were higher than that in healthy volunteers (p < 0.05). Tacrolimus 122-132 insulin Homo sapiens 55-62 25644968-11 2015 In conclusion, insulin resistance is the primary cause of tacrolimus-induced NODAT. Tacrolimus 58-68 insulin Homo sapiens 15-22 25631176-7 2015 In prostate cancer cell lines, CsA and FK506 inhibited NFATc1 expression and its nuclear translocation, NFAT transcriptional activity, and the expression of c-myc, a downstream target of NFAT. Tacrolimus 39-44 nuclear factor of activated T cells 1 Homo sapiens 55-61 25631176-7 2015 In prostate cancer cell lines, CsA and FK506 inhibited NFATc1 expression and its nuclear translocation, NFAT transcriptional activity, and the expression of c-myc, a downstream target of NFAT. Tacrolimus 39-44 MYC proto-oncogene, bHLH transcription factor Homo sapiens 157-162 25631176-12 2015 Thus, NFATc1 inactivation, especially using CsA and FK506, has the potential of being a therapeutic approach for not only hormone-naive but also castration-resistant prostate cancers. Tacrolimus 52-57 nuclear factor of activated T cells 1 Homo sapiens 6-12 25925267-8 2015 Although presence of CYP3A5 *1 was more frequently observed in the high dose group, initial trough concentration was the only significant factor for determining requirement of high dose of tacrolimus (OR = 28.0, 95% confidence interval 3.20 - 631). Tacrolimus 189-199 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 21-27 25966085-2 2015 One hundred patients who received tacrolimus-based therapy were included in the retrospective study in which the relationship between the tacrolimus blood trough concentration/dosage ratio and the CYP3A5 genotype of both donors and recipients was determined. Tacrolimus 34-44 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 197-203 25714622-6 2015 The mPEG113-PCC32-VE4 micelles significantly increased the absorption of P-gp substrate tacrolimus in the whole intestine. Tacrolimus 88-98 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 73-77 25681723-0 2015 Bovine serum albumin nanoparticles for delivery of tacrolimus to reduce its kidney uptake and functional nephrotoxicity. Tacrolimus 51-61 albumin Homo sapiens 7-20 25681723-2 2015 Tacrolimus (TAC)-loaded bovine serum albumin (BSA) nanoparticles (TAC-BSA-NPs) were prepared by emulsification-dispersion technique. Tacrolimus 0-10 albumin Homo sapiens 31-44 25966085-2 2015 One hundred patients who received tacrolimus-based therapy were included in the retrospective study in which the relationship between the tacrolimus blood trough concentration/dosage ratio and the CYP3A5 genotype of both donors and recipients was determined. Tacrolimus 138-148 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 197-203 25966085-3 2015 Subsequently, 106 patients were continuously enrolled in a prospective study and followed-up for 6 months; the relationship between tacrolimus dosage and CYP3A5 genotype was also determined. Tacrolimus 132-142 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 154-160 25966085-8 2015 A lower tacrolimus dose was suitable for about 25% of the liver transplant patients, as these patients did not have the CYP3A5*1 genotype and received a transplant from a donor with the same genotype. Tacrolimus 8-18 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 120-126 25966085-9 2015 Tailoring the tacrolimus dosage according to the CYP3A5 genotype could reduce rejection and adverse effects. Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 49-55 25394810-0 2015 Immunosuppression with tacrolimus improved reproductive outcome of women with repeated implantation failure and elevated peripheral blood TH1/TH2 cell ratios. Tacrolimus 23-33 negative elongation factor complex member C/D Homo sapiens 138-141 25652038-0 2015 Rapamycin-mediated mTORC2 inhibition is determined by the relative expression of FK506-binding proteins. Tacrolimus 81-86 CREB regulated transcription coactivator 2 Mus musculus 19-25 25780003-2 2015 METHODS: NMSC and peritumoral epidermis protein expression of Bcl-xL and Mcl-1 were assessed by immunohistochemistry in renal transplant recipients receiving tacrolimus or sirolimus and the general population not receiving immunosuppression. Tacrolimus 158-168 BCL2 like 1 Homo sapiens 62-68 25780003-4 2015 Mcl-1 expression intensity in NMSC was decreased in tacrolimus-treated patients compared with sirolimus-treated patients and the nonimmunosuppressed population (P = .024). Tacrolimus 52-62 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 0-5 25780003-7 2015 Importantly in NMSC, Bcl-xL expression was reduced with immunosuppression exposure, and Mcl-1 expression was reduced in tacrolimus-treated compared with sirolimus-treated patients. Tacrolimus 120-130 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 88-93 25394810-1 2015 PROBLEM: We evaluated the clinical efficacy of immunosuppressive treatment with tacrolimus for repeated implantation failure (RIF) patients who have elevated in T helper (Th1)/Th2 cytokine producing cell ratios. Tacrolimus 80-90 negative elongation factor complex member C/D Homo sapiens 171-174 25394810-5 2015 The daily dose of tacrolimus (1-3 mg) was determined based on the degree of the Th1/Th2 cell ratio. Tacrolimus 18-28 negative elongation factor complex member C/D Homo sapiens 80-83 25394810-10 2015 CONCLUSION: An immunosuppressive treatment using tacrolimus improved pregnancy outcome of repeated implantation failure patients with elevated Th1/Th2 ratios. Tacrolimus 49-59 negative elongation factor complex member C/D Homo sapiens 143-146 25416736-4 2015 CYP3A5 expressers require a 1.8-fold higher tacrolimus dose than non-expressers. Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 25416736-5 2015 A visual patient-tailored dosing chart, taking into consideration the child"s weight, recent haematocrit level and CYP3A5 genotype, was developed based on a population pharmacokinetic-pharmacogenetic model, and can be used routinely to individualise tacrolimus starting dose. Tacrolimus 250-260 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 115-121 25591474-8 2015 Similar results were obtained with the calcineurin inhibitor FK506, suggesting that most, but not all, effects on brain development and behavior are mediated by calcineurin inhibition. Tacrolimus 61-66 calcineurin binding protein 1 Homo sapiens 39-60 25891728-1 2015 BACKGROUND: We investigated the effect of FK506 in transcriptional activation of nuclear factor (erythroid-derived 2)-like2 (Nrf2) in human Jurkat T cells. Tacrolimus 42-47 NFE2 like bZIP transcription factor 2 Homo sapiens 125-129 25894154-0 2015 Polymorphism of the CYP3A5 gene and its effect on tacrolimus blood level. Tacrolimus 50-60 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 25894154-1 2015 OBJECTIVES: Tacrolimus is the cornerstone for immunosuppression in renal transplant and is metabolized by the cytochrome P 450 3A (CYP3A) subfamily of enzymes in the liver and small intestine. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 110-129 25894154-1 2015 OBJECTIVES: Tacrolimus is the cornerstone for immunosuppression in renal transplant and is metabolized by the cytochrome P 450 3A (CYP3A) subfamily of enzymes in the liver and small intestine. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 131-136 25894154-2 2015 A polymorphism in intron 3 of the CYP3A5 gene affects the expression of this enzyme and tacrolimus trough blood levels. Tacrolimus 88-98 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 25894154-3 2015 The purpose of this study was to identify the proportion of CYP3A5 gene polymorphisms in South Indian renal transplant patients and determine the effect of CYP3A5 gene polymorphisms on tacrolimus trough blood levels in patients with and without CYP3A5 expression. Tacrolimus 185-195 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 156-162 25894154-3 2015 The purpose of this study was to identify the proportion of CYP3A5 gene polymorphisms in South Indian renal transplant patients and determine the effect of CYP3A5 gene polymorphisms on tacrolimus trough blood levels in patients with and without CYP3A5 expression. Tacrolimus 185-195 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 156-162 25894154-9 2015 Mean tacrolimus level in the CYP3A5*1/*1 group was 5.154 ng/mL (range, 4.42 to 6.5 ng/mL), CYP3A5*1/*3 group was 5.348 ng/mL (range, 3.1 to 9.87 ng/mL), and CYP3A5*3/*3 group was 9.483 ng/mL (range, 4.5 to14.1 ng/mL). Tacrolimus 5-15 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 29-35 25894154-12 2015 Tacrolimus drug level correlated well with presence or absence of CYP3A5 polymorphisms. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 66-72 25894154-15 2015 Therefore, CYP3A5 polymorphism analysis before renal transplant may help determine the optimal dose of tacrolimus in this population and prevent acute rejection episodes or tacrolimus toxicity. Tacrolimus 103-113 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 11-17 25894154-15 2015 Therefore, CYP3A5 polymorphism analysis before renal transplant may help determine the optimal dose of tacrolimus in this population and prevent acute rejection episodes or tacrolimus toxicity. Tacrolimus 173-183 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 11-17 25287072-0 2015 The CYP3A4*22 C>T single nucleotide polymorphism is associated with reduced midazolam and tacrolimus clearance in stable renal allograft recipients. Tacrolimus 93-103 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 4-10 25287072-1 2015 Tacrolimus, a dual substrate of CYP3A4 and CYP3A5 has a narrow therapeutic index and is characterized by high between-subject variability in oral bioavailability. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-38 25287072-1 2015 Tacrolimus, a dual substrate of CYP3A4 and CYP3A5 has a narrow therapeutic index and is characterized by high between-subject variability in oral bioavailability. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 43-49 24414276-1 2015 Human FKBP25, a nuclear protein, is a member of FK506 binding protein family (FKBP) and binds to immunosuppressive drugs such as FK506 and rapamycin. Tacrolimus 48-53 FKBP prolyl isomerase 3 Homo sapiens 6-12 25587129-0 2015 Associations of HSD11B1 polymorphisms with tacrolimus concentrations in Chinese renal transplant recipients with prednisone combined therapy. Tacrolimus 43-53 hydroxysteroid 11-beta dehydrogenase 1 Homo sapiens 16-23 25587129-2 2015 In this study, we investigated the associations of polymorphisms in the gene encoding 11beta-hydroxysteroid dehydrogenase type 1 (HSD11B1) with tacrolimus concentrations in Chinese renal transplant recipients during the early posttransplantation stage. Tacrolimus 144-154 hydroxysteroid 11-beta dehydrogenase 1 Homo sapiens 130-137 25587129-6 2015 These findings illustrate that the HSD11B1 genotypes are closely correlated with tacrolimus trough concentrations, suggesting that these polymorphisms may be useful for safer dosing of tacrolimus. Tacrolimus 81-91 hydroxysteroid 11-beta dehydrogenase 1 Homo sapiens 35-42 25587129-6 2015 These findings illustrate that the HSD11B1 genotypes are closely correlated with tacrolimus trough concentrations, suggesting that these polymorphisms may be useful for safer dosing of tacrolimus. Tacrolimus 185-195 hydroxysteroid 11-beta dehydrogenase 1 Homo sapiens 35-42 25162212-1 2015 BACKGROUND: Tacrolimus (TAC) is a known substrate for cytochrome P450 (CYP) enzyme. Tacrolimus 12-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 25162212-1 2015 BACKGROUND: Tacrolimus (TAC) is a known substrate for cytochrome P450 (CYP) enzyme. Tacrolimus 12-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 25891728-5 2015 RESULTS: We found that a marked dissociation of Nrf 2 from Kelch-like ECH-associated protein-1 and subsequently Nrf 2 nuclear translocation occurred in Jurkat cells treated with FK506 during 48 hours. Tacrolimus 178-183 NFE2 like bZIP transcription factor 2 Homo sapiens 48-53 25891728-5 2015 RESULTS: We found that a marked dissociation of Nrf 2 from Kelch-like ECH-associated protein-1 and subsequently Nrf 2 nuclear translocation occurred in Jurkat cells treated with FK506 during 48 hours. Tacrolimus 178-183 kelch like ECH associated protein 1 Homo sapiens 59-94 25891728-5 2015 RESULTS: We found that a marked dissociation of Nrf 2 from Kelch-like ECH-associated protein-1 and subsequently Nrf 2 nuclear translocation occurred in Jurkat cells treated with FK506 during 48 hours. Tacrolimus 178-183 NFE2 like bZIP transcription factor 2 Homo sapiens 112-117 25891728-6 2015 Immunohistochemistry and Western blot analysis data revealed that the FK506 treatment increased expression of heme oxygenase-1 (HO-1) in Jurkat cells in a dose-dependent manner. Tacrolimus 70-75 heme oxygenase 1 Homo sapiens 110-126 25891728-6 2015 Immunohistochemistry and Western blot analysis data revealed that the FK506 treatment increased expression of heme oxygenase-1 (HO-1) in Jurkat cells in a dose-dependent manner. Tacrolimus 70-75 heme oxygenase 1 Homo sapiens 128-132 25891728-7 2015 HO-1 expression was induced after 6 hours of treatment of FK506 to Jurkat cells, peaked at 24 hours, and then decreased after 48 hours. Tacrolimus 58-63 heme oxygenase 1 Homo sapiens 0-4 25891728-8 2015 CONCLUSIONS: These results suggest that FK506 induces Nrf 2-driven transcriptional activation of the antioxidant response element by activating HO-1 and free radicals such as reactive oxygen species and nitric oxide. Tacrolimus 40-45 NFE2 like bZIP transcription factor 2 Homo sapiens 54-59 25891728-8 2015 CONCLUSIONS: These results suggest that FK506 induces Nrf 2-driven transcriptional activation of the antioxidant response element by activating HO-1 and free radicals such as reactive oxygen species and nitric oxide. Tacrolimus 40-45 heme oxygenase 1 Homo sapiens 144-148 25590378-15 2015 Tacrolimus disappearance was about two-fold higher in cell lines with the combined CYP3A5*1/ABCB1 3435TT genotype versus other genotype combinations. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 83-89 25889879-8 2015 Delivery of FK506 and tat-VIVIT to transgenic APP/PS1 mice attenuated spleen but not brain cytokine levels. Tacrolimus 12-17 presenilin 1 Mus musculus 50-53 25889879-9 2015 However, FK506 and tat-VIVIT significantly attenuated both microgliosis and Abeta plaque load in treated mice compared to controls. Tacrolimus 9-14 amyloid beta (A4) precursor protein Mus musculus 76-81 25590378-15 2015 Tacrolimus disappearance was about two-fold higher in cell lines with the combined CYP3A5*1/ABCB1 3435TT genotype versus other genotype combinations. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 92-97 25590378-19 2015 In particular, ciPTC with the combined CYP3A5*1/ABCB1 3435TT genotype demonstrated higher tacrolimus disappearance versus ciPTCs with a different pharmacogenetic profile. Tacrolimus 90-100 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 39-45 25590378-19 2015 In particular, ciPTC with the combined CYP3A5*1/ABCB1 3435TT genotype demonstrated higher tacrolimus disappearance versus ciPTCs with a different pharmacogenetic profile. Tacrolimus 90-100 ATP binding cassette subfamily B member 1 Homo sapiens 48-53 25660193-0 2015 FK506 reduces neuroinflammation and dopaminergic neurodegeneration in an alpha-synuclein-based rat model for Parkinson"s disease. Tacrolimus 0-5 synuclein alpha Rattus norvegicus 73-88 25588704-0 2015 Kidney transplant recipients carrying the CYP3A4*22 allelic variant have reduced tacrolimus clearance and often reach supratherapeutic tacrolimus concentrations. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 25588704-0 2015 Kidney transplant recipients carrying the CYP3A4*22 allelic variant have reduced tacrolimus clearance and often reach supratherapeutic tacrolimus concentrations. Tacrolimus 135-145 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 25187507-4 2015 PTHrP-mediated induction of RANKL expression was significantly inhibited by H89 and FK506, an inhibitor of PKA and calcineurin, respectively. Tacrolimus 84-89 parathyroid hormone-like peptide Mus musculus 0-5 25187507-4 2015 PTHrP-mediated induction of RANKL expression was significantly inhibited by H89 and FK506, an inhibitor of PKA and calcineurin, respectively. Tacrolimus 84-89 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 28-33 25675879-4 2015 Three animals received the AMR inducing regimen (CD3-IT/alefacept/tacrolimus) with TACI-Ig (atacicept), compared to five control animals treated with the AMR inducing regimen only. Tacrolimus 66-76 TNF receptor superfamily member 13B Homo sapiens 83-87 25660193-6 2015 These data suggest that the anti-inflammatory properties of FK506 decrease neurodegeneration in this alpha-synuclein-based PD model, pointing to a causal role of neuroinflammation in the pathogenesis of PD. Tacrolimus 60-65 synuclein alpha Rattus norvegicus 101-116 25500866-8 2015 The final model including two covariates, population (liver transplant recipients or volunteers) and serum ALT (alanine aminotransferase) level, was verified and adequately described the pharmacokinetic characteristics of tacrolimus. Tacrolimus 222-232 glutamic--pyruvic transaminase Homo sapiens 112-136 25580011-0 2015 Imp2, the PSTPIP homolog in fission yeast, affects sensitivity to the immunosuppressant FK506 and membrane trafficking in fission yeast. Tacrolimus 88-93 insulin like growth factor 2 mRNA binding protein 2 Homo sapiens 0-4 25580011-4 2015 The imp2-c3 mutants showed a defect in cytokinesis with multi-septated phenotypes, which was further enhanced upon treatment with the calcineurin inhibitor FK506. Tacrolimus 156-161 insulin like growth factor 2 mRNA binding protein 2 Homo sapiens 4-11 25442004-9 2015 In cell-based assays, ACY-738 increased the relative association of Hsp90 with FK506 binding protein 51 versus FK506 binding protein 52 and inhibited hormone-induced GR translocation. Tacrolimus 79-84 heat shock protein, 3 Mus musculus 68-73 25663111-2 2015 Mip exhibits peptidyl prolyl cistrans isomerase (PPIase) activity, which can be inhibited by Rapamycin and FK506. Tacrolimus 107-112 major intrinsic protein of lens fiber Homo sapiens 0-3 25377120-4 2015 Cyclosporine A, FK506, rapamycin and juglone are known PPIase inhibitors. Tacrolimus 16-21 FKBP prolyl isomerase 7 Homo sapiens 55-61 25487141-0 2015 Association between interleukin-18 promoter variants and tacrolimus pharmacokinetics in Chinese renal transplant patients. Tacrolimus 57-67 interleukin 18 Homo sapiens 20-34 25487141-2 2015 This study aimed to assess the potential influence of two functional single nucleotide polymorphisms (SNPs) in the IL-18 promoter region on the tacrolimus pharmacokinetics in Chinese renal transplant patients. Tacrolimus 144-154 interleukin 18 Homo sapiens 115-120 25487141-6 2015 RESULTS: The tacrolimus C/D ratio was significantly associated with the IL-18 rs1946518 gene polymorphism in the first month after transplantation (P = 0.0225). Tacrolimus 13-23 interleukin 18 Homo sapiens 72-77 25487141-9 2015 In a simple linear regression model, age, hemoglobin (Hb), CYP3A5 gene polymorphisms, and IL-18 A-607C gene polymorphisms were associated with log-transformed tacrolimus C/D ratios (P < 0.05). Tacrolimus 159-169 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 59-65 25487141-9 2015 In a simple linear regression model, age, hemoglobin (Hb), CYP3A5 gene polymorphisms, and IL-18 A-607C gene polymorphisms were associated with log-transformed tacrolimus C/D ratios (P < 0.05). Tacrolimus 159-169 interleukin 18 Homo sapiens 90-95 25487141-10 2015 In the final multiple linear regression model, CYP3A5 polymorphisms were the most important variant, accounting for 19.5 % of total variation involved in tacrolimus pharmacokinetics. Tacrolimus 154-164 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 47-53 25487141-11 2015 CONCLUSION: Our findings suggest that a combined analysis of CYP3A5 and IL-18 promoter polymorphisms may help clinicians develop individualized tacrolimus treatment, which is based on determining CYP3A5 genotype. Tacrolimus 144-154 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-67 25487141-11 2015 CONCLUSION: Our findings suggest that a combined analysis of CYP3A5 and IL-18 promoter polymorphisms may help clinicians develop individualized tacrolimus treatment, which is based on determining CYP3A5 genotype. Tacrolimus 144-154 interleukin 18 Homo sapiens 72-77 25487141-11 2015 CONCLUSION: Our findings suggest that a combined analysis of CYP3A5 and IL-18 promoter polymorphisms may help clinicians develop individualized tacrolimus treatment, which is based on determining CYP3A5 genotype. Tacrolimus 144-154 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 196-202 25338944-4 2015 The highest concentration/expression of CXCL10 was detected in allograft tissue compared with allograft treated with tacrolimus and isograft control. Tacrolimus 117-127 chemokine (C-X-C motif) ligand 10 Mus musculus 40-46 25638160-4 2015 In bladder cancer cell lines, cyclosporine A (CsA) and tacrolimus (FK506), immunosuppressant drugs/non-selective NFAT inhibitors, attenuated NFATc1 expression and its nuclear translocation, NFAT transcriptional activity, and the expression of cyclooxygenase-2 and c-myc, downstream targets of NFATc1. Tacrolimus 55-65 MYC proto-oncogene, bHLH transcription factor Homo sapiens 264-269 25810886-0 2015 Intra-CA1 administration of FK-506 (tacrolimus) in rat impairs learning and memory in an inhibitory avoidance paradigm. Tacrolimus 28-34 carbonic anhydrase 1 Rattus norvegicus 6-9 25150084-9 2015 Inhibition of CaMK with KN93 and CBD increased NO production but the calcineurin inhibitor FK 506 decreased iNOS expression. Tacrolimus 91-97 nitric oxide synthase 2 Rattus norvegicus 108-112 25810886-0 2015 Intra-CA1 administration of FK-506 (tacrolimus) in rat impairs learning and memory in an inhibitory avoidance paradigm. Tacrolimus 36-46 carbonic anhydrase 1 Rattus norvegicus 6-9 25810886-4 2015 MATERIALS AND METHODS: Using different doses of FK-506 (0.5, 5, and 50 nM) in the CA1 of hippocampus at different times (before, after the training and also before the test), the effect of drug was evaluated in a step-through inhibitory avoidance paradigm. Tacrolimus 48-54 carbonic anhydrase 1 Rattus norvegicus 82-85 25810886-6 2015 RESULTS: The pre-training intra-CA1 injections of FK-506 impaired inhibitory avoidance (IA) learning acquisition. Tacrolimus 50-56 carbonic anhydrase 1 Rattus norvegicus 32-35 25810886-7 2015 In addition, the post-training intra-CA1 injections of FK-506 at 1, 2, and 3 hr relative to training impaired memory consolidation. Tacrolimus 55-61 carbonic anhydrase 1 Rattus norvegicus 37-40 25810886-8 2015 Moreover, the pre-test intra-CA1 injections of FK-506 impaired memory retrieval. Tacrolimus 47-53 carbonic anhydrase 1 Rattus norvegicus 29-32 25810886-9 2015 CONCLUSION: These findings suggest that the FK-506 selectively interferes with acquisition, retention, and retrieval of information processing in CA1 of hippocampus. Tacrolimus 44-50 carbonic anhydrase 1 Rattus norvegicus 146-149 25201288-0 2015 Effect of CYP3A5*3 on kidney transplant recipients treated with tacrolimus: a systematic review and meta-analysis of observational studies. Tacrolimus 64-74 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 25201288-3 2015 We conducted a meta-analysis of studies evaluating the effect of CYP3A5 polymorphism on kidney transplant recipients with tacrolimus plasma concentration divided by daily dose per body weight (C/D) and clinical outcomes. Tacrolimus 122-132 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 65-71 25201288-7 2015 In conclusion, CYP3A5 6986A>G polymorphism can affect tacrolimus pharmacokinetics and the incidence of acute rejection and chronic nephrotoxicity on kidney transplant recipients. Tacrolimus 57-67 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 25975025-0 2015 [Associations of SUMO4 polymorphisms with tacrolimus concentrations in Chinese renal transplant recipients]. Tacrolimus 42-52 small ubiquitin like modifier 4 Homo sapiens 17-22 25975025-1 2015 The study aims to investigate the associations of SUMO4 polymorphisms with tacrolimus concentrations in Chinese renal transplant recipients. Tacrolimus 75-85 small ubiquitin like modifier 4 Homo sapiens 50-55 25975025-5 2015 The dose-adjusted concentration of tacrolimus in SUMO4 rs237024A-rs237025A (GA-GA +AA-AA) carriers was considerably higher than that in GG-GG carriers (P < 0.05). Tacrolimus 35-45 small ubiquitin like modifier 4 Homo sapiens 49-54 25975025-6 2015 After stratification by CYP3A5*3 genotypes, SUMO4 rs237024A-rs237025A carriers (GA-GA+AA-AA) had a higher dose-adjusted tacrolimus concentration than that in GG carriers in CYP3A5 expresser (P < 0.05). Tacrolimus 120-130 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-30 25975025-6 2015 After stratification by CYP3A5*3 genotypes, SUMO4 rs237024A-rs237025A carriers (GA-GA+AA-AA) had a higher dose-adjusted tacrolimus concentration than that in GG carriers in CYP3A5 expresser (P < 0.05). Tacrolimus 120-130 small ubiquitin like modifier 4 Homo sapiens 44-49 25975025-6 2015 After stratification by CYP3A5*3 genotypes, SUMO4 rs237024A-rs237025A carriers (GA-GA+AA-AA) had a higher dose-adjusted tacrolimus concentration than that in GG carriers in CYP3A5 expresser (P < 0.05). Tacrolimus 120-130 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 173-179 25975025-7 2015 The results illustrated that SUMO4 rs237024 and rs237025 polymorphisms were associated with tacrolimus concentrations, and the test of these genotypes may be useful for individualized medicine of tacrolimus. Tacrolimus 92-102 small ubiquitin like modifier 4 Homo sapiens 29-34 25975025-7 2015 The results illustrated that SUMO4 rs237024 and rs237025 polymorphisms were associated with tacrolimus concentrations, and the test of these genotypes may be useful for individualized medicine of tacrolimus. Tacrolimus 196-206 small ubiquitin like modifier 4 Homo sapiens 29-34 25638160-0 2015 Cyclosporine A and tacrolimus inhibit bladder cancer growth through down-regulation of NFATc1. Tacrolimus 19-29 nuclear factor of activated T cells 1 Homo sapiens 87-93 25638160-4 2015 In bladder cancer cell lines, cyclosporine A (CsA) and tacrolimus (FK506), immunosuppressant drugs/non-selective NFAT inhibitors, attenuated NFATc1 expression and its nuclear translocation, NFAT transcriptional activity, and the expression of cyclooxygenase-2 and c-myc, downstream targets of NFATc1. Tacrolimus 55-65 nuclear factor of activated T cells 1 Homo sapiens 141-147 25638160-4 2015 In bladder cancer cell lines, cyclosporine A (CsA) and tacrolimus (FK506), immunosuppressant drugs/non-selective NFAT inhibitors, attenuated NFATc1 expression and its nuclear translocation, NFAT transcriptional activity, and the expression of cyclooxygenase-2 and c-myc, downstream targets of NFATc1. Tacrolimus 55-65 prostaglandin-endoperoxide synthase 2 Homo sapiens 243-259 25638160-4 2015 In bladder cancer cell lines, cyclosporine A (CsA) and tacrolimus (FK506), immunosuppressant drugs/non-selective NFAT inhibitors, attenuated NFATc1 expression and its nuclear translocation, NFAT transcriptional activity, and the expression of cyclooxygenase-2 and c-myc, downstream targets of NFATc1. Tacrolimus 55-65 nuclear factor of activated T cells 1 Homo sapiens 293-299 25638160-4 2015 In bladder cancer cell lines, cyclosporine A (CsA) and tacrolimus (FK506), immunosuppressant drugs/non-selective NFAT inhibitors, attenuated NFATc1 expression and its nuclear translocation, NFAT transcriptional activity, and the expression of cyclooxygenase-2 and c-myc, downstream targets of NFATc1. Tacrolimus 67-72 nuclear factor of activated T cells 1 Homo sapiens 141-147 25638160-4 2015 In bladder cancer cell lines, cyclosporine A (CsA) and tacrolimus (FK506), immunosuppressant drugs/non-selective NFAT inhibitors, attenuated NFATc1 expression and its nuclear translocation, NFAT transcriptional activity, and the expression of cyclooxygenase-2 and c-myc, downstream targets of NFATc1. Tacrolimus 67-72 prostaglandin-endoperoxide synthase 2 Homo sapiens 243-259 25638160-4 2015 In bladder cancer cell lines, cyclosporine A (CsA) and tacrolimus (FK506), immunosuppressant drugs/non-selective NFAT inhibitors, attenuated NFATc1 expression and its nuclear translocation, NFAT transcriptional activity, and the expression of cyclooxygenase-2 and c-myc, downstream targets of NFATc1. Tacrolimus 67-72 MYC proto-oncogene, bHLH transcription factor Homo sapiens 264-269 25638160-4 2015 In bladder cancer cell lines, cyclosporine A (CsA) and tacrolimus (FK506), immunosuppressant drugs/non-selective NFAT inhibitors, attenuated NFATc1 expression and its nuclear translocation, NFAT transcriptional activity, and the expression of cyclooxygenase-2 and c-myc, downstream targets of NFATc1. Tacrolimus 67-72 nuclear factor of activated T cells 1 Homo sapiens 293-299 25638160-9 2015 Thus, NFATc1 inactivation, especially using CsA and FK506, has the potential of being a therapeutic approach for bladder cancer. Tacrolimus 52-57 nuclear factor of activated T cells 1 Homo sapiens 6-12 26439819-9 2015 (3) IHC staining of nephrin and podocin revealed unaffected expression and better linear distributions in the high-dose SPE and tacrolimus groups. Tacrolimus 128-138 NPHS1 adhesion molecule, nephrin Rattus norvegicus 20-27 25594874-0 2015 Capability of utilizing CYP3A5 polymorphisms to predict therapeutic dosage of tacrolimus at early stage post-renal transplantation. Tacrolimus 78-88 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-30 25594874-5 2015 The contribution of genetic factors (CYP3A5*1 or *3) for tacrolimus dosing showed increased variation from Day 14 to Day 28 after transplantation: 7.2%, 18.4% and 19.5% on Days 14, 21 and 28, respectively. Tacrolimus 57-67 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 25594874-6 2015 The influence of CYP3A5 polymorphisms on the tacrolimus maintenance dosage became evident after Day 14 post-transplantation, although the tacrolimus dosage was determined based only on patient body weight for the first three days after surgery. Tacrolimus 45-55 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 17-23 25594874-7 2015 Tacrolimus dosage starting with the initial administration should be individualized using the CYP3A5 genotype information. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 94-100 25799658-3 2015 Preemptive CYP3A5 genotyping and extended-release tacrolimus formulations should allow the prescription of a suitable dose of tacrolimus immediately and avoid overexposure as a source of nephrotoxicity. Tacrolimus 126-136 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 11-17 26439819-9 2015 (3) IHC staining of nephrin and podocin revealed unaffected expression and better linear distributions in the high-dose SPE and tacrolimus groups. Tacrolimus 128-138 NPHS2 stomatin family member, podocin Rattus norvegicus 32-39 26439819-11 2015 (4) The mRNA levels of nephrin and podocin in the tacrolimus and high-dose SPE groups were significantly higher than that in the others. Tacrolimus 50-60 NPHS1 adhesion molecule, nephrin Rattus norvegicus 23-30 26439819-11 2015 (4) The mRNA levels of nephrin and podocin in the tacrolimus and high-dose SPE groups were significantly higher than that in the others. Tacrolimus 50-60 NPHS2 stomatin family member, podocin Rattus norvegicus 35-42 26152630-8 2015 It has been also shown that antimycotics and tacrolimus suppress the induced production of IP-10 in human keratinocytes. Tacrolimus 45-55 C-X-C motif chemokine ligand 10 Homo sapiens 91-96 25614218-3 2015 We found that the temperature sensitivity of cells in which the conserved Verprolin VRP1 gene had been deleted, which gene is required for actin organization and endocytosis, was suppressed by either FK506 or by cnb1 deletion. Tacrolimus 200-205 Vrp1p Saccharomyces cerevisiae S288C 84-88 25975003-6 2015 Among CYP3A5 substrates are cyclosporine and tacrolimus prescribed after organ transplantations. Tacrolimus 45-55 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 6-12 25986565-7 2015 Finally, the fact that FKBP52 is already a validated target of the clinically approved immunosuppressive drug, FK506 (Tacrolimus), indicates that FKBP52 is a "druggable" protein. Tacrolimus 118-128 FK506 binding protein 4 Mus musculus 23-29 25968921-0 2015 Involvement of CYP 3A5 In the Interaction Between Tacrolimus and Nicardipine: A Case Report. Tacrolimus 50-60 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-22 25986565-7 2015 Finally, the fact that FKBP52 is already a validated target of the clinically approved immunosuppressive drug, FK506 (Tacrolimus), indicates that FKBP52 is a "druggable" protein. Tacrolimus 118-128 FK506 binding protein 4 Mus musculus 146-152 25986565-7 2015 Finally, the fact that FKBP52 is already a validated target of the clinically approved immunosuppressive drug, FK506 (Tacrolimus), indicates that FKBP52 is a "druggable" protein. Tacrolimus 111-116 FK506 binding protein 4 Mus musculus 23-29 25986565-7 2015 Finally, the fact that FKBP52 is already a validated target of the clinically approved immunosuppressive drug, FK506 (Tacrolimus), indicates that FKBP52 is a "druggable" protein. Tacrolimus 111-116 FK506 binding protein 4 Mus musculus 146-152 25986568-2 2015 The prototype of this protein family, FKBP12, is the binding partner for immunosuppressive drugs FK506 and rapamycin. Tacrolimus 97-102 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 38-44 25986569-3 2015 Most Cyps and FKBP family members catalyse peptidyl-prolyl cis/trans isomerase (PPIase) mediated reactions and form binary complexes with their ligands cyclosporine A and FK506. Tacrolimus 171-176 FKBP prolyl isomerase 5 Homo sapiens 43-78 25986569-3 2015 Most Cyps and FKBP family members catalyse peptidyl-prolyl cis/trans isomerase (PPIase) mediated reactions and form binary complexes with their ligands cyclosporine A and FK506. Tacrolimus 171-176 FKBP prolyl isomerase 5 Homo sapiens 80-86 25877445-0 2015 Reversible, time-dependent inhibition of CYP3A-mediated metabolism of midazolam and tacrolimus by telaprevir in human liver microsomes. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-46 25132599-9 2015 Knockdown of FKBP52 and treatment with the FK-binding immunosuppressant FK506 enhanced TDO expression and activity in glioblastoma cells. Tacrolimus 72-77 tryptophan 2,3-dioxygenase Homo sapiens 87-90 24471694-0 2015 The full electron structure of the FKBP12/FK506 complex. Tacrolimus 42-47 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 35-41 24471694-5 2015 The results indicate that the interactions occur only between the LUMOs of FKBP12 and the HOMO of FK506, not between the HOMOs of FKBP12 and the LUMO of FK506. Tacrolimus 98-103 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 75-81 24471694-7 2015 The electron structures of FKBP12/FK506 give us a clearer understanding of their interaction mechanism and will help us design new ligands of FKBP12. Tacrolimus 34-39 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 27-33 24471694-7 2015 The electron structures of FKBP12/FK506 give us a clearer understanding of their interaction mechanism and will help us design new ligands of FKBP12. Tacrolimus 34-39 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 142-148 25968921-1 2015 Tacrolimus is a calcineurin inhibitor primarily metabolized by CYP3A4 and secondarily by CYP3A5. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 63-69 25968921-1 2015 Tacrolimus is a calcineurin inhibitor primarily metabolized by CYP3A4 and secondarily by CYP3A5. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 89-95 25968921-8 2015 Increase of tacrolimus trough concentrations is due to the inhibition of CYP3A4. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 73-79 25968921-9 2015 Very high levels of tacrolimus suggest the non expression of CYP3A5. Tacrolimus 20-30 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-67 25684872-6 2015 Initiation of tacrolimus (TAC) was followed by dramatic response: Proteinuria decreased, serum albumin normalized and C-ANCA and anti-PR3 ANCA assays became negative. Tacrolimus 14-24 proteinase 3 Homo sapiens 118-124 26568012-0 2015 Successful Management of Lupus Nephritis with High Titers of Myeloperoxidase Anti-Neutrophil Cytoplasmic Antibodies Using Tacrolimus. Tacrolimus 122-132 myeloperoxidase Homo sapiens 61-76 25877445-3 2015 METHODS: We performed a static mechanistic DDI prediction to evaluate whether previously reported competitive inhibition of CYP3A by telaprevir and its diastereomeric metabolite - VRT-127394 is sufficient to explain the remarkable reduction in oral clearance observed with oral midazolam and tacrolimus. Tacrolimus 292-302 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 124-129 25712187-0 2015 Donor IL-18 rs5744247 polymorphism as a new biomarker of tacrolimus elimination in Chinese liver transplant patients during the early post-transplantation period: results from two cohort studies. Tacrolimus 57-67 interleukin 18 Homo sapiens 6-11 25333459-0 2015 IL-15 is decreased upon CsA and FK506 treatment of acute rejection following heart transplantation in mice. Tacrolimus 32-37 interleukin 15 Mus musculus 0-5 25333459-1 2015 The aim of this study was to investigate the effect of cyclosporine A (CsA) and tacrolimus (FK506) on interleukin-15 (IL-15) production during acute rejection following heart transplantation in mice. Tacrolimus 80-90 interleukin 15 Mus musculus 102-116 25333459-1 2015 The aim of this study was to investigate the effect of cyclosporine A (CsA) and tacrolimus (FK506) on interleukin-15 (IL-15) production during acute rejection following heart transplantation in mice. Tacrolimus 80-90 interleukin 15 Mus musculus 118-123 25333459-1 2015 The aim of this study was to investigate the effect of cyclosporine A (CsA) and tacrolimus (FK506) on interleukin-15 (IL-15) production during acute rejection following heart transplantation in mice. Tacrolimus 92-97 interleukin 15 Mus musculus 102-116 25333459-1 2015 The aim of this study was to investigate the effect of cyclosporine A (CsA) and tacrolimus (FK506) on interleukin-15 (IL-15) production during acute rejection following heart transplantation in mice. Tacrolimus 92-97 interleukin 15 Mus musculus 118-123 25333459-6 2015 Compared to the allogeneic acute rejection group, IL-15 expression was significantly decreased in the CsA- and FK506-treated allogeneic rejection groups. Tacrolimus 111-116 interleukin 15 Mus musculus 50-55 25333459-8 2015 IL-2 expression was increased in the allogeneic acute rejection group and was inhibited in mice treated with CsA and FK506. Tacrolimus 117-122 interleukin 2 Mus musculus 0-4 25333459-9 2015 In conclusion, increased IL-15 expression in rejected murine heart grafts may be reduced by CsA and FK506 in vivo. Tacrolimus 100-105 interleukin 15 Mus musculus 25-30 26299169-11 2015 Ins2 gene expressions in rats receiving tacrolimus-cyclosporin and rats receiving the vehicle were comparable. Tacrolimus 40-50 insulin 2 Rattus norvegicus 0-4 25916520-0 2015 Relationship between mRNA expression levels of CYP3A4, CYP3A5 and SXR in peripheral mononuclear blood cells and aging in young kidney transplant recipients under tacrolimus treatment. Tacrolimus 162-172 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-53 25916520-0 2015 Relationship between mRNA expression levels of CYP3A4, CYP3A5 and SXR in peripheral mononuclear blood cells and aging in young kidney transplant recipients under tacrolimus treatment. Tacrolimus 162-172 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 55-61 26199736-4 2015 This study was motivated by a patient with acute kidney injury associated with elevated serum tacrolimus levels in whom KIM-1 staining was present only in proximal tubules located in the medullary rays in the setting of otherwise normal light, immunofluorescent, and electron microscopy. Tacrolimus 94-104 hepatitis A virus cellular receptor 1 Homo sapiens 120-125 25712187-1 2015 AIM: This study evaluated the relationships between IL-18 polymorphisms and tacrolimus elimination in Chinese liver transplant patients. Tacrolimus 76-86 interleukin 18 Homo sapiens 52-57 26228923-1 2015 AIMS: To evaluate the influences of CYP3A4, CYP3A5, MDR1 and NR1I2 polymorphisms on tacrolimus concentration in early postrenal transplant recipients. Tacrolimus 84-94 nuclear receptor subfamily 1 group I member 2 Homo sapiens 61-66 25712187-4 2015 RESULTS: In training set, daily drug dose, total bilirubin, donor CYP3A5 rs776746 and IL-18 rs5744247 genotypes were screened to construct prediction model for tacrolimus elimination. Tacrolimus 160-170 interleukin 18 Homo sapiens 86-91 26228923-3 2015 RESULTS: CYP3A5*3 explained 32.8% of variability of tacrolimus C0D7/D. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 9-15 25712187-6 2015 Donor IL-18 rs5744247 polymorphism was an independent predictor of tacrolimus elimination in the first week after transplantation in both training (p = 0.008) and validating cohorts (p = 0.033). Tacrolimus 67-77 interleukin 18 Homo sapiens 6-11 26228923-4 2015 CYP3A4*1G, MDR1 1236-2677-3435 diplotype and NR1I2 -25385C > T explained 21.4% of variability of tacrolimus C0D7/D in CYP3A5 nonexpressers. Tacrolimus 100-110 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 26228923-4 2015 CYP3A4*1G, MDR1 1236-2677-3435 diplotype and NR1I2 -25385C > T explained 21.4% of variability of tacrolimus C0D7/D in CYP3A5 nonexpressers. Tacrolimus 100-110 ATP binding cassette subfamily B member 1 Homo sapiens 11-15 25916520-0 2015 Relationship between mRNA expression levels of CYP3A4, CYP3A5 and SXR in peripheral mononuclear blood cells and aging in young kidney transplant recipients under tacrolimus treatment. Tacrolimus 162-172 nuclear receptor subfamily 1 group I member 2 Homo sapiens 66-69 25712187-7 2015 CONCLUSION: Donor IL-18 rs5744247 polymorphism may influence on tacrolimus elimination. Tacrolimus 64-74 interleukin 18 Homo sapiens 18-23 25916520-5 2015 PATIENTS & METHODS: The correlation between the levels of mRNA specific for key enzymes SXR, CYP3A and ABCB1 involved in the metabolism of tacrolimus was evaluated in PBMCs obtained from a selected population of 29 young kidney transplant recipients. Tacrolimus 143-153 nuclear receptor subfamily 1 group I member 2 Homo sapiens 92-95 25916520-5 2015 PATIENTS & METHODS: The correlation between the levels of mRNA specific for key enzymes SXR, CYP3A and ABCB1 involved in the metabolism of tacrolimus was evaluated in PBMCs obtained from a selected population of 29 young kidney transplant recipients. Tacrolimus 143-153 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 97-102 26228923-4 2015 CYP3A4*1G, MDR1 1236-2677-3435 diplotype and NR1I2 -25385C > T explained 21.4% of variability of tacrolimus C0D7/D in CYP3A5 nonexpressers. Tacrolimus 100-110 nuclear receptor subfamily 1 group I member 2 Homo sapiens 45-50 26228923-4 2015 CYP3A4*1G, MDR1 1236-2677-3435 diplotype and NR1I2 -25385C > T explained 21.4% of variability of tacrolimus C0D7/D in CYP3A5 nonexpressers. Tacrolimus 100-110 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 121-127 26228923-5 2015 CONCLUSION: CYP3A5*3 was the predominant determinant affecting tacrolimus concentration. Tacrolimus 63-73 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 25916520-5 2015 PATIENTS & METHODS: The correlation between the levels of mRNA specific for key enzymes SXR, CYP3A and ABCB1 involved in the metabolism of tacrolimus was evaluated in PBMCs obtained from a selected population of 29 young kidney transplant recipients. Tacrolimus 143-153 ATP binding cassette subfamily B member 1 Homo sapiens 107-112 26228923-6 2015 Genotyping of CYP3A4/MDR1/NR1I2 polymorphisms may be helpful for better guiding tacrolimus dosing in CYP3A5 nonexpressers. Tacrolimus 80-90 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-20 25869250-14 2015 Population PK analysis of tacrolimus in Chinese adult liver transplant patients resulted in identification of the CYP3A5 genotype, POD, BUN, ALP, HCT, TBIL and HB as significant covariates on the PK parameters of tacrolimus. Tacrolimus 26-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 114-120 26228923-6 2015 Genotyping of CYP3A4/MDR1/NR1I2 polymorphisms may be helpful for better guiding tacrolimus dosing in CYP3A5 nonexpressers. Tacrolimus 80-90 ATP binding cassette subfamily B member 1 Homo sapiens 21-25 26228923-6 2015 Genotyping of CYP3A4/MDR1/NR1I2 polymorphisms may be helpful for better guiding tacrolimus dosing in CYP3A5 nonexpressers. Tacrolimus 80-90 nuclear receptor subfamily 1 group I member 2 Homo sapiens 26-31 26228923-6 2015 Genotyping of CYP3A4/MDR1/NR1I2 polymorphisms may be helpful for better guiding tacrolimus dosing in CYP3A5 nonexpressers. Tacrolimus 80-90 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 101-107 25916520-8 2015 tacrolimus-normalized daily dose was strongly correlated with patient"s age and multivariable regression indicates the CYP3A4-specific mRNA as the sole independent variable influencing tacrolimus concentration-to-dose ratio. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 119-125 25916520-8 2015 tacrolimus-normalized daily dose was strongly correlated with patient"s age and multivariable regression indicates the CYP3A4-specific mRNA as the sole independent variable influencing tacrolimus concentration-to-dose ratio. Tacrolimus 185-195 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 119-125 25869250-14 2015 Population PK analysis of tacrolimus in Chinese adult liver transplant patients resulted in identification of the CYP3A5 genotype, POD, BUN, ALP, HCT, TBIL and HB as significant covariates on the PK parameters of tacrolimus. Tacrolimus 213-223 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 114-120 26189776-8 2015 The average annual tacrolimus C12h was higher for patients with CYP3A5*3/*3 (PM) than for patients with the CYP3A5*1 allele (EM) [4.6 (3.2-6.6) versus 2.5 (2.0-3.1) ng/mL, respectively, p=0.002]. Tacrolimus 19-29 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 64-70 25491293-8 2014 The gene expression of IL-1beta and TNF-alpha in the FK-506 and vehicle-treated groups were 47.01% and 45.56%, 85.91% and 115.83% of that in the saline-treated group in the ocular surface, while in the lacrimal glands 49.16% and 67.60%, 94.91% and 95.77% of that in the saline-treated group, respectively. Tacrolimus 53-59 interleukin 1 beta Mus musculus 23-31 26858893-1 2015 The steroid receptor (SR) complex contains FKBP51 and FKBP52, which bind to tacrolimus (TAC) and cyclophilin 40, which, in turn, bind to cyclosporine (CYA); these influence the intranuclear mobility of steroid-SR complexes. Tacrolimus 76-86 FKBP prolyl isomerase 4 Homo sapiens 54-60 25491391-7 2014 The current study will test the hypothesis that MSC treatment, in combination with the mTOR inhibitor everolimus, facilitates tacrolimus withdrawal, reduces fibrosis and decreases the incidence of opportunistic infections compared to standard tacrolimus dose. Tacrolimus 126-136 mechanistic target of rapamycin kinase Homo sapiens 87-91 25491293-8 2014 The gene expression of IL-1beta and TNF-alpha in the FK-506 and vehicle-treated groups were 47.01% and 45.56%, 85.91% and 115.83% of that in the saline-treated group in the ocular surface, while in the lacrimal glands 49.16% and 67.60%, 94.91% and 95.77% of that in the saline-treated group, respectively. Tacrolimus 53-59 tumor necrosis factor Mus musculus 36-45 25491293-9 2014 The ratio of phosphorylated IkappaB-alpha to total IkappaB-alpha in the keratoconjunctival tissues was lower in the FK-506-treated group than in the vehicle- and saline-treated groups (both P < 0.05). Tacrolimus 116-122 nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha Mus musculus 28-41 25491293-9 2014 The ratio of phosphorylated IkappaB-alpha to total IkappaB-alpha in the keratoconjunctival tissues was lower in the FK-506-treated group than in the vehicle- and saline-treated groups (both P < 0.05). Tacrolimus 116-122 nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha Mus musculus 51-64 25471588-9 2014 FK506 inhibited the observed alphaSYN oligomerization both in cell culture and in mouse brain. Tacrolimus 0-5 synuclein, alpha Mus musculus 29-37 25407098-0 2014 Once-daily LCP-Tacro MeltDose tacrolimus for the prophylaxis of organ rejection in kidney and liver transplantations. Tacrolimus 15-20 kelch domain containing 2 Homo sapiens 11-14 25464008-0 2014 Tacrolimus (FK506) suppresses TREM-1 expression at an early but not at a late stage in a murine model of fungal keratitis. Tacrolimus 0-10 triggering receptor expressed on myeloid cells 1 Mus musculus 30-36 25464008-0 2014 Tacrolimus (FK506) suppresses TREM-1 expression at an early but not at a late stage in a murine model of fungal keratitis. Tacrolimus 12-17 triggering receptor expressed on myeloid cells 1 Mus musculus 30-36 25464008-1 2014 PURPOSE: To investigate the efficacy and mechanism of tacrolimus(FK506), which is a novel macrolide immunosuppressant, in inhibiting triggering receptor expressed on myeloid cells-1 (TREM-1) expression in a murine keratitis model induced by Aspergillus fumigatus. Tacrolimus 54-64 triggering receptor expressed on myeloid cells 1 Mus musculus 133-181 25464008-1 2014 PURPOSE: To investigate the efficacy and mechanism of tacrolimus(FK506), which is a novel macrolide immunosuppressant, in inhibiting triggering receptor expressed on myeloid cells-1 (TREM-1) expression in a murine keratitis model induced by Aspergillus fumigatus. Tacrolimus 54-64 triggering receptor expressed on myeloid cells 1 Mus musculus 183-189 25464008-1 2014 PURPOSE: To investigate the efficacy and mechanism of tacrolimus(FK506), which is a novel macrolide immunosuppressant, in inhibiting triggering receptor expressed on myeloid cells-1 (TREM-1) expression in a murine keratitis model induced by Aspergillus fumigatus. Tacrolimus 65-70 triggering receptor expressed on myeloid cells 1 Mus musculus 133-181 25464008-1 2014 PURPOSE: To investigate the efficacy and mechanism of tacrolimus(FK506), which is a novel macrolide immunosuppressant, in inhibiting triggering receptor expressed on myeloid cells-1 (TREM-1) expression in a murine keratitis model induced by Aspergillus fumigatus. Tacrolimus 65-70 triggering receptor expressed on myeloid cells 1 Mus musculus 183-189 25464008-9 2014 In contrast, FK506 reduced the expression of TREM-1, IL-1beta and TNFalpha in RAW264.7 macrophages stimulated with zymosan. Tacrolimus 13-18 triggering receptor expressed on myeloid cells 1 Mus musculus 45-51 25464008-9 2014 In contrast, FK506 reduced the expression of TREM-1, IL-1beta and TNFalpha in RAW264.7 macrophages stimulated with zymosan. Tacrolimus 13-18 interleukin 1 beta Mus musculus 53-61 25464008-9 2014 In contrast, FK506 reduced the expression of TREM-1, IL-1beta and TNFalpha in RAW264.7 macrophages stimulated with zymosan. Tacrolimus 13-18 tumor necrosis factor Mus musculus 66-74 25464008-13 2014 CONCLUSIONS: FK506 may inhibit the inflammation induced by fungi and alleviate the severity of corneal damage at an early stage of fungal keratitis by downregulating TREM-1 expression. Tacrolimus 13-18 triggering receptor expressed on myeloid cells 1 Mus musculus 166-172 25407098-0 2014 Once-daily LCP-Tacro MeltDose tacrolimus for the prophylaxis of organ rejection in kidney and liver transplantations. Tacrolimus 30-40 kelch domain containing 2 Homo sapiens 11-14 25407098-4 2014 LCP-Tacro is a novel, prolonged-release, MeltDose( ) formulation of tacrolimus designed for once-daily administration. Tacrolimus 68-78 kelch domain containing 2 Homo sapiens 0-3 25407098-6 2014 Considering the studies conducted to date, once-daily LCP-Tacro has shown improved pharmacokinetic properties, rapid achievement of therapeutic trough levels, consistent exposure, non-inferior efficacy and similar safety, with lower tacrolimus dose than other tacrolimus formulations. Tacrolimus 233-243 kelch domain containing 2 Homo sapiens 54-57 25407098-6 2014 Considering the studies conducted to date, once-daily LCP-Tacro has shown improved pharmacokinetic properties, rapid achievement of therapeutic trough levels, consistent exposure, non-inferior efficacy and similar safety, with lower tacrolimus dose than other tacrolimus formulations. Tacrolimus 260-270 kelch domain containing 2 Homo sapiens 54-57 25043904-8 2014 Accordingly, the inhibition of CaN by FK506 increased NF-kappaB activity in dorsal hippocampus. Tacrolimus 38-43 nuclear factor kappa B subunit 1 Homo sapiens 54-63 25043904-9 2014 The administration of the NF-kappaB signaling pathway inhibitor sulfasalazine (SSZ) impeded the enhancing effect of FK506. Tacrolimus 116-121 nuclear factor kappa B subunit 1 Homo sapiens 26-35 25043904-12 2014 The coadministration of SSZ and FK506 blocked the enhancement effect in reconsolidation, suggesting that this facilitation is also dependent on the NF-kappaB signaling pathway. Tacrolimus 32-37 nuclear factor kappa B subunit 1 Homo sapiens 148-157 25330904-5 2014 Pharmacological inhibitors of the catalytic subunit of protein phosphatase 2B (PP2B-A) such as cyclosporine A or tacrolimus (FK506) potentiated aggregation of human platelets. Tacrolimus 113-123 protein phosphatase 3, catalytic subunit, beta isoform Mus musculus 79-85 25219476-5 2014 EtOH conditioning significantly increased, compared to alcohol-naive control mice, both IP3 R-1 protein and the release of dopamine in the NAcc of mice after 3 days of withdrawal from EtOH vapor inhaled for 4 days, and this increase of IP3 R-1 protein was completely abolished by intracerebroventricular injection of FK506, an inhibitor for calcineurin. Tacrolimus 317-322 inositol 1,4,5-trisphosphate receptor 1 Mus musculus 236-243 25330904-5 2014 Pharmacological inhibitors of the catalytic subunit of protein phosphatase 2B (PP2B-A) such as cyclosporine A or tacrolimus (FK506) potentiated aggregation of human platelets. Tacrolimus 125-130 protein phosphatase 3, catalytic subunit, beta isoform Mus musculus 79-85 25505617-0 2014 Analysis of FK506, timcodar (VX-853) and FKBP51 and FKBP52 chaperones in control of glucocorticoid receptor activity and phosphorylation. Tacrolimus 12-17 nuclear receptor subfamily 3, group C, member 1 Mus musculus 84-107 25284168-6 2014 However, a higher mean tacrolimus level, particularly >=10 ng/mL in the first three months after transplantation, was associated with a slower rate of decline in GFR with time (r = 0.608, p = 0.004) and with a less likelihood of developing CKD five yr after transplant. Tacrolimus 23-33 Rap guanine nucleotide exchange factor 5 Homo sapiens 165-168 25505617-6 2014 In wild-type (WT) MEF cells, timcodar, like FK506, potentiated dexamethasone-induced GR transcriptional activity at two endogenous genes. Tacrolimus 44-49 nuclear receptor subfamily 3, group C, member 1 Mus musculus 85-87 25505617-7 2014 Using 52KO and 51KO MEF cells, FK506 potentiated GR activity in 51KO cells but could not do so in 52KO cells, suggesting FKBP52 as the major target of FK506 action. Tacrolimus 31-36 nuclear receptor subfamily 3, group C, member 1 Mus musculus 49-51 25505617-7 2014 Using 52KO and 51KO MEF cells, FK506 potentiated GR activity in 51KO cells but could not do so in 52KO cells, suggesting FKBP52 as the major target of FK506 action. Tacrolimus 31-36 FK506 binding protein 4 Mus musculus 121-127 25505617-7 2014 Using 52KO and 51KO MEF cells, FK506 potentiated GR activity in 51KO cells but could not do so in 52KO cells, suggesting FKBP52 as the major target of FK506 action. Tacrolimus 151-156 FK506 binding protein 4 Mus musculus 121-127 25322286-3 2014 RESULTS: Fast metabolizers (CYP3A5*1/POR*28T carriers) had two-fold to three-fold higher tacrolimus dose requirements compared with slow metabolizers (CYP3A5*3/*3/CYP3A4*22 carriers) and needed significantly more time to achieve the target tacrolimus C0 of a minimum 10 ng/ml (3.3+-1.7 vs. 1.34+-0.75 days; P<0.0001). Tacrolimus 89-99 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 28-34 25322286-3 2014 RESULTS: Fast metabolizers (CYP3A5*1/POR*28T carriers) had two-fold to three-fold higher tacrolimus dose requirements compared with slow metabolizers (CYP3A5*3/*3/CYP3A4*22 carriers) and needed significantly more time to achieve the target tacrolimus C0 of a minimum 10 ng/ml (3.3+-1.7 vs. 1.34+-0.75 days; P<0.0001). Tacrolimus 89-99 cytochrome p450 oxidoreductase Homo sapiens 37-40 25322286-6 2014 Multivariate analyses identified the CYP3A5*1/POR*28/CYP3A4*22 genotype combination as the single strongest determinant of tacrolimus dose requirements throughout the first year, explaining between 24-40% of its variability, whereas recipient age, hematocrit, and delayed graft function were additional nongenetic determinants of tacrolimus dose. Tacrolimus 123-133 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 25322286-6 2014 Multivariate analyses identified the CYP3A5*1/POR*28/CYP3A4*22 genotype combination as the single strongest determinant of tacrolimus dose requirements throughout the first year, explaining between 24-40% of its variability, whereas recipient age, hematocrit, and delayed graft function were additional nongenetic determinants of tacrolimus dose. Tacrolimus 123-133 cytochrome p450 oxidoreductase Homo sapiens 46-49 25322286-6 2014 Multivariate analyses identified the CYP3A5*1/POR*28/CYP3A4*22 genotype combination as the single strongest determinant of tacrolimus dose requirements throughout the first year, explaining between 24-40% of its variability, whereas recipient age, hematocrit, and delayed graft function were additional nongenetic determinants of tacrolimus dose. Tacrolimus 123-133 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 53-59 25322286-6 2014 Multivariate analyses identified the CYP3A5*1/POR*28/CYP3A4*22 genotype combination as the single strongest determinant of tacrolimus dose requirements throughout the first year, explaining between 24-40% of its variability, whereas recipient age, hematocrit, and delayed graft function were additional nongenetic determinants of tacrolimus dose. Tacrolimus 330-340 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 25322286-7 2014 CONCLUSION: Combining the CYP3A5*1, POR*28 and CYP3A4*22 genotypes allows partial differentiation of early tacrolimus dose requirements and the time to reach therapeutic target concentrations after transplantation, but without obvious clinical implications. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 26-32 25322286-7 2014 CONCLUSION: Combining the CYP3A5*1, POR*28 and CYP3A4*22 genotypes allows partial differentiation of early tacrolimus dose requirements and the time to reach therapeutic target concentrations after transplantation, but without obvious clinical implications. Tacrolimus 107-117 cytochrome p450 oxidoreductase Homo sapiens 36-39 25322286-7 2014 CONCLUSION: Combining the CYP3A5*1, POR*28 and CYP3A4*22 genotypes allows partial differentiation of early tacrolimus dose requirements and the time to reach therapeutic target concentrations after transplantation, but without obvious clinical implications. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-53 25505617-1 2014 The immunosuppressive ligand FK506 and the FK506-binding protein FKBP52 are stimulatory to glucocorticoid receptor (GR) activity. Tacrolimus 29-34 nuclear receptor subfamily 3, group C, member 1 Mus musculus 91-114 25505617-1 2014 The immunosuppressive ligand FK506 and the FK506-binding protein FKBP52 are stimulatory to glucocorticoid receptor (GR) activity. Tacrolimus 29-34 nuclear receptor subfamily 3, group C, member 1 Mus musculus 116-118 25505617-2 2014 Here, we explore the underlying mechanism by comparing GR activity and phosphorylation status in response to FK506 and the novel nonimmunosuppressive ligand timcodar (VX-853) and in the presence and absence of FKBP52 and the closely related protein FKBP51. Tacrolimus 109-114 nuclear receptor subfamily 3, group C, member 1 Mus musculus 55-57 25561870-9 2014 Therefore, using Ferula asafetida with CYP3A4 drug substrates should be cautioned especially those with narrow therapeutic index such as cyclosporine, tacrolimus and carbamazepine. Tacrolimus 151-161 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 39-45 25521359-0 2014 Donor ABCB1 genetic polymorphisms influence epithelial-to-mesenchyme transition in tacrolimus-treated kidney recipients. Tacrolimus 83-93 ATP binding cassette subfamily B member 1 Homo sapiens 6-11 28346678-0 2014 CORRIGENDUM: Identification and Characterization of a Defective CYP3A4 Genotype in a Kidney Transplant Patient With Severely Diminished Tacrolimus Clearance. Tacrolimus 136-146 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 64-70 24739669-3 2014 We thus assessed the role of genetic variation in CYP3A4, CYP3A5, POR, NR1I2, and ABCB1 acting jointly in immunosuppressive drug pathways in tacrolimus (TAC) and ciclosporin (CSA) dose requirement in HTx recipients. Tacrolimus 141-151 cytochrome p450 oxidoreductase Homo sapiens 66-69 24739669-3 2014 We thus assessed the role of genetic variation in CYP3A4, CYP3A5, POR, NR1I2, and ABCB1 acting jointly in immunosuppressive drug pathways in tacrolimus (TAC) and ciclosporin (CSA) dose requirement in HTx recipients. Tacrolimus 141-151 nuclear receptor subfamily 1 group I member 2 Homo sapiens 71-76 24739669-3 2014 We thus assessed the role of genetic variation in CYP3A4, CYP3A5, POR, NR1I2, and ABCB1 acting jointly in immunosuppressive drug pathways in tacrolimus (TAC) and ciclosporin (CSA) dose requirement in HTx recipients. Tacrolimus 141-151 ATP binding cassette subfamily B member 1 Homo sapiens 82-87 24739669-0 2014 CYP3A5*3 and POR*28 genetic variants influence the required dose of tacrolimus in heart transplant recipients. Tacrolimus 68-78 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 24739669-0 2014 CYP3A5*3 and POR*28 genetic variants influence the required dose of tacrolimus in heart transplant recipients. Tacrolimus 68-78 cytochrome p450 oxidoreductase Homo sapiens 13-16 24739669-3 2014 We thus assessed the role of genetic variation in CYP3A4, CYP3A5, POR, NR1I2, and ABCB1 acting jointly in immunosuppressive drug pathways in tacrolimus (TAC) and ciclosporin (CSA) dose requirement in HTx recipients. Tacrolimus 141-151 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-56 25199528-1 2014 CONTEXT: Tacrolimus (TAC), a calcineurin inhibitor, is commonly used as an immunosuppressive agent in organ transplantation, but its clinical use may be limited due to cardiotoxicity. Tacrolimus 9-19 calcineurin binding protein 1 Rattus norvegicus 29-50 25255841-6 2014 EXPERT OPINION: Tacrolimus is a potent calcineurin inhibitor, an important pathway that regulates pancreatic development. Tacrolimus 16-26 calcineurin binding protein 1 Homo sapiens 39-60 25352392-1 2014 Extended-release tacrolimus (tacrolimus ER) [Advagraf( ); Astagraf XL( ); Graceptor( ); Prograf XL( )] is a once-daily formulation of the immunosuppressive calcineurin inhibitor, which is approved in many countries worldwide for the prophylaxis of transplant rejection in adult de novo kidney transplant recipients. Tacrolimus 17-27 calcineurin binding protein 1 Homo sapiens 156-177 25162201-5 2014 A potentially clinically actionable genetic variant exists in the CYP3A5 gene, with the initial tacrolimus dose selection being optimized based on CYP3A5 genotype. Tacrolimus 96-106 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 66-72 25162201-5 2014 A potentially clinically actionable genetic variant exists in the CYP3A5 gene, with the initial tacrolimus dose selection being optimized based on CYP3A5 genotype. Tacrolimus 96-106 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 147-153 25255841-7 2014 Tacrolimus can induce beta-cell apoptosis, decrease insulin exocytosis and reduce insulin gene transcription, which ultimately lead to impaired functional beta-cell mass after pancreas transplant. Tacrolimus 0-10 insulin Homo sapiens 52-59 25255841-8 2014 Furthermore, insulin resistance can exacerbate the diabetogenic effect of tacrolimus due to inhibition of insulin gene transcription and beta-cell proliferation. Tacrolimus 74-84 insulin Homo sapiens 13-20 25255841-8 2014 Furthermore, insulin resistance can exacerbate the diabetogenic effect of tacrolimus due to inhibition of insulin gene transcription and beta-cell proliferation. Tacrolimus 74-84 insulin Homo sapiens 106-113 25340655-1 2014 The effective calcineurin inhibitor (CNI) tacrolimus (Tac) is an integral part of the standard immunosuppressive regimen after renal transplantation (RTx). Tacrolimus 42-52 calcineurin binding protein 1 Homo sapiens 14-35 25445585-10 2014 FK506, a calcineurin inhibitor, blocked the decrease in expression of inactivated pSer637 Drp1 and mitochondrial fission. Tacrolimus 0-5 dynamin 1 like Homo sapiens 90-94 25219597-7 2014 We also investigated the effects of such immunosuppressants as a corticosteroid (dexamethasone) and tacrolimus (FK506) on TARC production, and used various signaling inhibitors to evaluate the signaling pathways involved in TARC expression. Tacrolimus 100-110 C-C motif chemokine ligand 17 Homo sapiens 122-126 25219597-7 2014 We also investigated the effects of such immunosuppressants as a corticosteroid (dexamethasone) and tacrolimus (FK506) on TARC production, and used various signaling inhibitors to evaluate the signaling pathways involved in TARC expression. Tacrolimus 112-117 C-C motif chemokine ligand 17 Homo sapiens 122-126 25156476-7 2014 The lower tacrolimus bioavailability with Limustin( ) was observed in both expressers and non-expressers of the functional CYP3A5 protein. Tacrolimus 10-20 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 123-129 25225668-5 2014 In addition, cyclophilin A increased Crk SH3 domain-binding guanine-nucleotide releasing factor (C3G) binding to CrkII, whereas inhibitors of immunophilins, such as cyclosporine A (CsA) and FK506, inhibited CrkII, but not CrkI association with C3G. Tacrolimus 190-195 CRK proto-oncogene, adaptor protein Homo sapiens 37-40 25329716-0 2014 Urinary neutrophil gelatinase-associated lipocalin: a useful biomarker for tacrolimus-induced acute kidney injury in liver transplant patients. Tacrolimus 75-85 lipocalin 2 Homo sapiens 8-50 25329716-11 2014 In addition, the urinary levels of NGAL at postoperative day 1 (p = 0.0446) and day 7 (p = 0.0006) can be a good predictive marker for tacrolimus-induced AKI within next 6 days, respectively. Tacrolimus 135-145 lipocalin 2 Homo sapiens 35-39 25329716-12 2014 In conclusion, urinary NGAL is a sensitive biomarker for tacrolimus-induced AKI, and may help predict renal event caused by tacrolimus therapy in liver transplant patients. Tacrolimus 57-67 lipocalin 2 Homo sapiens 23-27 25329716-12 2014 In conclusion, urinary NGAL is a sensitive biomarker for tacrolimus-induced AKI, and may help predict renal event caused by tacrolimus therapy in liver transplant patients. Tacrolimus 124-134 lipocalin 2 Homo sapiens 23-27 25225668-5 2014 In addition, cyclophilin A increased Crk SH3 domain-binding guanine-nucleotide releasing factor (C3G) binding to CrkII, whereas inhibitors of immunophilins, such as cyclosporine A (CsA) and FK506, inhibited CrkII, but not CrkI association with C3G. Tacrolimus 190-195 Rap guanine nucleotide exchange factor 1 Homo sapiens 97-100 25225668-6 2014 Expression in Jurkat T cells of phosphorylation indicator of Crk chimeric unit plasmid, a plasmid encoding the human CrkII1-236 sandwiched between cyan fluorescent protein and yellow fluorescent protein, demonstrated a basal level of fluorescence resonance energy transfer, which increased in response to cell treatment with CsA and FK506, reflecting increased trans-to-cis conversion of CrkII. Tacrolimus 333-338 CRK proto-oncogene, adaptor protein Homo sapiens 61-64 25225668-6 2014 Expression in Jurkat T cells of phosphorylation indicator of Crk chimeric unit plasmid, a plasmid encoding the human CrkII1-236 sandwiched between cyan fluorescent protein and yellow fluorescent protein, demonstrated a basal level of fluorescence resonance energy transfer, which increased in response to cell treatment with CsA and FK506, reflecting increased trans-to-cis conversion of CrkII. Tacrolimus 333-338 CRK proto-oncogene, adaptor protein Homo sapiens 117-122 25225668-10 2014 The present data demonstrate that immunophilins regulate CrkII, but not CrkI activity in T cells and suggest that CsA and FK506 inhibit selected effector T cell functions via a CrkII-dependent mechanism. Tacrolimus 122-127 CRK proto-oncogene, adaptor protein Homo sapiens 177-182 25310192-0 2014 Personalized tacrolimus dose requirement by CYP3A5 but not ABCB1 or ACE genotyping in both recipient and donor after pediatric liver transplantation. Tacrolimus 13-23 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 44-50 25314078-5 2014 Progression is dramatically reduced by pharmacological activation of BMP pathway activity with low-dose FK506, suggesting an approach to management of human bladder cancer. Tacrolimus 104-109 bone morphogenetic protein 1 Homo sapiens 69-72 25142246-8 2014 Moreover, strong and statistically significant associations were found between tacrolimus pharmacodynamic parameters and polymorphisms in the genes coding cyclophilin A, the calcineurin catalytic subunit alpha isoenzyme, and CD25. Tacrolimus 79-89 interleukin 2 receptor subunit alpha Homo sapiens 174-229 25115303-11 2014 Expression of the codon-harmonized full-length ABCB5 DNA conferred increased resistance, relative to the host yeast strain, to the putative substrates rhodamine 123, daunorubicin, tetramethylrhodamine, FK506, or clorgyline. Tacrolimus 202-207 ATP binding cassette subfamily B member 5 Homo sapiens 47-52 25004245-0 2014 Cyclosporine A and tacrolimus reduce the amount of GLUT4 at the cell surface in human adipocytes: increased endocytosis as a potential mechanism for the diabetogenic effects of immunosuppressive agents. Tacrolimus 19-29 solute carrier family 2 member 4 Homo sapiens 51-56 25125128-1 2014 BACKGROUND: Tacrolimus is a CYP3A4 inhibitor and can alter colchicine metabolism. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 28-34 25878789-0 2014 C1q nephropathy in the setting of granulomatosis with polyangiitis treated with tacrolimus. Tacrolimus 80-90 complement C1q A chain Homo sapiens 0-3 25004245-4 2014 Furthermore, we studied effects of CsA and tacrolimus on the regulation of cellular trafficking of GLUT4 in differentiated human preadipocytes and L6 cells. Tacrolimus 43-53 solute carrier family 2 member 4 Homo sapiens 99-104 25004245-6 2014 Although phosphorylation at Tyr1146 of the insulin receptor was inhibited by tacrolimus, the phosphorylation and/or protein levels of the insulin signaling proteins IRS1/2, p85-PI3K, PKB, AS160, and mTORC1, as well as GLUT4 and GLUT1, were unchanged by CsA or tacrolimus. Tacrolimus 77-87 insulin receptor Homo sapiens 43-59 25004245-6 2014 Although phosphorylation at Tyr1146 of the insulin receptor was inhibited by tacrolimus, the phosphorylation and/or protein levels of the insulin signaling proteins IRS1/2, p85-PI3K, PKB, AS160, and mTORC1, as well as GLUT4 and GLUT1, were unchanged by CsA or tacrolimus. Tacrolimus 260-270 insulin receptor Homo sapiens 43-59 25004245-7 2014 Furthermore, CsA and tacrolimus reduced the GLUT4 amount localized at the cell surface of differentiated human preadipocytes and L6 cells in the presence of insulin. Tacrolimus 21-31 solute carrier family 2 member 4 Homo sapiens 44-49 25004245-9 2014 CONCLUSIONS: These results suggest that therapeutic concentrations of CsA and tacrolimus can inhibit glucose uptake independent of insulin signaling by removing GLUT4 from the cell surface via an increased rate of endocytosis. Tacrolimus 78-88 solute carrier family 2 member 4 Homo sapiens 161-166 25339327-4 2014 RESULTS: Tacrolimus at concentration of 0.06 mumol/L could promote collagen induced platelet aggregation, inhibit thrombin induced platelet aggregation, have no effect on ristocetin and vWF induced platelet aggregation function. Tacrolimus 9-19 coagulation factor II, thrombin Homo sapiens 114-122 24875272-0 2014 Effect of CYP3A5 genotype, steroids, and azoles on tacrolimus in a pediatric renal transplant population. Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 24875272-1 2014 BACKGROUND: Numerous studies have described the impact of cytochrome P450 3A5 (CYP3A5) genotype on Tacrolimus (TAC) exposure. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 58-77 24875272-1 2014 BACKGROUND: Numerous studies have described the impact of cytochrome P450 3A5 (CYP3A5) genotype on Tacrolimus (TAC) exposure. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 79-85 25339327-5 2014 Tacrolimus at concentration of 120 mumol/L and 240 mumol/L could reduce the platelet mitochondrial membrane potential and induce the expression of apoptosis protein caspase-3. Tacrolimus 0-10 caspase 3 Homo sapiens 165-174 24954089-0 2014 FK506-loaded chitosan conduit promotes the regeneration of injured sciatic nerves in the rat through the upregulation of brain-derived neurotrophic factor and TrkB. Tacrolimus 0-5 brain-derived neurotrophic factor Rattus norvegicus 121-154 25226517-9 2014 The expression of NFAT2 and T-plastin in keratinocytes was decreased under FK506 treatment, suggesting that T-plastin plays a role in keratinocyte migration downstream to the calcineurin/NFAT pathway. Tacrolimus 75-80 nuclear factor of activated T cells 1 Homo sapiens 18-23 25226517-9 2014 The expression of NFAT2 and T-plastin in keratinocytes was decreased under FK506 treatment, suggesting that T-plastin plays a role in keratinocyte migration downstream to the calcineurin/NFAT pathway. Tacrolimus 75-80 plastin 3 Homo sapiens 28-37 25226517-9 2014 The expression of NFAT2 and T-plastin in keratinocytes was decreased under FK506 treatment, suggesting that T-plastin plays a role in keratinocyte migration downstream to the calcineurin/NFAT pathway. Tacrolimus 75-80 plastin 3 Homo sapiens 108-117 25226517-11 2014 Actin lamellipodia formation as well as FAK and beta6-integrin expression were also significantly decreased after treatment with FK506 or siRNA, reinforcing that NFAT2-dependent T-plastin expression plays a role in keratinocyte migration. Tacrolimus 129-134 nuclear factor of activated T cells 1 Homo sapiens 162-167 25226517-11 2014 Actin lamellipodia formation as well as FAK and beta6-integrin expression were also significantly decreased after treatment with FK506 or siRNA, reinforcing that NFAT2-dependent T-plastin expression plays a role in keratinocyte migration. Tacrolimus 129-134 plastin 3 Homo sapiens 178-187 24291098-6 2014 FK506-binding proteins 1a and 1b (FKBP1a/1b, also known as FKBP12/12.6) are immunophilin proteins that bind the immunosuppressant drugs FK506 and rapamycin. Tacrolimus 0-5 FKBP prolyl isomerase 1A Rattus norvegicus 59-65 24954089-0 2014 FK506-loaded chitosan conduit promotes the regeneration of injured sciatic nerves in the rat through the upregulation of brain-derived neurotrophic factor and TrkB. Tacrolimus 0-5 neurotrophic receptor tyrosine kinase 2 Rattus norvegicus 159-163 24954089-6 2014 BDNF and TrkB levels in motor neurons were highest in rats treated with FK506-loaded chitosan. Tacrolimus 72-77 brain-derived neurotrophic factor Rattus norvegicus 0-4 24954089-6 2014 BDNF and TrkB levels in motor neurons were highest in rats treated with FK506-loaded chitosan. Tacrolimus 72-77 neurotrophic receptor tyrosine kinase 2 Rattus norvegicus 9-13 25268342-3 2014 OBJECTIVE: This study investigated the relationship between serum TARC levels and the dosage of topical agents, including corticosteroids and/or tacrolimus, in patients with AD. Tacrolimus 145-155 C-C motif chemokine ligand 17 Homo sapiens 66-70 25268342-7 2014 One gram of strong rank steroid or the equivalent amount of steroid/tacrolimus is required to reduce serum TARC levels by 9.94 pg/mL weekly in moderate to severe AD patients. Tacrolimus 68-78 C-C motif chemokine ligand 17 Homo sapiens 107-111 24951257-10 2014 Oral tacrolimus could be used in patients failing anti-TNF therapy. Tacrolimus 5-15 tumor necrosis factor Homo sapiens 55-58 25279405-6 2014 Tacrolimus whole blood clearance/bioavailability standardized to haematocrit of 45% and fat free mass of 60 kg was estimated to be 16.1 l h-1 [95% CI 12.6, 18.0 l h-1]. Tacrolimus 0-10 H1.5 linker histone, cluster member Homo sapiens 138-141 25279405-6 2014 Tacrolimus whole blood clearance/bioavailability standardized to haematocrit of 45% and fat free mass of 60 kg was estimated to be 16.1 l h-1 [95% CI 12.6, 18.0 l h-1]. Tacrolimus 0-10 H1.5 linker histone, cluster member Homo sapiens 163-166 25279405-7 2014 Tacrolimus clearance was 30% higher (95% CI 13, 46%) and bioavailability 18% lower (95% CI 2, 29%) in CYP3A5 expressers compared with non-expressers. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 102-108 24682043-4 2014 Searching for mechanisms, we found that AMD3100 combined with low-dose tacrolimus mobilized increased number of lineage-negative c-Kit+, CD34+, and CD133+ stem cells. Tacrolimus 71-81 CD34 molecule Homo sapiens 137-141 24682043-4 2014 Searching for mechanisms, we found that AMD3100 combined with low-dose tacrolimus mobilized increased number of lineage-negative c-Kit+, CD34+, and CD133+ stem cells. Tacrolimus 71-81 prominin 1 Homo sapiens 148-153 24682043-5 2014 Low-dose tacrolimus also increased the number of SDF-1-bearing macrophages in the wound sites amplifying the "pull" of mobilized stem cells into the wound. Tacrolimus 9-19 C-X-C motif chemokine ligand 12 Homo sapiens 49-54 24996284-13 2014 This study showed that polymorphisms in KCNJ11 might predispose the patients treated by tacrolimus to development of NODAT after liver transplantation. Tacrolimus 88-98 potassium inwardly rectifying channel subfamily J member 11 Homo sapiens 40-46 24838101-12 2014 In vitro, cardiomyocytes depleted of RCAN1 were more sensitive to simulated I/R and the calcineurin inhibitor, FK506, restored protection. Tacrolimus 111-116 regulator of calcineurin 1 Mus musculus 37-42 24838101-12 2014 In vitro, cardiomyocytes depleted of RCAN1 were more sensitive to simulated I/R and the calcineurin inhibitor, FK506, restored protection. Tacrolimus 111-116 regulator of calcineurin 1 Mus musculus 88-109 25242754-1 2014 BACKGROUND: The single-nucleotide polymorphisms (SNPs) of the Multidrug resistance 1 (MDR1) gene have been associated with changes in the pharmacokinetics of cyclosporine (CsA) and tacrolimus (FK506). Tacrolimus 181-191 ATP binding cassette subfamily B member 1 Homo sapiens 62-84 25799800-13 2014 Topical Tacrolimus 0.1% ointment could be better option for the treatment of localized vitiligo when compared to topical placental extract but in combination with a steroid cream. Tacrolimus 8-18 VAMAS6 Homo sapiens 87-95 24973091-10 2014 By contrast, FK506 induced limited effects only at 25-50muM and did not alter bile canaliculi. Tacrolimus 13-18 latexin Homo sapiens 56-59 25014506-2 2014 Calcineurin inhibitor (CNI) use is considered to play a major role in the development of CKD after LT. We studied the influence of single-nucleotide polymorphisms (SNPs) in the genes of the donor and recipient CNI-metabolizing enzyme CYP3A5 and the CNI-transporting ABCB1 on the development of CKD after LT. MATERIALS AND METHODS: Tacrolimus (Tac) predose concentrations at different time-points after transplantation and the CYP3A5 6986A>G and ABCB1 3435C>T SNPs were determined in 125 LT recipients and their respective donors to study the influence of Tac predose levels and genetics on the development of CKD. Tacrolimus 331-341 calcineurin binding protein 1 Homo sapiens 0-21 25242754-1 2014 BACKGROUND: The single-nucleotide polymorphisms (SNPs) of the Multidrug resistance 1 (MDR1) gene have been associated with changes in the pharmacokinetics of cyclosporine (CsA) and tacrolimus (FK506). Tacrolimus 181-191 ATP binding cassette subfamily B member 1 Homo sapiens 86-90 25242754-1 2014 BACKGROUND: The single-nucleotide polymorphisms (SNPs) of the Multidrug resistance 1 (MDR1) gene have been associated with changes in the pharmacokinetics of cyclosporine (CsA) and tacrolimus (FK506). Tacrolimus 193-198 ATP binding cassette subfamily B member 1 Homo sapiens 62-84 24981811-14 2014 Tacrolimus dose was increased to 3 mg bid (9/9/2010), awaiting tacrolimus levels. Tacrolimus 0-10 BH3 interacting domain death agonist Homo sapiens 38-41 25242754-1 2014 BACKGROUND: The single-nucleotide polymorphisms (SNPs) of the Multidrug resistance 1 (MDR1) gene have been associated with changes in the pharmacokinetics of cyclosporine (CsA) and tacrolimus (FK506). Tacrolimus 193-198 ATP binding cassette subfamily B member 1 Homo sapiens 86-90 24981811-15 2014 Subsequent levels (ng/mL) were 8.6 and 9.5, which made us resume the prior tacrolimus dose (2 mg bid). Tacrolimus 75-85 BH3 interacting domain death agonist Homo sapiens 97-100 25122638-6 2014 Histopathological evaluation, including anti-graft antibodies and complement C3, revealed significantly reduced immune responses in the tacrolimus-hydrogel group compared with tacrolimus only. Tacrolimus 136-146 complement C3 Rattus norvegicus 66-79 25017243-1 2014 BACKGROUND: We report a unique case which quantifies the effect of molecular adsorbent recirculating system (MARS [Gambro, Sweden]) therapy on blood concentrations of tacrolimus in a patient treated for refractory pruritus associated with recurrent hepatitis C of the liver allograft. Tacrolimus 167-177 methionyl-tRNA synthetase 1 Homo sapiens 109-113 25017243-2 2014 Tacrolimus is a low-molecular-weight, highly protein-bound drug with the potential to be removed during MARS therapy. Tacrolimus 0-10 methionyl-tRNA synthetase 1 Homo sapiens 104-108 25118278-3 2014 Through insertion of a modified FK506 binding protein (insertable FKBP12, iFKBP) into the protein kinase isoforms Fyn, Src, Lyn, and Yes, we engineered kinase analogs that can be activated within minutes in living cells (RapR analogs). Tacrolimus 32-37 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 66-72 25118278-3 2014 Through insertion of a modified FK506 binding protein (insertable FKBP12, iFKBP) into the protein kinase isoforms Fyn, Src, Lyn, and Yes, we engineered kinase analogs that can be activated within minutes in living cells (RapR analogs). Tacrolimus 32-37 FYN proto-oncogene, Src family tyrosine kinase Homo sapiens 114-117 25118278-3 2014 Through insertion of a modified FK506 binding protein (insertable FKBP12, iFKBP) into the protein kinase isoforms Fyn, Src, Lyn, and Yes, we engineered kinase analogs that can be activated within minutes in living cells (RapR analogs). Tacrolimus 32-37 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 119-122 25118278-3 2014 Through insertion of a modified FK506 binding protein (insertable FKBP12, iFKBP) into the protein kinase isoforms Fyn, Src, Lyn, and Yes, we engineered kinase analogs that can be activated within minutes in living cells (RapR analogs). Tacrolimus 32-37 LYN proto-oncogene, Src family tyrosine kinase Homo sapiens 124-127 25165975-0 2014 The calcineurin inhibitor tacrolimus reduces proteinuria in membranous nephropathy accompanied by a decrease in angiopoietin-like-4. Tacrolimus 26-36 calcineurin binding protein 1 Homo sapiens 4-25 25165975-0 2014 The calcineurin inhibitor tacrolimus reduces proteinuria in membranous nephropathy accompanied by a decrease in angiopoietin-like-4. Tacrolimus 26-36 angiopoietin like 4 Homo sapiens 112-131 25165975-8 2014 However, tacrolimus treatment markedly reduced glomerular and urinary Angptl4, accompanied by a reduction in the established proteinuria and the promotion of podocyte repair. Tacrolimus 9-19 angiopoietin like 4 Homo sapiens 70-77 25165975-10 2014 In conclusion, this is the first demonstration that the calcineurin inhibitor tacrolimus can reduce Angptl4 in podocytes accompanied by a decrease in established proteinuria and promotion of podocyte repair in MN. Tacrolimus 78-88 calcineurin binding protein 1 Homo sapiens 56-77 25165975-10 2014 In conclusion, this is the first demonstration that the calcineurin inhibitor tacrolimus can reduce Angptl4 in podocytes accompanied by a decrease in established proteinuria and promotion of podocyte repair in MN. Tacrolimus 78-88 angiopoietin like 4 Homo sapiens 100-107 24811606-12 2014 EXPERT OPINION: Combination of future T(H)2-specific systemic treatment with optimal topical treatment with TCI, especially tacrolimus ointment, could help to completely control the skin inflammation in AD. Tacrolimus 124-134 latexin Homo sapiens 108-111 24447269-8 2014 The high-sensitivity C-reactive protein was decreased (tacrolimus group, 1 mo) and increased (cyclosporine group, 6 and 12 mo) after the kidney transplant. Tacrolimus 55-65 C-reactive protein Homo sapiens 21-39 24998444-6 2014 Rapamycin or FK506 treatments of the polyribosomes isolated from porcine brain, HeLa and K568 cells caused a residual release of the endogenous FKBP25, which suggests that the immunophilin also binds to some proteins via its PPIase cavity. Tacrolimus 13-18 FKBP prolyl isomerase 3 Homo sapiens 144-150 24998444-6 2014 Rapamycin or FK506 treatments of the polyribosomes isolated from porcine brain, HeLa and K568 cells caused a residual release of the endogenous FKBP25, which suggests that the immunophilin also binds to some proteins via its PPIase cavity. Tacrolimus 13-18 FKBP prolyl isomerase 3 Homo sapiens 225-231 24528196-0 2014 Relationship of CYP3A5 genotype and ABCB1 diplotype to tacrolimus disposition in Brazilian kidney transplant patients. Tacrolimus 55-65 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 16-22 24528196-0 2014 Relationship of CYP3A5 genotype and ABCB1 diplotype to tacrolimus disposition in Brazilian kidney transplant patients. Tacrolimus 55-65 ATP binding cassette subfamily B member 1 Homo sapiens 36-41 24820765-0 2014 Influence of TLR4 rs1927907 locus polymorphisms on tacrolimus pharmacokinetics in the early stage after liver transplantation. Tacrolimus 51-61 toll like receptor 4 Homo sapiens 13-17 24820765-1 2014 PURPOSE: The aim of this study was to assess the potential influence of single nucleotide polymorphisms (SNPs) in the TLR4 gene on tacrolimus pharmacokinetics in the early stage after liver transplantation. Tacrolimus 131-141 toll like receptor 4 Homo sapiens 118-122 24820765-9 2014 CONCLUSIONS: Collectively, donor TLR4 rs1927907 SNPs were closely associated with tacrolimus elimination in our Chinese Han patient population. Tacrolimus 82-92 toll like receptor 4 Homo sapiens 33-37 24820765-10 2014 The combination of the donor TLR4 rs1927907 SNP and both donor and recipient CYP3A5 rs776746 SNP might have a greater effect on tacrolimus elimination than each SNP separately. Tacrolimus 128-138 toll like receptor 4 Homo sapiens 29-33 24820765-10 2014 The combination of the donor TLR4 rs1927907 SNP and both donor and recipient CYP3A5 rs776746 SNP might have a greater effect on tacrolimus elimination than each SNP separately. Tacrolimus 128-138 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 77-83 24737844-4 2014 Drugs with the highest contribution of CYP3A5 included atazanavir, vincristine, midazolam, vardenafil, otenabant, verapamil, and tacrolimus, whereas 17 of the 32 tested showed negligible CYP3A5 contribution. Tacrolimus 129-139 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 39-45 24921414-0 2014 Impact of PPARA and POR polymorphisms on tacrolimus pharmacokinetics and new-onset diabetes in kidney transplant recipients. Tacrolimus 41-51 cytochrome p450 oxidoreductase Homo sapiens 20-23 24921414-2 2014 The aim of the current study was to investigate the impact of polymorphism of PPARA and POR genes on tacrolimus (TAC) dose-adjusted trough concentration and risk of new-onset diabetes after transplantation (NODAT). Tacrolimus 101-111 peroxisome proliferator activated receptor alpha Homo sapiens 78-83 24921414-2 2014 The aim of the current study was to investigate the impact of polymorphism of PPARA and POR genes on tacrolimus (TAC) dose-adjusted trough concentration and risk of new-onset diabetes after transplantation (NODAT). Tacrolimus 101-111 cytochrome p450 oxidoreductase Homo sapiens 88-91 24378577-0 2014 Effects of combinational CYP3A5 6986A>G polymorphism in graft liver and native intestine on the pharmacokinetics of tacrolimus in liver transplant patients: a meta-analysis. Tacrolimus 119-129 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 25-31 24378577-1 2014 BACKGROUND: Many studies have reported reduced tacrolimus dose-adjusted exposure in individuals expressing the CYP3A5*1 allele. Tacrolimus 47-57 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 111-117 24378577-3 2014 METHODS: Structured searches, of studies that evaluated the association between CYP3A5*1 allele and tacrolimus pharmacokinetics in adult liver transplant recipients, were conducted using Embase and Medline. Tacrolimus 100-110 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 80-86 24378577-7 2014 CONCLUSIONS: Results of the meta-analysis demonstrated that, in adult liver transplant patients, CYP3A5 expression in either the donor or recipient resulted in a need for a higher mean tacrolimus daily dose to achieve the target drug exposure. Tacrolimus 185-195 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 97-103 24378577-8 2014 In the immediate posttransplant period, recipient expression of a CYP3A5*1 allele seemed to have the greatest influence on tacrolimus pharmacokinetics with donor expression of a CYP3A5*1 allelle possibly becoming more important with increasing time after transplant. Tacrolimus 123-133 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 66-72 24912495-0 2014 Tacrolimus promotes hepatocellular carcinoma and enhances CXCR4/SDF-1alpha expression in vivo. Tacrolimus 0-10 C-X-C motif chemokine receptor 4 Rattus norvegicus 58-63 24912495-4 2014 In vitro, FK506 (100-1,000 microg/l) significantly promoted the proliferation of MH3924A cells (P<0.01), and increased the expression of CXCR4 in MH3924A cells, albeit with no significance (P>0.05). Tacrolimus 10-15 C-X-C motif chemokine receptor 4 Rattus norvegicus 140-145 24912495-8 2014 Our results suggest that FK506 promotes the proliferation of MH3924A cells and the expression of CXCR4 and SDF-1alpha in vivo. Tacrolimus 25-30 C-X-C motif chemokine receptor 4 Rattus norvegicus 97-102 24912495-9 2014 Therefore, inhibiting the formation of the CXCR4/SDF-1alpha complex may partly reduce the promoting effect of FK506 on HCC. Tacrolimus 110-115 C-X-C motif chemokine receptor 4 Rattus norvegicus 43-48 24959755-0 2014 Dipeptidyl peptidase IV inhibitor MK-0626 attenuates pancreatic islet injury in tacrolimus-induced diabetic rats. Tacrolimus 80-90 dipeptidylpeptidase 4 Rattus norvegicus 0-23 24836681-0 2014 Tolerogenic dendritic cells modified by tacrolimus suppress CD4(+) T-cell proliferation and inhibit collagen-induced arthritis in mice. Tacrolimus 40-50 CD4 antigen Mus musculus 60-63 24836681-3 2014 STUDY DESIGN AND METHODS: DCs derived from human monocytes were induced in vitro by GM-CSF/IL-4 with tacrolimus. Tacrolimus 101-111 colony stimulating factor 2 Homo sapiens 84-90 24836681-3 2014 STUDY DESIGN AND METHODS: DCs derived from human monocytes were induced in vitro by GM-CSF/IL-4 with tacrolimus. Tacrolimus 101-111 interleukin 4 Homo sapiens 91-95 24836681-8 2014 The expression of mRNA encoding IL-10 and TGF-beta increased after 12h of tacrolimus stimulation, with the strongest responses being observed after 24h. Tacrolimus 74-84 interleukin 10 Mus musculus 32-37 24836681-8 2014 The expression of mRNA encoding IL-10 and TGF-beta increased after 12h of tacrolimus stimulation, with the strongest responses being observed after 24h. Tacrolimus 74-84 transforming growth factor, beta 1 Mus musculus 42-50 24836681-14 2014 CONCLUSIONS: tDCs modified by tacrolimus suppressed CD4(+) T cell proliferation and inhibited collagen-induced arthritis. Tacrolimus 30-40 CD4 antigen Mus musculus 52-55 24911663-1 2014 OBJECTIVE: We investigated whether the cytochrome P450 3A5*3 (CYP3A5*3) genotype affects tacrolimus pharmacokinetics and the risk of acute cellular rejection in living-donor liver transplant patients in Japan. Tacrolimus 89-99 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 62-68 24911663-7 2014 Consequently, graft liver CYP3A5*1 genotype might increase the risk for acute cellular rejection after living-donor liver transplantation, possibly by associating with the local hepatic tacrolimus concentration. Tacrolimus 186-196 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 26-32 24911663-8 2014 CONCLUSIONS: The target level of tacrolimus may be affected by the CYP3A5*3 genotype of the liver, rather than by that of the small intestine, after postoperative day 14. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-73 24749623-0 2014 Differential conformational dynamics in the closely homologous FK506-binding domains of FKBP51 and FKBP52. Tacrolimus 63-68 FKBP prolyl isomerase 4 Homo sapiens 99-105 24658827-0 2014 The influence of CYP3A, PPARA, and POR genetic variants on the pharmacokinetics of tacrolimus and cyclosporine in renal transplant recipients. Tacrolimus 83-93 cytochrome p450 oxidoreductase Homo sapiens 35-38 24704664-12 2014 Tubular epithelial cell-induced CD8 degranulation and CD8-mediated TEC lysis were preferentially inhibited by tacrolimus and prednisolone, and not by everolimus. Tacrolimus 110-120 CD8a molecule Homo sapiens 32-35 24704664-12 2014 Tubular epithelial cell-induced CD8 degranulation and CD8-mediated TEC lysis were preferentially inhibited by tacrolimus and prednisolone, and not by everolimus. Tacrolimus 110-120 CD8a molecule Homo sapiens 54-57 24658827-1 2014 PURPOSE: Tacrolimus (Tac) and cyclosporine (CsA) are mainly metabolized by CYP3A4 and CYP3A5. Tacrolimus 9-19 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 75-81 24658827-1 2014 PURPOSE: Tacrolimus (Tac) and cyclosporine (CsA) are mainly metabolized by CYP3A4 and CYP3A5. Tacrolimus 9-19 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 86-92 25141896-6 2014 Time post-transplantation, recipient age, donor CYP3A5 and CYP3A4 genotypes and fluconazole administration significantly influenced tacrolimus apparent clearance while bodyweight influenced volume of distribution. Tacrolimus 132-142 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 59-65 24495112-8 2014 In vitro, tacrolimus suppressed Th1 and Th2 cells but not Th17 cells. Tacrolimus 10-20 negative elongation factor complex member C/D Homo sapiens 32-35 25035868-5 2014 Here, we found that the immunosuppressants FK506 and cyclosporin A, both known as calcineurin inhibitors, complemented the high-temperature stress-induced growth defect of rsp5(A401E) strain. Tacrolimus 43-48 NEDD4 family E3 ubiquitin-protein ligase Saccharomyces cerevisiae S288C 172-176 25473571-3 2014 Tacrolimus, a calcineurin inhibitor, is one potential therapy. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 14-35 24985179-6 2014 RESULTS: tDCs induced by tacrolimus exhibit a typical tolerogenic phenotype, whose level of costimulatory molecules CD80, CD83, CD86 and HLA-DR is (2.95 +- 1.32)%, (2.33 +- 1.60)%, (90.02 +- 7.42)% and (91.80 +- 6.18)%, respectively. Tacrolimus 25-35 CD80 molecule Homo sapiens 116-120 24861504-2 2014 This review focuses on the pharmacokinetic interactions and exposure-response relationships of mTOR inhibitors and tacrolimus (TAC), the most widely used CNI. Tacrolimus 115-125 mechanistic target of rapamycin kinase Homo sapiens 95-99 24935317-6 2014 The calcineurin inhibitor was tacrolimus in 98.9% of alemtuzumab patients, 98.2% of thymoglobulin patients, and 87.0% of the noninduced (P < .001). Tacrolimus 30-40 calcineurin binding protein 1 Homo sapiens 4-25 24985179-6 2014 RESULTS: tDCs induced by tacrolimus exhibit a typical tolerogenic phenotype, whose level of costimulatory molecules CD80, CD83, CD86 and HLA-DR is (2.95 +- 1.32)%, (2.33 +- 1.60)%, (90.02 +- 7.42)% and (91.80 +- 6.18)%, respectively. Tacrolimus 25-35 CD83 molecule Homo sapiens 122-126 24985179-6 2014 RESULTS: tDCs induced by tacrolimus exhibit a typical tolerogenic phenotype, whose level of costimulatory molecules CD80, CD83, CD86 and HLA-DR is (2.95 +- 1.32)%, (2.33 +- 1.60)%, (90.02 +- 7.42)% and (91.80 +- 6.18)%, respectively. Tacrolimus 25-35 CD86 molecule Homo sapiens 128-132 24816588-9 2014 Si-RNA mediated knock-down of Nox4 expression prevented up-regulation of procollagen alpha1(V) mRNA in tacrolimus-treated cells, but induced procollagen alpha1(V) expression in control cells. Tacrolimus 103-113 collagen type V alpha 1 chain Homo sapiens 85-94 24846380-6 2014 Furthermore, we observed sirolimus (i.e., rapamycin)-inducible interactions between FRB and FKBP12 and a dose-dependent abolishment of such interactions by FK506, the ligand of FKBP12. Tacrolimus 156-161 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 92-98 24846380-6 2014 Furthermore, we observed sirolimus (i.e., rapamycin)-inducible interactions between FRB and FKBP12 and a dose-dependent abolishment of such interactions by FK506, the ligand of FKBP12. Tacrolimus 156-161 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 177-183 24816588-0 2014 Tacrolimus increases Nox4 expression in human renal fibroblasts and induces fibrosis-related genes by aberrant TGF-beta receptor signalling. Tacrolimus 0-10 NADPH oxidase 4 Homo sapiens 21-25 24816588-0 2014 Tacrolimus increases Nox4 expression in human renal fibroblasts and induces fibrosis-related genes by aberrant TGF-beta receptor signalling. Tacrolimus 0-10 transforming growth factor beta 1 Homo sapiens 111-119 24816588-3 2014 In the human renal fibroblast cell line TK-173, the macrolide calcineurin inhibitor tacrolimus (FK-506) induced TGF-beta-like effects, manifested by increased expression of NAD(P)H-oxidase 4 (Nox4), transgelin, tropomyosin 1, and procollagen alpha1(V) mRNA after three days. Tacrolimus 84-94 transforming growth factor beta 1 Homo sapiens 112-120 24526611-3 2014 A physiologically based pharmacokinetic (PBPK) model of tacrolimus was proposed, taking into account the body weight, the proportion of fat (P(fat)), hematocrit, lipid fraction of organs, typical intrinsic clearance (CLi(typ)), CYP3A5 genotype of liver donor, plasma unbound fraction of tacrolimus (fu(p)), and concomitant drugs (CYP3A4 inhibitors). Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 330-336 24816588-3 2014 In the human renal fibroblast cell line TK-173, the macrolide calcineurin inhibitor tacrolimus (FK-506) induced TGF-beta-like effects, manifested by increased expression of NAD(P)H-oxidase 4 (Nox4), transgelin, tropomyosin 1, and procollagen alpha1(V) mRNA after three days. Tacrolimus 84-94 NADPH oxidase 4 Homo sapiens 173-190 24816588-3 2014 In the human renal fibroblast cell line TK-173, the macrolide calcineurin inhibitor tacrolimus (FK-506) induced TGF-beta-like effects, manifested by increased expression of NAD(P)H-oxidase 4 (Nox4), transgelin, tropomyosin 1, and procollagen alpha1(V) mRNA after three days. Tacrolimus 84-94 NADPH oxidase 4 Homo sapiens 192-196 24816588-3 2014 In the human renal fibroblast cell line TK-173, the macrolide calcineurin inhibitor tacrolimus (FK-506) induced TGF-beta-like effects, manifested by increased expression of NAD(P)H-oxidase 4 (Nox4), transgelin, tropomyosin 1, and procollagen alpha1(V) mRNA after three days. Tacrolimus 84-94 transgelin Homo sapiens 199-209 24816588-3 2014 In the human renal fibroblast cell line TK-173, the macrolide calcineurin inhibitor tacrolimus (FK-506) induced TGF-beta-like effects, manifested by increased expression of NAD(P)H-oxidase 4 (Nox4), transgelin, tropomyosin 1, and procollagen alpha1(V) mRNA after three days. Tacrolimus 84-94 collagen type V alpha 1 chain Homo sapiens 242-251 24816588-3 2014 In the human renal fibroblast cell line TK-173, the macrolide calcineurin inhibitor tacrolimus (FK-506) induced TGF-beta-like effects, manifested by increased expression of NAD(P)H-oxidase 4 (Nox4), transgelin, tropomyosin 1, and procollagen alpha1(V) mRNA after three days. Tacrolimus 96-102 transforming growth factor beta 1 Homo sapiens 112-120 24816588-3 2014 In the human renal fibroblast cell line TK-173, the macrolide calcineurin inhibitor tacrolimus (FK-506) induced TGF-beta-like effects, manifested by increased expression of NAD(P)H-oxidase 4 (Nox4), transgelin, tropomyosin 1, and procollagen alpha1(V) mRNA after three days. Tacrolimus 96-102 NADPH oxidase 4 Homo sapiens 173-190 24816588-3 2014 In the human renal fibroblast cell line TK-173, the macrolide calcineurin inhibitor tacrolimus (FK-506) induced TGF-beta-like effects, manifested by increased expression of NAD(P)H-oxidase 4 (Nox4), transgelin, tropomyosin 1, and procollagen alpha1(V) mRNA after three days. Tacrolimus 96-102 NADPH oxidase 4 Homo sapiens 192-196 24816588-3 2014 In the human renal fibroblast cell line TK-173, the macrolide calcineurin inhibitor tacrolimus (FK-506) induced TGF-beta-like effects, manifested by increased expression of NAD(P)H-oxidase 4 (Nox4), transgelin, tropomyosin 1, and procollagen alpha1(V) mRNA after three days. Tacrolimus 96-102 transgelin Homo sapiens 199-209 24816588-3 2014 In the human renal fibroblast cell line TK-173, the macrolide calcineurin inhibitor tacrolimus (FK-506) induced TGF-beta-like effects, manifested by increased expression of NAD(P)H-oxidase 4 (Nox4), transgelin, tropomyosin 1, and procollagen alpha1(V) mRNA after three days. Tacrolimus 96-102 collagen type V alpha 1 chain Homo sapiens 242-251 24816588-6 2014 The effects were independent of extracellular TGF-beta as confirmed by the use of neutralizing antibodies, and thus most likely caused by aberrant TGF-beta receptor signaling, where binding of tacrolimus to the regulatory FKBP12 protein results in a "leaky" TGF-beta receptor. Tacrolimus 193-203 transforming growth factor beta 1 Homo sapiens 147-155 24816588-6 2014 The effects were independent of extracellular TGF-beta as confirmed by the use of neutralizing antibodies, and thus most likely caused by aberrant TGF-beta receptor signaling, where binding of tacrolimus to the regulatory FKBP12 protein results in a "leaky" TGF-beta receptor. Tacrolimus 193-203 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 222-228 24816588-6 2014 The effects were independent of extracellular TGF-beta as confirmed by the use of neutralizing antibodies, and thus most likely caused by aberrant TGF-beta receptor signaling, where binding of tacrolimus to the regulatory FKBP12 protein results in a "leaky" TGF-beta receptor. Tacrolimus 193-203 transforming growth factor beta 1 Homo sapiens 147-155 24816588-8 2014 Tacrolimus- and TGF-beta1-induced Nox4 protein upregulation was confirmed by Western blotting, and was accompanied by a rise in intracellular H2O2 concentration. Tacrolimus 0-10 NADPH oxidase 4 Homo sapiens 34-38 24816588-9 2014 Si-RNA mediated knock-down of Nox4 expression prevented up-regulation of procollagen alpha1(V) mRNA in tacrolimus-treated cells, but induced procollagen alpha1(V) expression in control cells. Tacrolimus 103-113 NADPH oxidase 4 Homo sapiens 30-34 24526611-3 2014 A physiologically based pharmacokinetic (PBPK) model of tacrolimus was proposed, taking into account the body weight, the proportion of fat (P(fat)), hematocrit, lipid fraction of organs, typical intrinsic clearance (CLi(typ)), CYP3A5 genotype of liver donor, plasma unbound fraction of tacrolimus (fu(p)), and concomitant drugs (CYP3A4 inhibitors). Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 228-234 24526611-8 2014 Values for initial dosing regimen of tacrolimus in order to reach a C0 of 10 ng/ml at day 5 (assuming a constant dosing schedule) as a function of CYP3A5 donor genotype and patient"s hematocrit and body weight are proposed. Tacrolimus 37-47 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 147-153 24582967-0 2014 Recombinant human soluble thrombomodulin attenuates FK506-induced endothelial dysfunction through prevention of Akt inactivation. Tacrolimus 52-57 thrombomodulin Homo sapiens 26-40 24238261-11 2014 Post-operative days and CYP3A5 genotype were confirmed as critical factors of tacrolimus pharmacokinetics. Tacrolimus 78-88 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-30 24610957-1 2014 Clinically used calcineurin inhibitors, including tacrolimus (FK506) and cyclosporine A, can induce calcineurin inhibitor-induced pain syndrome (CIPS), which is characterized as severe pain and pain hypersensitivity. Tacrolimus 50-60 calcineurin binding protein 1 Rattus norvegicus 16-37 24610957-1 2014 Clinically used calcineurin inhibitors, including tacrolimus (FK506) and cyclosporine A, can induce calcineurin inhibitor-induced pain syndrome (CIPS), which is characterized as severe pain and pain hypersensitivity. Tacrolimus 62-67 calcineurin binding protein 1 Rattus norvegicus 16-37 24610957-4 2014 In this study, we determined whether spinal CK2 is involved in increased NMDAR activity and pain hypersensitivity caused by systemic administration of FK506 in rats. Tacrolimus 151-156 casein kinase 2 beta Rattus norvegicus 44-47 24610957-6 2014 CK2 inhibition with either 5,6-dichloro-1-beta-d-ribofuranosylbenzimidazole (DRB) or 4,5,6,7-tetrabromobenzotriazole (TBB) completely normalized the amplitude of evoked NMDAR-EPSCs of dorsal horn neurons in FK506-treated rats. Tacrolimus 207-212 casein kinase 2 beta Rattus norvegicus 0-3 24709525-0 2014 Decrease of immature B cell and interleukin-10 during early-post-transplant period in renal transplant recipients under tacrolimus based immunosuppression. Tacrolimus 120-130 interleukin 10 Homo sapiens 32-46 24680768-2 2014 An immediate-release oral formulation of tacrolimus has been commercially available since 1994 that is administered orally BID. Tacrolimus 41-51 BH3 interacting domain death agonist Homo sapiens 123-126 24512120-6 2014 In this study, we investigated the effect of FK506 on mesangial cells via TGF-beta and Smads signal pathways. Tacrolimus 45-50 transforming growth factor, beta 1 Mus musculus 74-82 24512120-7 2014 Our results shows that FK506 effectively blocked the TGF-beta/Smad signaling pathway by downregulation of TGF-beta receptor, and played an important role in TGF-beta1-induced Smad2 expression in mice mesangial cells. Tacrolimus 23-28 transforming growth factor, beta 1 Mus musculus 53-61 24512120-7 2014 Our results shows that FK506 effectively blocked the TGF-beta/Smad signaling pathway by downregulation of TGF-beta receptor, and played an important role in TGF-beta1-induced Smad2 expression in mice mesangial cells. Tacrolimus 23-28 transforming growth factor, beta 1 Mus musculus 106-114 24512120-7 2014 Our results shows that FK506 effectively blocked the TGF-beta/Smad signaling pathway by downregulation of TGF-beta receptor, and played an important role in TGF-beta1-induced Smad2 expression in mice mesangial cells. Tacrolimus 23-28 transforming growth factor, beta 1 Mus musculus 157-166 24512120-7 2014 Our results shows that FK506 effectively blocked the TGF-beta/Smad signaling pathway by downregulation of TGF-beta receptor, and played an important role in TGF-beta1-induced Smad2 expression in mice mesangial cells. Tacrolimus 23-28 SMAD family member 2 Mus musculus 175-180 24512120-8 2014 FK506 can inhibit the TGF-beta1-stimulated cell proliferation via Smad-related pathways. Tacrolimus 0-5 transforming growth factor, beta 1 Mus musculus 22-31 24582967-0 2014 Recombinant human soluble thrombomodulin attenuates FK506-induced endothelial dysfunction through prevention of Akt inactivation. Tacrolimus 52-57 AKT serine/threonine kinase 1 Homo sapiens 112-115 24462213-7 2014 Metabolism study in the human recombinant CYP 3A showed that these lignans had higher affinity to CYP3A than that of FK506, and thus had a stronger CYP3A-mediated metabolism. Tacrolimus 117-122 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 24582967-3 2014 We reported that FK506 induced endothelial dysfunction through inactivation of Akt and extracellular-regulated kinase 1/2 using a three-dimensional culture blood vessel model. Tacrolimus 17-22 AKT serine/threonine kinase 1 Homo sapiens 79-82 24462213-7 2014 Metabolism study in the human recombinant CYP 3A showed that these lignans had higher affinity to CYP3A than that of FK506, and thus had a stronger CYP3A-mediated metabolism. Tacrolimus 117-122 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 148-153 24582967-6 2014 rTM prevented FK506-induced inactivation of Akt, but not of extracellular-regulated kinase 1/2. Tacrolimus 14-19 AKT serine/threonine kinase 1 Homo sapiens 44-47 24582967-7 2014 Akt inhibition by LY294002 abrogated the preventive effect of rTM on FK506-induced Akt inactivation and the suppressive effect of rTM on FK506-induced cell death. Tacrolimus 69-74 AKT serine/threonine kinase 1 Homo sapiens 0-3 24462213-8 2014 It was concluded that the blood concentrations of these lignans were decreased and their CYP3A-mediated metabolisms were increased in the presence of FK506 since these lignans had higher affinity to CYP3A. Tacrolimus 150-155 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 89-94 24462213-8 2014 It was concluded that the blood concentrations of these lignans were decreased and their CYP3A-mediated metabolisms were increased in the presence of FK506 since these lignans had higher affinity to CYP3A. Tacrolimus 150-155 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 199-204 24582967-7 2014 Akt inhibition by LY294002 abrogated the preventive effect of rTM on FK506-induced Akt inactivation and the suppressive effect of rTM on FK506-induced cell death. Tacrolimus 69-74 AKT serine/threonine kinase 1 Homo sapiens 83-86 24582967-7 2014 Akt inhibition by LY294002 abrogated the preventive effect of rTM on FK506-induced Akt inactivation and the suppressive effect of rTM on FK506-induced cell death. Tacrolimus 137-142 AKT serine/threonine kinase 1 Homo sapiens 0-3 24582967-8 2014 These results suggest that rTM attenuates FK506-induced endothelial dysfunction through prevention of Akt inactivation. Tacrolimus 42-47 AKT serine/threonine kinase 1 Homo sapiens 102-105 24297734-1 2014 Fpr3 and Fpr4 of Saccharomyces cerevisiae are nuclear FK506-binding proteins each containing an extended acidic domain in addition to the conserved FK506-binding/peptidylprolyl isomerase (PPIase) domain. Tacrolimus 54-59 peptidylprolyl isomerase FPR3 Saccharomyces cerevisiae S288C 0-4 24707135-4 2014 Active IBD at the time of LT, discontinuation of 5-aminosalicylic acid or azathioprine at the time of LT and use of tacrolimus-based immunosuppression may be associated with an unfavorable outcome of IBD after LT. Anti-tumor necrosis factor alpha (TNFalpha) therapy for refractory IBD may be an effective and safe therapeutic option after LT. Tacrolimus 116-126 tumor necrosis factor Homo sapiens 248-256 24822242-1 2014 In this study, an in vitro experimental system for evaluating the inhibitory effect of investigational drugs on the P-glycoprotein (P-gp, MDR1)-mediated transport of tacrolimus (FK506) was developed using LLC-PK1-MDR1 and LLC-PK1 wild-type (control) cells. Tacrolimus 166-176 ATP-binding cassette, sub-family B (MDR/TAP), member 1 Sus scrofa 138-142 24822242-1 2014 In this study, an in vitro experimental system for evaluating the inhibitory effect of investigational drugs on the P-glycoprotein (P-gp, MDR1)-mediated transport of tacrolimus (FK506) was developed using LLC-PK1-MDR1 and LLC-PK1 wild-type (control) cells. Tacrolimus 166-176 ATP-binding cassette, sub-family B (MDR/TAP), member 1 Sus scrofa 213-217 24822242-1 2014 In this study, an in vitro experimental system for evaluating the inhibitory effect of investigational drugs on the P-glycoprotein (P-gp, MDR1)-mediated transport of tacrolimus (FK506) was developed using LLC-PK1-MDR1 and LLC-PK1 wild-type (control) cells. Tacrolimus 178-183 ATP-binding cassette, sub-family B (MDR/TAP), member 1 Sus scrofa 138-142 24822242-1 2014 In this study, an in vitro experimental system for evaluating the inhibitory effect of investigational drugs on the P-glycoprotein (P-gp, MDR1)-mediated transport of tacrolimus (FK506) was developed using LLC-PK1-MDR1 and LLC-PK1 wild-type (control) cells. Tacrolimus 178-183 ATP-binding cassette, sub-family B (MDR/TAP), member 1 Sus scrofa 213-217 24126681-0 2014 Identification and characterization of a defective CYP3A4 genotype in a kidney transplant patient with severely diminished tacrolimus clearance. Tacrolimus 123-133 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 51-57 24126681-7 2014 In addition, the patient harbored inactive CYP3A5*3, resulting in loss of function of the entire CYP3A locus, explaining the deteriorated tacrolimus clearance. Tacrolimus 138-148 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 43-49 24126681-7 2014 In addition, the patient harbored inactive CYP3A5*3, resulting in loss of function of the entire CYP3A locus, explaining the deteriorated tacrolimus clearance. Tacrolimus 138-148 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 43-48 24502894-9 2014 Administration of tacrolimus improved uterine GSH levels and CAT activity in the tacrolimus-treated groups. Tacrolimus 18-28 catalase Rattus norvegicus 61-64 24502894-9 2014 Administration of tacrolimus improved uterine GSH levels and CAT activity in the tacrolimus-treated groups. Tacrolimus 81-91 catalase Rattus norvegicus 61-64 24569454-6 2014 Furthermore, IL-7 inhibition in combination with T cell depletion synergized with either CTLA-4Ig administration or suboptimal doses of tacrolimus to induce long-term skin graft acceptance in this stringent transplant model. Tacrolimus 136-146 interleukin 7 Mus musculus 13-17 24297734-1 2014 Fpr3 and Fpr4 of Saccharomyces cerevisiae are nuclear FK506-binding proteins each containing an extended acidic domain in addition to the conserved FK506-binding/peptidylprolyl isomerase (PPIase) domain. Tacrolimus 54-59 peptidylprolyl isomerase FPR4 Saccharomyces cerevisiae S288C 9-13 24468714-8 2014 The expression of IL-6 and Ki67 was significantly increased in the insulin and tacrolimus groups compared to the portal stenosis group. Tacrolimus 79-89 interleukin 6 Rattus norvegicus 18-22 24864437-8 2014 Tear ECP levels were significantly decreased from 3493.6 (median) ng/ml at instillation initiation to 205.6 ng/ml at the first month after initiation of Tacrolimus treatment (p < 0.05, Steel test). Tacrolimus 153-163 ribonuclease A family member 3 Homo sapiens 5-8 24507957-3 2014 The calcineurin inhibitor tacrolimus is diabetogenic by inhibiting insulin secretion, whereas cyclosporine causes hypertension and increases cholesterol levels. Tacrolimus 26-36 insulin Homo sapiens 67-74 24089076-1 2014 BACKGROUND: Both age and CYP3A5 genotype are important determinants of tacrolimus disposition in pediatric kidney transplant recipients. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 25-31 24089076-3 2014 The authors aimed to evaluate the additional contribution of POR*28 to early tacrolimus disposition in pediatric kidney transplant recipients. Tacrolimus 77-87 cytochrome p450 oxidoreductase Homo sapiens 61-64 24089076-6 2014 RESULTS: CYP3A5 expressers carrying at least 1 POR*28 allele had on average 18.3% lower tacrolimus predose concentrations and 20.2% lower concentration/dose ratios compared with CYP3A5 expressers with POR*1/*1 genotype (P = 0.002 and P = 0.001, respectively). Tacrolimus 88-98 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 9-15 24089076-6 2014 RESULTS: CYP3A5 expressers carrying at least 1 POR*28 allele had on average 18.3% lower tacrolimus predose concentrations and 20.2% lower concentration/dose ratios compared with CYP3A5 expressers with POR*1/*1 genotype (P = 0.002 and P = 0.001, respectively). Tacrolimus 88-98 cytochrome p450 oxidoreductase Homo sapiens 47-50 24089076-8 2014 CONCLUSIONS: In this small cohort of pediatric kidney transplant recipients, POR*28 genotype seems to explain part of the variability found in tacrolimus disposition, in addition to age and CYP3A5 genotype. Tacrolimus 143-153 cytochrome p450 oxidoreductase Homo sapiens 77-80 24623856-3 2014 Here, we assess the impact of FK506-binding protein with a molecular mass of ~52 kDa (FKBP52), an immunophilin protein that interacts with physiological Tau, on Tau-P301L activity. Tacrolimus 30-35 FKBP prolyl isomerase 4 Homo sapiens 86-92 24623856-3 2014 Here, we assess the impact of FK506-binding protein with a molecular mass of ~52 kDa (FKBP52), an immunophilin protein that interacts with physiological Tau, on Tau-P301L activity. Tacrolimus 30-35 microtubule associated protein tau Homo sapiens 153-156 24623856-3 2014 Here, we assess the impact of FK506-binding protein with a molecular mass of ~52 kDa (FKBP52), an immunophilin protein that interacts with physiological Tau, on Tau-P301L activity. Tacrolimus 30-35 microtubule associated protein tau Homo sapiens 161-164 24593903-0 2014 Genetic polymorphisms in warfarin and tacrolimus-related genes VKORC1, CYP2C9 and CYP3A5 in the Greek-Cypriot population. Tacrolimus 38-48 vitamin K epoxide reductase complex subunit 1 Homo sapiens 63-69 24658440-5 2014 Recently, different authors have demonstrated that P-gp inhibitors, such as cyclosporine A (CsA) and its analogue Tacrolimus, are able to reduce P-gp expression and or function in SLE, RA and PsA patients. Tacrolimus 114-124 ATP binding cassette subfamily B member 1 Homo sapiens 51-55 24658440-5 2014 Recently, different authors have demonstrated that P-gp inhibitors, such as cyclosporine A (CsA) and its analogue Tacrolimus, are able to reduce P-gp expression and or function in SLE, RA and PsA patients. Tacrolimus 114-124 ATP binding cassette subfamily B member 1 Homo sapiens 145-149 24621983-0 2014 ABCB1 1199G>A genetic polymorphism (Rs2229109) influences the intracellular accumulation of tacrolimus in HEK293 and K562 recombinant cell lines. Tacrolimus 95-105 ATP binding cassette subfamily B member 1 Homo sapiens 0-5 24621983-1 2014 OBJECTIVE: ATP-binding cassette, subfamily B, member 1 (ABCB1) transporter, or P-glycoprotein, is an efflux protein implicated in the absorption and the distribution of various compounds, including tacrolimus and cyclosporine A. Tacrolimus 198-208 ATP binding cassette subfamily B member 1 Homo sapiens 11-54 24621983-1 2014 OBJECTIVE: ATP-binding cassette, subfamily B, member 1 (ABCB1) transporter, or P-glycoprotein, is an efflux protein implicated in the absorption and the distribution of various compounds, including tacrolimus and cyclosporine A. Tacrolimus 198-208 ATP binding cassette subfamily B member 1 Homo sapiens 56-61 24621983-1 2014 OBJECTIVE: ATP-binding cassette, subfamily B, member 1 (ABCB1) transporter, or P-glycoprotein, is an efflux protein implicated in the absorption and the distribution of various compounds, including tacrolimus and cyclosporine A. Tacrolimus 198-208 ATP binding cassette subfamily B member 1 Homo sapiens 79-93 24621983-2 2014 In vivo studies suggest an association between the ABCB1 1199G>A single nucleotide polymorphism (SNP) and tacrolimus intracellular accumulation. Tacrolimus 109-119 ATP binding cassette subfamily B member 1 Homo sapiens 51-56 24621983-5 2014 The impact of the 1199G>A SNP on ABCB1 activity towards rhodamine (Rh123), doxorubicin, vinblastine, tacrolimus and cyclosporine A was assessed by accumulation, cytotoxicity and/or kinetic experiments. Tacrolimus 104-114 ATP binding cassette subfamily B member 1 Homo sapiens 36-41 24621983-6 2014 RESULTS: Tacrolimus accumulation was strongly decreased in cells overexpressing the wild-type protein (1199G) compared to control cells, confirming the ability of ABCB1 to transport tacrolimus. Tacrolimus 9-19 ATP binding cassette subfamily B member 1 Homo sapiens 163-168 24621983-6 2014 RESULTS: Tacrolimus accumulation was strongly decreased in cells overexpressing the wild-type protein (1199G) compared to control cells, confirming the ability of ABCB1 to transport tacrolimus. Tacrolimus 182-192 ATP binding cassette subfamily B member 1 Homo sapiens 163-168 24621983-9 2014 CONCLUSION: ABCB1 encoded by the 1199G wild-type allele transports more efficiently tacrolimus in comparison to the 1199A variant protein. Tacrolimus 84-94 ATP binding cassette subfamily B member 1 Homo sapiens 12-17 24621983-10 2014 This observation indicates that the amino-acid substitution (Ser400Asn) encoded by the 1199A allele drastically decreases the ability of ABCB1 to drive the efflux of tacrolimus in a substrate-specific manner, in agreement with our previously published clinical data. Tacrolimus 166-176 ATP binding cassette subfamily B member 1 Homo sapiens 137-142 24593903-3 2014 Another variant in the P450 3A5 enzyme (CYP3A5) gene is known to affect the metabolism of many drugs, including tacrolimus. Tacrolimus 112-122 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 40-46 24593903-0 2014 Genetic polymorphisms in warfarin and tacrolimus-related genes VKORC1, CYP2C9 and CYP3A5 in the Greek-Cypriot population. Tacrolimus 38-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 24593903-0 2014 Genetic polymorphisms in warfarin and tacrolimus-related genes VKORC1, CYP2C9 and CYP3A5 in the Greek-Cypriot population. Tacrolimus 38-48 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 82-88 24533735-0 2014 A Chinese girl with novel PLCE1 mutations and proliferation of the mesangium responded to tacrolimus therapy. Tacrolimus 90-100 phospholipase C epsilon 1 Homo sapiens 26-31 24438215-0 2014 CYP3A5 genotypes affect tacrolimus pharmacokinetics and infectious complications in Chinese pediatric liver transplant patients. Tacrolimus 24-34 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 24499793-8 2014 This FKBP12-overexpression-induced effect was reverted by FK506. Tacrolimus 58-63 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 5-11 24279588-8 2014 On multivariate analyses, significant independent predictors for long-term impaired CD4 T-cell reconstitution were recipient age, pretransplant CD4+ T-cell count, 12-month CD4+ T-cell count, and tacrolimus or MMF therapy. Tacrolimus 195-205 CD4 molecule Homo sapiens 84-87 24522145-5 2014 CYP3A4*22 carriers showed a significant lower clearance for cyclosporine (-15%), and a trend was observed for everolimus (-7%) and tacrolimus (-16%). Tacrolimus 131-141 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 24701168-2 2014 Thus, the aim of this study was to investigate whether tacrolimus (FK506) has synergistic antitumor effects with doxorubicin on two human hepatocellular carcinoma cell lines, Huh7 and HepG2. Tacrolimus 55-65 MIR7-3 host gene Homo sapiens 175-179 24701168-2 2014 Thus, the aim of this study was to investigate whether tacrolimus (FK506) has synergistic antitumor effects with doxorubicin on two human hepatocellular carcinoma cell lines, Huh7 and HepG2. Tacrolimus 67-72 MIR7-3 host gene Homo sapiens 175-179 24342979-7 2014 RESULTS: Cyclosporine A and tacrolimus significantly reduced IFNgamma production in a dose-dependent manner (53%-83%), but showed minimal effect on degranulation (20%). Tacrolimus 28-38 interferon gamma Homo sapiens 61-69 24522145-6 2014 Patients carrying at least one CYP3A5*1 allele had 1.5-fold higher tacrolimus clearance compared with noncarriers; however, CYP3A5*3 appeared to be nonpredictive for everolimus and cyclosporine. Tacrolimus 67-77 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 31-37 23301526-0 2014 Tacrolimus abrogates TGF-beta1-induced type I collagen production in normal human fibroblasts through suppressing p38MAPK signalling pathway: implications on treatment of chronic atopic dermatitis lesions. Tacrolimus 0-10 transforming growth factor beta 1 Homo sapiens 21-30 24249597-1 2014 The calcineurin inhibitor tacrolimus is the backbone of immunosuppressive drug therapy after solid organ transplantation. Tacrolimus 26-36 calcineurin binding protein 1 Homo sapiens 4-25 24249597-4 2014 With regard to its pharmacokinetic variability, a single nucleotide polymorphism (SNP) in cytochrome P450 (CYP) 3A5 (6986A>G) has been consistently associated with tacrolimus dose requirement. Tacrolimus 167-177 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 90-115 24249597-6 2014 A randomised controlled study in kidney transplant recipients has demonstrated that a CYP3A5 genotype-based approach to tacrolimus dosing leads to more patients reaching the target concentration early after transplantation. Tacrolimus 120-130 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 86-92 24249597-8 2014 In addition to CYP3A5 genotype, other genetic variants may also contribute to the variability in tacrolimus pharmacokinetics. Tacrolimus 97-107 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 24249597-10 2014 Individuals carrying the CYP3A4*22 T-variant allele have a lower tacrolimus dose requirement than individuals with the CYP3A4*22 CC genotype and this effect appears to be independent of CYP3A5 genotype status. Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-31 24249597-10 2014 Individuals carrying the CYP3A4*22 T-variant allele have a lower tacrolimus dose requirement than individuals with the CYP3A4*22 CC genotype and this effect appears to be independent of CYP3A5 genotype status. Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 119-125 24249597-11 2014 Individuals carrying the POR*28 T-variant allele have a higher tacrolimus dose requirement than POR*28 CC homozygotes but this association was only found in CYP3A5-expressing individuals. Tacrolimus 63-73 cytochrome p450 oxidoreductase Homo sapiens 25-28 24249597-20 2014 The CYP3A5*1 SNP is currently the most promising biomarker for tailoring tacrolimus treatment. Tacrolimus 73-83 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 23301526-13 2014 The increase in phosphorylated p38 mitogen-activated protein kinase (p38MAPK) expression stimulated by TGF-beta1 was down-regulated by tacrolimus. Tacrolimus 135-145 transforming growth factor beta 1 Homo sapiens 103-112 23301526-15 2014 CONCLUSIONS: The present results demonstrated that tacrolimus significantly inhibited physiological functions of fibroblasts enhanced by TGF-beta1 in vitro. Tacrolimus 51-61 transforming growth factor beta 1 Homo sapiens 137-146 23301526-0 2014 Tacrolimus abrogates TGF-beta1-induced type I collagen production in normal human fibroblasts through suppressing p38MAPK signalling pathway: implications on treatment of chronic atopic dermatitis lesions. Tacrolimus 0-10 mitogen-activated protein kinase 14 Homo sapiens 114-121 23301526-4 2014 Tacrolimus (FK506) ointment has been reported to be effective for treating AD as well as some TGF-beta1-induced fibrotic diseases. Tacrolimus 0-10 transforming growth factor beta 1 Homo sapiens 94-103 23301526-4 2014 Tacrolimus (FK506) ointment has been reported to be effective for treating AD as well as some TGF-beta1-induced fibrotic diseases. Tacrolimus 12-17 transforming growth factor beta 1 Homo sapiens 94-103 23301526-5 2014 OBJECTIVES: To evaluate the effect of tacrolimus on TGF-beta1-stimulated cultured normal human dermal fibroblasts and explore the potential signalling pathways involved. Tacrolimus 38-48 transforming growth factor beta 1 Homo sapiens 52-61 23301526-9 2014 RESULTS: Our results revealed that the increased expressions of transforming growth factor-beta receptor I (TGF-betaRI) and TGF-betaRII in TGF-beta1-treated fibroblasts were suppressed by tacrolimus treatment. Tacrolimus 188-198 transforming growth factor beta receptor 1 Homo sapiens 64-106 23301526-9 2014 RESULTS: Our results revealed that the increased expressions of transforming growth factor-beta receptor I (TGF-betaRI) and TGF-betaRII in TGF-beta1-treated fibroblasts were suppressed by tacrolimus treatment. Tacrolimus 188-198 transforming growth factor beta receptor 1 Homo sapiens 108-118 23301526-9 2014 RESULTS: Our results revealed that the increased expressions of transforming growth factor-beta receptor I (TGF-betaRI) and TGF-betaRII in TGF-beta1-treated fibroblasts were suppressed by tacrolimus treatment. Tacrolimus 188-198 transforming growth factor beta receptor 2 Homo sapiens 124-135 23301526-9 2014 RESULTS: Our results revealed that the increased expressions of transforming growth factor-beta receptor I (TGF-betaRI) and TGF-betaRII in TGF-beta1-treated fibroblasts were suppressed by tacrolimus treatment. Tacrolimus 188-198 transforming growth factor beta 1 Homo sapiens 139-148 24352002-0 2014 Significant association between CYP3A5 polymorphism and blood concentration of tacrolimus in patients with connective tissue diseases. Tacrolimus 79-89 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 32-38 23301526-10 2014 In addition, tacrolimus significantly inhibited fibroblast proliferation enhanced by TGF-beta1. Tacrolimus 13-23 transforming growth factor beta 1 Homo sapiens 85-94 24352002-1 2014 Although the association between CYP3A5 polymorphism and blood concentration of tacrolimus (TAC) in patients with solid organ transplantation was established, whether the association is also true in patients with connective tissue disease (CTD) who usually receive small amount of TAC is uncertain. Tacrolimus 80-90 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 23301526-11 2014 TGF-beta1 increased type I collagen production, and this enhancing effect was suppressed by tacrolimus. Tacrolimus 92-102 transforming growth factor beta 1 Homo sapiens 0-9 23301526-12 2014 The down-regulation of MMP-1 and up-regulation of TIMP-1 induced by TGF-beta1 were reversed by tacrolimus. Tacrolimus 95-105 matrix metallopeptidase 1 Homo sapiens 23-28 23301526-12 2014 The down-regulation of MMP-1 and up-regulation of TIMP-1 induced by TGF-beta1 were reversed by tacrolimus. Tacrolimus 95-105 TIMP metallopeptidase inhibitor 1 Homo sapiens 50-56 23301526-12 2014 The down-regulation of MMP-1 and up-regulation of TIMP-1 induced by TGF-beta1 were reversed by tacrolimus. Tacrolimus 95-105 transforming growth factor beta 1 Homo sapiens 68-77 23301526-13 2014 The increase in phosphorylated p38 mitogen-activated protein kinase (p38MAPK) expression stimulated by TGF-beta1 was down-regulated by tacrolimus. Tacrolimus 135-145 mitogen-activated protein kinase 14 Homo sapiens 31-67 23301526-13 2014 The increase in phosphorylated p38 mitogen-activated protein kinase (p38MAPK) expression stimulated by TGF-beta1 was down-regulated by tacrolimus. Tacrolimus 135-145 mitogen-activated protein kinase 14 Homo sapiens 69-76 24089074-1 2014 OBJECTIVES: The aims of this study were to develop a population pharmacokinetic model of tacrolimus in adult kidney transplant recipients, to use this model to compare cytochrome P450 3A5 (CYP3A5) genotype-based initial dosing of tacrolimus with standard per-kilogram-based dosing, and to predict the best starting dose of tacrolimus based on patient genotype to achieve a trough concentration between 6 and 10 microg/L by day 5 posttransplantation. Tacrolimus 230-240 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 189-195 24444408-0 2014 CYP3A5 and CYP3A4, but not ABCB1 polymorphisms affect tacrolimus dose-adjusted trough concentrations in kidney transplant recipients. Tacrolimus 54-64 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 24444408-0 2014 CYP3A5 and CYP3A4, but not ABCB1 polymorphisms affect tacrolimus dose-adjusted trough concentrations in kidney transplant recipients. Tacrolimus 54-64 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 11-17 24089074-1 2014 OBJECTIVES: The aims of this study were to develop a population pharmacokinetic model of tacrolimus in adult kidney transplant recipients, to use this model to compare cytochrome P450 3A5 (CYP3A5) genotype-based initial dosing of tacrolimus with standard per-kilogram-based dosing, and to predict the best starting dose of tacrolimus based on patient genotype to achieve a trough concentration between 6 and 10 microg/L by day 5 posttransplantation. Tacrolimus 230-240 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 189-195 24089074-6 2014 Patient CYP3A5 genotype (rs776746), weight, hematocrit, and postoperative day were identified as significant covariates effecting tacrolimus apparent oral clearance (CL/F), with higher CL/F in CYP3A5*1 allele carriers, heavier patients, patients with low hematocrit, and in the immediate posttransplantation period. Tacrolimus 130-140 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 8-14 24089074-6 2014 Patient CYP3A5 genotype (rs776746), weight, hematocrit, and postoperative day were identified as significant covariates effecting tacrolimus apparent oral clearance (CL/F), with higher CL/F in CYP3A5*1 allele carriers, heavier patients, patients with low hematocrit, and in the immediate posttransplantation period. Tacrolimus 130-140 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 193-199 24089074-9 2014 To reduce the risk of under immunosuppression in the immediate posttransplantation period, carriers of a CYP3A5*1 allele are likely to benefit from a tacrolimus starting dose of either 10 mg or 0.115 mg/kg twice daily. Tacrolimus 150-160 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 105-111 24434619-8 2014 Moreover, FK506 binding proteins (FKBPs), enzymes with peptidyl-prolyl cis-trans isomerase activity, still accelerate the aggregation of phosphorylated alpha-SYN in vitro, as was shown previously for WT alpha-SYN. Tacrolimus 10-15 synemin Homo sapiens 158-161 24061445-0 2014 Impact of POR*28 on the pharmacokinetics of tacrolimus and cyclosporine A in renal transplant patients. Tacrolimus 44-54 cytochrome p450 oxidoreductase Homo sapiens 10-13 24061445-2 2014 Both CYP3A5 and CYP3A4 are involved in the metabolism of calcineurin inhibitors and recent data show that POR*28 may explain part of the variability observed in tacrolimus (Tac) pharmacokinetics. Tacrolimus 161-171 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 5-11 24061445-2 2014 Both CYP3A5 and CYP3A4 are involved in the metabolism of calcineurin inhibitors and recent data show that POR*28 may explain part of the variability observed in tacrolimus (Tac) pharmacokinetics. Tacrolimus 161-171 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 16-22 24061445-2 2014 Both CYP3A5 and CYP3A4 are involved in the metabolism of calcineurin inhibitors and recent data show that POR*28 may explain part of the variability observed in tacrolimus (Tac) pharmacokinetics. Tacrolimus 161-171 cytochrome p450 oxidoreductase Homo sapiens 106-109 24465960-0 2014 Impact of the CYP3A5, CYP3A4, COMT, IL-10 and POR genetic polymorphisms on tacrolimus metabolism in Chinese renal transplant recipients. Tacrolimus 75-85 cytochrome p450 oxidoreductase Homo sapiens 46-49 24465960-11 2014 CYP3A5*3, CYP3A4 *1G, CYP3A4 rs4646437 T>C and IL-10 rs1800871 C>T might be potential polymorphisms affecting the interindividual variability in tacrolimus metabolism among Chinese renal transplant recipients. Tacrolimus 151-161 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 24465960-11 2014 CYP3A5*3, CYP3A4 *1G, CYP3A4 rs4646437 T>C and IL-10 rs1800871 C>T might be potential polymorphisms affecting the interindividual variability in tacrolimus metabolism among Chinese renal transplant recipients. Tacrolimus 151-161 interleukin 10 Homo sapiens 50-55 24434520-7 2014 Inhibition of calcineurin by FK506 restored AMPK function and LC3 expression, and decreased the extent of apoptosis caused by prolonged oxidative stress. Tacrolimus 29-34 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 44-48 24434520-7 2014 Inhibition of calcineurin by FK506 restored AMPK function and LC3 expression, and decreased the extent of apoptosis caused by prolonged oxidative stress. Tacrolimus 29-34 microtubule associated protein 1 light chain 3 alpha Homo sapiens 62-65 24434520-10 2014 Furthermore, inhibition of autophagy by 3-methylanine (3-MA) or by knockdown of the essential autophagy-related gene ATG7 abrogated the protective effect of FK506. Tacrolimus 157-162 autophagy related 7 Homo sapiens 117-121 24434619-8 2014 Moreover, FK506 binding proteins (FKBPs), enzymes with peptidyl-prolyl cis-trans isomerase activity, still accelerate the aggregation of phosphorylated alpha-SYN in vitro, as was shown previously for WT alpha-SYN. Tacrolimus 10-15 synuclein alpha Homo sapiens 152-161 26579923-6 2014 The methodology is applied, with encouraging results, to the calculation of the absolute binding free energy of some FK506-related ligands of the peptidyl prolyl cis-trans isomerase protein (FKBP12) with known dissociation constants. Tacrolimus 117-122 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 191-197 24157471-0 2014 Substantial proliferation of human renal tubular epithelial cell-reactive CD4+CD28null memory T cells, which is resistant to tacrolimus and everolimus. Tacrolimus 125-135 CD4 molecule Homo sapiens 74-77 24157471-9 2014 TEC-reactive CD4 T-cell proliferation was significantly suppressed by tacrolimus, everolimus, prednisolone, and MPA (P<0.05). Tacrolimus 70-80 CD4 molecule Homo sapiens 13-16 24369269-3 2014 Before initiation of triple therapy, all patients switched from tacrolimus to cyclosporine, which has a lower inhibitory effect on CYP3A4 and CYP3A5 than tacrolimus. Tacrolimus 64-74 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 131-137 24369269-3 2014 Before initiation of triple therapy, all patients switched from tacrolimus to cyclosporine, which has a lower inhibitory effect on CYP3A4 and CYP3A5 than tacrolimus. Tacrolimus 64-74 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 142-148 24987695-1 2014 Tacrolimus, a calcineurin inhibitor, is clinically used as an immunosuppressive agent in organ transplantation, but its use is limited due to its marked nephrotoxicity. Tacrolimus 0-10 calcineurin binding protein 1 Rattus norvegicus 14-35 24987695-7 2014 Tacrolimus significantly decreased GSH level and catalase activity while increasing MDA level. Tacrolimus 0-10 catalase Rattus norvegicus 49-57 24987695-8 2014 Olmesartan also attenuated the effects of tacrolimus on MDA, GSH, and catalase. Tacrolimus 42-52 catalase Rattus norvegicus 70-78 25131236-12 2014 The expression of c-KIT mRNA level in A375 cells exposed to tacrolimus (10(3) and 10(4) nmol/L) had significantly increased by 3.03-fold and 3.19-fold respectively compared with the control (P < 0.05). Tacrolimus 60-70 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 18-23 23738951-6 2014 CONCLUSION: Tacrolimus apparent clearance was influenced by time post-transplantation and CYP3A5 genotypes. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 90-96 24383873-0 2014 Pharmacogenotyping of CYP3A5 in predicting dose-adjusted trough levels of tacrolimus among Malaysian kidney-transplant patients. Tacrolimus 74-84 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 22-28 24383873-2 2014 Seventy-eight kidney transplant patients treated with tacrolimus were recruited to study the correlation of dose adjusted trough level (level/dose; L/D) of tacrolimus with CYP3A5 and ABCB1 genotypes, as well as the mRNA copy number of ABCB1 in blood. Tacrolimus 156-166 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 172-178 24383873-4 2014 CYP3A5*3 genotypes were found to be a good predictor of tacrolimus L/D in kidney-transplant patients. Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 24383873-8 2014 However, in multiple regression analysis, only CYP3A5*3 genotype groups were found to be significantly correlated with tacrolimus L/D (P < 0.001). Tacrolimus 119-129 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 47-53 24383873-9 2014 These findings highlight the importance of CYP3A5*3 pharmacogenotyping among kidney-transplant patients treated with tacrolimus, and confirm the role of blood cell P-glycoprotein in influencing the L/D for tacrolimus. Tacrolimus 117-127 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 43-49 25316243-8 2014 First, FK506 binds FK506 binding protein 12 and its related isoform 12.6 (FKBP12/12.6) and removes them from intracellular ryanodine receptors that induce a calcium ion leakage from the endoplasmic/sarcoplasmic reticulum. Tacrolimus 7-12 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 74-80 25316243-8 2014 First, FK506 binds FK506 binding protein 12 and its related isoform 12.6 (FKBP12/12.6) and removes them from intracellular ryanodine receptors that induce a calcium ion leakage from the endoplasmic/sarcoplasmic reticulum. Tacrolimus 19-24 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 74-80 25131236-14 2014 The expression of c-KIT mRNA and protein increased dose-dependently in tacrolimus-treated groups as compared with the control. Tacrolimus 71-81 KIT proto-oncogene, receptor tyrosine kinase Homo sapiens 18-23 25316243-10 2014 Second, transforming growth factor receptor/SMAD2/3 signaling activation plays an important role in Treg/Th17 cell imbalance in T cells which toget converge to cause inflammation, endothelial dysfunction, and hypertension following tacrolimus treatment. Tacrolimus 232-242 SMAD family member 2 Homo sapiens 44-51 26281147-2 2014 We assessed the clinical value of high-dose tacrolimus on post-transplant HLA and MICA antibodies and proteinuria after renal transplantation. Tacrolimus 44-54 MHC class I polypeptide-related sequence A Homo sapiens 82-86 25316243-11 2014 Third, the activation of with-no-K(Lys) kinases/STE20/SPS1-related proline/alanine-rich kinase/thiazide-sensitive sodium chloride co-transporter (WNKs/SPAK/NCC) pathway has a central role in tacrolimus-induced hypertension. Tacrolimus 191-201 serine/threonine kinase 24 Homo sapiens 48-53 25316243-11 2014 Third, the activation of with-no-K(Lys) kinases/STE20/SPS1-related proline/alanine-rich kinase/thiazide-sensitive sodium chloride co-transporter (WNKs/SPAK/NCC) pathway has a central role in tacrolimus-induced hypertension. Tacrolimus 191-201 selenophosphate synthetase 1 Homo sapiens 54-58 25316243-11 2014 Third, the activation of with-no-K(Lys) kinases/STE20/SPS1-related proline/alanine-rich kinase/thiazide-sensitive sodium chloride co-transporter (WNKs/SPAK/NCC) pathway has a central role in tacrolimus-induced hypertension. Tacrolimus 191-201 serine/threonine kinase 39 Homo sapiens 151-155 24325305-2 2014 Our aim was to analyze the rate of CMV infection in HTx patients receiving treatment with cyclosporine (CsA) or tacrolimus (Tac). Tacrolimus 112-122 Zic family member 3 Homo sapiens 52-55 24325305-2 2014 Our aim was to analyze the rate of CMV infection in HTx patients receiving treatment with cyclosporine (CsA) or tacrolimus (Tac). Tacrolimus 124-127 Zic family member 3 Homo sapiens 52-55 26281147-9 2014 RESULTS: In the HLA or MICA Ab+ patients, proteinuria decreased after 5 years in the high tacrolimus Ab+ group unlike the low tacrolimus Ab+ group. Tacrolimus 90-100 MHC class I polypeptide-related sequence A Homo sapiens 23-27 26281147-11 2014 CONCLUSIONS: High-dose tacrolimus might play a role in improving allograft survival in HLA or MICA Ab+ post-transplant patients. Tacrolimus 23-33 MHC class I polypeptide-related sequence A Homo sapiens 94-98 23955548-0 2014 Association between CYP3A5 genotypes in graft liver and increase in tacrolimus biotransformation from steroid treatment in living-donor liver transplant patients. Tacrolimus 68-78 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 25531359-9 2014 In patients not responding to anti-TNF therapy, thalidomide or tacrolimus may be considered. Tacrolimus 63-73 tumor necrosis factor Homo sapiens 35-38 23955548-3 2014 In patients engrafted with a CYP3A5*1-carrying liver but not with a CYP3A5*3/*3-carrying liver, the concentration/dose ratio of tacrolimus significantly fell after therapy, while ratios of M-I/tacrolimus, M-II/tacrolimus, and M-III/tacrolimus were significantly higher after therapy than before (p = 0.032, p = 0.023, and p = 0.0078, respectively). Tacrolimus 128-138 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 29-35 23955548-4 2014 After steroid pulse therapy, the concentration of tacrolimus measured by immunoassay was significantly higher than that measured by LC-MS/MS in patients engrafted with a CYP3A5*1-carrying liver, but not those engrafted with a CYP3A5*3/*3-carrying liver. Tacrolimus 50-60 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 170-176 23955548-4 2014 After steroid pulse therapy, the concentration of tacrolimus measured by immunoassay was significantly higher than that measured by LC-MS/MS in patients engrafted with a CYP3A5*1-carrying liver, but not those engrafted with a CYP3A5*3/*3-carrying liver. Tacrolimus 50-60 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 226-232 23955548-5 2014 This suggests that the increased ratio of tacrolimus metabolites/tacrolimus can be explained by induction of CYP3A5 via high-dose steroid pulse therapy. Tacrolimus 42-52 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 109-115 23955548-5 2014 This suggests that the increased ratio of tacrolimus metabolites/tacrolimus can be explained by induction of CYP3A5 via high-dose steroid pulse therapy. Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 109-115 24351870-0 2014 Association of hemoglobin levels, CYP3A5, and NR1I3 gene polymorphisms with tacrolimus pharmacokinetics in liver transplant patients. Tacrolimus 76-86 nuclear receptor subfamily 1 group I member 3 Homo sapiens 46-51 23955548-6 2014 Further, the concentrations of tacrolimus measured by the immunoassays were overestimated, partly because of cross-reactivity of the monoclonal antibody they incorporated to detect tacrolimus, with the increased metabolites in patients with a CYP3A5*1-carrying graft liver. Tacrolimus 31-41 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 243-249 24351870-3 2014 This study was undertaken to determine the association of clinical markers, cytochrome P450, family 3, subfamily A, polypeptide 5 (CYP3A5) and nuclear receptor subfamily 1, group I, member 3 (NR1I3) gene polymorphisms with tacrolimus pharmacokinetics. Tacrolimus 223-233 nuclear receptor subfamily 1 group I member 3 Homo sapiens 143-190 24674680-8 2014 Tacrolimus significantly inhibited cell migration, pulmonary edema, exudation, CRP and hyperglycemia. Tacrolimus 0-10 C-reactive protein, pentraxin-related Mus musculus 79-82 24351870-7 2014 In single variable analysis, hemoglobin (Hb), hematocrit (Hct), donor CYP3A5, NR1I3 gene polymorphisms and recipient CYP3A5 gene polymorphisms were associated with log-transformed tacrolimus C/D ratios. Tacrolimus 180-190 nuclear receptor subfamily 1 group I member 3 Homo sapiens 78-83 24351870-7 2014 In single variable analysis, hemoglobin (Hb), hematocrit (Hct), donor CYP3A5, NR1I3 gene polymorphisms and recipient CYP3A5 gene polymorphisms were associated with log-transformed tacrolimus C/D ratios. Tacrolimus 180-190 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 117-123 24351870-8 2014 Hb, donor CYP3A5, NR1I3 gene polymorphisms and recipient CYP3A5 gene polymorphisms showed association with log-transformed tacrolimus C/D ratios in the final multiple linear regression model. Tacrolimus 123-133 nuclear receptor subfamily 1 group I member 3 Homo sapiens 18-23 24351870-8 2014 Hb, donor CYP3A5, NR1I3 gene polymorphisms and recipient CYP3A5 gene polymorphisms showed association with log-transformed tacrolimus C/D ratios in the final multiple linear regression model. Tacrolimus 123-133 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 57-63 24351870-9 2014 Donor CYP3A5 polymorphisms were the most important variant, accounting for 14.3% of total variation involved in tacrolimus pharmacokinetics. Tacrolimus 112-122 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 6-12 24071959-11 2014 Fat-free mass, CYP3A5 genotype, sex, age and time after transplant influence the tacrolimus individual dose requirement. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 25268107-4 2014 MATERIALS AND METHODS: In the present study, we first detected the serum and renal ET-1 level of rats treated by tacrolimus and found strong positive correlations were existed between the ET-1 level and the tacrolimus dosage and treated time. Tacrolimus 113-123 endothelin 1 Rattus norvegicus 83-87 25268107-4 2014 MATERIALS AND METHODS: In the present study, we first detected the serum and renal ET-1 level of rats treated by tacrolimus and found strong positive correlations were existed between the ET-1 level and the tacrolimus dosage and treated time. Tacrolimus 113-123 endothelin 1 Rattus norvegicus 188-192 25268107-4 2014 MATERIALS AND METHODS: In the present study, we first detected the serum and renal ET-1 level of rats treated by tacrolimus and found strong positive correlations were existed between the ET-1 level and the tacrolimus dosage and treated time. Tacrolimus 207-217 endothelin 1 Rattus norvegicus 188-192 24899909-10 2014 In addition, the purple anthocyanins, such as cyanidin-3-glucoside and malvidin-3-glucoside, might be better than FK506 in regulating FKBP52 and treating Alzheimer"s disease. Tacrolimus 114-119 FKBP prolyl isomerase 4 Homo sapiens 134-140 24076965-10 2014 FK506 treatment prior to IR prevented Drp1-S637 dephosphorylation and preserved cardiac function. Tacrolimus 0-5 dynamin 1-like Rattus norvegicus 38-42 24734081-12 2014 CL/F, Vd/F, and t1/2beta of tacrolimus in this study were comparable to reported values from Italian heart transplant patients but somewhat different from reported ones from other solid organ transplant populations. Tacrolimus 28-38 interleukin 1 receptor like 1 Homo sapiens 16-24 24081160-8 2014 Furthermore, blocking GluN2A or GluN2B subunits similarly reduced the amplitude of evoked EPSCs and the frequency of miniature EPSCs in dorsal horn neurons of FK506-treated rats. Tacrolimus 159-164 glutamate ionotropic receptor NMDA type subunit 2A Rattus norvegicus 22-28 24033383-0 2014 Impact of CYP3A5 genetic polymorphisms on the pharmacokinetics and short-term remission in patients with ulcerative colitis treated with tacrolimus. Tacrolimus 137-147 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 24033383-1 2014 BACKGROUND AND AIM: The pharmacokinetics of tacrolimus (Tac) differ among individuals, and genetic polymorphisms of cytochrome P-450 (CYP) 3A4, CYP3A5, and ABCB1 are thought to be involved. Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-142 24033383-1 2014 BACKGROUND AND AIM: The pharmacokinetics of tacrolimus (Tac) differ among individuals, and genetic polymorphisms of cytochrome P-450 (CYP) 3A4, CYP3A5, and ABCB1 are thought to be involved. Tacrolimus 44-54 ATP binding cassette subfamily B member 1 Homo sapiens 156-161 24081160-8 2014 Furthermore, blocking GluN2A or GluN2B subunits similarly reduced the amplitude of evoked EPSCs and the frequency of miniature EPSCs in dorsal horn neurons of FK506-treated rats. Tacrolimus 159-164 glutamate ionotropic receptor NMDA type subunit 2B Rattus norvegicus 32-38 24656015-0 2014 Influence of cytochrome P450 3A5 polymorphisms on viral infection incidence in kidney transplant patients treated with tacrolimus. Tacrolimus 119-129 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-32 24934555-11 2014 The blood concentration of tacrolimus, a P-gp substrate, was lower during 0-20 min but was higher during 40-90 min post-administration compared with that in the sham-operated rats. Tacrolimus 27-37 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 41-45 24934555-12 2014 P-gp expression in the ileum was decreased at 6 h after I/R-15, due to abnormal localization of P-gp, resulting in a high blood tacrolimus concentration in rats reperfused for 6 h. CONCLUSIONS: ROS multimodally regulate P-gp expression depending on its amount. Tacrolimus 128-138 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 0-4 24934555-12 2014 P-gp expression in the ileum was decreased at 6 h after I/R-15, due to abnormal localization of P-gp, resulting in a high blood tacrolimus concentration in rats reperfused for 6 h. CONCLUSIONS: ROS multimodally regulate P-gp expression depending on its amount. Tacrolimus 128-138 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 96-100 24934555-12 2014 P-gp expression in the ileum was decreased at 6 h after I/R-15, due to abnormal localization of P-gp, resulting in a high blood tacrolimus concentration in rats reperfused for 6 h. CONCLUSIONS: ROS multimodally regulate P-gp expression depending on its amount. Tacrolimus 128-138 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 96-100 24463861-6 2014 It has been proposed that transplanted patients could receive an initial Tacrolimus dose based on the CYP3A5 genotype. Tacrolimus 73-83 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 102-108 24655974-0 2014 Reduced variability of tacrolimus trough level in once-daily tacrolimus-based Taiwanese kidney transplant recipients with high-expressive genotype of cytochrome P450 3A5. Tacrolimus 23-33 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 150-169 24655974-0 2014 Reduced variability of tacrolimus trough level in once-daily tacrolimus-based Taiwanese kidney transplant recipients with high-expressive genotype of cytochrome P450 3A5. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 150-169 24655974-3 2014 We aimed to clarify the impact of cytochrome P450 3A5 genetic polymorphism on the variability of tacrolimus blood trough level in Taiwanese kidney transplant recipients. Tacrolimus 97-107 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-53 25036063-3 2014 The predictive value of the evolution of anti-PLA2R antibody titre on the clinical response of idiopathic membranous nephropathy patients treated with tacrolimus is analysed in the following study. Tacrolimus 151-161 phospholipase A2 receptor 1 Homo sapiens 46-51 24656015-10 2014 Recipients with the CYP3A5 3/3 genotype (nonexpressors) showed significantly higher dose-adjusted tacrolimus trough concentrations than patients with the CYP3A5 1 allele (expressors; respectively, 104.6 +- 65.6 vs 52.6 +- 62.3 ng/mL per mg/kg/d). Tacrolimus 98-108 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 24656015-11 2014 CONCLUSIONS: The CYP3A5 1 allele is associated with viral infection, possibly as a result of higher peak concentrations of tacrolimus. Tacrolimus 123-133 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 17-23 24656020-1 2014 BACKGROUND: The bioavailability of oral tacrolimus is influenced by enterocyte metabolism, which involves CYP3A and P-glycoprotein. Tacrolimus 40-50 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 106-111 24656020-1 2014 BACKGROUND: The bioavailability of oral tacrolimus is influenced by enterocyte metabolism, which involves CYP3A and P-glycoprotein. Tacrolimus 40-50 ATP binding cassette subfamily B member 1 Homo sapiens 116-130 23528073-9 2013 CONCLUSIONS: Differences between maternal and umbilical cord tacrolimus concentrations may be explained in part by placental P-gp function, greater red blood cell partitioning and higher haematocrit levels in venous cord blood. Tacrolimus 61-71 phosphoglycolate phosphatase Homo sapiens 125-129 24392311-5 2013 Potential improvements to the calcineurin inhibitor class include a prolonged release tacrolimus formulation and voclosporin, a cyclosporine analog. Tacrolimus 86-96 calcineurin binding protein 1 Homo sapiens 30-51 24119294-7 2013 Use of specific immunosuppressive medications was variably associated with incidence; for example, tacrolimus, was associated with reduced incidence for some anogenital cancers (IRRs 0.4-0.7) but increased incidence of oropharyngeal cancer (IRR 2.1). Tacrolimus 99-109 insulin receptor related receptor Homo sapiens 178-181 24409044-7 2013 At present, research has been able to reliably show that the CYP3A5 genotype, but not the CYP3A4 or ABCB1 ones, can modify the pharmacokinetics of tacrolimus. Tacrolimus 147-157 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-67 24042126-0 2013 The effect of ABCB1 C3435T polymorphism on pharmacokinetics of tacrolimus in liver transplantation: a meta-analysis. Tacrolimus 63-73 ATP binding cassette subfamily B member 1 Homo sapiens 14-19 24136923-6 2013 In this study, a rapid, quantitative, image cytometry-based measurement of nuclear translocation of NFAT1 is used to evaluate NFAT activation in T cells and its tacrolimus-induced inhibition. Tacrolimus 161-171 nuclear factor of activated T cells 2 Homo sapiens 100-105 24136923-7 2013 A strong dose-dependent correlation between NFAT1 inhibition and tacrolimus dose is demonstrated in vitro. Tacrolimus 65-75 nuclear factor of activated T cells 2 Homo sapiens 44-49 24136923-8 2013 Time kinetic analysis of NFAT1 inhibition in plasma from stable renal transplant recipients before and after an in vivo dose with tacrolimus correlated with the expected pharmacokinetic profile of tacrolimus. Tacrolimus 130-140 nuclear factor of activated T cells 2 Homo sapiens 25-30 24136923-8 2013 Time kinetic analysis of NFAT1 inhibition in plasma from stable renal transplant recipients before and after an in vivo dose with tacrolimus correlated with the expected pharmacokinetic profile of tacrolimus. Tacrolimus 197-207 nuclear factor of activated T cells 2 Homo sapiens 25-30 24042126-1 2013 OBJECTIVES: The effect of ABCB1 C3435T SNP on the pharmacokinetics of immunosuppressive drug tacrolimus in different studies was conflicting. Tacrolimus 93-103 ATP binding cassette subfamily B member 1 Homo sapiens 26-31 24042126-2 2013 So a meta-analysis was employed to study the correlation of ABCB1 C3435T SNP and the pharmacokinetics of tacrolimus at different post-transplantation times. Tacrolimus 105-115 ATP binding cassette subfamily B member 1 Homo sapiens 60-65 24042126-3 2013 METHOD: Several studies about ABCB1 C3435T polymorphism and the pharmacokinetics of tacrolimus were collected through the search on PubMed and the Cochrane Library. Tacrolimus 84-94 ATP binding cassette subfamily B member 1 Homo sapiens 30-35 24042126-10 2013 CONCLUSION: Through this meta-analysis for the including studies about the pharmacokinetics of tacrolimus and ABCB1 C3435T SNP, several significant associations were obtained. Tacrolimus 95-105 ATP binding cassette subfamily B member 1 Homo sapiens 110-115 24118301-0 2013 Inclusion of CYP3A5 genotyping in a nonparametric population model improves dosing of tacrolimus early after transplantation. Tacrolimus 86-96 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 24136923-10 2013 This is the first report to show that the measurement of NFAT1 activation potential by nuclear translocation can be used as a direct, sensitive, reproducible and quantitative pharmacodynamic readout for tacrolimus action. Tacrolimus 203-213 nuclear factor of activated T cells 2 Homo sapiens 57-62 24263023-1 2013 BACKGROUND: We investigated cardiac proinflammatory, mitogenic, and apoptotic signaling events, and plasma biomarkers of inflammation and oxidative stress in de novo adult cardiac transplant (CTX) patients receiving tacrolimus (TAC) or cyclosporine A (CsA). Tacrolimus 216-226 cytochrome P450 family 27 subfamily A member 1 Homo sapiens 192-195 24113216-0 2013 Single-nucleotide polymorphisms in P450 oxidoreductase and peroxisome proliferator-activated receptor-alpha are associated with the development of new-onset diabetes after transplantation in kidney transplant recipients treated with tacrolimus. Tacrolimus 233-243 cytochrome p450 oxidoreductase Homo sapiens 35-54 24113216-14 2013 CONCLUSION: This candidate-gene study shows that polymorphisms in PPARalpha and POR might predispose patients being treated with tacrolimus to the development of NODAT after kidney transplantation. Tacrolimus 129-139 peroxisome proliferator activated receptor alpha Homo sapiens 66-75 24113216-14 2013 CONCLUSION: This candidate-gene study shows that polymorphisms in PPARalpha and POR might predispose patients being treated with tacrolimus to the development of NODAT after kidney transplantation. Tacrolimus 129-139 cytochrome p450 oxidoreductase Homo sapiens 80-83 24041534-8 2013 Moreover, TNF-alpha significantly enhanced apoptotic cell death induced by cisplatin, cyclosporine A, tacrolimus and azidothymidine. Tacrolimus 102-112 tumor necrosis factor Mus musculus 10-19 24337847-1 2013 The effects of tacrolimus postconditioning on protein-serine-threonine kinases (Akt) phosphorylation and apoptotic cell death in rats after spinal cord ischemia-reperfusion injury were investigated. Tacrolimus 15-25 AKT serine/threonine kinase 1 Rattus norvegicus 80-83 24337847-9 2013 The Akt activities reached the peak at 15 min after reperfusion in ischemia-reperfusion group and tacrolimus postconditioning group. Tacrolimus 98-108 AKT serine/threonine kinase 1 Rattus norvegicus 4-7 24337847-11 2013 It is concluded that tacrolimus post-conditioning can increase Akt activity in spinal cord tissue of rats, inhibit apoptosis of neural cells as well as tissue edema, and thereby alleviate spinal cord ischemia-reperfusion injury. Tacrolimus 21-31 AKT serine/threonine kinase 1 Rattus norvegicus 63-66 24036158-7 2013 With regard to the apical efflux transporters, mycophenolic acid, cyclophosphamide, hydrocortisone, and tacrolimus inhibited MRP2 and MRP4, whereas mitoxantrone and dexamethasone stimulated [(3)H]-MTX transport by both transporters. Tacrolimus 104-114 ATP binding cassette subfamily C member 2 Homo sapiens 125-129 24118301-2 2013 The bioavailability and elimination of tacrolimus are affected by the patients CYP3A5 genotype. Tacrolimus 39-49 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 79-85 24036158-7 2013 With regard to the apical efflux transporters, mycophenolic acid, cyclophosphamide, hydrocortisone, and tacrolimus inhibited MRP2 and MRP4, whereas mitoxantrone and dexamethasone stimulated [(3)H]-MTX transport by both transporters. Tacrolimus 104-114 ATP binding cassette subfamily C member 4 Homo sapiens 134-138 24303040-3 2013 We found that depletion of calstabin1 with the calstabin1-dissociation compound FK506 increased the release of calcium from the SR by 14 % during tetanic stimulation (50 Hz, 300 ms) and delayed cytosolic calcium removal. Tacrolimus 80-85 FKBP prolyl isomerase 1A Homo sapiens 27-37 24036158-7 2013 With regard to the apical efflux transporters, mycophenolic acid, cyclophosphamide, hydrocortisone, and tacrolimus inhibited MRP2 and MRP4, whereas mitoxantrone and dexamethasone stimulated [(3)H]-MTX transport by both transporters. Tacrolimus 104-114 metaxin 1 Homo sapiens 197-200 24303040-3 2013 We found that depletion of calstabin1 with the calstabin1-dissociation compound FK506 increased the release of calcium from the SR by 14 % during tetanic stimulation (50 Hz, 300 ms) and delayed cytosolic calcium removal. Tacrolimus 80-85 FKBP prolyl isomerase 1A Homo sapiens 47-57 23062024-1 2013 This study aimed to investigate in vivo absorption of tacrolimus formulated as a solid dispersion using Eudragit E /HCl (E-SD). Tacrolimus 54-64 esterase D Rattus norvegicus 121-125 24120259-0 2013 Personalized tacrolimus doses determined by CYP3A5 genotype for induction and maintenance phases of kidney transplantation. Tacrolimus 13-23 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 44-50 24120259-1 2013 BACKGROUND: Cytochrome P450 (CYP) 3A4 and 3A5 are major isoforms involved in the metabolism of tacrolimus, with the CYP3A5 gene being more polymorphic. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 116-122 24120259-2 2013 It is hypothesized that individual variation in the metabolism of tacrolimus drug may result from genetic polymorphism of CYP3A5. Tacrolimus 66-76 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 122-128 24120259-3 2013 It has been reported that the clearance of tacrolimus in patients with the CYP3A5*1 allele was ~2.5-fold greater than that in those with the CYP3A5*3/*3 genotype. Tacrolimus 43-53 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 75-81 24120259-3 2013 It has been reported that the clearance of tacrolimus in patients with the CYP3A5*1 allele was ~2.5-fold greater than that in those with the CYP3A5*3/*3 genotype. Tacrolimus 43-53 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 141-147 24120259-4 2013 Recent data have also shown that polymorphism in exon 26 (C3435T) of the multidrug resistance gene (MDR1) was correlated with the expression level and function of P-glycoprotein in the lower duodenum, making the relationship between polymorphism of MDR1 and the effective dose of tacrolimus a source of controversy. Tacrolimus 280-290 ATP binding cassette subfamily B member 1 Homo sapiens 100-104 24120259-4 2013 Recent data have also shown that polymorphism in exon 26 (C3435T) of the multidrug resistance gene (MDR1) was correlated with the expression level and function of P-glycoprotein in the lower duodenum, making the relationship between polymorphism of MDR1 and the effective dose of tacrolimus a source of controversy. Tacrolimus 280-290 ATP binding cassette subfamily B member 1 Homo sapiens 163-177 24120259-4 2013 Recent data have also shown that polymorphism in exon 26 (C3435T) of the multidrug resistance gene (MDR1) was correlated with the expression level and function of P-glycoprotein in the lower duodenum, making the relationship between polymorphism of MDR1 and the effective dose of tacrolimus a source of controversy. Tacrolimus 280-290 ATP binding cassette subfamily B member 1 Homo sapiens 249-253 24120259-5 2013 OBJECTIVES: This study investigated the influence of genetic polymorphisms of CYP3A5 and MDR1 on the dose requirements for the induction and maintenance phases of tacrolimus therapy in kidney transplant recipients. Tacrolimus 163-173 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 78-84 24120259-5 2013 OBJECTIVES: This study investigated the influence of genetic polymorphisms of CYP3A5 and MDR1 on the dose requirements for the induction and maintenance phases of tacrolimus therapy in kidney transplant recipients. Tacrolimus 163-173 ATP binding cassette subfamily B member 1 Homo sapiens 89-93 24120259-9 2013 The mean dose of tacrolimus required for the induction phase was significantly greater in the CYP3A5*1/*1 group (0.142 [0.050] mg/kg/d) than that required in the CYP3A5*1/*3 group (0.097 [0.040] mg/kg/d; P = 0.072) and in the CYP3A5*3/*3 group (0.077 [0.020] mg/kg/d; P = 0.005). Tacrolimus 17-27 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 94-100 24120259-9 2013 The mean dose of tacrolimus required for the induction phase was significantly greater in the CYP3A5*1/*1 group (0.142 [0.050] mg/kg/d) than that required in the CYP3A5*1/*3 group (0.097 [0.040] mg/kg/d; P = 0.072) and in the CYP3A5*3/*3 group (0.077 [0.020] mg/kg/d; P = 0.005). Tacrolimus 17-27 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 162-168 24120259-9 2013 The mean dose of tacrolimus required for the induction phase was significantly greater in the CYP3A5*1/*1 group (0.142 [0.050] mg/kg/d) than that required in the CYP3A5*1/*3 group (0.097 [0.040] mg/kg/d; P = 0.072) and in the CYP3A5*3/*3 group (0.077 [0.020] mg/kg/d; P = 0.005). Tacrolimus 17-27 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 162-168 24120259-12 2013 CONCLUSION: Determination of the CYP3A5 genotype would be helpful in the design of adequate immunosuppressive treatment and in lowering toxicity by predicting the doses of tacrolimus required for the induction and maintenance phases in individual kidney transplant recipients. Tacrolimus 172-182 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 23933011-5 2013 The immunophilin FKBP52 is composed of three domains, an FK506-binding domain with peptidyl-prolyl isomerase activity, an FKBP-like domain of unknown function and a TPR-clamp domain, which recognizes the C-terminal peptide of Hsp90 with high affinity. Tacrolimus 57-62 FKBP prolyl isomerase 4 Homo sapiens 17-23 23933011-6 2013 The herein reported crystal structures of FKBP52 reveal that the short linker connecting the FK506-binding domain and the FKBP-like domain acts as a flexible hinge. Tacrolimus 93-98 FKBP prolyl isomerase 4 Homo sapiens 42-48 23933011-8 2013 We further present two co-crystal structures of FKBP52 in complex with the prototypic ligand FK506 and a synthetic analog thereof. Tacrolimus 93-98 FKBP prolyl isomerase 4 Homo sapiens 48-54 23062024-4 2013 Tacrolimus was formulated with E-SD at several different ratios. Tacrolimus 0-10 esterase D Rattus norvegicus 31-35 24064349-5 2013 Co-stimulation with BMP-9 and FK506 induced gene expression of runx2, osterix, and bone sialoprotein, and ALP activity compared with BMP-9 alone, BMP-2 alone and BMP-2+FK506 in rDFAT cells. Tacrolimus 30-35 RUNX family transcription factor 2 Rattus norvegicus 63-68 24085506-2 2013 However, current immunosuppression regimens containing tacrolimus and sirolimus have been shown to induce insulin resistance in rodents. Tacrolimus 55-65 insulin Homo sapiens 106-113 23271170-1 2013 OBJECTIVES: A multicenter, randomized, double-blind, placebo-controlled study of the oral calcineurin inhibitor tacrolimus was performed in patients with early rheumatoid arthritis who had responded poorly to disease-modifying antirheumatic drugs (DMARDs), and factors related to suppression of joint destruction were investigated. Tacrolimus 112-122 calcineurin binding protein 1 Homo sapiens 90-111 23271170-4 2013 RESULTS: Univariate analysis showed that a baseline C-reactive protein (CRP) level of <1.5 mg/dL was the major determinant of TSS <0.5 at week 52 in the tacrolimus group. Tacrolimus 160-170 C-reactive protein Homo sapiens 52-70 23271170-4 2013 RESULTS: Univariate analysis showed that a baseline C-reactive protein (CRP) level of <1.5 mg/dL was the major determinant of TSS <0.5 at week 52 in the tacrolimus group. Tacrolimus 160-170 C-reactive protein Homo sapiens 72-75 23271170-8 2013 CONCLUSIONS: Adding tacrolimus to DMARDs significantly suppressed disease activity and joint destruction in patients with early rheumatoid arthritis, a disease duration <=3 years, a CRP <1.5 mg/dL, and a poor response to oral DMARDs. Tacrolimus 20-30 C-reactive protein Homo sapiens 185-188 24049110-5 2013 Pharmacological inhibition of this interaction using FK506 or siRNA-mediated Fkbp4/5 depletion leads to decreased Ago2 protein levels. Tacrolimus 53-58 argonaute RISC catalytic component 2 Homo sapiens 114-118 23957229-2 2013 Natural compounds FK506, rapamycin and ascomycin, are FKBP12 ligands used for treating organ transplant rejection and other diseases. Tacrolimus 18-23 FKBP prolyl isomerase 1A Homo sapiens 54-60 23900887-0 2013 Impact of donor and recipient CYP3A5 and ABCB1 genetic polymorphisms on tacrolimus dosage requirements and rejection in Caucasian Spanish liver transplant patients. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 30-36 23900887-0 2013 Impact of donor and recipient CYP3A5 and ABCB1 genetic polymorphisms on tacrolimus dosage requirements and rejection in Caucasian Spanish liver transplant patients. Tacrolimus 72-82 ATP binding cassette subfamily B member 1 Homo sapiens 41-46 23900887-4 2013 Three months post-LT, patients carrying a liver with CYP3A5*1 had significantly lower C0 /D (P = .03) and took significantly higher tacrolimus doses (P = .03) than the corresponding *3/*3 homozygotes. Tacrolimus 132-142 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 53-59 23900887-7 2013 We conclude that in Caucasian Spanish LT patients, a native or graft-borne CYP3A5*1 allele tends to lower tacrolimus concentrations and increase dosage needs, but has no significant impact on the incidence of BPAR. Tacrolimus 106-116 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 75-81 23989449-2 2013 In yeasts and animals, FKBP12 can interact with rapamycin and FK506 to form rapamycin-FKBP12 and FK506-FKBP12 complex, respectively. Tacrolimus 62-67 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 23-29 23989449-2 2013 In yeasts and animals, FKBP12 can interact with rapamycin and FK506 to form rapamycin-FKBP12 and FK506-FKBP12 complex, respectively. Tacrolimus 62-67 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 86-92 23989449-2 2013 In yeasts and animals, FKBP12 can interact with rapamycin and FK506 to form rapamycin-FKBP12 and FK506-FKBP12 complex, respectively. Tacrolimus 62-67 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 86-92 23989449-3 2013 In higher plants, FKBP12 protein lost its function to bind rapamycin and FK506. Tacrolimus 73-78 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 18-24 24049110-6 2013 We find FK506 treatment inhibits, whereas Fkbp4/5 overexpression promotes, miRNA-mediated stabilization of Ago2 expression. Tacrolimus 8-13 argonaute RISC catalytic component 2 Homo sapiens 107-111 24049110-7 2013 Simultaneous treatment with a lysosome inhibitor revealed the accumulation of unloaded Ago2 complexes in FK506-treated cells. Tacrolimus 105-110 argonaute RISC catalytic component 2 Homo sapiens 87-91 24231473-0 2013 The impact of CYP3A5 and MDR1 polymorphisms on tacrolimus dosage requirements and trough concentrations in pediatric renal transplant recipients. Tacrolimus 47-57 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 24231473-0 2013 The impact of CYP3A5 and MDR1 polymorphisms on tacrolimus dosage requirements and trough concentrations in pediatric renal transplant recipients. Tacrolimus 47-57 ATP binding cassette subfamily B member 1 Homo sapiens 25-29 24064349-5 2013 Co-stimulation with BMP-9 and FK506 induced gene expression of runx2, osterix, and bone sialoprotein, and ALP activity compared with BMP-9 alone, BMP-2 alone and BMP-2+FK506 in rDFAT cells. Tacrolimus 30-35 Sp7 transcription factor Rattus norvegicus 70-77 24064349-5 2013 Co-stimulation with BMP-9 and FK506 induced gene expression of runx2, osterix, and bone sialoprotein, and ALP activity compared with BMP-9 alone, BMP-2 alone and BMP-2+FK506 in rDFAT cells. Tacrolimus 30-35 PDZ and LIM domain 3 Rattus norvegicus 106-109 24064349-5 2013 Co-stimulation with BMP-9 and FK506 induced gene expression of runx2, osterix, and bone sialoprotein, and ALP activity compared with BMP-9 alone, BMP-2 alone and BMP-2+FK506 in rDFAT cells. Tacrolimus 30-35 bone morphogenetic protein 2 Rattus norvegicus 146-151 24064349-5 2013 Co-stimulation with BMP-9 and FK506 induced gene expression of runx2, osterix, and bone sialoprotein, and ALP activity compared with BMP-9 alone, BMP-2 alone and BMP-2+FK506 in rDFAT cells. Tacrolimus 30-35 bone morphogenetic protein 2 Rattus norvegicus 162-167 24064349-7 2013 The ALP activity induced by BMP-9+FK506 was not influenced by addition of noggin, a BMP antagonist. Tacrolimus 34-39 PDZ and LIM domain 3 Rattus norvegicus 4-7 26589173-0 2013 Absolute Free Energy of Binding and Entropy of the FKBP12-FK506 Complex: Effects of the Force Field. Tacrolimus 58-63 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 51-57 26589173-5 2013 In this paper, we check the performance of HSMD-TI by applying it to the relatively large ligand FK506 (126 atoms) complexed with the protein FKBP12, where DeltaA(0) = -12.8 kcal/mol is known experimentally as well as the crystal structure of the complex. Tacrolimus 97-102 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 142-148 23990505-0 2013 A Markov chain model to evaluate the effect of CYP3A5 and ABCB1 polymorphisms on adverse events associated with tacrolimus in pediatric renal transplantation. Tacrolimus 112-122 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 47-53 24092409-7 2013 We extend the studies to mammalian cells and examine the FK-506 inhibition of the rapamycin-induced association of FRB/FKBP12. Tacrolimus 57-63 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 119-125 23990505-7 2013 The CYP3A5 non-expressors had higher corresponding normalized tacrolimus levels compared to the expressors in the first 3 months. Tacrolimus 62-72 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 23990505-8 2013 The correlation trend between probability of adverse events and normalized tacrolimus concentrations for the two CYP3A5 phenotypes persisted for the first 9 months of therapy. Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 113-119 23990505-9 2013 The differences among ABCB1 genotypes in terms of adverse events and normalized tacrolimus levels were only observed in the first 3 months of therapy. Tacrolimus 80-90 ATP binding cassette subfamily B member 1 Homo sapiens 22-27 23990505-10 2013 The information on CYP3A5 genotypes and tacrolimus dose requirement is important in designing effective programs toward management of tacrolimus side effects particularly for the initial dose when tacrolimus blood levels are not available for therapeutic drug monitoring. Tacrolimus 134-144 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 19-25 23990505-10 2013 The information on CYP3A5 genotypes and tacrolimus dose requirement is important in designing effective programs toward management of tacrolimus side effects particularly for the initial dose when tacrolimus blood levels are not available for therapeutic drug monitoring. Tacrolimus 134-144 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 19-25 23817982-6 2013 We found that MyD88-mediated innate TLR signals were essential for the B-cell response; transgenic B cells expressing surface immunoglobulin specific for insulin reacted to EXO stimulation, and addition of a calcineurin inhibitor FK506 abrogated the EXO-induced B-cell response, suggesting that both innate and antigen-specific signals may be involved. Tacrolimus 230-235 myeloid differentiation primary response gene 88 Mus musculus 14-19 23732701-1 2013 Cyclosporine A and FK506 produce immunosuppression by blocking calcineurin phosphatase activity and consequently activation of cytosolic Nuclear Factor of Activated T-cell (NFATc) transcription factor. Tacrolimus 19-24 nuclear factor of activated T cells 1 Homo sapiens 173-178 23873120-0 2013 Meta-analysis and systematic review of the effect of the donor and recipient CYP3A5 6986A>G genotype on tacrolimus dose requirements in liver transplantation. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 77-83 24097149-0 2013 [A case of calcineurin-inhibitor induced pain syndrome associated with tacrolimus therapy for ulcerative colitis]. Tacrolimus 71-81 calcineurin binding protein 1 Homo sapiens 11-32 24097149-6 2013 We suspected calcineurin-inhibitor induced pain syndrome (CIPS) due to tacrolimus therapy. Tacrolimus 71-81 calcineurin binding protein 1 Homo sapiens 13-34 23873120-1 2013 OBJECTIVE: A meta-analysis was carried out of published studies on the effect of the CYP3A5 6986A>G polymorphism in liver donors and transplant recipients on tacrolimus pharmacokinetics. Tacrolimus 161-171 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 85-91 23873120-2 2013 METHODS: Cohort studies that evaluated the relationship between the CYP3A5 polymorphism in liver donors and transplant recipients and tacrolimus, trough blood concentration normalized for the daily dose (C) per kilogram body weight (D) (C/D, ng/ml/mg/kg/day) up to 1 year after transplantation, were included. Tacrolimus 134-144 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 68-74 23873120-9 2013 CONCLUSION: The presence of the CYP3A5 6986A>G polymorphism in the donor affects tacrolimus pharmacokinetics in the recipient, although only the evidence available for the first month after transplantation was of adequate quality for demonstrating a significant difference. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 32-38 24052064-0 2013 Impact of CYP3A4*22 allele on tacrolimus pharmacokinetics in early period after renal transplantation: toward updated genotype-based dosage guidelines. Tacrolimus 30-40 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 10-16 24052064-1 2013 BACKGROUND: Tacrolimus (Tac) metabolism is mainly mediated by the cytochrome P450 3A (CYP3A) subfamily. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 66-84 24052064-1 2013 BACKGROUND: Tacrolimus (Tac) metabolism is mainly mediated by the cytochrome P450 3A (CYP3A) subfamily. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 86-91 23305195-1 2013 AIMS: An algorithm based on the CYP3A5 genotype to predict tacrolimus clearance to inform the optimal initial dose was derived using data from the DeKAF study (Passey et al. Tacrolimus 59-69 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 32-38 24011263-9 2013 In the nerve graft, miR-125-3b and miR-672 were significantly upregulated in the mice that received an allograft with FK506 only at 7 d after nerve allotransplantation. Tacrolimus 118-123 microRNA 672 Mus musculus 35-42 23800841-3 2013 We found that FK506 markedly reduced the abnormal form of prion protein (PRNP(Sc)) in the cell cultures (N2a58 and MG20) infected with Fukuoka-1 prion. Tacrolimus 14-19 prion protein Mus musculus 73-77 23800841-5 2013 Upregulation of the autophagy-related molecules was also seen in the brains of FK506-treated mice and the accumulation of PRNP(Sc) was delayed. Tacrolimus 79-84 prion protein Mus musculus 122-126 23800841-7 2013 These findings provide evidence that FK506 could constitute a novel antiprion drug, capable of enhancing the degradation of PRNP(Sc) in addition to attenuation of microgliosis and neuroprotection. Tacrolimus 37-42 prion protein Mus musculus 124-128 23633119-0 2013 Multidrug resistance-associated protein 2 (MRP2/ABCC2) haplotypes significantly affect the pharmacokinetics of tacrolimus in kidney transplant recipients. Tacrolimus 111-121 ATP binding cassette subfamily C member 2 Homo sapiens 0-41 23224428-3 2013 Human FKBP12 and some of its paralogues bind to different macrocyclic antibiotics such as FK506 or rapamycin and their derivatives. Tacrolimus 90-95 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 6-12 23707520-0 2013 FK506 induces endothelial dysfunction through attenuation of Akt and ERK1/2 independently of calcineurin inhibition and the caspase pathway. Tacrolimus 0-5 AKT serine/threonine kinase 1 Homo sapiens 61-64 23707520-0 2013 FK506 induces endothelial dysfunction through attenuation of Akt and ERK1/2 independently of calcineurin inhibition and the caspase pathway. Tacrolimus 0-5 mitogen-activated protein kinase 3 Homo sapiens 69-75 23707520-7 2013 FK506 induced attenuation of Akt and extracellular-regulated kinase 1/2 (ERK1/2). Tacrolimus 0-5 AKT serine/threonine kinase 1 Homo sapiens 29-32 23707520-7 2013 FK506 induced attenuation of Akt and extracellular-regulated kinase 1/2 (ERK1/2). Tacrolimus 0-5 mitogen-activated protein kinase 3 Homo sapiens 73-79 23707520-9 2013 Present results suggest that FK506 induces endothelial dysfunction through attenuation of Akt and ERK1/2 independently of calcineurin inhibition and the caspase pathway. Tacrolimus 29-34 AKT serine/threonine kinase 1 Homo sapiens 90-93 23707520-9 2013 Present results suggest that FK506 induces endothelial dysfunction through attenuation of Akt and ERK1/2 independently of calcineurin inhibition and the caspase pathway. Tacrolimus 29-34 mitogen-activated protein kinase 3 Homo sapiens 98-104 23633119-0 2013 Multidrug resistance-associated protein 2 (MRP2/ABCC2) haplotypes significantly affect the pharmacokinetics of tacrolimus in kidney transplant recipients. Tacrolimus 111-121 ATP binding cassette subfamily C member 2 Homo sapiens 43-47 23633119-0 2013 Multidrug resistance-associated protein 2 (MRP2/ABCC2) haplotypes significantly affect the pharmacokinetics of tacrolimus in kidney transplant recipients. Tacrolimus 111-121 ATP binding cassette subfamily C member 2 Homo sapiens 48-53 23633119-3 2013 Tacrolimus is mainly metabolized by cytochrome P450 (CYP) 3A4 and 3A5 and effluxed via ATP-binding cassette (ABC) transporters such as P-glycoprotein (P-gp), encoded by ABCB1 gene. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 135-149 23633119-3 2013 Tacrolimus is mainly metabolized by cytochrome P450 (CYP) 3A4 and 3A5 and effluxed via ATP-binding cassette (ABC) transporters such as P-glycoprotein (P-gp), encoded by ABCB1 gene. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 151-155 23633119-3 2013 Tacrolimus is mainly metabolized by cytochrome P450 (CYP) 3A4 and 3A5 and effluxed via ATP-binding cassette (ABC) transporters such as P-glycoprotein (P-gp), encoded by ABCB1 gene. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 169-174 23633119-4 2013 The influence of CYP3A5*3 on the pharmacokinetics of tacrolimus has been well characterized. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 17-23 23633119-6 2013 In addition, the involvement of other efflux transporters than P-gp in tacrolimus disposition is uncertain. Tacrolimus 71-81 ATP binding cassette subfamily B member 1 Homo sapiens 63-67 23633119-7 2013 The present study was designed to investigate the effects of genetic polymorphisms of CYP3As and efflux transporters on the pharmacokinetics of tacrolimus. Tacrolimus 144-154 peptidylprolyl isomerase F Homo sapiens 86-90 23633119-13 2013 RESULTS: Analyses revealed that the CYP3A5 expressers (CYP3A5*1 carriers) and MRP2 high-activity group (ABCC2 H2/H2 and H1/H2) showed a decreased dose-normalized trough concentration of tacrolimus by 2.3-fold (p < 0.001) and 1.5-fold (p = 0.007), respectively. Tacrolimus 186-196 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 23633119-13 2013 RESULTS: Analyses revealed that the CYP3A5 expressers (CYP3A5*1 carriers) and MRP2 high-activity group (ABCC2 H2/H2 and H1/H2) showed a decreased dose-normalized trough concentration of tacrolimus by 2.3-fold (p < 0.001) and 1.5-fold (p = 0.007), respectively. Tacrolimus 186-196 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 55-61 23633119-13 2013 RESULTS: Analyses revealed that the CYP3A5 expressers (CYP3A5*1 carriers) and MRP2 high-activity group (ABCC2 H2/H2 and H1/H2) showed a decreased dose-normalized trough concentration of tacrolimus by 2.3-fold (p < 0.001) and 1.5-fold (p = 0.007), respectively. Tacrolimus 186-196 ATP binding cassette subfamily C member 2 Homo sapiens 78-82 23633119-13 2013 RESULTS: Analyses revealed that the CYP3A5 expressers (CYP3A5*1 carriers) and MRP2 high-activity group (ABCC2 H2/H2 and H1/H2) showed a decreased dose-normalized trough concentration of tacrolimus by 2.3-fold (p < 0.001) and 1.5-fold (p = 0.007), respectively. Tacrolimus 186-196 ATP binding cassette subfamily C member 2 Homo sapiens 104-109 23633119-13 2013 RESULTS: Analyses revealed that the CYP3A5 expressers (CYP3A5*1 carriers) and MRP2 high-activity group (ABCC2 H2/H2 and H1/H2) showed a decreased dose-normalized trough concentration of tacrolimus by 2.3-fold (p < 0.001) and 1.5-fold (p = 0.007), respectively. Tacrolimus 186-196 relaxin 2 Homo sapiens 110-125 23633119-15 2013 In the population pharmacokinetic analysis, CYP3A5 expressers and MRP2 high-activity groups were identified as the significant covariates for tacrolimus apparent clearance expressed as 20.7 x (age/50)(-0.78) x 2.03 (CYP3A5 expressers) x 1.40 (MRP2 high-activity group). Tacrolimus 142-152 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 44-50 23633119-15 2013 In the population pharmacokinetic analysis, CYP3A5 expressers and MRP2 high-activity groups were identified as the significant covariates for tacrolimus apparent clearance expressed as 20.7 x (age/50)(-0.78) x 2.03 (CYP3A5 expressers) x 1.40 (MRP2 high-activity group). Tacrolimus 142-152 ATP binding cassette subfamily C member 2 Homo sapiens 66-70 23633119-15 2013 In the population pharmacokinetic analysis, CYP3A5 expressers and MRP2 high-activity groups were identified as the significant covariates for tacrolimus apparent clearance expressed as 20.7 x (age/50)(-0.78) x 2.03 (CYP3A5 expressers) x 1.40 (MRP2 high-activity group). Tacrolimus 142-152 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 216-222 23633119-15 2013 In the population pharmacokinetic analysis, CYP3A5 expressers and MRP2 high-activity groups were identified as the significant covariates for tacrolimus apparent clearance expressed as 20.7 x (age/50)(-0.78) x 2.03 (CYP3A5 expressers) x 1.40 (MRP2 high-activity group). Tacrolimus 142-152 ATP binding cassette subfamily C member 2 Homo sapiens 243-247 23633119-17 2013 CONCLUSIONS: This is the first report showing that MRP2/ABCC2 has a crucial impact on the pharmacokinetics of tacrolimus in a haplotype-specific manner. Tacrolimus 110-120 ATP binding cassette subfamily C member 2 Homo sapiens 51-55 23633119-17 2013 CONCLUSIONS: This is the first report showing that MRP2/ABCC2 has a crucial impact on the pharmacokinetics of tacrolimus in a haplotype-specific manner. Tacrolimus 110-120 ATP binding cassette subfamily C member 2 Homo sapiens 56-61 23633119-18 2013 Determination of the ABCC2 as well as CYP3A5 genotype may be useful for more accurate tacrolimus dosage adjustment. Tacrolimus 86-96 ATP binding cassette subfamily C member 2 Homo sapiens 21-26 23633119-18 2013 Determination of the ABCC2 as well as CYP3A5 genotype may be useful for more accurate tacrolimus dosage adjustment. Tacrolimus 86-96 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 38-44 23991193-0 2013 Novel single nucleotide polymorphisms in interleukin 6 affect tacrolimus metabolism in liver transplant patients. Tacrolimus 62-72 interleukin 6 Homo sapiens 41-54 23733010-6 2013 Consequently, tacrolimus absolute bioavailability was lowest for recipients with the CYP3A5*1 allele taking tacrolimus QD. Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 85-91 23733010-6 2013 Consequently, tacrolimus absolute bioavailability was lowest for recipients with the CYP3A5*1 allele taking tacrolimus QD. Tacrolimus 108-118 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 85-91 23733010-8 2013 CONCLUSION: The larger interindividual variability of tacrolimus bioavailability for oral formulations appears to be due to the effects of the CYP3A5 polymorphism and the tacrolimus oral formulation. Tacrolimus 54-64 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 143-149 24128104-7 2013 The visa was applied to 170 drugs and nutrition products different, being the majority Tacrolimus. Tacrolimus 87-97 mitochondrial antiviral signaling protein Homo sapiens 4-8 24024898-1 2013 BACKGROUND & AIM: In vitro studies have identified both midazolam and tacrolimus as dual CYP3A4 and CYP3A5 substrates. Tacrolimus 74-84 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-99 24024898-1 2013 BACKGROUND & AIM: In vitro studies have identified both midazolam and tacrolimus as dual CYP3A4 and CYP3A5 substrates. Tacrolimus 74-84 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 104-110 24024898-2 2013 In vivo; however, the CYP3A5 genotype has a marked impact on tacrolimus pharmacokinetics, whereas it seems not to affect midazolam pharmacokinetics. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 22-28 24024898-5 2013 RESULTS: CYP3A5 expressers display an approximately 2.4-fold higher tacrolimus clearance as compared with CYP3A5 nonexpressers, whereas there are no differences in systemic and apparent oral midazolam clearance. Tacrolimus 68-78 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 9-15 24024898-6 2013 CONCLUSION: These data confirm that in vivo CYP3A5 plays an important role in tacrolimus metabolism, while its contribution to midazolam metabolism in a relevant study population is limited. Tacrolimus 78-88 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 44-50 24687344-4 2013 In regard to the clinical implications of MDR1 SNPs, it was found in large meta-analysis that C3435T SNP was associated with a slight increase in the susceptibility to ulcerative colitis and cancer and was related with slight modifications in tacrolimus pharmacokinetics and platinum-based chemotherapy response in lung cancer. Tacrolimus 243-253 ATP binding cassette subfamily B member 1 Homo sapiens 42-46 23921892-0 2013 Evolution to twice daily bolus intravenous tacrolimus: optimizing efficacy and safety of calcineurin inhibitor delivery early post lung transplant. Tacrolimus 43-53 calcineurin binding protein 1 Homo sapiens 89-110 23647800-8 2013 Microarray gene expression of NME/NM23 nucleoside diphosphate kinase 1 and heterogeneous nuclear ribonucleoprotein H3-2H9 were significantly downregulated (P < 0.05) by tacrolimus in both HTS and keloid fibroblast populations but not normal fibroblasts. Tacrolimus 172-182 NME/NM23 nucleoside diphosphate kinase 1 Homo sapiens 30-121 23647800-10 2013 Microarray gene expression of NME/NM23 nucleoside diphosphate kinase 1 and heterogeneous nuclear ribonucleoprotein H3-2H9 are blocked by tacrolimus in pathologic fibroblasts but not normal fibroblasts, and may represent novel genes underlying HTS and keloid pathogenesis. Tacrolimus 137-147 NME/NM23 nucleoside diphosphate kinase 1 Homo sapiens 30-121 24024898-0 2013 Impact of CYP3A5 genotype on tacrolimus versus midazolam clearance in renal transplant recipients: new insights in CYP3A5-mediated drug metabolism. Tacrolimus 29-39 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 23823650-9 2013 In addition, the amount of CnAbeta-CaM corresponds positively with T-cell activity and negatively with tacrolimus level. Tacrolimus 103-113 protein phosphatase 3, catalytic subunit, beta isoform Mus musculus 27-34 23991193-6 2013 We speculated that IL6 rs1800796 polymorphisms may lead to individual differences in tacrolimus metabolism by affecting CYP3A enzymes levels and liver function after liver transplantation. Tacrolimus 85-95 interleukin 6 Homo sapiens 19-22 23991193-11 2013 Donor CYP3A5 rs776746 allele A, IL6 rs1800796 allele C, and recipient CYP3A5 rs776746 allele A were associated with fast tacrolimus metabolism. Tacrolimus 121-131 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 6-12 23991193-11 2013 Donor CYP3A5 rs776746 allele A, IL6 rs1800796 allele C, and recipient CYP3A5 rs776746 allele A were associated with fast tacrolimus metabolism. Tacrolimus 121-131 interleukin 6 Homo sapiens 32-35 23991193-11 2013 Donor CYP3A5 rs776746 allele A, IL6 rs1800796 allele C, and recipient CYP3A5 rs776746 allele A were associated with fast tacrolimus metabolism. Tacrolimus 121-131 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 70-76 23991193-14 2013 CONCLUSIONS: Combined analysis of donor CYP3A5 rs776746, IL6 rs1800796, and recipient CYP3A5 rs776746 polymorphisms may distinguish tacrolimus metabolism better than CYP3A5 rs776746 alone. Tacrolimus 132-142 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 40-46 23991193-14 2013 CONCLUSIONS: Combined analysis of donor CYP3A5 rs776746, IL6 rs1800796, and recipient CYP3A5 rs776746 polymorphisms may distinguish tacrolimus metabolism better than CYP3A5 rs776746 alone. Tacrolimus 132-142 interleukin 6 Homo sapiens 57-60 23991193-14 2013 CONCLUSIONS: Combined analysis of donor CYP3A5 rs776746, IL6 rs1800796, and recipient CYP3A5 rs776746 polymorphisms may distinguish tacrolimus metabolism better than CYP3A5 rs776746 alone. Tacrolimus 132-142 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 86-92 23991193-14 2013 CONCLUSIONS: Combined analysis of donor CYP3A5 rs776746, IL6 rs1800796, and recipient CYP3A5 rs776746 polymorphisms may distinguish tacrolimus metabolism better than CYP3A5 rs776746 alone. Tacrolimus 132-142 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 86-92 23991193-15 2013 IL6 may lead to individual differences in tacrolimus metabolism mainly by affecting liver function. Tacrolimus 42-52 interleukin 6 Homo sapiens 0-3 23839048-5 2013 Recent studies have also shown that FK506-binding proteins can modulate Akt-mTOR signaling in the absence of rapamycin. Tacrolimus 36-41 AKT serine/threonine kinase 1 Homo sapiens 72-75 23696253-1 2013 The mammalian target of rapamycin inhibitor everolimus (Zortress , Certican ) was recently approved in the USA and a number of EU countries for use in combination with a reduced dosage of tacrolimus and corticosteroids for the prophylaxis of organ rejection in adult liver transplant recipients. Tacrolimus 188-198 mechanistic target of rapamycin kinase Homo sapiens 4-33 23657856-0 2013 Scavengers of reactive oxygen species, paracalcitol, RhoA, and Rac-1 inhibitors and tacrolimus inhibit angiotensin II-induced actions on glomerular permeability. Tacrolimus 84-94 angiotensinogen Rattus norvegicus 103-117 23657856-9 2013 Paracalcitol, RhoA, and Rac-1 inhibition, and, to some extent tacrolimus, but not prostacyclin, could also inhibit the glomerular permeability actions of ANG II. Tacrolimus 62-72 angiogenin Rattus norvegicus 154-157 23839048-5 2013 Recent studies have also shown that FK506-binding proteins can modulate Akt-mTOR signaling in the absence of rapamycin. Tacrolimus 36-41 mechanistic target of rapamycin kinase Homo sapiens 76-80 23665867-0 2013 Response to "CYP3A5 genotype, but not CYP3A4*1b, CYP3A4*22, or hematocrit, predicts tacrolimus dose requirements in Brazilian renal transplant patients". Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 23588314-0 2013 CYP3A5 genotype, but not CYP3A4*1b, CYP3A4*22, or hematocrit, predicts tacrolimus dose requirements in Brazilian renal transplant patients. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 23574377-1 2013 WHAT IS KNOWN AND OBJECTIVE: Tacrolimus has a narrow therapeutic index and shows large interindividual variations in pharmacokinetics, which may be partly explained by genetic variability in metabolic enzymes of the cytochrome P450 (mainly CYP3A4 and CYP3A5) and transport P-glycoprotein (encoded by the ABCB1 gene). Tacrolimus 29-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 240-246 23574377-1 2013 WHAT IS KNOWN AND OBJECTIVE: Tacrolimus has a narrow therapeutic index and shows large interindividual variations in pharmacokinetics, which may be partly explained by genetic variability in metabolic enzymes of the cytochrome P450 (mainly CYP3A4 and CYP3A5) and transport P-glycoprotein (encoded by the ABCB1 gene). Tacrolimus 29-39 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 251-257 23574377-1 2013 WHAT IS KNOWN AND OBJECTIVE: Tacrolimus has a narrow therapeutic index and shows large interindividual variations in pharmacokinetics, which may be partly explained by genetic variability in metabolic enzymes of the cytochrome P450 (mainly CYP3A4 and CYP3A5) and transport P-glycoprotein (encoded by the ABCB1 gene). Tacrolimus 29-39 ATP binding cassette subfamily B member 1 Homo sapiens 273-287 23574377-1 2013 WHAT IS KNOWN AND OBJECTIVE: Tacrolimus has a narrow therapeutic index and shows large interindividual variations in pharmacokinetics, which may be partly explained by genetic variability in metabolic enzymes of the cytochrome P450 (mainly CYP3A4 and CYP3A5) and transport P-glycoprotein (encoded by the ABCB1 gene). Tacrolimus 29-39 ATP binding cassette subfamily B member 1 Homo sapiens 304-309 23597509-4 2013 A selective D2R agonist, quinpirole, up-regulated IP3R-1 protein following its mRNA increase, which was significantly inhibited by gallein (a Gbetagamma modulator), U73122 (a phospholipase C inhibitor), BAPTA-AM (an intracellular calcium chelating reagent), W7 (a calmodulin inhibitor), KN-93 (a calmodulin-dependent protein kinases inhibitor), and FK506 (a calcineurin inhibitor). Tacrolimus 349-354 inositol 1,4,5-trisphosphate receptor 1 Mus musculus 50-56 23867624-0 2013 FK506 activates BMPR2, rescues endothelial dysfunction, and reverses pulmonary hypertension. Tacrolimus 0-5 bone morphogenetic protein receptor type 2 Rattus norvegicus 16-21 23867624-3 2013 The best response was achieved with FK506 (tacrolimus), via a dual mechanism of action as a calcineurin inhibitor that also binds FK-binding protein-12 (FKBP12), a repressor of BMP signaling. Tacrolimus 36-41 FKBP prolyl isomerase 1A Rattus norvegicus 130-151 23867624-3 2013 The best response was achieved with FK506 (tacrolimus), via a dual mechanism of action as a calcineurin inhibitor that also binds FK-binding protein-12 (FKBP12), a repressor of BMP signaling. Tacrolimus 36-41 FKBP prolyl isomerase 1A Rattus norvegicus 153-159 23867624-3 2013 The best response was achieved with FK506 (tacrolimus), via a dual mechanism of action as a calcineurin inhibitor that also binds FK-binding protein-12 (FKBP12), a repressor of BMP signaling. Tacrolimus 36-41 bone morphogenetic protein receptor, type II (serine/threonine kinase) Mus musculus 177-180 23867624-3 2013 The best response was achieved with FK506 (tacrolimus), via a dual mechanism of action as a calcineurin inhibitor that also binds FK-binding protein-12 (FKBP12), a repressor of BMP signaling. Tacrolimus 43-53 FKBP prolyl isomerase 1A Rattus norvegicus 130-151 23867624-3 2013 The best response was achieved with FK506 (tacrolimus), via a dual mechanism of action as a calcineurin inhibitor that also binds FK-binding protein-12 (FKBP12), a repressor of BMP signaling. Tacrolimus 43-53 FKBP prolyl isomerase 1A Rattus norvegicus 153-159 23867624-3 2013 The best response was achieved with FK506 (tacrolimus), via a dual mechanism of action as a calcineurin inhibitor that also binds FK-binding protein-12 (FKBP12), a repressor of BMP signaling. Tacrolimus 43-53 bone morphogenetic protein receptor, type II (serine/threonine kinase) Mus musculus 177-180 23867624-4 2013 FK506 released FKBP12 from type I receptors activin receptor-like kinase 1 (ALK1), ALK2, and ALK3 and activated downstream SMAD1/5 and MAPK signaling and ID1 gene regulation in a manner superior to the calcineurin inhibitor cyclosporine and the FKBP12 ligand rapamycin. Tacrolimus 0-5 FKBP prolyl isomerase 1A Rattus norvegicus 15-21 23867624-4 2013 FK506 released FKBP12 from type I receptors activin receptor-like kinase 1 (ALK1), ALK2, and ALK3 and activated downstream SMAD1/5 and MAPK signaling and ID1 gene regulation in a manner superior to the calcineurin inhibitor cyclosporine and the FKBP12 ligand rapamycin. Tacrolimus 0-5 bone morphogenetic protein receptor type 1A Rattus norvegicus 93-97 23867624-4 2013 FK506 released FKBP12 from type I receptors activin receptor-like kinase 1 (ALK1), ALK2, and ALK3 and activated downstream SMAD1/5 and MAPK signaling and ID1 gene regulation in a manner superior to the calcineurin inhibitor cyclosporine and the FKBP12 ligand rapamycin. Tacrolimus 0-5 SMAD family member 1 Rattus norvegicus 123-130 23867624-4 2013 FK506 released FKBP12 from type I receptors activin receptor-like kinase 1 (ALK1), ALK2, and ALK3 and activated downstream SMAD1/5 and MAPK signaling and ID1 gene regulation in a manner superior to the calcineurin inhibitor cyclosporine and the FKBP12 ligand rapamycin. Tacrolimus 0-5 inhibitor of DNA binding 1, HLH protein Rattus norvegicus 154-157 23867624-4 2013 FK506 released FKBP12 from type I receptors activin receptor-like kinase 1 (ALK1), ALK2, and ALK3 and activated downstream SMAD1/5 and MAPK signaling and ID1 gene regulation in a manner superior to the calcineurin inhibitor cyclosporine and the FKBP12 ligand rapamycin. Tacrolimus 0-5 FKBP prolyl isomerase 1A Rattus norvegicus 245-251 23867624-5 2013 In pulmonary artery endothelial cells (ECs) from patients with idiopathic PAH, low-dose FK506 reversed dysfunctional BMPR2 signaling. Tacrolimus 88-93 bone morphogenetic protein receptor type 2 Homo sapiens 117-122 23867624-6 2013 In mice with conditional Bmpr2 deletion in ECs, low-dose FK506 prevented exaggerated chronic hypoxic PAH associated with induction of EC targets of BMP signaling, such as apelin. Tacrolimus 57-62 bone morphogenetic protein receptor, type II (serine/threonine kinase) Mus musculus 25-30 23867624-6 2013 In mice with conditional Bmpr2 deletion in ECs, low-dose FK506 prevented exaggerated chronic hypoxic PAH associated with induction of EC targets of BMP signaling, such as apelin. Tacrolimus 57-62 bone morphogenetic protein receptor, type II (serine/threonine kinase) Mus musculus 148-151 22996305-1 2013 In our previous reports, Wuzhi tablet (an herbal preparation of ethanol extract of Wuweizi (Schisandra sphenanthera)) can significantly increase the blood concentration of tacrolimus and paclitaxel in rats by inhibiting the CYP3A-mediated metabolism and the P-gp-mediated efflux. Tacrolimus 172-182 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 224-229 22996305-1 2013 In our previous reports, Wuzhi tablet (an herbal preparation of ethanol extract of Wuweizi (Schisandra sphenanthera)) can significantly increase the blood concentration of tacrolimus and paclitaxel in rats by inhibiting the CYP3A-mediated metabolism and the P-gp-mediated efflux. Tacrolimus 172-182 phosphoglycolate phosphatase Rattus norvegicus 258-262 23743668-0 2013 Genetic polymorphisms in ABCB1 influence the pharmacodynamics of tacrolimus. Tacrolimus 65-75 ATP binding cassette subfamily B member 1 Homo sapiens 25-30 22447331-8 2013 IL-2 and IL-6 were significantly decreased after the administration of tacrolimus (p = 0.027 and p = 0.024). Tacrolimus 71-81 interleukin 2 Homo sapiens 0-4 22447331-8 2013 IL-2 and IL-6 were significantly decreased after the administration of tacrolimus (p = 0.027 and p = 0.024). Tacrolimus 71-81 interleukin 6 Homo sapiens 9-13 23778326-0 2013 A search for new CYP3A4 variants as determinants of tacrolimus dose requirements in renal-transplanted patients. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 17-23 23778326-1 2013 The CYP3A5*3 and CYP3A4*1B alleles have been related with tacrolimus (Tac) dose requirements. Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 23778326-1 2013 The CYP3A5*3 and CYP3A4*1B alleles have been related with tacrolimus (Tac) dose requirements. Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 17-23 23743668-7 2013 In addition, the ABCB1 genotype of 36 tacrolimus-treated renal transplant patients was related to ABCB1 activity and tacrolimus efficacy. Tacrolimus 38-48 ATP binding cassette subfamily B member 1 Homo sapiens 17-22 23743668-7 2013 In addition, the ABCB1 genotype of 36 tacrolimus-treated renal transplant patients was related to ABCB1 activity and tacrolimus efficacy. Tacrolimus 38-48 ATP binding cassette subfamily B member 1 Homo sapiens 98-103 23743668-7 2013 In addition, the ABCB1 genotype of 36 tacrolimus-treated renal transplant patients was related to ABCB1 activity and tacrolimus efficacy. Tacrolimus 117-127 ATP binding cassette subfamily B member 1 Homo sapiens 17-22 23743668-10 2013 Tacrolimus 10 ng/mL reduced the production of IL-2 in CD4 and CD8 T cells by 28.9% and 45.4% (P < 0.05). Tacrolimus 0-10 interleukin 2 Homo sapiens 46-50 23312000-2 2013 The immunosuppressant tacrolimus (TAC) is a substrate of cytochrome P450 3A2 (CYP3A2) and P-glycoprotein (P-gp) in rats. Tacrolimus 22-32 cytochrome P450, family 3, subfamily a, polypeptide 2 Rattus norvegicus 57-76 23743668-10 2013 Tacrolimus 10 ng/mL reduced the production of IL-2 in CD4 and CD8 T cells by 28.9% and 45.4% (P < 0.05). Tacrolimus 0-10 CD4 molecule Homo sapiens 54-57 23312000-2 2013 The immunosuppressant tacrolimus (TAC) is a substrate of cytochrome P450 3A2 (CYP3A2) and P-glycoprotein (P-gp) in rats. Tacrolimus 22-32 cytochrome P450, family 3, subfamily a, polypeptide 2 Rattus norvegicus 78-84 23743668-10 2013 Tacrolimus 10 ng/mL reduced the production of IL-2 in CD4 and CD8 T cells by 28.9% and 45.4% (P < 0.05). Tacrolimus 0-10 CD8a molecule Homo sapiens 62-65 23312000-2 2013 The immunosuppressant tacrolimus (TAC) is a substrate of cytochrome P450 3A2 (CYP3A2) and P-glycoprotein (P-gp) in rats. Tacrolimus 22-32 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 90-104 23743668-11 2013 Tacrolimus-mediated inhibition of IL-2 was enhanced by verapamil (P < 0.05). Tacrolimus 0-10 interleukin 2 Homo sapiens 34-38 23312000-2 2013 The immunosuppressant tacrolimus (TAC) is a substrate of cytochrome P450 3A2 (CYP3A2) and P-glycoprotein (P-gp) in rats. Tacrolimus 22-32 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 106-110 23743668-12 2013 This effect on tacrolimus pharmacodynamics was associated with ABCB1 3435C>T SNP in renal transplant patients: verapamil reduced the percentage of IL-2-producing CD4 and CD8 T cells by 14% and 22% in patients with the CC genotype (P < 0.05) but not in patients with the TT genotype. Tacrolimus 15-25 ATP binding cassette subfamily B member 1 Homo sapiens 63-68 23743668-12 2013 This effect on tacrolimus pharmacodynamics was associated with ABCB1 3435C>T SNP in renal transplant patients: verapamil reduced the percentage of IL-2-producing CD4 and CD8 T cells by 14% and 22% in patients with the CC genotype (P < 0.05) but not in patients with the TT genotype. Tacrolimus 15-25 interleukin 2 Homo sapiens 150-154 23743668-12 2013 This effect on tacrolimus pharmacodynamics was associated with ABCB1 3435C>T SNP in renal transplant patients: verapamil reduced the percentage of IL-2-producing CD4 and CD8 T cells by 14% and 22% in patients with the CC genotype (P < 0.05) but not in patients with the TT genotype. Tacrolimus 15-25 CD4 molecule Homo sapiens 165-168 23743668-12 2013 This effect on tacrolimus pharmacodynamics was associated with ABCB1 3435C>T SNP in renal transplant patients: verapamil reduced the percentage of IL-2-producing CD4 and CD8 T cells by 14% and 22% in patients with the CC genotype (P < 0.05) but not in patients with the TT genotype. Tacrolimus 15-25 CD8a molecule Homo sapiens 173-176 23743668-13 2013 Moreover, the ratio of tacrolimus C0 over the percent of IL-2-producing CD8 T cells in CC genotype patients was significantly higher compared with TT genotype patients (P < 0.05), showing a smaller pharmacodynamic effect in CC genotype patients. Tacrolimus 23-33 interleukin 2 Homo sapiens 57-61 23743668-13 2013 Moreover, the ratio of tacrolimus C0 over the percent of IL-2-producing CD8 T cells in CC genotype patients was significantly higher compared with TT genotype patients (P < 0.05), showing a smaller pharmacodynamic effect in CC genotype patients. Tacrolimus 23-33 CD8a molecule Homo sapiens 72-75 23743668-14 2013 CONCLUSION: The ABCB1 3435C>T SNP influences ABCB1 activity of T cells and the pharmacodynamic effect of tacrolimus in kidney transplant patients. Tacrolimus 108-118 ATP binding cassette subfamily B member 1 Homo sapiens 16-21 23743668-14 2013 CONCLUSION: The ABCB1 3435C>T SNP influences ABCB1 activity of T cells and the pharmacodynamic effect of tacrolimus in kidney transplant patients. Tacrolimus 108-118 ATP binding cassette subfamily B member 1 Homo sapiens 48-53 22660906-1 2013 Tacrolimus, a reversible calcineurin inhibitor, is known for its diabetogenic potential. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 25-46 23651473-8 2013 In obese animals, tacrolimus reduced beta-cell proliferation and Ins2 gene expression compared with cyclosporine-A. Tacrolimus 18-28 insulin 2 Rattus norvegicus 65-69 23651473-12 2013 This may be explained by greater inhibition of Ins2 gene and beta-cell proliferation by tacrolimus in the insulin resistant state. Tacrolimus 88-98 insulin 2 Rattus norvegicus 47-51 23890011-2 2013 We demonstrated that FK506 binds to the C-terminal region of spartin and thereby inhibits the interaction of spartin with TIP47, one of the lipid droplet-associated proteins. Tacrolimus 21-26 spartin Homo sapiens 61-68 23890011-2 2013 We demonstrated that FK506 binds to the C-terminal region of spartin and thereby inhibits the interaction of spartin with TIP47, one of the lipid droplet-associated proteins. Tacrolimus 21-26 spartin Homo sapiens 109-116 23890011-2 2013 We demonstrated that FK506 binds to the C-terminal region of spartin and thereby inhibits the interaction of spartin with TIP47, one of the lipid droplet-associated proteins. Tacrolimus 21-26 perilipin 3 Homo sapiens 122-127 23890011-3 2013 We further confirmed that FK506 inhibits localization of spartin and its binder, an E3 ubiquitin ligase AIP4, in lipid droplets and increases the protein level of ADRP (adipose differentiation-related protein), which is a regulator of lipid homeostasis. Tacrolimus 26-31 spartin Homo sapiens 57-64 23890011-3 2013 We further confirmed that FK506 inhibits localization of spartin and its binder, an E3 ubiquitin ligase AIP4, in lipid droplets and increases the protein level of ADRP (adipose differentiation-related protein), which is a regulator of lipid homeostasis. Tacrolimus 26-31 itchy E3 ubiquitin protein ligase Homo sapiens 104-108 23890011-3 2013 We further confirmed that FK506 inhibits localization of spartin and its binder, an E3 ubiquitin ligase AIP4, in lipid droplets and increases the protein level of ADRP (adipose differentiation-related protein), which is a regulator of lipid homeostasis. Tacrolimus 26-31 perilipin 2 Homo sapiens 163-167 23890011-4 2013 These results strongly suggest that FK506 suppresses the proteasomal degradation of ADRP, a substrate of AIP4, by inhibiting the spartin-TIP47 interaction and thereby blocking the localization of spartin and AIP4 in lipid droplets. Tacrolimus 36-41 perilipin 2 Homo sapiens 84-88 23890011-4 2013 These results strongly suggest that FK506 suppresses the proteasomal degradation of ADRP, a substrate of AIP4, by inhibiting the spartin-TIP47 interaction and thereby blocking the localization of spartin and AIP4 in lipid droplets. Tacrolimus 36-41 itchy E3 ubiquitin protein ligase Homo sapiens 105-109 23890011-4 2013 These results strongly suggest that FK506 suppresses the proteasomal degradation of ADRP, a substrate of AIP4, by inhibiting the spartin-TIP47 interaction and thereby blocking the localization of spartin and AIP4 in lipid droplets. Tacrolimus 36-41 spartin Homo sapiens 129-136 23890011-4 2013 These results strongly suggest that FK506 suppresses the proteasomal degradation of ADRP, a substrate of AIP4, by inhibiting the spartin-TIP47 interaction and thereby blocking the localization of spartin and AIP4 in lipid droplets. Tacrolimus 36-41 perilipin 3 Homo sapiens 137-142 23890011-4 2013 These results strongly suggest that FK506 suppresses the proteasomal degradation of ADRP, a substrate of AIP4, by inhibiting the spartin-TIP47 interaction and thereby blocking the localization of spartin and AIP4 in lipid droplets. Tacrolimus 36-41 spartin Homo sapiens 196-203 23890011-4 2013 These results strongly suggest that FK506 suppresses the proteasomal degradation of ADRP, a substrate of AIP4, by inhibiting the spartin-TIP47 interaction and thereby blocking the localization of spartin and AIP4 in lipid droplets. Tacrolimus 36-41 itchy E3 ubiquitin protein ligase Homo sapiens 208-212 24059111-0 2013 [Association of CYP3A5 and MDR1 genetic polymorphisms with the blood concentration of tacrolimus in Chinese liver and renal transplant recipients]. Tacrolimus 86-96 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 16-22 23837476-0 2013 Tacrolimus dose requirement in pediatric liver transplantation: influence of CYP3A5 gene polymorphism. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 77-83 23837476-3 2013 RESULTS: Mean stable tacrolimus daily dose requirement was higher among children who received a liver expressing CYP3A5 (carrying the CYPA3A5*1 allele) compared with those with a liver that did not express CYP3A5 (CYP3A5*3/*3 genotype): 0.29 +- 0.20 vs 0.18 +- 0.13 mg.kg(-1).d(-1), p = 0.005, respectively. Tacrolimus 21-31 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 113-119 23837476-5 2013 Time to reach stable tacrolimus therapeutic trough concentrations was prolonged among patients with a CYP3A5-expressing graft (26 vs 21 days, p = 0.04). Tacrolimus 21-31 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 102-108 23837476-6 2013 CONCLUSION: Donor CYP3A5 genotype partially explains tacrolimus dose requirement. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 18-24 23837477-0 2013 CYP3A4*22 and CYP3A combined genotypes both correlate with tacrolimus disposition in pediatric heart transplant recipients. Tacrolimus 59-69 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 23837477-0 2013 CYP3A4*22 and CYP3A combined genotypes both correlate with tacrolimus disposition in pediatric heart transplant recipients. Tacrolimus 59-69 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-5 23837477-1 2013 BACKGROUND: Tacrolimus metabolism depends on CYP3A4 and CYP3A5. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 45-51 23837477-1 2013 BACKGROUND: Tacrolimus metabolism depends on CYP3A4 and CYP3A5. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 56-62 23837477-2 2013 We aimed to determine the relationship between the CYP3A4*22 polymorphism and combined CYP3A genotypes with tacrolimus disposition in pediatric heart transplant recipients. Tacrolimus 108-118 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 51-57 23837477-2 2013 We aimed to determine the relationship between the CYP3A4*22 polymorphism and combined CYP3A genotypes with tacrolimus disposition in pediatric heart transplant recipients. Tacrolimus 108-118 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 51-56 23837477-6 2013 RESULTS: CYP3A4*22 carriers needed 30% less tacrolimus (p = 0.016) to reach similar target concentrations compared with CYP3A4*1/*1 (n = 56) carriers. Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 9-15 23837477-7 2013 Poor CYP3A metabolizers required 17% (p = 0.023) less tacrolimus than intermediate and 48% less (p < 0.0001) than extensive metabolizers. Tacrolimus 54-64 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 5-10 23837477-9 2013 CONCLUSION: Analysis of CYP3A4*22, either alone or in combination with CYP3A5*3, may help towards individualization of tacrolimus therapy in pediatric heart transplant patients. Tacrolimus 119-129 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-30 23837477-9 2013 CONCLUSION: Analysis of CYP3A4*22, either alone or in combination with CYP3A5*3, may help towards individualization of tacrolimus therapy in pediatric heart transplant patients. Tacrolimus 119-129 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 71-77 24059111-0 2013 [Association of CYP3A5 and MDR1 genetic polymorphisms with the blood concentration of tacrolimus in Chinese liver and renal transplant recipients]. Tacrolimus 86-96 ATP binding cassette subfamily B member 1 Homo sapiens 27-31 24059111-1 2013 OBJECTIVE: To investigate the association of CYP3A5 and MDR1 genetic polymorphisms with the concentration/ dose (C/D) ratio of tacrolimus for the feasibility of individualized medication. Tacrolimus 127-137 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 24059111-1 2013 OBJECTIVE: To investigate the association of CYP3A5 and MDR1 genetic polymorphisms with the concentration/ dose (C/D) ratio of tacrolimus for the feasibility of individualized medication. Tacrolimus 127-137 ATP binding cassette subfamily B member 1 Homo sapiens 56-60 24059111-10 2013 CONCLUSIONS: Determination of CYP3A5 genotype could help individualize tacrolimus dose regimen prospectively. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 30-36 24059111-11 2013 The patients with CYP3A5 *3 *3 require less dose of tacrolimus to reach the same concentrations comparing with the patients with at least one CYP3A5 * 1 allele. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 18-24 24059111-11 2013 The patients with CYP3A5 *3 *3 require less dose of tacrolimus to reach the same concentrations comparing with the patients with at least one CYP3A5 * 1 allele. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 142-148 23825664-6 2013 Induction of PAI-1 by NGF was sensitive to the calcineurin inhibitor FK506 and the specific inhibition of NFAT activation by the cell permeable VIVIT peptide. Tacrolimus 69-74 serpin family E member 1 Rattus norvegicus 13-18 23278282-8 2013 CYP3A4*1B carriers and CYP3A5 expressers, independently or when assessed as a combined CYP3A4-3A5 genotype, had significantly lower dose-normalized pre-dose (C0/dose) and 2-hour post-dose (C2/dose) concentrations of tacrolimus and metabolites. Tacrolimus 216-226 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-9 23278282-8 2013 CYP3A4*1B carriers and CYP3A5 expressers, independently or when assessed as a combined CYP3A4-3A5 genotype, had significantly lower dose-normalized pre-dose (C0/dose) and 2-hour post-dose (C2/dose) concentrations of tacrolimus and metabolites. Tacrolimus 216-226 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 23-29 23278282-8 2013 CYP3A4*1B carriers and CYP3A5 expressers, independently or when assessed as a combined CYP3A4-3A5 genotype, had significantly lower dose-normalized pre-dose (C0/dose) and 2-hour post-dose (C2/dose) concentrations of tacrolimus and metabolites. Tacrolimus 216-226 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 23278282-11 2013 Genetic polymorphism of CYP3A5 or CYP3A4 influence tacrolimus or metabolites dose-normalized concentrations but not metabolite to parent concentration ratios. Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-30 23278282-11 2013 Genetic polymorphism of CYP3A5 or CYP3A4 influence tacrolimus or metabolites dose-normalized concentrations but not metabolite to parent concentration ratios. Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 34-40 23583290-5 2013 Moreover, we also found that Abeta-induced both presynaptic and postsynaptic dysfunction can be reversed by the inhibitor of protein phosphatase 2B (PP2B), FK506, whereas only postsynaptic disruption can be ameliorated by the inhibitor of PP1/PP2A, Okadaic acid (OA). Tacrolimus 156-161 amyloid beta precursor protein Rattus norvegicus 29-34 23583290-5 2013 Moreover, we also found that Abeta-induced both presynaptic and postsynaptic dysfunction can be reversed by the inhibitor of protein phosphatase 2B (PP2B), FK506, whereas only postsynaptic disruption can be ameliorated by the inhibitor of PP1/PP2A, Okadaic acid (OA). Tacrolimus 156-161 neuropeptide Y receptor Y4 Rattus norvegicus 239-242 23769045-11 2013 Splenocytes isolated from rats treated with IL-10-imDCs plus and FK-506 produced significant amounts of IL-10 and IL-4 cytokines upon alloantigen stimulation, as confirmed using ELISPOT and ELISA. Tacrolimus 65-71 interleukin 10 Rattus norvegicus 104-109 23755290-0 2013 Tacrolimus (FK506) prevents early stages of ethanol induced hepatic fibrosis by targeting LARP6 dependent mechanism of collagen synthesis. Tacrolimus 0-10 La ribonucleoprotein 6, translational regulator Rattus norvegicus 90-95 23755290-0 2013 Tacrolimus (FK506) prevents early stages of ethanol induced hepatic fibrosis by targeting LARP6 dependent mechanism of collagen synthesis. Tacrolimus 12-17 La ribonucleoprotein 6, translational regulator Rattus norvegicus 90-95 23755290-8 2013 FK506 binding protein 3 (FKBP3) is one of cellular proteins which binds FK506 with high affinity. Tacrolimus 0-5 FKBP prolyl isomerase 3 Rattus norvegicus 25-30 23755290-10 2013 In the presence of FK506 the interaction between FKBP3 and LARP6 is weakened and so is the pull down of collagen mRNAs with FKBP3. Tacrolimus 19-24 FKBP prolyl isomerase 3 Rattus norvegicus 49-54 23755290-10 2013 In the presence of FK506 the interaction between FKBP3 and LARP6 is weakened and so is the pull down of collagen mRNAs with FKBP3. Tacrolimus 19-24 La ribonucleoprotein 6, translational regulator Rattus norvegicus 59-64 23755290-10 2013 In the presence of FK506 the interaction between FKBP3 and LARP6 is weakened and so is the pull down of collagen mRNAs with FKBP3. Tacrolimus 19-24 FKBP prolyl isomerase 3 Rattus norvegicus 124-129 23755290-11 2013 We postulate that FK506 inactivates FKBP3 and that lack of interaction of LARP6 and FKBP3 results in aberrant translation of collagen mRNAs and prevention of fibrosis. Tacrolimus 18-23 FKBP prolyl isomerase 3 Rattus norvegicus 36-41 23163400-0 2013 The CYP3A4*22 allele affects the predictive value of a pharmacogenetic algorithm predicting tacrolimus predose concentrations. Tacrolimus 92-102 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 4-10 23354810-10 2013 Prospective analysis of the CYP3A5 polymorphism may be important to ensure safe and reliable immunosuppressive therapy with tacrolimus in patients treated with ITCZ. Tacrolimus 124-134 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 28-34 23666583-2 2013 CYP3A5 genotype has a significant influence on the oral bioavailability of tacrolimus and is a potential influence on variability of exposure. Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 23666583-7 2013 Tacrolimus dose requirement was significantly higher in patients expressing CYP3A5, confirming earlier observations (P < 0.0001). Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 76-82 23762456-6 2013 After treatment with the NFAT antagonist cyclosporin A (CsA) and FK506, levels of NFAT1 in the nuclei of U87 GBM cells were dramatically reduced. Tacrolimus 65-70 nuclear factor of activated T cells 2 Homo sapiens 82-87 23557867-0 2013 CYP3A4/5 polymorphisms affect the blood level of cyclosporine and tacrolimus in Chinese renal transplant recipients. Tacrolimus 66-76 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 23557867-5 2013 RESULTS: Both CYP3A4*18B and CYP3A5*3 polymorphisms affected the tacrolimus dose-adjusted trough concentration (C0/D). Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-20 23557867-5 2013 RESULTS: Both CYP3A4*18B and CYP3A5*3 polymorphisms affected the tacrolimus dose-adjusted trough concentration (C0/D). Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 29-35 23557867-10 2013 CONCLUSIONS: Genetic polymorphisms of CYP3A5*3 and CYP3A4*18B may be partly responsible in large interindividual variability of cyclosporine and tacrolimus blood levels in Chinese renal transplant patients during the first month after transplantation. Tacrolimus 145-155 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 38-44 23557867-10 2013 CONCLUSIONS: Genetic polymorphisms of CYP3A5*3 and CYP3A4*18B may be partly responsible in large interindividual variability of cyclosporine and tacrolimus blood levels in Chinese renal transplant patients during the first month after transplantation. Tacrolimus 145-155 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 51-57 23557867-11 2013 A patient carried combined genotype of CYP3A4*1/*1-CYP3A5* 3/*3 might require lower tacrolimus doses to achieve target concentration levels. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 39-45 23557867-11 2013 A patient carried combined genotype of CYP3A4*1/*1-CYP3A5* 3/*3 might require lower tacrolimus doses to achieve target concentration levels. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 51-57 23769045-11 2013 Splenocytes isolated from rats treated with IL-10-imDCs plus and FK-506 produced significant amounts of IL-10 and IL-4 cytokines upon alloantigen stimulation, as confirmed using ELISPOT and ELISA. Tacrolimus 65-71 interleukin 4 Rattus norvegicus 114-118 23769096-7 2013 Among heart transplant recipients, VAP-1 correlated with age, presence of diabetes, insulin therapy, ejection fraction, estimated glomerular filtration rate by MDRD (Modification of Diet in Renal Disease), eGFR by CKD-EPI (Chronic Kidney Disease-Epidemiological Collaboration), use of tacrolimus, LVIDd (left ventricular internal end-diastolic dimension), New York Heart Association class and NT-proBNP. Tacrolimus 285-295 amine oxidase copper containing 3 Homo sapiens 35-40 23769096-8 2013 VAP-1 was significantly lower among patients treated with tacrolimus than cyclosporine. Tacrolimus 58-68 amine oxidase copper containing 3 Homo sapiens 0-5 23769096-10 2013 Multiple regression analysis revealed VAP-1 to be predicted in 25% by LVIDd, and use of tacrolimus in OHT. Tacrolimus 88-98 amine oxidase copper containing 3 Homo sapiens 38-43 23769096-12 2013 CONCLUDING: VAP-1 elevations in heart transplant recipients were predominantly dependent on left ventricular diameter and use of tacrolimus; however, the precise associations with the immunosuppressive regimen warrant further studies. Tacrolimus 129-139 amine oxidase copper containing 3 Homo sapiens 12-17 23518805-0 2013 Tacrolimus salvage in anti-tumor necrosis factor antibody treatment-refractory Crohn"s disease. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 27-48 24028719-9 2013 Compared with control group, the expression of TNF-beta was significantly higher in IL-2 group (73.36% +- 16.73% vs 66.61% +- 16.20%, P < 0.05), significantly lower in FK506 and FK506 plus CsA groups (P < 0.05). Tacrolimus 171-176 lymphotoxin alpha Homo sapiens 47-55 24028719-9 2013 Compared with control group, the expression of TNF-beta was significantly higher in IL-2 group (73.36% +- 16.73% vs 66.61% +- 16.20%, P < 0.05), significantly lower in FK506 and FK506 plus CsA groups (P < 0.05). Tacrolimus 171-176 interleukin 2 Homo sapiens 84-88 24028719-9 2013 Compared with control group, the expression of TNF-beta was significantly higher in IL-2 group (73.36% +- 16.73% vs 66.61% +- 16.20%, P < 0.05), significantly lower in FK506 and FK506 plus CsA groups (P < 0.05). Tacrolimus 181-186 lymphotoxin alpha Homo sapiens 47-55 24028719-9 2013 Compared with control group, the expression of TNF-beta was significantly higher in IL-2 group (73.36% +- 16.73% vs 66.61% +- 16.20%, P < 0.05), significantly lower in FK506 and FK506 plus CsA groups (P < 0.05). Tacrolimus 181-186 interleukin 2 Homo sapiens 84-88 23432535-0 2013 Individualization of tacrolimus dosage basing on cytochrome P450 3A5 polymorphism--a prospective, randomized, controlled study. Tacrolimus 21-31 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 49-68 23432535-9 2013 Results showed that CYP3A5 expressers needed more tacrolimus to reach therapeutic concentration window and were more susceptible to diltiazem-induced concentration increase than CYP3A5 non-expressers. Tacrolimus 50-60 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 23432535-10 2013 CYP3A5 polymorphism-guided dosing equation helped to determine appropriate initial doses of tacrolimus in individuals. Tacrolimus 92-102 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 23432535-11 2013 In conclusion, CYP3A5 polymorphism profoundly influences pharmacokinetics of tacrolimus and helps to individualize tacrolimus dose. Tacrolimus 77-87 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 23432535-11 2013 In conclusion, CYP3A5 polymorphism profoundly influences pharmacokinetics of tacrolimus and helps to individualize tacrolimus dose. Tacrolimus 115-125 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 23585572-1 2013 We used site-directed labeling of the type 1 ryanodine receptor (RyR1) and fluorescence resonance energy transfer (FRET) measurements to map RyR1 sequence elements forming the binding site of the 12-kDa binding protein for the immunosuppressant drug, FK506. Tacrolimus 251-256 ryanodine receptor 1 Homo sapiens 141-145 23614749-8 2013 In the tacrolimus-treated group, mRNA expression levels of IL-1alpha and TGF-beta were slightly decreased, while the others were similar with the vehicle-treated group. Tacrolimus 7-17 interleukin 1 alpha Mus musculus 59-68 23614749-8 2013 In the tacrolimus-treated group, mRNA expression levels of IL-1alpha and TGF-beta were slightly decreased, while the others were similar with the vehicle-treated group. Tacrolimus 7-17 transforming growth factor, beta 1 Mus musculus 73-81 23518805-2 2013 METHODS: We conducted a retrospective study of the use of oral tacrolimus for severe CD refractory to anti-tumor necrosis factor agents. Tacrolimus 63-73 tumor necrosis factor Homo sapiens 107-128 23518805-14 2013 CONCLUSIONS: Oral tacrolimus seems to be safe and effective in some patients with severe CD refractory to anti-tumor necrosis factor therapy, particularly at a mean trough level of 10 to 15 ng/mL. Tacrolimus 18-28 tumor necrosis factor Homo sapiens 111-132 22404745-11 2013 Moreover, the greater activities of MMP-2 and MMP-9 induced by tacrolimus than ET-1, reflected tacrolimus would enhance migration stimulatory effect in cultured NHMs. Tacrolimus 63-73 matrix metallopeptidase 2 Homo sapiens 36-41 23577651-1 2013 The use of the mammal target of rapamycin (mTOR) inhibitors has been consolidated as the therapy of election for preventing graft rejection in kidney transplant patients, despite their immunosuppressive activity is less strong than anti-calcineurin agents like tacrolimus and cyclosporine A. Tacrolimus 261-271 mechanistic target of rapamycin kinase Homo sapiens 43-47 22404745-11 2013 Moreover, the greater activities of MMP-2 and MMP-9 induced by tacrolimus than ET-1, reflected tacrolimus would enhance migration stimulatory effect in cultured NHMs. Tacrolimus 63-73 matrix metallopeptidase 9 Homo sapiens 46-51 22404745-11 2013 Moreover, the greater activities of MMP-2 and MMP-9 induced by tacrolimus than ET-1, reflected tacrolimus would enhance migration stimulatory effect in cultured NHMs. Tacrolimus 95-105 matrix metallopeptidase 2 Homo sapiens 36-41 22404745-11 2013 Moreover, the greater activities of MMP-2 and MMP-9 induced by tacrolimus than ET-1, reflected tacrolimus would enhance migration stimulatory effect in cultured NHMs. Tacrolimus 95-105 matrix metallopeptidase 9 Homo sapiens 46-51 22404745-11 2013 Moreover, the greater activities of MMP-2 and MMP-9 induced by tacrolimus than ET-1, reflected tacrolimus would enhance migration stimulatory effect in cultured NHMs. Tacrolimus 95-105 endothelin 1 Homo sapiens 79-83 22404745-12 2013 CONCLUSIONS: Topical tacrolimus can be used an effective agent for vitiligo treatment as monotherapy, maybe due to its migration stimulatory action or TNF-alpha inhibitory property, and also as a component in combination therapy with UV treatment, considering the more upregulated MMPs activities are induced and the more effective migrations are feasible by itself than ET-1. Tacrolimus 21-31 tumor necrosis factor Homo sapiens 151-160 22404745-12 2013 CONCLUSIONS: Topical tacrolimus can be used an effective agent for vitiligo treatment as monotherapy, maybe due to its migration stimulatory action or TNF-alpha inhibitory property, and also as a component in combination therapy with UV treatment, considering the more upregulated MMPs activities are induced and the more effective migrations are feasible by itself than ET-1. Tacrolimus 21-31 matrix metallopeptidase 2 Homo sapiens 281-285 22404745-12 2013 CONCLUSIONS: Topical tacrolimus can be used an effective agent for vitiligo treatment as monotherapy, maybe due to its migration stimulatory action or TNF-alpha inhibitory property, and also as a component in combination therapy with UV treatment, considering the more upregulated MMPs activities are induced and the more effective migrations are feasible by itself than ET-1. Tacrolimus 21-31 endothelin 1 Homo sapiens 371-375 23459029-0 2013 Effects of CYP3A4 and CYP3A5 polymorphisms on tacrolimus pharmacokinetics in Chinese adult renal transplant recipients: a population pharmacokinetic analysis. Tacrolimus 46-56 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 22-28 23459029-9 2013 CONCLUSION: This is the first study to extensively determine the effect of CYP3A4*1G and CYP3A5*3 genetic polymorphisms and hematocrit value on tacrolimus pharmacokinetics in Chinese renal transplant recipients. Tacrolimus 144-154 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 89-95 23483701-2 2013 The aim of the present study was to evaluate the safety and efficacy of conversion from a tacrolimus (TAC) twice daily (bid) formulation to a once daily (qd) formulation in a large cohort of adult liver transplantation (LT) patients. Tacrolimus 90-100 BH3 interacting domain death agonist Homo sapiens 120-123 23459029-2 2013 The aim of this study was to examine the association between tacrolimus pharmacokinetic variability and CYP3A4 and CYP3A5 genotypes by a population pharmacokinetic analysis based on routine drug monitoring data in adult renal transplant recipients. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-110 23459029-10 2013 The findings suggest that CYP3A5*3 and CYP3A4*1G polymorphisms and hematocrit are determinant factors in the apparent clearance of tacrolimus. Tacrolimus 131-141 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 26-32 23459029-2 2013 The aim of this study was to examine the association between tacrolimus pharmacokinetic variability and CYP3A4 and CYP3A5 genotypes by a population pharmacokinetic analysis based on routine drug monitoring data in adult renal transplant recipients. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 115-121 23459029-10 2013 The findings suggest that CYP3A5*3 and CYP3A4*1G polymorphisms and hematocrit are determinant factors in the apparent clearance of tacrolimus. Tacrolimus 131-141 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 39-45 23459029-8 2013 The apparent clearance of tacrolimus was about two-fold higher in kidney transplant patients with higher enzymatic activity of CYP3A5*1 and CYP3A4*1G (with the CYP3A5*1/*1 or *1/*3 and CYP3A4*1/*1G or CYP3A4*1G/*1G) compared with those with lower enzymatic activity (CYP3A5*3/*3 and CYP3A4*1/*1). Tacrolimus 26-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 127-133 23459029-8 2013 The apparent clearance of tacrolimus was about two-fold higher in kidney transplant patients with higher enzymatic activity of CYP3A5*1 and CYP3A4*1G (with the CYP3A5*1/*1 or *1/*3 and CYP3A4*1/*1G or CYP3A4*1G/*1G) compared with those with lower enzymatic activity (CYP3A5*3/*3 and CYP3A4*1/*1). Tacrolimus 26-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 140-146 23459029-8 2013 The apparent clearance of tacrolimus was about two-fold higher in kidney transplant patients with higher enzymatic activity of CYP3A5*1 and CYP3A4*1G (with the CYP3A5*1/*1 or *1/*3 and CYP3A4*1/*1G or CYP3A4*1G/*1G) compared with those with lower enzymatic activity (CYP3A5*3/*3 and CYP3A4*1/*1). Tacrolimus 26-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 160-166 23947183-3 2013 Tacrolimus, a calcineurin inhibitor, is usually used for immunosuppressive therapy after transplantation. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 14-35 23459029-8 2013 The apparent clearance of tacrolimus was about two-fold higher in kidney transplant patients with higher enzymatic activity of CYP3A5*1 and CYP3A4*1G (with the CYP3A5*1/*1 or *1/*3 and CYP3A4*1/*1G or CYP3A4*1G/*1G) compared with those with lower enzymatic activity (CYP3A5*3/*3 and CYP3A4*1/*1). Tacrolimus 26-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 185-191 23947183-4 2013 Tacrolimus is mainly metabolized by cytochrome P450 isozymes, CYP3A4 and CYP3A5, expressed in the intestine as well as in the liver. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 23459029-8 2013 The apparent clearance of tacrolimus was about two-fold higher in kidney transplant patients with higher enzymatic activity of CYP3A5*1 and CYP3A4*1G (with the CYP3A5*1/*1 or *1/*3 and CYP3A4*1/*1G or CYP3A4*1G/*1G) compared with those with lower enzymatic activity (CYP3A5*3/*3 and CYP3A4*1/*1). Tacrolimus 26-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 185-191 23947183-4 2013 Tacrolimus is mainly metabolized by cytochrome P450 isozymes, CYP3A4 and CYP3A5, expressed in the intestine as well as in the liver. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 73-79 23459029-8 2013 The apparent clearance of tacrolimus was about two-fold higher in kidney transplant patients with higher enzymatic activity of CYP3A5*1 and CYP3A4*1G (with the CYP3A5*1/*1 or *1/*3 and CYP3A4*1/*1G or CYP3A4*1G/*1G) compared with those with lower enzymatic activity (CYP3A5*3/*3 and CYP3A4*1/*1). Tacrolimus 26-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 160-166 23947183-7 2013 In living-donor liver transplantation, CYP3A5 phenotypes could predict the blood concentration of tacrolimus. Tacrolimus 98-108 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 39-45 23947183-8 2013 In particular, preoperative assessment of CYP3A5 genotypes in both recipients (intestine) and donors (graft liver) is required for predicting tacrolimus pharmacokinetics. Tacrolimus 142-152 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 42-48 23459029-8 2013 The apparent clearance of tacrolimus was about two-fold higher in kidney transplant patients with higher enzymatic activity of CYP3A5*1 and CYP3A4*1G (with the CYP3A5*1/*1 or *1/*3 and CYP3A4*1/*1G or CYP3A4*1G/*1G) compared with those with lower enzymatic activity (CYP3A5*3/*3 and CYP3A4*1/*1). Tacrolimus 26-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 185-191 23947183-11 2013 Furthermore, phenotypes of CYP3A5 could predict the initial dose of tacrolimus. Tacrolimus 68-78 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 27-33 23459029-9 2013 CONCLUSION: This is the first study to extensively determine the effect of CYP3A4*1G and CYP3A5*3 genetic polymorphisms and hematocrit value on tacrolimus pharmacokinetics in Chinese renal transplant recipients. Tacrolimus 144-154 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 75-81 23576755-3 2013 We screened a collection of drugs approved for human use and identified astemizole and tacrolimus, which reduced cell-surface PrP and inhibited prion replication in neuroblastoma cells. Tacrolimus 87-97 prion protein Homo sapiens 126-129 23576755-4 2013 Tacrolimus reduced total cellular PrP levels by a nontranscriptional mechanism. Tacrolimus 0-10 prion protein Homo sapiens 34-37 23617933-2 2013 The CYP3A4*18, CYP3A5*3, and MDR1-3435 variant alleles are very important, particularly in tacrolimus metabolism in organ transplant rejection. Tacrolimus 91-101 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 4-10 23613708-10 2013 Although the expression of Vegfa (p<0.05) and Ccnd1 (p<0.05) was significantly upregulated in the tacrolimus-treated group compared with that of the control group, no differences were observed between the groups in terms of other types of gene expression. Tacrolimus 104-114 vascular endothelial growth factor A Mus musculus 27-32 23617933-2 2013 The CYP3A4*18, CYP3A5*3, and MDR1-3435 variant alleles are very important, particularly in tacrolimus metabolism in organ transplant rejection. Tacrolimus 91-101 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 23617933-2 2013 The CYP3A4*18, CYP3A5*3, and MDR1-3435 variant alleles are very important, particularly in tacrolimus metabolism in organ transplant rejection. Tacrolimus 91-101 ATP binding cassette subfamily B member 1 Homo sapiens 29-33 23613708-10 2013 Although the expression of Vegfa (p<0.05) and Ccnd1 (p<0.05) was significantly upregulated in the tacrolimus-treated group compared with that of the control group, no differences were observed between the groups in terms of other types of gene expression. Tacrolimus 104-114 cyclin D1 Mus musculus 49-54 23097010-1 2013 PURPOSE: To assess the influence of the P450 oxidoreductase 28 SNP (POR 28) on tacrolimus pharmacokinetics in the Chinese population. Tacrolimus 79-89 cytochrome p450 oxidoreductase Homo sapiens 68-71 23274970-7 2013 The lower fetal blood concentrations are likely due to active efflux transport of tacrolimus from the fetus toward the mother by placental P-glycoprotein. Tacrolimus 82-92 ATP binding cassette subfamily B member 1 Homo sapiens 139-153 23691335-0 2013 Colon Mucosa Exhibits Loss of Ectopic MUC5AC Expression in Patients with Ulcerative Colitis Treated with Oral Tacrolimus. Tacrolimus 110-120 mucin 5AC, oligomeric mucus/gel-forming Homo sapiens 38-44 23435727-1 2013 The immunosuppressive drug FK506 binding proteins (FKBPs), an immunophilin family with the immunosuppressive drug FK506 binding property, exhibit peptidylprolyl cis-trans isomerase (PPIase) activity. Tacrolimus 27-32 FKBP prolyl isomerase 5 Homo sapiens 146-180 23097010-9 2013 CONCLUSIONS: The POR 28 CT genotype presented a significantly lower level of tacrolimus exposure (AUC, Cmax) compared with the POR 28 CC genotype in CYP3A5-expressing subjects. Tacrolimus 77-87 cytochrome p450 oxidoreductase Homo sapiens 17-20 23435727-1 2013 The immunosuppressive drug FK506 binding proteins (FKBPs), an immunophilin family with the immunosuppressive drug FK506 binding property, exhibit peptidylprolyl cis-trans isomerase (PPIase) activity. Tacrolimus 27-32 FKBP prolyl isomerase 5 Homo sapiens 182-188 23435727-1 2013 The immunosuppressive drug FK506 binding proteins (FKBPs), an immunophilin family with the immunosuppressive drug FK506 binding property, exhibit peptidylprolyl cis-trans isomerase (PPIase) activity. Tacrolimus 114-119 FKBP prolyl isomerase 5 Homo sapiens 146-180 23435727-1 2013 The immunosuppressive drug FK506 binding proteins (FKBPs), an immunophilin family with the immunosuppressive drug FK506 binding property, exhibit peptidylprolyl cis-trans isomerase (PPIase) activity. Tacrolimus 114-119 FKBP prolyl isomerase 5 Homo sapiens 182-188 23097010-10 2013 It suggested that the POR 28 genetic polymorphism might also be responsible for the marked interindividual variability of tacrolimus besides the CYP3A5 3 genetic polymorphism. Tacrolimus 122-132 cytochrome p450 oxidoreductase Homo sapiens 22-25 23334350-1 2013 BACKGROUND: We aimed to compare the effects of intraperitoneal ghrelin and tacrolimus on vitreous levels of tumor necrosis factor-alpha (TNF-alpha), interleukin-1 (IL-1), and interleukin-6 (IL-6) in an experimental autoimmune uveitis model. Tacrolimus 75-85 tumor necrosis factor Rattus norvegicus 108-135 23334350-14 2013 CONCLUSIONS: Tacrolimus could be effective in uveitis treatment by neutralizing or decreasing the levels of cytokines such as TNF-alpha, IL-1 and IL-6 that have a critical part in the pathogenesis of uveitis. Tacrolimus 13-23 tumor necrosis factor Rattus norvegicus 126-135 23334350-14 2013 CONCLUSIONS: Tacrolimus could be effective in uveitis treatment by neutralizing or decreasing the levels of cytokines such as TNF-alpha, IL-1 and IL-6 that have a critical part in the pathogenesis of uveitis. Tacrolimus 13-23 interleukin 6 Rattus norvegicus 146-150 23622581-0 2013 Effect of MDR1 polymorphisms on the blood concentrations of tacrolimus in Turkish renal transplant patients. Tacrolimus 60-70 ATP binding cassette subfamily B member 1 Homo sapiens 10-14 23386250-10 2013 In addition, treatment with FK-506 or 11R-VIVIT significantly reduced the mRNA levels of NFATc4 and CCR2 but not large-conductance Ca(2+)-activated K(+) channels, in the DRG after nerve injury. Tacrolimus 28-34 nuclear factor of activated T-cells 4 Rattus norvegicus 89-95 23386250-10 2013 In addition, treatment with FK-506 or 11R-VIVIT significantly reduced the mRNA levels of NFATc4 and CCR2 but not large-conductance Ca(2+)-activated K(+) channels, in the DRG after nerve injury. Tacrolimus 28-34 C-C motif chemokine receptor 2 Rattus norvegicus 100-104 23622676-12 2013 CONCLUSION: We observed that in a real-world setting far from LT centers, the switch from BID tacrolimus to QD tacrolimus in stable LT recipients is efficient (safe and effective) to improve quality of medical care, with possibly better IS adherence and improvement of renal function. Tacrolimus 94-104 BH3 interacting domain death agonist Homo sapiens 90-93 23622581-1 2013 BACKGROUND: Tacrolimus, a calcineurin inhibitor, is prescribed to prevent allograft rejection in renal transplantation. Tacrolimus 12-22 calcineurin binding protein 1 Homo sapiens 26-47 23622676-12 2013 CONCLUSION: We observed that in a real-world setting far from LT centers, the switch from BID tacrolimus to QD tacrolimus in stable LT recipients is efficient (safe and effective) to improve quality of medical care, with possibly better IS adherence and improvement of renal function. Tacrolimus 111-121 BH3 interacting domain death agonist Homo sapiens 90-93 23622581-4 2013 This study investigated associations of MDR1 gene C3435T polymorphism with tacrolimus blood concentrations and dose requirements as well as acute rejection episodes among Turkish renal transplant patients. Tacrolimus 75-85 ATP binding cassette subfamily B member 1 Homo sapiens 40-44 23622581-13 2013 CONCLUSIONS: Determination of MDR1 polymorphism may help to achieve target of tacrolimus blood concentrations. Tacrolimus 78-88 ATP binding cassette subfamily B member 1 Homo sapiens 30-34 23364483-0 2013 CYP3A and ABCB1 genetic polymorphisms on the pharmacokinetics and pharmacodynamics of tacrolimus and its metabolites (M-I and M-III). Tacrolimus 86-96 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-5 23418784-1 2013 The aryl hydrocarbon receptor-interacting protein (AIP) has been predicted to consist of an N-terminal FKBP-type peptidyl-prolyl cis/trans isomerase (PPIase) domain and a C-terminal tetratricopeptide repeat (TPR) domain, as typically found in FK506-binding immunophilins. Tacrolimus 243-248 aryl hydrocarbon receptor interacting protein Homo sapiens 4-49 23418784-1 2013 The aryl hydrocarbon receptor-interacting protein (AIP) has been predicted to consist of an N-terminal FKBP-type peptidyl-prolyl cis/trans isomerase (PPIase) domain and a C-terminal tetratricopeptide repeat (TPR) domain, as typically found in FK506-binding immunophilins. Tacrolimus 243-248 aryl hydrocarbon receptor interacting protein Homo sapiens 51-54 23364483-0 2013 CYP3A and ABCB1 genetic polymorphisms on the pharmacokinetics and pharmacodynamics of tacrolimus and its metabolites (M-I and M-III). Tacrolimus 86-96 ATP binding cassette subfamily B member 1 Homo sapiens 10-15 23364483-3 2013 RESULTS: The dose-adjusted C0 of tacrolimus and its metabolites and AUC0-12 were significantly higher and the mean fluorescence intensity (MFI) of HLA/DR in monocytes was significantly lower in patients with CYP3A5*3/*3 than in patients with CYP3A5*1/*1 or CYP3A5*1/*3. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 208-214 23364483-6 2013 In a multiple regression analysis, the presence of the CYP3A5*3/*3 genotype was a significant independent variable determining the dose-adjusted C0 of tacrolimus and its metabolites, AUC0-12, and the MFI of HLA/DR in monocytes. Tacrolimus 151-161 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 55-61 23364483-7 2013 CONCLUSIONS: This study demonstrates that the CYP3A5 genetic polymorphisms are associated with the individual differences in PK and PD as well as in C0 of tacrolimus and its metabolites. Tacrolimus 155-165 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 23470533-8 2013 Pretreatment of cells with JNK inhibitor, SP600125, or ERK inhibitor, PD98059, could inhibit FK506-induced fibroblast apoptosis, respectively. Tacrolimus 93-98 mitogen-activated protein kinase 8 Rattus norvegicus 27-30 23626544-3 2013 Interestingly, immunohistochemical staining revealed that prominent CD163(+) proinflammatory macrophages and IL-17-producing cells were infiltrating around plasma cells, which might suggest the reason for the therapeutic effect of topical tacrolimus on PLC. Tacrolimus 239-249 CD163 molecule Homo sapiens 68-73 23626544-3 2013 Interestingly, immunohistochemical staining revealed that prominent CD163(+) proinflammatory macrophages and IL-17-producing cells were infiltrating around plasma cells, which might suggest the reason for the therapeutic effect of topical tacrolimus on PLC. Tacrolimus 239-249 interleukin 17A Homo sapiens 109-114 23470533-8 2013 Pretreatment of cells with JNK inhibitor, SP600125, or ERK inhibitor, PD98059, could inhibit FK506-induced fibroblast apoptosis, respectively. Tacrolimus 93-98 Eph receptor B1 Rattus norvegicus 55-58 23470533-10 2013 These results suggest that FK506-induced fibroblast apoptosis contributes to the suppression of fibroblast proliferation and then results in the reduction of scar formation in sciatic nerve-injured rat, and that JNK and ERK are involved in FK506-induced fibroblast apoptosis. Tacrolimus 27-32 mitogen-activated protein kinase 8 Rattus norvegicus 212-215 23470533-10 2013 These results suggest that FK506-induced fibroblast apoptosis contributes to the suppression of fibroblast proliferation and then results in the reduction of scar formation in sciatic nerve-injured rat, and that JNK and ERK are involved in FK506-induced fibroblast apoptosis. Tacrolimus 27-32 Eph receptor B1 Rattus norvegicus 220-223 23470533-10 2013 These results suggest that FK506-induced fibroblast apoptosis contributes to the suppression of fibroblast proliferation and then results in the reduction of scar formation in sciatic nerve-injured rat, and that JNK and ERK are involved in FK506-induced fibroblast apoptosis. Tacrolimus 240-245 mitogen-activated protein kinase 8 Rattus norvegicus 212-215 23470533-10 2013 These results suggest that FK506-induced fibroblast apoptosis contributes to the suppression of fibroblast proliferation and then results in the reduction of scar formation in sciatic nerve-injured rat, and that JNK and ERK are involved in FK506-induced fibroblast apoptosis. Tacrolimus 240-245 Eph receptor B1 Rattus norvegicus 220-223 23497558-3 2013 Experimental data have shown that the calcineurin inhibitor tacrolimus exerts protective effects on hepatic IRI when applied intravenously or directly as a hepatic rinse. Tacrolimus 60-70 calcineurin binding protein 1 Homo sapiens 38-59 23228564-7 2013 FK506 and rapamycin reduced the association between TRPC1 and Orai1 with FK506 binding protein (52) (FKBP52) in human platelets, and between TRPC1 and the type II IP(3)R, which association is known to be crucial for the maintenance of SOCE in human platelets. Tacrolimus 0-5 transient receptor potential cation channel subfamily C member 1 Homo sapiens 52-57 23228564-7 2013 FK506 and rapamycin reduced the association between TRPC1 and Orai1 with FK506 binding protein (52) (FKBP52) in human platelets, and between TRPC1 and the type II IP(3)R, which association is known to be crucial for the maintenance of SOCE in human platelets. Tacrolimus 0-5 ORAI calcium release-activated calcium modulator 1 Homo sapiens 62-67 23228564-7 2013 FK506 and rapamycin reduced the association between TRPC1 and Orai1 with FK506 binding protein (52) (FKBP52) in human platelets, and between TRPC1 and the type II IP(3)R, which association is known to be crucial for the maintenance of SOCE in human platelets. Tacrolimus 0-5 FKBP prolyl isomerase 4 Homo sapiens 101-107 23228564-7 2013 FK506 and rapamycin reduced the association between TRPC1 and Orai1 with FK506 binding protein (52) (FKBP52) in human platelets, and between TRPC1 and the type II IP(3)R, which association is known to be crucial for the maintenance of SOCE in human platelets. Tacrolimus 0-5 transient receptor potential cation channel subfamily C member 1 Homo sapiens 141-146 23228564-10 2013 Finally, in MEG 01 incubated with FK506 we observed a reduction in TRPC1/FKBP52 coupling, and similarly, FKBP52 silencing reduced the association between IP3R type II and TRPC1 during SOCE. Tacrolimus 34-39 transient receptor potential cation channel subfamily C member 1 Homo sapiens 67-72 23228564-10 2013 Finally, in MEG 01 incubated with FK506 we observed a reduction in TRPC1/FKBP52 coupling, and similarly, FKBP52 silencing reduced the association between IP3R type II and TRPC1 during SOCE. Tacrolimus 34-39 FKBP prolyl isomerase 4 Homo sapiens 73-79 23228564-10 2013 Finally, in MEG 01 incubated with FK506 we observed a reduction in TRPC1/FKBP52 coupling, and similarly, FKBP52 silencing reduced the association between IP3R type II and TRPC1 during SOCE. Tacrolimus 34-39 FKBP prolyl isomerase 4 Homo sapiens 105-111 23228564-10 2013 Finally, in MEG 01 incubated with FK506 we observed a reduction in TRPC1/FKBP52 coupling, and similarly, FKBP52 silencing reduced the association between IP3R type II and TRPC1 during SOCE. Tacrolimus 34-39 inositol 1,4,5-trisphosphate receptor type 3 Homo sapiens 154-158 23228564-10 2013 Finally, in MEG 01 incubated with FK506 we observed a reduction in TRPC1/FKBP52 coupling, and similarly, FKBP52 silencing reduced the association between IP3R type II and TRPC1 during SOCE. Tacrolimus 34-39 transient receptor potential cation channel subfamily C member 1 Homo sapiens 171-176 23269193-10 2013 In vivo combination therapy of DTCM-G plus either tacrolimus or DHMEQ significantly suppressed alloreactive interferon-gamma-producing precursors and markedly prolonged cardiac allograft survival. Tacrolimus 50-60 interferon gamma Mus musculus 108-124 23227980-2 2013 Specifically, the use of the calcineurin inhibitor tacrolimus is problematic because of a narrow therapeutic range, a high interindividual variability of trough levels, and multiple interactions with combination antiretroviral therapy (cART). Tacrolimus 51-61 calcineurin binding protein 1 Homo sapiens 29-50 23218920-10 2013 Uteri of the nontreated transplanted group showed elevated mRNA expression of IL-1alpha and IP-10 and reduced galectin-1, compared with the tacrolimus-treated transplanted group. Tacrolimus 140-150 C-X-C motif chemokine ligand 10 Rattus norvegicus 92-97 23218920-12 2013 CONCLUSION(S): Tacrolimus monotherapy suppresses rejection of an allotransplanted uterus and normalizes the expression of IL-1alpha and IP-10 and prevents T-lymphocyte infiltration. Tacrolimus 15-25 interleukin 1 alpha Rattus norvegicus 122-131 23218920-12 2013 CONCLUSION(S): Tacrolimus monotherapy suppresses rejection of an allotransplanted uterus and normalizes the expression of IL-1alpha and IP-10 and prevents T-lymphocyte infiltration. Tacrolimus 15-25 C-X-C motif chemokine ligand 10 Rattus norvegicus 136-141 23473526-8 2013 The independent risk factors for UTI with concomitant bacteremia in multivariate analysis were immunosuppression with tacrolimus (adjusted odds ratio [AOR] 3.17; 95% confidence interval [CI] 1.29-7.75; P = 0.011) and baseline serum creatinine level >1.3 mg/dL before first UTI (AOR 2.55; 95% CI 1.02-6.36; P = 0.045). Tacrolimus 118-128 alpha-1-microglobulin/bikunin precursor Homo sapiens 33-36 23473526-8 2013 The independent risk factors for UTI with concomitant bacteremia in multivariate analysis were immunosuppression with tacrolimus (adjusted odds ratio [AOR] 3.17; 95% confidence interval [CI] 1.29-7.75; P = 0.011) and baseline serum creatinine level >1.3 mg/dL before first UTI (AOR 2.55; 95% CI 1.02-6.36; P = 0.045). Tacrolimus 118-128 alpha-1-microglobulin/bikunin precursor Homo sapiens 276-279 23473526-10 2013 CONCLUSION: From this study, we found that E coli tends to have resistance to commonly used empirical antibiotics in this modern era and that patients who use the immunosuppressant tacrolimus and have baseline serum creatinine level >1.3 mg/dL before their first UTI have a tendency to suffer from concomitant bacteremia and even sepsis. Tacrolimus 181-191 alpha-1-microglobulin/bikunin precursor Homo sapiens 266-269 23481136-1 2013 BACKGROUND AND AIMS: Tacrolimus is a macrolide immunosuppressant used for prevention of allograft rejection in organ transplantation and metabolized in the liver and intestine by cytochrome P450 3A4 (CYP3A4) enzyme. Tacrolimus 21-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 179-198 23481136-1 2013 BACKGROUND AND AIMS: Tacrolimus is a macrolide immunosuppressant used for prevention of allograft rejection in organ transplantation and metabolized in the liver and intestine by cytochrome P450 3A4 (CYP3A4) enzyme. Tacrolimus 21-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 200-206 23274966-8 2013 For example, tacrolimus was more potent in inhibiting CMV-specific Th1 cytokines versus Th2, whereas MPA preferentially inhibited Th2 cytokines. Tacrolimus 13-23 negative elongation factor complex member C/D Homo sapiens 67-70 23238171-3 2013 Upon action of acid (p-toluenesulfonic acid in toluene), tacrolimus is dehydrated by loss of water from the beta-hydroxyketone moiety with partial inversion of configuration at C-8, resulting in formation of 5-deoxy-Delta(5,6)-tacrolimus and 5-deoxy-Delta(5,6)-8-epitacrolimus. Tacrolimus 57-67 homeobox C8 Homo sapiens 177-180 23274966-9 2013 In a comparison of the relative potency of each drug at different dosing ranges, tacrolimus had the strongest Th1 inhibitory effect (median inhibition of interferon-gamma at 97.5%; P=0.004-0.008) followed by sirolimus (median inhibition at 82.4%). Tacrolimus 81-91 negative elongation factor complex member C/D Homo sapiens 110-113 23274966-9 2013 In a comparison of the relative potency of each drug at different dosing ranges, tacrolimus had the strongest Th1 inhibitory effect (median inhibition of interferon-gamma at 97.5%; P=0.004-0.008) followed by sirolimus (median inhibition at 82.4%). Tacrolimus 81-91 interferon gamma Homo sapiens 154-170 23175667-2 2013 Tacrolimus is mainly metabolized via CYP3A4/5, whereas CYP2C19 and CYP3A4/5 are responsible for omeprazole metabolism. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-45 23313512-6 2013 Here we screened for genes whose expression in the tail stumps was altered by FK506 treatment during the refractory period and identified a Xenopus homolog of phytanoyl-CoA dioxygenase (PhyH)-like. Tacrolimus 78-83 phytanoyl-CoA 2-hydroxylase S homeolog Xenopus laevis 186-190 23390586-1 2013 FK506 (Tacrolimus) is a potent inhibitor of calcineurin that blocks IL2 production and is widely used to prevent transplant rejection and treat autoimmunity. Tacrolimus 0-5 interleukin 2 Mus musculus 68-71 23390586-1 2013 FK506 (Tacrolimus) is a potent inhibitor of calcineurin that blocks IL2 production and is widely used to prevent transplant rejection and treat autoimmunity. Tacrolimus 7-17 interleukin 2 Mus musculus 68-71 23153588-5 2013 Cyclosporine A, FK506, and pimecrolimus inhibited ABCA1-mediated cholesterol efflux in a concentration-dependent manner, with IC(50) of 7.6, 13.6, and 7.0muM, respectively. Tacrolimus 16-21 phospholipid-transporting ATPase ABCA1 Mesocricetus auratus 50-55 23406906-0 2013 Regulatory effect of calcineurin inhibitor, tacrolimus, on IL-6/sIL-6R-mediated RANKL expression through JAK2-STAT3-SOCS3 signaling pathway in fibroblast-like synoviocytes. Tacrolimus 44-54 interleukin 6 Mus musculus 59-63 23406906-0 2013 Regulatory effect of calcineurin inhibitor, tacrolimus, on IL-6/sIL-6R-mediated RANKL expression through JAK2-STAT3-SOCS3 signaling pathway in fibroblast-like synoviocytes. Tacrolimus 44-54 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 80-85 23406906-0 2013 Regulatory effect of calcineurin inhibitor, tacrolimus, on IL-6/sIL-6R-mediated RANKL expression through JAK2-STAT3-SOCS3 signaling pathway in fibroblast-like synoviocytes. Tacrolimus 44-54 Janus kinase 2 Mus musculus 105-109 23406906-0 2013 Regulatory effect of calcineurin inhibitor, tacrolimus, on IL-6/sIL-6R-mediated RANKL expression through JAK2-STAT3-SOCS3 signaling pathway in fibroblast-like synoviocytes. Tacrolimus 44-54 signal transducer and activator of transcription 3 Mus musculus 110-115 23406906-0 2013 Regulatory effect of calcineurin inhibitor, tacrolimus, on IL-6/sIL-6R-mediated RANKL expression through JAK2-STAT3-SOCS3 signaling pathway in fibroblast-like synoviocytes. Tacrolimus 44-54 suppressor of cytokine signaling 3 Mus musculus 116-121 23406906-1 2013 INTRODUCTION: This study investigated whether the calcineurin inhibitor, tacrolimus, suppresses receptor activator of NF-kappaB ligand (RANKL) expression in fibroblast-like synoviocytes (FLS) through regulation of IL-6/Janus activated kinase (JAK2)/signal transducer and activator of transcription-3 (STAT3) and suppressor of cytokine signaling (SOCS3) signaling. Tacrolimus 73-83 calcineurin binding protein 1 Mus musculus 50-71 23406906-1 2013 INTRODUCTION: This study investigated whether the calcineurin inhibitor, tacrolimus, suppresses receptor activator of NF-kappaB ligand (RANKL) expression in fibroblast-like synoviocytes (FLS) through regulation of IL-6/Janus activated kinase (JAK2)/signal transducer and activator of transcription-3 (STAT3) and suppressor of cytokine signaling (SOCS3) signaling. Tacrolimus 73-83 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 136-141 23406906-1 2013 INTRODUCTION: This study investigated whether the calcineurin inhibitor, tacrolimus, suppresses receptor activator of NF-kappaB ligand (RANKL) expression in fibroblast-like synoviocytes (FLS) through regulation of IL-6/Janus activated kinase (JAK2)/signal transducer and activator of transcription-3 (STAT3) and suppressor of cytokine signaling (SOCS3) signaling. Tacrolimus 73-83 interleukin 6 Mus musculus 214-218 23406906-1 2013 INTRODUCTION: This study investigated whether the calcineurin inhibitor, tacrolimus, suppresses receptor activator of NF-kappaB ligand (RANKL) expression in fibroblast-like synoviocytes (FLS) through regulation of IL-6/Janus activated kinase (JAK2)/signal transducer and activator of transcription-3 (STAT3) and suppressor of cytokine signaling (SOCS3) signaling. Tacrolimus 73-83 Janus kinase 2 Mus musculus 243-247 23406906-1 2013 INTRODUCTION: This study investigated whether the calcineurin inhibitor, tacrolimus, suppresses receptor activator of NF-kappaB ligand (RANKL) expression in fibroblast-like synoviocytes (FLS) through regulation of IL-6/Janus activated kinase (JAK2)/signal transducer and activator of transcription-3 (STAT3) and suppressor of cytokine signaling (SOCS3) signaling. Tacrolimus 73-83 signal transducer and activator of transcription 3 Mus musculus 249-299 23406906-1 2013 INTRODUCTION: This study investigated whether the calcineurin inhibitor, tacrolimus, suppresses receptor activator of NF-kappaB ligand (RANKL) expression in fibroblast-like synoviocytes (FLS) through regulation of IL-6/Janus activated kinase (JAK2)/signal transducer and activator of transcription-3 (STAT3) and suppressor of cytokine signaling (SOCS3) signaling. Tacrolimus 73-83 signal transducer and activator of transcription 3 Mus musculus 301-306 23406906-1 2013 INTRODUCTION: This study investigated whether the calcineurin inhibitor, tacrolimus, suppresses receptor activator of NF-kappaB ligand (RANKL) expression in fibroblast-like synoviocytes (FLS) through regulation of IL-6/Janus activated kinase (JAK2)/signal transducer and activator of transcription-3 (STAT3) and suppressor of cytokine signaling (SOCS3) signaling. Tacrolimus 73-83 suppressor of cytokine signaling 3 Mus musculus 346-351 23406906-4 2013 Immunofluorescent staining was performed to identify the effect of tacrolimus on RANKL and SOCS3. Tacrolimus 67-77 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 81-86 23406906-4 2013 Immunofluorescent staining was performed to identify the effect of tacrolimus on RANKL and SOCS3. Tacrolimus 67-77 suppressor of cytokine signaling 3 Mus musculus 91-96 23406906-7 2013 Inhibitory effects of tacrolimus on RANKL expression in a serum-induced arthritis mice model were identified. Tacrolimus 22-32 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 36-41 23406906-8 2013 Tacrolimus inhibits RANKL expression in IL-6/sIL-6R-stimulated FLS by suppressing STAT3. Tacrolimus 0-10 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 20-25 23406906-8 2013 Tacrolimus inhibits RANKL expression in IL-6/sIL-6R-stimulated FLS by suppressing STAT3. Tacrolimus 0-10 interleukin 6 Mus musculus 40-44 23406906-8 2013 Tacrolimus inhibits RANKL expression in IL-6/sIL-6R-stimulated FLS by suppressing STAT3. Tacrolimus 0-10 signal transducer and activator of transcription 3 Mus musculus 82-87 23406906-9 2013 Among negative regulators of the JAK/STAT pathway, such as CIS1, SOCS1, and SOCS3, only SOCS3 is significantly induced by tacrolimus. Tacrolimus 122-132 cytokine inducible SH2-containing protein Mus musculus 59-63 23406906-9 2013 Among negative regulators of the JAK/STAT pathway, such as CIS1, SOCS1, and SOCS3, only SOCS3 is significantly induced by tacrolimus. Tacrolimus 122-132 suppressor of cytokine signaling 1 Mus musculus 65-70 23406906-9 2013 Among negative regulators of the JAK/STAT pathway, such as CIS1, SOCS1, and SOCS3, only SOCS3 is significantly induced by tacrolimus. Tacrolimus 122-132 suppressor of cytokine signaling 3 Mus musculus 76-81 23406906-9 2013 Among negative regulators of the JAK/STAT pathway, such as CIS1, SOCS1, and SOCS3, only SOCS3 is significantly induced by tacrolimus. Tacrolimus 122-132 suppressor of cytokine signaling 3 Mus musculus 88-93 23406906-10 2013 As compared to dexamethasone and methotrexate, tacrolimus more potently suppresses RANKL expression in FLS. Tacrolimus 47-57 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 83-88 23406906-11 2013 By up-regulating SOCS3, tacrolimus down-regulates activation of the JAK-STAT pathway by IL-6/sIL-6R trans-signaling, thus decreasing RANKL expression in FLS. Tacrolimus 24-34 suppressor of cytokine signaling 3 Mus musculus 17-22 23406906-11 2013 By up-regulating SOCS3, tacrolimus down-regulates activation of the JAK-STAT pathway by IL-6/sIL-6R trans-signaling, thus decreasing RANKL expression in FLS. Tacrolimus 24-34 interleukin 6 Mus musculus 88-92 23406906-11 2013 By up-regulating SOCS3, tacrolimus down-regulates activation of the JAK-STAT pathway by IL-6/sIL-6R trans-signaling, thus decreasing RANKL expression in FLS. Tacrolimus 24-34 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 133-138 23406906-12 2013 CONCLUSIONS: These data suggest that tacrolimus might affect the RANKL expression in IL-6 stimulated FLS through STAT3 suppression, together with up-regulation of SOCS3. Tacrolimus 37-47 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 65-70 23406906-12 2013 CONCLUSIONS: These data suggest that tacrolimus might affect the RANKL expression in IL-6 stimulated FLS through STAT3 suppression, together with up-regulation of SOCS3. Tacrolimus 37-47 interleukin 6 Mus musculus 85-89 23406906-12 2013 CONCLUSIONS: These data suggest that tacrolimus might affect the RANKL expression in IL-6 stimulated FLS through STAT3 suppression, together with up-regulation of SOCS3. Tacrolimus 37-47 signal transducer and activator of transcription 3 Mus musculus 113-118 23406906-12 2013 CONCLUSIONS: These data suggest that tacrolimus might affect the RANKL expression in IL-6 stimulated FLS through STAT3 suppression, together with up-regulation of SOCS3. Tacrolimus 37-47 suppressor of cytokine signaling 3 Mus musculus 163-168 22706623-1 2013 BACKGROUND AND OBJECTIVES: Tacrolimus(PR) is a new prolonged-release once-daily formulation of the calcineurin inhibitor tacrolimus, currently used in adult transplant patients. Tacrolimus 27-37 calcineurin binding protein 1 Homo sapiens 99-120 22706623-1 2013 BACKGROUND AND OBJECTIVES: Tacrolimus(PR) is a new prolonged-release once-daily formulation of the calcineurin inhibitor tacrolimus, currently used in adult transplant patients. Tacrolimus 121-131 calcineurin binding protein 1 Homo sapiens 99-120 23175667-2 2013 Tacrolimus is mainly metabolized via CYP3A4/5, whereas CYP2C19 and CYP3A4/5 are responsible for omeprazole metabolism. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-43 23175667-3 2013 Omeprazole inhibits tacrolimus metabolism via CYP3A5 in patients carrying variant alleles of CYP2C19, increasing tacrolimus blood concentrations. Tacrolimus 20-30 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 23175667-3 2013 Omeprazole inhibits tacrolimus metabolism via CYP3A5 in patients carrying variant alleles of CYP2C19, increasing tacrolimus blood concentrations. Tacrolimus 20-30 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 93-100 23175667-3 2013 Omeprazole inhibits tacrolimus metabolism via CYP3A5 in patients carrying variant alleles of CYP2C19, increasing tacrolimus blood concentrations. Tacrolimus 113-123 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 23175667-3 2013 Omeprazole inhibits tacrolimus metabolism via CYP3A5 in patients carrying variant alleles of CYP2C19, increasing tacrolimus blood concentrations. Tacrolimus 113-123 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 93-100 23175667-7 2013 Genotyping of CYP3A5 and CYP2C19 in renal transplantation should be considered to be of interest when treating with tacrolimus and omeprazole, because CYP2C19*2/*2 variant indirectly elicits an increase of tacrolimus blood levels and, in our study population, the adverse effects described. Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 23175667-7 2013 Genotyping of CYP3A5 and CYP2C19 in renal transplantation should be considered to be of interest when treating with tacrolimus and omeprazole, because CYP2C19*2/*2 variant indirectly elicits an increase of tacrolimus blood levels and, in our study population, the adverse effects described. Tacrolimus 116-126 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 25-32 23175667-7 2013 Genotyping of CYP3A5 and CYP2C19 in renal transplantation should be considered to be of interest when treating with tacrolimus and omeprazole, because CYP2C19*2/*2 variant indirectly elicits an increase of tacrolimus blood levels and, in our study population, the adverse effects described. Tacrolimus 116-126 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 151-158 23175667-7 2013 Genotyping of CYP3A5 and CYP2C19 in renal transplantation should be considered to be of interest when treating with tacrolimus and omeprazole, because CYP2C19*2/*2 variant indirectly elicits an increase of tacrolimus blood levels and, in our study population, the adverse effects described. Tacrolimus 206-216 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 23175667-7 2013 Genotyping of CYP3A5 and CYP2C19 in renal transplantation should be considered to be of interest when treating with tacrolimus and omeprazole, because CYP2C19*2/*2 variant indirectly elicits an increase of tacrolimus blood levels and, in our study population, the adverse effects described. Tacrolimus 206-216 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 25-32 23175667-7 2013 Genotyping of CYP3A5 and CYP2C19 in renal transplantation should be considered to be of interest when treating with tacrolimus and omeprazole, because CYP2C19*2/*2 variant indirectly elicits an increase of tacrolimus blood levels and, in our study population, the adverse effects described. Tacrolimus 206-216 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 151-158 23134420-6 2013 When we chronically expose WT cells to cyclosporine A and FK-506 we find that catecholamine release per vesicle and pre-spike foot (PSF) signal parameters are decreased, similar to that in RCAN1(ox) cells. Tacrolimus 58-64 regulator of calcineurin 1 Homo sapiens 189-194 23627041-3 2013 During the preparation, amphiphilic copolymer MPEG-PCL was chosen to form the unique core-shell structure, and tacrolimus was loaded into the hydrophobic core due to its great hydrophobicity. Tacrolimus 111-121 polycystic kidney disease 2-like 1 Mus musculus 51-54 23627041-5 2013 For the tacrolimus loaded MPEG-PCL (2000-2000) micelles, the mean particle size and drug entrapment efficiency were ca. Tacrolimus 8-18 polycystic kidney disease 2-like 1 Mus musculus 31-34 23627041-12 2013 Therefore, micelles of MPEG-PCL could be a very promising novel vehicle for tacrolimus. Tacrolimus 76-86 polycystic kidney disease 2-like 1 Mus musculus 28-31 23160140-1 2013 Cyclosporin A (CsA), tacrolimus and rapamycin are immunosuppressive agents (IAs) associated with insulin resistance and dyslipidemia, although their molecular effects on lipid metabolism in adipose tissue are unknown. Tacrolimus 21-31 insulin Homo sapiens 97-104 23089672-9 2013 There is evidence for the influence of CYP3A5 polymorphism on tacrolimus dose, although the influence on response is less studied. Tacrolimus 62-72 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 39-45 23305836-4 2013 Although FKBP12 was first identified as the principal intracellular target for the immunosuppressive drugs, FK506 and rapamycin, new insights into the role of FKBPs have since emerged. Tacrolimus 108-113 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 9-15 23160140-7 2013 These findings suggest that CsA, tacrolimus and rapamycin enhance lipolysis, inhibit lipid storage and expression of lipogenic genes in adipose tissue, which may contribute to the development of dyslipidemia and insulin resistance associated with immunosuppressive therapy. Tacrolimus 33-43 insulin Homo sapiens 212-219 23220162-9 2013 The hydroxyproline content, the number of fibroblasts, the mRNA expression level of IL-2 and TGF-beta1 in 0.1mg/ml FK506 group were significantly less than those of 0.05 mg/ml FK506 group, 0.01 mg/ml FK506 group and control group. Tacrolimus 115-120 interleukin 2 Rattus norvegicus 84-88 23220162-9 2013 The hydroxyproline content, the number of fibroblasts, the mRNA expression level of IL-2 and TGF-beta1 in 0.1mg/ml FK506 group were significantly less than those of 0.05 mg/ml FK506 group, 0.01 mg/ml FK506 group and control group. Tacrolimus 115-120 transforming growth factor, beta 1 Rattus norvegicus 93-102 23224887-4 2013 This suggests that CypA has a role in acquisition of function competence of NCX1 protein.Unlike CsA treatment, which affects the functional expression of all three mammalian NCX proteins similarly, FK506 and rapamycin treatment modulates only the functional expression of NCX2 and NCX3 proteins. Tacrolimus 198-203 peptidylprolyl isomerase A Homo sapiens 19-23 23107770-0 2013 Influence of CYP3A4, CYP3A5 and MDR-1 polymorphisms on tacrolimus pharmacokinetics and early renal dysfunction in liver transplant recipients. Tacrolimus 55-65 ATP binding cassette subfamily B member 1 Homo sapiens 32-37 23107770-2 2013 The oral bioavailability of tacrolimus varies greatly between individuals and depends largely on the activity of both the cytochrome P450 3A (CYP3A) subfamily and P-glycoprotein (P-gp). Tacrolimus 28-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 122-140 23107770-2 2013 The oral bioavailability of tacrolimus varies greatly between individuals and depends largely on the activity of both the cytochrome P450 3A (CYP3A) subfamily and P-glycoprotein (P-gp). Tacrolimus 28-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 142-147 23107770-2 2013 The oral bioavailability of tacrolimus varies greatly between individuals and depends largely on the activity of both the cytochrome P450 3A (CYP3A) subfamily and P-glycoprotein (P-gp). Tacrolimus 28-38 ATP binding cassette subfamily B member 1 Homo sapiens 163-177 23107770-2 2013 The oral bioavailability of tacrolimus varies greatly between individuals and depends largely on the activity of both the cytochrome P450 3A (CYP3A) subfamily and P-glycoprotein (P-gp). Tacrolimus 28-38 ATP binding cassette subfamily B member 1 Homo sapiens 179-183 23107770-3 2013 The possible influence of single nucleotide polymorphisms (SNPs) of CYP3A subfamily and P-gp (MDR-1) in liver transplant recipients has recently been indicated as one of the most important variables affecting the pharmacokinetics of tacrolimus and the renal injury induced by tacrolimus. Tacrolimus 233-243 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-73 23107770-3 2013 The possible influence of single nucleotide polymorphisms (SNPs) of CYP3A subfamily and P-gp (MDR-1) in liver transplant recipients has recently been indicated as one of the most important variables affecting the pharmacokinetics of tacrolimus and the renal injury induced by tacrolimus. Tacrolimus 233-243 ATP binding cassette subfamily B member 1 Homo sapiens 88-92 23107770-3 2013 The possible influence of single nucleotide polymorphisms (SNPs) of CYP3A subfamily and P-gp (MDR-1) in liver transplant recipients has recently been indicated as one of the most important variables affecting the pharmacokinetics of tacrolimus and the renal injury induced by tacrolimus. Tacrolimus 233-243 ATP binding cassette subfamily B member 1 Homo sapiens 94-99 23107770-3 2013 The possible influence of single nucleotide polymorphisms (SNPs) of CYP3A subfamily and P-gp (MDR-1) in liver transplant recipients has recently been indicated as one of the most important variables affecting the pharmacokinetics of tacrolimus and the renal injury induced by tacrolimus. Tacrolimus 276-286 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-73 23107770-3 2013 The possible influence of single nucleotide polymorphisms (SNPs) of CYP3A subfamily and P-gp (MDR-1) in liver transplant recipients has recently been indicated as one of the most important variables affecting the pharmacokinetics of tacrolimus and the renal injury induced by tacrolimus. Tacrolimus 276-286 ATP binding cassette subfamily B member 1 Homo sapiens 88-92 23107770-3 2013 The possible influence of single nucleotide polymorphisms (SNPs) of CYP3A subfamily and P-gp (MDR-1) in liver transplant recipients has recently been indicated as one of the most important variables affecting the pharmacokinetics of tacrolimus and the renal injury induced by tacrolimus. Tacrolimus 276-286 ATP binding cassette subfamily B member 1 Homo sapiens 94-99 23107770-9 2013 RESULTS: The daily dose of tacrolimus was higher for recipients with CYP3A5 1/1 (AA) genotype than those with CYP3A5 3/ 3 (GG) genotype [3.0 (2.0-4.0) versus 2.0 (1.5-2.5) mg/d, P<0.05]. Tacrolimus 27-37 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 69-79 23107770-9 2013 RESULTS: The daily dose of tacrolimus was higher for recipients with CYP3A5 1/1 (AA) genotype than those with CYP3A5 3/ 3 (GG) genotype [3.0 (2.0-4.0) versus 2.0 (1.5-2.5) mg/d, P<0.05]. Tacrolimus 27-37 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 69-75 23107770-13 2013 CONCLUSION: CYP3A5 6986A>G genetic polymorphism affected daily dose requirements, concentration and nephrotoxicity of tacrolimus. Tacrolimus 121-131 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 23224887-7 2013 Exchange of the large cytosolic loop of NCX3 with that of NCX1 renders the mutant protein insensitive to FK506. Tacrolimus 105-110 solute carrier family 8 member A3 Homo sapiens 40-44 23224887-7 2013 Exchange of the large cytosolic loop of NCX3 with that of NCX1 renders the mutant protein insensitive to FK506. Tacrolimus 105-110 solute carrier family 8 member A1 Homo sapiens 58-62 23224887-4 2013 This suggests that CypA has a role in acquisition of function competence of NCX1 protein.Unlike CsA treatment, which affects the functional expression of all three mammalian NCX proteins similarly, FK506 and rapamycin treatment modulates only the functional expression of NCX2 and NCX3 proteins. Tacrolimus 198-203 solute carrier family 8 member A1 Homo sapiens 76-80 23224887-4 2013 This suggests that CypA has a role in acquisition of function competence of NCX1 protein.Unlike CsA treatment, which affects the functional expression of all three mammalian NCX proteins similarly, FK506 and rapamycin treatment modulates only the functional expression of NCX2 and NCX3 proteins. Tacrolimus 198-203 T cell leukemia homeobox 2 Homo sapiens 76-79 23224887-5 2013 FK506 reduces NCX2 and NCX3 surface expression and transport activity without affecting cell NCX protein. Tacrolimus 0-5 solute carrier family 8 member A2 Homo sapiens 14-18 23224887-5 2013 FK506 reduces NCX2 and NCX3 surface expression and transport activity without affecting cell NCX protein. Tacrolimus 0-5 solute carrier family 8 member A3 Homo sapiens 23-27 23224887-5 2013 FK506 reduces NCX2 and NCX3 surface expression and transport activity without affecting cell NCX protein. Tacrolimus 0-5 T cell leukemia homeobox 2 Homo sapiens 14-17 24189425-2 2013 The concentration/dose (C/D) ratio of tacrolimus in patients carrying graft liver with CYP3A4*1/*1 was significantly higher during 7 d after surgery than in that with CYP3A4*1/*1G (214 vs. 157 [ng/mL]/[mg/kg/day], p<0.01). Tacrolimus 38-48 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 87-93 23902969-9 2013 Basic experimental reports indicated that the antipruritic effect of tacrolimus is probably dependent on depleting substance P, followed by transient induction. Tacrolimus 69-79 tachykinin 1 Mus musculus 115-126 24189425-8 2013 Thus, we elucidated that CYP3A4*1G genotype in the intestine was an important indicator of the pharmacokinetics of tacrolimus, whereas this genotype in the graft liver tended to influence the frequency of acute cellular rejection after transplantation. Tacrolimus 115-125 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-31 23689569-6 2013 In progressive IgAN patients whose clinical and pathological manifestations are more severe, active therapy may be considered including glucocorticoid therapy, cyclophosphamide, azathioprine, mycophenolate mofetil, tacrolimus, and other immunosuppressants. Tacrolimus 215-225 IGAN1 Homo sapiens 15-19 24308005-3 2013 Here, we report that the Schistosoma japonicum recombinant protein (SjGST-32) combined with tacrolimus (FK506) augmented the induction of Tfh cells, which expressed the canonical markers CXCR5, BCL6, and IL-21, and enhanced the humoral immune responses in BALB/c mice. Tacrolimus 104-109 interleukin 21 Mus musculus 204-209 23676788-6 2013 Because the intracellular receptor of sirolimus and tacrolimus is FK506-binding protein 12, we switched tacrolimus to cyclosporine at a dose of 60 mg/d to avoid competitive inhibition between these 2 drugs. Tacrolimus 52-62 FKBP prolyl isomerase 1A Homo sapiens 66-90 27121561-0 2013 Influence of CYP3A5 6986A > G and ABCB1 3435C > T Polymorphisms on Adverse Events Associated With Tacrolimus in Jordanian Pediatric Renal Transplant Patients. Tacrolimus 104-114 ATP binding cassette subfamily B member 1 Homo sapiens 37-42 27121561-1 2013 The aim of the study is to investigate the influence of ABCB1(3435) and CYP3A5(6986) polymorphisms, tacrolimus troughs and clinical factors on the time of adverse events associated with tacrolimus in pediatric kidney transplant patients. Tacrolimus 186-196 ATP binding cassette subfamily B member 1 Homo sapiens 56-61 27121561-1 2013 The aim of the study is to investigate the influence of ABCB1(3435) and CYP3A5(6986) polymorphisms, tacrolimus troughs and clinical factors on the time of adverse events associated with tacrolimus in pediatric kidney transplant patients. Tacrolimus 186-196 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 72-78 23689577-10 2013 All of the side effects were mild and controlled, and there were fewer side effects in the tacrolimus group compared with the CTX group, indicating a better treatment tolerance in the tacrolimus group. Tacrolimus 184-194 cytochrome P450 family 27 subfamily A member 1 Homo sapiens 126-129 23149872-0 2013 Influence of SLCO1B3 genetic variations on tacrolimus pharmacokinetics in renal transplant recipients. Tacrolimus 43-53 solute carrier organic anion transporter family member 1B3 Homo sapiens 13-20 23149872-3 2013 Genetic variations within the solute carrier (SLCO) 1B3 gene that encodes OATP1B3 may contribute to interindividual differences in tacrolimus disposition. Tacrolimus 131-141 solute carrier organic anion transporter family member 1B3 Homo sapiens 30-55 23149872-3 2013 Genetic variations within the solute carrier (SLCO) 1B3 gene that encodes OATP1B3 may contribute to interindividual differences in tacrolimus disposition. Tacrolimus 131-141 solute carrier organic anion transporter family member 1B3 Homo sapiens 74-81 23149872-4 2013 The purpose of the present study is to investigate the association between SLCO1B3 polymorphisms and tacrolimus pharmacokinetics in renal transplant recipients. Tacrolimus 101-111 solute carrier organic anion transporter family member 1B3 Homo sapiens 75-82 23149872-7 2013 This study shows, for the first time, that SLCO1B3 polymorphism is associated with tacrolimus exposure in the early post-transplant period. Tacrolimus 83-93 solute carrier organic anion transporter family member 1B3 Homo sapiens 43-50 23174045-8 2013 Western blot and real-time PCR revealed that clobetasol significantly decreased claudin-1,-4 and occludin, whereas tacrolimus did not significantly affect claudin-1 and -4 but downregulated occludin to a lesser extent compared to clobetasol. Tacrolimus 115-125 occludin Mus musculus 190-198 23438946-0 2013 Effect of CYP3A5*3 polymorphism on pharmacokinetic drug interaction between tacrolimus and amlodipine. Tacrolimus 76-86 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 23438946-1 2013 The objective of this study was to evaluate the effect of the CYP3A5*3 allele on the pharmacokinetics of tacrolimus and amlodipine, and drug-drug interactions between them in healthy subjects. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 62-68 23438946-5 2013 In the single-dose study, apparent oral clearance (CL/F) of tacrolimus (5 mg) in CYP3A5 expressers was 3.8-fold (p = 0.008) higher than that in CYP3A5 non-expressers. Tacrolimus 60-70 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 81-87 23438946-6 2013 Amlodipine decreased mean tacrolimus CL/F in CYP3A5 expressers by 2.2-fold (p = 0.005), while it had no effect on that in CYP3A5 non-expressers. Tacrolimus 26-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 23438946-8 2013 Tacrolimus increased mean amlodipine CL/F in CYP3A5 expressers by 1.4-fold (p = 0.016) while it had no effect on that in CYP3A5 non-expressers. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 23438946-9 2013 Tacrolimus slightly reduced the AUC0- of amlodipine in both CYP3A5 expressers and non-expressers. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-67 23438946-10 2013 Dose adjustment of tacrolimus should be considered according to CYP3A5*3 genetic polymorphism when tacrolimus is coadministered with amlodipine. Tacrolimus 19-29 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 64-70 23438946-10 2013 Dose adjustment of tacrolimus should be considered according to CYP3A5*3 genetic polymorphism when tacrolimus is coadministered with amlodipine. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 64-70 23174045-7 2013 Tacrolimus also caused morphological alteration of occludin. Tacrolimus 0-10 occludin Mus musculus 51-59 23448773-7 2013 Our experience in this case suggests that treatment with tacrolimus might be useful for treating refractory LD-CTD even when histopathologically chronic fibrotic interstitial pneumonia is evident. Tacrolimus 57-67 CTD Homo sapiens 111-114 23291669-1 2013 OBJECTIVE: The calcineurin inhibitor tacrolimus has been shown to be safe and effective as salvage therapy for steroid-refractory ulcerative colitis (UC). Tacrolimus 37-47 calcineurin binding protein 1 Homo sapiens 15-36 22660440-0 2013 Prediction of the tacrolimus population pharmacokinetic parameters according to CYP3A5 genotype and clinical factors using NONMEM in adult kidney transplant recipients. Tacrolimus 18-28 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 80-86 24229832-0 2013 Addition of mycophenolate mofetil to tacrolimus is associated with decreases in food-specific IgE levels in a pediatric patient with liver transplantation-associated food allergy. Tacrolimus 37-47 immunoglobulin heavy constant epsilon Homo sapiens 94-97 23142694-13 2013 FK506, which interacts with ALK2, increased ET-1 levels and ID1 levels, and this was blocked by ALK2 knockdown. Tacrolimus 0-5 activin A receptor type 1 Homo sapiens 96-100 22972310-0 2013 Effect of tacrolimus on myocardial infarction is associated with inflammation, ROS, MAP kinase and Akt pathways in mini-pigs. Tacrolimus 10-20 AKT serine/threonine kinase 1 Sus scrofa 99-102 22972310-1 2013 AIM: This study tested the hypothesis that tacrolimus therapy limited left ventricular (LV) infarct and remodeling by suppressing the inflammatory response, oxidative stress and regulating the mitogen-activated protein kinase (MAPK) and Akt signaling pathways in an acute myocardial infarction (AMI) mini-pig model by ligating the left anterior descending coronary artery (LAD). Tacrolimus 43-53 AKT serine/threonine kinase 1 Sus scrofa 237-240 23594830-9 2013 Of the three SNPs, the rs4762 of the AGT gene was significantly associated with the development of PTDM in the dominant models (p = 0.03) after adjusting for age and tacrolimus usage. Tacrolimus 166-176 angiotensinogen Homo sapiens 37-40 23142694-13 2013 FK506, which interacts with ALK2, increased ET-1 levels and ID1 levels, and this was blocked by ALK2 knockdown. Tacrolimus 0-5 activin A receptor type 1 Homo sapiens 28-32 23142694-13 2013 FK506, which interacts with ALK2, increased ET-1 levels and ID1 levels, and this was blocked by ALK2 knockdown. Tacrolimus 0-5 endothelin 1 Homo sapiens 44-48 23142694-13 2013 FK506, which interacts with ALK2, increased ET-1 levels and ID1 levels, and this was blocked by ALK2 knockdown. Tacrolimus 0-5 inhibitor of DNA binding 1, HLH protein Homo sapiens 60-63 23573283-0 2013 Tacrolimus inhibits NF-kappaB activation in peripheral human T cells. Tacrolimus 0-10 nuclear factor kappa B subunit 1 Homo sapiens 20-29 23469007-5 2013 The FKBP51-Akt1 interaction was not affected by FK506 analogs or Akt active site inhibitors, but was abolished by the allosteric Akt inhibitor VIII. Tacrolimus 48-53 AKT serine/threonine kinase 1 Homo sapiens 11-15 23252948-5 2013 A new CYP3A4 allele (CYP3A4*22; rs35599367 C>T in intron 6) with a frequency of 5-7% in the Caucasian population was recently discovered through its association with low hepatic CYP3A4 expression and CYP3A4 activity, and showing effects on statin, tacrolimus and cyclosporine metabolism. Tacrolimus 251-261 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 6-12 23252948-5 2013 A new CYP3A4 allele (CYP3A4*22; rs35599367 C>T in intron 6) with a frequency of 5-7% in the Caucasian population was recently discovered through its association with low hepatic CYP3A4 expression and CYP3A4 activity, and showing effects on statin, tacrolimus and cyclosporine metabolism. Tacrolimus 251-261 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-27 23252948-5 2013 A new CYP3A4 allele (CYP3A4*22; rs35599367 C>T in intron 6) with a frequency of 5-7% in the Caucasian population was recently discovered through its association with low hepatic CYP3A4 expression and CYP3A4 activity, and showing effects on statin, tacrolimus and cyclosporine metabolism. Tacrolimus 251-261 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-27 23252948-5 2013 A new CYP3A4 allele (CYP3A4*22; rs35599367 C>T in intron 6) with a frequency of 5-7% in the Caucasian population was recently discovered through its association with low hepatic CYP3A4 expression and CYP3A4 activity, and showing effects on statin, tacrolimus and cyclosporine metabolism. Tacrolimus 251-261 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-27 22875481-6 2013 RESULTS: Chronic microinjection of CsA or FK506 into mPFC increased depressive-like behaviors and decreased mTOR activity, but acute CsA or FK506 had no effects on both behavioral phenotype and mTOR activity. Tacrolimus 42-47 mechanistic target of rapamycin kinase Homo sapiens 108-112 22875481-7 2013 Furthermore, activation of mTOR by NMDA reversed the depressive-like behavior induced by chronic CsA or FK506 administration. Tacrolimus 104-109 mechanistic target of rapamycin kinase Homo sapiens 27-31 23560366-1 2013 Tacrolimus, a calcineurin inhibitor, is a potent immunosuppressive agent used by a majority of transplanters across the globe. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 14-35 22889968-0 2012 Impact of CYP3A5 genetic polymorphism on cross-reactivity in tacrolimus chemiluminescent immunoassay in kidney transplant recipients. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 23328499-6 2013 SN-38 glucuronidation in human and rat liver microsomes was inhibited dose-dependently by the presence of tacrolimus and the 50% inhibition concentration (IC50) values of tacrolimus in rat and human liver microsomes were 10.33 muM and 3.58 muM, respectively. Tacrolimus 171-181 latexin Homo sapiens 227-230 23328499-6 2013 SN-38 glucuronidation in human and rat liver microsomes was inhibited dose-dependently by the presence of tacrolimus and the 50% inhibition concentration (IC50) values of tacrolimus in rat and human liver microsomes were 10.33 muM and 3.58 muM, respectively. Tacrolimus 171-181 latexin Homo sapiens 240-243 23328499-8 2013 These findings suggest that tacrolimus inhibits UGT1A1-mediated SN-38 glucuronidation. Tacrolimus 28-38 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 48-54 22889968-5 2012 The influence of CYP3A5 genotypes on the cross-reactivity in tacrolimus CLIA was evaluated by interaction plots. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 17-23 22889968-7 2012 The dose-normalized C(12) of tacrolimus was significantly higher in the CYP3A5*3/*3 genotype than in the CYP3A5*1/*3 genotype. Tacrolimus 29-39 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 72-78 22889968-7 2012 The dose-normalized C(12) of tacrolimus was significantly higher in the CYP3A5*3/*3 genotype than in the CYP3A5*1/*3 genotype. Tacrolimus 29-39 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 105-111 22889968-8 2012 The C(12) ratio of 13-O-demethylate to tacrolimus was significantly lower in the CYP3A5*3/*3 genotype than in the CYP3A5*1/*3 genotype. Tacrolimus 39-49 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 81-87 22889968-8 2012 The C(12) ratio of 13-O-demethylate to tacrolimus was significantly lower in the CYP3A5*3/*3 genotype than in the CYP3A5*1/*3 genotype. Tacrolimus 39-49 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 114-120 22889968-12 2012 High metabolic capacity associated with the CYP3A5*1 genotype affected the cross-reactivity in patients with low tacrolimus levels. Tacrolimus 113-123 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 44-50 22947444-7 2012 Age and CYP3A5*1 genotype had the largest effect on tacrolimus troughs. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 8-14 24175257-6 2012 Cyclosporine A (CsA) and tacrolimus (Tac) are T-cell-specific calcineurin inhibitors that prevent the activation of helper T cells, thereby inhibiting the transcription of the early activation genes of interleukin (IL)-2 and suppressing T cell-induced activation of tumor necrosis factor-alpha, IL-1beta and IL-6. Tacrolimus 25-35 interleukin 2 Homo sapiens 202-220 24175257-6 2012 Cyclosporine A (CsA) and tacrolimus (Tac) are T-cell-specific calcineurin inhibitors that prevent the activation of helper T cells, thereby inhibiting the transcription of the early activation genes of interleukin (IL)-2 and suppressing T cell-induced activation of tumor necrosis factor-alpha, IL-1beta and IL-6. Tacrolimus 25-35 interleukin 1 beta Homo sapiens 295-303 24175257-6 2012 Cyclosporine A (CsA) and tacrolimus (Tac) are T-cell-specific calcineurin inhibitors that prevent the activation of helper T cells, thereby inhibiting the transcription of the early activation genes of interleukin (IL)-2 and suppressing T cell-induced activation of tumor necrosis factor-alpha, IL-1beta and IL-6. Tacrolimus 25-35 interleukin 6 Homo sapiens 308-312 24175257-6 2012 Cyclosporine A (CsA) and tacrolimus (Tac) are T-cell-specific calcineurin inhibitors that prevent the activation of helper T cells, thereby inhibiting the transcription of the early activation genes of interleukin (IL)-2 and suppressing T cell-induced activation of tumor necrosis factor-alpha, IL-1beta and IL-6. Tacrolimus 37-40 interleukin 2 Homo sapiens 202-220 24175257-6 2012 Cyclosporine A (CsA) and tacrolimus (Tac) are T-cell-specific calcineurin inhibitors that prevent the activation of helper T cells, thereby inhibiting the transcription of the early activation genes of interleukin (IL)-2 and suppressing T cell-induced activation of tumor necrosis factor-alpha, IL-1beta and IL-6. Tacrolimus 37-40 interleukin 1 beta Homo sapiens 295-303 24175257-6 2012 Cyclosporine A (CsA) and tacrolimus (Tac) are T-cell-specific calcineurin inhibitors that prevent the activation of helper T cells, thereby inhibiting the transcription of the early activation genes of interleukin (IL)-2 and suppressing T cell-induced activation of tumor necrosis factor-alpha, IL-1beta and IL-6. Tacrolimus 37-40 interleukin 6 Homo sapiens 308-312 22885734-7 2012 RESULTS: The mRNA expressions of CCL3, CCL4, and CXCL8 were up-regulated by IL-1beta and down-regulated by tacrolimus. Tacrolimus 107-117 C-C motif chemokine ligand 4 Homo sapiens 39-43 23073208-0 2012 Measurement and compartmental modeling of the effect of CYP3A5 gene variation on systemic and intrarenal tacrolimus disposition. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 56-62 23073208-1 2012 We evaluated the hypothesis that cytochrome P450 3A5 (CYP3A5) expression can affect intrarenal tacrolimus accumulation. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-52 23073208-1 2012 We evaluated the hypothesis that cytochrome P450 3A5 (CYP3A5) expression can affect intrarenal tacrolimus accumulation. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 54-60 23073208-2 2012 Tacrolimus was administered orally to 24 healthy volunteers who were selected on the basis of their CYP3A5 genotype. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 100-106 23073208-3 2012 As compared with CYP3A5 nonexpressors, expressors had a 1.6-fold higher oral tacrolimus clearance and 2.0- to 2.7-fold higher metabolite/parent area under the curve (AUC) ratios for 31-desmethyl tacrolimus (31-DMT), 12-hydroxy tacrolimus, and 13-desmethyl tacrolimus (13-DMT). Tacrolimus 77-87 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 17-23 23073208-4 2012 In addition, the apparent urinary tacrolimus clearance was 36% lower in CYP3A5 expressors as compared with nonexpressors. Tacrolimus 34-44 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 72-78 23073208-5 2012 To explore the mechanism behind this observation, we developed a semiphysiological model of renal tacrolimus disposition and predicted that tacrolimus exposure in the renal epithelium of CYP3A5 expressors is 53% of that for CYP3A5 nonexpressors, when normalized to blood AUC. Tacrolimus 98-108 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 187-193 23073208-5 2012 To explore the mechanism behind this observation, we developed a semiphysiological model of renal tacrolimus disposition and predicted that tacrolimus exposure in the renal epithelium of CYP3A5 expressors is 53% of that for CYP3A5 nonexpressors, when normalized to blood AUC. Tacrolimus 140-150 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 187-193 23073208-6 2012 These data suggest that, at steady state, intrarenal accumulation of tacrolimus and its primary metabolites will depend on the CYP3A5 genotype of the liver and kidneys. Tacrolimus 69-79 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 127-133 23082898-1 2012 OBJECTIVES: The once-daily prolonged-release formulation of tacrolimus (tacrolimus QD) is expected to demonstrate equivalent efficacy and safety to the twice-daily formulation (tacrolimus BID). Tacrolimus 60-70 BH3 interacting domain death agonist Homo sapiens 188-191 23082898-10 2012 CONCLUSIONS: Patients taking tacrolimus QD tended to have lower trough levels and require higher dosages than those taking tacrolimus BID during the early posttransplant period, though the differences decreased with increasing time after transplant. Tacrolimus 123-133 BH3 interacting domain death agonist Homo sapiens 134-137 22917533-10 2012 Both p38 responses as well as hypertrophy were abrogated by KB-R7943 as well as the calcineurin inhibitor FK-506 although ERK1/2 phosphorylation was unaffected. Tacrolimus 106-112 mitogen activated protein kinase 14 Rattus norvegicus 5-8 23003106-6 2012 RESULTS: Cyclosporine and tacrolimus decreased insulin sensitivity by 22% (P = 0.02) and 13% (P = 0.048), respectively. Tacrolimus 26-36 insulin Homo sapiens 47-54 22885734-6 2012 The effects of tacrolimus on nuclear translocation of nuclear factor-kappaB (NF-kappaB) were also examined. Tacrolimus 15-25 nuclear factor kappa B subunit 1 Homo sapiens 54-75 22885734-7 2012 RESULTS: The mRNA expressions of CCL3, CCL4, and CXCL8 were up-regulated by IL-1beta and down-regulated by tacrolimus. Tacrolimus 107-117 C-C motif chemokine ligand 3 Homo sapiens 33-37 22885734-7 2012 RESULTS: The mRNA expressions of CCL3, CCL4, and CXCL8 were up-regulated by IL-1beta and down-regulated by tacrolimus. Tacrolimus 107-117 C-X-C motif chemokine ligand 8 Homo sapiens 49-54 22885734-8 2012 The levels of these IL-1beta-induced chemokines in culture supernatant were decreased by a therapeutic concentration of tacrolimus. Tacrolimus 120-130 interleukin 1 beta Homo sapiens 20-28 23032068-4 2012 The effect of FK506 treatment (10 mug per eye for one week) was evaluated by TNF-a, VEGF, iNOS and COX-2 protein levels measurement, neovascularization, and the activation of NF-kB in the retina. Tacrolimus 14-19 tumor necrosis factor Mus musculus 77-82 22885734-9 2012 Tumor necrosis factor-alpha as well as IL-1beta induced these chemokines, while tacrolimus inhibited their production and mRNA expression. Tacrolimus 80-90 tumor necrosis factor Homo sapiens 0-27 23032068-4 2012 The effect of FK506 treatment (10 mug per eye for one week) was evaluated by TNF-a, VEGF, iNOS and COX-2 protein levels measurement, neovascularization, and the activation of NF-kB in the retina. Tacrolimus 14-19 nitric oxide synthase 2, inducible Mus musculus 90-94 22885734-10 2012 Chemotaxis of polymorphonuclear cells in response to IL-1beta was also inhibited by tacrolimus. Tacrolimus 84-94 interleukin 1 beta Homo sapiens 53-61 22885734-11 2012 Nuclear translocation of p50 and p65 NF-kappaB in response to IL-1beta was decreased by tacrolimus. Tacrolimus 88-98 nuclear factor kappa B subunit 1 Homo sapiens 25-28 23032068-4 2012 The effect of FK506 treatment (10 mug per eye for one week) was evaluated by TNF-a, VEGF, iNOS and COX-2 protein levels measurement, neovascularization, and the activation of NF-kB in the retina. Tacrolimus 14-19 cytochrome c oxidase II, mitochondrial Mus musculus 99-104 23032068-6 2012 FK506 treatment significantly lowered retinal TNF-a, VEGF, iNOS and COX-2. Tacrolimus 0-5 tumor necrosis factor Mus musculus 46-51 22885734-11 2012 Nuclear translocation of p50 and p65 NF-kappaB in response to IL-1beta was decreased by tacrolimus. Tacrolimus 88-98 nuclear factor kappa B subunit 1 Homo sapiens 37-46 22885734-11 2012 Nuclear translocation of p50 and p65 NF-kappaB in response to IL-1beta was decreased by tacrolimus. Tacrolimus 88-98 interleukin 1 beta Homo sapiens 62-70 23032068-6 2012 FK506 treatment significantly lowered retinal TNF-a, VEGF, iNOS and COX-2. Tacrolimus 0-5 vascular endothelial growth factor A Mus musculus 53-57 23032068-6 2012 FK506 treatment significantly lowered retinal TNF-a, VEGF, iNOS and COX-2. Tacrolimus 0-5 nitric oxide synthase 2, inducible Mus musculus 59-63 22885734-12 2012 CONCLUSION: IL-1beta-induced chemokine expressions were down-regulated by tacrolimus, suggesting that tacrolimus exerts its anti-inflammatory effect partly through inhibiting chemokine production by RSF. Tacrolimus 74-84 interleukin 1 beta Homo sapiens 12-20 23032068-6 2012 FK506 treatment significantly lowered retinal TNF-a, VEGF, iNOS and COX-2. Tacrolimus 0-5 cytochrome c oxidase II, mitochondrial Mus musculus 68-73 22885734-12 2012 CONCLUSION: IL-1beta-induced chemokine expressions were down-regulated by tacrolimus, suggesting that tacrolimus exerts its anti-inflammatory effect partly through inhibiting chemokine production by RSF. Tacrolimus 102-112 interleukin 1 beta Homo sapiens 12-20 22806180-5 2012 While both drugs efficiently reduced the expression of iNOS and the release of nitric oxide, only FK506 strongly inhibited the expression of Cox-2 and secretion of the mature form of IL-1beta. Tacrolimus 98-103 cytochrome c oxidase II, mitochondrial Rattus norvegicus 141-146 22806180-5 2012 While both drugs efficiently reduced the expression of iNOS and the release of nitric oxide, only FK506 strongly inhibited the expression of Cox-2 and secretion of the mature form of IL-1beta. Tacrolimus 98-103 interleukin 1 beta Rattus norvegicus 183-191 23095803-0 2012 NR1I2 polymorphisms are related to tacrolimus dose-adjusted exposure and BK viremia in adult kidney transplantation. Tacrolimus 35-45 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-5 23149441-10 2012 High IIV of tacrolimus was an independent risk factor of biopsy-proven acute rejection after adjusting for mean tacrolimus concentration, HLA mismatch, induction therapy, donor type, and CYP3A5 polymorphism (hazard ratio 2.655, 95% confidence interval 1.394-5.056). Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 187-193 23149441-11 2012 Interestingly, the impact of tacrolimus IIV on acute rejection was significant in CYP3A5 expressers, whereas it was not in CYP3A5 nonexpressers. Tacrolimus 29-39 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 82-88 23095803-4 2012 RESULTS: In the 158 subjects studied, median (interquartile range) dose-adjusted exposure to tacrolimus was significantly higher in individuals carrying the NR1I2 8055T variant allele, when compared with exposure in wild-type individuals [20 (14, 22) mug h/L/mg versus 15 (9, 24) mug h/L/mg; P =0.0007]. Tacrolimus 93-103 nuclear receptor subfamily 1 group I member 2 Homo sapiens 157-162 23095803-5 2012 Using multivariable logistic regression, NR1I2 8055T carrier status was independently predictive of higher dose-adjusted tacrolimus exposure (P=0.0005). Tacrolimus 121-131 nuclear receptor subfamily 1 group I member 2 Homo sapiens 41-46 23095803-9 2012 CONCLUSIONS: These data demonstrate an impact of pregnane X receptor polymorphisms on tacrolimus pharmacokinetics. Tacrolimus 86-96 nuclear receptor subfamily 1 group I member 2 Homo sapiens 49-68 22895606-0 2012 Tacrolimus (FK506) suppresses TNF-alpha-induced CCL2 (MCP-1) and CXCL10 (IP-10) expression via the inhibition of p38 MAP kinase activation in human colonic myofibroblasts. Tacrolimus 0-10 mitogen-activated protein kinase 14 Homo sapiens 125-128 22576324-2 2012 Cyclosporine and tacrolimus are calcineurin inhibitor immunosuppressants used to prevent organ rejection after liver transplantation; both are substrates of CYP3A4. Tacrolimus 17-27 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 157-163 22895606-0 2012 Tacrolimus (FK506) suppresses TNF-alpha-induced CCL2 (MCP-1) and CXCL10 (IP-10) expression via the inhibition of p38 MAP kinase activation in human colonic myofibroblasts. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 30-39 22895606-0 2012 Tacrolimus (FK506) suppresses TNF-alpha-induced CCL2 (MCP-1) and CXCL10 (IP-10) expression via the inhibition of p38 MAP kinase activation in human colonic myofibroblasts. Tacrolimus 0-10 C-C motif chemokine ligand 2 Homo sapiens 48-52 22895606-0 2012 Tacrolimus (FK506) suppresses TNF-alpha-induced CCL2 (MCP-1) and CXCL10 (IP-10) expression via the inhibition of p38 MAP kinase activation in human colonic myofibroblasts. Tacrolimus 12-17 tumor necrosis factor Homo sapiens 30-39 22895606-0 2012 Tacrolimus (FK506) suppresses TNF-alpha-induced CCL2 (MCP-1) and CXCL10 (IP-10) expression via the inhibition of p38 MAP kinase activation in human colonic myofibroblasts. Tacrolimus 0-10 C-C motif chemokine ligand 2 Homo sapiens 54-59 22895606-0 2012 Tacrolimus (FK506) suppresses TNF-alpha-induced CCL2 (MCP-1) and CXCL10 (IP-10) expression via the inhibition of p38 MAP kinase activation in human colonic myofibroblasts. Tacrolimus 12-17 C-C motif chemokine ligand 2 Homo sapiens 48-52 22895606-0 2012 Tacrolimus (FK506) suppresses TNF-alpha-induced CCL2 (MCP-1) and CXCL10 (IP-10) expression via the inhibition of p38 MAP kinase activation in human colonic myofibroblasts. Tacrolimus 0-10 C-X-C motif chemokine ligand 10 Homo sapiens 65-71 22895606-0 2012 Tacrolimus (FK506) suppresses TNF-alpha-induced CCL2 (MCP-1) and CXCL10 (IP-10) expression via the inhibition of p38 MAP kinase activation in human colonic myofibroblasts. Tacrolimus 0-10 C-X-C motif chemokine ligand 10 Homo sapiens 85-90 22895606-0 2012 Tacrolimus (FK506) suppresses TNF-alpha-induced CCL2 (MCP-1) and CXCL10 (IP-10) expression via the inhibition of p38 MAP kinase activation in human colonic myofibroblasts. Tacrolimus 12-17 C-C motif chemokine ligand 2 Homo sapiens 54-59 22895606-0 2012 Tacrolimus (FK506) suppresses TNF-alpha-induced CCL2 (MCP-1) and CXCL10 (IP-10) expression via the inhibition of p38 MAP kinase activation in human colonic myofibroblasts. Tacrolimus 12-17 C-X-C motif chemokine ligand 10 Homo sapiens 65-71 22895606-0 2012 Tacrolimus (FK506) suppresses TNF-alpha-induced CCL2 (MCP-1) and CXCL10 (IP-10) expression via the inhibition of p38 MAP kinase activation in human colonic myofibroblasts. Tacrolimus 12-17 C-X-C motif chemokine ligand 10 Homo sapiens 85-90 22895606-0 2012 Tacrolimus (FK506) suppresses TNF-alpha-induced CCL2 (MCP-1) and CXCL10 (IP-10) expression via the inhibition of p38 MAP kinase activation in human colonic myofibroblasts. Tacrolimus 12-17 mitogen-activated protein kinase 14 Homo sapiens 125-128 22895606-5 2012 Tacrolimus (1 microM) suppressed tumor necrosis factor (TNF)-alpha-induced CCL2 and CXCL10 mRNA expression, but did not modulate TNF-alpha-induced interleukin (IL)-6 or CXCL8 mRNA expression. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 33-66 22895606-5 2012 Tacrolimus (1 microM) suppressed tumor necrosis factor (TNF)-alpha-induced CCL2 and CXCL10 mRNA expression, but did not modulate TNF-alpha-induced interleukin (IL)-6 or CXCL8 mRNA expression. Tacrolimus 0-10 C-C motif chemokine ligand 2 Homo sapiens 75-79 22895606-5 2012 Tacrolimus (1 microM) suppressed tumor necrosis factor (TNF)-alpha-induced CCL2 and CXCL10 mRNA expression, but did not modulate TNF-alpha-induced interleukin (IL)-6 or CXCL8 mRNA expression. Tacrolimus 0-10 C-X-C motif chemokine ligand 10 Homo sapiens 84-90 22895606-6 2012 Dose-dependent, inhibitory effects of tacrolimus on CCL2 and CXCL10 expression were observed at the mRNA and protein levels. Tacrolimus 38-48 C-C motif chemokine ligand 2 Homo sapiens 52-56 22895606-6 2012 Dose-dependent, inhibitory effects of tacrolimus on CCL2 and CXCL10 expression were observed at the mRNA and protein levels. Tacrolimus 38-48 C-X-C motif chemokine ligand 10 Homo sapiens 61-67 22895606-7 2012 Significant inhibitory effects of tacrolimus were observed at concentrations as low as 0.5 microM for CCL2 and 0.1 microM for CXCL10, respectively. Tacrolimus 34-44 C-C motif chemokine ligand 2 Homo sapiens 102-106 22895606-7 2012 Significant inhibitory effects of tacrolimus were observed at concentrations as low as 0.5 microM for CCL2 and 0.1 microM for CXCL10, respectively. Tacrolimus 34-44 C-X-C motif chemokine ligand 10 Homo sapiens 126-132 22895606-8 2012 TNF-alpha-induced CCL2 and CXCL10 expression depended on p38 MAP kinase activation, and tacrolimus strongly inhibited the TNF-alpha-induced phosphorylation of p38 MAP kinase. Tacrolimus 88-98 tumor necrosis factor Homo sapiens 122-131 22895606-8 2012 TNF-alpha-induced CCL2 and CXCL10 expression depended on p38 MAP kinase activation, and tacrolimus strongly inhibited the TNF-alpha-induced phosphorylation of p38 MAP kinase. Tacrolimus 88-98 mitogen-activated protein kinase 14 Homo sapiens 159-162 22895606-10 2012 In conclusion, tacrolimus suppressed CCL2 and CXCL10 expression in human colonic myofibroblasts. Tacrolimus 15-25 C-C motif chemokine ligand 2 Homo sapiens 37-41 22895606-10 2012 In conclusion, tacrolimus suppressed CCL2 and CXCL10 expression in human colonic myofibroblasts. Tacrolimus 15-25 C-X-C motif chemokine ligand 10 Homo sapiens 46-52 22672741-3 2012 The aim of our study was to examine expression patterns of TLR4 and TLR9 in normal oral mucosa and OLP and describe the effect of topical tacrolimus treatment on the expression of TLR4 and TLR9 in OLP. Tacrolimus 138-148 toll like receptor 4 Homo sapiens 180-184 22865594-0 2012 FK506 alleviates proteinuria in rats with adriamycin-induced nephropathy by down-regulating TRPC6 and CaN expression. Tacrolimus 0-5 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 92-97 22865594-8 2012 FK506 can inhibit CaN activity in the renal tissues and further decrease TRPC6 expression and therefore reduce renal damage and proteinuria in a dose-dependent manner. Tacrolimus 0-5 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 73-78 22865594-10 2012 FK506 had a therapeutic effect on the progression of proteinuria and renal damage by down-regulating of TRPC6 and CaN in the renal tissues. Tacrolimus 0-5 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 104-109 22672741-3 2012 The aim of our study was to examine expression patterns of TLR4 and TLR9 in normal oral mucosa and OLP and describe the effect of topical tacrolimus treatment on the expression of TLR4 and TLR9 in OLP. Tacrolimus 138-148 toll like receptor 9 Homo sapiens 189-193 22672741-5 2012 We evaluated also the effect of topical tacrolimus on TLR4 and TLR9 expression in a patient with OLP. Tacrolimus 40-50 toll like receptor 4 Homo sapiens 54-58 22672741-5 2012 We evaluated also the effect of topical tacrolimus on TLR4 and TLR9 expression in a patient with OLP. Tacrolimus 40-50 toll like receptor 9 Homo sapiens 63-67 23022192-10 2012 An inhibitor (FK506) against calcineurin, which is a serine/threonine specific phosphatase enhanced the resistance of ATBF1 against the digestion by calpain-1. Tacrolimus 14-19 zinc finger homeobox 3 Homo sapiens 118-123 23140765-0 2012 Differential regulation of nuclear factor-kappa B subunits on epidermal keratinocytes by ultraviolet B and tacrolimus. Tacrolimus 107-117 nuclear factor kappa B subunit 1 Homo sapiens 27-49 23140765-2 2012 Recently, we have shown that direct regulation of NF-kappaB/p65 subunit may account for tacrolimus ointment"s remarkable clinical efficacy on treating inflammatory dermatoses. Tacrolimus 88-98 nuclear factor kappa B subunit 1 Homo sapiens 50-59 23140765-2 2012 Recently, we have shown that direct regulation of NF-kappaB/p65 subunit may account for tacrolimus ointment"s remarkable clinical efficacy on treating inflammatory dermatoses. Tacrolimus 88-98 RELA proto-oncogene, NF-kB subunit Homo sapiens 60-63 23140765-10 2012 Furthermore, treatment with tacrolimus didn"t affect the nuclear activation and translocation of NF-kappaB/p50, while the UVB induced NF-kappaB/p65 nuclear expression was suppressed by tacrolimus. Tacrolimus 185-195 nuclear factor kappa B subunit 1 Homo sapiens 134-143 23140765-10 2012 Furthermore, treatment with tacrolimus didn"t affect the nuclear activation and translocation of NF-kappaB/p50, while the UVB induced NF-kappaB/p65 nuclear expression was suppressed by tacrolimus. Tacrolimus 185-195 RELA proto-oncogene, NF-kB subunit Homo sapiens 144-147 23140765-12 2012 The clinical efficacy of tacrolimus may be attributed to its specific regulatory effect on NF-kappaB/p65 but not NF-kappaB/p50 of the NF-kappaB pathway. Tacrolimus 25-35 nuclear factor kappa B subunit 1 Homo sapiens 91-100 23140765-12 2012 The clinical efficacy of tacrolimus may be attributed to its specific regulatory effect on NF-kappaB/p65 but not NF-kappaB/p50 of the NF-kappaB pathway. Tacrolimus 25-35 RELA proto-oncogene, NF-kB subunit Homo sapiens 101-104 23146479-0 2012 Effect of CYP3A5, CYP3A4, and ABCB1 genotypes as determinants of tacrolimus dose and clinical outcomes after heart transplantation. Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 23146479-0 2012 Effect of CYP3A5, CYP3A4, and ABCB1 genotypes as determinants of tacrolimus dose and clinical outcomes after heart transplantation. Tacrolimus 65-75 ATP binding cassette subfamily B member 1 Homo sapiens 30-35 22992768-0 2012 Combinational effect of intestinal and hepatic CYP3A5 genotypes on tacrolimus pharmacokinetics in recipients of living donor liver transplantation. Tacrolimus 67-77 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 47-53 22992768-1 2012 BACKGROUND: For living donor liver transplantation, the genetic association of CYP3A5 genotype of recipient"s native intestine and donor"s liver allograft with tacrolimus pharmacokinetics has not been explained completely considering liver regeneration time. Tacrolimus 160-170 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 79-85 22992768-2 2012 The goal of the study was to investigate the longitudinal effects of recipient-donor combinational CYP3A5 genotypes on tacrolimus dose-normalized concentration (C/D ratio) in blood. Tacrolimus 119-129 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 99-105 22992768-7 2012 CONCLUSIONS: CYP3A5 genotypes of both recipient and donor were important factors influencing pharmacokinetic variability of tacrolimus. Tacrolimus 124-134 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 23095847-7 2012 The calcineurin inhibitor, FK506, significantly attenuated the injury-induced NF200 dephosphorylation of NF200 at 3 dpi and axonal degeneration at 3 and 7 dpi but did not affect the decrease in NF200 protein levels or impaired axonal transport. Tacrolimus 27-32 calcineurin binding protein 1 Rattus norvegicus 4-25 23677660-1 2012 Calcineurin inhibitor encephalopathy (CIE) is a rare condition occurring in patients who are undergoing treatment with drugs from the calcineurin inhibitor (CI) family of immunosuppressants, either cyclosporine (CsA) or tacrolimus (TAC, FK506). Tacrolimus 220-230 calcineurin binding protein 1 Homo sapiens 0-21 23677660-1 2012 Calcineurin inhibitor encephalopathy (CIE) is a rare condition occurring in patients who are undergoing treatment with drugs from the calcineurin inhibitor (CI) family of immunosuppressants, either cyclosporine (CsA) or tacrolimus (TAC, FK506). Tacrolimus 220-230 calcineurin binding protein 1 Homo sapiens 134-155 23677660-1 2012 Calcineurin inhibitor encephalopathy (CIE) is a rare condition occurring in patients who are undergoing treatment with drugs from the calcineurin inhibitor (CI) family of immunosuppressants, either cyclosporine (CsA) or tacrolimus (TAC, FK506). Tacrolimus 220-230 calcineurin binding protein 1 Homo sapiens 38-40 23677660-1 2012 Calcineurin inhibitor encephalopathy (CIE) is a rare condition occurring in patients who are undergoing treatment with drugs from the calcineurin inhibitor (CI) family of immunosuppressants, either cyclosporine (CsA) or tacrolimus (TAC, FK506). Tacrolimus 232-235 calcineurin binding protein 1 Homo sapiens 0-21 23677660-1 2012 Calcineurin inhibitor encephalopathy (CIE) is a rare condition occurring in patients who are undergoing treatment with drugs from the calcineurin inhibitor (CI) family of immunosuppressants, either cyclosporine (CsA) or tacrolimus (TAC, FK506). Tacrolimus 232-235 calcineurin binding protein 1 Homo sapiens 38-40 23677660-1 2012 Calcineurin inhibitor encephalopathy (CIE) is a rare condition occurring in patients who are undergoing treatment with drugs from the calcineurin inhibitor (CI) family of immunosuppressants, either cyclosporine (CsA) or tacrolimus (TAC, FK506). Tacrolimus 237-242 calcineurin binding protein 1 Homo sapiens 0-21 23677660-1 2012 Calcineurin inhibitor encephalopathy (CIE) is a rare condition occurring in patients who are undergoing treatment with drugs from the calcineurin inhibitor (CI) family of immunosuppressants, either cyclosporine (CsA) or tacrolimus (TAC, FK506). Tacrolimus 237-242 calcineurin binding protein 1 Homo sapiens 38-40 23146466-3 2012 Our objective was to investigate whether CYP3A5 1/3 polymorphism was associated with ambulatory BP among a population of renal transplant recipients receiving the calcineurin inhibitor tacrolimus for immunosuppression. Tacrolimus 185-195 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 41-47 22835622-8 2012 Both GA and FK-506 significantly increased the rate of axonal regeneration following crush injury in Thy1-GFP rats. Tacrolimus 12-18 Thy-1 cell surface antigen Rattus norvegicus 101-105 23022192-10 2012 An inhibitor (FK506) against calcineurin, which is a serine/threonine specific phosphatase enhanced the resistance of ATBF1 against the digestion by calpain-1. Tacrolimus 14-19 calpain 1 Homo sapiens 149-158 23000414-5 2012 Treatment with calcineurin-NFAT inhibitors, Tacrolimus (FK506) and the 11R-VIVIT (VIVIT) peptide, reduces NFATc1 expression consistent with a reduction in osteoclast differentiation and activity. Tacrolimus 44-54 nuclear factor of activated T cells 1 Homo sapiens 106-112 23000414-5 2012 Treatment with calcineurin-NFAT inhibitors, Tacrolimus (FK506) and the 11R-VIVIT (VIVIT) peptide, reduces NFATc1 expression consistent with a reduction in osteoclast differentiation and activity. Tacrolimus 56-61 nuclear factor of activated T cells 1 Homo sapiens 106-112 23000414-9 2012 Quantitative real-time polymerase chain reaction (qRT-PCR) analysis demonstrated that FK506 treatment significantly (p<0.05) reduced the expression of NFATc1, CathK, OSCAR, FcRgamma, TREM2 and DAP12 during the terminal stage of osteoclast formation. Tacrolimus 86-91 nuclear factor of activated T cells 1 Homo sapiens 154-160 23000414-9 2012 Quantitative real-time polymerase chain reaction (qRT-PCR) analysis demonstrated that FK506 treatment significantly (p<0.05) reduced the expression of NFATc1, CathK, OSCAR, FcRgamma, TREM2 and DAP12 during the terminal stage of osteoclast formation. Tacrolimus 86-91 cathepsin K Homo sapiens 162-167 23000414-9 2012 Quantitative real-time polymerase chain reaction (qRT-PCR) analysis demonstrated that FK506 treatment significantly (p<0.05) reduced the expression of NFATc1, CathK, OSCAR, FcRgamma, TREM2 and DAP12 during the terminal stage of osteoclast formation. Tacrolimus 86-91 osteoclast associated Ig-like receptor Homo sapiens 169-174 23000414-9 2012 Quantitative real-time polymerase chain reaction (qRT-PCR) analysis demonstrated that FK506 treatment significantly (p<0.05) reduced the expression of NFATc1, CathK, OSCAR, FcRgamma, TREM2 and DAP12 during the terminal stage of osteoclast formation. Tacrolimus 86-91 Fc epsilon receptor Ig Homo sapiens 176-184 23000414-9 2012 Quantitative real-time polymerase chain reaction (qRT-PCR) analysis demonstrated that FK506 treatment significantly (p<0.05) reduced the expression of NFATc1, CathK, OSCAR, FcRgamma, TREM2 and DAP12 during the terminal stage of osteoclast formation. Tacrolimus 86-91 triggering receptor expressed on myeloid cells 2 Homo sapiens 186-191 23000414-9 2012 Quantitative real-time polymerase chain reaction (qRT-PCR) analysis demonstrated that FK506 treatment significantly (p<0.05) reduced the expression of NFATc1, CathK, OSCAR, FcRgamma, TREM2 and DAP12 during the terminal stage of osteoclast formation. Tacrolimus 86-91 transmembrane immune signaling adaptor TYROBP Homo sapiens 196-201 22469198-1 2012 The purpose of this study was to identify and characterize the association of various clinical variables and CYP3A5 genotypes with the pharmacokinetics of tacrolimus and outcome over 1-5 years in kidney transplantation patients in Korea. Tacrolimus 155-165 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 109-115 22469198-6 2012 After adjusting for sex, body-weight and doses of corticosteroids and mycophenolate mofetil, we noted that CYP3A5 genotypes had the most profound effect on the dose and dose-adjusted trough levels of tacrolimus 1-5 years after transplantation (p < 0.001). Tacrolimus 200-210 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 107-113 22469198-8 2012 Therefore, the CYP3A5 genotype is a variable marker for tacrolimus dose requirement, and the trough level of tacrolimus should be controlled to minimize post-transplant hyperlipidaemia to achieve optimum outcome. Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 22495293-2 2012 In addition to increasing transforming growth factor (TGF)-beta levels, tacrolimus inhibits calcineurin by binding to FK506-binding protein 12 (FKBP12). Tacrolimus 72-82 FK506 binding protein 1a Mus musculus 118-142 23580932-4 2012 Tacrolimus has a large inter-/intra-patient variability in pharmacokinetics profile and a poor oral bioavailability because of its poor solubility, P-gp efflux, marked pre-systemic metabolism by CYP3A in the enterocytes and liver first pass effect. Tacrolimus 0-10 phosphoglycolate phosphatase Homo sapiens 148-152 23580932-4 2012 Tacrolimus has a large inter-/intra-patient variability in pharmacokinetics profile and a poor oral bioavailability because of its poor solubility, P-gp efflux, marked pre-systemic metabolism by CYP3A in the enterocytes and liver first pass effect. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 195-200 22221776-2 2012 To our knowledge, there have been no prospective trials examining the use of oral tacrolimus, a calcineurin inhibitor, in AD. Tacrolimus 82-92 calcineurin binding protein 1 Homo sapiens 96-117 22495293-2 2012 In addition to increasing transforming growth factor (TGF)-beta levels, tacrolimus inhibits calcineurin by binding to FK506-binding protein 12 (FKBP12). Tacrolimus 72-82 FK506 binding protein 1a Mus musculus 144-150 22495293-4 2012 Here we tested whether tacrolimus binding to FKBP12 removes an inhibition of the TGF-beta receptor, allowing ligand binding, ultimately leading to receptor activation and arteriolar hyalinosis. Tacrolimus 23-33 FK506 binding protein 1a Mus musculus 45-51 22495293-4 2012 Here we tested whether tacrolimus binding to FKBP12 removes an inhibition of the TGF-beta receptor, allowing ligand binding, ultimately leading to receptor activation and arteriolar hyalinosis. Tacrolimus 23-33 transforming growth factor, beta 1 Mus musculus 81-89 22495293-6 2012 Tacrolimus-treated and knockout mice exhibited significantly increased levels of aortic TGF-beta receptor activation as evidenced by SMAD2/3 phosphorylation, along with increased collagen and fibronectin expression compared to controls. Tacrolimus 0-10 transforming growth factor, beta 1 Mus musculus 88-96 22495293-6 2012 Tacrolimus-treated and knockout mice exhibited significantly increased levels of aortic TGF-beta receptor activation as evidenced by SMAD2/3 phosphorylation, along with increased collagen and fibronectin expression compared to controls. Tacrolimus 0-10 SMAD family member 2 Mus musculus 133-140 22495293-6 2012 Tacrolimus-treated and knockout mice exhibited significantly increased levels of aortic TGF-beta receptor activation as evidenced by SMAD2/3 phosphorylation, along with increased collagen and fibronectin expression compared to controls. Tacrolimus 0-10 fibronectin 1 Mus musculus 192-203 22495293-7 2012 Treatment of isolated mouse aortas with tacrolimus increased TGF-beta receptor activation and collagen and fibronectin expression. Tacrolimus 40-50 transforming growth factor, beta 1 Mus musculus 61-69 22495293-7 2012 Treatment of isolated mouse aortas with tacrolimus increased TGF-beta receptor activation and collagen and fibronectin expression. Tacrolimus 40-50 fibronectin 1 Mus musculus 107-118 22897149-0 2012 Insulin independence after conversion from tacrolimus to cyclosporine in islet transplantation. Tacrolimus 43-53 insulin Homo sapiens 0-7 22863028-5 2012 Steroid-free tacrolimus monotherapy with rATG or IL-2RA induction is effective if adequate tacrolimus exposure is maintained. Tacrolimus 13-23 interleukin 2 receptor subunit alpha Homo sapiens 49-55 22863028-5 2012 Steroid-free tacrolimus monotherapy with rATG or IL-2RA induction is effective if adequate tacrolimus exposure is maintained. Tacrolimus 91-101 interleukin 2 receptor subunit alpha Homo sapiens 49-55 22960764-3 2012 METHODS: After whole-blood activation with the gamma(c) cytokines IL-2, IL-7, and IL-15, STAT5 phosphorylation was determined in T cells of de novo kidney transplantation patients treated with tofacitinib/basiliximab (n=5), calcineurin inhibitor (CNI) (cyclosporine A)/basiliximab (n=4) or CNI (tacrolimus)-based immunosuppression (n=6). Tacrolimus 295-305 signal transducer and activator of transcription 5A Homo sapiens 89-94 22922441-5 2012 In contrast, administration of FK506 significantly suppressed interferon (IFN)-gamma but increased IL-10 expression as compared with that of SAHA-treated animals, and this effect was independent of SAHA. Tacrolimus 31-36 interferon gamma Mus musculus 62-84 22037511-3 2012 A key factor in the inter-individual variability for tacrolimus, and probably sirolimus, is whether an individual is genetically predicted to express the drug metabolising enzyme cytochrome P450 3A5 (CYP3A5). Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 179-198 22037511-3 2012 A key factor in the inter-individual variability for tacrolimus, and probably sirolimus, is whether an individual is genetically predicted to express the drug metabolising enzyme cytochrome P450 3A5 (CYP3A5). Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 200-206 22037511-4 2012 Individuals predicted to express CYP3A5 by possession of at least one wild-type CYP3A5*1 allele require 1.5-2 times higher doses of tacrolimus to achieve target blood concentrations than individuals homozygous for the CYP3A5*3 allele who are functional non-expressers of CYP3A5. Tacrolimus 132-142 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 22037511-4 2012 Individuals predicted to express CYP3A5 by possession of at least one wild-type CYP3A5*1 allele require 1.5-2 times higher doses of tacrolimus to achieve target blood concentrations than individuals homozygous for the CYP3A5*3 allele who are functional non-expressers of CYP3A5. Tacrolimus 132-142 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 80-86 22037511-4 2012 Individuals predicted to express CYP3A5 by possession of at least one wild-type CYP3A5*1 allele require 1.5-2 times higher doses of tacrolimus to achieve target blood concentrations than individuals homozygous for the CYP3A5*3 allele who are functional non-expressers of CYP3A5. Tacrolimus 132-142 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 80-86 22037511-4 2012 Individuals predicted to express CYP3A5 by possession of at least one wild-type CYP3A5*1 allele require 1.5-2 times higher doses of tacrolimus to achieve target blood concentrations than individuals homozygous for the CYP3A5*3 allele who are functional non-expressers of CYP3A5. Tacrolimus 132-142 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 80-86 22037511-5 2012 Planning the initial tacrolimus dose based on the CYP3A5 genotype has been shown to allow more rapid achievement of target blood concentrations after transplantation than a standard dose given to all patients. Tacrolimus 21-31 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 50-56 22327003-4 2012 Among the currently available immunosuppressive agents, cyclosporine, tacrolimus and mycophenolic acid are in vitro substrates of the UGT1A and 2B families of glucuronidation enzymes. Tacrolimus 70-80 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 134-146 22540338-0 2012 FK506 inhibits the enhancing effects of transforming growth factor (TGF)-beta1 on collagen expression and TGF-beta/Smad signalling in keloid fibroblasts: implication for new therapeutic approach. Tacrolimus 0-5 transforming growth factor beta 1 Homo sapiens 40-78 22540338-0 2012 FK506 inhibits the enhancing effects of transforming growth factor (TGF)-beta1 on collagen expression and TGF-beta/Smad signalling in keloid fibroblasts: implication for new therapeutic approach. Tacrolimus 0-5 transforming growth factor beta 1 Homo sapiens 106-114 22540338-0 2012 FK506 inhibits the enhancing effects of transforming growth factor (TGF)-beta1 on collagen expression and TGF-beta/Smad signalling in keloid fibroblasts: implication for new therapeutic approach. Tacrolimus 0-5 SMAD family member 4 Homo sapiens 115-119 22540338-4 2012 As tacrolimus (FK506) has been reported to inhibit the effects of TGF-beta1 on cultured fibroblasts, we hypothesized that FK506 may be useful in treating keloids. Tacrolimus 3-13 transforming growth factor beta 1 Homo sapiens 66-75 22540338-4 2012 As tacrolimus (FK506) has been reported to inhibit the effects of TGF-beta1 on cultured fibroblasts, we hypothesized that FK506 may be useful in treating keloids. Tacrolimus 15-20 transforming growth factor beta 1 Homo sapiens 66-75 22540338-4 2012 As tacrolimus (FK506) has been reported to inhibit the effects of TGF-beta1 on cultured fibroblasts, we hypothesized that FK506 may be useful in treating keloids. Tacrolimus 122-127 transforming growth factor beta 1 Homo sapiens 66-75 22540338-5 2012 OBJECTIVES: To explore the effects of FK506 on TGF-beta1-stimulated keloid fibroblasts (KFs) in terms of proliferation, migration and collagen production and to investigate the regulatory pathways involved. Tacrolimus 38-43 transforming growth factor beta 1 Homo sapiens 47-56 22540338-9 2012 FK506 markedly inhibited KF proliferation, migration and collagen production enhanced by TGF-beta1. Tacrolimus 0-5 transforming growth factor beta 1 Homo sapiens 89-98 22540338-10 2012 The increase in TGF-beta receptor I and II expression in TGF-beta1-treated KFs was suppressed by FK506 treatment. Tacrolimus 97-102 transforming growth factor beta 1 Homo sapiens 16-24 22540338-10 2012 The increase in TGF-beta receptor I and II expression in TGF-beta1-treated KFs was suppressed by FK506 treatment. Tacrolimus 97-102 transforming growth factor beta 1 Homo sapiens 57-66 22540338-11 2012 TGF-beta1 increased the phosphorylation of Smad2/3 and Smad4 in KFs, and this enhancing effect was abrogated by FK506. Tacrolimus 112-117 transforming growth factor beta 1 Homo sapiens 0-9 22540338-11 2012 TGF-beta1 increased the phosphorylation of Smad2/3 and Smad4 in KFs, and this enhancing effect was abrogated by FK506. Tacrolimus 112-117 SMAD family member 2 Homo sapiens 43-50 22540338-11 2012 TGF-beta1 increased the phosphorylation of Smad2/3 and Smad4 in KFs, and this enhancing effect was abrogated by FK506. Tacrolimus 112-117 SMAD family member 4 Homo sapiens 55-60 22540338-12 2012 In addition, FK506 significantly increased the expression of Smad7 which was suppressed by TGF-beta1 treatment. Tacrolimus 13-18 SMAD family member 7 Homo sapiens 61-66 22540338-12 2012 In addition, FK506 significantly increased the expression of Smad7 which was suppressed by TGF-beta1 treatment. Tacrolimus 13-18 transforming growth factor beta 1 Homo sapiens 91-100 22540338-13 2012 CONCLUSIONS: Our results demonstrate that FK506 effectively blocks the TGF-beta/Smad signalling pathway in KFs by downregulation of TGF-beta receptors and suggest that FK506 may be included in the armamentarium for treating keloids. Tacrolimus 42-47 transforming growth factor beta 1 Homo sapiens 71-79 22540338-13 2012 CONCLUSIONS: Our results demonstrate that FK506 effectively blocks the TGF-beta/Smad signalling pathway in KFs by downregulation of TGF-beta receptors and suggest that FK506 may be included in the armamentarium for treating keloids. Tacrolimus 42-47 SMAD family member 4 Homo sapiens 80-84 22540338-13 2012 CONCLUSIONS: Our results demonstrate that FK506 effectively blocks the TGF-beta/Smad signalling pathway in KFs by downregulation of TGF-beta receptors and suggest that FK506 may be included in the armamentarium for treating keloids. Tacrolimus 42-47 transforming growth factor beta 1 Homo sapiens 132-140 22540338-13 2012 CONCLUSIONS: Our results demonstrate that FK506 effectively blocks the TGF-beta/Smad signalling pathway in KFs by downregulation of TGF-beta receptors and suggest that FK506 may be included in the armamentarium for treating keloids. Tacrolimus 168-173 transforming growth factor beta 1 Homo sapiens 71-79 22540338-13 2012 CONCLUSIONS: Our results demonstrate that FK506 effectively blocks the TGF-beta/Smad signalling pathway in KFs by downregulation of TGF-beta receptors and suggest that FK506 may be included in the armamentarium for treating keloids. Tacrolimus 168-173 SMAD family member 4 Homo sapiens 80-84 22540338-13 2012 CONCLUSIONS: Our results demonstrate that FK506 effectively blocks the TGF-beta/Smad signalling pathway in KFs by downregulation of TGF-beta receptors and suggest that FK506 may be included in the armamentarium for treating keloids. Tacrolimus 168-173 transforming growth factor beta 1 Homo sapiens 132-140 22922441-5 2012 In contrast, administration of FK506 significantly suppressed interferon (IFN)-gamma but increased IL-10 expression as compared with that of SAHA-treated animals, and this effect was independent of SAHA. Tacrolimus 31-36 interleukin 10 Mus musculus 99-104 22922441-6 2012 Interestingly, SAHA synergizes with FK506 to promote Foxp3 and CTLA4 expression. Tacrolimus 36-41 forkhead box P3 Mus musculus 53-58 22922441-6 2012 Interestingly, SAHA synergizes with FK506 to promote Foxp3 and CTLA4 expression. Tacrolimus 36-41 cytotoxic T-lymphocyte-associated protein 4 Mus musculus 63-68 22386321-2 2012 Besides immunosuppression, tacrolimus has been reported to have the potential to increase muscle strength by enhancing ryanodine receptor (RyR) function. Tacrolimus 27-37 ryanodine receptor 1 Homo sapiens 119-137 22386321-2 2012 Besides immunosuppression, tacrolimus has been reported to have the potential to increase muscle strength by enhancing ryanodine receptor (RyR) function. Tacrolimus 27-37 ryanodine receptor 1 Homo sapiens 139-142 22260488-6 2012 TCF2 analysis might, therefore, be of interest in patients with congenital abnormalities of the kidney and the urinary tract in order to improve posttransplant management in terms of steroid and tacrolimus exposure. Tacrolimus 195-205 HNF1 homeobox B Homo sapiens 0-4 22386321-3 2012 However, few attempts have been made to demonstrate the early effect of tacrolimus as an RyR enhancer in clinical investigation. Tacrolimus 72-82 ryanodine receptor 1 Homo sapiens 89-92 22386321-14 2012 SIGNIFICANCE: This early effect of tacrolimus may imply a pharmacological enhancement of RyR function to improve E-C coupling in MG. Tacrolimus 35-45 ryanodine receptor 1 Homo sapiens 89-92 22428934-1 2012 BACKGROUND: The aim of this study was to explore effects of erythropoietin and pentoxifylline in tacrolimus-induced pancreatic beta cell and renal injury in rats. Tacrolimus 97-107 erythropoietin Rattus norvegicus 60-74 22428934-5 2012 In rats subjected to tacrolimus and pretreated with Epo, there was significant decrease in apoptotic nuclei staining than those in tacrolimus group. Tacrolimus 21-31 erythropoietin Rattus norvegicus 52-55 22428934-5 2012 In rats subjected to tacrolimus and pretreated with Epo, there was significant decrease in apoptotic nuclei staining than those in tacrolimus group. Tacrolimus 131-141 erythropoietin Rattus norvegicus 52-55 22428934-6 2012 Blood trough levels of tacrolimus were significantly higher in erythropoietin-pretreated group, although same amount of tacrolimus was injected with other groups. Tacrolimus 23-33 erythropoietin Rattus norvegicus 63-77 22428934-7 2012 CONCLUSION: Results of our study demonstrated significant antiapoptotic effects of erythropoietin on renal tubules, increasing effect of erythropoietin on tacrolimus blood levels, and insignificant antioxidant effects of both erythropoietin and pentoxifylline on renal and pancreas tissues. Tacrolimus 155-165 erythropoietin Rattus norvegicus 137-151 22428934-7 2012 CONCLUSION: Results of our study demonstrated significant antiapoptotic effects of erythropoietin on renal tubules, increasing effect of erythropoietin on tacrolimus blood levels, and insignificant antioxidant effects of both erythropoietin and pentoxifylline on renal and pancreas tissues. Tacrolimus 155-165 erythropoietin Rattus norvegicus 137-151 22871995-0 2012 In vivo CYP3A4 activity, CYP3A5 genotype, and hematocrit predict tacrolimus dose requirements and clearance in renal transplant patients. Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 8-14 22871995-0 2012 In vivo CYP3A4 activity, CYP3A5 genotype, and hematocrit predict tacrolimus dose requirements and clearance in renal transplant patients. Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 25-31 22871995-1 2012 Tacrolimus is metabolized by CYP3A4 and CYP3A5 and is characterized by a narrow therapeutic index and highly variable pharmacokinetics. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-35 22871995-1 2012 Tacrolimus is metabolized by CYP3A4 and CYP3A5 and is characterized by a narrow therapeutic index and highly variable pharmacokinetics. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 40-46 22871995-3 2012 In vivo CYP3A4 activity and CYP3A5 genotype explain 56-59% of variability in tacrolimus dose requirements and clearance, contributing ~25 and 30%, respectively. Tacrolimus 77-87 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 8-14 22871995-3 2012 In vivo CYP3A4 activity and CYP3A5 genotype explain 56-59% of variability in tacrolimus dose requirements and clearance, contributing ~25 and 30%, respectively. Tacrolimus 77-87 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 28-34 22871995-5 2012 These data indicate that CYP3A4- and CYP3A5-mediated tacrolimus metabolisms are major determinants of tacrolimus disposition in vivo and explain a substantial part of the clinically observed high interindividual variability in tacrolimus pharmacokinetics. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-31 22871995-5 2012 These data indicate that CYP3A4- and CYP3A5-mediated tacrolimus metabolisms are major determinants of tacrolimus disposition in vivo and explain a substantial part of the clinically observed high interindividual variability in tacrolimus pharmacokinetics. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 22871995-5 2012 These data indicate that CYP3A4- and CYP3A5-mediated tacrolimus metabolisms are major determinants of tacrolimus disposition in vivo and explain a substantial part of the clinically observed high interindividual variability in tacrolimus pharmacokinetics. Tacrolimus 102-112 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-31 22871995-5 2012 These data indicate that CYP3A4- and CYP3A5-mediated tacrolimus metabolisms are major determinants of tacrolimus disposition in vivo and explain a substantial part of the clinically observed high interindividual variability in tacrolimus pharmacokinetics. Tacrolimus 102-112 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 22871995-5 2012 These data indicate that CYP3A4- and CYP3A5-mediated tacrolimus metabolisms are major determinants of tacrolimus disposition in vivo and explain a substantial part of the clinically observed high interindividual variability in tacrolimus pharmacokinetics. Tacrolimus 102-112 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-31 22871995-5 2012 These data indicate that CYP3A4- and CYP3A5-mediated tacrolimus metabolisms are major determinants of tacrolimus disposition in vivo and explain a substantial part of the clinically observed high interindividual variability in tacrolimus pharmacokinetics. Tacrolimus 102-112 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 21883747-1 2012 The shared metabolism of PPIs and tacrolimus through the CYP enzyme system has been associated with clinically significant drug interactions, especially in patients who are classified as CYP 2C19 PMs. Tacrolimus 34-44 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 187-195 21883747-3 2012 A drug interaction between tacrolimus and omeprazole, esomeprazole, but not lansoprazole, occurred in an 18-yr-old female kidney transplant recipient classified as a CYP 2C19 extensive (normal) metabolizer. Tacrolimus 27-37 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 166-174 22023701-5 2012 The temporal resolution of the patient"s symptoms suggests that prompt recognition of central pontine and EPM and conversion from tacrolimus to rapamycin during the early post-operative course may have therapeutic benefits for patients undergoing pediatric transplant with CNI-related neurotoxicity. Tacrolimus 130-140 5'-nucleotidase, cytosolic IA Homo sapiens 273-276 22670785-6 2012 Expression of the peripheral homing receptors CXCR3 (r = 0.44, P < 0.05) and CCR5 (r = 0.45, P < 0.05) on FOXP3+ Tregs correlated with renal allograft function (eGFR) in patients receiving tacrolimus (n = 28), but not cyclosporine A (CsA) (n = 26). Tacrolimus 195-205 C-X-C motif chemokine receptor 3 Homo sapiens 46-51 23016456-0 2012 Tacrolimus strongly inhibits multiple human UDP-glucuronosyltransferase (UGT) isoforms. Tacrolimus 0-10 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 44-71 23016456-0 2012 Tacrolimus strongly inhibits multiple human UDP-glucuronosyltransferase (UGT) isoforms. Tacrolimus 0-10 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 73-76 23016456-1 2012 The objective of the present study is to clearly evaluate the inhibitory effects of tacrolimus (tacro) on important UGT isoforms in human liver, including determination of inhibition kinetic type and calculation of inhibition kinetic parameters. Tacrolimus 84-94 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 116-119 23016456-1 2012 The objective of the present study is to clearly evaluate the inhibitory effects of tacrolimus (tacro) on important UGT isoforms in human liver, including determination of inhibition kinetic type and calculation of inhibition kinetic parameters. Tacrolimus 84-89 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 116-119 23016456-2 2012 An in vitro incubation system was used to investigate the inhibitory effect of tacro on UGT isoforms. Tacrolimus 79-84 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 88-91 23016456-4 2012 Among the tested UGT isoforms, UGT1A1, UGT1A3, UGT2B7 and UGT2B15 were strongly inhibited by tacro in a concentration-dependent manner. Tacrolimus 93-98 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 17-20 23016456-4 2012 Among the tested UGT isoforms, UGT1A1, UGT1A3, UGT2B7 and UGT2B15 were strongly inhibited by tacro in a concentration-dependent manner. Tacrolimus 93-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 23016456-4 2012 Among the tested UGT isoforms, UGT1A1, UGT1A3, UGT2B7 and UGT2B15 were strongly inhibited by tacro in a concentration-dependent manner. Tacrolimus 93-98 UDP glucuronosyltransferase family 1 member A3 Homo sapiens 39-45 23016456-4 2012 Among the tested UGT isoforms, UGT1A1, UGT1A3, UGT2B7 and UGT2B15 were strongly inhibited by tacro in a concentration-dependent manner. Tacrolimus 93-98 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 47-53 23016456-4 2012 Among the tested UGT isoforms, UGT1A1, UGT1A3, UGT2B7 and UGT2B15 were strongly inhibited by tacro in a concentration-dependent manner. Tacrolimus 93-98 UDP glucuronosyltransferase family 2 member B15 Homo sapiens 58-65 23016456-7 2012 Inhibition of these important UGT isoforms in human liver might be an important reason for clinically frequent drug-drug interaction between tacro and other drugs. Tacrolimus 141-146 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 30-33 22670785-6 2012 Expression of the peripheral homing receptors CXCR3 (r = 0.44, P < 0.05) and CCR5 (r = 0.45, P < 0.05) on FOXP3+ Tregs correlated with renal allograft function (eGFR) in patients receiving tacrolimus (n = 28), but not cyclosporine A (CsA) (n = 26). Tacrolimus 195-205 C-C motif chemokine receptor 5 Homo sapiens 80-84 22670785-6 2012 Expression of the peripheral homing receptors CXCR3 (r = 0.44, P < 0.05) and CCR5 (r = 0.45, P < 0.05) on FOXP3+ Tregs correlated with renal allograft function (eGFR) in patients receiving tacrolimus (n = 28), but not cyclosporine A (CsA) (n = 26). Tacrolimus 195-205 forkhead box P3 Homo sapiens 112-117 22729026-1 2012 PURPOSE OF REVIEW: The purpose of the review is to review the pathophysiology, available data, and our current recommendations for calcineurin inhibitor (cyclosporine and tacrolimus) treatment in antihistamine refractory chronic idiopathic urticaria (CIU) patients. Tacrolimus 171-181 calcineurin binding protein 1 Homo sapiens 131-152 22974867-0 2012 Long-term interleukin-2 assessment after conversion from a twice-daily to once-daily tacrolimus regimen in stable kidney recipients. Tacrolimus 85-95 interleukin 2 Homo sapiens 10-23 22568654-3 2012 Cyclosporine A (CsA) and tacrolimus (TAC) were associated with greater levels of Nox2 mRNA and epithelial to mesenchymal transition (EMT) in NRK52E cells. Tacrolimus 25-35 cytochrome b-245 beta chain Rattus norvegicus 81-85 22568654-3 2012 Cyclosporine A (CsA) and tacrolimus (TAC) were associated with greater levels of Nox2 mRNA and epithelial to mesenchymal transition (EMT) in NRK52E cells. Tacrolimus 37-40 cytochrome b-245 beta chain Rattus norvegicus 81-85 22786571-0 2012 The effect of CYP3A5 6986A>G and ABCB1 3435C>T on tacrolimus dose-adjusted trough levels and acute rejection rates in renal transplant patients: a systematic review and meta-analysis. Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 22579702-0 2012 Tacrolimus potently inhibits human osteoclastogenesis induced by IL-17 from human monocytes alone and suppresses human Th17 differentiation. Tacrolimus 0-10 interleukin 17A Homo sapiens 65-70 22579702-4 2012 The present study was undertaken to assess the effect of tacrolimus on IL-17-induced human osteoclastogenesis and human Th17 differentiation. Tacrolimus 57-67 interleukin 17A Homo sapiens 71-76 22579702-10 2012 Tacrolimus potently inhibited IL-17-induced osteoclastogenesis from human monocytes and osteoclast activation. Tacrolimus 0-10 interleukin 17A Homo sapiens 30-35 22579702-11 2012 Addition of tacrolimus also reduced production of IL-17 in human activated T cells stimulated with IL-23. Tacrolimus 12-22 interleukin 17A Homo sapiens 50-55 22579702-11 2012 Addition of tacrolimus also reduced production of IL-17 in human activated T cells stimulated with IL-23. Tacrolimus 12-22 interleukin 23 subunit alpha Homo sapiens 99-104 22579702-12 2012 Interestingly, the population of human IL-17(+)IFN-gamma(-) CD4 T cells or IL-17(+)TNF-alpha(+) CD4 T cells were decreased by adding of tacrolimus. Tacrolimus 136-146 interleukin 17A Homo sapiens 39-44 22579702-12 2012 Interestingly, the population of human IL-17(+)IFN-gamma(-) CD4 T cells or IL-17(+)TNF-alpha(+) CD4 T cells were decreased by adding of tacrolimus. Tacrolimus 136-146 interleukin 17A Homo sapiens 75-80 22579702-12 2012 Interestingly, the population of human IL-17(+)IFN-gamma(-) CD4 T cells or IL-17(+)TNF-alpha(+) CD4 T cells were decreased by adding of tacrolimus. Tacrolimus 136-146 tumor necrosis factor Homo sapiens 83-92 22579702-13 2012 The present study demonstrates that the inhibitory effect of tacrolimus on IL-17-induced osteoclastogenesis from human monocytes. Tacrolimus 61-71 interleukin 17A Homo sapiens 75-80 22579702-14 2012 Tacrolimus also inhibited expression of IL-17 or TNF-alpha by reducing the proportion of Th17, suggesting that therapeutic effect on Th17-associated disease such as RA, inflammatory bowel disease, multiple sclerosis, psoriasis, or allograft rejection. Tacrolimus 0-10 interleukin 17A Homo sapiens 40-45 22579702-14 2012 Tacrolimus also inhibited expression of IL-17 or TNF-alpha by reducing the proportion of Th17, suggesting that therapeutic effect on Th17-associated disease such as RA, inflammatory bowel disease, multiple sclerosis, psoriasis, or allograft rejection. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 49-58 21631587-7 2012 Cyclosporine and TAC led to a comparable and significant decrease in the efflux ratio of ImTORs, suggesting inhibition of a P-gp-mediated efflux transport. Tacrolimus 17-20 phosphoglycolate phosphatase Homo sapiens 124-128 22688515-5 2012 In neonatal rat cardiomyocytes, NHE1 stimulated hypertrophic gene expression and the NFAT pathway, which were inhibited by a CaN inhibitor, FK506. Tacrolimus 140-145 solute carrier family 9 member A1 Rattus norvegicus 32-36 22688515-5 2012 In neonatal rat cardiomyocytes, NHE1 stimulated hypertrophic gene expression and the NFAT pathway, which were inhibited by a CaN inhibitor, FK506. Tacrolimus 140-145 nuclear factor of activated T-cells 5 Rattus norvegicus 85-89 22786571-1 2012 In the present study, we performed a systematic review and meta-analysis on published data to examine the impact of CYP3A5 6986A>G and ABCB1 3435C>T polymorphisms on tacrolimus dose-adjusted trough levels (C0/D) and acute rejection rates in adult renal transplant patients. Tacrolimus 172-182 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 116-122 23018257-0 2012 Influence of conversion from cyclosporine A to tacrolimus on insulin sensitivity assessed by euglicaemic hyperinsulinemic clamp technique in patients after kidney transplantation. Tacrolimus 47-57 insulin Homo sapiens 61-68 22504573-0 2012 Meta-analysis of the effect of MDR1 C3435 polymorphism on tacrolimus pharmacokinetics in renal transplant recipients. Tacrolimus 58-68 ATP binding cassette subfamily B member 1 Homo sapiens 31-35 22504573-1 2012 BACKGROUND: The published data revealed conflicting results of the polymorphism of MDR1 exon 26 SNP C3435T on the pharmacokinetics of tacrolimus in different post transplant times; thus, the aim was to perform a meta-analysis of different post transplant times to investigate the influence of SNP C3435T on the tacrolimus pharmacokinetics. Tacrolimus 134-144 ATP binding cassette subfamily B member 1 Homo sapiens 83-87 22504573-6 2012 CONCLUSIONS: Our meta-analysis of available studies has demonstrated a definite correlation between the SNP C3435T in MDR1 gene and pharmacokinetics of tacrolimus. Tacrolimus 152-162 ATP binding cassette subfamily B member 1 Homo sapiens 118-122 22814600-7 2012 Tacrolimus exposure reduced Cn/NFAT-dependent expression of factors essential for insulin dense core granule formation and secretion and neonatal beta cell proliferation, consistent with our genetic studies. Tacrolimus 0-10 insulin Homo sapiens 82-89 22841242-10 2012 Within the rejection group the frequency of patients with the GSTM1 null genotype was higher among subjects prescribed cyclosporine A versus tacrolimus (P = .029). Tacrolimus 141-151 glutathione S-transferase mu 1 Homo sapiens 62-67 22841242-12 2012 Anti-GSTT1 antibody was observed more frequently albeit not significantly, among the cyclosporine versus tacrolimus patient group (P = .16). Tacrolimus 105-115 glutathione S-transferase theta 1 Homo sapiens 5-10 22841206-6 2012 In addition, patients with low CNI blood levels showed higher serum levels of IL-9, an effect that was greater with tacrolimus, albeit not significantly. Tacrolimus 116-126 interleukin 9 Homo sapiens 78-82 22643331-0 2012 Inhibitory effect of tacrolimus on p38 mitogen-activated protein kinase signaling in kidney transplant recipients measured by whole-blood phosphospecific flow cytometry. Tacrolimus 21-31 mitogen-activated protein kinase 14 Homo sapiens 35-38 22993851-6 2012 While after the effects of CYP3A4*18B genotype were eliminated, the C/D" ratio of tacrolimus in patients with CYP3A5*1/*1 and *1/*3 genotype group was significantly lower than those with CYP3A5*3/*3 genotype groups (P < 0.01). Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 27-33 22993851-6 2012 While after the effects of CYP3A4*18B genotype were eliminated, the C/D" ratio of tacrolimus in patients with CYP3A5*1/*1 and *1/*3 genotype group was significantly lower than those with CYP3A5*3/*3 genotype groups (P < 0.01). Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 110-116 22993851-6 2012 While after the effects of CYP3A4*18B genotype were eliminated, the C/D" ratio of tacrolimus in patients with CYP3A5*1/*1 and *1/*3 genotype group was significantly lower than those with CYP3A5*3/*3 genotype groups (P < 0.01). Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 187-193 22993851-0 2012 [Effect of CYP3A4*18B, CYP3A5*3 gene polymorphism on dosage and concentration of tacrolimus in renal transplant patients]. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 11-17 22993851-0 2012 [Effect of CYP3A4*18B, CYP3A5*3 gene polymorphism on dosage and concentration of tacrolimus in renal transplant patients]. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 23-29 22421346-6 2012 Treatment of osteoblast precursor cells with rapamycin or FK-506, either alone, or synergistically with BMP-2, increased levels of phospho-Smad 1/5/8 protein and transcription of Runx-2, Osx and Smad-7, consistent with a role in promoting osteoblast differentiation. Tacrolimus 58-64 bone morphogenetic protein 2 Homo sapiens 104-109 22615397-5 2012 Our method successfully identified specific disruption of the Hom3:Fpr1 interaction by the immunosuppressant FK506, illustrating the assay"s capacity to identify chemical inhibitors of protein-protein interactions. Tacrolimus 109-114 aspartate kinase Saccharomyces cerevisiae S288C 62-66 22615397-5 2012 Our method successfully identified specific disruption of the Hom3:Fpr1 interaction by the immunosuppressant FK506, illustrating the assay"s capacity to identify chemical inhibitors of protein-protein interactions. Tacrolimus 109-114 peptidylprolyl isomerase FPR1 Saccharomyces cerevisiae S288C 67-71 22421346-6 2012 Treatment of osteoblast precursor cells with rapamycin or FK-506, either alone, or synergistically with BMP-2, increased levels of phospho-Smad 1/5/8 protein and transcription of Runx-2, Osx and Smad-7, consistent with a role in promoting osteoblast differentiation. Tacrolimus 58-64 RUNX family transcription factor 2 Homo sapiens 179-185 22421346-6 2012 Treatment of osteoblast precursor cells with rapamycin or FK-506, either alone, or synergistically with BMP-2, increased levels of phospho-Smad 1/5/8 protein and transcription of Runx-2, Osx and Smad-7, consistent with a role in promoting osteoblast differentiation. Tacrolimus 58-64 Sp7 transcription factor Homo sapiens 187-190 22421346-6 2012 Treatment of osteoblast precursor cells with rapamycin or FK-506, either alone, or synergistically with BMP-2, increased levels of phospho-Smad 1/5/8 protein and transcription of Runx-2, Osx and Smad-7, consistent with a role in promoting osteoblast differentiation. Tacrolimus 58-64 SMAD family member 7 Homo sapiens 195-201 22421346-7 2012 Only FK-506 was able to enhance osteocalcin transcripts and Alizarin Red staining, both late markers for differentiation. Tacrolimus 5-11 bone gamma-carboxyglutamate protein Homo sapiens 32-43 22160488-0 2012 Effect of the calcineurin inhibitor FK506 on K+-Cl- cotransporter 2 expression in the mouse hippocampus after kainic acid-induced status epilepticus. Tacrolimus 36-41 solute carrier family 12, member 5 Mus musculus 45-67 22476221-6 2012 Both protease inhibitors can modify the levels of drugs metabolized by the CYP3A/4 pathway, and in posttransplant patients, the protease inhibitors increase the levels of cyclosporine and tacrolimus, with the magnitude of the drug-drug interactions varying with protease inhibitor and type of calcineurin inhibitor (CNI). Tacrolimus 188-198 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 75-82 22476221-6 2012 Both protease inhibitors can modify the levels of drugs metabolized by the CYP3A/4 pathway, and in posttransplant patients, the protease inhibitors increase the levels of cyclosporine and tacrolimus, with the magnitude of the drug-drug interactions varying with protease inhibitor and type of calcineurin inhibitor (CNI). Tacrolimus 188-198 calcineurin binding protein 1 Homo sapiens 293-314 22476221-6 2012 Both protease inhibitors can modify the levels of drugs metabolized by the CYP3A/4 pathway, and in posttransplant patients, the protease inhibitors increase the levels of cyclosporine and tacrolimus, with the magnitude of the drug-drug interactions varying with protease inhibitor and type of calcineurin inhibitor (CNI). Tacrolimus 188-198 calcineurin binding protein 1 Homo sapiens 316-319 22304537-0 2012 Frequencies of CYP3A5*1/*3 variants in a Moroccan population and effect on tacrolimus daily dose requirements in renal transplant patients. Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 22304537-7 2012 In the second part of this work, we assessed the influence of the CYP3A5 polymorphism on tacrolimus doses required for 10 renal transplant patients who are receiving tacrolimus as immunosuppressive therapy. Tacrolimus 89-99 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 66-72 22304537-7 2012 In the second part of this work, we assessed the influence of the CYP3A5 polymorphism on tacrolimus doses required for 10 renal transplant patients who are receiving tacrolimus as immunosuppressive therapy. Tacrolimus 166-176 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 66-72 22304537-8 2012 Our results showed that, during the first 3 months after kidney transplantation, the tacrolimus daily requirements for heterozygous patients (CYP3A5*3/*1) were higher compared with homozygous patients (CYP3A5*3/*3) (0.133 +- 0.026 vs. 0.21 +- 0.037 mg/kg/day). Tacrolimus 85-95 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 142-148 22304537-9 2012 After the third month the difference was also observed, whereby the mean of tacrolimus daily requirements for patients with CYP3A5*3/*3 and CYP3A5*1/*3 was 0.053 +- 0.013 and 0.08 +- 0.014 mg/kg/day, respectively. Tacrolimus 76-86 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 124-130 22304537-9 2012 After the third month the difference was also observed, whereby the mean of tacrolimus daily requirements for patients with CYP3A5*3/*3 and CYP3A5*1/*3 was 0.053 +- 0.013 and 0.08 +- 0.014 mg/kg/day, respectively. Tacrolimus 76-86 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 140-146 22160488-4 2012 In the present study, we investigated the altered expression of KCC2 and the effects of the CaN inhibitor FK506 on KCC2 expression in the mouse hippocampus following kainic acid (KA) treatment. Tacrolimus 106-111 solute carrier family 12, member 5 Mus musculus 115-119 22160488-10 2012 In particular, Western blot analysis showed that KCC2 expression was time dependent, with a peak at 6 h and a return to decreased levels at 48 h, whereas FK506 pretreatment inhibited the KA-induced decrease in KCC2 expression in the hippocampus. Tacrolimus 154-159 solute carrier family 12, member 5 Mus musculus 210-214 22160488-11 2012 Immunofluorescence showed that FK506 pretreatment protected the loss of inhibitory GABAergic KCC2-expressing neurons following KA treatment. Tacrolimus 31-36 solute carrier family 12, member 5 Mus musculus 93-97 22803008-11 2012 RT-PCR results of the FK506 groups showed that several bone-related genes-osteopontin, osteonectin, and type I collagen (Col-I)-were expressed more than the control in the beginning, but the intensity of expression decreased over time. Tacrolimus 22-27 secreted phosphoprotein 1 Rattus norvegicus 74-85 22803008-11 2012 RT-PCR results of the FK506 groups showed that several bone-related genes-osteopontin, osteonectin, and type I collagen (Col-I)-were expressed more than the control in the beginning, but the intensity of expression decreased over time. Tacrolimus 22-27 secreted protein acidic and cysteine rich Rattus norvegicus 87-98 22405967-2 2012 The aim of this study was to investigate the effect of Eudragit E/HCl (E-SD) on the reprecipitation of a poorly water-soluble drug, tacrolimus. Tacrolimus 132-142 esterase D Homo sapiens 71-75 22365914-4 2012 Tacrolimus treatment did not modify neither systolic nor diastolic arterial pressure but increased ET-1 content, ET(A)- and ET(B)-type receptor expression in aorta. Tacrolimus 0-10 endothelin 1 Rattus norvegicus 99-103 22365914-4 2012 Tacrolimus treatment did not modify neither systolic nor diastolic arterial pressure but increased ET-1 content, ET(A)- and ET(B)-type receptor expression in aorta. Tacrolimus 0-10 endothelin receptor type A Rattus norvegicus 113-119 22365914-5 2012 Proteomic study revealed that tacrolimus treatment modified the expression of aortic proteins associated with the cytoskeleton as some isotypes of lamin A and beta-tropomyosin; and energetic metabolism such as ATP synthase gamma chain, NADH dehydrogenase ubiquinone, acyl CoA dehydrogenase long chain mitochondrial and phosphatidylinositol 3-kinase regulatory subunit gamma. Tacrolimus 30-40 lamin A/C Rattus norvegicus 147-154 22365914-5 2012 Proteomic study revealed that tacrolimus treatment modified the expression of aortic proteins associated with the cytoskeleton as some isotypes of lamin A and beta-tropomyosin; and energetic metabolism such as ATP synthase gamma chain, NADH dehydrogenase ubiquinone, acyl CoA dehydrogenase long chain mitochondrial and phosphatidylinositol 3-kinase regulatory subunit gamma. Tacrolimus 30-40 tropomyosin 2 Rattus norvegicus 159-175 22365914-6 2012 Aortic expression of gp91-phox and MnSOD was also increased by tacrolimus. Tacrolimus 63-73 cytochrome b-245 beta chain Rattus norvegicus 21-30 22365914-6 2012 Aortic expression of gp91-phox and MnSOD was also increased by tacrolimus. Tacrolimus 63-73 superoxide dismutase 2 Rattus norvegicus 35-40 22365914-7 2012 Bosentan co-administration with tacrolimus prevented also changes in ET-1 content and the expression of proteins associated with energetic metabolism. Tacrolimus 32-42 endothelin 1 Rattus norvegicus 69-73 22365914-9 2012 As conclusion, tacrolimus treatment increased ET-1 content in aortic wall and modified the expression of proteins associated with the cytoskeleton and energetic metabolism independently of changes on blood pressure. Tacrolimus 15-25 endothelin 1 Rattus norvegicus 46-50 22405967-5 2012 Supersaturation profiles of tacrolimus were observed, and were maintained for 24h only with E-SD. Tacrolimus 28-38 esterase D Homo sapiens 92-96 22405967-7 2012 Solid dispersions prepared with E-SD showed higher solubility of tacrolimus compared with that of HPMC. Tacrolimus 65-75 esterase D Homo sapiens 32-36 22455398-3 2012 Rapamycin and FK506 are two macrocyclic natural products, which tightly bind to most FKBP family members, including FKBP51 and FKBP52. Tacrolimus 14-19 FKBP prolyl isomerase 4 Homo sapiens 127-133 22495472-8 2012 The association between IL-28B polymorphism and ACR occurrence was evident in tacrolimus but not in cyclosporine-treated patients. Tacrolimus 78-88 interferon lambda 3 Homo sapiens 24-30 30532941-10 2012 This case demonstrates that conversion to the extended-release formulation of tacrolimus from other calcineurin inhibitor preparations is a reasonable choice to consider in the management of compromised immunosuppressive therapy adherence in heart transplant patients during the late posttransplant period. Tacrolimus 78-88 calcineurin binding protein 1 Homo sapiens 100-121 22091865-1 2012 BACKGROUND AND PURPOSE The immunosuppressive macrolide FK506 (tacrolimus) shows neuroregenerative action by a mechanism that appears to involve the Hsp90-binding immunophilin FKBP52. Tacrolimus 55-60 heat shock protein 86, pseudogene 1 Mus musculus 148-153 22408025-9 2012 Treatment of the isolated knockout myocytes with FK506, which inhibits the association of FKBP12.6 with the ryanodine receptor, restored contractile function. Tacrolimus 49-54 FK506 binding protein 1b Mus musculus 90-98 22704849-0 2012 CYP3A5 polymorphism in Mexican renal transplant recipients and its association with tacrolimus dosing. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 22704849-1 2012 BACKGROUND AND AIMS: Variability in CYP3A5 expression associated with differences in tacrolimus bioavailability has been documented. Tacrolimus 85-95 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 22704849-9 2012 The CYP3A5 phenotype had a significant impact in tacrolimus bioavailability, as wild-type carriers required higher dosing compared to mutated carriers to achieve similar drug trough levels. Tacrolimus 49-59 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 22704849-10 2012 Patients with CYP3A5*1*1 genotype had a median dose requirement of 0.16 mg/kg/day, CYP3A5*1*3 patients had a median tacrolimus dose of 0.13 mg/kg/day and CYP3A5*3*3 had a median dose of 0.07 mg/kg/day (Kruskal-Wallis, p <0.0001). Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 83-89 22704849-10 2012 Patients with CYP3A5*1*1 genotype had a median dose requirement of 0.16 mg/kg/day, CYP3A5*1*3 patients had a median tacrolimus dose of 0.13 mg/kg/day and CYP3A5*3*3 had a median dose of 0.07 mg/kg/day (Kruskal-Wallis, p <0.0001). Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 83-89 22704849-11 2012 CONCLUSIONS: Of the Mexican transplant recipients, 52.2% were CYP3A5*3*3 and required significantly lower tacrolimus dose than those with CYP3A5*1 allele. Tacrolimus 106-116 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 62-68 22091865-1 2012 BACKGROUND AND PURPOSE The immunosuppressive macrolide FK506 (tacrolimus) shows neuroregenerative action by a mechanism that appears to involve the Hsp90-binding immunophilin FKBP52. Tacrolimus 55-60 FK506 binding protein 4 Mus musculus 175-181 22091865-1 2012 BACKGROUND AND PURPOSE The immunosuppressive macrolide FK506 (tacrolimus) shows neuroregenerative action by a mechanism that appears to involve the Hsp90-binding immunophilin FKBP52. Tacrolimus 62-72 heat shock protein 86, pseudogene 1 Mus musculus 148-153 22091865-1 2012 BACKGROUND AND PURPOSE The immunosuppressive macrolide FK506 (tacrolimus) shows neuroregenerative action by a mechanism that appears to involve the Hsp90-binding immunophilin FKBP52. Tacrolimus 62-72 FK506 binding protein 4 Mus musculus 175-181 22091865-6 2012 KEY RESULTS In undifferentiated cells, FKBP52, Hsp90 and p23 are located in the cell nucleus, forming an annular structure that disassembles when the differentiation process is triggered by FK506. Tacrolimus 190-195 FK506 binding protein 4 Mus musculus 39-45 22091865-6 2012 KEY RESULTS In undifferentiated cells, FKBP52, Hsp90 and p23 are located in the cell nucleus, forming an annular structure that disassembles when the differentiation process is triggered by FK506. Tacrolimus 190-195 heat shock protein 86, pseudogene 1 Mus musculus 47-52 22091865-6 2012 KEY RESULTS In undifferentiated cells, FKBP52, Hsp90 and p23 are located in the cell nucleus, forming an annular structure that disassembles when the differentiation process is triggered by FK506. Tacrolimus 190-195 prostaglandin E synthase 3 Mus musculus 57-60 22101623-4 2012 Patients with a small tacrolimus requirement were at increased risk for multiple infections (OR 1.533, p = 0.0008) and higher systolic blood pressure (OR 1.385, p = 0.022) and showed a significant association with the CYP3A5*3/*3 genotype adjusted by MDR1 polymorphisms C3435T and C1236T (OR 8.104, p = 0.0001). Tacrolimus 22-32 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 218-224 22101623-4 2012 Patients with a small tacrolimus requirement were at increased risk for multiple infections (OR 1.533, p = 0.0008) and higher systolic blood pressure (OR 1.385, p = 0.022) and showed a significant association with the CYP3A5*3/*3 genotype adjusted by MDR1 polymorphisms C3435T and C1236T (OR 8.104, p = 0.0001). Tacrolimus 22-32 ATP binding cassette subfamily B member 1 Homo sapiens 251-255 22093061-6 2012 Children with AHR had a lower mean tacrolimus trough level and were more likely to have a sub-therapeutic trough at six months (3.5 vs. 5.5 ng/mL, p = 0.05); mean MMF doses were lower at all times points except three months in the AHR group (not statistically significant). Tacrolimus 35-45 aryl hydrocarbon receptor Homo sapiens 14-17 22183771-4 2012 RESULTS: A donor source-adjusted linear mixed model with multilevel analysis adjusting for age, body weight, hematocrit, and serum creatinine showed that CYP3A5 genotype is associated with dose-adjusted level of tacrolimus (p < 0.001). Tacrolimus 212-222 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 154-160 22183771-5 2012 The influence of ABCB1 polymorphisms on the pharmacokinetics or complications of tacrolimus was less certain in our study. Tacrolimus 81-91 ATP binding cassette subfamily B member 1 Homo sapiens 17-22 22183771-8 2012 Genotype of CYP3A5 variants along with significant clinical covariates may be useful in individualizing tacrolimus therapy in kidney transplantation patients. Tacrolimus 104-114 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 22493070-7 2012 Ang II effects were blunted by an Ang II type 1 receptor antagonist (candesartan) and inhibitors of calcineurin (cyclosporine A and FK506) and nuclear factor of activated T-cells (VIVIT). Tacrolimus 132-137 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Mus musculus 0-6 22277940-7 2012 Finally, the increased expression of IL-33 and ST2 caused by irritant, allergen, or SEB challenge was suppressed by topical tacrolimus treatment. Tacrolimus 124-134 interleukin 33 Mus musculus 37-42 22277940-7 2012 Finally, the increased expression of IL-33 and ST2 caused by irritant, allergen, or SEB challenge was suppressed by topical tacrolimus treatment. Tacrolimus 124-134 interleukin 1 receptor-like 1 Mus musculus 47-50 21988494-4 2012 WHAT THIS STUDY ADDS: This is the first trial to investigate beta cell function and insulin sensitivity using gold standard methodology in healthy human volunteers treated with clinically relevant doses of ciclosporin and tacrolimus. Tacrolimus 222-232 insulin Homo sapiens 84-91 22743729-3 2012 Variation in disposition of tacrolimus is associated with genetic variation in CYP3A5. Tacrolimus 28-38 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 79-85 22743729-4 2012 Hence, could genetic variation in CYP3A5 or other genes involved in tacrolimus disposition and effect be associated with a risk for tacrolimus-induced nephrotoxicity? Tacrolimus 68-78 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 22743729-4 2012 Hence, could genetic variation in CYP3A5 or other genes involved in tacrolimus disposition and effect be associated with a risk for tacrolimus-induced nephrotoxicity? Tacrolimus 132-142 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 22743729-5 2012 To perform a review of the literature and to identify if genetic variation in CYP3A5 or other genes involved in tacrolimus disposition or effect may be associated with tacrolimus-induced nephrotoxicity and/or renal dysfunction in solid organ transplant recipients. Tacrolimus 112-122 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 78-84 22743729-5 2012 To perform a review of the literature and to identify if genetic variation in CYP3A5 or other genes involved in tacrolimus disposition or effect may be associated with tacrolimus-induced nephrotoxicity and/or renal dysfunction in solid organ transplant recipients. Tacrolimus 168-178 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 78-84 22743729-9 2012 In kidney recipients, associations between donor ABCB1, recipient CCR5 genotype and tacrolimus-induced nephrotoxicity were found. Tacrolimus 84-94 ATP binding cassette subfamily B member 1 Homo sapiens 49-54 22743729-9 2012 In kidney recipients, associations between donor ABCB1, recipient CCR5 genotype and tacrolimus-induced nephrotoxicity were found. Tacrolimus 84-94 C-C motif chemokine receptor 5 Homo sapiens 66-70 22743729-11 2012 In liver recipients, a possible association between recipient ACE, CYP3A5, ABCB1 and CYP2C8 genetic polymorphisms and tacrolimus-induced nephrotoxicity was suggested. Tacrolimus 118-128 angiotensin I converting enzyme Homo sapiens 62-65 22743729-11 2012 In liver recipients, a possible association between recipient ACE, CYP3A5, ABCB1 and CYP2C8 genetic polymorphisms and tacrolimus-induced nephrotoxicity was suggested. Tacrolimus 118-128 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-73 22743729-11 2012 In liver recipients, a possible association between recipient ACE, CYP3A5, ABCB1 and CYP2C8 genetic polymorphisms and tacrolimus-induced nephrotoxicity was suggested. Tacrolimus 118-128 ATP binding cassette subfamily B member 1 Homo sapiens 75-80 22743729-11 2012 In liver recipients, a possible association between recipient ACE, CYP3A5, ABCB1 and CYP2C8 genetic polymorphisms and tacrolimus-induced nephrotoxicity was suggested. Tacrolimus 118-128 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 85-91 22743729-12 2012 In heart recipients, TGF-beta genetic polymorphisms were associated with tacrolimus-induced nephrotoxicity. Tacrolimus 73-83 transforming growth factor beta 1 Homo sapiens 21-29 22743729-14 2012 CONCLUSIONS: Limited evidence suggests that variation in genes involved in pharmacokinetics (ABCB1 and CYP3A5) and pharmacodynamics (TGF-beta, CYP2C8, ACE, CCR5) of tacrolimus may impact a transplant recipients" risk to develop tacrolimus-induced nephrotoxicity across different transplant organ groups. Tacrolimus 165-175 ATP binding cassette subfamily B member 1 Homo sapiens 93-98 22743729-14 2012 CONCLUSIONS: Limited evidence suggests that variation in genes involved in pharmacokinetics (ABCB1 and CYP3A5) and pharmacodynamics (TGF-beta, CYP2C8, ACE, CCR5) of tacrolimus may impact a transplant recipients" risk to develop tacrolimus-induced nephrotoxicity across different transplant organ groups. Tacrolimus 165-175 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 103-109 22743729-14 2012 CONCLUSIONS: Limited evidence suggests that variation in genes involved in pharmacokinetics (ABCB1 and CYP3A5) and pharmacodynamics (TGF-beta, CYP2C8, ACE, CCR5) of tacrolimus may impact a transplant recipients" risk to develop tacrolimus-induced nephrotoxicity across different transplant organ groups. Tacrolimus 165-175 transforming growth factor beta 1 Homo sapiens 133-141 22743729-14 2012 CONCLUSIONS: Limited evidence suggests that variation in genes involved in pharmacokinetics (ABCB1 and CYP3A5) and pharmacodynamics (TGF-beta, CYP2C8, ACE, CCR5) of tacrolimus may impact a transplant recipients" risk to develop tacrolimus-induced nephrotoxicity across different transplant organ groups. Tacrolimus 165-175 C-C motif chemokine receptor 5 Homo sapiens 156-160 22743729-14 2012 CONCLUSIONS: Limited evidence suggests that variation in genes involved in pharmacokinetics (ABCB1 and CYP3A5) and pharmacodynamics (TGF-beta, CYP2C8, ACE, CCR5) of tacrolimus may impact a transplant recipients" risk to develop tacrolimus-induced nephrotoxicity across different transplant organ groups. Tacrolimus 228-238 ATP binding cassette subfamily B member 1 Homo sapiens 93-98 22743729-14 2012 CONCLUSIONS: Limited evidence suggests that variation in genes involved in pharmacokinetics (ABCB1 and CYP3A5) and pharmacodynamics (TGF-beta, CYP2C8, ACE, CCR5) of tacrolimus may impact a transplant recipients" risk to develop tacrolimus-induced nephrotoxicity across different transplant organ groups. Tacrolimus 228-238 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 103-109 22743729-14 2012 CONCLUSIONS: Limited evidence suggests that variation in genes involved in pharmacokinetics (ABCB1 and CYP3A5) and pharmacodynamics (TGF-beta, CYP2C8, ACE, CCR5) of tacrolimus may impact a transplant recipients" risk to develop tacrolimus-induced nephrotoxicity across different transplant organ groups. Tacrolimus 228-238 transforming growth factor beta 1 Homo sapiens 133-141 22743729-14 2012 CONCLUSIONS: Limited evidence suggests that variation in genes involved in pharmacokinetics (ABCB1 and CYP3A5) and pharmacodynamics (TGF-beta, CYP2C8, ACE, CCR5) of tacrolimus may impact a transplant recipients" risk to develop tacrolimus-induced nephrotoxicity across different transplant organ groups. Tacrolimus 228-238 C-C motif chemokine receptor 5 Homo sapiens 156-160 22491658-2 2012 Continuous intravenous nicardipine (CIVN), prescribed for posttransplant hypertension, inhibits tacrolimus metabolism by cytochrome P450 (CYP) 3A4 and could lead to tacrolimus overexposure in patients genetically lacking the alternative pathway for tacrolimus metabolism, CYP3A5. Tacrolimus 96-106 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 121-146 22491658-6 2012 Compared with the other two groups combined, CYP3A5*3/*3 cases had higher median dose-adjusted MaxC0 (330 vs. 175, P=0.012), less time to MaxC0 (42 vs. 72 hr, P<0.001), and more scheduled tacrolimus doses held per patient (1.75 vs. 0.4, P=0.007). Tacrolimus 191-201 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 22491658-8 2012 CONCLUSION: CYP3A5 nonexpressors simultaneously treated with tacrolimus and CIVN may be at increased risk for tacrolimus toxicity. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 21720923-7 2012 Tacrolimus could be an additional or alternative modality for treating refractory HPS. Tacrolimus 0-10 HPS1 biogenesis of lysosomal organelles complex 3 subunit 1 Homo sapiens 82-85 22205779-0 2012 Cyclosporine A- and tacrolimus-mediated inhibition of CYP3A4 and CYP3A5 in vitro. Tacrolimus 20-30 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 54-60 22205779-0 2012 Cyclosporine A- and tacrolimus-mediated inhibition of CYP3A4 and CYP3A5 in vitro. Tacrolimus 20-30 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 65-71 22112437-10 2012 When TNCB-sensitized GATA-3 Tg mice were treated with a high dose of tacrolimus, ear swelling was not significantly decreased, compared with the results in WT mice. Tacrolimus 69-79 GATA binding protein 3 Mus musculus 21-27 22422348-11 2012 CONCLUSIONS: CYP3A5 genotyping may represent a useful tool to better evaluate the appropriate initial dose of tacrolimus for patients carrying a liver with the CYP3A5*1/*1 genotype. Tacrolimus 110-120 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 22422348-11 2012 CONCLUSIONS: CYP3A5 genotyping may represent a useful tool to better evaluate the appropriate initial dose of tacrolimus for patients carrying a liver with the CYP3A5*1/*1 genotype. Tacrolimus 110-120 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 160-166 22328519-0 2012 Disparities in the association of the ryanodine receptor and the FK506-binding proteins in mammalian heart. Tacrolimus 65-70 ryanodine receptor 1 Homo sapiens 38-56 22328519-9 2012 FKBP interactions with RyR2 are very strong and resistant to drug (FK506, rapamycin and cyclic ADPribose) and redox (H(2)O(2) and diamide) treatment. Tacrolimus 67-72 ryanodine receptor 2 Homo sapiens 23-27 22328519-12 2012 Our results challenge the widespread perception that RyR2 associates exclusively with FKBP12.6 to near saturation, with important implications for the role of the FK506-binding proteins in RyR2 pathophysiology and cardiac disease. Tacrolimus 163-168 ryanodine receptor 2 Homo sapiens 53-57 22369694-1 2012 We retrospectively examined the association of polymorphisms in the CYP3A, CYP2J2, CYP2C8, and ABCB1 genes with pharmacokinetic (PKs) and pharmacodynamic (PDs) parameters of tacrolimus in 103 renal transplant recipients for a period of 1 year. Tacrolimus 174-184 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 75-81 22406656-3 2012 In a previous study of the pharmacokinetics (PK) of these formulations, mean systemic exposure [area under the curve from 0 to 24 hours (AUC0-24)] of tacrolimus on day 1 was approximately 30% lower for Tacrolimus QD than for Tacrolimus BID; by day 14, systemic exposure was similar; however, the mean dose of Tacrolimus QD was higher to achieve similar systemic exposure as Tacrolimus BID. Tacrolimus 150-160 BH3 interacting domain death agonist Homo sapiens 236-239 22406656-3 2012 In a previous study of the pharmacokinetics (PK) of these formulations, mean systemic exposure [area under the curve from 0 to 24 hours (AUC0-24)] of tacrolimus on day 1 was approximately 30% lower for Tacrolimus QD than for Tacrolimus BID; by day 14, systemic exposure was similar; however, the mean dose of Tacrolimus QD was higher to achieve similar systemic exposure as Tacrolimus BID. Tacrolimus 150-160 BH3 interacting domain death agonist Homo sapiens 385-388 22406656-3 2012 In a previous study of the pharmacokinetics (PK) of these formulations, mean systemic exposure [area under the curve from 0 to 24 hours (AUC0-24)] of tacrolimus on day 1 was approximately 30% lower for Tacrolimus QD than for Tacrolimus BID; by day 14, systemic exposure was similar; however, the mean dose of Tacrolimus QD was higher to achieve similar systemic exposure as Tacrolimus BID. Tacrolimus 202-212 BH3 interacting domain death agonist Homo sapiens 236-239 22406656-3 2012 In a previous study of the pharmacokinetics (PK) of these formulations, mean systemic exposure [area under the curve from 0 to 24 hours (AUC0-24)] of tacrolimus on day 1 was approximately 30% lower for Tacrolimus QD than for Tacrolimus BID; by day 14, systemic exposure was similar; however, the mean dose of Tacrolimus QD was higher to achieve similar systemic exposure as Tacrolimus BID. Tacrolimus 202-212 BH3 interacting domain death agonist Homo sapiens 385-388 22406656-7 2012 RESULTS: Mean AUC0-24 of tacrolimus on day 1 was approximately 16% lower for Tacrolimus QD than for Tacrolimus BID, although by day 3 onward, the exposure was similar between treatment groups. Tacrolimus 25-35 BH3 interacting domain death agonist Homo sapiens 111-114 22406656-9 2012 There was a good correlation between AUC0-24 and concentration of tacrolimus at 24 hours postdose for both formulations (Tacrolimus QD, r = 0.87; Tacrolimus BID, r = 0.92), and the slope of the line of best fit was similar. Tacrolimus 146-156 BH3 interacting domain death agonist Homo sapiens 157-160 22406656-10 2012 CONCLUSIONS: These results suggest that initiating tacrolimus therapy before transplant reduces the difference in exposure between Tacrolimus QD and Tacrolimus BID. Tacrolimus 51-61 BH3 interacting domain death agonist Homo sapiens 160-163 22406656-10 2012 CONCLUSIONS: These results suggest that initiating tacrolimus therapy before transplant reduces the difference in exposure between Tacrolimus QD and Tacrolimus BID. Tacrolimus 149-159 BH3 interacting domain death agonist Homo sapiens 160-163 22369694-6 2012 Our findings indicate that the CYP3A4*1B-CYP3A5*1 haplotype may have a more profound impact in tacrolimus PKs than the CYP3A5*1 allele. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 41-47 22369694-6 2012 Our findings indicate that the CYP3A4*1B-CYP3A5*1 haplotype may have a more profound impact in tacrolimus PKs than the CYP3A5*1 allele. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 119-125 22369694-1 2012 We retrospectively examined the association of polymorphisms in the CYP3A, CYP2J2, CYP2C8, and ABCB1 genes with pharmacokinetic (PKs) and pharmacodynamic (PDs) parameters of tacrolimus in 103 renal transplant recipients for a period of 1 year. Tacrolimus 174-184 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 83-89 22369694-6 2012 Our findings indicate that the CYP3A4*1B-CYP3A5*1 haplotype may have a more profound impact in tacrolimus PKs than the CYP3A5*1 allele. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 22264780-0 2012 Association between a common KCNJ11 polymorphism (rs5219) and new-onset posttransplant diabetes in patients treated with Tacrolimus. Tacrolimus 121-131 potassium inwardly rectifying channel subfamily J member 11 Homo sapiens 29-35 22260742-10 2012 Even after predonisolone and tacrolimus were tapered gradually and discontinued because of her good response, CRP and DAS-CRP became 0.0 mg/dL and 1.5, respectively. Tacrolimus 29-39 C-reactive protein Homo sapiens 110-113 22260742-10 2012 Even after predonisolone and tacrolimus were tapered gradually and discontinued because of her good response, CRP and DAS-CRP became 0.0 mg/dL and 1.5, respectively. Tacrolimus 29-39 C-reactive protein Homo sapiens 122-125 21964739-6 2012 These effects were inhibited by FK506 (10 muM), a specific inhibitor of calcineurin. Tacrolimus 32-37 latexin Homo sapiens 42-45 21550286-0 2012 Tacrolimus (FK506) inhibits interleukin-1beta-induced angiopoietin-1, Tie-2 receptor, and vascular endothelial growth factor through down-regulation of JNK and p38 pathway in human rheumatoid fibroblast-like synoviocytes. Tacrolimus 0-10 interleukin 1 beta Homo sapiens 28-45 21550286-0 2012 Tacrolimus (FK506) inhibits interleukin-1beta-induced angiopoietin-1, Tie-2 receptor, and vascular endothelial growth factor through down-regulation of JNK and p38 pathway in human rheumatoid fibroblast-like synoviocytes. Tacrolimus 0-10 angiopoietin 1 Homo sapiens 54-68 21550286-0 2012 Tacrolimus (FK506) inhibits interleukin-1beta-induced angiopoietin-1, Tie-2 receptor, and vascular endothelial growth factor through down-regulation of JNK and p38 pathway in human rheumatoid fibroblast-like synoviocytes. Tacrolimus 0-10 TEK receptor tyrosine kinase Homo sapiens 70-75 21550286-0 2012 Tacrolimus (FK506) inhibits interleukin-1beta-induced angiopoietin-1, Tie-2 receptor, and vascular endothelial growth factor through down-regulation of JNK and p38 pathway in human rheumatoid fibroblast-like synoviocytes. Tacrolimus 0-10 vascular endothelial growth factor A Homo sapiens 90-124 21550286-0 2012 Tacrolimus (FK506) inhibits interleukin-1beta-induced angiopoietin-1, Tie-2 receptor, and vascular endothelial growth factor through down-regulation of JNK and p38 pathway in human rheumatoid fibroblast-like synoviocytes. Tacrolimus 0-10 mitogen-activated protein kinase 8 Homo sapiens 152-155 21550286-0 2012 Tacrolimus (FK506) inhibits interleukin-1beta-induced angiopoietin-1, Tie-2 receptor, and vascular endothelial growth factor through down-regulation of JNK and p38 pathway in human rheumatoid fibroblast-like synoviocytes. Tacrolimus 0-10 mitogen-activated protein kinase 14 Homo sapiens 160-163 21550286-0 2012 Tacrolimus (FK506) inhibits interleukin-1beta-induced angiopoietin-1, Tie-2 receptor, and vascular endothelial growth factor through down-regulation of JNK and p38 pathway in human rheumatoid fibroblast-like synoviocytes. Tacrolimus 12-17 interleukin 1 beta Homo sapiens 28-45 21550286-0 2012 Tacrolimus (FK506) inhibits interleukin-1beta-induced angiopoietin-1, Tie-2 receptor, and vascular endothelial growth factor through down-regulation of JNK and p38 pathway in human rheumatoid fibroblast-like synoviocytes. Tacrolimus 12-17 angiopoietin 1 Homo sapiens 54-68 21550286-0 2012 Tacrolimus (FK506) inhibits interleukin-1beta-induced angiopoietin-1, Tie-2 receptor, and vascular endothelial growth factor through down-regulation of JNK and p38 pathway in human rheumatoid fibroblast-like synoviocytes. Tacrolimus 12-17 TEK receptor tyrosine kinase Homo sapiens 70-75 21550286-0 2012 Tacrolimus (FK506) inhibits interleukin-1beta-induced angiopoietin-1, Tie-2 receptor, and vascular endothelial growth factor through down-regulation of JNK and p38 pathway in human rheumatoid fibroblast-like synoviocytes. Tacrolimus 12-17 vascular endothelial growth factor A Homo sapiens 90-124 21550286-0 2012 Tacrolimus (FK506) inhibits interleukin-1beta-induced angiopoietin-1, Tie-2 receptor, and vascular endothelial growth factor through down-regulation of JNK and p38 pathway in human rheumatoid fibroblast-like synoviocytes. Tacrolimus 12-17 mitogen-activated protein kinase 8 Homo sapiens 152-155 21550286-0 2012 Tacrolimus (FK506) inhibits interleukin-1beta-induced angiopoietin-1, Tie-2 receptor, and vascular endothelial growth factor through down-regulation of JNK and p38 pathway in human rheumatoid fibroblast-like synoviocytes. Tacrolimus 12-17 mitogen-activated protein kinase 14 Homo sapiens 160-163 21550286-1 2012 OBJECT: This study aimed to identify the regulatory effect of tacrolimus on the interleukin-1beta (IL-1beta)-induced expressions of angiopoietin-1 (Ang-1), Tie-2 receptor (Tie-2), and vascular endothelial growth factor (VEGF) in human rheumatoid fibroblast-like synoviocytes (FLS) and to determine the regulatory mechanism in the mitogen-activated protein kinases (MAPKs) signaling pathway. Tacrolimus 62-72 interleukin 1 beta Homo sapiens 80-97 21550286-1 2012 OBJECT: This study aimed to identify the regulatory effect of tacrolimus on the interleukin-1beta (IL-1beta)-induced expressions of angiopoietin-1 (Ang-1), Tie-2 receptor (Tie-2), and vascular endothelial growth factor (VEGF) in human rheumatoid fibroblast-like synoviocytes (FLS) and to determine the regulatory mechanism in the mitogen-activated protein kinases (MAPKs) signaling pathway. Tacrolimus 62-72 interleukin 1 beta Homo sapiens 99-107 21550286-1 2012 OBJECT: This study aimed to identify the regulatory effect of tacrolimus on the interleukin-1beta (IL-1beta)-induced expressions of angiopoietin-1 (Ang-1), Tie-2 receptor (Tie-2), and vascular endothelial growth factor (VEGF) in human rheumatoid fibroblast-like synoviocytes (FLS) and to determine the regulatory mechanism in the mitogen-activated protein kinases (MAPKs) signaling pathway. Tacrolimus 62-72 angiopoietin 1 Homo sapiens 132-146 21550286-1 2012 OBJECT: This study aimed to identify the regulatory effect of tacrolimus on the interleukin-1beta (IL-1beta)-induced expressions of angiopoietin-1 (Ang-1), Tie-2 receptor (Tie-2), and vascular endothelial growth factor (VEGF) in human rheumatoid fibroblast-like synoviocytes (FLS) and to determine the regulatory mechanism in the mitogen-activated protein kinases (MAPKs) signaling pathway. Tacrolimus 62-72 angiopoietin 1 Homo sapiens 148-153 21550286-1 2012 OBJECT: This study aimed to identify the regulatory effect of tacrolimus on the interleukin-1beta (IL-1beta)-induced expressions of angiopoietin-1 (Ang-1), Tie-2 receptor (Tie-2), and vascular endothelial growth factor (VEGF) in human rheumatoid fibroblast-like synoviocytes (FLS) and to determine the regulatory mechanism in the mitogen-activated protein kinases (MAPKs) signaling pathway. Tacrolimus 62-72 TEK receptor tyrosine kinase Homo sapiens 156-161 21550286-1 2012 OBJECT: This study aimed to identify the regulatory effect of tacrolimus on the interleukin-1beta (IL-1beta)-induced expressions of angiopoietin-1 (Ang-1), Tie-2 receptor (Tie-2), and vascular endothelial growth factor (VEGF) in human rheumatoid fibroblast-like synoviocytes (FLS) and to determine the regulatory mechanism in the mitogen-activated protein kinases (MAPKs) signaling pathway. Tacrolimus 62-72 TEK receptor tyrosine kinase Homo sapiens 172-177 21550286-1 2012 OBJECT: This study aimed to identify the regulatory effect of tacrolimus on the interleukin-1beta (IL-1beta)-induced expressions of angiopoietin-1 (Ang-1), Tie-2 receptor (Tie-2), and vascular endothelial growth factor (VEGF) in human rheumatoid fibroblast-like synoviocytes (FLS) and to determine the regulatory mechanism in the mitogen-activated protein kinases (MAPKs) signaling pathway. Tacrolimus 62-72 vascular endothelial growth factor A Homo sapiens 184-218 21550286-1 2012 OBJECT: This study aimed to identify the regulatory effect of tacrolimus on the interleukin-1beta (IL-1beta)-induced expressions of angiopoietin-1 (Ang-1), Tie-2 receptor (Tie-2), and vascular endothelial growth factor (VEGF) in human rheumatoid fibroblast-like synoviocytes (FLS) and to determine the regulatory mechanism in the mitogen-activated protein kinases (MAPKs) signaling pathway. Tacrolimus 62-72 vascular endothelial growth factor A Homo sapiens 220-224 21550286-2 2012 METHODS: IL-1beta-induced Ang-1, Tie-2, and VEGF expressions with and without tacrolimus were measured in cultured FLS using real time-polymerase chain reaction, enzyme-linked immunosorbent assay, Western blotting, and immunofluorescence staining. Tacrolimus 78-88 interleukin 1 beta Homo sapiens 9-17 21550286-3 2012 The effect of tacrolimus on the regulation of Ang-1, Tie-2 and VEGF expressions through the MAPK signaling pathway was identified by Western blotting and immunofluorescence staining. Tacrolimus 14-24 angiopoietin 1 Homo sapiens 46-51 21550286-3 2012 The effect of tacrolimus on the regulation of Ang-1, Tie-2 and VEGF expressions through the MAPK signaling pathway was identified by Western blotting and immunofluorescence staining. Tacrolimus 14-24 TEK receptor tyrosine kinase Homo sapiens 53-58 21550286-3 2012 The effect of tacrolimus on the regulation of Ang-1, Tie-2 and VEGF expressions through the MAPK signaling pathway was identified by Western blotting and immunofluorescence staining. Tacrolimus 14-24 vascular endothelial growth factor A Homo sapiens 63-67 21550286-3 2012 The effect of tacrolimus on the regulation of Ang-1, Tie-2 and VEGF expressions through the MAPK signaling pathway was identified by Western blotting and immunofluorescence staining. Tacrolimus 14-24 mitogen-activated protein kinase 1 Homo sapiens 92-96 21550286-5 2012 Tacrolimus significantly inhibited Ang-1, Tie-2, and VEGF mRNA and protein in cultured FLS treated with 10 ng/ml IL-1beta. Tacrolimus 0-10 angiopoietin 1 Homo sapiens 35-40 21550286-5 2012 Tacrolimus significantly inhibited Ang-1, Tie-2, and VEGF mRNA and protein in cultured FLS treated with 10 ng/ml IL-1beta. Tacrolimus 0-10 TEK receptor tyrosine kinase Homo sapiens 42-47 21550286-5 2012 Tacrolimus significantly inhibited Ang-1, Tie-2, and VEGF mRNA and protein in cultured FLS treated with 10 ng/ml IL-1beta. Tacrolimus 0-10 vascular endothelial growth factor A Homo sapiens 53-57 21550286-5 2012 Tacrolimus significantly inhibited Ang-1, Tie-2, and VEGF mRNA and protein in cultured FLS treated with 10 ng/ml IL-1beta. Tacrolimus 0-10 interleukin 1 beta Homo sapiens 113-121 21550286-7 2012 However, the inhibitory effects of tacrolimus on Ang-1, Tie-2, and VEGF proteins were regulated by blocking the phosphorylations of JNK and p38 MAPK, but not that of ERK. Tacrolimus 35-45 angiopoietin 1 Homo sapiens 49-54 21550286-7 2012 However, the inhibitory effects of tacrolimus on Ang-1, Tie-2, and VEGF proteins were regulated by blocking the phosphorylations of JNK and p38 MAPK, but not that of ERK. Tacrolimus 35-45 TEK receptor tyrosine kinase Homo sapiens 56-61 21550286-7 2012 However, the inhibitory effects of tacrolimus on Ang-1, Tie-2, and VEGF proteins were regulated by blocking the phosphorylations of JNK and p38 MAPK, but not that of ERK. Tacrolimus 35-45 vascular endothelial growth factor A Homo sapiens 67-71 21550286-7 2012 However, the inhibitory effects of tacrolimus on Ang-1, Tie-2, and VEGF proteins were regulated by blocking the phosphorylations of JNK and p38 MAPK, but not that of ERK. Tacrolimus 35-45 mitogen-activated protein kinase 8 Homo sapiens 132-135 21550286-7 2012 However, the inhibitory effects of tacrolimus on Ang-1, Tie-2, and VEGF proteins were regulated by blocking the phosphorylations of JNK and p38 MAPK, but not that of ERK. Tacrolimus 35-45 mitogen-activated protein kinase 14 Homo sapiens 140-143 21550286-8 2012 CONCLUSION: This study demonstrates that tacrolimus inhibits the expressions of Ang-1, Tie-2, and VEGF by blocking the activations of the IL-1beta-mediated JNK and p38 MAPK pathways in human FLS. Tacrolimus 41-51 angiopoietin 1 Homo sapiens 80-85 21550286-8 2012 CONCLUSION: This study demonstrates that tacrolimus inhibits the expressions of Ang-1, Tie-2, and VEGF by blocking the activations of the IL-1beta-mediated JNK and p38 MAPK pathways in human FLS. Tacrolimus 41-51 TEK receptor tyrosine kinase Homo sapiens 87-92 21550286-8 2012 CONCLUSION: This study demonstrates that tacrolimus inhibits the expressions of Ang-1, Tie-2, and VEGF by blocking the activations of the IL-1beta-mediated JNK and p38 MAPK pathways in human FLS. Tacrolimus 41-51 vascular endothelial growth factor A Homo sapiens 98-102 21550286-8 2012 CONCLUSION: This study demonstrates that tacrolimus inhibits the expressions of Ang-1, Tie-2, and VEGF by blocking the activations of the IL-1beta-mediated JNK and p38 MAPK pathways in human FLS. Tacrolimus 41-51 interleukin 1 beta Homo sapiens 138-146 21550286-8 2012 CONCLUSION: This study demonstrates that tacrolimus inhibits the expressions of Ang-1, Tie-2, and VEGF by blocking the activations of the IL-1beta-mediated JNK and p38 MAPK pathways in human FLS. Tacrolimus 41-51 mitogen-activated protein kinase 8 Homo sapiens 156-159 21550286-8 2012 CONCLUSION: This study demonstrates that tacrolimus inhibits the expressions of Ang-1, Tie-2, and VEGF by blocking the activations of the IL-1beta-mediated JNK and p38 MAPK pathways in human FLS. Tacrolimus 41-51 mitogen-activated protein kinase 14 Homo sapiens 164-167 22264780-2 2012 Our aim was to define the contribution of KCNJ11 to new-onset diabetes after transplantation (NODAT) among patients treated with Tacrolimus (Tac). Tacrolimus 129-139 potassium inwardly rectifying channel subfamily J member 11 Homo sapiens 42-48 22265013-5 2012 FKBP14 belongs to the family of FK506-binding peptidyl-prolyl cis-trans isomerases (PPIases). Tacrolimus 32-37 FKBP prolyl isomerase 14 Homo sapiens 0-6 22239105-5 2012 Mean (SD) tacrolimus dose was 3.7 (1.7) mg/day during BID and at conversion, and 3.9 (1.8) mg/day at Week 12. Tacrolimus 10-20 BH3 interacting domain death agonist Homo sapiens 54-57 22239105-7 2012 Mean tacrolimus whole blood trough levels were at the lower end of the recommended range during tacrolimus BID and QD; the difference between mean steady-state trough levels was statistically significant (7.5ng/ml vs. 6.5ng/ml; P<0.0001). Tacrolimus 5-15 BH3 interacting domain death agonist Homo sapiens 107-110 22170067-5 2012 We report that IL-22 induction by TPA/A23187 (T/A) or alphaCD3 is inhibited by CsA or related FK506. Tacrolimus 94-99 interleukin 22 Homo sapiens 15-20 21680259-9 2012 We detected multiple reaction monitoring (MRM) transitions of m/z 1220>1203 and 1231.9>1215.1 for CsA and d12 CsA respectively which co-eluted at 1.30min, and 821.6>768.5 and 809.6>756.5 for tacrolimus and ascomycin respectively which co-eluted at 1.17 min. Tacrolimus 203-213 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 104-107 22309416-0 2012 Association between tacrolimus concentration and genetic polymorphisms of CYP3A5 and ABCB1 during the early stage after liver transplant in an Iranian population. Tacrolimus 20-30 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 74-80 22309416-0 2012 Association between tacrolimus concentration and genetic polymorphisms of CYP3A5 and ABCB1 during the early stage after liver transplant in an Iranian population. Tacrolimus 20-30 ATP binding cassette subfamily B member 1 Homo sapiens 85-90 22309416-2 2012 Tacrolimus is a substrate of cytochrome P-450 3A enzyme and the drug transporter, P-glycoprotein. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 82-96 21680259-9 2012 We detected multiple reaction monitoring (MRM) transitions of m/z 1220>1203 and 1231.9>1215.1 for CsA and d12 CsA respectively which co-eluted at 1.30min, and 821.6>768.5 and 809.6>756.5 for tacrolimus and ascomycin respectively which co-eluted at 1.17 min. Tacrolimus 203-213 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 116-119 22249344-12 2012 CONCLUSIONS: The results of this study provide evidence for safe conversion from Tacrolimus BID to QD with appropriate trough concentration monitoring. Tacrolimus 81-91 BH3 interacting domain death agonist Homo sapiens 92-95 22184128-3 2012 We investigated effects of two calcineurin inhibitors, cyclosporine A (CsA) and tacrolimus (FK506) (10-10-10-6 mol/L, 20 mins) on activation of MAP kinases and on growth, pro-fibrotic and pro-inflammatory responses in Ang II-stimulated rat cardiac fibroblasts. Tacrolimus 80-90 angiotensinogen Rattus norvegicus 218-224 22184128-5 2012 FK506, but not CsA, attenuated Ang II-stimulated MAP kinase activation. Tacrolimus 0-5 angiotensinogen Rattus norvegicus 31-37 22184128-7 2012 FK506 and CsA inhibited PCNA effects. Tacrolimus 0-5 proliferating cell nuclear antigen Rattus norvegicus 24-28 22456060-0 2012 Tacrolimus improves the proteinuria remission in patients with refractory IgA nephropathy. Tacrolimus 0-10 IGAN1 Homo sapiens 74-89 22184128-11 2012 p38MAP kinase and ERK1/2 are regulated in a FK506-sensitive manner, whereas fibrosis and inflammation are CsA-sensitive. Tacrolimus 44-49 mitogen activated protein kinase 14 Rattus norvegicus 0-3 22184128-11 2012 p38MAP kinase and ERK1/2 are regulated in a FK506-sensitive manner, whereas fibrosis and inflammation are CsA-sensitive. Tacrolimus 44-49 mitogen activated protein kinase 3 Rattus norvegicus 18-24 22158517-9 2012 Percent FoxP3+ cells in the CD4+CD25+ double positive population (T regulatory cells) in peripheral blood monocyte cells decreased in recipients with FK506 induction monotherapy while no reduction was observed in recipients treated with FK506 and HGF. Tacrolimus 150-155 forkhead box P3 Sus scrofa 8-13 22158517-9 2012 Percent FoxP3+ cells in the CD4+CD25+ double positive population (T regulatory cells) in peripheral blood monocyte cells decreased in recipients with FK506 induction monotherapy while no reduction was observed in recipients treated with FK506 and HGF. Tacrolimus 150-155 CD4 molecule Sus scrofa 28-31 22158517-9 2012 Percent FoxP3+ cells in the CD4+CD25+ double positive population (T regulatory cells) in peripheral blood monocyte cells decreased in recipients with FK506 induction monotherapy while no reduction was observed in recipients treated with FK506 and HGF. Tacrolimus 150-155 interleukin 2 receptor subunit alpha Sus scrofa 32-36 22158517-9 2012 Percent FoxP3+ cells in the CD4+CD25+ double positive population (T regulatory cells) in peripheral blood monocyte cells decreased in recipients with FK506 induction monotherapy while no reduction was observed in recipients treated with FK506 and HGF. Tacrolimus 150-155 hepatocyte growth factor Sus scrofa 247-250 22100870-0 2012 Cyclosporin A and tacrolimus induce renal Erk1/2 pathway via ROS-induced and metalloproteinase-dependent EGF-receptor signaling. Tacrolimus 18-28 mitogen activated protein kinase 3 Rattus norvegicus 42-48 22100870-1 2012 We previously demonstrated that the widely used immunosuppressive drugs cyclosporin A (CsA) and tacrolimus (FK506), independent of immunophilin binding, can activate profibrogenic transforming growth factor beta (TGFbeta)/Smad signaling cascades in rat renal mesangial cells (MC). Tacrolimus 96-106 transforming growth factor, beta 1 Rattus norvegicus 213-220 22100870-1 2012 We previously demonstrated that the widely used immunosuppressive drugs cyclosporin A (CsA) and tacrolimus (FK506), independent of immunophilin binding, can activate profibrogenic transforming growth factor beta (TGFbeta)/Smad signaling cascades in rat renal mesangial cells (MC). Tacrolimus 108-113 transforming growth factor, beta 1 Rattus norvegicus 213-220 22100870-8 2012 Our data suggest that CsA and FK506, via ROS-dependent and ADAM17-catalyzed HB-EGF shedding induce the mitogenic ERK1/2 signaling cascade in renal MC. Tacrolimus 30-35 ADAM metallopeptidase domain 17 Rattus norvegicus 59-65 22100870-8 2012 Our data suggest that CsA and FK506, via ROS-dependent and ADAM17-catalyzed HB-EGF shedding induce the mitogenic ERK1/2 signaling cascade in renal MC. Tacrolimus 30-35 heparin-binding EGF-like growth factor Rattus norvegicus 76-82 22100870-8 2012 Our data suggest that CsA and FK506, via ROS-dependent and ADAM17-catalyzed HB-EGF shedding induce the mitogenic ERK1/2 signaling cascade in renal MC. Tacrolimus 30-35 mitogen activated protein kinase 3 Rattus norvegicus 113-119 22008665-0 2012 The associations of IL-18 serum levels and promoter polymorphism with tacrolimus pharmacokinetics and hepatic allograft dysfunction in Chinese liver transplantation recipients. Tacrolimus 70-80 interleukin 18 Homo sapiens 20-25 22008665-9 2012 We found the recipients with higher IL-18 and IFN-gamma serum levels had lower tacrolimus concentration/dose (C/D) ratios (P<0.05). Tacrolimus 79-89 interleukin 18 Homo sapiens 36-41 22008665-9 2012 We found the recipients with higher IL-18 and IFN-gamma serum levels had lower tacrolimus concentration/dose (C/D) ratios (P<0.05). Tacrolimus 79-89 interferon gamma Homo sapiens 46-55 22008665-12 2012 This study identifies IL-18 reduced tacrolimus concentration/dose (C/D) ratio through up regulation of P-glycoprotein (P-gp). Tacrolimus 36-46 interleukin 18 Homo sapiens 22-27 22008665-12 2012 This study identifies IL-18 reduced tacrolimus concentration/dose (C/D) ratio through up regulation of P-glycoprotein (P-gp). Tacrolimus 36-46 ATP binding cassette subfamily B member 1 Homo sapiens 103-117 22008665-12 2012 This study identifies IL-18 reduced tacrolimus concentration/dose (C/D) ratio through up regulation of P-glycoprotein (P-gp). Tacrolimus 36-46 ATP binding cassette subfamily B member 1 Homo sapiens 119-123 22456060-3 2012 The efficacy and safety of tacrolimus in such refractory IgAN patients are extremely ambiguous, and the mechanism of tacrolimus improving proteinuria remission needs to be investigated. Tacrolimus 27-37 IGAN1 Homo sapiens 57-61 22456060-13 2012 CONCLUSIONS: Tacrolimus showed a rapid proteinuria remission in refractory IgAN patients. Tacrolimus 13-23 IGAN1 Homo sapiens 75-79 22100703-5 2012 Using NMR chemical shift mapping, we show that the important binding residues in FKBP12 are located in its hydrophobic FK506 binding region. Tacrolimus 119-124 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 81-87 22058159-9 2012 Atorvastatin downregulated beta-catenin, Erk1 and Erk2, and cyclin B in tacrolimus-stimulated VSMC. Tacrolimus 72-82 catenin beta 1 Homo sapiens 27-39 22058159-14 2012 In contrast, atorvastatin acts in concert with tacrolimus in HUVEC to stimulate production of endoglin, a factor that has an important role in endothelial repair. Tacrolimus 47-57 endoglin Homo sapiens 94-102 22100703-6 2012 Consistently, we demonstrate that FK506 can competitively inhibit the interaction between FKBP12 and the dipeptide motifs of the calcium channels. Tacrolimus 34-39 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 90-96 22132896-13 2012 Finally, inflammatory cytokines, such as tumour necrosis factor (TNF)-alpha and interleukin (IL)-6, showed lower levels in the graft of those animals that had been pretreated with rapamycin or tacrolimus. Tacrolimus 193-203 tumor necrosis factor Homo sapiens 41-75 22132896-13 2012 Finally, inflammatory cytokines, such as tumour necrosis factor (TNF)-alpha and interleukin (IL)-6, showed lower levels in the graft of those animals that had been pretreated with rapamycin or tacrolimus. Tacrolimus 193-203 interleukin 6 Homo sapiens 80-98 22108237-2 2012 Previous studies have established that CYP3A5 rs776746 is associated with tacrolimus clearance, blood concentration, and dose requirement. Tacrolimus 74-84 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 39-45 23269082-0 2012 Epstein-Barr virus-negative, CD5-positive diffuse large B-cell lymphoma developing after treatment with oral tacrolimus for mixed connective tissue disease : a case report and review of the literature. Tacrolimus 109-119 CD5 molecule Homo sapiens 29-32 22576566-3 2012 Recently, new immunomodulators and anti-TNFa agents, such as tacrolimus, infliximab, and adalimumab have been developed and these treatments are available to be treated for patients with refractory UC and CD. Tacrolimus 61-71 tumor necrosis factor Homo sapiens 40-44 22108237-9 2012 RESULTS: In analyses, which adjusted for race and other clinical factors, we replicated the association of tacrolimus blood concentration to dose ratio with CYP3A5 rs776746 (P=7.15x10), and identified associations with nine variants in linkage disequilibrium with rs776746, including eight CYP3A4 variants. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 157-163 22108237-9 2012 RESULTS: In analyses, which adjusted for race and other clinical factors, we replicated the association of tacrolimus blood concentration to dose ratio with CYP3A5 rs776746 (P=7.15x10), and identified associations with nine variants in linkage disequilibrium with rs776746, including eight CYP3A4 variants. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 290-296 23251520-7 2012 We tracked the subcellular localization of Crz1-GFP in WT C. neoformans and Deltacna1 in response to different stimuli, in the presence and absence of the calcineurin inhibitor, FK506. Tacrolimus 178-183 DNA-binding transcription factor CRZ1 Saccharomyces cerevisiae S288C 43-47 22848688-6 2012 In vitro, tacrolimus suppressed Th1 and Th2 cells in a concentration-dependent manner, but did not suppress Th17 cells even at high concentration. Tacrolimus 10-20 negative elongation factor complex member C/D Homo sapiens 32-35 22479554-5 2012 Treatment of bone marrow derived macrophages (BMDMs) with tacrolimus or 11R-VIVIT significantly inhibited LPS and LPS plus IFN-gamma induced IL-12 p40 mRNA and protein expression. Tacrolimus 58-68 interleukin 12b Mus musculus 141-150 22019501-13 2012 While tacrolimus and antithymocyte globulin additionally induced endothelial nitric oxide synthase and inducible nitric oxide synthase expression, cyclosporine A influenced only endothelial inducible nitric oxide synthase expression. Tacrolimus 6-16 nitric oxide synthase 3, endothelial cell Mus musculus 65-98 23196601-7 2012 These results indicate the contribution of anti-ryanodine receptor (RyR) antibody to E-C coupling impairment, and the early effect of tacrolimus as a pharmacological enhancement of RyR function to improve E-C coupling in MG. Tacrolimus 134-144 ryanodine receptor 2 Homo sapiens 181-184 21941688-0 2012 Effects of FK506 on Hippocampal CA1 Cells Following Transient Global Ischemia/Reperfusion in Wistar Rat. Tacrolimus 11-16 carbonic anhydrase 1 Rattus norvegicus 32-35 21941688-7 2012 The cell number and size of the CA1 pyramidal cells were increased, also the number of cell death decreased in this region when FK506 was administrated 48 h after reperfusion. Tacrolimus 128-133 carbonic anhydrase 1 Rattus norvegicus 32-35 22310591-0 2012 Impact of cytochrome P450 3A and ATP-binding cassette subfamily B member 1 polymorphisms on tacrolimus dose-adjusted trough concentrations among Korean renal transplant recipients. Tacrolimus 92-102 ATP binding cassette subfamily B member 1 Homo sapiens 33-74 21992068-10 2012 Tacrolimus BID to tacrolimus QD conversion is straightforward and does not compromise renal function in stable kidney transplant patients in the short term. Tacrolimus 0-10 BH3 interacting domain death agonist Homo sapiens 11-14 22310591-1 2012 BACKGROUND: Tacrolimus is a substrate of cytochrome P450 3A (CYP3A) and P-glycoprotein (P-gp), encoded by the CYP3A and ATP-binding cassette subfamily B member 1 (ABCB1) genes, respectively. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-59 22310591-1 2012 BACKGROUND: Tacrolimus is a substrate of cytochrome P450 3A (CYP3A) and P-glycoprotein (P-gp), encoded by the CYP3A and ATP-binding cassette subfamily B member 1 (ABCB1) genes, respectively. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 61-66 22310591-1 2012 BACKGROUND: Tacrolimus is a substrate of cytochrome P450 3A (CYP3A) and P-glycoprotein (P-gp), encoded by the CYP3A and ATP-binding cassette subfamily B member 1 (ABCB1) genes, respectively. Tacrolimus 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 72-86 22310591-1 2012 BACKGROUND: Tacrolimus is a substrate of cytochrome P450 3A (CYP3A) and P-glycoprotein (P-gp), encoded by the CYP3A and ATP-binding cassette subfamily B member 1 (ABCB1) genes, respectively. Tacrolimus 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 88-92 22310591-1 2012 BACKGROUND: Tacrolimus is a substrate of cytochrome P450 3A (CYP3A) and P-glycoprotein (P-gp), encoded by the CYP3A and ATP-binding cassette subfamily B member 1 (ABCB1) genes, respectively. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 110-115 22310591-1 2012 BACKGROUND: Tacrolimus is a substrate of cytochrome P450 3A (CYP3A) and P-glycoprotein (P-gp), encoded by the CYP3A and ATP-binding cassette subfamily B member 1 (ABCB1) genes, respectively. Tacrolimus 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 120-161 22310591-1 2012 BACKGROUND: Tacrolimus is a substrate of cytochrome P450 3A (CYP3A) and P-glycoprotein (P-gp), encoded by the CYP3A and ATP-binding cassette subfamily B member 1 (ABCB1) genes, respectively. Tacrolimus 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 163-168 22310591-2 2012 This study was aimed to investigate the impact of CYP3A and ABCB1 polymorphisms on the tacrolimus pharmacokinetics and clinical outcomes in Korean renal transplant recipients. Tacrolimus 87-97 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-55 22310591-2 2012 This study was aimed to investigate the impact of CYP3A and ABCB1 polymorphisms on the tacrolimus pharmacokinetics and clinical outcomes in Korean renal transplant recipients. Tacrolimus 87-97 ATP binding cassette subfamily B member 1 Homo sapiens 60-65 22310591-5 2012 RESULTS: Patients with the CYP3A5*3 alleles showed higher dose-adjusted tacrolimus concentrations for 12 months and higher trough levels until 6 months after transplantation. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 27-33 22310591-7 2012 In a multivariate analysis, the presence of >=1 CYP3A5*3 allele was a significant independent variable affecting dose-adjusted tacrolimus concentrations. Tacrolimus 130-140 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 51-57 22310591-9 2012 Calcineurin inhibitor toxicity, which showed higher tendency in patients with CYP3A5*1 alleles, might be associated with higher tacrolimus dose per kilogram. Tacrolimus 128-138 calcineurin binding protein 1 Homo sapiens 0-21 23023417-8 2012 Our experimental results demonstrate that preoperative assessment of cytochrome P450 3A5 (CYP3A5) genotypes in both recipients and donors and an immune cell function assay would be useful not only for predicting tacrolimus pharmacokinetics but also for defining groups at high-risk of infectious complications after LDLT. Tacrolimus 212-222 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 69-88 22310591-9 2012 Calcineurin inhibitor toxicity, which showed higher tendency in patients with CYP3A5*1 alleles, might be associated with higher tacrolimus dose per kilogram. Tacrolimus 128-138 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 78-84 23023417-8 2012 Our experimental results demonstrate that preoperative assessment of cytochrome P450 3A5 (CYP3A5) genotypes in both recipients and donors and an immune cell function assay would be useful not only for predicting tacrolimus pharmacokinetics but also for defining groups at high-risk of infectious complications after LDLT. Tacrolimus 212-222 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 90-96 22310591-10 2012 CONCLUSIONS: The CYP3A5 genotype is a major factor in determining the dose requirement of tacrolimus, and genotyping may be of value in individualization of immunosuppressive therapy of renal transplant patients. Tacrolimus 90-100 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 17-23 22310593-3 2012 The main aim of this study was to evaluate the effect of conversion from twice-daily tacrolimus (tacrolimus-BID) to once-daily tacrolimus (tacrolimus-OD) on glucose intolerance in stable kidney transplant patients. Tacrolimus 85-95 BH3 interacting domain death agonist Homo sapiens 108-111 22310593-3 2012 The main aim of this study was to evaluate the effect of conversion from twice-daily tacrolimus (tacrolimus-BID) to once-daily tacrolimus (tacrolimus-OD) on glucose intolerance in stable kidney transplant patients. Tacrolimus 97-107 BH3 interacting domain death agonist Homo sapiens 108-111 22310593-3 2012 The main aim of this study was to evaluate the effect of conversion from twice-daily tacrolimus (tacrolimus-BID) to once-daily tacrolimus (tacrolimus-OD) on glucose intolerance in stable kidney transplant patients. Tacrolimus 97-107 BH3 interacting domain death agonist Homo sapiens 108-111 22310593-3 2012 The main aim of this study was to evaluate the effect of conversion from twice-daily tacrolimus (tacrolimus-BID) to once-daily tacrolimus (tacrolimus-OD) on glucose intolerance in stable kidney transplant patients. Tacrolimus 97-107 BH3 interacting domain death agonist Homo sapiens 108-111 22310593-13 2012 CONCLUSION: The results of this study suggests that conversion from tacrolimus-BID to tacrolimus-OD may benefit kidney transplant patients with glucose intolerance because of improved insulin secretion. Tacrolimus 68-78 BH3 interacting domain death agonist Homo sapiens 79-82 22310596-6 2012 CONCLUSIONS: This study demonstrated a significant reduction in tacrolimus trough levels after switching from Tac BID to Tac OD, which increased insulin secretion and decreased HbA1c, suggesting that it may decrease the frequency of PTDM among stable renal transplant recipients. Tacrolimus 64-74 insulin Homo sapiens 145-152 21922127-0 2011 Influence of CYP3A5 and ABCB1 gene polymorphisms and other factors on tacrolimus dosing in Caucasian liver and kidney transplant patients. Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 21698374-11 2011 CONCLUSION: CYP3A5 genotype may explain variation in tacrolimus disposition early after transplant in pediatric kidney recipients, independent of age-related variation. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 21698374-12 2011 In contrast, in pediatric liver recipients, variation in tacrolimus disposition appears related to age and ABCB1 genotype. Tacrolimus 57-67 ATP binding cassette subfamily B member 1 Homo sapiens 107-112 22028295-6 2011 The number of JLTs required to detect the influence of CYP3A5 genotype and sex were estimated to be about 50 and > 600, respectively, which was consistent with the results of previous population pharmacokinetic studies for tacrolimus. Tacrolimus 226-236 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 55-61 21922127-0 2011 Influence of CYP3A5 and ABCB1 gene polymorphisms and other factors on tacrolimus dosing in Caucasian liver and kidney transplant patients. Tacrolimus 82-92 ATP binding cassette subfamily B member 1 Homo sapiens 24-29 21922127-1 2011 Tacrolimus is a substrate of cytochrome P4503A (CYP3A) enzymes as well as of the drug transporter ABCB1. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 98-103 21922127-9 2011 In the case of liver transplant patients the tacrolimus dose requirements were dominantly influenced by the polymorphisms of the CYP3A5 gene in the donors. Tacrolimus 45-55 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 129-135 21922127-11 2011 Identification of CYP3A5 single nucleotide polymorphisms prior to transplantation could contribute to evaluate the appropriate initial dosage of tacrolimus in the patients. Tacrolimus 145-155 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 18-24 22399088-10 2011 Conversely, LPO content decreased by 18.97% and 24.06% and MPO level by 42.86% and 18.66% after FK-506 and CsA treatment. Tacrolimus 96-102 myeloperoxidase Rattus norvegicus 59-62 21930396-0 2011 Age and CYP3A5 genotype affect tacrolimus dosing requirements after transplant in pediatric heart recipients. Tacrolimus 31-41 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 8-14 21930396-7 2011 CYP3A5 expressors required median (interquartile range) higher doses of tacrolimus (0.14 [0.09] vs 0.06 [0.04] mg/kg/12 hours, p = 0.001), and had lower concentration/dose ratios (45.34 [44.54] vs 177.78 [145.38] ng/ml per mg/kg/12 hours, p < 0.0001). Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 21930396-9 2011 Age and CYP3A5 genotype predicted the tacrolimus dosing requirements as well as the concentration/dose ratio (R(2) = 0.351, p = 0.001 and R(2) = 0.521, p < 0.001). Tacrolimus 38-48 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 8-14 21930396-11 2011 CONCLUSION: Younger age and CYP3A5 expressor genotype were independently associated with higher dosing requirements and lower tacrolimus concentration/dose ratios. Tacrolimus 126-136 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 28-34 21995851-0 2011 FK506 neuroprotection after cavernous nerve injury is mediated by thioredoxin and glutathione redox systems. Tacrolimus 0-5 thioredoxin 1 Rattus norvegicus 66-77 21917492-6 2011 We chose FK506-binding protein 51(FKBP5) (6p21.31) for our clinical target because of its role in modulating pharmacological responses to physiological and synthetic glucocorticoids and because of the complexity of the genomic region. Tacrolimus 9-14 FKBP prolyl isomerase 5 Homo sapiens 34-39 22090023-11 2011 The presence of CD23 with EBV in the presence of FK506, a T cell immunosuppressant. Tacrolimus 49-54 Fc epsilon receptor II Homo sapiens 16-20 21903774-0 2011 A new functional CYP3A4 intron 6 polymorphism significantly affects tacrolimus pharmacokinetics in kidney transplant recipients. Tacrolimus 68-78 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 17-23 21824470-4 2011 Using a microarray with 4504 human cDNAs and quantitative RT-PCR, two genes as apoptotic markers, transmembrane protein 100 (TMEM100) and limb-bud and heart (LBH), were identified because the thapsigargin-induced elevations in their mRNA levels were reversed by both FK506 and diclofenac. Tacrolimus 267-272 transmembrane protein 100 Homo sapiens 98-123 21940781-8 2011 Calcineurin inhibitors (FK506, cyclosporin A, and a peptide calcineurin inhibitor [CAIN]) abolished glucose-induced IRS-2 mRNA and protein levels, whereas expression of a constitutively active calcineurin increased them. Tacrolimus 24-29 insulin receptor substrate 2 Rattus norvegicus 116-121 21824470-8 2011 In conclusion, a novel neuroprotecitve compound, AS1219164, was discovered by high-throughput chemical screening using a reporter assay with the TMEM100 gene promoter regulated by both FK506 and diclofenac. Tacrolimus 185-190 transmembrane protein 100 Homo sapiens 145-152 21824470-4 2011 Using a microarray with 4504 human cDNAs and quantitative RT-PCR, two genes as apoptotic markers, transmembrane protein 100 (TMEM100) and limb-bud and heart (LBH), were identified because the thapsigargin-induced elevations in their mRNA levels were reversed by both FK506 and diclofenac. Tacrolimus 267-272 transmembrane protein 100 Homo sapiens 125-132 21824470-4 2011 Using a microarray with 4504 human cDNAs and quantitative RT-PCR, two genes as apoptotic markers, transmembrane protein 100 (TMEM100) and limb-bud and heart (LBH), were identified because the thapsigargin-induced elevations in their mRNA levels were reversed by both FK506 and diclofenac. Tacrolimus 267-272 LBH regulator of WNT signaling pathway Homo sapiens 138-156 21824470-4 2011 Using a microarray with 4504 human cDNAs and quantitative RT-PCR, two genes as apoptotic markers, transmembrane protein 100 (TMEM100) and limb-bud and heart (LBH), were identified because the thapsigargin-induced elevations in their mRNA levels were reversed by both FK506 and diclofenac. Tacrolimus 267-272 LBH regulator of WNT signaling pathway Homo sapiens 158-161 22210422-0 2011 Impact of CYP3A5 genotype of recipients as well as donors on the tacrolimus pharmacokinetics and infectious complications after living-donor liver transplantation for Japanese adult recipients. Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 21471328-1 2011 BACKGROUND: New-onset diabetes after transplantation may be associated with the use of tacrolimus (Tac) causing impaired insulin release or reduced insulin sensitivity. Tacrolimus 87-97 insulin Homo sapiens 121-128 21886016-0 2011 Lower tacrolimus daily dose requirements and acute rejection rates in the CYP3A5 nonexpressers than expressers. Tacrolimus 6-16 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 74-80 21886016-1 2011 BACKGROUND: CYP3A5 genetic polymorphisms contribute to marked interindividual differences in the metabolism of and response to tacrolimus in humans. Tacrolimus 127-137 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 21886016-2 2011 OBJECTIVE: This study was aimed to clarify the impact of the CYP3A5*3 variant on tacrolimus dose requirements and acute rejection rates in patients with organ transplantation. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-67 21886016-6 2011 Results of meta-analysis demonstrated that, in renal transplant patients, despite the presence of significant heterogeneity, CYP3A5 expressers required higher mean tacrolimus daily doses by 0.045 mg/kg (95% confidence interval (CI), 0.033-0.056) than nonexpressers. Tacrolimus 164-174 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 125-131 21886016-8 2011 Subset analysis of the ethnicity of organ recipients indicated that mean tacrolimus daily doses were 0.056, 0.037, and 0.077 mg/kg higher in CYP3A5 expressers than non- expressers for white, Chinese, and Japanese patients, respectively. Tacrolimus 73-83 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 141-147 21886016-9 2011 In contrast, for liver transplant patients, higher tacrolimus daily doses were required not only in CYP3A5 expressers of the organ donors than nonexpressers by 0.024 mg/kg (95% CI, 0.019-0.028), but also in CYP3A5 expresser of the organ recipients than nonexpresser by 0.012 mg/kg (95% CI, 0.005-0.018). Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 100-106 21886016-9 2011 In contrast, for liver transplant patients, higher tacrolimus daily doses were required not only in CYP3A5 expressers of the organ donors than nonexpressers by 0.024 mg/kg (95% CI, 0.019-0.028), but also in CYP3A5 expresser of the organ recipients than nonexpresser by 0.012 mg/kg (95% CI, 0.005-0.018). Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 207-213 21886016-11 2011 CONCLUSION: Tacrolimus daily dose requirements may vary with the presence of the CYP3A5*3 variant, ethnicity of the organ recipients, and the time of posttransplantation. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 81-87 21692771-0 2011 A topical combination of rapamycin and tacrolimus for the treatment of angiofibroma due to tuberous sclerosis complex (TSC): a pilot study of nine Japanese patients with TSC of different disease severity. Tacrolimus 39-49 TSC complex subunit 1 Homo sapiens 119-122 21692771-5 2011 OBJECTIVES: The efficacy of rapamycin-tacrolimus ointment was examined for TSC-related angiofibroma. Tacrolimus 38-48 TSC complex subunit 1 Homo sapiens 75-78 21692771-11 2011 CONCLUSIONS: Topical application of rapamycin-tacrolimus ointment is a safe and useful treatment for TSC-related angiofibroma. Tacrolimus 46-56 TSC complex subunit 1 Homo sapiens 101-104 21615281-3 2011 In order to inhibit the efflux of P-glycoprotein (P-gp) for tacrolimus, which is the substrate of P-gp, the excipients which show the inhibition effect to P-gp, such as tocopheryl polyethylene glycol succinate (TPGS) and Cremophor EL40, were chosen in the SMEDDS formulations. Tacrolimus 60-70 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 34-48 21615281-3 2011 In order to inhibit the efflux of P-glycoprotein (P-gp) for tacrolimus, which is the substrate of P-gp, the excipients which show the inhibition effect to P-gp, such as tocopheryl polyethylene glycol succinate (TPGS) and Cremophor EL40, were chosen in the SMEDDS formulations. Tacrolimus 60-70 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 50-54 21615281-3 2011 In order to inhibit the efflux of P-glycoprotein (P-gp) for tacrolimus, which is the substrate of P-gp, the excipients which show the inhibition effect to P-gp, such as tocopheryl polyethylene glycol succinate (TPGS) and Cremophor EL40, were chosen in the SMEDDS formulations. Tacrolimus 60-70 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 98-102 21615281-3 2011 In order to inhibit the efflux of P-glycoprotein (P-gp) for tacrolimus, which is the substrate of P-gp, the excipients which show the inhibition effect to P-gp, such as tocopheryl polyethylene glycol succinate (TPGS) and Cremophor EL40, were chosen in the SMEDDS formulations. Tacrolimus 60-70 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 98-102 21640200-8 2011 The expression of 1alpha (IV) collagen, p65, p-p65, OPN, alpha-SMA and TGF-beta1 protein in kidney was significantly increased in diabetic rats and reduced by FK506 treatment (p<0.05, 0.01). Tacrolimus 159-164 synaptotagmin 1 Rattus norvegicus 40-43 21640200-8 2011 The expression of 1alpha (IV) collagen, p65, p-p65, OPN, alpha-SMA and TGF-beta1 protein in kidney was significantly increased in diabetic rats and reduced by FK506 treatment (p<0.05, 0.01). Tacrolimus 159-164 synaptotagmin 1 Rattus norvegicus 47-50 21640200-8 2011 The expression of 1alpha (IV) collagen, p65, p-p65, OPN, alpha-SMA and TGF-beta1 protein in kidney was significantly increased in diabetic rats and reduced by FK506 treatment (p<0.05, 0.01). Tacrolimus 159-164 transforming growth factor, beta 1 Rattus norvegicus 71-80 21916909-0 2011 Association of ABCB1, CYP3A4*18B and CYP3A5*3 genotypes with the pharmacokinetics of tacrolimus in healthy Chinese subjects: a population pharmacokinetic analysis. Tacrolimus 85-95 ATP binding cassette subfamily B member 1 Homo sapiens 15-20 21916909-0 2011 Association of ABCB1, CYP3A4*18B and CYP3A5*3 genotypes with the pharmacokinetics of tacrolimus in healthy Chinese subjects: a population pharmacokinetic analysis. Tacrolimus 85-95 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 22-28 21916909-0 2011 Association of ABCB1, CYP3A4*18B and CYP3A5*3 genotypes with the pharmacokinetics of tacrolimus in healthy Chinese subjects: a population pharmacokinetic analysis. Tacrolimus 85-95 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 21916909-1 2011 WHAT IS KNOWN AND OBJECTIVE: Tacrolimus (TAC) is metabolized mainly by the CYP3A subfamily and extruded into the intestine by P-glycoprotein, which is encoded by the ABCB1 gene. Tacrolimus 29-39 ATP binding cassette subfamily B member 1 Homo sapiens 166-171 21830095-0 2011 Preventive and therapeutic effects of tacrolimus in an interleukin-10-deficient mouse model of colitis. Tacrolimus 38-48 interleukin 10 Mus musculus 55-69 21830095-2 2011 METHODS: Tacrolimus or prednisolone, an anti-inflammatory glucocorticoid, was administered to IL-10(-/-) mice with pre- or post-symptomatic colitis. Tacrolimus 9-19 interleukin 10 Mus musculus 94-99 21830095-7 2011 Tacrolimus completely inhibited IFN-gamma and TNF-alpha production of activated T-cells in LPMCs, but only partially inhibited IFN-gamma, TNF-alpha, and IL-12 production of activated monocytes/macrophages in LPMCs. Tacrolimus 0-10 interferon gamma Mus musculus 32-41 21830095-7 2011 Tacrolimus completely inhibited IFN-gamma and TNF-alpha production of activated T-cells in LPMCs, but only partially inhibited IFN-gamma, TNF-alpha, and IL-12 production of activated monocytes/macrophages in LPMCs. Tacrolimus 0-10 tumor necrosis factor Mus musculus 46-55 21830095-7 2011 Tacrolimus completely inhibited IFN-gamma and TNF-alpha production of activated T-cells in LPMCs, but only partially inhibited IFN-gamma, TNF-alpha, and IL-12 production of activated monocytes/macrophages in LPMCs. Tacrolimus 0-10 interferon gamma Mus musculus 127-136 21830095-7 2011 Tacrolimus completely inhibited IFN-gamma and TNF-alpha production of activated T-cells in LPMCs, but only partially inhibited IFN-gamma, TNF-alpha, and IL-12 production of activated monocytes/macrophages in LPMCs. Tacrolimus 0-10 tumor necrosis factor Mus musculus 138-147 21830095-9 2011 CONCLUSION: These results suggest that the preventive and therapeutic effect of tacrolimus in IL-10(-/-) mice colitis might be attributed to the inhibition of colonic T-cell activation rather than monocyte/macrophage activation. Tacrolimus 80-90 interleukin 10 Mus musculus 94-99 22099811-4 2011 Since studies have demonstrated that single nucleotide polymorphisms (SNPs) of ATF6 are associated with type 2 diabetes, we sought to determine whether their mutations were associated with NODAT among renal transplant recipients treated with tacrolimus. Tacrolimus 242-252 activating transcription factor 6 Homo sapiens 79-83 22099811-13 2011 However, these data underscore the role of ATF6 and endoplasmic reticulum stress in the regulation of metabolic flux among patients treated with tacrolimus, suggesting that inherited disturbances of endoplasmic reticulum stress signaling could predispose people to obesity. Tacrolimus 145-155 activating transcription factor 6 Homo sapiens 43-47 22016125-8 2011 Calcineurin inhibitors (cyclosporine, tacrolimus) and mTOR inhibitors (sirolimus, everolimus) are particularly susceptible to the effects of substances that inhibit or induce cytochrome P450 (CYP) 3A4 and P-glycoprotein. Tacrolimus 38-48 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 175-200 22016125-8 2011 Calcineurin inhibitors (cyclosporine, tacrolimus) and mTOR inhibitors (sirolimus, everolimus) are particularly susceptible to the effects of substances that inhibit or induce cytochrome P450 (CYP) 3A4 and P-glycoprotein. Tacrolimus 38-48 ATP binding cassette subfamily B member 1 Homo sapiens 205-219 21963515-0 2011 The calcineurin inhibitor tacrolimus activates the renal sodium chloride cotransporter to cause hypertension. Tacrolimus 26-36 calcineurin binding protein 1 Mus musculus 4-25 21963515-5 2011 In wild-type mice, the CNI tacrolimus caused salt-sensitive hypertension and increased the abundance of phosphorylated NCC and the NCC-regulatory kinases WNK3, WNK4 and SPAK. Tacrolimus 27-37 WNK lysine deficient protein kinase 3 Mus musculus 154-158 21963515-5 2011 In wild-type mice, the CNI tacrolimus caused salt-sensitive hypertension and increased the abundance of phosphorylated NCC and the NCC-regulatory kinases WNK3, WNK4 and SPAK. Tacrolimus 27-37 WNK lysine deficient protein kinase 4 Mus musculus 160-164 21963515-5 2011 In wild-type mice, the CNI tacrolimus caused salt-sensitive hypertension and increased the abundance of phosphorylated NCC and the NCC-regulatory kinases WNK3, WNK4 and SPAK. Tacrolimus 27-37 serine/threonine kinase 39 Mus musculus 169-173 22210422-1 2011 BACKGROUND: The impact of cytochrome P450 3A5 (CYP3A5) genotype of recipients (intestine) as well as donors (graft liver) on the tacrolimus pharmacokinetics and the incidence of infectious complications was assessed in Japanese living-donor liver transplant (LDLT) adult recipients. Tacrolimus 129-139 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 26-45 22210422-1 2011 BACKGROUND: The impact of cytochrome P450 3A5 (CYP3A5) genotype of recipients (intestine) as well as donors (graft liver) on the tacrolimus pharmacokinetics and the incidence of infectious complications was assessed in Japanese living-donor liver transplant (LDLT) adult recipients. Tacrolimus 129-139 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 47-53 22210422-10 2011 CONCLUSIONS: Preoperative assessment of CYP3A5 genotypes in both recipients and donors would be useful not only for predicting tacrolimus pharmacokinetics but also defining high-risk group of infectious complications after LDLT. Tacrolimus 127-137 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 40-46 22004713-10 2011 In addition, FK506 markedly reduced microglial activation (P<0 01) and TNF-alpha expression (P<0 01). Tacrolimus 13-18 tumor necrosis factor Rattus norvegicus 75-84 21703318-3 2011 In the present study, we show the protective effects of FK506 in two striatal HD models, primary rat striatal neurons treated with 3-nitropropionic acid (3-NP) and immortalized striatal STHdh cells derived from HD knock-in mice expressing normal (STHdh(7/7)) or full-length mutant huntingtin (FL-mHtt) with 111 glutamines (STHdh(111/111)), under basal conditions and after exposure to 3-NP or staurosporine (STS). Tacrolimus 56-61 huntingtin Mus musculus 281-291 21773745-0 2011 Different effects of CsA and FK506 on aquaporin-2 abundance in rat primary cultured collecting duct cells. Tacrolimus 29-34 aquaporin 2 Rattus norvegicus 38-49 21703318-4 2011 In rat striatal neurons, FK506 abolished 3-NP-induced increase in caspase-3 activation, DNA fragmentation/condensation and necrosis. Tacrolimus 25-30 caspase 3 Rattus norvegicus 66-75 21902502-1 2011 AIMS: CYP3A4 is involved in the oxidative metabolism of many drugs and xenobiotics including the immunosuppressants tacrolimus (Tac) and cyclosporine (CsA). Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 6-12 21902502-1 2011 AIMS: CYP3A4 is involved in the oxidative metabolism of many drugs and xenobiotics including the immunosuppressants tacrolimus (Tac) and cyclosporine (CsA). Tacrolimus 128-131 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 6-12 21753749-1 2011 In vitro studies have identified cyclosporine and tacrolimus as CYP3A inhibitors. Tacrolimus 50-60 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 64-69 21677149-7 2011 Addition of AP20187 (an analog of FK506) enhanced the homodimerization of Fv-SLK 1-373. Tacrolimus 34-39 STE20 like kinase Homo sapiens 77-80 21640771-5 2011 After administration, FK506 and CsA mainly distribute within erythrocytes owing to the presence into these cells of immunophilins that bind the drugs with very high affinity (FKBP12 for FK506 and cyclophilin A for CsA); therefore, a new strategy aimed to increase the amount of FK506/CsA carried by erythrocytes by increasing the intra-erythrocytic concentration of the respective binding proteins has been developed. Tacrolimus 22-27 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 175-181 21640771-5 2011 After administration, FK506 and CsA mainly distribute within erythrocytes owing to the presence into these cells of immunophilins that bind the drugs with very high affinity (FKBP12 for FK506 and cyclophilin A for CsA); therefore, a new strategy aimed to increase the amount of FK506/CsA carried by erythrocytes by increasing the intra-erythrocytic concentration of the respective binding proteins has been developed. Tacrolimus 186-191 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 175-181 21640771-5 2011 After administration, FK506 and CsA mainly distribute within erythrocytes owing to the presence into these cells of immunophilins that bind the drugs with very high affinity (FKBP12 for FK506 and cyclophilin A for CsA); therefore, a new strategy aimed to increase the amount of FK506/CsA carried by erythrocytes by increasing the intra-erythrocytic concentration of the respective binding proteins has been developed. Tacrolimus 186-191 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 175-181 21640771-9 2011 Moreover, FK506 released from FKBP12-RBC is able to be up-taken by T lymphocytes and inhibit IL-2 expression in vitro as free administered drug. Tacrolimus 10-15 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 30-36 21640771-9 2011 Moreover, FK506 released from FKBP12-RBC is able to be up-taken by T lymphocytes and inhibit IL-2 expression in vitro as free administered drug. Tacrolimus 10-15 RNA, 7SL, cytoplasmic 263, pseudogene Homo sapiens 37-40 21640771-9 2011 Moreover, FK506 released from FKBP12-RBC is able to be up-taken by T lymphocytes and inhibit IL-2 expression in vitro as free administered drug. Tacrolimus 10-15 interleukin 2 Homo sapiens 93-97 22141281-5 2011 TREC contents within double-positive cells and CD4+ single-positive cells were significantly decreased 4 M after tacrolimus therapy (p < 0.05) in PM/DM patients. Tacrolimus 113-123 CD4 molecule Homo sapiens 47-50 22141281-6 2011 Tacrolimus treatment significantly attenuated TREC content within cultured CD4+CD8- cells from PM/DMpatients (p < 0.05), but total cell counts were not significantly changed. Tacrolimus 0-10 CD4 molecule Homo sapiens 75-78 22141281-6 2011 Tacrolimus treatment significantly attenuated TREC content within cultured CD4+CD8- cells from PM/DMpatients (p < 0.05), but total cell counts were not significantly changed. Tacrolimus 0-10 CD8a molecule Homo sapiens 79-82 21753749-0 2011 In vivo CYP3A activity is significantly lower in cyclosporine-treated as compared with tacrolimus-treated renal allograft recipients. Tacrolimus 87-97 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 8-13 21753749-5 2011 This implies that in vivo hepatic and first-pass CYP3A activities are significantly lower in patients receiving cyclosporine than in those receiving tacrolimus, indicating that, at the doses generally used in clinical practice, cyclosporine is the stronger of the two with respect to CYP3A inhibition. Tacrolimus 149-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 49-54 21725799-5 2011 The inhibitory potencies of known P-gp inhibitors viz verapamil, ketoconazole, tacrolimus and cyclosporine A, determined over a wide concentration range, showed low apparent IC(50) values. Tacrolimus 79-89 phosphoglycolate phosphatase Homo sapiens 34-38 21677300-0 2011 CYP3A5 and ABCB1 polymorphisms in donor and recipient: impact on Tacrolimus dose requirements and clinical outcome after renal transplantation. Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 21493098-10 2011 CONCLUSIONS: This study provides evidence for successful conversion from tacrolimus BID to QD on a 1:1 (mg/mg) total daily dose basis. Tacrolimus 73-83 BH3 interacting domain death agonist Homo sapiens 84-87 21493098-13 2011 Adverse events were reported by approximately one-tenth of patients receiving tacrolimus BID and a quarter of those who received QD. Tacrolimus 78-88 BH3 interacting domain death agonist Homo sapiens 89-92 21677300-0 2011 CYP3A5 and ABCB1 polymorphisms in donor and recipient: impact on Tacrolimus dose requirements and clinical outcome after renal transplantation. Tacrolimus 65-75 ATP binding cassette subfamily B member 1 Homo sapiens 11-16 21677300-1 2011 BACKGROUND: The effect of potentially relevant genetic polymorphisms, CYP3A5 6986A>G and ABCB1 3435C>T, on Tacrolimus pharmacokinetics and graft clinical outcome was investigated in donor and recipient DNA samples from 209 kidney transplant patients. Tacrolimus 113-123 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 70-76 21677300-1 2011 BACKGROUND: The effect of potentially relevant genetic polymorphisms, CYP3A5 6986A>G and ABCB1 3435C>T, on Tacrolimus pharmacokinetics and graft clinical outcome was investigated in donor and recipient DNA samples from 209 kidney transplant patients. Tacrolimus 113-123 ATP binding cassette subfamily B member 1 Homo sapiens 92-97 21677300-3 2011 The Tacrolimus dose, trough blood concentrations (C0) and C0/dose ratio were only statistically correlated with the recipient CYP3A5 genotype. Tacrolimus 4-14 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 126-132 21677300-8 2011 CONCLUSIONS/SIGNIFICANCE: Recipient CYP3A5 6986A>G polymorphism explains part of the interindividual variability of the pharmacokinetics of Tacrolimus. Tacrolimus 143-153 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 21806386-1 2011 AIM: Polymorphisms in the CYP3A5 and ABCB1 genes have been investigated as modulators of the pharmacokinetics and clinical effects of cyclosporine (CSA) and tacrolimus (TAC) in European, North American and Asian populations, with controversial results. Tacrolimus 169-172 ATP binding cassette subfamily B member 1 Homo sapiens 37-42 21672049-2 2011 Tacrolimus, a very potent and effective calcineurin inhibitor, was selected because of its ability to ameliorate I/R injury. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 40-61 21770725-0 2011 The P450 oxidoreductase *28 SNP is associated with low initial tacrolimus exposure and increased dose requirements in CYP3A5-expressing renal recipients. Tacrolimus 63-73 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 118-124 21770725-3 2011 MATERIALS & METHODS: To test this hypothesis we performed a study in a cohort of 298 de novo renal allograft recipients stratified according to CYP3A5 genotype, which has a known impact on tacrolimus pharmacokinetics. Tacrolimus 193-203 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 148-154 21770725-4 2011 RESULTS: We found that in CYP3A5 expressers (CYP3A5*1 allele carriers) receiving a standard loading dose of 0.2 mg/kg, POR*28T allele carriers had lower tacrolimus C0 levels in the first days post-transplantation and reached target C0 levels significantly later as compared with POR*28CC homozygous patients. Tacrolimus 153-163 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 21770725-4 2011 RESULTS: We found that in CYP3A5 expressers (CYP3A5*1 allele carriers) receiving a standard loading dose of 0.2 mg/kg, POR*28T allele carriers had lower tacrolimus C0 levels in the first days post-transplantation and reached target C0 levels significantly later as compared with POR*28CC homozygous patients. Tacrolimus 153-163 cytochrome p450 oxidoreductase Homo sapiens 119-122 21770725-5 2011 The POR*28T allele carriers had significantly higher tacrolimus dose requirements throughout the first year. Tacrolimus 53-63 cytochrome p450 oxidoreductase Homo sapiens 4-7 21770725-7 2011 CONCLUSION: These data indicate that the POR*28 SNP is associated with additional increases in early tacrolimus dose-requirements in patients carrying a CYP3A5*1 allele. Tacrolimus 101-111 cytochrome p450 oxidoreductase Homo sapiens 41-44 21770725-7 2011 CONCLUSION: These data indicate that the POR*28 SNP is associated with additional increases in early tacrolimus dose-requirements in patients carrying a CYP3A5*1 allele. Tacrolimus 101-111 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 153-159 21806386-1 2011 AIM: Polymorphisms in the CYP3A5 and ABCB1 genes have been investigated as modulators of the pharmacokinetics and clinical effects of cyclosporine (CSA) and tacrolimus (TAC) in European, North American and Asian populations, with controversial results. Tacrolimus 157-167 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 26-32 21806386-1 2011 AIM: Polymorphisms in the CYP3A5 and ABCB1 genes have been investigated as modulators of the pharmacokinetics and clinical effects of cyclosporine (CSA) and tacrolimus (TAC) in European, North American and Asian populations, with controversial results. Tacrolimus 157-167 ATP binding cassette subfamily B member 1 Homo sapiens 37-42 21806386-1 2011 AIM: Polymorphisms in the CYP3A5 and ABCB1 genes have been investigated as modulators of the pharmacokinetics and clinical effects of cyclosporine (CSA) and tacrolimus (TAC) in European, North American and Asian populations, with controversial results. Tacrolimus 169-172 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 26-32 21818055-5 2011 RESULTS: MPA and tacrolimus exhibited a comparable impact on T-cell response, dampening most Th1-related genes transcription and preserving regulatory T cells/Th2 molecular phenotypes. Tacrolimus 17-27 negative elongation factor complex member C/D Homo sapiens 93-96 21911153-1 2011 AIMS: The aims of this study were to study the role of histone deacetylase 11 (HDAC11) in tolerance induction in orthotopic liver transplantation (OLT) in rats and to assess the advantages of gene therapy over the immunosuppressant FK506. Tacrolimus 232-237 histone deacetylase 11 Rattus norvegicus 79-85 21911157-0 2011 Tacrolimus enhances the invasion potential of hepatocellular carcinoma cells and promotes lymphatic metastasis in a rat model of hepatocellular carcinoma: involvement of vascular endothelial growth factor-C. Tacrolimus 0-10 vascular endothelial growth factor C Rattus norvegicus 170-206 21911157-8 2011 qRT-PCR, immunohistochemisty, and Western blot revealed that tacrolimus increased the levels of expression of VEGF-C in HCC. Tacrolimus 61-71 vascular endothelial growth factor C Rattus norvegicus 110-116 21820512-9 2011 Reduced mRNA expression levels of CD4, IFN-gamma, IL-6, IL-10, iNOS, NFkappaB, TNF-alpha and MCP-1 (p <= 0.05) were observed in tacrolimus treated animals compared to sirolimus. Tacrolimus 131-141 Cd4 molecule Rattus norvegicus 34-37 21820512-9 2011 Reduced mRNA expression levels of CD4, IFN-gamma, IL-6, IL-10, iNOS, NFkappaB, TNF-alpha and MCP-1 (p <= 0.05) were observed in tacrolimus treated animals compared to sirolimus. Tacrolimus 131-141 interferon gamma Rattus norvegicus 39-48 21820512-9 2011 Reduced mRNA expression levels of CD4, IFN-gamma, IL-6, IL-10, iNOS, NFkappaB, TNF-alpha and MCP-1 (p <= 0.05) were observed in tacrolimus treated animals compared to sirolimus. Tacrolimus 131-141 interleukin 6 Rattus norvegicus 50-54 21820512-9 2011 Reduced mRNA expression levels of CD4, IFN-gamma, IL-6, IL-10, iNOS, NFkappaB, TNF-alpha and MCP-1 (p <= 0.05) were observed in tacrolimus treated animals compared to sirolimus. Tacrolimus 131-141 interleukin 10 Rattus norvegicus 56-61 21820512-9 2011 Reduced mRNA expression levels of CD4, IFN-gamma, IL-6, IL-10, iNOS, NFkappaB, TNF-alpha and MCP-1 (p <= 0.05) were observed in tacrolimus treated animals compared to sirolimus. Tacrolimus 131-141 nitric oxide synthase 2 Rattus norvegicus 63-67 21820512-9 2011 Reduced mRNA expression levels of CD4, IFN-gamma, IL-6, IL-10, iNOS, NFkappaB, TNF-alpha and MCP-1 (p <= 0.05) were observed in tacrolimus treated animals compared to sirolimus. Tacrolimus 131-141 tumor necrosis factor Rattus norvegicus 79-88 21820512-9 2011 Reduced mRNA expression levels of CD4, IFN-gamma, IL-6, IL-10, iNOS, NFkappaB, TNF-alpha and MCP-1 (p <= 0.05) were observed in tacrolimus treated animals compared to sirolimus. Tacrolimus 131-141 mast cell protease 1-like 1 Rattus norvegicus 93-98 21818055-7 2011 Accordingly, renal transplant patients treated with MPA in combination with minimized dose of tacrolimus tended to have lower circulating IL-17 levels than patients treated with tacrolimus alone given at conventional dose. Tacrolimus 94-104 interleukin 17A Homo sapiens 138-143 21102498-6 2011 The CYP3A5 rs776746 and rs4646450 genotypes were also associated with tacrolimus concentration (P=0.013 and P=0.0058, respectively). Tacrolimus 70-80 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 21102498-7 2011 The dosage of tacrolimus was remarkably reduced from day -1 to day 28 when the patient had the CYP3A5 rs4646450 C/C and/or rs776746 G/G genotype (P=0.0010 and P=0.0021, respectively). Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 95-101 21622531-4 2011 CN(+) similarly promoted podocyte apoptosis, and apoptosis induced by either angiotensin II or endothelin-1 was blocked by FK506. Tacrolimus 123-128 endothelin 1 Mus musculus 95-107 21514407-7 2011 Cyclosporin A and FK506, inhibitors of calcineurin phosphatase, blocked high [Ca(2+)](o)-induced expression of NFAT and RANKL. Tacrolimus 18-23 TNF superfamily member 11 Homo sapiens 120-125 21545550-9 2011 CONCLUSION AND CLINICAL RELEVANCE: Our results suggest that post-transplant immunosuppression with tacrolimus is associated with an increased occurrence of IgE-mediated sensitization and probably manifestation of allergic disease, which has to be treated specifically despite immunosuppressive therapy. Tacrolimus 99-109 immunoglobulin heavy constant epsilon Homo sapiens 156-159 21802143-0 2011 Hepatic drug interaction between tacrolimus and lansoprazole in a bone marrow transplant patient receiving voriconazole and harboring CYP2C19 and CYP3A5 heterozygous mutations. Tacrolimus 33-43 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 134-141 21802143-0 2011 Hepatic drug interaction between tacrolimus and lansoprazole in a bone marrow transplant patient receiving voriconazole and harboring CYP2C19 and CYP3A5 heterozygous mutations. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 146-152 21802143-1 2011 BACKGROUND: A drug interaction between oral tacrolimus (TAC) and lansoprazole (LAN) has been reported in patients with CYP2C19 hetero/homozygous mutations and the CYP3A5 *3/*3 genotype. Tacrolimus 44-54 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 119-126 21802143-1 2011 BACKGROUND: A drug interaction between oral tacrolimus (TAC) and lansoprazole (LAN) has been reported in patients with CYP2C19 hetero/homozygous mutations and the CYP3A5 *3/*3 genotype. Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 163-169 21331500-5 2011 RESULTS: CYP3A5 genotype, body weight, haematocrit, haemoglobin and total bilirubin significantly influenced the maintenance tacrolimus dose. Tacrolimus 125-135 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 9-15 21359536-0 2011 Impact of interleukin-10 gene polymorphisms on tacrolimus dosing requirements in Chinese liver transplant patients during the early posttransplantation period. Tacrolimus 47-57 interleukin 10 Homo sapiens 10-24 21359536-2 2011 Impacts of CYP3A5 and ABCB1 gene polymorphisms on the immunosuppressant tacrolimus have been reported in previous studies of liver transplantation. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 11-17 21359536-2 2011 Impacts of CYP3A5 and ABCB1 gene polymorphisms on the immunosuppressant tacrolimus have been reported in previous studies of liver transplantation. Tacrolimus 72-82 ATP binding cassette subfamily B member 1 Homo sapiens 22-27 21359536-8 2011 RESULTS: Recipients who received organs from CYP3A5*3/*3 donors had higher tacrolimus C/D ratios. Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 21359536-9 2011 In the first 2 weeks, the tacrolimus C/D ratios of the recipients with donors who were CYP3A5 nonexpressors and had a low IL-10 production genotype (-819TT, -592 AA) were higher than those with donors who were CYP3A5 nonexpressors and had a high IL-10 production genotype (-819CC or CT, -592CC or AC). Tacrolimus 26-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 87-93 21359536-11 2011 CONCLUSION: Determining IL-10 and CYP3A5 polymorphisms of donors may allow individualized tacrolimus dosage regimens to be determined for liver transplant patients during the early posttransplantation period. Tacrolimus 90-100 interleukin 10 Homo sapiens 24-29 21359536-11 2011 CONCLUSION: Determining IL-10 and CYP3A5 polymorphisms of donors may allow individualized tacrolimus dosage regimens to be determined for liver transplant patients during the early posttransplantation period. Tacrolimus 90-100 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 21799260-4 2011 Binding of the immunosuppressant molecule FK506 to this domain inhibits their PPIase activity while mediating immune suppression through inhibition of calcineurin. Tacrolimus 42-47 FKBP prolyl isomerase 4 Homo sapiens 78-84 21399973-1 2011 FK506 binding protein 65 (FKBP65) belongs to a group of proteins termed immunophilins that have a high binding affinity to immunosuppressant drugs as FK506 (tacrolimus) and rapamycin (sirolimus). Tacrolimus 0-5 FKBP prolyl isomerase 10 Homo sapiens 26-32 21523562-1 2011 The Hsp90-associated cis-trans peptidyl-prolyl isomerase--FK506 binding protein 51 (FKBP51)--was recently found to co-localize with the microtubule (MT)-associated protein tau in neurons and physically interact with tau in brain tissues from humans who died from Alzheimer"s disease (AD). Tacrolimus 58-63 heat shock protein 90 alpha family class A member 1 Homo sapiens 4-9 20514078-0 2011 Effects of diltiazem on pharmacokinetics of tacrolimus in relation to CYP3A5 genotype status in renal recipients: from retrospective to prospective. Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 70-76 20514078-2 2011 In retrospective study, when coadministered with diltiazem, mean increments in dose-adjusted C(0D7), C(max) and AUC(0-12 h) for tacrolimus were larger in CYP3A5 expressers than in CYP3A5 nonexpressers (48.7 vs 3.7%, 31.7 vs 17.2% and 38.2 vs 18.5%, respectively). Tacrolimus 128-138 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 154-160 20514078-2 2011 In retrospective study, when coadministered with diltiazem, mean increments in dose-adjusted C(0D7), C(max) and AUC(0-12 h) for tacrolimus were larger in CYP3A5 expressers than in CYP3A5 nonexpressers (48.7 vs 3.7%, 31.7 vs 17.2% and 38.2 vs 18.5%, respectively). Tacrolimus 128-138 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 180-186 20514078-4 2011 For CYP3A5 expressers, an algorithm guided by CYP3A5 and diltiazem significantly reduced tacrolimus maintenance dosage (P=0.009) and improved the accuracy of tacrolimus initial dose, resulting in reduction in out-of-range C(0) after initial dose (P=0.002) and dose adjustments (P=0.004). Tacrolimus 89-99 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 20514078-4 2011 For CYP3A5 expressers, an algorithm guided by CYP3A5 and diltiazem significantly reduced tacrolimus maintenance dosage (P=0.009) and improved the accuracy of tacrolimus initial dose, resulting in reduction in out-of-range C(0) after initial dose (P=0.002) and dose adjustments (P=0.004). Tacrolimus 89-99 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 20514078-4 2011 For CYP3A5 expressers, an algorithm guided by CYP3A5 and diltiazem significantly reduced tacrolimus maintenance dosage (P=0.009) and improved the accuracy of tacrolimus initial dose, resulting in reduction in out-of-range C(0) after initial dose (P=0.002) and dose adjustments (P=0.004). Tacrolimus 158-168 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 20514078-4 2011 For CYP3A5 expressers, an algorithm guided by CYP3A5 and diltiazem significantly reduced tacrolimus maintenance dosage (P=0.009) and improved the accuracy of tacrolimus initial dose, resulting in reduction in out-of-range C(0) after initial dose (P=0.002) and dose adjustments (P=0.004). Tacrolimus 158-168 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 20514078-6 2011 Our study results show that CYP3A5 genotype-guided tacrolimus-diltiazem combination is a promising therapy in renal transplant recipients in the early postoperative stage. Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 28-34 21796700-3 2011 It was reported recently that WZ could significantly increase the blood concentrations of tacrolimus, which might be due to the inhibitory effect of WZ and its ingredients on P-gp and/or CYP450 activity. Tacrolimus 90-100 phosphoglycolate phosphatase Rattus norvegicus 175-179 21711429-0 2011 Influence of CYP3A5 polymorphism on tacrolimus maintenance doses and serum levels after renal transplantation: age dependency and pharmacological interaction with steroids. Tacrolimus 36-46 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 21399973-1 2011 FK506 binding protein 65 (FKBP65) belongs to a group of proteins termed immunophilins that have a high binding affinity to immunosuppressant drugs as FK506 (tacrolimus) and rapamycin (sirolimus). Tacrolimus 157-167 FKBP prolyl isomerase 10 Homo sapiens 0-24 21399973-1 2011 FK506 binding protein 65 (FKBP65) belongs to a group of proteins termed immunophilins that have a high binding affinity to immunosuppressant drugs as FK506 (tacrolimus) and rapamycin (sirolimus). Tacrolimus 157-167 FKBP prolyl isomerase 10 Homo sapiens 26-32 21399973-2 2011 Treatment of female premenopausal women with tacrolimus, which binds to FKBP65, has been reported to be followed by a strongly increased risk of ovarian cysts. Tacrolimus 45-55 FKBP prolyl isomerase 10 Homo sapiens 72-78 21677130-6 2011 In this study, we show that pCD40L surface mobilization is resistant to cyclosporine or FK506 treatment, while de novo CD40L and cytokine expression are completely inhibited. Tacrolimus 88-93 CD40 ligand Homo sapiens 29-34 21708136-4 2011 We find that in primary cultured or T98G GBM cells exposed to therapeutic plasma concentrations of FK506 (tacrolimus), the expression of Mrp1 was decreased in a dose-dependent manner. Tacrolimus 99-104 ATP binding cassette subfamily C member 1 Homo sapiens 137-141 21708136-4 2011 We find that in primary cultured or T98G GBM cells exposed to therapeutic plasma concentrations of FK506 (tacrolimus), the expression of Mrp1 was decreased in a dose-dependent manner. Tacrolimus 106-116 ATP binding cassette subfamily C member 1 Homo sapiens 137-141 21528942-0 2011 Influence of cytochrome P450 3A5 (CYP3A5) genetic polymorphism on the pharmacokinetics of the prolonged-release, once-daily formulation of tacrolimus in stable renal transplant recipients. Tacrolimus 139-149 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-32 21540458-6 2011 Inhibition by IL-2 occurred via a NFAT cell-independent component of the calcineurin pathway, and CD94/NKG2A induction was markedly enhanced in the presence of calcineurin blockers, such as FK506 or using calcineurin-deficient T cells, both in vitro and in vivo. Tacrolimus 190-195 interleukin 2 Homo sapiens 14-18 21540458-6 2011 Inhibition by IL-2 occurred via a NFAT cell-independent component of the calcineurin pathway, and CD94/NKG2A induction was markedly enhanced in the presence of calcineurin blockers, such as FK506 or using calcineurin-deficient T cells, both in vitro and in vivo. Tacrolimus 190-195 killer cell lectin like receptor D1 Homo sapiens 98-102 21540458-6 2011 Inhibition by IL-2 occurred via a NFAT cell-independent component of the calcineurin pathway, and CD94/NKG2A induction was markedly enhanced in the presence of calcineurin blockers, such as FK506 or using calcineurin-deficient T cells, both in vitro and in vivo. Tacrolimus 190-195 killer cell lectin like receptor C1 Homo sapiens 103-108 21528942-0 2011 Influence of cytochrome P450 3A5 (CYP3A5) genetic polymorphism on the pharmacokinetics of the prolonged-release, once-daily formulation of tacrolimus in stable renal transplant recipients. Tacrolimus 139-149 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 21528942-2 2011 The objective of this study was to investigate the influence of the genetic polymorphism of CYP3A5 on the pharmacokinetics of a new modified-release, once-daily formulation of tacrolimus (Advagraf ) after a switch from the immediate-release formulation (Prograf ). Tacrolimus 176-186 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 92-98 21528942-2 2011 The objective of this study was to investigate the influence of the genetic polymorphism of CYP3A5 on the pharmacokinetics of a new modified-release, once-daily formulation of tacrolimus (Advagraf ) after a switch from the immediate-release formulation (Prograf ). Tacrolimus 188-196 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 92-98 21528942-2 2011 The objective of this study was to investigate the influence of the genetic polymorphism of CYP3A5 on the pharmacokinetics of a new modified-release, once-daily formulation of tacrolimus (Advagraf ) after a switch from the immediate-release formulation (Prograf ). Tacrolimus 254-261 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 92-98 21528942-11 2011 CONCLUSION: Tacrolimus exposure significantly decreases after a switch from Prograf to Advagraf , on a milligram-for-milligram basis, in CYP3A5 expressor recipients. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 138-144 21528942-11 2011 CONCLUSION: Tacrolimus exposure significantly decreases after a switch from Prograf to Advagraf , on a milligram-for-milligram basis, in CYP3A5 expressor recipients. Tacrolimus 76-83 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 138-144 21623908-0 2011 A case of tacrolimus-associated thrombotic microangiopathy after ABO-blood-type-incompatible renal transplantation. Tacrolimus 10-20 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 65-68 21528942-11 2011 CONCLUSION: Tacrolimus exposure significantly decreases after a switch from Prograf to Advagraf , on a milligram-for-milligram basis, in CYP3A5 expressor recipients. Tacrolimus 88-96 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 138-144 21623908-3 2011 We present a case of tacrolimus-associated TMA soon after ABO blood type incompatible renal transplantation that was difficult to differentiate from acute AMR. Tacrolimus 21-31 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 58-61 21487055-0 2011 In vitro investigation of human UDP-glucuronosyltransferase isoforms responsible for tacrolimus glucuronidation: predominant contribution of UGT1A4. Tacrolimus 85-95 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 32-59 21606250-1 2011 The immunosuppressive calcineurin inhibitors (CNIs) cyclosporine A (CsA) and tacrolimus are widely used in transplant organ recipients, but in the kidney allograft, they may cause tubulointerstitial as well as mesangial fibrosis, with TGF-beta believed to be a central inductor. Tacrolimus 77-87 transforming growth factor, beta 1 Rattus norvegicus 235-243 21288594-1 2011 FK506 binding protein 12 (FK506BP) is an immunophilin that acts as a receptor for the immunosuppressant drug FK506. Tacrolimus 0-5 FK506 binding protein 1a Mus musculus 26-33 21430698-1 2011 Immunophilin, FK506-binding protein 12 (FK506BP), is a receptor protein for the immunosuppressive drug FK506 by the FK506BP/FK506 complex. Tacrolimus 14-19 FK506 binding protein 1a Mus musculus 40-47 21430698-1 2011 Immunophilin, FK506-binding protein 12 (FK506BP), is a receptor protein for the immunosuppressive drug FK506 by the FK506BP/FK506 complex. Tacrolimus 14-19 FK506 binding protein 1a Mus musculus 116-123 21430698-1 2011 Immunophilin, FK506-binding protein 12 (FK506BP), is a receptor protein for the immunosuppressive drug FK506 by the FK506BP/FK506 complex. Tacrolimus 40-45 FK506 binding protein 1a Mus musculus 14-38 21430698-1 2011 Immunophilin, FK506-binding protein 12 (FK506BP), is a receptor protein for the immunosuppressive drug FK506 by the FK506BP/FK506 complex. Tacrolimus 40-45 FK506 binding protein 1a Mus musculus 116-123 21844990-0 2011 Tacrolimus and 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors: An interaction study in CYP3A5 non-expressors, renal transplant recipients. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 99-105 21844990-4 2011 The aim of this study was to investigate possible pharmacological interactions between tacrolimus and statins in CYP3A5 non-expressors, renal transplant recipients. Tacrolimus 87-97 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 113-119 21844990-13 2011 No significant clinical interaction or effect was observed, even with the use of atorvastatin or simvastatin, which are metabolized by CYP3A4 such as tacrolimus. Tacrolimus 150-160 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 135-141 21606250-2 2011 In this study, we report that the cold-shock protein Y-box binding protein-1 (YB-1) is a TGF-beta independent downstream effector in CsA- as well as in tacrolimus- but not in rapamycin-mediated activation of rat mesangial cells (rMCs). Tacrolimus 152-162 Y box binding protein 1 Rattus norvegicus 53-76 21606250-2 2011 In this study, we report that the cold-shock protein Y-box binding protein-1 (YB-1) is a TGF-beta independent downstream effector in CsA- as well as in tacrolimus- but not in rapamycin-mediated activation of rat mesangial cells (rMCs). Tacrolimus 152-162 Y box binding protein 1 Rattus norvegicus 78-82 21606250-2 2011 In this study, we report that the cold-shock protein Y-box binding protein-1 (YB-1) is a TGF-beta independent downstream effector in CsA- as well as in tacrolimus- but not in rapamycin-mediated activation of rat mesangial cells (rMCs). Tacrolimus 152-162 transforming growth factor, beta 1 Rattus norvegicus 89-97 20238216-0 2011 FK506 inhibition of gliostatin/thymidine phosphorylase production induced by tumor necrosis factor-alpha in rheumatoid fibroblast-like synoviocytes. Tacrolimus 0-5 thymidine phosphorylase Homo sapiens 20-30 21635144-0 2011 Impact of the CYP3A4*1G polymorphism and its combination with CYP3A5 genotypes on tacrolimus pharmacokinetics in renal transplant patients. Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-20 21635144-1 2011 AIM: Tacrolimus is a substrate of CYP3A4 and CYP3A5. Tacrolimus 5-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 34-40 21635144-1 2011 AIM: Tacrolimus is a substrate of CYP3A4 and CYP3A5. Tacrolimus 5-15 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 21635144-2 2011 The present study investigated the impact of the CYP3A4*1/*1G polymorphism compared with CYP3A5 genotypes on the dose-adjusted pharmacokinetics of tacrolimus. Tacrolimus 147-157 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 49-55 21635144-5 2011 RESULTS: The dose-adjusted AUC0-12 and C0 of tacrolimus were significantly lower in patients with the CYP3A4*1G allele and CYP3A5 expressers than those with the CYP3A4*1/*1 genotype and nonexpressers, respectively. Tacrolimus 45-55 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 102-108 21635144-5 2011 RESULTS: The dose-adjusted AUC0-12 and C0 of tacrolimus were significantly lower in patients with the CYP3A4*1G allele and CYP3A5 expressers than those with the CYP3A4*1/*1 genotype and nonexpressers, respectively. Tacrolimus 45-55 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 123-129 21635144-5 2011 RESULTS: The dose-adjusted AUC0-12 and C0 of tacrolimus were significantly lower in patients with the CYP3A4*1G allele and CYP3A5 expressers than those with the CYP3A4*1/*1 genotype and nonexpressers, respectively. Tacrolimus 45-55 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 161-167 21635144-7 2011 The standardized regression coefficient for the AUC0-12 of tacrolimus was approximately twofold less for CYP3A4*1/*1 than CYP3A5*3/*3. Tacrolimus 59-69 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-111 21635144-7 2011 The standardized regression coefficient for the AUC0-12 of tacrolimus was approximately twofold less for CYP3A4*1/*1 than CYP3A5*3/*3. Tacrolimus 59-69 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 122-128 21635144-9 2011 CONCLUSION: The CYP3A4*1/*1G polymorphism was associated with the pharmacokinetics of tacrolimus, however, its contribution to dose-adjusted pharmacokinetics was approximately twofold less than that of the CYP3A5*1/*3 polymorphism. Tacrolimus 86-96 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 16-22 21635144-9 2011 CONCLUSION: The CYP3A4*1/*1G polymorphism was associated with the pharmacokinetics of tacrolimus, however, its contribution to dose-adjusted pharmacokinetics was approximately twofold less than that of the CYP3A5*1/*3 polymorphism. Tacrolimus 86-96 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 206-212 21635144-10 2011 Although its effect on CYP3A4 activity is not clear, CYP3A4*1/*1G may be a candidate for a polymorphism affecting the interindividual variability in tacrolimus pharmacokinetics among CYP3A5 expressers. Tacrolimus 149-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 23-29 21635144-10 2011 Although its effect on CYP3A4 activity is not clear, CYP3A4*1/*1G may be a candidate for a polymorphism affecting the interindividual variability in tacrolimus pharmacokinetics among CYP3A5 expressers. Tacrolimus 149-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 53-59 21635144-10 2011 Although its effect on CYP3A4 activity is not clear, CYP3A4*1/*1G may be a candidate for a polymorphism affecting the interindividual variability in tacrolimus pharmacokinetics among CYP3A5 expressers. Tacrolimus 149-159 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 183-189 20238216-0 2011 FK506 inhibition of gliostatin/thymidine phosphorylase production induced by tumor necrosis factor-alpha in rheumatoid fibroblast-like synoviocytes. Tacrolimus 0-5 tumor necrosis factor Homo sapiens 77-104 20238216-8 2011 GLS levels in TNF-alpha-stimulated FLSs were reduced by FK506 treatment. Tacrolimus 56-61 tumor necrosis factor Homo sapiens 14-23 21635950-7 2011 CD3(+)CD56(+)NKT cells were also lower during immunosuppressive therapy with tacrolimus (0.03+-0.01/HPF) than with cyclosporine (0.1+-0.003/HPF), cyclosporine/MMF (0.1+-0.003/HPF) or sirolimus (0.1+-0.01/HPF) treatment. Tacrolimus 77-87 neural cell adhesion molecule 1 Homo sapiens 6-10 21544031-8 2011 CONCLUSIONS: Based on retrospectively collected data of 103 renal transplant recipients receiving tacrolimus in combination with mycophenolate mofetil and corticosteroids, we found that renal expression and localization of CYP3A5 but not P-gp is associated with the occurrence of CNIT. Tacrolimus 98-108 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 223-229 21839244-0 2011 Pharmacogenetic study of ABCB1 and CYP3A5 genes during the first year following heart transplantation regarding tacrolimus or cyclosporine levels. Tacrolimus 112-122 ATP binding cassette subfamily B member 1 Homo sapiens 25-30 21839244-0 2011 Pharmacogenetic study of ABCB1 and CYP3A5 genes during the first year following heart transplantation regarding tacrolimus or cyclosporine levels. Tacrolimus 112-122 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 35-41 21839244-3 2011 We evaluated the SNPs in ABCB1 (glycoprotein P) and CYP3A5 (metabolic enzyme) genes, seeking correlate them with tacrolimus or cyclosporine levels during the first year after heart transplantation. Tacrolimus 113-123 ATP binding cassette subfamily B member 1 Homo sapiens 25-30 21839244-3 2011 We evaluated the SNPs in ABCB1 (glycoprotein P) and CYP3A5 (metabolic enzyme) genes, seeking correlate them with tacrolimus or cyclosporine levels during the first year after heart transplantation. Tacrolimus 113-123 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 52-58 22010353-5 2011 It is proved that the network server has the basic functions for database management and three levels of prediction to aid doctor to optimize the regimen of tacrolimus for Chinese renal transplant patients. Tacrolimus 157-167 activation induced cytidine deaminase Homo sapiens 119-122 21518963-1 2011 Patients treated with the immunosuppressive drug tacrolimus (FK506), which binds FK506 binding protein 12 (FKBP12) and then inhibits the calcium-dependent phosphatase calcineurin, exhibit decreased regulatory T cells, endothelial dysfunction, and hypertension; however, the mechanisms and whether altered T-cell polarization play a role are unknown. Tacrolimus 49-59 FK506 binding protein 1a Mus musculus 81-105 21518963-1 2011 Patients treated with the immunosuppressive drug tacrolimus (FK506), which binds FK506 binding protein 12 (FKBP12) and then inhibits the calcium-dependent phosphatase calcineurin, exhibit decreased regulatory T cells, endothelial dysfunction, and hypertension; however, the mechanisms and whether altered T-cell polarization play a role are unknown. Tacrolimus 49-59 FK506 binding protein 1a Mus musculus 107-113 21518963-1 2011 Patients treated with the immunosuppressive drug tacrolimus (FK506), which binds FK506 binding protein 12 (FKBP12) and then inhibits the calcium-dependent phosphatase calcineurin, exhibit decreased regulatory T cells, endothelial dysfunction, and hypertension; however, the mechanisms and whether altered T-cell polarization play a role are unknown. Tacrolimus 61-66 FK506 binding protein 1a Mus musculus 81-105 21518963-1 2011 Patients treated with the immunosuppressive drug tacrolimus (FK506), which binds FK506 binding protein 12 (FKBP12) and then inhibits the calcium-dependent phosphatase calcineurin, exhibit decreased regulatory T cells, endothelial dysfunction, and hypertension; however, the mechanisms and whether altered T-cell polarization play a role are unknown. Tacrolimus 61-66 FK506 binding protein 1a Mus musculus 107-113 21518963-2 2011 Tacrolimus treatment of mice for 1 week dose-dependently decreased splenic CD4(+)/FoxP3(+) (regulatory T cells), increased splenic CD4(+)/IL-17(+) (T-helper 17) cells, and caused endothelial dysfunction and hypertension. Tacrolimus 0-10 CD4 antigen Mus musculus 75-78 21518963-2 2011 Tacrolimus treatment of mice for 1 week dose-dependently decreased splenic CD4(+)/FoxP3(+) (regulatory T cells), increased splenic CD4(+)/IL-17(+) (T-helper 17) cells, and caused endothelial dysfunction and hypertension. Tacrolimus 0-10 forkhead box P3 Mus musculus 82-87 21518963-2 2011 Tacrolimus treatment of mice for 1 week dose-dependently decreased splenic CD4(+)/FoxP3(+) (regulatory T cells), increased splenic CD4(+)/IL-17(+) (T-helper 17) cells, and caused endothelial dysfunction and hypertension. Tacrolimus 0-10 CD4 antigen Mus musculus 131-134 21518963-2 2011 Tacrolimus treatment of mice for 1 week dose-dependently decreased splenic CD4(+)/FoxP3(+) (regulatory T cells), increased splenic CD4(+)/IL-17(+) (T-helper 17) cells, and caused endothelial dysfunction and hypertension. Tacrolimus 0-10 interleukin 17A Mus musculus 138-143 21518964-0 2011 Tacrolimus-induced hypertension: what"s endothelial and hematopoietic FKBP12 got to do with it? Tacrolimus 0-10 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 70-76 21383022-0 2011 Protein kinase CbetaII-mediated phosphorylation of endothelial nitric oxide synthase threonine 495 mediates the endothelial dysfunction induced by FK506 (tacrolimus). Tacrolimus 147-152 nitric oxide synthase 3 Homo sapiens 51-84 21383022-0 2011 Protein kinase CbetaII-mediated phosphorylation of endothelial nitric oxide synthase threonine 495 mediates the endothelial dysfunction induced by FK506 (tacrolimus). Tacrolimus 154-164 nitric oxide synthase 3 Homo sapiens 51-84 21383022-2 2011 We have previously shown that FK506 activates Ca(2+)/diacylglycerol-dependent conventional protein kinase C (cPKC), which phosphorylates endothelial nitric oxide synthase (eNOS) at one of its inhibitory sites, Thr495. Tacrolimus 30-35 nitric oxide synthase 3 Homo sapiens 137-170 21515370-7 2011 Cell proliferative responses and IFN-gamma production were suppressed to a significant extent in the FK506 group compared with the TAK-779 group. Tacrolimus 101-106 interferon gamma Rattus norvegicus 33-42 21515370-9 2011 The numbers of both CD4(+) and CD8(+) cells were significantly suppressed in the blood of the recipients of both the FK506 and the TAK-779 groups, and in Peyer"s patches of the graft of the TAK-779 group, but the FK506 group was not, as evidenced by FACS analysis. Tacrolimus 117-122 Cd4 molecule Rattus norvegicus 20-23 21515370-9 2011 The numbers of both CD4(+) and CD8(+) cells were significantly suppressed in the blood of the recipients of both the FK506 and the TAK-779 groups, and in Peyer"s patches of the graft of the TAK-779 group, but the FK506 group was not, as evidenced by FACS analysis. Tacrolimus 213-218 Cd4 molecule Rattus norvegicus 20-23 20351753-6 2011 Both tacrolimus and rapamycin caused similar suppression of GSIS in cells expressing ZnT-8 R325. Tacrolimus 5-15 solute carrier family 30 member 8 Homo sapiens 85-90 20351753-7 2011 However, cells expressing ZnT-8 W325 were resistant to tacrolimus, but not to rapamycin. Tacrolimus 55-65 solute carrier family 30 member 8 Homo sapiens 26-31 21355884-0 2011 KCNQ1 gene variants and risk of new-onset diabetes in tacrolimus-treated renal-transplanted patients. Tacrolimus 54-64 potassium voltage-gated channel subfamily Q member 1 Homo sapiens 0-5 21273094-3 2011 In this we study examined the effect of tacrolimus (Tac) in an identical model. Tacrolimus 40-50 tachykinin, precursor 1 Rattus norvegicus 52-55 21310938-0 2011 Immunosuppressor FK506 increases endoglin and activin receptor-like kinase 1 expression and modulates transforming growth factor-beta1 signaling in endothelial cells. Tacrolimus 17-22 endoglin Homo sapiens 33-41 21310938-0 2011 Immunosuppressor FK506 increases endoglin and activin receptor-like kinase 1 expression and modulates transforming growth factor-beta1 signaling in endothelial cells. Tacrolimus 17-22 transforming growth factor beta 1 Homo sapiens 102-134 21310938-7 2011 We report the efficacy of tacrolimus (FK506) in increasing ENG and ALK1 expression. Tacrolimus 26-36 endoglin Homo sapiens 59-62 21310938-7 2011 We report the efficacy of tacrolimus (FK506) in increasing ENG and ALK1 expression. Tacrolimus 26-36 activin A receptor like type 1 Homo sapiens 67-71 21310938-7 2011 We report the efficacy of tacrolimus (FK506) in increasing ENG and ALK1 expression. Tacrolimus 38-43 endoglin Homo sapiens 59-62 21310938-7 2011 We report the efficacy of tacrolimus (FK506) in increasing ENG and ALK1 expression. Tacrolimus 38-43 activin A receptor like type 1 Homo sapiens 67-71 21310938-11 2011 Here, we find that the immunosuppressor FK506 increases the protein and mRNA expression of ENG and ALK1 in cultured endothelial cells and enhances the TGF-beta1/ALK1 signaling pathway and endothelial cell functions like tubulogenesis and migration. Tacrolimus 40-45 endoglin Homo sapiens 91-94 21310938-11 2011 Here, we find that the immunosuppressor FK506 increases the protein and mRNA expression of ENG and ALK1 in cultured endothelial cells and enhances the TGF-beta1/ALK1 signaling pathway and endothelial cell functions like tubulogenesis and migration. Tacrolimus 40-45 activin A receptor like type 1 Homo sapiens 99-103 21310938-11 2011 Here, we find that the immunosuppressor FK506 increases the protein and mRNA expression of ENG and ALK1 in cultured endothelial cells and enhances the TGF-beta1/ALK1 signaling pathway and endothelial cell functions like tubulogenesis and migration. Tacrolimus 40-45 transforming growth factor beta 1 Homo sapiens 151-160 21310938-11 2011 Here, we find that the immunosuppressor FK506 increases the protein and mRNA expression of ENG and ALK1 in cultured endothelial cells and enhances the TGF-beta1/ALK1 signaling pathway and endothelial cell functions like tubulogenesis and migration. Tacrolimus 40-45 activin A receptor like type 1 Homo sapiens 161-165 21310938-12 2011 These results suggest that the mechanism of action of FK506 involves a partial correction of endoglin and ALK1 haploinsufficiency and may therefore be an interesting drug for use in patients with HHT who undergo transplantation. Tacrolimus 54-59 endoglin Homo sapiens 93-101 21541276-0 2011 Eosinophil cationic protein as a marker for assessing the efficacy of tacrolimus ophthalmic solution in the treatment of atopic keratoconjunctivitis. Tacrolimus 70-80 ribonuclease A family member 3 Homo sapiens 0-27 21620040-0 2011 Interleukin-2 profiles shortly after tacrolimus conversion from a twice-daily to once-daily regimen. Tacrolimus 37-47 interleukin 2 Homo sapiens 0-13 21195813-10 2011 We found that tacrolimus reduced levels of the IFN-gamma, IL-2, IL-10, and IL-13 cytokines and induced the proliferation of tolerogenic plasmacytoid dendritic cells after treatment. Tacrolimus 14-24 interferon gamma Homo sapiens 47-56 21368751-3 2011 We have shown in vitro that ezetimibe and tacrolimus may interact in competition for intestinal UGT1A1 and ABCB1 at concentrations reached in gut lumen after oral administration. Tacrolimus 42-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 21368751-3 2011 We have shown in vitro that ezetimibe and tacrolimus may interact in competition for intestinal UGT1A1 and ABCB1 at concentrations reached in gut lumen after oral administration. Tacrolimus 42-52 ATP binding cassette subfamily B member 1 Homo sapiens 107-112 21172435-0 2011 Surfactant protein A (SP-A)-tacrolimus complexes have a greater anti-inflammatory effect than either SP-A or tacrolimus alone on human macrophage-like U937 cells. Tacrolimus 28-38 surfactant protein A1 Homo sapiens 0-20 21172435-0 2011 Surfactant protein A (SP-A)-tacrolimus complexes have a greater anti-inflammatory effect than either SP-A or tacrolimus alone on human macrophage-like U937 cells. Tacrolimus 28-38 surfactant protein A1 Homo sapiens 22-26 21172435-0 2011 Surfactant protein A (SP-A)-tacrolimus complexes have a greater anti-inflammatory effect than either SP-A or tacrolimus alone on human macrophage-like U937 cells. Tacrolimus 109-119 surfactant protein A1 Homo sapiens 0-20 21172435-0 2011 Surfactant protein A (SP-A)-tacrolimus complexes have a greater anti-inflammatory effect than either SP-A or tacrolimus alone on human macrophage-like U937 cells. Tacrolimus 109-119 surfactant protein A1 Homo sapiens 22-26 21172435-2 2011 Nanoparticles of tacrolimus (FK506) might interact with human SP-A, which is the most abundant lipoprotein in the alveolar fluid. Tacrolimus 17-27 surfactant protein A1 Homo sapiens 62-66 21172435-2 2011 Nanoparticles of tacrolimus (FK506) might interact with human SP-A, which is the most abundant lipoprotein in the alveolar fluid. Tacrolimus 29-34 surfactant protein A1 Homo sapiens 62-66 21172435-3 2011 This study was undertaken to determine whether the formation of FK506/SP-A complexes interferes with FK506 immunosuppressive actions on stimulated human macrophage-like U937 cells. Tacrolimus 64-69 surfactant protein A1 Homo sapiens 70-74 21172435-3 2011 This study was undertaken to determine whether the formation of FK506/SP-A complexes interferes with FK506 immunosuppressive actions on stimulated human macrophage-like U937 cells. Tacrolimus 101-106 surfactant protein A1 Homo sapiens 70-74 21172435-4 2011 We found that SP-A was avidly bound to FK506 (K(d) = 35 +- 4nM), as determined by solid phase-binding assays and dynamic light scattering. Tacrolimus 39-44 surfactant protein A1 Homo sapiens 14-18 21172435-6 2011 However, coincubation of FK506 and SP-A, at concentrations where each component alone did not affect LPS-stimulated macrophage response, significantly inhibited LPS-induced NF-kappaB activation and TNF-alpha secretion. Tacrolimus 25-30 tumor necrosis factor Homo sapiens 198-207 21172435-8 2011 FK506 bound to SP-A was delivered to macrophages by endocytosis, since several endocytosis inhibitors blocked FK506/SP-A anti-inflammatory effects. Tacrolimus 0-5 surfactant protein A1 Homo sapiens 15-19 21172435-8 2011 FK506 bound to SP-A was delivered to macrophages by endocytosis, since several endocytosis inhibitors blocked FK506/SP-A anti-inflammatory effects. Tacrolimus 0-5 surfactant protein A1 Homo sapiens 116-120 21172435-8 2011 FK506 bound to SP-A was delivered to macrophages by endocytosis, since several endocytosis inhibitors blocked FK506/SP-A anti-inflammatory effects. Tacrolimus 110-115 surfactant protein A1 Homo sapiens 15-19 21434949-0 2011 Tacrolimus suppresses IL-12/IL23 p40 in Crohn"s disease and heals fistulae refractory to anti-TNF-alpha therapy. Tacrolimus 0-10 interleukin 23 subunit alpha Homo sapiens 28-34 21434949-0 2011 Tacrolimus suppresses IL-12/IL23 p40 in Crohn"s disease and heals fistulae refractory to anti-TNF-alpha therapy. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 94-103 21070333-11 2011 CONCLUSION: Short-term therapy with tacrolimus ointment reduced expression of TLR-1, which may inhibit the antimicrobial potential of TLR-2, and also reversed the impairment of TLR-2 in AD lesions. Tacrolimus 36-46 toll like receptor 1 Homo sapiens 78-83 21070333-11 2011 CONCLUSION: Short-term therapy with tacrolimus ointment reduced expression of TLR-1, which may inhibit the antimicrobial potential of TLR-2, and also reversed the impairment of TLR-2 in AD lesions. Tacrolimus 36-46 toll like receptor 2 Homo sapiens 134-139 21070333-11 2011 CONCLUSION: Short-term therapy with tacrolimus ointment reduced expression of TLR-1, which may inhibit the antimicrobial potential of TLR-2, and also reversed the impairment of TLR-2 in AD lesions. Tacrolimus 36-46 toll like receptor 2 Homo sapiens 177-182 21195813-10 2011 We found that tacrolimus reduced levels of the IFN-gamma, IL-2, IL-10, and IL-13 cytokines and induced the proliferation of tolerogenic plasmacytoid dendritic cells after treatment. Tacrolimus 14-24 interleukin 2 Homo sapiens 58-62 21195813-10 2011 We found that tacrolimus reduced levels of the IFN-gamma, IL-2, IL-10, and IL-13 cytokines and induced the proliferation of tolerogenic plasmacytoid dendritic cells after treatment. Tacrolimus 14-24 interleukin 10 Homo sapiens 64-69 21195813-10 2011 We found that tacrolimus reduced levels of the IFN-gamma, IL-2, IL-10, and IL-13 cytokines and induced the proliferation of tolerogenic plasmacytoid dendritic cells after treatment. Tacrolimus 14-24 interleukin 13 Homo sapiens 75-80 21195813-11 2011 Tacrolimus did not change the population of T cell subtypes but did steadily reduce the population of BAFF-R(+) CD19(+) B cells over the course of the study. Tacrolimus 0-10 TNF receptor superfamily member 13C Homo sapiens 102-108 21195813-11 2011 Tacrolimus did not change the population of T cell subtypes but did steadily reduce the population of BAFF-R(+) CD19(+) B cells over the course of the study. Tacrolimus 0-10 CD19 molecule Homo sapiens 112-116 21195813-13 2011 The decrease in IL-13 and reduction of BAFF-R(+) CD19(+) B cells may be related to the therapeutic effect of tacrolimus. Tacrolimus 109-119 interleukin 13 Homo sapiens 16-21 21195813-13 2011 The decrease in IL-13 and reduction of BAFF-R(+) CD19(+) B cells may be related to the therapeutic effect of tacrolimus. Tacrolimus 109-119 TNF receptor superfamily member 13C Homo sapiens 39-45 21195813-13 2011 The decrease in IL-13 and reduction of BAFF-R(+) CD19(+) B cells may be related to the therapeutic effect of tacrolimus. Tacrolimus 109-119 CD19 molecule Homo sapiens 49-53 21366650-3 2011 The objective of this study was to investigate how, 1 year after renal transplantation, CYP3A5 and CYP2C19 polymorphisms, biochemical parameters and coadministration with lansoprazole, influenced tacrolimus pharmacokinetics. Tacrolimus 196-206 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 88-94 20571034-0 2011 Do drug transporter (ABCB1) SNPs influence cyclosporine and tacrolimus dose requirements and renal allograft outcome in the posttransplantation period? Tacrolimus 60-70 ATP binding cassette subfamily B member 1 Homo sapiens 21-26 20571034-1 2011 Polymorphisms in the drug transporter gene (ABCB1) may play a significant role in individualizing cyclosporine (CsA) and tacrolimus (Tac) dosage and subsequently the allograft outcome in renal transplant recipients. Tacrolimus 121-131 ATP binding cassette subfamily B member 1 Homo sapiens 44-49 20571034-1 2011 Polymorphisms in the drug transporter gene (ABCB1) may play a significant role in individualizing cyclosporine (CsA) and tacrolimus (Tac) dosage and subsequently the allograft outcome in renal transplant recipients. Tacrolimus 133-136 ATP binding cassette subfamily B member 1 Homo sapiens 44-49 21366650-3 2011 The objective of this study was to investigate how, 1 year after renal transplantation, CYP3A5 and CYP2C19 polymorphisms, biochemical parameters and coadministration with lansoprazole, influenced tacrolimus pharmacokinetics. Tacrolimus 196-206 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 99-106 21366650-6 2011 In a multiple regression analysis, the dose-adjusted AUC(0-12) and C(max) of tacrolimus were associated with CYP3A5*3/*3 and co-administration with lansoprazole. Tacrolimus 77-87 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 109-115 21366650-8 2011 The long-term combination of tacrolimus and lansoprazole requires careful monitoring of patients with the CYP3A5*3/*3 genotype. Tacrolimus 29-39 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 106-112 21720672-7 2011 RESULTS: We observed a higher rate of insulin independence in PTA (75%) versus ITA (59%), with the longer insulin independence among PTA patients receiving tacrolimus. Tacrolimus 156-166 insulin Homo sapiens 38-45 20818295-0 2011 Differential impact of the CYP3A5*1 and CYP3A5*3 alleles on pre-dose concentrations of two tacrolimus formulations. Tacrolimus 91-101 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 27-33 20818295-0 2011 Differential impact of the CYP3A5*1 and CYP3A5*3 alleles on pre-dose concentrations of two tacrolimus formulations. Tacrolimus 91-101 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 40-46 21720672-7 2011 RESULTS: We observed a higher rate of insulin independence in PTA (75%) versus ITA (59%), with the longer insulin independence among PTA patients receiving tacrolimus. Tacrolimus 156-166 insulin Homo sapiens 106-113 21076890-0 2011 Protective effect of FK506 on myocardial ischemia/reperfusion injury by suppression of CaN and ASK1 signaling circuitry. Tacrolimus 21-26 mitogen-activated protein kinase kinase kinase 5 Rattus norvegicus 95-99 21200364-12 2011 PGC1-alpha mRNA was down-regulated by FK506. Tacrolimus 38-43 PPARG coactivator 1 alpha Rattus norvegicus 0-10 21255669-8 2011 Tacrolimus diminished both pancreas and liver iNOS and nNOS expression. Tacrolimus 0-10 nitric oxide synthase 2 Rattus norvegicus 46-50 21255669-8 2011 Tacrolimus diminished both pancreas and liver iNOS and nNOS expression. Tacrolimus 0-10 nitric oxide synthase 1 Rattus norvegicus 55-59 21255669-11 2011 Tacrolimus treatment diminished TNF-alpha but not IL-1 expression increase after LPS challenge in hepatic tissue. Tacrolimus 0-10 tumor necrosis factor Rattus norvegicus 32-41 21255669-13 2011 Plasmatic NO, CO, TNF-alpha, and IL-1 concentrations increase after LPS challenge was diminished when highest doses of tacrolimus were given. Tacrolimus 119-129 tumor necrosis factor Rattus norvegicus 18-27 21199863-10 2011 In summary, the evidence provided here argues for a previously unanticipated role of calcineurin in CBM complex formation as a molecular basis of the inhibitory function of CsA or FK506 on TCR-induced NF-kappaB activity. Tacrolimus 180-185 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 189-192 21039414-10 2011 ET B receptor, which was not present in NCCmelb4 cells, was induced after FK506 treatment. Tacrolimus 74-79 endothelin receptor type B Mus musculus 0-4 21389905-1 2011 BACKGROUND: CYP3A5 genotyping might be useful to guide tacrolimus and sirolimus dosing. Tacrolimus 55-65 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 21389905-6 2011 On the other hand, no effect of CYP3A5 polymorphism was observed on the intrinsic clearance of everolimus by human liver microsomes, whereas that of tacrolimus (positive control) was 1.5-fold higher in microsomes carrying the CYP3A5*1 allele than in noncarriers. Tacrolimus 149-159 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 226-232 21423799-4 2011 Specifically, CsA and FK506 bind to intracellular proteins, principally cyclophilin A and FKBP12, respectively, and thereby inhibit the phosphatase calcineurin (Cn). Tacrolimus 22-27 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 90-96 21076890-8 2011 Increase in Bcl-2/Bax ratio in FK506-IR group indicated that FK506 protected myocardium from apoptosis induced by IR. Tacrolimus 31-36 BCL2, apoptosis regulator Rattus norvegicus 12-17 21076890-8 2011 Increase in Bcl-2/Bax ratio in FK506-IR group indicated that FK506 protected myocardium from apoptosis induced by IR. Tacrolimus 31-36 BCL2 associated X, apoptosis regulator Rattus norvegicus 18-21 21076890-8 2011 Increase in Bcl-2/Bax ratio in FK506-IR group indicated that FK506 protected myocardium from apoptosis induced by IR. Tacrolimus 61-66 BCL2, apoptosis regulator Rattus norvegicus 12-17 21076890-8 2011 Increase in Bcl-2/Bax ratio in FK506-IR group indicated that FK506 protected myocardium from apoptosis induced by IR. Tacrolimus 61-66 BCL2 associated X, apoptosis regulator Rattus norvegicus 18-21 21076890-9 2011 The activity of CaN and ASK1 protein level decreased significantly after I/R injury in FK506-treated I/R heart. Tacrolimus 87-92 mitogen-activated protein kinase kinase kinase 5 Rattus norvegicus 24-28 21076890-10 2011 FK506 suppresses the activation of CaN and ASK1 through CaN-mediated apoptosis pathway, and ASK1 negatively regulates CaN activity. Tacrolimus 0-5 mitogen-activated protein kinase kinase kinase 5 Rattus norvegicus 43-47 21076890-11 2011 Suppression of CaN and ASK1 signaling circuitry are involved in protective effect of FK506 on rat myocardium I/R injury. Tacrolimus 85-90 mitogen-activated protein kinase kinase kinase 5 Rattus norvegicus 23-27 21107352-0 2011 IL-1alpha stimulation restores epidermal permeability and antimicrobial barriers compromised by topical tacrolimus. Tacrolimus 104-114 interleukin 1 alpha Mus musculus 0-9 21289623-0 2011 ABCB1 single-nucleotide polymorphisms determine tacrolimus response in patients with ulcerative colitis. Tacrolimus 48-58 ATP binding cassette subfamily B member 1 Homo sapiens 0-5 21107352-2 2011 Tacrolimus decreases lipid synthesis and the expressions of antimicrobial peptide (AMP) and IL-1alpha in the epidermis. Tacrolimus 0-10 interleukin 1 alpha Mus musculus 92-101 21107352-4 2011 We aimed to evaluate whether IL-1alpha stimulation could restore the barrier dysfunction observed in tacrolimus-treated skin. Tacrolimus 101-111 interleukin 1 alpha Mus musculus 29-38 21107352-9 2011 In conclusion, IL-1alpha stimulation induced positive effects on epidermal permeability and antimicrobial barrier functions in tacrolimus-treated skin. Tacrolimus 127-137 interleukin 1 alpha Mus musculus 15-24 21280190-1 2011 Tacrolimus, a cornerstone immunosuppressant, is available as a twice-daily formulation (tacrolimus bid). Tacrolimus 0-10 BH3 interacting domain death agonist Homo sapiens 99-102 21416322-5 2011 FK506 treatment combined with gradual withdrawal of NTBC before cell transplantation ensured that Fah (-/-) Nod/Scid mice were susceptible to liver xeno-repopulation by human hepatocytes; the proportion of engrafted human hepatocytes reached 33.6%. Tacrolimus 0-5 fumarylacetoacetate hydrolase Mus musculus 98-101 21206424-1 2011 BACKGROUND: The CYP4503A5*1 genotype is associated with lower tacrolimus concentrations. Tacrolimus 62-72 peptidylprolyl isomerase G Homo sapiens 16-19 21206424-10 2011 CONCLUSION: We identified variants beyond CYP3A5*3, which may further explain pharmacokinetic variability of tacrolimus and demonstrated that important variants differ by race. Tacrolimus 109-119 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 42-48 21289178-4 2011 In cardiomyocytes, FK506-binding protein 1b/12.6 (FKBP1b) binds and stabilizes RyR2 in the closed state, inhibiting RyR-mediated Ca(2+) release. Tacrolimus 19-24 FKBP prolyl isomerase 1B Rattus norvegicus 50-56 21289178-4 2011 In cardiomyocytes, FK506-binding protein 1b/12.6 (FKBP1b) binds and stabilizes RyR2 in the closed state, inhibiting RyR-mediated Ca(2+) release. Tacrolimus 19-24 ryanodine receptor 2 Rattus norvegicus 79-83 21289178-4 2011 In cardiomyocytes, FK506-binding protein 1b/12.6 (FKBP1b) binds and stabilizes RyR2 in the closed state, inhibiting RyR-mediated Ca(2+) release. Tacrolimus 19-24 ryanodine receptor 2 Rattus norvegicus 79-82 20937549-0 2011 Bidirectional immunoregulation of calcineurin inhibitor tacrolimus on FOXP3 transcription? Tacrolimus 56-66 calcineurin binding protein 1 Homo sapiens 34-55 20524872-12 2011 In tacrolimus-treated cases, there was a decrease in CD4 and MHC II, with no change in CD8 between the pre- and post-treated areas. Tacrolimus 3-13 CD4 molecule Homo sapiens 53-56 21280190-1 2011 Tacrolimus, a cornerstone immunosuppressant, is available as a twice-daily formulation (tacrolimus bid). Tacrolimus 88-98 BH3 interacting domain death agonist Homo sapiens 99-102 21280190-6 2011 Tacrolimus area under the curve (AUC) from 0 to 24 hours (AUC(0-24) ) at equivalent doses was approximately 50% lower for tacrolimus qd than for bid on day 1 (146 versus 264 ng h/mL, respectively), but by day 14 was comparable between treatments (324 and 287 ng h/mL, respectively) with higher tacrolimus qd doses. Tacrolimus 0-10 BH3 interacting domain death agonist Homo sapiens 145-148 20937549-0 2011 Bidirectional immunoregulation of calcineurin inhibitor tacrolimus on FOXP3 transcription? Tacrolimus 56-66 forkhead box P3 Homo sapiens 70-75 21280190-7 2011 There was a strong correlation between AUC(0-24) and concentration at 24 hours for tacrolimus qd and bid (r = 0.92 and r = 0.76, respectively). Tacrolimus 83-93 BH3 interacting domain death agonist Homo sapiens 101-104 20937549-4 2011 Data show that tacrolimus has multiple impacts on FOXP3, but the exact pharmacological mechanism of tacrolimus on FOXP3 have yet to be elucidated. Tacrolimus 15-25 forkhead box P3 Homo sapiens 50-55 21099736-6 2011 All animals received intravenous Tacrolimus (0.025 mg/kg) BID during the course of the study. Tacrolimus 33-43 BH3 interacting domain death agonist Homo sapiens 58-61 20937549-4 2011 Data show that tacrolimus has multiple impacts on FOXP3, but the exact pharmacological mechanism of tacrolimus on FOXP3 have yet to be elucidated. Tacrolimus 100-110 forkhead box P3 Homo sapiens 114-119 20937549-5 2011 We herein suggest the bidirectional immunoregulation of tacrolimus on FOXP3. Tacrolimus 56-66 forkhead box P3 Homo sapiens 70-75 20937549-6 2011 High concentration of tacrolimus renders the cooperation of NFAT with STAT6 and NF-kappaB to activate GATA3 transcription. Tacrolimus 22-32 signal transducer and activator of transcription 6 Homo sapiens 70-75 20937549-6 2011 High concentration of tacrolimus renders the cooperation of NFAT with STAT6 and NF-kappaB to activate GATA3 transcription. Tacrolimus 22-32 GATA binding protein 3 Homo sapiens 102-107 20937549-7 2011 On the contrary, low concentration of tacrolimus results in higher nucleus level of NFAT, which directly binds to FOXP3 enhancer and/or cooperates with Smad3 to activate FOXP3 transcription. Tacrolimus 38-48 forkhead box P3 Homo sapiens 114-119 20937549-7 2011 On the contrary, low concentration of tacrolimus results in higher nucleus level of NFAT, which directly binds to FOXP3 enhancer and/or cooperates with Smad3 to activate FOXP3 transcription. Tacrolimus 38-48 SMAD family member 3 Homo sapiens 152-157 20937549-7 2011 On the contrary, low concentration of tacrolimus results in higher nucleus level of NFAT, which directly binds to FOXP3 enhancer and/or cooperates with Smad3 to activate FOXP3 transcription. Tacrolimus 38-48 forkhead box P3 Homo sapiens 170-175 20937549-8 2011 Further studies using loss of function and over-expression methods are needed to determine the detailed molecules involved in this bidirectional immunoregulation of tacrolimus on FOXP3. Tacrolimus 165-175 forkhead box P3 Homo sapiens 179-184 21176018-6 2011 The use of anti-interleukin-2 inhibitors as induction therapy, with low-dose tacrolimus or in combination with mycophenolate mofetil, has a key role in preventing significant renal dysfunction and reducing infection and rejection. Tacrolimus 77-87 interleukin 2 Homo sapiens 16-29 21094139-6 2011 Dephosphorylation and nuclear translocation of NFAT1 caused by ILY were prevented by [Ca(2+)]i calcium chelator, BAPTA/AM, and calcineurin inhibitors, cyclosporine A and tacrolimus. Tacrolimus 170-180 nuclear factor of activated T cells 2 Homo sapiens 47-52 21255728-3 2011 A variety of inhibitors of the PI activity of FKBP12, including FK506, rapamycin, and cycloheximide, increase steady-state palmitoylation. Tacrolimus 64-69 FKBP prolyl isomerase 1A Rattus norvegicus 46-52 21255728-4 2011 FK506 inhibits retrograde trafficking of H-Ras from the plasma membrane to the Golgi in a proline 179-dependent fashion, augments early GTP loading of Ras in response to growth factors, and promotes H-Ras-dependent neurite outgrowth from PC12 cells. Tacrolimus 0-5 HRas proto-oncogene, GTPase Rattus norvegicus 41-46 21255728-4 2011 FK506 inhibits retrograde trafficking of H-Ras from the plasma membrane to the Golgi in a proline 179-dependent fashion, augments early GTP loading of Ras in response to growth factors, and promotes H-Ras-dependent neurite outgrowth from PC12 cells. Tacrolimus 0-5 HRas proto-oncogene, GTPase Rattus norvegicus 199-204 21452412-9 2011 Tacrolimus blood levels in the early stage posttransplantation were higher in nonexpressers than CYP3A5 expressers in both regimens 1 and 2, despite therapeutic drug monitoring. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 97-103 21669047-11 2011 In contrast to animals treated with a combination of FK506 + mycophenolate, robust CD4/8 immunoreactivity was identified in the vicinity of the injection tract in animals treated with FK506 only. Tacrolimus 184-189 CD48 molecule Homo sapiens 83-88 21691056-6 2011 CsA and tacrolimus reduced mRNA abundance in TRPV5 (CsA: 64 +- 3% of control; tacrolimus: 50 +- 3%) calbindin-D28k (CsA: 62 +- 4%; tacrolimus: 43 +- 3%), and vitamin D receptor (CsA: 52 +- 3%; tacrolimus: 58 +- 2%, all p < 0.05). Tacrolimus 8-18 transient receptor potential cation channel subfamily V member 5 Homo sapiens 45-50 21691056-6 2011 CsA and tacrolimus reduced mRNA abundance in TRPV5 (CsA: 64 +- 3% of control; tacrolimus: 50 +- 3%) calbindin-D28k (CsA: 62 +- 4%; tacrolimus: 43 +- 3%), and vitamin D receptor (CsA: 52 +- 3%; tacrolimus: 58 +- 2%, all p < 0.05). Tacrolimus 8-18 vitamin D receptor Homo sapiens 158-176 21691056-6 2011 CsA and tacrolimus reduced mRNA abundance in TRPV5 (CsA: 64 +- 3% of control; tacrolimus: 50 +- 3%) calbindin-D28k (CsA: 62 +- 4%; tacrolimus: 43 +- 3%), and vitamin D receptor (CsA: 52 +- 3%; tacrolimus: 58 +- 2%, all p < 0.05). Tacrolimus 78-88 transient receptor potential cation channel subfamily V member 5 Homo sapiens 45-50 21691056-6 2011 CsA and tacrolimus reduced mRNA abundance in TRPV5 (CsA: 64 +- 3% of control; tacrolimus: 50 +- 3%) calbindin-D28k (CsA: 62 +- 4%; tacrolimus: 43 +- 3%), and vitamin D receptor (CsA: 52 +- 3%; tacrolimus: 58 +- 2%, all p < 0.05). Tacrolimus 78-88 transient receptor potential cation channel subfamily V member 5 Homo sapiens 45-50 21691056-6 2011 CsA and tacrolimus reduced mRNA abundance in TRPV5 (CsA: 64 +- 3% of control; tacrolimus: 50 +- 3%) calbindin-D28k (CsA: 62 +- 4%; tacrolimus: 43 +- 3%), and vitamin D receptor (CsA: 52 +- 3%; tacrolimus: 58 +- 2%, all p < 0.05). Tacrolimus 78-88 transient receptor potential cation channel subfamily V member 5 Homo sapiens 45-50 21691056-8 2011 The immunofluorescence staining study demonstrated a 50% reduction of TRPV5 and calbindin-D28k by CsA and tacrolimus. Tacrolimus 106-116 transient receptor potential cation channel subfamily V member 5 Homo sapiens 70-75 21437550-0 2011 Tacrolimus 0,1% ointment in the treatment of vitiligo: a series of cases. Tacrolimus 0-10 VAMAS6 Homo sapiens 45-53 21437550-2 2011 The objective of this study was to evaluate the response to tacrolimus 0,1% ointment for vitiligo treatment. Tacrolimus 60-70 VAMAS6 Homo sapiens 89-97 21860784-8 2011 The pharmacological treatment does not differ from nonthymoma MG, except for tacrolimus which is an option in difficult thymoma and nonthymoma MG cases with RyR antibodies. Tacrolimus 77-87 ryanodine receptor 1 Homo sapiens 157-160 22130237-0 2011 Tacrolimus ameliorates dextran sulfate sodium-induced colitis in mice: implication of interferon-gamma and interleukin-1beta suppression. Tacrolimus 0-10 interferon gamma Mus musculus 86-102 22130237-0 2011 Tacrolimus ameliorates dextran sulfate sodium-induced colitis in mice: implication of interferon-gamma and interleukin-1beta suppression. Tacrolimus 0-10 interleukin 1 beta Mus musculus 107-124 22130237-5 2011 Treatment with tacrolimus for 14 d reduced the colon weight per unit length and suppressed the release of IFN-gamma and IL-1beta, but not other cytokines, in inflamed colons of colitic mice compared with vehicle-treated mice. Tacrolimus 15-25 interferon gamma Mus musculus 106-115 22130237-5 2011 Treatment with tacrolimus for 14 d reduced the colon weight per unit length and suppressed the release of IFN-gamma and IL-1beta, but not other cytokines, in inflamed colons of colitic mice compared with vehicle-treated mice. Tacrolimus 15-25 interleukin 1 beta Mus musculus 120-128 22130237-7 2011 The release of IFN-gamma and IL-1beta was also suppressed after single dosing with tacrolimus to colitic mice. Tacrolimus 83-93 interferon gamma Mus musculus 15-24 22130237-7 2011 The release of IFN-gamma and IL-1beta was also suppressed after single dosing with tacrolimus to colitic mice. Tacrolimus 83-93 interleukin 1 beta Mus musculus 29-37 22130237-8 2011 Taken together, these results suggested that tacrolimus ameliorated DSS-induced colitis by suppressing release of IFN-gamma and IL-1beta from inflamed colon. Tacrolimus 45-55 interferon gamma Mus musculus 114-123 22130237-8 2011 Taken together, these results suggested that tacrolimus ameliorated DSS-induced colitis by suppressing release of IFN-gamma and IL-1beta from inflamed colon. Tacrolimus 45-55 interleukin 1 beta Mus musculus 128-136 21358177-10 2011 Tacrolimus and cyclosporine increased fibronectin and TGF-beta expression and matrix deposition. Tacrolimus 0-10 fibronectin 1 Rattus norvegicus 38-49 21358177-10 2011 Tacrolimus and cyclosporine increased fibronectin and TGF-beta expression and matrix deposition. Tacrolimus 0-10 transforming growth factor, beta 1 Rattus norvegicus 54-62 21436775-0 2011 Polymorphisms in CYP3A5*3 and MDR1, and haplotype modulate response to plasma levels of tacrolimus in Chinese renal transplant patients. Tacrolimus 88-98 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 17-23 21436775-0 2011 Polymorphisms in CYP3A5*3 and MDR1, and haplotype modulate response to plasma levels of tacrolimus in Chinese renal transplant patients. Tacrolimus 88-98 ATP binding cassette subfamily B member 1 Homo sapiens 30-34 21436775-1 2011 BACKGROUND: The purpose of this study was to investigate the effects of polymorphisms in CYP3A5*3, CYP3AP1, and MDR1, and of haplotype, on plasma levels of tacrolimus in Chinese patients after renal transplantation, and to assess the relationship between polymorphisms and the variability of concentration/dose of tacrolimus for optimization and individualization regimens. Tacrolimus 156-166 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 89-95 21436775-1 2011 BACKGROUND: The purpose of this study was to investigate the effects of polymorphisms in CYP3A5*3, CYP3AP1, and MDR1, and of haplotype, on plasma levels of tacrolimus in Chinese patients after renal transplantation, and to assess the relationship between polymorphisms and the variability of concentration/dose of tacrolimus for optimization and individualization regimens. Tacrolimus 156-166 cytochrome P450 family 3 subfamily A member 51, pseudogene Homo sapiens 99-106 21436775-1 2011 BACKGROUND: The purpose of this study was to investigate the effects of polymorphisms in CYP3A5*3, CYP3AP1, and MDR1, and of haplotype, on plasma levels of tacrolimus in Chinese patients after renal transplantation, and to assess the relationship between polymorphisms and the variability of concentration/dose of tacrolimus for optimization and individualization regimens. Tacrolimus 156-166 ATP binding cassette subfamily B member 1 Homo sapiens 112-116 21436775-2 2011 MATERIAL/METHODS: The MALDI-TOF method was used to detect the genotype of CYP3A5*3, CYP3AP1, and MDR-1 in kidney transplant recipients (n=63) receiving tacrolimus. Tacrolimus 152-162 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 74-80 21436775-6 2011 RESULTS: We found that tacrolimus dose-adjusted C0 was larger in CYP3A5*3 and CYP3AP1 non-expressing renal transplant patients than in those who expressed the genes. Tacrolimus 23-33 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 65-71 21436775-6 2011 RESULTS: We found that tacrolimus dose-adjusted C0 was larger in CYP3A5*3 and CYP3AP1 non-expressing renal transplant patients than in those who expressed the genes. Tacrolimus 23-33 cytochrome P450 family 3 subfamily A member 51, pseudogene Homo sapiens 78-85 21436775-9 2011 CONCLUSIONS: The CYP3A5 genotype shows the most important association with tacrolimus concentrations. Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 17-23 21163525-1 2011 Immunophilins are receptors for immunosuppressive drugs such as the macrolides cyclosporin A (CsA) and FK506; correspondingly these immunophilins are referred to as cyclophilins and FK506-binding proteins (FKBPs). Tacrolimus 103-108 peptidylprolyl isomerase F (cyclophilin F) Mus musculus 165-177 21163525-5 2011 Furthermore, acute treatment with CsA to inhibit CypD modulation of mitochondrial Ca(2+) buffering, or with FK506 to inhibit FKBP12 interaction with inositol-trisphosphate receptor of the endoplasmic reticulum, led to similar reductive effects on astrocytic Ca(cyt)(2+) dynamics, but also to an enhanced Ca(2+)-dependent exocytotic release of glutamate in wild-type astrocytes. Tacrolimus 108-113 FK506 binding protein 1a Mus musculus 125-131 23213600-7 2011 We suspect CYP2C19*2 (poor metaboliser) genotype status and concomitant treatment with lansoprazole, tacrolimus, and antiretroviral (ARV) medications resulted in hepatic decompensation. Tacrolimus 101-111 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 11-18 21669047-12 2011 These data suggest that a combined, systemically delivered immunosuppression regimen including FK506 and mycophenolate can significantly improve survival of human spinal stem cells after intraspinal transplantation in SOD1 (G93A) rats. Tacrolimus 96-101 superoxide dismutase 1 Homo sapiens 219-223 21138364-0 2011 The impact of tacrolimus on vascular endothelial growth factor in experimental corneal neovascularization. Tacrolimus 14-24 vascular endothelial growth factor A Rattus norvegicus 28-62 20041908-0 2011 The role of CYP3A5 genotypes in dose requirements of tacrolimus and everolimus after heart transplantation. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 20041908-2 2011 A common variant of the CYP3A5 gene, CYP3A5*3, results in strongly decreased CYP3A5 activity and has been shown to influence Tacrolimus blood concentrations, but its role for the pharmacogenetics of Everolimus remains unclear. Tacrolimus 125-135 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-30 20041908-2 2011 A common variant of the CYP3A5 gene, CYP3A5*3, results in strongly decreased CYP3A5 activity and has been shown to influence Tacrolimus blood concentrations, but its role for the pharmacogenetics of Everolimus remains unclear. Tacrolimus 125-135 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 20041908-2 2011 A common variant of the CYP3A5 gene, CYP3A5*3, results in strongly decreased CYP3A5 activity and has been shown to influence Tacrolimus blood concentrations, but its role for the pharmacogenetics of Everolimus remains unclear. Tacrolimus 125-135 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 20041908-3 2011 Aim of the study was to examine the role of CYP3A5*3 variant in tacrolimus and everolimus dose and drug levels after heart transplantation. Tacrolimus 64-74 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 44-50 20041908-7 2011 Average Tacrolimus dose was significantly higher in subjects expressing CYP3A5 compared to non-expressors. Tacrolimus 8-18 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 72-78 20041908-11 2011 DISCUSSION: We conclude that in adult patients after heart transplantation, CYP3A5 genotypes have a strong influence on Tacrolimus, but not Everolimus dose requirement. Tacrolimus 120-130 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 76-82 21138364-1 2011 PURPOSE: To investigate the impact of tacrolimus on vascular endothelial growth factor (VEGF) in experimental corneal neovascularization (NV) immunohistochemically. Tacrolimus 38-48 vascular endothelial growth factor A Rattus norvegicus 52-86 21138364-1 2011 PURPOSE: To investigate the impact of tacrolimus on vascular endothelial growth factor (VEGF) in experimental corneal neovascularization (NV) immunohistochemically. Tacrolimus 38-48 vascular endothelial growth factor A Rattus norvegicus 88-92 21138364-11 2011 The mean intensity of the epithelial VEGF immunostaining of the intraperitoneally tacrolimus-treated group was less than that of its sham group (p = 0.002), while the mean intensity of the stromal VEGF staining of the topically tacrolimus-treated group was lesser than that of its sham group (p = 0.042). Tacrolimus 82-92 vascular endothelial growth factor A Rattus norvegicus 37-41 21138364-11 2011 The mean intensity of the epithelial VEGF immunostaining of the intraperitoneally tacrolimus-treated group was less than that of its sham group (p = 0.002), while the mean intensity of the stromal VEGF staining of the topically tacrolimus-treated group was lesser than that of its sham group (p = 0.042). Tacrolimus 82-92 vascular endothelial growth factor A Rattus norvegicus 197-201 21138364-12 2011 The intensities of the endothelial VEGF immunostaining of the intraperitoneally and topically tacrolimus-treated groups were less than those of the sham groups (p = 0.038, p = 0.032). Tacrolimus 94-104 vascular endothelial growth factor A Rattus norvegicus 35-39 21138364-13 2011 CONCLUSION: Systemic and topical administration of tacrolimus may be beneficial in the prevention of corneal NV because of its effect on VEGF. Tacrolimus 51-61 vascular endothelial growth factor A Rattus norvegicus 137-141 21980965-2 2011 This was due to atazanavir and ritonavir therapy for her HIV inhibiting the CYP3A4 hepatic enzyme resulting in accumulation of tacrolimus. Tacrolimus 127-137 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 76-82 21466465-4 2011 In this quest, efforts were made to determine three-dimensional structures of Plasmodium falciparum and Plasmodium vivax FK506 binding proteins which bind the macrolides (FK506 and rapamycin) and also demonstrate peptidylprolyl cis-trans isomerase activity in a similar manner as human FKBP12. Tacrolimus 121-126 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 286-292 21466465-4 2011 In this quest, efforts were made to determine three-dimensional structures of Plasmodium falciparum and Plasmodium vivax FK506 binding proteins which bind the macrolides (FK506 and rapamycin) and also demonstrate peptidylprolyl cis-trans isomerase activity in a similar manner as human FKBP12. Tacrolimus 171-176 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 286-292 21318106-8 2011 LS 180 cell line study indicated that Zhi Shi increased the efflux activity of P-gp, enabling us to explain the decreased oral bioavailability of tacrolimus caused by Zhi Shi. Tacrolimus 146-156 ATP binding cassette subfamily B member 1 Homo sapiens 79-83 21182091-6 2011 Intradermal injection of a blocking antibody against IL-23 as well as treatment with the immunosuppressant FK506 reversed these skin phenotypes, which was accompanied by suppression of the IL-22-producing T-cell infiltration. Tacrolimus 107-112 interleukin 22 Mus musculus 189-194 21227887-0 2011 Effects of tacrolimus on IFN-gamma signaling in keratinocytes: possible mechanisms by which tacrolimus affects IFN-gamma-dependent skin inflammation. Tacrolimus 11-21 interferon gamma Homo sapiens 25-34 21227887-0 2011 Effects of tacrolimus on IFN-gamma signaling in keratinocytes: possible mechanisms by which tacrolimus affects IFN-gamma-dependent skin inflammation. Tacrolimus 92-102 interferon gamma Homo sapiens 25-34 21227887-0 2011 Effects of tacrolimus on IFN-gamma signaling in keratinocytes: possible mechanisms by which tacrolimus affects IFN-gamma-dependent skin inflammation. Tacrolimus 92-102 interferon gamma Homo sapiens 111-120 21227887-3 2011 This study explored the mechanism of tacrolimus-modulated IFN-gamma signal transduction in HaCaT keratinocytes and the effects of tacrolimus on IFN-gamma-associated cytokine production in HaCaT cells. Tacrolimus 37-47 interferon gamma Homo sapiens 58-67 21227887-3 2011 This study explored the mechanism of tacrolimus-modulated IFN-gamma signal transduction in HaCaT keratinocytes and the effects of tacrolimus on IFN-gamma-associated cytokine production in HaCaT cells. Tacrolimus 130-140 interferon gamma Homo sapiens 144-153 21227887-4 2011 Tacrolimus down-regulated the recombinant human IFN-gamma (rhIFN-gamma)-induced expression of IFN-gamma receptor alpha (IFN-gammaRalpha). Tacrolimus 0-10 interferon gamma Homo sapiens 48-57 21227887-4 2011 Tacrolimus down-regulated the recombinant human IFN-gamma (rhIFN-gamma)-induced expression of IFN-gamma receptor alpha (IFN-gammaRalpha). Tacrolimus 0-10 interferon gamma Homo sapiens 61-70 21227887-5 2011 The IFN-gamma induced expression of phosphorylated Janus kinase 2 (pJAK2) and phosphorylated signal transducer and activator of transcription-1 (pSTAT-1) was also inhibited by tacrolimus. Tacrolimus 176-186 interferon gamma Homo sapiens 4-13 21227887-5 2011 The IFN-gamma induced expression of phosphorylated Janus kinase 2 (pJAK2) and phosphorylated signal transducer and activator of transcription-1 (pSTAT-1) was also inhibited by tacrolimus. Tacrolimus 176-186 Janus kinase 2 Homo sapiens 51-65 21227887-5 2011 The IFN-gamma induced expression of phosphorylated Janus kinase 2 (pJAK2) and phosphorylated signal transducer and activator of transcription-1 (pSTAT-1) was also inhibited by tacrolimus. Tacrolimus 176-186 signal transducer and activator of transcription 1 Homo sapiens 93-143 21227887-6 2011 Tacrolimus up-regulated the IFN-gamma-induced expression of suppressor of cytokine signaling-1 (SOCS-1). Tacrolimus 0-10 interferon gamma Homo sapiens 28-37 21227887-6 2011 Tacrolimus up-regulated the IFN-gamma-induced expression of suppressor of cytokine signaling-1 (SOCS-1). Tacrolimus 0-10 suppressor of cytokine signaling 1 Homo sapiens 60-94 21227887-6 2011 Tacrolimus up-regulated the IFN-gamma-induced expression of suppressor of cytokine signaling-1 (SOCS-1). Tacrolimus 0-10 suppressor of cytokine signaling 1 Homo sapiens 96-102 21227887-7 2011 Tacrolimus was also demonstrated to down-regulate IFN-gamma-induced the secretion of chemotactic factor CXCL-8 and the expression of intercellular adhesion molecule-1 and human leucocyte antigen HLA-DR. Tacrolimus 0-10 interferon gamma Homo sapiens 50-59 21227887-7 2011 Tacrolimus was also demonstrated to down-regulate IFN-gamma-induced the secretion of chemotactic factor CXCL-8 and the expression of intercellular adhesion molecule-1 and human leucocyte antigen HLA-DR. Tacrolimus 0-10 C-X-C motif chemokine ligand 8 Homo sapiens 104-110 21227887-7 2011 Tacrolimus was also demonstrated to down-regulate IFN-gamma-induced the secretion of chemotactic factor CXCL-8 and the expression of intercellular adhesion molecule-1 and human leucocyte antigen HLA-DR. Tacrolimus 0-10 intercellular adhesion molecule 1 Homo sapiens 133-166 21227887-8 2011 The findings in this work indicate that the direct effects of tacrolimus on IFN-gamma signaling in keratinocytes may contribute to its therapeutic efficacy as a topical ointment in the treatment of IFN-gamma-dependent skin inflammation. Tacrolimus 62-72 interferon gamma Homo sapiens 76-85 20932935-3 2011 Heat shock protein 56 (HSP56) belongs to the family of FK506-binding immunophilin proteins and is found in steroid receptor complexes, notably the glucocorticoid receptor. Tacrolimus 55-60 nuclear receptor subfamily 3, group C, member 1 Mus musculus 147-170 21227887-8 2011 The findings in this work indicate that the direct effects of tacrolimus on IFN-gamma signaling in keratinocytes may contribute to its therapeutic efficacy as a topical ointment in the treatment of IFN-gamma-dependent skin inflammation. Tacrolimus 62-72 interferon gamma Homo sapiens 198-207 20629603-0 2011 Impact of ATP-binding cassette, subfamily B, member 1 pharmacogenetics on tacrolimus-associated nephrotoxicity and dosage requirements in paediatric patients with liver transplant. Tacrolimus 74-84 ATP binding cassette subfamily B member 1 Homo sapiens 10-53 20629603-3 2011 Studies examining interindividual differences in the expression of the ABCB1 (ATP-binding cassette, subfamily B, member 1) gene (which encodes the drug transporter, P-gp) and its genetic polymorphisms have attempted to elucidate variations in tacrolimus response and disposition in children. Tacrolimus 243-253 ATP binding cassette subfamily B member 1 Homo sapiens 71-76 20629603-3 2011 Studies examining interindividual differences in the expression of the ABCB1 (ATP-binding cassette, subfamily B, member 1) gene (which encodes the drug transporter, P-gp) and its genetic polymorphisms have attempted to elucidate variations in tacrolimus response and disposition in children. Tacrolimus 243-253 ATP binding cassette subfamily B member 1 Homo sapiens 78-121 20629603-3 2011 Studies examining interindividual differences in the expression of the ABCB1 (ATP-binding cassette, subfamily B, member 1) gene (which encodes the drug transporter, P-gp) and its genetic polymorphisms have attempted to elucidate variations in tacrolimus response and disposition in children. Tacrolimus 243-253 phosphoglycolate phosphatase Homo sapiens 165-169 20629603-4 2011 AREAS COVERED IN THIS REVIEW: This review explores pharmacogenetic knowledge developed over the last decade regarding the impact of ABCB1 polymorphisms on tacrolimus toxicity and dosage requirements in children. Tacrolimus 155-165 ATP binding cassette subfamily B member 1 Homo sapiens 132-137 20629603-5 2011 WHAT THE READER WILL GAIN: A better understanding of the role of ABCB1 genetic polymorphisms (and corresponding haplotypes) and ABCB1 expression levels in various tissues and organs on tacrolimus outcomes in children with liver transplant. Tacrolimus 185-195 ATP binding cassette subfamily B member 1 Homo sapiens 65-70 20629603-5 2011 WHAT THE READER WILL GAIN: A better understanding of the role of ABCB1 genetic polymorphisms (and corresponding haplotypes) and ABCB1 expression levels in various tissues and organs on tacrolimus outcomes in children with liver transplant. Tacrolimus 185-195 ATP binding cassette subfamily B member 1 Homo sapiens 128-133 22353651-2 2011 METHODS: Tacrolimus was administered if the effect of biologics was unsatisfactory for 24 weeks at least in terms of laboratory data or DAS28 level: ESR, CRP level and DAS28 level were not below 15 mm/h, 0.2 mg/dl or 2.6, respectively. Tacrolimus 9-19 C-reactive protein Homo sapiens 154-157 22211008-1 2011 Single nucleotide polymorphisms in CYP3A5 (A6986G) and MDR-1 (C3435T) genes have been shown to be associated with the pharmacokinetics of tacrolimus in case of renal transplant recipients. Tacrolimus 138-148 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 35-41 22211008-1 2011 Single nucleotide polymorphisms in CYP3A5 (A6986G) and MDR-1 (C3435T) genes have been shown to be associated with the pharmacokinetics of tacrolimus in case of renal transplant recipients. Tacrolimus 138-148 ATP binding cassette subfamily B member 1 Homo sapiens 55-60 21850191-10 2011 Pre-treatment with tacrolimus markedly protected RGC-5 cells from NMDA-induced neurotoxicity, and then both spontaneous RGC death and degenerative changes to the optic nerve in p50-deficient mice were significantly reduced by the chronic administration of tacrolimus. Tacrolimus 19-29 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 177-180 21869564-4 2011 The general CaM kinase (CaMK) inhibitor KN-93 and CaM-dependent phosphatase calcineurin inhibitor FK-506 also inhibited s-RANKL-induced osteoclastogenesis. Tacrolimus 98-104 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 122-127 21617754-5 2011 FK506 achieves these properties through interaction with FK506 binding proteins (FKBP), including FK506 binding protein 51 (FKBP51). Tacrolimus 0-5 FK506 binding protein 5 Mus musculus 98-122 21617754-5 2011 FK506 achieves these properties through interaction with FK506 binding proteins (FKBP), including FK506 binding protein 51 (FKBP51). Tacrolimus 0-5 FK506 binding protein 5 Mus musculus 124-130 21617754-14 2011 FK506 increased FKBP51, NF-kappaB p65, and levels of activated NF-kappaB p65 protein. Tacrolimus 0-5 FK506 binding protein 5 Mus musculus 16-22 19815232-0 2011 Allograft inflammatory factor-1 is up-regulated in warm and cold ischemia-reperfusion injury in rat liver and may be inhibited by FK506. Tacrolimus 130-135 allograft inflammatory factor 1 Rattus norvegicus 0-31 19815232-5 2011 The potential inhibitory effect of FK506 on AIF-1 in a rat macrophage cell line and in these three models was also assessed. Tacrolimus 35-40 allograft inflammatory factor 1 Rattus norvegicus 44-49 19815232-7 2011 Real-time RT-PCR and Western blotting showed that pretreatment with low-dose FK506 partially inhibited AIF-1 activation as well as its inducers (IFN-gamma and IL-1beta) in these three models. Tacrolimus 77-82 allograft inflammatory factor 1 Rattus norvegicus 103-108 19815232-7 2011 Real-time RT-PCR and Western blotting showed that pretreatment with low-dose FK506 partially inhibited AIF-1 activation as well as its inducers (IFN-gamma and IL-1beta) in these three models. Tacrolimus 77-82 interferon gamma Rattus norvegicus 145-154 19815232-7 2011 Real-time RT-PCR and Western blotting showed that pretreatment with low-dose FK506 partially inhibited AIF-1 activation as well as its inducers (IFN-gamma and IL-1beta) in these three models. Tacrolimus 77-82 interleukin 1 beta Rattus norvegicus 159-167 19815232-10 2011 CONCLUSION: AIF-1 was activated in warm and cold IR injury, and pretreatment with low-dose FK506 partly inhibited AIF-1 activation and reduced warm and cold IR injury. Tacrolimus 91-96 allograft inflammatory factor 1 Rattus norvegicus 12-17 19815232-10 2011 CONCLUSION: AIF-1 was activated in warm and cold IR injury, and pretreatment with low-dose FK506 partly inhibited AIF-1 activation and reduced warm and cold IR injury. Tacrolimus 91-96 allograft inflammatory factor 1 Rattus norvegicus 114-119 21617754-14 2011 FK506 increased FKBP51, NF-kappaB p65, and levels of activated NF-kappaB p65 protein. Tacrolimus 0-5 v-rel reticuloendotheliosis viral oncogene homolog A (avian) Mus musculus 34-37 21617754-14 2011 FK506 increased FKBP51, NF-kappaB p65, and levels of activated NF-kappaB p65 protein. Tacrolimus 0-5 v-rel reticuloendotheliosis viral oncogene homolog A (avian) Mus musculus 73-76 21617754-17 2011 Both FK506 and FKBP51 appear to act through activation of NF-kappaB p65 protein, suggesting a common pathway for neuroprotection. Tacrolimus 5-10 v-rel reticuloendotheliosis viral oncogene homolog A (avian) Mus musculus 68-71 21850191-10 2011 Pre-treatment with tacrolimus markedly protected RGC-5 cells from NMDA-induced neurotoxicity, and then both spontaneous RGC death and degenerative changes to the optic nerve in p50-deficient mice were significantly reduced by the chronic administration of tacrolimus. Tacrolimus 256-266 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 177-180 21850191-12 2011 CONCLUSIONS: Research findings show that the chronic administration of tacrolimus significantly reduces spontaneous optic neuropathy in p50-deficient mice. Tacrolimus 71-81 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 136-139 21857094-0 2011 Association of transcription factor 7-like 2 (TCF7L2) gene polymorphism with posttransplant diabetes mellitus in kidney transplant patients medicated with tacrolimus. Tacrolimus 155-165 transcription factor 7 like 2 Homo sapiens 15-44 21857094-0 2011 Association of transcription factor 7-like 2 (TCF7L2) gene polymorphism with posttransplant diabetes mellitus in kidney transplant patients medicated with tacrolimus. Tacrolimus 155-165 transcription factor 7 like 2 Homo sapiens 46-52 21857094-6 2011 Therefore, the present study aimed to evaluate TCF7L2 polymorphisms in PTDM in kidney transplant patients medicated with tacrolimus. Tacrolimus 121-131 transcription factor 7 like 2 Homo sapiens 47-53 21857094-12 2011 If the application of TCF7L2 rs7903146 SNPs as a marker for PTDM is confirmed by further independent studies, replacing tacrolimus with other immunosuppressants could be warranted in patients at high risk of PTDM, as diagnosed by TCF7L2 genotyping. Tacrolimus 120-130 transcription factor 7 like 2 Homo sapiens 22-28 22110582-0 2011 Association of MDR1 gene SNPs and haplotypes with the tacrolimus dose requirements in Han Chinese liver transplant recipients. Tacrolimus 54-64 ATP binding cassette subfamily B member 1 Homo sapiens 15-19 22110582-7 2011 Tacrolimus C/D ratios of liver transplant recipients varied significantly among different haplotype groups of MDR1 gene. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 110-114 21076384-0 2010 CYP3A5 *1 allele: impacts on early acute rejection and graft function in tacrolimus-based renal transplant recipients. Tacrolimus 73-83 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 21673995-0 2011 The role of calcineurin/NFAT in SFRP2 induced angiogenesis--a rationale for breast cancer treatment with the calcineurin inhibitor tacrolimus. Tacrolimus 131-141 secreted frizzled related protein 2 Homo sapiens 32-37 21673995-2 2011 The FK506-FKBP12 complex associates with calcineurin and inhibits its phosphatase activity, resulting in inhibition of nuclear translocation of nuclear factor of activated T-cells (NFAT). Tacrolimus 4-9 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 10-16 21673995-4 2011 Since both VEGF and SFRP2 are expressed in breast carcinomas, we hypothesized that tacrolimus would inhibit breast carcinoma growth. Tacrolimus 83-93 vascular endothelial growth factor A Homo sapiens 11-15 21673995-4 2011 Since both VEGF and SFRP2 are expressed in breast carcinomas, we hypothesized that tacrolimus would inhibit breast carcinoma growth. Tacrolimus 83-93 secreted frizzled related protein 2 Homo sapiens 20-25 21673995-8 2011 Tacrolimus (1 microM) inhibited SFRP2 induced endothelial tube formation by 71% (p = 0.005) and inhibited VEGF induced endothelial tube formation by 67% (p = 0.004). Tacrolimus 0-10 secreted frizzled related protein 2 Homo sapiens 32-37 21673995-8 2011 Tacrolimus (1 microM) inhibited SFRP2 induced endothelial tube formation by 71% (p = 0.005) and inhibited VEGF induced endothelial tube formation by 67% (p = 0.004). Tacrolimus 0-10 vascular endothelial growth factor A Homo sapiens 106-110 21076384-2 2010 The effects of genetic polymorphisms of cytochrome P450 3A (CYP3A) 5 and Adenosine triphosphate-binding cassette subfamily B member 1 (ABCB1) genes on the achievement of target tacrolimus trough levels and clinical outcomes in renal transplants were evaluated. Tacrolimus 177-187 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 60-68 21076384-2 2010 The effects of genetic polymorphisms of cytochrome P450 3A (CYP3A) 5 and Adenosine triphosphate-binding cassette subfamily B member 1 (ABCB1) genes on the achievement of target tacrolimus trough levels and clinical outcomes in renal transplants were evaluated. Tacrolimus 177-187 ATP binding cassette subfamily B member 1 Homo sapiens 73-133 21076384-2 2010 The effects of genetic polymorphisms of cytochrome P450 3A (CYP3A) 5 and Adenosine triphosphate-binding cassette subfamily B member 1 (ABCB1) genes on the achievement of target tacrolimus trough levels and clinical outcomes in renal transplants were evaluated. Tacrolimus 177-187 ATP binding cassette subfamily B member 1 Homo sapiens 135-140 21076384-7 2010 RESULTS: Those subjects expressing CYP3A5 (n=29) evidenced significantly lower tacrolimus trough levels between days 1 and 5 after transplantation, when compared with nonexpressers (n=33). Tacrolimus 79-89 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 35-41 21076384-13 2010 CONCLUSION: We confirmed the significant effects of CYP3A5 polymorphism on the achievement of target tacrolimus trough levels and the development of acute rejection in early period after transplantation and consequent renal allograft function. Tacrolimus 101-111 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 52-58 20848491-8 2010 Tacrolimus suppressed LPS-induced activation of both NF-kappaB and MAPK in macrophages and induced apoptosis of macrophages via activation of caspases 3 and 9. Tacrolimus 0-10 caspase 3 Mus musculus 142-158 20840480-3 2010 Biopsy-proven acute rejection rate at 24 weeks (primary endpoint, per-protocol analysis) was 15.8% for Tacrolimus BID versus 20.4% for Tacrolimus QD (p = 0.182; treatment difference 4.5%, 95% confidence interval-1.8%, 10.9%, just outside the prespecified 10% noninferiority margin). Tacrolimus 103-113 BH3 interacting domain death agonist Homo sapiens 114-117 20840480-4 2010 Kaplan-Meier 12-month patient and graft survival rates were 97.5% and 92.8% for Tacrolimus BID and 96.9% and 91.5% for QD. Tacrolimus 80-90 BH3 interacting domain death agonist Homo sapiens 91-94 20943662-8 2010 We found that tacrolimus decreased Akt phosphorylation, suggesting that calcineurin could regulate replication and survival via the PI3K/Akt pathway. Tacrolimus 14-24 AKT serine/threonine kinase 1 Homo sapiens 35-38 20943662-8 2010 We found that tacrolimus decreased Akt phosphorylation, suggesting that calcineurin could regulate replication and survival via the PI3K/Akt pathway. Tacrolimus 14-24 AKT serine/threonine kinase 1 Homo sapiens 137-140 20943662-12 2010 Ex-4 restored Irs2 expression in tacrolimus-treated rodent and human islets nearly to baseline. Tacrolimus 33-43 insulin receptor substrate 2 Homo sapiens 14-18 20848491-9 2010 Rectal administration of tacrolimus evoked apoptosis of colonic macrophages in IL-10-KO mice. Tacrolimus 25-35 interleukin 10 Mus musculus 79-84 20848491-10 2010 Moreover, the rectal administration of tacrolimus ameliorated colitis in IL-10-KO mice and DSS-induced colitis in CB.17/SCID mice. Tacrolimus 39-49 interleukin 10 Mus musculus 73-78 21507867-2 2010 SUMMARY: Drug interactions with tacrolimus due to its metabolism through the CYP 450 3A4 enzymatic pathway have led to several reports of altered tacrolimus levels, which can lead to acute rejection in renal transplant recipients. Tacrolimus 32-42 peptidylprolyl isomerase G Homo sapiens 77-80 20671192-5 2010 Likewise, FK506 caused profound inhibition of NK cell proliferation in vitro and suppressed NK cytotoxicity and cytokine secretion in response to IL-2. Tacrolimus 10-15 interleukin 2 Homo sapiens 146-150 20671192-7 2010 Furthermore, FK506 specifically inhibited expression of NKG2D, CD48, and DNAM1 receptors without affecting that of 2B4, NKp30, NKp44, and NKp46. Tacrolimus 13-18 killer cell lectin like receptor K1 Homo sapiens 56-61 20671192-7 2010 Furthermore, FK506 specifically inhibited expression of NKG2D, CD48, and DNAM1 receptors without affecting that of 2B4, NKp30, NKp44, and NKp46. Tacrolimus 13-18 CD48 molecule Homo sapiens 63-67 20671192-7 2010 Furthermore, FK506 specifically inhibited expression of NKG2D, CD48, and DNAM1 receptors without affecting that of 2B4, NKp30, NKp44, and NKp46. Tacrolimus 13-18 CD226 molecule Homo sapiens 73-78 20671192-9 2010 Finally, FK506-treated NK cells showed impaired IL-2R signaling and inhibition of STAT3. Tacrolimus 9-14 interleukin 2 receptor subunit alpha Homo sapiens 48-53 20671192-9 2010 Finally, FK506-treated NK cells showed impaired IL-2R signaling and inhibition of STAT3. Tacrolimus 9-14 signal transducer and activator of transcription 3 Homo sapiens 82-87 21507867-2 2010 SUMMARY: Drug interactions with tacrolimus due to its metabolism through the CYP 450 3A4 enzymatic pathway have led to several reports of altered tacrolimus levels, which can lead to acute rejection in renal transplant recipients. Tacrolimus 146-156 peptidylprolyl isomerase G Homo sapiens 77-80 21040278-5 2010 For all organ types, increased standard deviation in intrapatient tacrolimus blood levels was an independent risk factor for late rejection (OR 1.6 [CI 1.1-2.1]; p = 0.02). Tacrolimus 66-76 olfactory receptor family 10 subfamily K member 1 Homo sapiens 141-147 21182472-4 2010 Of note, rapamycin and FK506 bind to FKBP12, and the resulting complexes interfere with distinct intracellular signaling pathways driven, respectively, by the mammalian target of rapamycin and calcineurin phosphatase. Tacrolimus 23-28 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 37-43 20951600-8 2010 Rp-8-Br-cGMPS, nicotinamide, RRed, BAPTA, CsA or FK506 paralled the cbl9 and atrbohD/F mutants to abolish the 8-Br-cGMP response. Tacrolimus 49-54 calcineurin B-like protein 9 Arabidopsis thaliana 68-72 20864901-0 2010 A systematic review of the effect of CYP3A5 genotype on the apparent oral clearance of tacrolimus in renal transplant recipients. Tacrolimus 87-97 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 20864901-5 2010 An association has been established between CYP3A5 genotype (expressors versus nonexpressors) and tacrolimus dose requirements to achieve target concentrations. Tacrolimus 98-108 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 44-50 20864901-7 2010 The focus of this review was to use a systematic method to investigate whether the CYP3A5 genotype has an effect on the apparent oral clearance of tacrolimus in renal transplant recipients. Tacrolimus 147-157 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 83-89 21068335-7 2010 Importantly, pretreatment with FK506 and overexpression of the GluR1 mutants, S845D (phospho-mimic) or S845A (phospho-blocking) attenuated the morphine-induced GluR1 endocytosis. Tacrolimus 31-36 glutamate ionotropic receptor AMPA type subunit 1 Homo sapiens 160-165 20876828-8 2010 Tacrolimus, an immunosuppressant, is also a substrate of CYP3A and P-GP. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 57-62 20876828-8 2010 Tacrolimus, an immunosuppressant, is also a substrate of CYP3A and P-GP. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 67-71 21182472-4 2010 Of note, rapamycin and FK506 bind to FKBP12, and the resulting complexes interfere with distinct intracellular signaling pathways driven, respectively, by the mammalian target of rapamycin and calcineurin phosphatase. Tacrolimus 23-28 mechanistic target of rapamycin kinase Homo sapiens 159-188 21182472-9 2010 Interestingly, derivatives of FK506 that have the same FKBP12-binding properties as FK506 but lack functional immunosuppressant activity, exert the same apoptotic effect as FK506 in chronic lymphocytic leukemia.These findings suggest that a direct FKBP inhibition represents a further mechanism of immunosuppressants." Tacrolimus 30-35 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 55-61 20030680-8 2010 In the FK506 patient group (n = 33), CYP3A5 A6986G (p < 0.001), and ABCB1 C1236T (p = 0.002) were associated with neurotoxicity. Tacrolimus 7-12 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 21104927-7 2010 Simulations indicated that for a compound with greater metabolism by CYP3A5 than CYP3A4, such as tacrolimus, incorporation of the correlation between CYP3A4 and CYP3A5 does have an impact on the prediction of oral clearance. Tacrolimus 97-107 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 69-75 21104927-7 2010 Simulations indicated that for a compound with greater metabolism by CYP3A5 than CYP3A4, such as tacrolimus, incorporation of the correlation between CYP3A4 and CYP3A5 does have an impact on the prediction of oral clearance. Tacrolimus 97-107 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-87 21104927-7 2010 Simulations indicated that for a compound with greater metabolism by CYP3A5 than CYP3A4, such as tacrolimus, incorporation of the correlation between CYP3A4 and CYP3A5 does have an impact on the prediction of oral clearance. Tacrolimus 97-107 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 150-156 21104927-7 2010 Simulations indicated that for a compound with greater metabolism by CYP3A5 than CYP3A4, such as tacrolimus, incorporation of the correlation between CYP3A4 and CYP3A5 does have an impact on the prediction of oral clearance. Tacrolimus 97-107 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 161-167 20980962-10 2010 RESULTS: In patients treated with tacrolimus, we observed a higher eGFR (all formulae), a greater 24-hour creatinine clearance, and lower serum cholesterol, LDL-cholesterol and triglyceride values than in patients treated with cyclosporine. Tacrolimus 34-44 epidermal growth factor receptor Homo sapiens 67-71 20980962-13 2010 The prevalence of CKD (eGFR <60 ml/min) in the tacrolimus-treated patients was 45%, whereas in the cyclosporine treated group it was 87% (p<0.01). Tacrolimus 50-60 epidermal growth factor receptor Homo sapiens 23-27 21118736-17 2010 In homozygous CYP3A5*3 carriers (n = 252), mean (SD) tacrolimus dose requirements remained significantly lower during DGF, while in CYP3A5*1 carriers with DGF (n = 52), lower mean dose requirements were observed only after postoperative day 4. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 20826153-3 2010 Finally, the effects of glucocorticoids, cyclosporine, and tacrolimus on eotaxin production by corneal fibroblasts were assessed. Tacrolimus 59-69 C-C motif chemokine ligand 11 Homo sapiens 73-80 21094856-1 2010 Tacrolimus (Tac, FK506) is a widely used T-cell-targeted immunosuppression drug known as a calcineurin inhibitor. Tacrolimus 0-10 calcineurin binding protein 1 Mus musculus 91-112 21094796-0 2010 Influence of CYP3A5 and MDR1(ABCB1) polymorphisms on the pharmacokinetics of tacrolimus in Chinese renal transplant recipients. Tacrolimus 77-87 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 21094856-1 2010 Tacrolimus (Tac, FK506) is a widely used T-cell-targeted immunosuppression drug known as a calcineurin inhibitor. Tacrolimus 17-22 calcineurin binding protein 1 Mus musculus 91-112 21094796-0 2010 Influence of CYP3A5 and MDR1(ABCB1) polymorphisms on the pharmacokinetics of tacrolimus in Chinese renal transplant recipients. Tacrolimus 77-87 ATP binding cassette subfamily B member 1 Homo sapiens 24-28 21094796-0 2010 Influence of CYP3A5 and MDR1(ABCB1) polymorphisms on the pharmacokinetics of tacrolimus in Chinese renal transplant recipients. Tacrolimus 77-87 ATP binding cassette subfamily B member 1 Homo sapiens 29-34 21094796-1 2010 The aims of this study were to investigate the influence of CYP3A5 and MDR1 genetic polymorphisms on tacrolimus pharmacokinetics in Chinese renal transplant recipients, so as to help rational administration in clinical practice. Tacrolimus 101-111 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 60-66 21094796-1 2010 The aims of this study were to investigate the influence of CYP3A5 and MDR1 genetic polymorphisms on tacrolimus pharmacokinetics in Chinese renal transplant recipients, so as to help rational administration in clinical practice. Tacrolimus 101-111 ATP binding cassette subfamily B member 1 Homo sapiens 71-75 21094796-7 2010 Renal transplant recipients who were CYP3A5*1 carriers required a higher dose of tacrolimus than CYP3A5*3/*3, indicating a significantly lower dose-adjusted AUC(0-12) of tacrolimus. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 21094796-7 2010 Renal transplant recipients who were CYP3A5*1 carriers required a higher dose of tacrolimus than CYP3A5*3/*3, indicating a significantly lower dose-adjusted AUC(0-12) of tacrolimus. Tacrolimus 170-180 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 20818834-13 2010 The apparent clearance of tacrolimus was 2-fold higher in expressers (with the CYP3A5*1/*1 and CYP3A5*1/*3 genotypes) than in non-expressers (with the CYP3A5*3/*3 genotype). Tacrolimus 26-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 79-85 21094796-7 2010 Renal transplant recipients who were CYP3A5*1 carriers required a higher dose of tacrolimus than CYP3A5*3/*3, indicating a significantly lower dose-adjusted AUC(0-12) of tacrolimus. Tacrolimus 170-180 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 97-103 21094797-0 2010 Value of CYP3A5 genotyping on determining initial dosages of tacrolimus for Chinese renal transplant recipients. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 9-15 21094797-10 2010 In conclusion, CYP3A5 polymorphism plays an important role in influencing tacrolimus blood levels. Tacrolimus 74-84 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 21094797-11 2010 Initial tacrolimus dosage selection based on CYP3A5 genotyping can improve drug blood levels in the early stage following renal transplantation. Tacrolimus 8-18 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 21094806-1 2010 OBJECTIVE: The present study was designed to investigate the effect of low-dose calcineurin inhibitor (CNI) tacrolimus combined with a mammalian target of rapamycin (mTOR) inhibitor on renal function in transplant recipients without allograft nephropathy. Tacrolimus 108-118 calcineurin binding protein 1 Homo sapiens 80-101 21094806-1 2010 OBJECTIVE: The present study was designed to investigate the effect of low-dose calcineurin inhibitor (CNI) tacrolimus combined with a mammalian target of rapamycin (mTOR) inhibitor on renal function in transplant recipients without allograft nephropathy. Tacrolimus 108-118 calcineurin binding protein 1 Homo sapiens 103-106 20674565-2 2010 Tacrolimus forms a complex with FK506 binding protein (FKBP), and Cys A forms a complex with cyclophilin. Tacrolimus 0-10 FK506 binding protein 1a Mus musculus 32-53 20674565-2 2010 Tacrolimus forms a complex with FK506 binding protein (FKBP), and Cys A forms a complex with cyclophilin. Tacrolimus 0-10 FK506 binding protein 1a Mus musculus 55-59 19880819-13 2010 Tacrolimus AER was superior to TAC MED at preventing AE IFN-gamma, IL-10, IL-13, monocyte chemoattractant protein-1 chemokine (C-C motif) ligand 5 (RANTES) and TNF-alpha up-regulation. Tacrolimus 0-10 interferon gamma Homo sapiens 56-65 19880819-13 2010 Tacrolimus AER was superior to TAC MED at preventing AE IFN-gamma, IL-10, IL-13, monocyte chemoattractant protein-1 chemokine (C-C motif) ligand 5 (RANTES) and TNF-alpha up-regulation. Tacrolimus 0-10 interleukin 10 Homo sapiens 67-72 19880819-13 2010 Tacrolimus AER was superior to TAC MED at preventing AE IFN-gamma, IL-10, IL-13, monocyte chemoattractant protein-1 chemokine (C-C motif) ligand 5 (RANTES) and TNF-alpha up-regulation. Tacrolimus 0-10 interleukin 13 Homo sapiens 74-79 19880819-13 2010 Tacrolimus AER was superior to TAC MED at preventing AE IFN-gamma, IL-10, IL-13, monocyte chemoattractant protein-1 chemokine (C-C motif) ligand 5 (RANTES) and TNF-alpha up-regulation. Tacrolimus 0-10 C-C motif chemokine ligand 5 Homo sapiens 148-154 19880819-13 2010 Tacrolimus AER was superior to TAC MED at preventing AE IFN-gamma, IL-10, IL-13, monocyte chemoattractant protein-1 chemokine (C-C motif) ligand 5 (RANTES) and TNF-alpha up-regulation. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 160-169 20840481-3 2010 The primary endpoint, event rate of biopsy-proven acute rejection (BPAR) at 24 weeks, was 33.7% for tacrolimus BID versus 36.3% for tacrolimus QD (Per-protocol set; p = 0.512; treatment difference 2.6%, 95% confidence interval -7.3%, 12.4%), falling within the predefined 15% noninferiority margin. Tacrolimus 100-110 BH3 interacting domain death agonist Homo sapiens 111-114 20840481-5 2010 Twelve-month patient and graft survival was 90.8% and 85.6% for tacrolimus BID and 89.2% and 85.3% for tacrolimus QD. Tacrolimus 64-74 BH3 interacting domain death agonist Homo sapiens 75-78 20842569-11 2010 The calcineurin inhibitor (CNI) tacrolimus is being used as the cornerstone of maintenance therapy in lieu of cyclosporine in more and more centers. Tacrolimus 32-42 calcineurin binding protein 1 Homo sapiens 4-25 20818834-13 2010 The apparent clearance of tacrolimus was 2-fold higher in expressers (with the CYP3A5*1/*1 and CYP3A5*1/*3 genotypes) than in non-expressers (with the CYP3A5*3/*3 genotype). Tacrolimus 26-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 95-101 20818834-13 2010 The apparent clearance of tacrolimus was 2-fold higher in expressers (with the CYP3A5*1/*1 and CYP3A5*1/*3 genotypes) than in non-expressers (with the CYP3A5*3/*3 genotype). Tacrolimus 26-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 95-101 20818834-16 2010 CONCLUSIONS: Population pharmacokinetic analysis of once-daily tacrolimus in renal transplant recipients resulted in identification of the CYP3A5*1/*3 genotype as a significant covariate on the apparent clearance of tacrolimus, and the design of an accurate maximum a posteriori Bayesian estimator based on three blood concentration measurements and this covariate. Tacrolimus 63-73 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 139-145 20818834-16 2010 CONCLUSIONS: Population pharmacokinetic analysis of once-daily tacrolimus in renal transplant recipients resulted in identification of the CYP3A5*1/*3 genotype as a significant covariate on the apparent clearance of tacrolimus, and the design of an accurate maximum a posteriori Bayesian estimator based on three blood concentration measurements and this covariate. Tacrolimus 216-226 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 139-145 20726688-5 2010 Amongst the last is the calcineurin inhibitor (CNI) (cyclosporine A (CsA) and tacrolimus)-related nephrotoxicity. Tacrolimus 78-88 calcineurin binding protein 1 Homo sapiens 24-45 20970601-9 2010 Tacrolimus concentration/dose ratios in heterozygote patients for CYP3A5*3 genotypes was >120% lower than for the homozygote CYP3A5*3 genotype (0.65+-0.04 vs 1.45+-0.05; P<.0001). Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 66-72 20858726-4 2010 Upon treatment with Rap, a bipartite transactivator was reconstituted, and transcription of a genomic firefly luciferase reporter was activated in a concentration-dependent (K(d) = 2.3 nmol/L) and FK506-competitive (K(i) = 17.1 nmol/L) manner in cellulo, as well as in a temporal and specific manner in vivo. Tacrolimus 197-202 regulatory associated protein of MTOR, complex 1 Mus musculus 20-23 20567855-11 2010 Cyclosporine (versus tacrolimus) was independently associated with greater SBP-Z and DBP-Z (p=0.001). Tacrolimus 21-31 selenium binding protein 1 Homo sapiens 75-78 21047202-4 2010 RESULTS: Four polymorphisms in CYP3A5 and one polymorphism in CYP3A4 were identified to be significantly associated with tacrolimus stable dose (p < 8.46 x 10(-5)). Tacrolimus 121-131 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 31-37 21047202-4 2010 RESULTS: Four polymorphisms in CYP3A5 and one polymorphism in CYP3A4 were identified to be significantly associated with tacrolimus stable dose (p < 8.46 x 10(-5)). Tacrolimus 121-131 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 21047202-9 2010 Age, ethnicity and CYP3A inhibitor use could predict 30% of tacrolimus dosing variability. Tacrolimus 60-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-24 21047202-13 2010 CONCLUSION: CYP3A5 genotype is the most significant genetic factor that impacts tacrolimus dose among the genes studied. Tacrolimus 80-90 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 20970601-9 2010 Tacrolimus concentration/dose ratios in heterozygote patients for CYP3A5*3 genotypes was >120% lower than for the homozygote CYP3A5*3 genotype (0.65+-0.04 vs 1.45+-0.05; P<.0001). Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 128-134 20970601-11 2010 CONCLUSION: Intestinal absorption and metabolism of tacrolimus was significantly affected by the SNPs in the CYP3A5 and MDR1 genes, which may offer a useful tool to optimize tacrolimus dosing after renal transplantation. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 109-115 20970601-11 2010 CONCLUSION: Intestinal absorption and metabolism of tacrolimus was significantly affected by the SNPs in the CYP3A5 and MDR1 genes, which may offer a useful tool to optimize tacrolimus dosing after renal transplantation. Tacrolimus 52-62 ATP binding cassette subfamily B member 1 Homo sapiens 120-124 20970601-11 2010 CONCLUSION: Intestinal absorption and metabolism of tacrolimus was significantly affected by the SNPs in the CYP3A5 and MDR1 genes, which may offer a useful tool to optimize tacrolimus dosing after renal transplantation. Tacrolimus 174-184 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 109-115 20970601-11 2010 CONCLUSION: Intestinal absorption and metabolism of tacrolimus was significantly affected by the SNPs in the CYP3A5 and MDR1 genes, which may offer a useful tool to optimize tacrolimus dosing after renal transplantation. Tacrolimus 174-184 ATP binding cassette subfamily B member 1 Homo sapiens 120-124 20651238-5 2010 In this study, we successfully apply immunosuppressive drug treatments of anti-asialo GM1 and FK506 in Fah(-/-)Rag2(-/-) mice, resulting in significant liver xeno-repopulation from human hepatocytes and human fetal liver cells. Tacrolimus 94-99 fumarylacetoacetate hydrolase Mus musculus 103-106 20651238-5 2010 In this study, we successfully apply immunosuppressive drug treatments of anti-asialo GM1 and FK506 in Fah(-/-)Rag2(-/-) mice, resulting in significant liver xeno-repopulation from human hepatocytes and human fetal liver cells. Tacrolimus 94-99 recombination activating gene 2 Mus musculus 111-115 20678096-7 2010 Tacrolimus ointment, a TCI, is approved for patients aged 2 years and older. Tacrolimus 0-10 latexin Homo sapiens 23-26 20388639-6 2010 Addition of FK506, which dissociates FKBP12.6 from RyR2, to normal SR reduced the CaM-binding affinity. Tacrolimus 12-17 ryanodine receptor 2 Canis lupus familiaris 51-55 20519340-1 2010 The aim of this study was to investigate the potential of calcineurin inhibitors [cyclosporine A (CsA) and tacrolimus (Tac)] to inhibit cellular uptake of atorvastatin mediated by the liver-specific organic anion-transporting polypeptide 1B1 (OATP1B1) in vitro. Tacrolimus 107-117 solute carrier organic anion transporter family member 1B1 Homo sapiens 243-250 20519340-1 2010 The aim of this study was to investigate the potential of calcineurin inhibitors [cyclosporine A (CsA) and tacrolimus (Tac)] to inhibit cellular uptake of atorvastatin mediated by the liver-specific organic anion-transporting polypeptide 1B1 (OATP1B1) in vitro. Tacrolimus 119-122 solute carrier organic anion transporter family member 1B1 Homo sapiens 243-250 20812435-0 2010 Falsely elevated tacrolimus levels caused by immunoassay interference secondary to beta-galactosidase antibodies in an infected liver transplant recipient. Tacrolimus 17-27 galactosidase beta 1 Homo sapiens 83-101 20832577-3 2010 Both tacrolimus and micafungin are substrates of cytochrome P450 3A4 in vitro. Tacrolimus 5-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 49-68 20528886-6 2010 Interestingly, FoxP3(+)T cells were also induced when tacrolimus was present in the rATG cultures. Tacrolimus 54-64 forkhead box P3 Homo sapiens 15-20 20718999-5 2010 Tibolone is known to be a weak competitive inhibitor of CYP3A4, which is involved in tacrolimus metabolism. Tacrolimus 85-95 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 56-62 20568249-0 2010 Effect of oral itraconazole on the pharmacokinetics of tacrolimus in a hematopoietic stem cell transplant recipient with CYP3A5*3/*3. Tacrolimus 55-65 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 121-127 20528886-0 2010 The calcineurin inhibitor tacrolimus allows the induction of functional CD4CD25 regulatory T cells by rabbit anti-thymocyte globulins. Tacrolimus 26-36 calcineurin binding protein 1 Homo sapiens 4-25 20528886-8 2010 The rATG tacrolimus-induced CD25(+) T cells inhibited proliferative responses of alloantigen-stimulated effector T cells as vigorously as rATG-induced and natural CD4(+)CD25(+)FoxP3(+)CD127(-/low) T cells (67% +/- 18% versus 69% +/- 16% versus 45% +/- 20%, mean +/- standard error of the mean, respectively). Tacrolimus 9-19 interleukin 2 receptor subunit alpha Homo sapiens 28-32 20528886-8 2010 The rATG tacrolimus-induced CD25(+) T cells inhibited proliferative responses of alloantigen-stimulated effector T cells as vigorously as rATG-induced and natural CD4(+)CD25(+)FoxP3(+)CD127(-/low) T cells (67% +/- 18% versus 69% +/- 16% versus 45% +/- 20%, mean +/- standard error of the mean, respectively). Tacrolimus 9-19 CD4 molecule Homo sapiens 163-166 20528886-8 2010 The rATG tacrolimus-induced CD25(+) T cells inhibited proliferative responses of alloantigen-stimulated effector T cells as vigorously as rATG-induced and natural CD4(+)CD25(+)FoxP3(+)CD127(-/low) T cells (67% +/- 18% versus 69% +/- 16% versus 45% +/- 20%, mean +/- standard error of the mean, respectively). Tacrolimus 9-19 interleukin 2 receptor subunit alpha Homo sapiens 169-173 20528886-8 2010 The rATG tacrolimus-induced CD25(+) T cells inhibited proliferative responses of alloantigen-stimulated effector T cells as vigorously as rATG-induced and natural CD4(+)CD25(+)FoxP3(+)CD127(-/low) T cells (67% +/- 18% versus 69% +/- 16% versus 45% +/- 20%, mean +/- standard error of the mean, respectively). Tacrolimus 9-19 forkhead box P3 Homo sapiens 176-181 20528886-8 2010 The rATG tacrolimus-induced CD25(+) T cells inhibited proliferative responses of alloantigen-stimulated effector T cells as vigorously as rATG-induced and natural CD4(+)CD25(+)FoxP3(+)CD127(-/low) T cells (67% +/- 18% versus 69% +/- 16% versus 45% +/- 20%, mean +/- standard error of the mean, respectively). Tacrolimus 9-19 interleukin 7 receptor Homo sapiens 184-189 20434440-0 2010 The calcineurin inhibitor, FK506, does not alter glutamate transport in the ischaemic mouse retina. Tacrolimus 27-32 calcineurin binding protein 1 Mus musculus 4-25 20491945-5 2010 Inactivation of this pathway by deletions, or by FK506, caused hypersensitivity to the peptide, demonstrating the importance of this pathway to the defense of C. albicans against the MUC7 peptide. Tacrolimus 49-54 mucin 7, secreted Homo sapiens 183-187 20438729-9 2010 One of these, a CN feedback inhibitor Rcan1, was induced >50 fold during 1-8 hours course of pancreatic growth and strongly inhibited (>99%) by FK506. Tacrolimus 150-155 regulator of calcineurin 1 Mus musculus 38-43 20430366-2 2010 The present study was conducted to assess the association between IL-6 (-174 G/C) gene polymorphism and GO in renal transplant recipients under cyclosporine (CsA), tacrolimus (Tcr), or sirolimus (Sir)-based regimens. Tacrolimus 164-174 interleukin 6 Homo sapiens 66-70 20056734-0 2010 Taurine reduces FK506-induced generation of ROS and activation of JNK and Bax in Madin Darby canine kidney cells. Tacrolimus 16-21 BCL2 associated X, apoptosis regulator Canis lupus familiaris 74-77 20056734-7 2010 This increase in intracellular ROS promoted JNK and Bax activation, which increased FK506-induced MDCK cell death. Tacrolimus 84-89 BCL2 associated X, apoptosis regulator Canis lupus familiaris 52-55 20056734-8 2010 Taurine reduced the FK506-induced generation of ROS and activation of JNK and Bax. Tacrolimus 20-25 BCL2 associated X, apoptosis regulator Canis lupus familiaris 78-81 20214743-0 2010 Effect of combined treatment with FK506, FTY720, and ex vivo graft irradiation in rat small bowel transplantation: expression of mucosal addressin cell adhesion molecule-1. Tacrolimus 34-39 mucosal vascular addressin cell adhesion molecule 1 Rattus norvegicus 129-171 20214743-6 2010 SBT allografts treated by FK506 and FTY720 demonstrated less infiltration of CD4 positive cells, but the irradiation group did not show any effects on its expression. Tacrolimus 26-31 Cd4 molecule Rattus norvegicus 77-80 20214743-7 2010 In FK506- and FTY720-treated groups, MAdCAM-1 expression on the HEVs in PPs was up-regulated, and its expression on the ECVs in the LP was down-regulated compared with other allograft groups. Tacrolimus 3-8 mucosal vascular addressin cell adhesion molecule 1 Rattus norvegicus 37-45 20214743-9 2010 FK506 and FTY720 prevented the infiltration of CD4 positive cells, the down-regulation of MAdCAM-1 expression on HEVs in PPs, and the up-regulation of MAdCAM-1 expression on ECVs in LP during the early phase of SBT. Tacrolimus 0-5 Cd4 molecule Rattus norvegicus 47-50 20214743-9 2010 FK506 and FTY720 prevented the infiltration of CD4 positive cells, the down-regulation of MAdCAM-1 expression on HEVs in PPs, and the up-regulation of MAdCAM-1 expression on ECVs in LP during the early phase of SBT. Tacrolimus 0-5 mucosal vascular addressin cell adhesion molecule 1 Rattus norvegicus 90-98 20214743-9 2010 FK506 and FTY720 prevented the infiltration of CD4 positive cells, the down-regulation of MAdCAM-1 expression on HEVs in PPs, and the up-regulation of MAdCAM-1 expression on ECVs in LP during the early phase of SBT. Tacrolimus 0-5 mucosal vascular addressin cell adhesion molecule 1 Rattus norvegicus 151-159 20526235-10 2010 CONCLUSIONS: High early tacrolimus dose requirements, predominantly but not exclusively encountered in CYP3A5*1 expressers, are associated with the development of calcineurin inhibitor-related nephrotoxicity, especially in recipients who continue corticosteroid therapy. Tacrolimus 24-34 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 103-109 20526235-10 2010 CONCLUSIONS: High early tacrolimus dose requirements, predominantly but not exclusively encountered in CYP3A5*1 expressers, are associated with the development of calcineurin inhibitor-related nephrotoxicity, especially in recipients who continue corticosteroid therapy. Tacrolimus 24-34 calcineurin binding protein 1 Homo sapiens 163-184 20571464-1 2010 Chronic calcineurin inhibitor (CNI)-induced nephrotoxicity is associated with prolonged use of cyclosporine and tacrolimus and has been observed after all types of transplantation, as well as during treatment of autoimmune disease. Tacrolimus 112-122 calcineurin binding protein 1 Homo sapiens 8-29 20624462-8 2010 Additional immunosuppression with tacrolimus or deoxyspergualin after transplantation delayed post-transplant proliferation of effector memory CD8(+) T cells but did not promote chimerism. Tacrolimus 34-44 CD8a molecule Homo sapiens 143-146 20410216-8 2010 Drp1 dephosphorylation could be suppressed by cyclosporine A and FK506, two calcineurin inhibitors. Tacrolimus 65-70 collapsin response mediator protein 1 Rattus norvegicus 0-4 20410216-9 2010 Importantly, cyclosporine A and FK506 could also prevent mitochondrial fragmentation, Bax accumulation, cytochrome c release, and apoptosis following ATP depletion in RPTC. Tacrolimus 32-37 BCL2 associated X, apoptosis regulator Rattus norvegicus 86-89 20596503-0 2010 Investigation of clinical interaction between omeprazole and tacrolimus in CYP3A5 non-expressors, renal transplant recipients. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 75-81 20302583-4 2010 METHODS: We examined the impact of tacrolimus on substance P (SP) release in an in vitro model of cutaneous neurogenic inflammation. Tacrolimus 35-45 tachykinin precursor 1 Homo sapiens 49-60 20302583-5 2010 Because phosphorylation of TRPV1 (transient receptor potential subtype vanilloid 1) plays a role in the induction of pain, we investigated whether tacrolimus regulates the phosphorylation state of TRPV1. Tacrolimus 147-157 transient receptor potential cation channel subfamily V member 1 Homo sapiens 197-202 20302583-8 2010 Analysis of TRPV1 phosphorylation by Western blot confirmed the capacity of tacrolimus to favour phosphorylation. Tacrolimus 76-86 transient receptor potential cation channel subfamily V member 1 Homo sapiens 12-17 20302583-10 2010 CONCLUSIONS: The efficacy of tacrolimus in pruritus, as well as the sensory side-effects, could be explained by a direct effect on neurons through an effect on calcineurin, possibly by a desensitization of TRPV1 and calcium currents through the PIP(2) regulation pathway. Tacrolimus 29-39 transient receptor potential cation channel subfamily V member 1 Homo sapiens 206-211 20302583-10 2010 CONCLUSIONS: The efficacy of tacrolimus in pruritus, as well as the sensory side-effects, could be explained by a direct effect on neurons through an effect on calcineurin, possibly by a desensitization of TRPV1 and calcium currents through the PIP(2) regulation pathway. Tacrolimus 29-39 prolactin induced protein Homo sapiens 245-248 21072155-11 2010 However, median L/D ratio for tacrolimus was significantly higher in subjects with CYP3A5*3/*3 (n = 24) (P = 0.011) and MDR- 1 3435TT (n = 18) (P = 0.0122). Tacrolimus 30-40 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 83-89 21072155-11 2010 However, median L/D ratio for tacrolimus was significantly higher in subjects with CYP3A5*3/*3 (n = 24) (P = 0.011) and MDR- 1 3435TT (n = 18) (P = 0.0122). Tacrolimus 30-40 ATP binding cassette subfamily B member 1 Homo sapiens 120-126 21072155-12 2010 The findings from this study show that homozygous mutant patients for CYP3A5 and MDR-1 gene SNPs could be managed with lower tacrolimus dose to avoid nephrotoxicity. Tacrolimus 125-135 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 70-76 21072155-12 2010 The findings from this study show that homozygous mutant patients for CYP3A5 and MDR-1 gene SNPs could be managed with lower tacrolimus dose to avoid nephrotoxicity. Tacrolimus 125-135 ATP binding cassette subfamily B member 1 Homo sapiens 81-86 20413672-4 2010 First, CN inhibitors, cyclosporine A (CysA) and tacrolimus (FK506) inhibited the Ang II-induced elevation of CYP11B2 mRNA level. Tacrolimus 48-58 angiotensinogen Homo sapiens 81-87 20413672-4 2010 First, CN inhibitors, cyclosporine A (CysA) and tacrolimus (FK506) inhibited the Ang II-induced elevation of CYP11B2 mRNA level. Tacrolimus 48-58 cytochrome P450 family 11 subfamily B member 2 Homo sapiens 109-116 20413672-4 2010 First, CN inhibitors, cyclosporine A (CysA) and tacrolimus (FK506) inhibited the Ang II-induced elevation of CYP11B2 mRNA level. Tacrolimus 60-65 angiotensinogen Homo sapiens 81-87 20413672-4 2010 First, CN inhibitors, cyclosporine A (CysA) and tacrolimus (FK506) inhibited the Ang II-induced elevation of CYP11B2 mRNA level. Tacrolimus 60-65 cytochrome P450 family 11 subfamily B member 2 Homo sapiens 109-116 20458432-5 2010 Both low- and high-dose FK506 treatment significantly reduced systemic CD3+ and CD4+CD25+ T-cell populations, and showed similar suppression of FoxP3 regulatory T-cell populations. Tacrolimus 24-29 forkhead box P3 Mus musculus 144-149 20931787-3 2010 It was observed that besides CL1/F, the index of sensitivity for all of the other four parameters (V1/F, V2/F, CL2/F and k(a)) in tacrolimus PopPK model showed relatively high level and changed fast with the time passing. Tacrolimus 130-140 adhesion G protein-coupled receptor L1 Homo sapiens 29-32 20931787-3 2010 It was observed that besides CL1/F, the index of sensitivity for all of the other four parameters (V1/F, V2/F, CL2/F and k(a)) in tacrolimus PopPK model showed relatively high level and changed fast with the time passing. Tacrolimus 130-140 immunoglobulin kappa variable 1-5 Homo sapiens 99-109 20931787-3 2010 It was observed that besides CL1/F, the index of sensitivity for all of the other four parameters (V1/F, V2/F, CL2/F and k(a)) in tacrolimus PopPK model showed relatively high level and changed fast with the time passing. Tacrolimus 130-140 endogenous retrovirus group W member 5 Homo sapiens 111-114 20580688-4 2010 Mechanical stresses for 2weeks in vivo and for 24h in vitro significantly induced upregulation of calcineurin expression and hypertrophic responses, such as the increases in cardiomyocytes size and specific gene expressions, in cardiomyocytes of angiotensinogen gene knockout (ATG(-/-)) mice, both of which were significantly suppressed by a specific calcineurin inhibitor FK506, suggesting a critical role of calcineurin in mechanical stress-induced cardiac hypertrophy in the ATG(-/-) mice. Tacrolimus 373-378 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Mus musculus 246-261 20596503-1 2010 BACKGROUND: As proton pump inhibitors share CYP3A4 enzyme with tacrolimus for their hepatic elimination, they potentially affect its pharmacokinetics, most prominently in patients with CYP2C19 or CYP3A5 gene mutations. Tacrolimus 63-73 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 185-192 20596503-1 2010 BACKGROUND: As proton pump inhibitors share CYP3A4 enzyme with tacrolimus for their hepatic elimination, they potentially affect its pharmacokinetics, most prominently in patients with CYP2C19 or CYP3A5 gene mutations. Tacrolimus 63-73 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 196-202 20485320-0 2010 Dosing tacrolimus based on CYP3A5 genotype: will it improve clinical outcome? Tacrolimus 7-17 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 27-33 20494024-3 2010 Thrombin inhibition with hirudin, and calcineurin inhibition with tacrolimus have independently been shown to ameliorate lung ischemia-reperfusion injury by reducing activator protein-1 and nuclear factor kappa B activation, respectively. Tacrolimus 66-76 Jun proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 166-212 20393454-1 2010 Retrospective studies have demonstrated that patients who are expressors of cytochrome P4503A5 (CYP3A5) require a higher tacrolimus dose to achieve a therapeutic trough concentration (C(0)). Tacrolimus 121-131 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 76-94 20393454-1 2010 Retrospective studies have demonstrated that patients who are expressors of cytochrome P4503A5 (CYP3A5) require a higher tacrolimus dose to achieve a therapeutic trough concentration (C(0)). Tacrolimus 121-131 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 96-102 20207170-0 2010 Lung transplantation under a tacrolimus/mycophenolate mofetil-based immunosuppressive regimen results in low titers of HLA and MICA IgG antibodies which are not related to development of BOS. Tacrolimus 29-39 MHC class I polypeptide-related sequence A Homo sapiens 127-131 20082110-0 2010 Improvement of rapidly progressive lupus nephritis associated MPO-ANCA with tacrolimus. Tacrolimus 76-86 myeloperoxidase Homo sapiens 62-65 20082110-8 2010 Furthermore, we propose that tacrolimus is an effective immunosuppressant for MPO-ANCA-related renal crisis in diffuse proliferative lupus nephritis. Tacrolimus 29-39 myeloperoxidase Homo sapiens 78-81 21079895-7 2010 RESULTS: Rats receiving tacrolimus showed statistically significant higher levels of liver regeneration when compared to placebo according to Kwon"s formula, mitotic index and PCNA marker. Tacrolimus 24-34 proliferating cell nuclear antigen Rattus norvegicus 176-180 20226231-5 2010 Using cresyl violet staining, we also observed that an increased number of CA3 neurons survived in the 0.1 microM FK506 group compared to the KA only group. Tacrolimus 114-119 carbonic anhydrase 3 Homo sapiens 75-78 20226231-6 2010 Based on the results of the Western blot analysis, the expressions of 5-lipoxygenase and caspase-3 were reduced 24h after 0.1 microM FK506 treatment. Tacrolimus 133-138 arachidonate 5-lipoxygenase Homo sapiens 70-84 20226231-6 2010 Based on the results of the Western blot analysis, the expressions of 5-lipoxygenase and caspase-3 were reduced 24h after 0.1 microM FK506 treatment. Tacrolimus 133-138 caspase 3 Homo sapiens 89-98 20226231-7 2010 The levels of superoxide dismutase (SOD) and phospho-Akt expression were increased by treatment with 0.1 microM FK506. Tacrolimus 112-117 AKT serine/threonine kinase 1 Homo sapiens 53-56 20451249-6 2010 Dissociation studies demonstrated that application of cADPR resulted in significant removal of FKBP12.6 proteins from sarcoplasmic reticulum (SR) microsomes, and that treatment of the RyR2 immunoprecipitation complexes with cADPR or FK506 disrupted the interaction between RyR2 and FKBP12.6. Tacrolimus 233-238 ryanodine receptor 2, cardiac Mus musculus 184-188 20480792-4 2010 In this article, the authors review clinical trials using tacrolimus ointment, a topical calcineurin inhibitor, to treat adult and pediatric patients with AD over a wide range of disease severity focusing on its efficacy in rapidly reducing pruritus. Tacrolimus 58-68 calcineurin binding protein 1 Homo sapiens 89-110 20233105-1 2010 Tacrolimus is an immunosuppressive medication in the class of calcineurin inhibitors that acts by inhibiting T-cell and interleukin-2 activity, and is commonly used after allogeneic organ transplant. Tacrolimus 0-10 interleukin 2 Homo sapiens 120-133 20431509-2 2010 To date, the only strategy to have been tested in a clinical trial is the use of the cytochrome P450 3A5 (CYP3A5) genotype to predict tacrolimus dose. Tacrolimus 134-144 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 85-104 20431509-2 2010 To date, the only strategy to have been tested in a clinical trial is the use of the cytochrome P450 3A5 (CYP3A5) genotype to predict tacrolimus dose. Tacrolimus 134-144 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 106-112 20479955-1 2010 BACKGROUND: The aim of our study was to determine the impact of CYP3A5*1 and CYP3A5*3 on the kinetics of tacrolimus in renal transplant recipients. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 64-70 20479955-1 2010 BACKGROUND: The aim of our study was to determine the impact of CYP3A5*1 and CYP3A5*3 on the kinetics of tacrolimus in renal transplant recipients. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 77-83 20479955-12 2010 Carriers of CYP3A5*1 allele had lower predicted measures for tacrolimus dose adjusted concentration and higher predicted measures for volume of distribution. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 20479955-13 2010 CONCLUSION: We proved that CYP3A5*1 carriers need higher tacrolimus dose than CYP3A5*3 homozygotes to achieve the target blood concentration. Tacrolimus 57-67 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 27-33 20175769-1 2010 BACKGROUND: The calcineurin inhibitor tacrolimus and the anti-TNF-antibody infliximab are established options in steroid-refractory ulcerative colitis (UC). Tacrolimus 38-48 calcineurin binding protein 1 Homo sapiens 16-37 20394493-7 2010 CD1a, CD11b and CCR7 expression were all significantly down-regulated after topical treatment with tacrolimus. Tacrolimus 99-109 CD1a molecule Homo sapiens 0-4 20394493-7 2010 CD1a, CD11b and CCR7 expression were all significantly down-regulated after topical treatment with tacrolimus. Tacrolimus 99-109 integrin subunit alpha M Homo sapiens 6-11 20394493-7 2010 CD1a, CD11b and CCR7 expression were all significantly down-regulated after topical treatment with tacrolimus. Tacrolimus 99-109 C-C motif chemokine receptor 7 Homo sapiens 16-20 20202085-0 2010 Prolonged activation of ERK triggers glutamate-induced apoptosis of astrocytes: neuroprotective effect of FK506. Tacrolimus 106-111 mitogen-activated protein kinase 1 Homo sapiens 24-27 19965585-5 2010 We further demonstrate that calcineurin activity, the downstream target of Ca(2+) influx, was essential; inhibition of calcineurin activity by cyclosporine A or FK506 completely abolished apoA-I lipidation. Tacrolimus 161-166 apolipoprotein A1 Homo sapiens 188-194 20202085-5 2010 Treatment with UO126, inhibitor of MEK1, threo-beta-benzyloxyaspartic acid, glutamate transporter inhibitor, and FK506, a cytoprotective drug prevented ERK activation and glutamate-induced apoptosis. Tacrolimus 113-118 mitogen-activated protein kinase 1 Homo sapiens 152-155 20202085-8 2010 Global gene expression profiling in the cortex revealed that FK506 blocks middle cerebral artery occlusion-induced expression of numerous genes associated with ERK signaling pathway and apoptosis. Tacrolimus 61-66 mitogen-activated protein kinase 1 Homo sapiens 160-163 20383867-9 2010 However, patients receiving tacrolimus more strongly preferred to continue on this CNI than those receiving cyclosporine (67.4% vs. 44.4%, p=0.009), while more patients on cyclosporine wished to stop taking it (23.4 vs. 2.3%, p=0.004). Tacrolimus 28-38 5'-nucleotidase, cytosolic IA Homo sapiens 83-86 20091056-2 2010 We report a renal transplant recipient who suffered from severe nephrotoxicity related to a toxic tacrolimus trough concentration in both conditions, diarrhoea and CCB co-administration, and with genotyped CYP3A system and P-glycoprotein (P-gp) polymorphisms. Tacrolimus 98-108 ATP binding cassette subfamily B member 1 Homo sapiens 239-243 20091056-5 2010 It also highlights the key role in tacrolimus pharmacokinetics of the CYP3A system and P-gp polymorphisms, and their influence in high-risk situations when enzyme activity is already affected by enterocyte damage due to diarrhoea and CCB competition. Tacrolimus 35-45 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 70-75 20091056-5 2010 It also highlights the key role in tacrolimus pharmacokinetics of the CYP3A system and P-gp polymorphisms, and their influence in high-risk situations when enzyme activity is already affected by enterocyte damage due to diarrhoea and CCB competition. Tacrolimus 35-45 ATP binding cassette subfamily B member 1 Homo sapiens 87-91 20346916-4 2010 The magnitude of increase in cell ATP caused by inhibition of Pdr5p pump by compound 23.1 and the Pdr5p pump inhibitor FK506 used for comparison reflects the activity and hence the energy demand of the pump. Tacrolimus 119-124 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 98-103 20415563-0 2010 CYP3A5 and ABCB1 polymorphisms influence tacrolimus concentrations in peripheral blood mononuclear cells after renal transplantation. Tacrolimus 41-51 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 20415563-0 2010 CYP3A5 and ABCB1 polymorphisms influence tacrolimus concentrations in peripheral blood mononuclear cells after renal transplantation. Tacrolimus 41-51 ATP binding cassette subfamily B member 1 Homo sapiens 11-16 20415563-1 2010 AIMS: This prospective study investigated the effect of genetic polymorphisms in a biotransformation enzyme (CYP3A5) and a transporter protein (ABCB1) on tacrolimus (Tac) whole blood concentrations in renal transplantation, and more specifically on peripheral blood mononuclear cell (PBMC) drug concentrations, after renal transplantation. Tacrolimus 154-164 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 109-115 20415563-1 2010 AIMS: This prospective study investigated the effect of genetic polymorphisms in a biotransformation enzyme (CYP3A5) and a transporter protein (ABCB1) on tacrolimus (Tac) whole blood concentrations in renal transplantation, and more specifically on peripheral blood mononuclear cell (PBMC) drug concentrations, after renal transplantation. Tacrolimus 154-164 ATP binding cassette subfamily B member 1 Homo sapiens 144-149 20214406-9 2010 Despite a strong association between the CYP3A5 6986A>G SNP and tacrolimus pharmacokinetics, there is no consistent evidence of organ rejection as a result of genotype-related under-immunosuppression. Tacrolimus 67-77 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 41-47 19912168-10 2010 In human embryonic kidney 293 cells, however, only supratherapeutic tacrolimus concentrations (>0.1 mumol/l) inhibited hERG K(+) current with a maximum inhibition of 28% at 10 mumol/l, indicating that other mechanisms might have also operated besides direct block of ionic channel function. Tacrolimus 68-78 ETS transcription factor ERG Homo sapiens 122-126 20097278-5 2010 In the Caco-2 cell transport study, the transport ratio of FK506 with WZ extract was significantly lower than that of FK506 alone, which suggested WZ extract inhibited P-gp-mediated efflux of FK506. Tacrolimus 59-64 phosphoglycolate phosphatase Homo sapiens 168-172 20097278-5 2010 In the Caco-2 cell transport study, the transport ratio of FK506 with WZ extract was significantly lower than that of FK506 alone, which suggested WZ extract inhibited P-gp-mediated efflux of FK506. Tacrolimus 118-123 phosphoglycolate phosphatase Homo sapiens 168-172 20097278-5 2010 In the Caco-2 cell transport study, the transport ratio of FK506 with WZ extract was significantly lower than that of FK506 alone, which suggested WZ extract inhibited P-gp-mediated efflux of FK506. Tacrolimus 118-123 phosphoglycolate phosphatase Homo sapiens 168-172 20097278-6 2010 Furthermore, 100 microM of WZ extract almost completely inhibited FK506 metabolism in rat and human liver microsomes, indicating WZ extract potently inhibited the CYP3A-mediated metabolism of FK506. Tacrolimus 192-197 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 163-168 20097278-7 2010 In conclusion, WZ inhibited P-gp-mediated efflux and CYP3A-mediated metabolism of FK506, and the reduction of intestinal first-pass effect by WZ was the major cause of the increased FK506 oral bioavailability. Tacrolimus 82-87 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 53-58 20359353-0 2010 HMG-CoA reductase inhibitors in kidney transplant recipients receiving tacrolimus: statins not associated with improved patient or graft survival. Tacrolimus 71-81 3-hydroxy-3-methylglutaryl-CoA reductase Homo sapiens 0-17 20214406-18 2010 A large standardized clinical trial is now required to evaluate the relationship between the pharmacokinetics and pharmacodynamics of CYP3A5-mediated tacrolimus metabolism, particularly in regard to the outcomes of acute rejection and nephrotoxicity. Tacrolimus 150-160 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 134-140 20149694-6 2010 Further, FK506, an immunosuppressant, partially rescued the neuron death and attenuated the expression of NO, nNOS, iNOS, MDA and activated caspase-3. Tacrolimus 9-14 nitric oxide synthase 1 Rattus norvegicus 110-114 20373462-2 2010 Recent findings have suggested that the calcineurin inhibitor tacrolimus (Tac), unlike cyclosporine A (CsA), interferes with the antiviral activity of interferon-alpha (IFN-alpha) in vitro. Tacrolimus 62-72 calcineurin binding protein 1 Homo sapiens 40-61 20373462-2 2010 Recent findings have suggested that the calcineurin inhibitor tacrolimus (Tac), unlike cyclosporine A (CsA), interferes with the antiviral activity of interferon-alpha (IFN-alpha) in vitro. Tacrolimus 62-72 interferon alpha 1 Homo sapiens 169-178 20373462-2 2010 Recent findings have suggested that the calcineurin inhibitor tacrolimus (Tac), unlike cyclosporine A (CsA), interferes with the antiviral activity of interferon-alpha (IFN-alpha) in vitro. Tacrolimus 74-77 calcineurin binding protein 1 Homo sapiens 40-61 20373462-2 2010 Recent findings have suggested that the calcineurin inhibitor tacrolimus (Tac), unlike cyclosporine A (CsA), interferes with the antiviral activity of interferon-alpha (IFN-alpha) in vitro. Tacrolimus 74-77 interferon alpha 1 Homo sapiens 169-178 19941067-6 2010 FK-506, a calcineurin inhibitor caused AATYK hyperphosphorylation under high KCl conditions. Tacrolimus 0-6 apoptosis-associated tyrosine kinase Mus musculus 39-44 20216109-5 2010 Immunoabsorption studies showed that the cause of the interference was an endogenous antibody present in the patient"s plasma that recognized a unique epitope present on the antibody-enzyme (beta-galactosidase) conjugate used in the Siemens tacrolimus immunoassay but not on the antibody or beta-galactosidase alone. Tacrolimus 241-251 galactosidase beta 1 Homo sapiens 191-209 20216109-5 2010 Immunoabsorption studies showed that the cause of the interference was an endogenous antibody present in the patient"s plasma that recognized a unique epitope present on the antibody-enzyme (beta-galactosidase) conjugate used in the Siemens tacrolimus immunoassay but not on the antibody or beta-galactosidase alone. Tacrolimus 241-251 galactosidase beta 1 Homo sapiens 291-309 19752882-0 2010 Association of calpain-10 gene polymorphism and posttransplant diabetes mellitus in kidney transplant patients medicated with tacrolimus. Tacrolimus 126-136 calpain 10 Homo sapiens 15-25 19752882-4 2010 Therefore, the present study was aimed at evaluation of CAPN10 gene polymorphisms in PTDM in kidney transplant patients medicated with tacrolimus. Tacrolimus 135-145 calpain 10 Homo sapiens 56-62 20149694-6 2010 Further, FK506, an immunosuppressant, partially rescued the neuron death and attenuated the expression of NO, nNOS, iNOS, MDA and activated caspase-3. Tacrolimus 9-14 nitric oxide synthase 2 Rattus norvegicus 116-120 20149694-6 2010 Further, FK506, an immunosuppressant, partially rescued the neuron death and attenuated the expression of NO, nNOS, iNOS, MDA and activated caspase-3. Tacrolimus 9-14 caspase 3 Rattus norvegicus 140-149 20465949-12 2010 CONCLUSION: The administration of FK506 has dramatically neuroprotective effects against the development of ACR neuropathy, which may be related to up-regulating the expression of HSP70 and Bcl-2 with down-regulating the expression of Bax. Tacrolimus 34-39 BCL2 associated X, apoptosis regulator Rattus norvegicus 235-238 20121702-6 2010 Intriguingly, the Ca2+-induced expression of GLO1 was enhanced in the presence of FK506, a potent inhibitor of calcineurin. Tacrolimus 82-87 lactoylglutathione lyase GLO1 Saccharomyces cerevisiae S288C 45-49 20465949-0 2010 [Effects of tacrolimus (FK506) on heat-shock proteins 70, Bcl-2 and Bax expression in nervous tissue of acrylamide-induced rat]. Tacrolimus 12-22 BCL2 associated X, apoptosis regulator Rattus norvegicus 68-71 20465949-0 2010 [Effects of tacrolimus (FK506) on heat-shock proteins 70, Bcl-2 and Bax expression in nervous tissue of acrylamide-induced rat]. Tacrolimus 24-29 BCL2 associated X, apoptosis regulator Rattus norvegicus 68-71 20465949-7 2010 In the cerebrum and sciatic nerve pellet, the level of HSP70 in the FK506 groups increased by 11.6%, 33.3% and 56.3%, 58.5% (P < 0.01), but no significant changes existed in spinal cord. Tacrolimus 68-73 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 55-60 20465949-8 2010 The level of Bcl-2 in the sciatic nerve pellet increased by 39.1% (P < 0.01) but no significant changes existed in the cerebrum and spinal cord from low dose of FK506 group. Tacrolimus 164-169 BCL2, apoptosis regulator Rattus norvegicus 13-18 20465949-9 2010 And the level of Bax in the spinal cord pellet markedly increased by 46.8% but not in cerebrum and sciatic nerve pellet; Whereas in the tissues mentioned above, the levels of Bcl-2 were enhanced remarkably by 16.3%, 14.8% and 56.0% (P < 0.01) in the high dose of FK506 group. Tacrolimus 266-271 BCL2 associated X, apoptosis regulator Rattus norvegicus 17-20 20465949-9 2010 And the level of Bax in the spinal cord pellet markedly increased by 46.8% but not in cerebrum and sciatic nerve pellet; Whereas in the tissues mentioned above, the levels of Bcl-2 were enhanced remarkably by 16.3%, 14.8% and 56.0% (P < 0.01) in the high dose of FK506 group. Tacrolimus 266-271 BCL2, apoptosis regulator Rattus norvegicus 175-180 20465949-11 2010 The values of Bcl-2/Bax in low and high doses of FK506 groups clearly increased by 15.9%, 33.3%, 36.9% and 30.1%, 49.1%, 60.1% (P < 0.01). Tacrolimus 49-54 BCL2, apoptosis regulator Rattus norvegicus 14-19 20465949-11 2010 The values of Bcl-2/Bax in low and high doses of FK506 groups clearly increased by 15.9%, 33.3%, 36.9% and 30.1%, 49.1%, 60.1% (P < 0.01). Tacrolimus 49-54 BCL2 associated X, apoptosis regulator Rattus norvegicus 20-23 20465949-12 2010 CONCLUSION: The administration of FK506 has dramatically neuroprotective effects against the development of ACR neuropathy, which may be related to up-regulating the expression of HSP70 and Bcl-2 with down-regulating the expression of Bax. Tacrolimus 34-39 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 180-185 20465949-12 2010 CONCLUSION: The administration of FK506 has dramatically neuroprotective effects against the development of ACR neuropathy, which may be related to up-regulating the expression of HSP70 and Bcl-2 with down-regulating the expression of Bax. Tacrolimus 34-39 BCL2, apoptosis regulator Rattus norvegicus 190-195 20121702-10 2010 We also found that the levels of Msn2 and Msn4 proteins in Ca2+-treated cells decreased gradually and that FK506 blocked the degradation of Msn2/Msn4. Tacrolimus 107-112 stress-responsive transcriptional activator MSN2 Saccharomyces cerevisiae S288C 33-37 20121702-10 2010 We also found that the levels of Msn2 and Msn4 proteins in Ca2+-treated cells decreased gradually and that FK506 blocked the degradation of Msn2/Msn4. Tacrolimus 107-112 stress-responsive transcriptional activator MSN2 Saccharomyces cerevisiae S288C 140-144 20121702-10 2010 We also found that the levels of Msn2 and Msn4 proteins in Ca2+-treated cells decreased gradually and that FK506 blocked the degradation of Msn2/Msn4. Tacrolimus 107-112 stress-responsive transcriptional activator MSN4 Saccharomyces cerevisiae S288C 145-149 20121702-7 2010 Consequently, the Ca2+-induced expression of GLO1 in a mutant that is defective in calcineurin or Crz1, the sole transcription factor downstream of calcineurin, was much greater than that in the wild-type strain even without FK506. Tacrolimus 225-230 lactoylglutathione lyase GLO1 Saccharomyces cerevisiae S288C 45-49 20121702-9 2010 The level of Ca2+-induced expression of GLO1 reached a maximum in cells overexpressing MSN2 even when FK506 was not present, whereas in cells overexpressing CRZ1 the level was greatly reduced and increased markedly when FK506 was present. Tacrolimus 102-107 lactoylglutathione lyase GLO1 Saccharomyces cerevisiae S288C 40-44 20121702-9 2010 The level of Ca2+-induced expression of GLO1 reached a maximum in cells overexpressing MSN2 even when FK506 was not present, whereas in cells overexpressing CRZ1 the level was greatly reduced and increased markedly when FK506 was present. Tacrolimus 220-225 lactoylglutathione lyase GLO1 Saccharomyces cerevisiae S288C 40-44 20023700-7 2010 Under knockdown conditions, the inhibitory effects of TPR ligands, cyclosporine A (CsA) and FK506, on AR activity were not observed, indicating that Cyp40 and FKBP51 are the targets of CsA and FK506, respectively. Tacrolimus 92-97 androgen receptor Homo sapiens 102-104 20023700-0 2010 FKBP51 and Cyp40 are positive regulators of androgen-dependent prostate cancer cell growth and the targets of FK506 and cyclosporin A. Tacrolimus 110-115 peptidylprolyl isomerase D Homo sapiens 11-16 20023700-7 2010 Under knockdown conditions, the inhibitory effects of TPR ligands, cyclosporine A (CsA) and FK506, on AR activity were not observed, indicating that Cyp40 and FKBP51 are the targets of CsA and FK506, respectively. Tacrolimus 193-198 peptidylprolyl isomerase D Homo sapiens 149-154 20023700-8 2010 Our findings show that FKBP51 and Cyp40 are positive regulators of AR that can be selectively targeted by CsA and FK506 to achieve inhibition of androgen-induced cell proliferation. Tacrolimus 114-119 peptidylprolyl isomerase D Homo sapiens 34-39 20023700-8 2010 Our findings show that FKBP51 and Cyp40 are positive regulators of AR that can be selectively targeted by CsA and FK506 to achieve inhibition of androgen-induced cell proliferation. Tacrolimus 114-119 androgen receptor Homo sapiens 67-69 20169301-7 2010 Long-term treatment with topical tacrolimus or a corticosteroid regimen improves atopic dermatitis and recall antigen reactivity, suggesting an improvement in the Th1/Th2-balance. Tacrolimus 33-43 negative elongation factor complex member C/D Homo sapiens 163-166 20170205-10 2010 The CYP3A5 6986A>G SNP has a well established influence on the pharmacokinetics of tacrolimus. Tacrolimus 86-96 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 20170205-11 2010 Several studies in kidney, heart and liver transplant recipients have reported an approximate halving of tacrolimus dose-adjusted trough concentrations and doubling of tacrolimus dose requirements in heterozygous or homozygous carriers of a CYP3A5*1 wild-type allele compared with homozygous carriers of a CYP3A5*3 variant allele. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 241-247 20170205-11 2010 Several studies in kidney, heart and liver transplant recipients have reported an approximate halving of tacrolimus dose-adjusted trough concentrations and doubling of tacrolimus dose requirements in heterozygous or homozygous carriers of a CYP3A5*1 wild-type allele compared with homozygous carriers of a CYP3A5*3 variant allele. Tacrolimus 168-178 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 241-247 19659514-3 2010 We describe a case of a patient with alpha-1-antitrypsin deficiency who underwent single lung transplantation in 2005 and was maintained on tacrolimus, cytoxan and prednisone. Tacrolimus 140-150 serpin family A member 1 Homo sapiens 37-56 20170205-12 2010 Carriers of a CYP3A5*1 allele take a longer time to reach target blood tacrolimus concentrations. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 20164329-0 2010 Inhibition of FK506 binding proteins reduces alpha-synuclein aggregation and Parkinson"s disease-like pathology. Tacrolimus 14-19 synuclein, alpha Mus musculus 45-60 20199368-5 2010 The homeostasis model assessment of insulin sensitivity showed intermediate values for patients who underwent a simultaneous pancreas-kidney transplant and correlated with prednisone dosages (in those who underwent kidney transplant alone) and tacrolimus levels (in patients who underwent a simultaneous pancreas-kidney transplant). Tacrolimus 244-254 insulin Homo sapiens 36-43 19714763-10 2010 In agreement, immunohistochemistry demonstrated that tacrolimus inhibited production of the neutrophil chemoattractants CXCL1 and CXCL2. Tacrolimus 53-63 chemokine (C-X-C motif) ligand 1 Mus musculus 120-125 19714763-10 2010 In agreement, immunohistochemistry demonstrated that tacrolimus inhibited production of the neutrophil chemoattractants CXCL1 and CXCL2. Tacrolimus 53-63 chemokine (C-X-C motif) ligand 2 Mus musculus 130-135 20036165-11 2010 Cardiac allografts treated with tacrolimus showed decreased expression of both ASC and IL-1 beta similar to that seen in isografts. Tacrolimus 32-42 interleukin 1 beta Mus musculus 87-96 20080907-0 2010 Tacrolimus, a calcineurin inhibitor, overcomes treatment unresponsiveness mediated by P-glycoprotein on lymphocytes in refractory rheumatoid arthritis. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 14-35 20080907-0 2010 Tacrolimus, a calcineurin inhibitor, overcomes treatment unresponsiveness mediated by P-glycoprotein on lymphocytes in refractory rheumatoid arthritis. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 86-100 20080907-1 2010 OBJECTIVE: Tacrolimus, a calcineurin inhibitor, is used for treatment of rheumatoid arthritis (RA). Tacrolimus 11-21 calcineurin binding protein 1 Homo sapiens 25-46 20080907-10 2010 The response to tacrolimus correlated with P-glycoprotein expression and C/M ratio. Tacrolimus 16-26 ATP binding cassette subfamily B member 1 Homo sapiens 43-57 20080907-13 2010 CONCLUSION: Early efficacy of tacrolimus treatment depended on its inhibitory effect on the drug exclusion function of P-glycoprotein, leading to restoration of intracellular therapeutic levels of corticosteroids and clinical improvement. Tacrolimus 30-40 ATP binding cassette subfamily B member 1 Homo sapiens 119-133 19686443-6 2010 Young, female, non-black, children without ATN and acute rejection in the first 30 days, TX after 1995, those with better eGFR at day 30, and receiving tacrolimus had better long-term eGFR. Tacrolimus 152-162 epidermal growth factor receptor Homo sapiens 184-188 20164329-6 2010 FK506, a specific inhibitor of this family of proteins, inhibited alpha-SYN aggregation and neuronal cell death in this synucleinopathy model dose dependently. Tacrolimus 0-5 synuclein, alpha Mus musculus 66-75 20164329-8 2010 Thus, FK506 likely targets FKBP members in the cell culture model. Tacrolimus 6-11 FK506 binding protein 1a Mus musculus 27-31 20164329-9 2010 Furthermore, oral administration of FK506 after viral vector-mediated overexpression of alpha-SYN in adult mouse brain significantly reduced alpha-SYN aggregate formation and neuronal cell death. Tacrolimus 36-41 synuclein, alpha Mus musculus 88-97 20164329-9 2010 Furthermore, oral administration of FK506 after viral vector-mediated overexpression of alpha-SYN in adult mouse brain significantly reduced alpha-SYN aggregate formation and neuronal cell death. Tacrolimus 36-41 synuclein, alpha Mus musculus 141-150 20145524-0 2010 Insulin resistance indexes in renal transplant recipients maintained on tacrolimus immunosuppression. Tacrolimus 72-82 insulin Homo sapiens 0-7 20304212-1 2010 BACKGROUND: Mammalian target of rapamycin (mTOR) inhibitors behave as potent immunosuppressants, which have the advantages, with respect to calcineurin inhibitors (CNI; cyclosporine or tacrolimus), of no nephrotoxicity but inhibition of cell proliferation. Tacrolimus 185-195 mechanistic target of rapamycin kinase Homo sapiens 12-41 20304212-1 2010 BACKGROUND: Mammalian target of rapamycin (mTOR) inhibitors behave as potent immunosuppressants, which have the advantages, with respect to calcineurin inhibitors (CNI; cyclosporine or tacrolimus), of no nephrotoxicity but inhibition of cell proliferation. Tacrolimus 185-195 mechanistic target of rapamycin kinase Homo sapiens 43-47 20145524-12 2010 CONCLUSIONS: Insulin resistance indexes are valid in transplant recipients taking tacrolimus, with McAuley"s index the strongest surrogate. Tacrolimus 82-92 insulin Homo sapiens 13-20 20133804-6 2010 FKBP52 is a member of the FKBP (FK506-binding protein) family that comprises intracellular protein effectors of immunosuppressive drugs (such as FK506 and rapamycin). Tacrolimus 32-37 FKBP prolyl isomerase 4 Homo sapiens 0-6 20159692-4 2010 RESULTS: Compared with the control group, high-concentration FK506 (20 ng/ml) significantly inhibited the secretions of IL-2, IL-6, IL-12, IL-17, IFN-gamma, TNF-alpha, GM-CSF and G-CSF. Tacrolimus 61-66 interleukin 2 Homo sapiens 120-124 20197746-13 2010 Efficacy of calcineurin inhibitor combinations also vary based on the type used and UV light combined, with tacrolimus being more effective with excimer laser. Tacrolimus 108-118 calcineurin binding protein 1 Homo sapiens 12-33 20175845-0 2010 Efficacy and safety of topical tacrolimus for the treatment of face and neck vitiligo. Tacrolimus 31-41 VAMAS6 Homo sapiens 77-85 20175845-3 2010 The aim of the present study was to determine the efficacy and safety of topical tacrolimus as monotherapy for the treatment of face/neck vitiligo in Taiwan. Tacrolimus 81-91 VAMAS6 Homo sapiens 138-146 20175845-14 2010 Tacrolimus ointment is effective and well tolerated for the treatment of patients with vitiligo in Taiwan. Tacrolimus 0-10 VAMAS6 Homo sapiens 87-95 19786111-9 2010 We conclude that the combination of tacrolimus/sirolimus+/-MTX for GVHD prophylaxis in the setting of RIC HCT for MF appears to reduce the incidence of severe aGVHD and NRM, and leads to improved OS compared to CSA/MMF+/-MTX. Tacrolimus 36-46 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 211-214 20159692-4 2010 RESULTS: Compared with the control group, high-concentration FK506 (20 ng/ml) significantly inhibited the secretions of IL-2, IL-6, IL-12, IL-17, IFN-gamma, TNF-alpha, GM-CSF and G-CSF. Tacrolimus 61-66 tumor necrosis factor Homo sapiens 157-166 20159692-4 2010 RESULTS: Compared with the control group, high-concentration FK506 (20 ng/ml) significantly inhibited the secretions of IL-2, IL-6, IL-12, IL-17, IFN-gamma, TNF-alpha, GM-CSF and G-CSF. Tacrolimus 61-66 colony stimulating factor 2 Homo sapiens 168-174 20159692-4 2010 RESULTS: Compared with the control group, high-concentration FK506 (20 ng/ml) significantly inhibited the secretions of IL-2, IL-6, IL-12, IL-17, IFN-gamma, TNF-alpha, GM-CSF and G-CSF. Tacrolimus 61-66 colony stimulating factor 3 Homo sapiens 179-184 20159692-4 2010 RESULTS: Compared with the control group, high-concentration FK506 (20 ng/ml) significantly inhibited the secretions of IL-2, IL-6, IL-12, IL-17, IFN-gamma, TNF-alpha, GM-CSF and G-CSF. Tacrolimus 61-66 interleukin 6 Homo sapiens 126-130 20159692-5 2010 At a moderate concentration (5 ng/ml), FK506 inhibited the secretion of GM-CSF significantly. Tacrolimus 39-44 colony stimulating factor 2 Homo sapiens 72-78 20159692-4 2010 RESULTS: Compared with the control group, high-concentration FK506 (20 ng/ml) significantly inhibited the secretions of IL-2, IL-6, IL-12, IL-17, IFN-gamma, TNF-alpha, GM-CSF and G-CSF. Tacrolimus 61-66 interleukin 17A Homo sapiens 139-144 20159692-6 2010 CONCLUSION: FK506 effectively inhibits the secretion of proinflammatory cytokines including IL-6, IFN-gamma and TNF-alpha and also the secretion of IL-2, IL-12, IL-17, GM-CSF and G-CSF. Tacrolimus 12-17 interleukin 6 Homo sapiens 92-96 20159692-6 2010 CONCLUSION: FK506 effectively inhibits the secretion of proinflammatory cytokines including IL-6, IFN-gamma and TNF-alpha and also the secretion of IL-2, IL-12, IL-17, GM-CSF and G-CSF. Tacrolimus 12-17 interferon gamma Homo sapiens 98-107 20159692-4 2010 RESULTS: Compared with the control group, high-concentration FK506 (20 ng/ml) significantly inhibited the secretions of IL-2, IL-6, IL-12, IL-17, IFN-gamma, TNF-alpha, GM-CSF and G-CSF. Tacrolimus 61-66 interferon gamma Homo sapiens 146-155 20159692-6 2010 CONCLUSION: FK506 effectively inhibits the secretion of proinflammatory cytokines including IL-6, IFN-gamma and TNF-alpha and also the secretion of IL-2, IL-12, IL-17, GM-CSF and G-CSF. Tacrolimus 12-17 tumor necrosis factor Homo sapiens 112-121 20010459-0 2010 CYP3A5*3 genotype associated with intrasubject pharmacokinetic variation toward tacrolimus in bioequivalence study. Tacrolimus 80-90 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 20159692-6 2010 CONCLUSION: FK506 effectively inhibits the secretion of proinflammatory cytokines including IL-6, IFN-gamma and TNF-alpha and also the secretion of IL-2, IL-12, IL-17, GM-CSF and G-CSF. Tacrolimus 12-17 interleukin 2 Homo sapiens 148-152 20159692-6 2010 CONCLUSION: FK506 effectively inhibits the secretion of proinflammatory cytokines including IL-6, IFN-gamma and TNF-alpha and also the secretion of IL-2, IL-12, IL-17, GM-CSF and G-CSF. Tacrolimus 12-17 interleukin 17A Homo sapiens 161-166 20159692-6 2010 CONCLUSION: FK506 effectively inhibits the secretion of proinflammatory cytokines including IL-6, IFN-gamma and TNF-alpha and also the secretion of IL-2, IL-12, IL-17, GM-CSF and G-CSF. Tacrolimus 12-17 colony stimulating factor 2 Homo sapiens 168-174 20159692-6 2010 CONCLUSION: FK506 effectively inhibits the secretion of proinflammatory cytokines including IL-6, IFN-gamma and TNF-alpha and also the secretion of IL-2, IL-12, IL-17, GM-CSF and G-CSF. Tacrolimus 12-17 colony stimulating factor 3 Homo sapiens 179-184 20010459-6 2010 The estimated total sample size for the bioequivalence study of tacrolimus with a 2 x 2 cross-over design was increased by 93.3% for AUClast (n = 30 versus 58) and 121.4% for Cmax (n = 28 versus 62) in the CYP3A5*3/*3 group compared with the CYP3A5*1/*1+*1/*3 group. Tacrolimus 64-74 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 206-212 20010459-6 2010 The estimated total sample size for the bioequivalence study of tacrolimus with a 2 x 2 cross-over design was increased by 93.3% for AUClast (n = 30 versus 58) and 121.4% for Cmax (n = 28 versus 62) in the CYP3A5*3/*3 group compared with the CYP3A5*1/*1+*1/*3 group. Tacrolimus 64-74 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 242-248 20010459-7 2010 The intraindividual variability of tacrolimus PK parameters may be associated with the CYP3A5 genotype. Tacrolimus 35-45 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 87-93 19818345-10 2010 Treatment with tacrolimus before the final challenge completely inhibited the recovery of substance P content, whereas dexamethasone facilitated the recovery. Tacrolimus 15-25 tachykinin 1 Mus musculus 90-101 19818345-0 2010 Depletion of substance P, a mechanism for inhibition of mouse scratching behavior by tacrolimus. Tacrolimus 85-95 tachykinin 1 Mus musculus 13-24 19755159-4 2010 This study was conducted to investigate the inhibiting capability of RAPA or FK-506 against transferred alloreactive CD4(+) Tm cells in a mouse cardiac transplant model. Tacrolimus 77-83 CD4 antigen Mus musculus 117-120 19818345-12 2010 Therefore, substance P seems to participate in the induction of ear swelling and scratching behavior upon final challenge with DNFB, and depletion of substance P by tacrolimus in the dermis contributes to its inhibition of ear swelling and scratching behavior at least in part. Tacrolimus 165-175 tachykinin 1 Mus musculus 150-161 19818345-5 2010 Both tacrolimus and dexamethasone given topically for 10 days before the final challenge significantly inhibited the ear swelling and reduced the expression of interferon-gamma mRNA. Tacrolimus 5-15 interferon gamma Mus musculus 160-176 20084280-10 2010 Finally, we also found that FKBP52 formed stable complexes with APP through its FK506 interacting domain. Tacrolimus 80-85 FKBP prolyl isomerase 4 Homo sapiens 28-34 19682688-0 2010 Tacrolimus-eluting stent inhibits neointimal hyperplasia via calcineurin/NFAT signaling in porcine coronary artery model. Tacrolimus 0-10 nuclear factor of activated T-cells 5 Rattus norvegicus 73-77 20484893-8 2010 Statistically significant correlations between CD4+CD25(high)Foxp3+ Tregs and tacrolimus levels and CD4+CD25(high)Foxp3- Tregs and HLA-DR mismatching were detected. Tacrolimus 78-88 CD4 molecule Homo sapiens 47-50 20484893-8 2010 Statistically significant correlations between CD4+CD25(high)Foxp3+ Tregs and tacrolimus levels and CD4+CD25(high)Foxp3- Tregs and HLA-DR mismatching were detected. Tacrolimus 78-88 interleukin 2 receptor subunit alpha Homo sapiens 51-55 20484893-8 2010 Statistically significant correlations between CD4+CD25(high)Foxp3+ Tregs and tacrolimus levels and CD4+CD25(high)Foxp3- Tregs and HLA-DR mismatching were detected. Tacrolimus 78-88 forkhead box P3 Homo sapiens 61-66 19910577-9 2010 This effect of AT1 receptor blockade was mimicked by inhibition of calcineurin by FK506. Tacrolimus 82-87 angiotensin II receptor, type 1a Rattus norvegicus 15-18 20305320-1 2010 BACKGROUND: Induction of the hepatic and intestinal cytochrome P450-3A4 system and intestinal P-glycoprotein is an unavoidable consequence of rifampin administration which requires substantial increase in tacrolimus dose when given concurrently. Tacrolimus 205-215 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 52-71 20305320-1 2010 BACKGROUND: Induction of the hepatic and intestinal cytochrome P450-3A4 system and intestinal P-glycoprotein is an unavoidable consequence of rifampin administration which requires substantial increase in tacrolimus dose when given concurrently. Tacrolimus 205-215 ATP binding cassette subfamily B member 1 Homo sapiens 94-108 20305320-11 2010 CONCLUSIONS: We conclude that chronic diarrhea may cause toxic tacrolimus blood levels even in presence of rifampin, this would be due to its significant cytochrome P450-3A4 and P-glycoprotein enzyme inhibitory effect. Tacrolimus 63-73 ATP binding cassette subfamily B member 1 Homo sapiens 178-192 19682688-8 2010 Western blotting demonstrated that tacrolimus decreased the expression of calcineurin, NFATc4, and IL-2 of cultured VSMCs. Tacrolimus 35-45 nuclear factor of activated T-cells 4 Rattus norvegicus 87-93 19682688-8 2010 Western blotting demonstrated that tacrolimus decreased the expression of calcineurin, NFATc4, and IL-2 of cultured VSMCs. Tacrolimus 35-45 interleukin 2 Rattus norvegicus 99-103 20346231-8 2010 The serum complement hemolytic activity (CH50), complement C3 level, and CRP level were also significantly improved after treatment with tacrolimus (p<0.05). Tacrolimus 137-147 complement C3 Homo sapiens 48-61 19661110-1 2010 AIMS: Our objective was to explore the functional interdependence of protein kinase A (PKA) phosphorylation with binding of modulatory FK506 binding proteins (FKBP12/12.6) to the ryanodine receptor (RyR). Tacrolimus 135-140 peptidyl-prolyl cis-trans isomerase FKBP1A Oryctolagus cuniculus 159-165 19661110-1 2010 AIMS: Our objective was to explore the functional interdependence of protein kinase A (PKA) phosphorylation with binding of modulatory FK506 binding proteins (FKBP12/12.6) to the ryanodine receptor (RyR). Tacrolimus 135-140 LOC100009439 Oryctolagus cuniculus 179-197 19661110-1 2010 AIMS: Our objective was to explore the functional interdependence of protein kinase A (PKA) phosphorylation with binding of modulatory FK506 binding proteins (FKBP12/12.6) to the ryanodine receptor (RyR). Tacrolimus 135-140 LOC100009439 Oryctolagus cuniculus 199-202 19890249-4 2010 The pharmacokinetics of both MPA and MPAG were significantly influenced by the OATP1B3 polymorphism 334T>G/699G>A in 70 renal transplant patients receiving combination treatment of MMF with either tacrolimus or sirolimus, but not in 115 patients receiving MMF and cyclosporine. Tacrolimus 203-213 solute carrier organic anion transporter family member 1B3 Homo sapiens 79-86 20573295-9 2010 Among the observed immunosuppressive agents, corticosteroids, prednisolone, 6alpha-methylprednisolone, dexamethasone, and beclomethasone inhibited PS-PLA(1) expression with half-maximal inhibitory concentrations less than 3.0 nM, while methotrexate, cyclosporine A, tacrolimus, 6-mercaptopurine, and mycophenoic acid showed either a weak or moderate inhibition. Tacrolimus 266-276 phospholipase A1 member A Homo sapiens 147-156 20205663-3 2010 Patients carrying the CYP3A5*1 allele have an increased tacrolimus metabolism, hence lower drug exposure. Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 22-28 20847451-0 2010 FK506 reduces amyloid plaque burden and induces MMP-9 in AbetaPP/PS1 double transgenic mice. Tacrolimus 0-5 matrix metallopeptidase 9 Mus musculus 48-53 20357482-8 2010 The inhibitory effect became saturated (50-70% reduction) at concentrations higher than 10(-10)M. Cyclosporin A inhibited the release of TSLP by 50-60% at 10(-5) and 10(-4)M. FK506 had no effect at 10(-5)M or less, but almost completely inhibited the release of TSLP at 10(-4)M. No synergistic effect was obtained with a glucocorticoid plus either of the calcineurin inhibitors. Tacrolimus 175-180 thymic stromal lymphopoietin Homo sapiens 137-141 20357482-8 2010 The inhibitory effect became saturated (50-70% reduction) at concentrations higher than 10(-10)M. Cyclosporin A inhibited the release of TSLP by 50-60% at 10(-5) and 10(-4)M. FK506 had no effect at 10(-5)M or less, but almost completely inhibited the release of TSLP at 10(-4)M. No synergistic effect was obtained with a glucocorticoid plus either of the calcineurin inhibitors. Tacrolimus 175-180 thymic stromal lymphopoietin Homo sapiens 262-266 20847451-0 2010 FK506 reduces amyloid plaque burden and induces MMP-9 in AbetaPP/PS1 double transgenic mice. Tacrolimus 0-5 amyloid beta (A4) precursor protein Mus musculus 57-64 20847451-0 2010 FK506 reduces amyloid plaque burden and induces MMP-9 in AbetaPP/PS1 double transgenic mice. Tacrolimus 0-5 presenilin 1 Mus musculus 65-68 20847451-4 2010 Treatment with FK506, a CaN inhibitor, significantly reduces Abeta burden and restores synaptic proteins (synaptophysin and postsynaptic density protein-95; PSD-95) while inducing matrix metallopeptidase-9 (MMP-9) expression in GFAP-positive astrocytes in the brain. Tacrolimus 15-20 amyloid beta (A4) precursor protein Mus musculus 61-66 20847451-4 2010 Treatment with FK506, a CaN inhibitor, significantly reduces Abeta burden and restores synaptic proteins (synaptophysin and postsynaptic density protein-95; PSD-95) while inducing matrix metallopeptidase-9 (MMP-9) expression in GFAP-positive astrocytes in the brain. Tacrolimus 15-20 synaptophysin Mus musculus 106-119 20847451-4 2010 Treatment with FK506, a CaN inhibitor, significantly reduces Abeta burden and restores synaptic proteins (synaptophysin and postsynaptic density protein-95; PSD-95) while inducing matrix metallopeptidase-9 (MMP-9) expression in GFAP-positive astrocytes in the brain. Tacrolimus 15-20 discs large MAGUK scaffold protein 4 Mus musculus 124-155 20847451-4 2010 Treatment with FK506, a CaN inhibitor, significantly reduces Abeta burden and restores synaptic proteins (synaptophysin and postsynaptic density protein-95; PSD-95) while inducing matrix metallopeptidase-9 (MMP-9) expression in GFAP-positive astrocytes in the brain. Tacrolimus 15-20 discs large MAGUK scaffold protein 4 Mus musculus 157-163 20847451-4 2010 Treatment with FK506, a CaN inhibitor, significantly reduces Abeta burden and restores synaptic proteins (synaptophysin and postsynaptic density protein-95; PSD-95) while inducing matrix metallopeptidase-9 (MMP-9) expression in GFAP-positive astrocytes in the brain. Tacrolimus 15-20 matrix metallopeptidase 9 Mus musculus 180-205 20847451-4 2010 Treatment with FK506, a CaN inhibitor, significantly reduces Abeta burden and restores synaptic proteins (synaptophysin and postsynaptic density protein-95; PSD-95) while inducing matrix metallopeptidase-9 (MMP-9) expression in GFAP-positive astrocytes in the brain. Tacrolimus 15-20 matrix metallopeptidase 9 Mus musculus 207-212 20847451-4 2010 Treatment with FK506, a CaN inhibitor, significantly reduces Abeta burden and restores synaptic proteins (synaptophysin and postsynaptic density protein-95; PSD-95) while inducing matrix metallopeptidase-9 (MMP-9) expression in GFAP-positive astrocytes in the brain. Tacrolimus 15-20 glial fibrillary acidic protein Mus musculus 228-232 20847451-5 2010 These results suggest a role of FK506 and control of CaN activity in neuroprotection associated with Abeta deposition in AD. Tacrolimus 32-37 amyloid beta (A4) precursor protein Mus musculus 101-106 19609312-0 2010 Decrease of skin infiltrating and circulating CCR10+ T cells coincides with clinical improvement after topical tacrolimus in Omenn syndrome. Tacrolimus 111-121 C-C motif chemokine receptor 10 Homo sapiens 46-51 20402659-10 2010 Treatment with all three drugs (tacrolimus, somatostatin and glucocorticoids) resulted in a significant decrease of serum amylase, lung edema, and serum TNF-alpha and IL-1beta levels. Tacrolimus 32-42 tumor necrosis factor Rattus norvegicus 153-162 20402659-10 2010 Treatment with all three drugs (tacrolimus, somatostatin and glucocorticoids) resulted in a significant decrease of serum amylase, lung edema, and serum TNF-alpha and IL-1beta levels. Tacrolimus 32-42 interleukin 1 beta Rattus norvegicus 167-175 20172323-1 2010 The tacrolimus is metabolized primarily by CYP3A5, a member of the single nucleotide polymorphism family. Tacrolimus 4-14 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 43-49 20090409-6 2010 Betamethasone and, to a lesser extent, tacrolimus led to a marked reduction of chemical-induced IL-8 expression by TNBS and CAld. Tacrolimus 39-49 C-X-C motif chemokine ligand 8 Homo sapiens 96-100 19604038-5 2010 Inhibition of Cn activity by FK506 increased the expression of chondrogenic markers collagen type 2, aggrecan, and SOX9 in culture-expanded cells. Tacrolimus 29-34 SRY-box transcription factor 9 Homo sapiens 115-119 19604038-6 2010 Addition of FK506 increased endogenous transforming growth factor 2 (TGF) beta1 expression on both mRNA and protein level. Tacrolimus 12-17 transforming growth factor beta 1 Homo sapiens 39-79 19604038-7 2010 The effect of FK506 on chondrogenic markers was abolished by addition of anti-TGFbeta1 antibody, indicating that the endogenous TGFbeta1 was necessary to increase chondrogenic marker expression. Tacrolimus 14-19 transforming growth factor beta 1 Homo sapiens 78-86 19604038-7 2010 The effect of FK506 on chondrogenic markers was abolished by addition of anti-TGFbeta1 antibody, indicating that the endogenous TGFbeta1 was necessary to increase chondrogenic marker expression. Tacrolimus 14-19 transforming growth factor beta 1 Homo sapiens 128-136 19604038-9 2010 In conclusion, inhibition of Cn activity by FK506 increases the expression of chondrogenic markers via endogenous TGFbeta1 production in human articular chondrocytes. Tacrolimus 44-49 transforming growth factor beta 1 Homo sapiens 114-122 19820002-1 2010 ABO-incompatible kidney transplantation is possible after pre-treatment with rituximab, intravenous immunoglobulin and basiliximab combined with tacrolimus, mycophenolate mofetil and prednisolone. Tacrolimus 145-155 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 0-3 19747209-3 2009 Because the calcineurin inhibitor FK506 inhibits the hyperresponsiveness of Tgfb1(-/-) thymocytes, and because calcineurin Abeta (CNAbeta)-deficient mice do not reject allogenic tumours, we have generated Tgfb1(-/-) Cnab(-/-) mice to address whether CNAbeta deficiency prevents T cell activation and inflammation in Tgfb1(-/-) mice. Tacrolimus 34-39 transforming growth factor, beta 1 Mus musculus 76-81 19843528-3 2009 We show here that hTERT is expressed soon after lymphocyte activation and that its expression is inhibited by rapamycin, wortmannin, and FK506, which was the most potent inhibitor. Tacrolimus 137-142 telomerase reverse transcriptase Homo sapiens 18-23 20128966-6 2010 RESULTS: High dose of FK506 resulted in the immunosuppressed in LT rats, as evident by low production of IL-2 and IFN-gamma, and without liver rejection compared to rats in the control group. Tacrolimus 22-27 interleukin 2 Rattus norvegicus 105-109 20128966-6 2010 RESULTS: High dose of FK506 resulted in the immunosuppressed in LT rats, as evident by low production of IL-2 and IFN-gamma, and without liver rejection compared to rats in the control group. Tacrolimus 22-27 interferon gamma Rattus norvegicus 114-123 19852929-4 2009 TRESK is highly activated by Ca(2+), calcineurin, acetylcholine, and histamine which induce hypertrophy, whereas TRESK is inhibited by immunosuppressants, such as cyclosporin A and FK506. Tacrolimus 181-186 potassium two pore domain channel subfamily K member 18 Homo sapiens 0-5 19852929-4 2009 TRESK is highly activated by Ca(2+), calcineurin, acetylcholine, and histamine which induce hypertrophy, whereas TRESK is inhibited by immunosuppressants, such as cyclosporin A and FK506. Tacrolimus 181-186 potassium two pore domain channel subfamily K member 18 Homo sapiens 113-118 19728359-5 2009 Between 24 hr and 5 days after MCAo, FK506 reduced the T-cell infiltration in the infarct area as well as the presence of activated and/or phagocytic OX-18, OX-42, GSA-IB4, Iba1, and ED1 positive microglia/macrophages. Tacrolimus 37-42 allograft inflammatory factor 1 Rattus norvegicus 173-177 19728359-6 2009 FK506 also lowered the protein levels of TNFalpha and IL-2 in ischemic brain areas. Tacrolimus 0-5 tumor necrosis factor Rattus norvegicus 41-49 19728359-6 2009 FK506 also lowered the protein levels of TNFalpha and IL-2 in ischemic brain areas. Tacrolimus 0-5 interleukin 2 Rattus norvegicus 54-58 20005364-6 2009 It was observed that tacrolimus significantly decreased triglyceride, Apo A1, Apo B, LDL, HDL, and total cholesterol levels (P < .001; P = .006; P = .01; P < .001; P = .03; P </= .001, respectively), but had no effect on homocysteine, Apo A1/B, HDL 2, HDL 3, or HDL 2/3 levels (P > .05). Tacrolimus 21-31 apolipoprotein A1 Homo sapiens 70-76 19865079-6 2009 The population pharmacokinetic-pharmacogenetic model developed in de novo pediatric kidney transplant patients demonstrated that, in children, tacrolimus dosage should be based on weight, hematocrit levels, and CYP3A5 polymorphism. Tacrolimus 143-153 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 211-217 19930321-1 2009 The inhibitory effect of Cyclosporine A (CsA) and Tacrolimus (Tacr) on interleukin 2 (IL-2) are well known, and the importance of Th1-type (IL-2, interferon gamma), and Th2-type (IL-4, IL-6, and IL-10) cytokine secretion in preventing allograft rejection is a controversial issue. Tacrolimus 50-60 interleukin 2 Homo sapiens 71-84 19930321-1 2009 The inhibitory effect of Cyclosporine A (CsA) and Tacrolimus (Tacr) on interleukin 2 (IL-2) are well known, and the importance of Th1-type (IL-2, interferon gamma), and Th2-type (IL-4, IL-6, and IL-10) cytokine secretion in preventing allograft rejection is a controversial issue. Tacrolimus 50-60 interleukin 2 Homo sapiens 86-90 19930321-1 2009 The inhibitory effect of Cyclosporine A (CsA) and Tacrolimus (Tacr) on interleukin 2 (IL-2) are well known, and the importance of Th1-type (IL-2, interferon gamma), and Th2-type (IL-4, IL-6, and IL-10) cytokine secretion in preventing allograft rejection is a controversial issue. Tacrolimus 50-54 interleukin 2 Homo sapiens 71-84 19930321-1 2009 The inhibitory effect of Cyclosporine A (CsA) and Tacrolimus (Tacr) on interleukin 2 (IL-2) are well known, and the importance of Th1-type (IL-2, interferon gamma), and Th2-type (IL-4, IL-6, and IL-10) cytokine secretion in preventing allograft rejection is a controversial issue. Tacrolimus 50-54 interleukin 2 Homo sapiens 86-90 19930321-6 2009 A significantly impaired T-cell IL-10 secretion (p = 0.0001) and decreased Th function of whole T cells was found in Tacr-treated patients only, whereas unstimulated Th1 responses and SAC-I-stimulated B-cell IL-6 responses were reduced in CsA-treated patients. Tacrolimus 117-121 interleukin 6 Homo sapiens 208-212 19628039-2 2009 OBJECTIVE: This study examined the effect of FK506 on interleukin-2 (IL-2) and insulin secretion, establishing a novel characteristic of this drug that could explain its diverse adverse effects, and developed an experimental model for the simultaneous analysis of mRNA expression and protein secretion affected by this drug. Tacrolimus 45-50 interleukin 2 Homo sapiens 54-67 19628039-2 2009 OBJECTIVE: This study examined the effect of FK506 on interleukin-2 (IL-2) and insulin secretion, establishing a novel characteristic of this drug that could explain its diverse adverse effects, and developed an experimental model for the simultaneous analysis of mRNA expression and protein secretion affected by this drug. Tacrolimus 45-50 interleukin 2 Homo sapiens 69-73 19628039-3 2009 METHODS: The IL-2 levels when tacrolimus was administered were analysed by Western blot, immunocytochemistry and RT-PCR in a T lymphocyte cellular line (Jurkat) 24 h post-stimulation. Tacrolimus 30-40 interleukin 2 Homo sapiens 13-17 19628039-4 2009 The insulin levels when tacrolimus was administered were analysed 4 h after stimulation of glucose-induced insulin secretion in a pancreatic cellular line (MIN6). Tacrolimus 24-34 insulin Homo sapiens 4-11 19628039-5 2009 RESULTS: The previously published information describes tacrolimus as only capable of partially blocking IL-2 mRNA expression. Tacrolimus 56-66 interleukin 2 Homo sapiens 105-109 19628039-9 2009 Tacrolimus added before stimulation inhibits only IL-2 synthesis, but blocks IL-2 protein secretion when added 2 h after stimulation. Tacrolimus 0-10 interleukin 2 Homo sapiens 50-54 19628039-9 2009 Tacrolimus added before stimulation inhibits only IL-2 synthesis, but blocks IL-2 protein secretion when added 2 h after stimulation. Tacrolimus 0-10 interleukin 2 Homo sapiens 77-81 19628039-10 2009 Similarly, tacrolimus is also capable of blocking the glucose-stimulated secretion of insulin by MIN6 cells. Tacrolimus 11-21 insulin Homo sapiens 86-93 19628039-12 2009 CONCLUSIONS: Results of this study indicate that tacrolimus possesses another important effect in addition to the inhibition of IL-2 gene transcription, namely the ability to act as a general inhibitor of the protein secretory pathway. Tacrolimus 49-59 interleukin 2 Homo sapiens 128-132 20073093-0 2009 Topical tacrolimus suppresses the expression of vascular endothelial growth factor and insulin-like growth factor-1 in late anagen. Tacrolimus 8-18 vascular endothelial growth factor A Mus musculus 48-82 20073093-0 2009 Topical tacrolimus suppresses the expression of vascular endothelial growth factor and insulin-like growth factor-1 in late anagen. Tacrolimus 8-18 insulin-like growth factor 1 Mus musculus 87-115 19766187-10 2009 K combined with a half-dose of FK-506 reduced the expression levels of IL-2 and IFN-gamma (both within the graft and in the recipients" serum) more effectively than a full-dose of FK-506. Tacrolimus 31-37 interleukin 2 Mus musculus 71-75 19766187-10 2009 K combined with a half-dose of FK-506 reduced the expression levels of IL-2 and IFN-gamma (both within the graft and in the recipients" serum) more effectively than a full-dose of FK-506. Tacrolimus 31-37 interferon gamma Mus musculus 80-89 19952532-5 2009 Pretreatment with tacrolimus also produced significant (p<0.05) reduction in TBARS, total calcium, TNF-alpha, IL-8 and MPO whereas it showed an increase in GSH level at higher dose. Tacrolimus 18-28 tumor necrosis factor Rattus norvegicus 102-111 20005364-6 2009 It was observed that tacrolimus significantly decreased triglyceride, Apo A1, Apo B, LDL, HDL, and total cholesterol levels (P < .001; P = .006; P = .01; P < .001; P = .03; P </= .001, respectively), but had no effect on homocysteine, Apo A1/B, HDL 2, HDL 3, or HDL 2/3 levels (P > .05). Tacrolimus 21-31 apolipoprotein B Homo sapiens 78-83 19952532-5 2009 Pretreatment with tacrolimus also produced significant (p<0.05) reduction in TBARS, total calcium, TNF-alpha, IL-8 and MPO whereas it showed an increase in GSH level at higher dose. Tacrolimus 18-28 myeloperoxidase Rattus norvegicus 122-125 20005364-6 2009 It was observed that tacrolimus significantly decreased triglyceride, Apo A1, Apo B, LDL, HDL, and total cholesterol levels (P < .001; P = .006; P = .01; P < .001; P = .03; P </= .001, respectively), but had no effect on homocysteine, Apo A1/B, HDL 2, HDL 3, or HDL 2/3 levels (P > .05). Tacrolimus 21-31 apolipoprotein A1 Homo sapiens 244-252 20005364-6 2009 It was observed that tacrolimus significantly decreased triglyceride, Apo A1, Apo B, LDL, HDL, and total cholesterol levels (P < .001; P = .006; P = .01; P < .001; P = .03; P </= .001, respectively), but had no effect on homocysteine, Apo A1/B, HDL 2, HDL 3, or HDL 2/3 levels (P > .05). Tacrolimus 21-31 junctophilin 3 Homo sapiens 254-259 20005364-6 2009 It was observed that tacrolimus significantly decreased triglyceride, Apo A1, Apo B, LDL, HDL, and total cholesterol levels (P < .001; P = .006; P = .01; P < .001; P = .03; P </= .001, respectively), but had no effect on homocysteine, Apo A1/B, HDL 2, HDL 3, or HDL 2/3 levels (P > .05). Tacrolimus 21-31 HDL3 Homo sapiens 261-266 20005364-6 2009 It was observed that tacrolimus significantly decreased triglyceride, Apo A1, Apo B, LDL, HDL, and total cholesterol levels (P < .001; P = .006; P = .01; P < .001; P = .03; P </= .001, respectively), but had no effect on homocysteine, Apo A1/B, HDL 2, HDL 3, or HDL 2/3 levels (P > .05). Tacrolimus 21-31 junctophilin 3 Homo sapiens 271-276 19724275-8 2009 Regulation of COX-2 expression by NFAT was investigated using NFAT-targeted siRNA, calcineurin inhibitors cyclosporin A and FK506, in addition to COX-2 luciferase reporter vectors that selectively lacked NFAT binding sites. Tacrolimus 124-129 mitochondrially encoded cytochrome c oxidase II Homo sapiens 14-19 19759356-3 2009 Tacrolimus (FK506) is one of the most widely used immunosuppressive agents in SOT recipients, and its immunosuppressive effects are largely dependent on its interaction with the 12-kDa FK506-binding protein (FKBP12). Tacrolimus 0-10 FKBP prolyl isomerase 1A Homo sapiens 178-206 19759356-3 2009 Tacrolimus (FK506) is one of the most widely used immunosuppressive agents in SOT recipients, and its immunosuppressive effects are largely dependent on its interaction with the 12-kDa FK506-binding protein (FKBP12). Tacrolimus 0-10 FKBP prolyl isomerase 1A Homo sapiens 208-214 19759356-3 2009 Tacrolimus (FK506) is one of the most widely used immunosuppressive agents in SOT recipients, and its immunosuppressive effects are largely dependent on its interaction with the 12-kDa FK506-binding protein (FKBP12). Tacrolimus 12-17 FKBP prolyl isomerase 1A Homo sapiens 178-206 19759356-3 2009 Tacrolimus (FK506) is one of the most widely used immunosuppressive agents in SOT recipients, and its immunosuppressive effects are largely dependent on its interaction with the 12-kDa FK506-binding protein (FKBP12). Tacrolimus 12-17 FKBP prolyl isomerase 1A Homo sapiens 208-214 19759356-4 2009 We have knocked down the expression of FKBP12 in EBV-CTLs using a specific small interfering RNA (siRNA) stably expressed from a retroviral vector and found that FKBP12-silenced EBV-CTLs are FK506 resistant. Tacrolimus 191-196 FKBP prolyl isomerase 1A Homo sapiens 39-45 19759356-4 2009 We have knocked down the expression of FKBP12 in EBV-CTLs using a specific small interfering RNA (siRNA) stably expressed from a retroviral vector and found that FKBP12-silenced EBV-CTLs are FK506 resistant. Tacrolimus 191-196 FKBP prolyl isomerase 1A Homo sapiens 162-168 19681813-1 2009 Tacrolimus, a cornerstone immunosuppressant, is widely available as a twice-daily formulation (Tacrolimus BID). Tacrolimus 0-10 BH3 interacting domain death agonist Homo sapiens 106-109 19681813-1 2009 Tacrolimus, a cornerstone immunosuppressant, is widely available as a twice-daily formulation (Tacrolimus BID). Tacrolimus 95-105 BH3 interacting domain death agonist Homo sapiens 106-109 19681813-3 2009 This study compared the pharmacokinetics (PK) of tacrolimus in de novo kidney transplant patients treated with Tacrolimus QD or Tacrolimus BID. Tacrolimus 49-59 BH3 interacting domain death agonist Homo sapiens 139-142 19681813-9 2009 Mean AUC(0-24) of tacrolimus on day 1 was approximately 30% lower for Tacrolimus QD than Tacrolimus BID (232 and 361 ng.h/mL, respectively), but was comparable by day 4. Tacrolimus 18-28 BH3 interacting domain death agonist Homo sapiens 100-103 19681813-12 2009 Tacrolimus QD can be administered once daily in the morning on the basis of the same systemic exposure and therapeutic drug monitoring concept as Tacrolimus BID. Tacrolimus 0-10 BH3 interacting domain death agonist Homo sapiens 157-160 19737140-9 2009 In vitro, methylprednisolone and gabexate mesilate had no effect and cyclosporin A and tacrolimus only a moderate effect on production of granzyme b by CD8(+) T cells. Tacrolimus 87-97 granzyme B Homo sapiens 138-148 19737140-9 2009 In vitro, methylprednisolone and gabexate mesilate had no effect and cyclosporin A and tacrolimus only a moderate effect on production of granzyme b by CD8(+) T cells. Tacrolimus 87-97 CD8a molecule Homo sapiens 152-155 21436979-3 2009 Tacrolimus, a calcineurin inhibitor, not only complements existing treatment options but also overcomes some of the drawbacks of topical steroid therapy when given topically and thus meets the long-term needs of patients in preventing disease progression. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 14-35 21436979-5 2009 The recent proactive use of topical tacrolimus may emphasize a long-term benefit of this calcineurin inhibitor for AD treatment. Tacrolimus 36-46 calcineurin binding protein 1 Homo sapiens 89-110 19860865-8 2009 We found that pharmacological inhibition of calcineurin by FK506 led to sustained phosphorylation of mutant huntingtin at S421. Tacrolimus 59-64 huntingtin Homo sapiens 108-118 19860865-9 2009 FK506 restored BDNF transport in two complementary models: rat primary neuronal cultures expressing mutant huntingtin and mouse cortical neurons from Hdh(Q111/Q111) HD knock-in mice. Tacrolimus 0-5 brain-derived neurotrophic factor Rattus norvegicus 15-19 19860865-9 2009 FK506 restored BDNF transport in two complementary models: rat primary neuronal cultures expressing mutant huntingtin and mouse cortical neurons from Hdh(Q111/Q111) HD knock-in mice. Tacrolimus 0-5 huntingtin Rattus norvegicus 107-117 19860865-9 2009 FK506 restored BDNF transport in two complementary models: rat primary neuronal cultures expressing mutant huntingtin and mouse cortical neurons from Hdh(Q111/Q111) HD knock-in mice. Tacrolimus 0-5 huntingtin Mus musculus 150-153 19785652-6 2009 KEY RESULTS: FK506 which displaces FKBP from each receptor (to inhibit calcineurin) increased the [Ca(2+)](cyto) rise evoked by activation of either RyR or IP(3)R. Tacrolimus 13-18 ryanodine receptor 1 Homo sapiens 149-152 18957484-6 2009 RESULTS: Treatment of PBL with leflunomide, cyclosporin A and FK-506 inhibited the IL-15-induced expression of both CD54 and CD69 by PBL, as well as TNF production in co-cultures of activated PBL and THP-1 cells. Tacrolimus 62-68 interleukin 15 Homo sapiens 83-88 18957484-6 2009 RESULTS: Treatment of PBL with leflunomide, cyclosporin A and FK-506 inhibited the IL-15-induced expression of both CD54 and CD69 by PBL, as well as TNF production in co-cultures of activated PBL and THP-1 cells. Tacrolimus 62-68 intercellular adhesion molecule 1 Homo sapiens 116-120 18957484-6 2009 RESULTS: Treatment of PBL with leflunomide, cyclosporin A and FK-506 inhibited the IL-15-induced expression of both CD54 and CD69 by PBL, as well as TNF production in co-cultures of activated PBL and THP-1 cells. Tacrolimus 62-68 CD69 molecule Homo sapiens 125-129 18957484-6 2009 RESULTS: Treatment of PBL with leflunomide, cyclosporin A and FK-506 inhibited the IL-15-induced expression of both CD54 and CD69 by PBL, as well as TNF production in co-cultures of activated PBL and THP-1 cells. Tacrolimus 62-68 tumor necrosis factor Homo sapiens 149-152 18957484-6 2009 RESULTS: Treatment of PBL with leflunomide, cyclosporin A and FK-506 inhibited the IL-15-induced expression of both CD54 and CD69 by PBL, as well as TNF production in co-cultures of activated PBL and THP-1 cells. Tacrolimus 62-68 GLI family zinc finger 2 Homo sapiens 200-205 18957484-8 2009 Likewise, leflunomide, cyclosporin A and FK-506 all inhibited IL-17 production in IL-15-activated PBL. Tacrolimus 41-47 interleukin 17A Homo sapiens 62-67 18957484-8 2009 Likewise, leflunomide, cyclosporin A and FK-506 all inhibited IL-17 production in IL-15-activated PBL. Tacrolimus 41-47 interleukin 15 Homo sapiens 82-87 19785652-6 2009 KEY RESULTS: FK506 which displaces FKBP from each receptor (to inhibit calcineurin) increased the [Ca(2+)](cyto) rise evoked by activation of either RyR or IP(3)R. Tacrolimus 13-18 inositol 1,4,5-trisphosphate receptor type 3 Homo sapiens 156-162 19730841-5 2009 D/BW-adjusted C(max), C(0) and AUC(0-12) values of tacrolimus were significantly greater in patients with the CYP3A5*3/*3 genotype than in those with the CYP3A5*1 allele in young and middle-aged patients as previously reported, but not in the elderly. Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 110-116 19082716-4 2009 The results showed that in ABO-C groups I and II and in ABO-I group I, effector cells remained above the pretransplant levels after tacrolimus administration. Tacrolimus 132-142 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 56-59 19730841-5 2009 D/BW-adjusted C(max), C(0) and AUC(0-12) values of tacrolimus were significantly greater in patients with the CYP3A5*3/*3 genotype than in those with the CYP3A5*1 allele in young and middle-aged patients as previously reported, but not in the elderly. Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 154-160 19730841-8 2009 However, a larger number of subjects need to be studied to confirm the impact of age on the CYP3A5 pharmacogenetics of tacrolimus in the elderly. Tacrolimus 119-129 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 92-98 19468783-0 2009 Efficacy of tacrolimus in Sjogren"s syndrome-associated CNS disease with aquaporin-4 autoantibodies. Tacrolimus 12-22 aquaporin 4 Homo sapiens 73-84 19729569-2 2009 Tacrolimus is a substrate for P-glycoprotein, the product of the ABCB1 gene. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 30-44 19604035-3 2009 The extent of tacrolimus metabolism, when normalized to the cytochrome P-450 content, was in the order: rat < hamster < rabbit < pig < guinea pig < dog < human < baboon. Tacrolimus 14-24 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 60-76 19729569-2 2009 Tacrolimus is a substrate for P-glycoprotein, the product of the ABCB1 gene. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 65-70 19729569-0 2009 Effect of the ABCB1 3435C>T polymorphism on tacrolimus concentrations and dosage requirements in liver transplant recipients. Tacrolimus 47-57 ATP binding cassette subfamily B member 1 Homo sapiens 14-19 19729569-1 2009 PURPOSE: The effect of ABCB1 3435C>T on tacrolimus concentrations in liver transplant recipients was studied. Tacrolimus 43-53 ATP binding cassette subfamily B member 1 Homo sapiens 23-28 19729569-3 2009 To determine whether the ABCB1 single-nucleotide polymorphism (SNP) 3435C>T was associated with variation in the tacrolimus concentration:dose ratio (C:D) in 42 liver transplant recipients during three months after transplantation. Tacrolimus 116-126 ATP binding cassette subfamily B member 1 Homo sapiens 25-30 19729569-18 2009 CONCLUSION: The ABCB1 3435C>T polymorphism influenced the tacrolimus C:D in the first days after liver transplantation. Tacrolimus 61-71 ATP binding cassette subfamily B member 1 Homo sapiens 16-21 19438859-0 2009 Analysis of interleukin-10 levels in lesions of vitiligo following treatment with topical tacrolimus. Tacrolimus 90-100 interleukin 10 Homo sapiens 12-26 19438859-4 2009 We propose that the successful treatment of vitiligo with topical tacrolimus involves the unique immunosuppressive actions of the T lymphocyte T-helper (Th) 2 cytokine, interleukin (IL)-10. Tacrolimus 66-76 interleukin 10 Homo sapiens 169-188 19438859-11 2009 In addition, there was a statistically significant difference between IL-10 expression in vitiligo lesions following treatment for 3 months with topical tacrolimus compared with untreated vitiligo lesions (P = 0.017) and normal skin (P = 0.004). Tacrolimus 153-163 interleukin 10 Homo sapiens 70-75 19438859-13 2009 We propose that topical tacrolimus increases IL-10 expression in vitiligo lesions, and thereby inhibits melanocyte destruction triggered by unchecked Th1 pathways in vitiligo. Tacrolimus 24-34 interleukin 10 Homo sapiens 45-50 19438859-13 2009 We propose that topical tacrolimus increases IL-10 expression in vitiligo lesions, and thereby inhibits melanocyte destruction triggered by unchecked Th1 pathways in vitiligo. Tacrolimus 24-34 negative elongation factor complex member C/D Homo sapiens 150-153 19405107-3 2009 To achieve precise pharmacological modulation of neurotrophin activity, we have generated a chimeric trkA receptor (ItrkA) by fusing the entire intracellular domain of the trkA high-affinity NGF receptor to two intracellular, modified FK506 binding domains for the synthetic small molecule dimerization ligand AP20187. Tacrolimus 235-240 neurotrophic receptor tyrosine kinase 1 Rattus norvegicus 101-105 19740399-0 2009 Influence of ABCB1 polymorphisms and haplotypes on tacrolimus nephrotoxicity and dosage requirements in children with liver transplant. Tacrolimus 51-61 ATP binding cassette subfamily B member 1 Homo sapiens 13-18 19740399-1 2009 AIMS: The aim of this study was to investigate the influence of genetic polymorphisms in ABCB1 on the incidence of nephrotoxicity and tacrolimus dosage-requirements in paediatric patients following liver transplantation. Tacrolimus 134-144 ATP binding cassette subfamily B member 1 Homo sapiens 89-94 19740399-8 2009 CONCLUSIONS: These findings suggest that ABCB1 polymorphisms in the native intestine significantly influence tacrolimus dosage-requirement in the stable phase after transplantation. Tacrolimus 109-119 ATP binding cassette subfamily B member 1 Homo sapiens 41-46 19740399-10 2009 Genotyping future transplant recipients for ABCB1 polymorphisms, therefore, could have the potential to individualize better tacrolimus immunosuppressive therapy and enhance drug safety. Tacrolimus 125-135 ATP binding cassette subfamily B member 1 Homo sapiens 44-49 19494809-9 2009 UGT1A9 -275T>A and/or -2152C>T genotyping may identify patients at risk of MPA underexposure and acute rejection when receiving treatment with MMF and tacrolimus. Tacrolimus 157-167 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 0-6 19553475-5 2009 32P-labeling demonstrated direct phosphorylation of TRPV1 or TRPV1t in anterior lingual epithelium by PMA, cyclosporin A, or FK-506. Tacrolimus 125-131 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 52-57 19553475-5 2009 32P-labeling demonstrated direct phosphorylation of TRPV1 or TRPV1t in anterior lingual epithelium by PMA, cyclosporin A, or FK-506. Tacrolimus 125-131 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 61-66 19494809-0 2009 UGT1A9 -275T>A/-2152C>T polymorphisms correlate with low MPA exposure and acute rejection in MMF/tacrolimus-treated kidney transplant patients. Tacrolimus 103-113 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 0-6 19494809-5 2009 Tacrolimus-treated patients who were UGT1A9 -275T>A and/or -2152C>T carriers displayed a 20% lower MPA area under the concentration-time curve from 0 to 12 h (AUC(0-12)) (P = 0.012). Tacrolimus 0-10 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 37-43 19494809-6 2009 UGT1A9*3 carriers displayed a 49% higher MPA AUC(0-12) when treated with tacrolimus and a 54% higher MPA AUC(0-12) when treated with cyclosporine (P < 0.005). Tacrolimus 73-83 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 0-6 19494809-8 2009 Carrying the UGT1A9 -275T>A and/or -2152C>T polymorphism significantly predicted acute rejection in fixed-dose (FD) MMF-treated patients receiving tacrolimus (odds ratio 13.3, 95% confidence interval 1.1-162.3; P < 0.05). Tacrolimus 153-163 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 13-19 19813492-3 2009 Tacrolimus is metabolized by cytochrome P450 3A4 in both the liver and small intestine. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-48 19813492-4 2009 Drugs that are substrates of cytochrome P450 3A4, as well as inhibitors and inducers of cytochrome P450 3A4, can cause significant interactions with tacrolimus. Tacrolimus 149-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-48 19813492-4 2009 Drugs that are substrates of cytochrome P450 3A4, as well as inhibitors and inducers of cytochrome P450 3A4, can cause significant interactions with tacrolimus. Tacrolimus 149-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-107 19420299-8 2009 Calcineurin activity increased significantly from 1214 +/- 111 to 1652 +/- 138 fmol/microg/min; addition of either tacrolimus or CsA (500 ng/ml) blocked CD3/CD28 stimulation. Tacrolimus 115-125 CD28 molecule Homo sapiens 157-161 19153737-0 2009 Adding low dose tacrolimus in rheumatoid arthritis patients with an inadequate response to tumor necrosis factor inhibitor therapies. Tacrolimus 16-26 tumor necrosis factor Homo sapiens 91-112 19545546-5 2009 We noted that FK506 altered nuclear localization of the GR and inhibited expression of GR-responsive genes. Tacrolimus 14-19 nuclear receptor subfamily 3 group C member 1 Homo sapiens 56-58 19545546-5 2009 We noted that FK506 altered nuclear localization of the GR and inhibited expression of GR-responsive genes. Tacrolimus 14-19 nuclear receptor subfamily 3 group C member 1 Homo sapiens 87-89 19667964-8 2009 Cyclosporine A and Tac are metabolized by CYP3A4 and CYP3A5, and several single nucleotide polymorphisms in the two genes have been associated with differences in drug clearance. Tacrolimus 19-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 19667964-8 2009 Cyclosporine A and Tac are metabolized by CYP3A4 and CYP3A5, and several single nucleotide polymorphisms in the two genes have been associated with differences in drug clearance. Tacrolimus 19-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 53-59 19439519-8 2009 In parallel, protein expression of AE1 was reduced at baseline and increased together with pendrin during NH(4)Cl loading in FK506 rats. Tacrolimus 125-130 solute carrier family 4 member 1 (Diego blood group) Rattus norvegicus 35-38 19714314-0 2009 Tacrolimus differentially regulates the proliferation of conventional and regulatory CD4(+) T cells. Tacrolimus 0-10 CD4 molecule Homo sapiens 85-88 19714314-1 2009 Tacrolimus is a widely used T cell targeted immunosuppressive drug, known as a calcineurin inhibitor. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 79-100 19714314-2 2009 However, the exact pharmacological effects of tacrolimus on CD4(+) T cells have yet to be elucidated. Tacrolimus 46-56 CD4 molecule Homo sapiens 60-63 19714314-3 2009 This study investigated the effects of tacrolimus on CD4(+) T cell subsets. Tacrolimus 39-49 CD4 molecule Homo sapiens 53-56 19714314-4 2009 Mouse or human CD4(+) T cells were cultured with immobilized anti-CD3/CD28 antibodies in the presence of tacrolimus. Tacrolimus 105-115 CD28 molecule Homo sapiens 70-74 19714314-7 2009 In the case of conventional CD4(+) T cells (Tconv cells), tacrolimus inhibited T cell receptor stimulation-induced cell division. Tacrolimus 58-68 CD4 molecule Homo sapiens 28-31 19714314-8 2009 In contrast, the cell division of regulatory CD4(+) T cells (Treg cells) was even promoted in the presence of tacrolimus, especially in humans. Tacrolimus 110-120 CD4 molecule Homo sapiens 45-48 19714314-10 2009 Furthermore, tacrolimus modified the expression levels of Foxp3-regulated T cell receptor signal related-genes, PTPN22 and Itk, in human Treg cells. Tacrolimus 13-23 forkhead box P3 Homo sapiens 58-63 19714314-10 2009 Furthermore, tacrolimus modified the expression levels of Foxp3-regulated T cell receptor signal related-genes, PTPN22 and Itk, in human Treg cells. Tacrolimus 13-23 protein tyrosine phosphatase non-receptor type 22 Homo sapiens 112-118 19714314-10 2009 Furthermore, tacrolimus modified the expression levels of Foxp3-regulated T cell receptor signal related-genes, PTPN22 and Itk, in human Treg cells. Tacrolimus 13-23 IL2 inducible T cell kinase Homo sapiens 123-126 19539216-1 2009 Previous studies have shown that adding sirolimus to a tacrolimus/mini-methotrexate regimen (Tac/Sir/MTX) as graft-versus-host disease (GVHD) prophylaxis produces low rates of acute GVHD (aGVHD) after reduced-intensity conditioning (RIC) allogeneic stem cell transplantation (SCT). Tacrolimus 55-65 metaxin 1 Homo sapiens 101-104 19333617-0 2009 The effects of over-expression of the FK506-binding protein FKBP12.6 on K(+) currents in adult rabbit ventricular myocytes. Tacrolimus 38-43 peptidyl-prolyl cis-trans isomerase FKBP1B Oryctolagus cuniculus 60-68 19333617-5 2009 FKBP12.6 over-expression decreased the amplitude of the inward rectifier current (I (K1)) by ~25% in the voltage range -70 to -30 mV, an effect prevented by FK506 or lowering intracellular [Ca(2+)] below 1 nM. Tacrolimus 157-162 peptidyl-prolyl cis-trans isomerase FKBP1B Oryctolagus cuniculus 0-8 19584682-0 2009 Tacrolimus-induced elevation in plasma triglyceride concentrations after administration to renal transplant patients is partially due to a decrease in lipoprotein lipase activity and plasma concentrations. Tacrolimus 0-10 lipoprotein lipase Homo sapiens 151-169 19560701-10 2009 CONCLUSIONS: These results suggest that methylprednisolone and tacrolimus inhibit hypothermia-induced endothelial gap formation by phosphorylated ERK 1/2 inhibition and connexin 43 stabilization. Tacrolimus 63-73 mitogen-activated protein kinase 3 Homo sapiens 146-153 19393753-0 2009 Effect of FK506 and cyclosporine A on the expression of BDNF, tyrosine kinase B and p75 neurotrophin receptors in astrocytes exposed to simulated ischemia in vitro. Tacrolimus 10-15 brain-derived neurotrophic factor Rattus norvegicus 56-60 19393753-0 2009 Effect of FK506 and cyclosporine A on the expression of BDNF, tyrosine kinase B and p75 neurotrophin receptors in astrocytes exposed to simulated ischemia in vitro. Tacrolimus 10-15 nerve growth factor receptor Rattus norvegicus 84-87 19393753-1 2009 We investigated whether the immunosuppressive drugs, FK506 and cyclosporine A, increase BDNF protein and/or mRNA expression in ischemic astrocytes and if an increase could be related to changes in the nuclear expression of p-CREB, p-Erk1/2 and p-Akt. Tacrolimus 53-58 brain-derived neurotrophic factor Rattus norvegicus 88-92 19393753-4 2009 FK506 and cyclosporine A (at 1000nM and 0.25microM, respectively) stimulated the expression and release of BDNF in cultured rat cerebral cortical astrocytes exposed to OGD. Tacrolimus 0-5 brain-derived neurotrophic factor Rattus norvegicus 107-111 19828092-10 2009 Immunosuppressive therapy with FK506, subsequent to liver transplantation, restored CD81 expression at normal levels. Tacrolimus 31-36 CD81 molecule Homo sapiens 84-88 19571380-9 2009 Tacrolimus in doses of 3% and 10% significantly inhibited tryptase-induced scratching behavior. Tacrolimus 0-10 tryptase alpha/beta 1 Mus musculus 58-66 19560701-10 2009 CONCLUSIONS: These results suggest that methylprednisolone and tacrolimus inhibit hypothermia-induced endothelial gap formation by phosphorylated ERK 1/2 inhibition and connexin 43 stabilization. Tacrolimus 63-73 gap junction protein alpha 1 Homo sapiens 169-180 19447772-9 2009 SAA-induced CCL20 production was also suppressed by dexamethasone or FK506. Tacrolimus 69-74 serum amyloid A1 cluster Homo sapiens 0-3 19267185-1 2009 PURPOSE: This study investigated whether haplotypes in the multidrug resistance 1 (MDR1) gene had effects on mRNA expression levels of MDR1 and cytochrome P450 (CYP) 3A4, and on the pharmacokinetics of tacrolimus in living-donor liver transplant (LDLT) patients, considering the gender difference. Tacrolimus 202-212 ATP binding cassette subfamily B member 1 Homo sapiens 59-81 19267185-1 2009 PURPOSE: This study investigated whether haplotypes in the multidrug resistance 1 (MDR1) gene had effects on mRNA expression levels of MDR1 and cytochrome P450 (CYP) 3A4, and on the pharmacokinetics of tacrolimus in living-donor liver transplant (LDLT) patients, considering the gender difference. Tacrolimus 202-212 ATP binding cassette subfamily B member 1 Homo sapiens 83-87 19267185-6 2009 CONCLUSION: MDR1 haplotype may have a minor association with the tacrolimus pharmacokinetics after LDLT, but could be a good predictor of the inter-individual variation of intestinal expression of CYP3A4 in women. Tacrolimus 65-75 ATP binding cassette subfamily B member 1 Homo sapiens 12-16 19447772-9 2009 SAA-induced CCL20 production was also suppressed by dexamethasone or FK506. Tacrolimus 69-74 C-C motif chemokine ligand 20 Homo sapiens 12-17 19254242-0 2009 Tacrolimus and angiotensin receptor blockers associated with changes in serum adiponectin level in new-onset diabetes after renal transplantation: single-center cross-sectional analysis. Tacrolimus 0-10 adiponectin, C1Q and collagen domain containing Homo sapiens 78-89 19454720-6 2009 Induction of CCL2, CCL7, CXCL3, and CXCL8 by anti-IgE was significantly inhibited by dexamethasone but was enhanced by FK506. Tacrolimus 119-124 C-C motif chemokine ligand 7 Homo sapiens 19-23 19254242-6 2009 In addition, the post-transplant serum adiponectin level in patients treated with tacrolimus (TAC) was significantly lower than that in patients with cyclosporine (14.3 vs. 18.7 microg/ml, P = 0.01), while, that level in patients treated with angiotensin receptor blockers (ARB) was significantly higher than that in patients without treatment of ARB (17.9 vs. 14.7 microg/ml, P = 0.01). Tacrolimus 82-92 adiponectin, C1Q and collagen domain containing Homo sapiens 39-50 19715850-5 2009 VLP was associated with older recipients (P = .002), HLA incompatibilities (P = .001), older donors (P = .000), nontraumatic cause of brain death (P = .033) and previous hypertension (P = .030), tacrolimus (P = .000) and induction treatment with Thymoglobulin (P = .018) or anti-CD25 monoclonal antibodies (P = .000), as well as delayed graft function (DGF; P = .000). Tacrolimus 195-205 VHL like Homo sapiens 0-3 19715850-7 2009 Multivariate analysis confirmed the impact of donor age, HLA incompatibilities, DGF, and tacrolimus treatment to predict the presence of VLP. Tacrolimus 89-99 VHL like Homo sapiens 137-140 19715866-1 2009 BACKGROUND: Mammalian target of rapamycin (mTOR) inhibitors behave as potent immunosuppressants which have the advantages, with respect to calcineurin inhibitors (CNI: cyclosporine or tacrolimus), of no nephrotoxicity and inhibition of cell proliferation. Tacrolimus 184-194 mechanistic target of rapamycin kinase Homo sapiens 12-41 19715866-1 2009 BACKGROUND: Mammalian target of rapamycin (mTOR) inhibitors behave as potent immunosuppressants which have the advantages, with respect to calcineurin inhibitors (CNI: cyclosporine or tacrolimus), of no nephrotoxicity and inhibition of cell proliferation. Tacrolimus 184-194 mechanistic target of rapamycin kinase Homo sapiens 43-47 19458075-5 2009 Tacrolimus, a calcineurin inhibitor that inhibits dephosphorylation of NFAT, inhibited SFRP2-induced endothelial tube formation. Tacrolimus 0-10 secreted frizzled-related protein 2 Mus musculus 87-92 19454717-0 2009 Suppression of NF-kappaB by cyclosporin a and tacrolimus (FK506) via induction of the C/EBP family: implication for unfolded protein response. Tacrolimus 46-56 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 15-24 19454717-0 2009 Suppression of NF-kappaB by cyclosporin a and tacrolimus (FK506) via induction of the C/EBP family: implication for unfolded protein response. Tacrolimus 46-56 CCAAT/enhancer binding protein (C/EBP), beta Mus musculus 86-91 19454717-0 2009 Suppression of NF-kappaB by cyclosporin a and tacrolimus (FK506) via induction of the C/EBP family: implication for unfolded protein response. Tacrolimus 58-63 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 15-24 19454717-0 2009 Suppression of NF-kappaB by cyclosporin a and tacrolimus (FK506) via induction of the C/EBP family: implication for unfolded protein response. Tacrolimus 58-63 CCAAT/enhancer binding protein (C/EBP), beta Mus musculus 86-91 19454717-3 2009 We found that, in renal tubular cells, induction of MCP-1 by inflammatory cytokines was blunted by CsA and FK506. Tacrolimus 107-112 mast cell protease 1 Mus musculus 52-57 19398001-1 2009 We evaluated whether the regulatory function of CD4(+)CD25(high+)FoxP3(+) T-cells from patients on tacrolimus and mycophenolate mofetil (MMF) is affected by preceding steroid and anti-CD25 mAb induction therapy and whether this function is associated with rejection after kidney transplantation. Tacrolimus 99-109 CD4 molecule Homo sapiens 48-51 19398001-1 2009 We evaluated whether the regulatory function of CD4(+)CD25(high+)FoxP3(+) T-cells from patients on tacrolimus and mycophenolate mofetil (MMF) is affected by preceding steroid and anti-CD25 mAb induction therapy and whether this function is associated with rejection after kidney transplantation. Tacrolimus 99-109 interleukin 2 receptor subunit alpha Homo sapiens 54-58 19398001-1 2009 We evaluated whether the regulatory function of CD4(+)CD25(high+)FoxP3(+) T-cells from patients on tacrolimus and mycophenolate mofetil (MMF) is affected by preceding steroid and anti-CD25 mAb induction therapy and whether this function is associated with rejection after kidney transplantation. Tacrolimus 99-109 forkhead box P3 Homo sapiens 65-70 19543067-0 2009 Insulin hyposecretion in nondiabetic, tacrolimus-treated renal transplant recipients more than 6 months posttransplantation. Tacrolimus 38-48 insulin Homo sapiens 0-7 19011160-6 2009 Modification of RyR2 activity by FK506-binding protein 12.6 homozygous or RyR2 heterozygous gene deletion did not prevent the effect of PKCepsilon inhibition or activation. Tacrolimus 33-38 ryanodine receptor 2 Homo sapiens 16-20 19481021-4 2009 The aim of this prospective study was to evaluate the safety and efficacy of conversion to tacrolimus and atorvastatin in CsA-treated heart transplant recipients and dyslipidemia refractory to fluvastatin. Tacrolimus 91-101 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 122-125 19481021-11 2009 CONCLUSIONS: Conversion to tacrolimus and atorvastatin appears to be a safe and effective lipid-lowering therapy in CsA-treated heart transplant recipients with dyslipidemia refractory to fluvastatin. Tacrolimus 27-37 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 116-119 19454720-6 2009 Induction of CCL2, CCL7, CXCL3, and CXCL8 by anti-IgE was significantly inhibited by dexamethasone but was enhanced by FK506. Tacrolimus 119-124 C-X-C motif chemokine ligand 3 Homo sapiens 25-30 19454717-7 2009 In TNF-alpha-treated cells, suppression of MCP-1 by CsA or FK506 was associated with blunted responses of NF-kappaB, the crucial regulator of MCP-1. Tacrolimus 59-64 tumor necrosis factor Mus musculus 3-12 19454717-7 2009 In TNF-alpha-treated cells, suppression of MCP-1 by CsA or FK506 was associated with blunted responses of NF-kappaB, the crucial regulator of MCP-1. Tacrolimus 59-64 mast cell protease 1 Mus musculus 43-48 19454720-6 2009 Induction of CCL2, CCL7, CXCL3, and CXCL8 by anti-IgE was significantly inhibited by dexamethasone but was enhanced by FK506. Tacrolimus 119-124 C-X-C motif chemokine ligand 8 Homo sapiens 36-41 19454717-7 2009 In TNF-alpha-treated cells, suppression of MCP-1 by CsA or FK506 was associated with blunted responses of NF-kappaB, the crucial regulator of MCP-1. Tacrolimus 59-64 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 106-115 19454717-9 2009 CsA and FK506, as well as other UPR inducers, caused up-regulation of C/EBP family members, especially C/EBPbeta and CHOP (C/EBP homologous protein), and overexpression of either C/EBPbeta or CHOP significantly attenuated TNF-alpha-triggered NF-kappaB activation. Tacrolimus 8-13 CCAAT/enhancer binding protein (C/EBP), beta Mus musculus 70-75 19454720-7 2009 In contrast, induction of CCL1, CCL3, CCL4, and CCL18 was significantly inhibited by FK506 but, with the exception of CCL1, was enhanced by dexamethasone. Tacrolimus 85-90 C-C motif chemokine ligand 1 Homo sapiens 26-30 19454717-9 2009 CsA and FK506, as well as other UPR inducers, caused up-regulation of C/EBP family members, especially C/EBPbeta and CHOP (C/EBP homologous protein), and overexpression of either C/EBPbeta or CHOP significantly attenuated TNF-alpha-triggered NF-kappaB activation. Tacrolimus 8-13 CCAAT/enhancer binding protein (C/EBP), beta Mus musculus 103-112 19454717-9 2009 CsA and FK506, as well as other UPR inducers, caused up-regulation of C/EBP family members, especially C/EBPbeta and CHOP (C/EBP homologous protein), and overexpression of either C/EBPbeta or CHOP significantly attenuated TNF-alpha-triggered NF-kappaB activation. Tacrolimus 8-13 DNA-damage inducible transcript 3 Mus musculus 117-121 19454720-7 2009 In contrast, induction of CCL1, CCL3, CCL4, and CCL18 was significantly inhibited by FK506 but, with the exception of CCL1, was enhanced by dexamethasone. Tacrolimus 85-90 C-C motif chemokine ligand 3 Homo sapiens 32-36 19454717-9 2009 CsA and FK506, as well as other UPR inducers, caused up-regulation of C/EBP family members, especially C/EBPbeta and CHOP (C/EBP homologous protein), and overexpression of either C/EBPbeta or CHOP significantly attenuated TNF-alpha-triggered NF-kappaB activation. Tacrolimus 8-13 DNA-damage inducible transcript 3 Mus musculus 123-147 19454717-9 2009 CsA and FK506, as well as other UPR inducers, caused up-regulation of C/EBP family members, especially C/EBPbeta and CHOP (C/EBP homologous protein), and overexpression of either C/EBPbeta or CHOP significantly attenuated TNF-alpha-triggered NF-kappaB activation. Tacrolimus 8-13 CCAAT/enhancer binding protein (C/EBP), beta Mus musculus 179-188 19454720-7 2009 In contrast, induction of CCL1, CCL3, CCL4, and CCL18 was significantly inhibited by FK506 but, with the exception of CCL1, was enhanced by dexamethasone. Tacrolimus 85-90 C-C motif chemokine ligand 4 Homo sapiens 38-42 19454717-9 2009 CsA and FK506, as well as other UPR inducers, caused up-regulation of C/EBP family members, especially C/EBPbeta and CHOP (C/EBP homologous protein), and overexpression of either C/EBPbeta or CHOP significantly attenuated TNF-alpha-triggered NF-kappaB activation. Tacrolimus 8-13 DNA-damage inducible transcript 3 Mus musculus 192-196 19454720-7 2009 In contrast, induction of CCL1, CCL3, CCL4, and CCL18 was significantly inhibited by FK506 but, with the exception of CCL1, was enhanced by dexamethasone. Tacrolimus 85-90 C-C motif chemokine ligand 18 Homo sapiens 48-53 19454717-9 2009 CsA and FK506, as well as other UPR inducers, caused up-regulation of C/EBP family members, especially C/EBPbeta and CHOP (C/EBP homologous protein), and overexpression of either C/EBPbeta or CHOP significantly attenuated TNF-alpha-triggered NF-kappaB activation. Tacrolimus 8-13 tumor necrosis factor Mus musculus 222-231 19454717-9 2009 CsA and FK506, as well as other UPR inducers, caused up-regulation of C/EBP family members, especially C/EBPbeta and CHOP (C/EBP homologous protein), and overexpression of either C/EBPbeta or CHOP significantly attenuated TNF-alpha-triggered NF-kappaB activation. Tacrolimus 8-13 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 242-251 19454717-11 2009 These results suggested that CsA and FK506 confer insensitiveness to TNF-alpha on resident cells through UPR-dependent induction of the C/EBP family members. Tacrolimus 37-42 CCAAT/enhancer binding protein (C/EBP), beta Mus musculus 136-141 19454720-2 2009 The objective of this study was to comprehensively investigate FcepsilonRI-mediated chemokine induction in human mast cells and the effect of a corticosteroid (dexamethasone) and a calcineurin inhibitor (FK506). Tacrolimus 204-209 calcineurin binding protein 1 Homo sapiens 181-202 19454720-7 2009 In contrast, induction of CCL1, CCL3, CCL4, and CCL18 was significantly inhibited by FK506 but, with the exception of CCL1, was enhanced by dexamethasone. Tacrolimus 85-90 C-C motif chemokine ligand 1 Homo sapiens 48-52 19084239-2 2009 Previously, intestines from donors receiving Tacrolimus revealed improved morphology and abrogated nuclear factor kappa B (NF-kappaB) activation. Tacrolimus 45-55 nuclear factor kappa B subunit 1 Homo sapiens 99-121 19454720-6 2009 Induction of CCL2, CCL7, CXCL3, and CXCL8 by anti-IgE was significantly inhibited by dexamethasone but was enhanced by FK506. Tacrolimus 119-124 C-C motif chemokine ligand 2 Homo sapiens 13-17 19084239-2 2009 Previously, intestines from donors receiving Tacrolimus revealed improved morphology and abrogated nuclear factor kappa B (NF-kappaB) activation. Tacrolimus 45-55 nuclear factor kappa B subunit 1 Homo sapiens 123-132 19400811-2 2009 Recently in Japan tacrolimus ointment (0.03%) as a TCI was approved for use in children aged > or =2 years. Tacrolimus 18-28 latexin Homo sapiens 51-54 19235203-8 2009 Treatment of BPAEC with Cn inhibitors (cyclosporin A and FK506) hindered recovery of the cells from thrombin-induced EC dysfunction. Tacrolimus 57-62 coagulation factor II, thrombin Bos taurus 100-108 19545678-0 2009 Effect of CYP3A5 genotype on renal allograft recipients treated with tacrolimus. Tacrolimus 69-79 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 19545678-1 2009 OBJECTIVE: Tacrolimus concentrations are associated with CYP3A5 genotype. Tacrolimus 11-21 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 57-63 19545678-14 2009 CONCLUSION: CYP3A5 influenced the blood concentrations of tacrolimus. Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 19384264-0 2009 CYP3A5*1/*3 genotype influences the blood concentration of tacrolimus in response to metabolic inhibition by ketoconazole. Tacrolimus 59-69 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 19384264-10 2009 CONCLUSION: We show that the CYP3A5*1/*3 polymorphism is an important determinant of the response to inhibition of tacrolimus metabolism by ketoconazole, with a 30% greater inhibition in those lacking *1 allele. Tacrolimus 115-125 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 29-35 19492085-9 2009 Accordingly, the calcineurin inhibitor FK506 blocks both Drp1 dephosphorylation and loss of mitochondrial integrity in PINK1 deficient cells but does not fully rescue mitochondrial membrane potential. Tacrolimus 39-44 dynamin 1 like Homo sapiens 57-61 19492085-9 2009 Accordingly, the calcineurin inhibitor FK506 blocks both Drp1 dephosphorylation and loss of mitochondrial integrity in PINK1 deficient cells but does not fully rescue mitochondrial membrane potential. Tacrolimus 39-44 PTEN induced kinase 1 Homo sapiens 119-124 19125240-0 2009 CYP3A5 *1 allele associated with tacrolimus trough concentrations but not subclinical acute rejection or chronic allograft nephropathy in Japanese renal transplant recipients. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 19125240-1 2009 PURPOSE: We assessed reported associations of CYP3A5 *1 allele with a delay in achieving target tacrolimus concentrations, and occurrence of biopsy-confirmed subclinical acute rejection (SAR) and chronic allograft nephropathy (CAN) in Japanese subjects. Tacrolimus 96-106 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 19539558-8 2009 However, combined therapy with low-dose CsA (5 mg/kg) or tacrolimus (1 mg/kg) reduced significantly the expression of Foxp3 and failed to prolong the allograft survival. Tacrolimus 57-67 forkhead box P3 Mus musculus 118-123 19435458-4 2009 Many retrospective studies have shown a clear relationship between CYP3A5*1/*3 polymorphism and tacrolimus pharmacokinetics, while the influence of CYP3A5*1/*3 or CYP3A4*/*1B on ciclosporin and sirolimus exposure are still questionable. Tacrolimus 96-106 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-73 19557878-0 2009 Low-dose of tacrolimus favors the induction of functional CD4(+)CD25(+)FoxP3(+) regulatory T cells in solid-organ transplantation. Tacrolimus 12-22 CD4 molecule Homo sapiens 58-61 19557878-0 2009 Low-dose of tacrolimus favors the induction of functional CD4(+)CD25(+)FoxP3(+) regulatory T cells in solid-organ transplantation. Tacrolimus 12-22 forkhead box P3 Homo sapiens 71-76 19204148-11 2009 Finally, pretreatment with two pharmacological inhibitors of calcineurin, cyclosporin A, and FK-506, but not with MAPK inhibitors, blocked JAM-C induction in activated T cells. Tacrolimus 93-99 junctional adhesion molecule 3 Homo sapiens 139-144 19627009-6 2009 Simultaneously, the level of IL-2 and IL-4 in leukemia mice treated with FK506 and CSA decreased significantly than those of intact or leukemia control mice. Tacrolimus 73-78 interleukin 2 Mus musculus 29-33 19442221-5 2009 The equation of multiple stepwise regression was: Y (tacrolimus" blood concentration) = 34.534 - 0.247 (age) - 0.510 (weight) + 1.688 (dose) + 6.876 (recipient"s CYP3A5 genotype) - 3.097 (donor"s CYP3A5 genotype), P < 0.01. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 162-168 19442221-5 2009 The equation of multiple stepwise regression was: Y (tacrolimus" blood concentration) = 34.534 - 0.247 (age) - 0.510 (weight) + 1.688 (dose) + 6.876 (recipient"s CYP3A5 genotype) - 3.097 (donor"s CYP3A5 genotype), P < 0.01. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 196-202 19442221-6 2009 The factors impacting patient"s tacrolimus blood concentrations in a descending order are weight, recipient"s CYP3A5 genotype, dose, age, donor"s CYP3A5 genotype. Tacrolimus 32-42 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 110-116 19442221-6 2009 The factors impacting patient"s tacrolimus blood concentrations in a descending order are weight, recipient"s CYP3A5 genotype, dose, age, donor"s CYP3A5 genotype. Tacrolimus 32-42 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 146-152 19442221-7 2009 Among those, patient"s weight and recipient"s CYP3A5 genotype could significantly impact the blood concentration of tacrolimus. Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 19627009-6 2009 Simultaneously, the level of IL-2 and IL-4 in leukemia mice treated with FK506 and CSA decreased significantly than those of intact or leukemia control mice. Tacrolimus 73-78 interleukin 4 Mus musculus 38-42 19233271-11 2009 On the other hand, in vitro assays showed that cyclosporine (5.0 ug/mL) and tacrolimus (100 ng/mL) diminished the percentages of CD8+CD28- cells, with no significant effect on natural T regulatory cells. Tacrolimus 76-86 CD8a molecule Homo sapiens 129-132 19627009-7 2009 CONCLUSION: FK506 can promote the survival of HSCs in host, and the mechanism may be associated with the down regulation of IL-2 and IL-4. Tacrolimus 12-17 interleukin 2 Mus musculus 124-128 19627009-7 2009 CONCLUSION: FK506 can promote the survival of HSCs in host, and the mechanism may be associated with the down regulation of IL-2 and IL-4. Tacrolimus 12-17 interleukin 4 Mus musculus 133-137 19384171-2 2009 We have determined the genotypic frequencies of the CYP3A and ATP-binding cassette sub-family B member 1 (ABCB1) genes, which encode the CYP3A and P-gp proteins, respectively, in Korean organ transplant recipients and donors, and have assessed the influence of CYP3A and ABCB1 polymorphisms on tacrolimus concentrations. Tacrolimus 294-304 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 52-57 19688973-1 2009 AIM: P-glycoprotein, the product of the ATP-binding cassette subfamily B member 1 (ABCB1) gene (or the so-called multidrug resistance 1 gene), is an ATP-driven efflux pump contributing to the pharmacokinetics as well as the pharmacokinetics of drugs that are P-glycoprotein substrates, such as tacrolimus. Tacrolimus 294-304 ATP binding cassette subfamily B member 1 Homo sapiens 5-19 19688973-1 2009 AIM: P-glycoprotein, the product of the ATP-binding cassette subfamily B member 1 (ABCB1) gene (or the so-called multidrug resistance 1 gene), is an ATP-driven efflux pump contributing to the pharmacokinetics as well as the pharmacokinetics of drugs that are P-glycoprotein substrates, such as tacrolimus. Tacrolimus 294-304 ATP binding cassette subfamily B member 1 Homo sapiens 40-81 19688973-1 2009 AIM: P-glycoprotein, the product of the ATP-binding cassette subfamily B member 1 (ABCB1) gene (or the so-called multidrug resistance 1 gene), is an ATP-driven efflux pump contributing to the pharmacokinetics as well as the pharmacokinetics of drugs that are P-glycoprotein substrates, such as tacrolimus. Tacrolimus 294-304 ATP binding cassette subfamily B member 1 Homo sapiens 83-88 19688973-1 2009 AIM: P-glycoprotein, the product of the ATP-binding cassette subfamily B member 1 (ABCB1) gene (or the so-called multidrug resistance 1 gene), is an ATP-driven efflux pump contributing to the pharmacokinetics as well as the pharmacokinetics of drugs that are P-glycoprotein substrates, such as tacrolimus. Tacrolimus 294-304 ATP binding cassette subfamily B member 1 Homo sapiens 113-135 19688973-1 2009 AIM: P-glycoprotein, the product of the ATP-binding cassette subfamily B member 1 (ABCB1) gene (or the so-called multidrug resistance 1 gene), is an ATP-driven efflux pump contributing to the pharmacokinetics as well as the pharmacokinetics of drugs that are P-glycoprotein substrates, such as tacrolimus. Tacrolimus 294-304 ATP binding cassette subfamily B member 1 Homo sapiens 259-273 19384171-2 2009 We have determined the genotypic frequencies of the CYP3A and ATP-binding cassette sub-family B member 1 (ABCB1) genes, which encode the CYP3A and P-gp proteins, respectively, in Korean organ transplant recipients and donors, and have assessed the influence of CYP3A and ABCB1 polymorphisms on tacrolimus concentrations. Tacrolimus 294-304 ATP binding cassette subfamily B member 1 Homo sapiens 62-104 19384171-0 2009 Tacrolimus concentrations in relation to CYP3A and ABCB1 polymorphisms among solid organ transplant recipients in Korea. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-46 19384171-0 2009 Tacrolimus concentrations in relation to CYP3A and ABCB1 polymorphisms among solid organ transplant recipients in Korea. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 51-56 19384171-2 2009 We have determined the genotypic frequencies of the CYP3A and ATP-binding cassette sub-family B member 1 (ABCB1) genes, which encode the CYP3A and P-gp proteins, respectively, in Korean organ transplant recipients and donors, and have assessed the influence of CYP3A and ABCB1 polymorphisms on tacrolimus concentrations. Tacrolimus 294-304 ATP binding cassette subfamily B member 1 Homo sapiens 106-111 19384171-1 2009 BACKGROUND: Cytochrome P450 3A (CYP3A) and the drug transporter P-glycoprotein (P-gp) affect the bioavailability of tacrolimus, the most commonly used immunosuppressive agent in organ transplant recipients. Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 12-30 19384171-2 2009 We have determined the genotypic frequencies of the CYP3A and ATP-binding cassette sub-family B member 1 (ABCB1) genes, which encode the CYP3A and P-gp proteins, respectively, in Korean organ transplant recipients and donors, and have assessed the influence of CYP3A and ABCB1 polymorphisms on tacrolimus concentrations. Tacrolimus 294-304 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 137-142 19384171-1 2009 BACKGROUND: Cytochrome P450 3A (CYP3A) and the drug transporter P-glycoprotein (P-gp) affect the bioavailability of tacrolimus, the most commonly used immunosuppressive agent in organ transplant recipients. Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-37 19384171-1 2009 BACKGROUND: Cytochrome P450 3A (CYP3A) and the drug transporter P-glycoprotein (P-gp) affect the bioavailability of tacrolimus, the most commonly used immunosuppressive agent in organ transplant recipients. Tacrolimus 116-126 ATP binding cassette subfamily B member 1 Homo sapiens 64-78 19384171-2 2009 We have determined the genotypic frequencies of the CYP3A and ATP-binding cassette sub-family B member 1 (ABCB1) genes, which encode the CYP3A and P-gp proteins, respectively, in Korean organ transplant recipients and donors, and have assessed the influence of CYP3A and ABCB1 polymorphisms on tacrolimus concentrations. Tacrolimus 294-304 ATP binding cassette subfamily B member 1 Homo sapiens 147-151 19384171-1 2009 BACKGROUND: Cytochrome P450 3A (CYP3A) and the drug transporter P-glycoprotein (P-gp) affect the bioavailability of tacrolimus, the most commonly used immunosuppressive agent in organ transplant recipients. Tacrolimus 116-126 ATP binding cassette subfamily B member 1 Homo sapiens 80-84 19384171-2 2009 We have determined the genotypic frequencies of the CYP3A and ATP-binding cassette sub-family B member 1 (ABCB1) genes, which encode the CYP3A and P-gp proteins, respectively, in Korean organ transplant recipients and donors, and have assessed the influence of CYP3A and ABCB1 polymorphisms on tacrolimus concentrations. Tacrolimus 294-304 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 137-142 19384171-6 2009 Patients with the CYP3A5*3 and CYP3A5P1*3 alleles showed higher blood tacrolimus concentrations per adjusted dose ratio than did patients with wild-type alleles, among both liver transplant donors and renal transplant recipients. Tacrolimus 70-80 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 18-24 19384171-6 2009 Patients with the CYP3A5*3 and CYP3A5P1*3 alleles showed higher blood tacrolimus concentrations per adjusted dose ratio than did patients with wild-type alleles, among both liver transplant donors and renal transplant recipients. Tacrolimus 70-80 cytochrome P450 family 3 subfamily A member 51, pseudogene Homo sapiens 31-39 19384171-7 2009 CONCLUSION: The CYP3A5 genotype of the liver is considered to show the most important association with tacrolimus concentrations. Tacrolimus 103-113 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 16-22 19154728-6 2009 When cell lysate was treated with and without MTI, tacrolimus and FKBP12, FKBP12 could increase calcineurin inhibition by tacrolimus and reverse the MTI antagonism for both MTIs. Tacrolimus 51-61 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 74-80 19051071-1 2009 CONCLUSION: The calcineurin inhibitor tacrolimus (TCR) and the pineal gland hormone and antioxidant melatonin (MLT) have been shown to possess otoprotective properties against noise-induced hearing loss (NIHL). Tacrolimus 38-48 calcineurin binding protein 1 Rattus norvegicus 16-37 19154728-6 2009 When cell lysate was treated with and without MTI, tacrolimus and FKBP12, FKBP12 could increase calcineurin inhibition by tacrolimus and reverse the MTI antagonism for both MTIs. Tacrolimus 122-132 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 66-72 19154728-6 2009 When cell lysate was treated with and without MTI, tacrolimus and FKBP12, FKBP12 could increase calcineurin inhibition by tacrolimus and reverse the MTI antagonism for both MTIs. Tacrolimus 122-132 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 74-80 19154728-7 2009 These results demonstrate that FKBP12 can be rate limiting for calcineurin inhibition at high tacrolimus concentrations and that the antagonism of sirolimus and everolimus on tacrolimus based immune suppression is mediated via saturation of FKBP12. Tacrolimus 94-104 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 31-37 19154728-7 2009 These results demonstrate that FKBP12 can be rate limiting for calcineurin inhibition at high tacrolimus concentrations and that the antagonism of sirolimus and everolimus on tacrolimus based immune suppression is mediated via saturation of FKBP12. Tacrolimus 175-185 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 241-247 19161445-12 2009 In combined tacrolimus-SIR and SIR-treated patients, CTL alloreactivity was associated with less insulin independence and C-peptide production (P = 0.03). Tacrolimus 12-22 insulin Homo sapiens 97-104 19081230-0 2009 Tacrolimus causes reduced GLI1 expression and phenotypic changes in the TE 354.T basal cell carcinoma cell line. Tacrolimus 0-10 GLI family zinc finger 1 Homo sapiens 26-30 19139162-0 2009 Impact of intestinal CYP2C19 genotypes on the interaction between tacrolimus and omeprazole, but not lansoprazole, in adult living-donor liver transplant patients. Tacrolimus 66-76 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 21-28 19139162-3 2009 Conversely, the CYP2C19 polymorphisms both in the native intestine and in the graft liver little influenced the interaction between tacrolimus and lansoprazole, but CYP3A5*1 noncarriers showed higher tacrolimus concentration/dose ratio than CYP3A5*1 carriers. Tacrolimus 200-210 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 165-171 19139162-4 2009 Furthermore, our experiments in vitro revealed that lansoprazole had a stronger inhibitory effect on the CYP3A5-mediated metabolism of tacrolimus than omeprazole, although not significantly (IC(50) = 19.9 +/- 13.8 microM for lansoprazole, 53.7 +/- 6.1 microM for omeprazole). Tacrolimus 135-145 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 105-111 19139162-5 2009 Our findings suggest that intestinal and graft liver CYP2C19 plays a relatively greater role in the metabolism of omeprazole than it does for lansoprazole, so that the effects of CYP3A5 on the metabolism of tacrolimus might be masked by the interaction with omeprazole associated with the CYP2C19 genotype. Tacrolimus 207-217 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 53-60 19139162-5 2009 Our findings suggest that intestinal and graft liver CYP2C19 plays a relatively greater role in the metabolism of omeprazole than it does for lansoprazole, so that the effects of CYP3A5 on the metabolism of tacrolimus might be masked by the interaction with omeprazole associated with the CYP2C19 genotype. Tacrolimus 207-217 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 179-185 19139162-5 2009 Our findings suggest that intestinal and graft liver CYP2C19 plays a relatively greater role in the metabolism of omeprazole than it does for lansoprazole, so that the effects of CYP3A5 on the metabolism of tacrolimus might be masked by the interaction with omeprazole associated with the CYP2C19 genotype. Tacrolimus 207-217 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 289-296 19162238-5 2009 Interestingly, repeated application of tacrolimus ointment potently inhibited DfE-induced atopic dermatitis in NC/Nga mice concomitant with the inhibition of these changes in chemokine gene and protein expression levels particularly of CCL20/LARC, CCL17/TARC, and CCL11/eotaxin-1. Tacrolimus 39-49 chemokine (C-C motif) ligand 20 Mus musculus 236-241 19162238-5 2009 Interestingly, repeated application of tacrolimus ointment potently inhibited DfE-induced atopic dermatitis in NC/Nga mice concomitant with the inhibition of these changes in chemokine gene and protein expression levels particularly of CCL20/LARC, CCL17/TARC, and CCL11/eotaxin-1. Tacrolimus 39-49 chemokine (C-C motif) ligand 20 Mus musculus 242-246 19162238-5 2009 Interestingly, repeated application of tacrolimus ointment potently inhibited DfE-induced atopic dermatitis in NC/Nga mice concomitant with the inhibition of these changes in chemokine gene and protein expression levels particularly of CCL20/LARC, CCL17/TARC, and CCL11/eotaxin-1. Tacrolimus 39-49 chemokine (C-C motif) ligand 17 Mus musculus 248-253 19162238-5 2009 Interestingly, repeated application of tacrolimus ointment potently inhibited DfE-induced atopic dermatitis in NC/Nga mice concomitant with the inhibition of these changes in chemokine gene and protein expression levels particularly of CCL20/LARC, CCL17/TARC, and CCL11/eotaxin-1. Tacrolimus 39-49 chemokine (C-C motif) ligand 17 Mus musculus 254-258 19162238-5 2009 Interestingly, repeated application of tacrolimus ointment potently inhibited DfE-induced atopic dermatitis in NC/Nga mice concomitant with the inhibition of these changes in chemokine gene and protein expression levels particularly of CCL20/LARC, CCL17/TARC, and CCL11/eotaxin-1. Tacrolimus 39-49 chemokine (C-C motif) ligand 11 Mus musculus 264-269 19335435-0 2009 Effects of tacrolimus ointment on the expression of substance P, nerve growth factor, and neurotrophin-3 in atopic dermatitis. Tacrolimus 11-21 tachykinin precursor 1 Homo sapiens 52-63 19335435-0 2009 Effects of tacrolimus ointment on the expression of substance P, nerve growth factor, and neurotrophin-3 in atopic dermatitis. Tacrolimus 11-21 nerve growth factor Homo sapiens 65-84 19335435-0 2009 Effects of tacrolimus ointment on the expression of substance P, nerve growth factor, and neurotrophin-3 in atopic dermatitis. Tacrolimus 11-21 neurotrophin 3 Homo sapiens 90-104 19335435-2 2009 Objectives To investigate the effects of tacrolimus on the neuropeptides substance P (SP), nerve growth factor (NGF), and neurotrophin-3 (NT-3) in the skin, and SP and NGF in the serum, of patients with AD. Tacrolimus 41-51 neurotrophin 3 Homo sapiens 138-142 19150448-10 2009 Specific calcineurin inhibition with cyclosporine A and FK506 or NFAT inhibition with 11R-VIVIT reversed the stretch or alpha-1-adrenergic induced decrease of NGF. Tacrolimus 56-61 nerve growth factor Rattus norvegicus 159-162 19361439-0 2009 Differential responses of the backbone and side-chain conformational dynamics in FKBP12 upon binding the transition-state analog FK506: implications for transition-state stabilization and target protein recognition. Tacrolimus 129-134 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 81-87 19177155-0 2009 Tacrolimus reduces nitric oxide synthase function by binding to FKBP rather than by its calcineurin effect. Tacrolimus 0-10 FK506 binding protein 1a Mus musculus 64-68 19177155-4 2009 In mouse aortas, incubation with tacrolimus increased protein kinase C activity and basal endothelial nitric oxide synthase phosphorylation at threonine 495 but reduced basal and agonist-induced endothelial nitric oxide synthase phosphorylation at serine 1177, a mechanism known to inhibit synthase activity. Tacrolimus 33-43 nitric oxide synthase 3, endothelial cell Mus musculus 90-123 19177155-4 2009 In mouse aortas, incubation with tacrolimus increased protein kinase C activity and basal endothelial nitric oxide synthase phosphorylation at threonine 495 but reduced basal and agonist-induced endothelial nitric oxide synthase phosphorylation at serine 1177, a mechanism known to inhibit synthase activity. Tacrolimus 33-43 nitric oxide synthase 3, endothelial cell Mus musculus 195-228 19177155-7 2009 Since it is known that the FK506 binding protein (FKBP12/12.6) interacts with the ryanodine receptor to regulate calcium release, we propose this as the mechanism by which tacrolimus alters intracellular calcium and endothelial nitric oxide synthase rather than by its effect on calcineurin. Tacrolimus 172-182 FK506 binding protein 1a Mus musculus 50-56 19177155-7 2009 Since it is known that the FK506 binding protein (FKBP12/12.6) interacts with the ryanodine receptor to regulate calcium release, we propose this as the mechanism by which tacrolimus alters intracellular calcium and endothelial nitric oxide synthase rather than by its effect on calcineurin. Tacrolimus 172-182 nitric oxide synthase 3, endothelial cell Mus musculus 216-249 19188906-8 2009 The increased Th1/Th2 ratio found in I/R livers after 2 h was reverted by immunosuppressants, which also amplified the proportion of CD4(+)CD25(+)Foxp3(+) regulatory T lymphocytes at 24 h. The protective effects of both tacrolimus and sirolimus correlated well with a decreased ratio of proinflammatory to anti-inflammatory T lymphocytes, and with an increase in the Treg proportion. Tacrolimus 220-230 Cd4 molecule Rattus norvegicus 133-136 19162189-9 2009 In situ MMP-2/MMP-9 activity was significantly reduced in grafts treated with tacrolimus and rapamycin compared to controls (p<0.05). Tacrolimus 78-88 matrix metallopeptidase 2 Homo sapiens 8-13 19162189-9 2009 In situ MMP-2/MMP-9 activity was significantly reduced in grafts treated with tacrolimus and rapamycin compared to controls (p<0.05). Tacrolimus 78-88 matrix metallopeptidase 9 Homo sapiens 14-19 19162189-10 2009 Immunohistochemistry revealed a high number of CD4+ cells and high CD4/CD8 ratio in grafts pre-treated with tacrolimus. Tacrolimus 108-118 CD4 molecule Homo sapiens 47-50 19162189-10 2009 Immunohistochemistry revealed a high number of CD4+ cells and high CD4/CD8 ratio in grafts pre-treated with tacrolimus. Tacrolimus 108-118 CD4 molecule Homo sapiens 67-70 19162189-10 2009 Immunohistochemistry revealed a high number of CD4+ cells and high CD4/CD8 ratio in grafts pre-treated with tacrolimus. Tacrolimus 108-118 CD8a molecule Homo sapiens 71-74 19162189-11 2009 Donor pre-treatment with tacrolimus significantly reduces transplant arteriosclerosis and is associated with reduced in situ MMP-2/MMP-9 activity and increased number of CD4+ cells. Tacrolimus 25-35 matrix metallopeptidase 2 Homo sapiens 125-130 19162189-11 2009 Donor pre-treatment with tacrolimus significantly reduces transplant arteriosclerosis and is associated with reduced in situ MMP-2/MMP-9 activity and increased number of CD4+ cells. Tacrolimus 25-35 matrix metallopeptidase 9 Homo sapiens 131-136 19162189-11 2009 Donor pre-treatment with tacrolimus significantly reduces transplant arteriosclerosis and is associated with reduced in situ MMP-2/MMP-9 activity and increased number of CD4+ cells. Tacrolimus 25-35 CD4 molecule Homo sapiens 170-173 19376366-0 2009 Genetic polymorphisms in CYP3A5 and MDR1 genes and their correlations with plasma levels of tacrolimus and cyclosporine in renal transplant recipients. Tacrolimus 92-102 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 25-31 19376366-0 2009 Genetic polymorphisms in CYP3A5 and MDR1 genes and their correlations with plasma levels of tacrolimus and cyclosporine in renal transplant recipients. Tacrolimus 92-102 ATP binding cassette subfamily B member 1 Homo sapiens 36-40 19376366-3 2009 The bioavailability of CsA and FK506 seems to be associated with the cytocrhome P450 IIIA (CYP3A) gene. Tacrolimus 31-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 91-96 19376366-7 2009 This study showed that subjects in the FK506 group who had encoded the 1236C>T substitution in the MDR1 gene displayed 44.4% higher drug concentrations compared with ("wild-type") individuals. Tacrolimus 39-44 ATP binding cassette subfamily B member 1 Homo sapiens 102-106 19356587-6 2009 In the FKBP domain, Tyr68 points into the active site, which might explain the lowered intrinsic prolyl isomerase activity and the much lower FK506 binding affinity of the protein compared with archetype human FKBP12 (human FK506 binding protein with 12 kDa). Tacrolimus 142-147 FKBP prolyl isomerase 1A Homo sapiens 224-257 19361439-2 2009 The macrolide FK506 is a transition-state analog of the catalyzed reaction and displaces FKBP12 from its natural target proteins. Tacrolimus 14-19 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 89-95 19361439-3 2009 We compared the conformational exchange dynamics of the backbone and methyl-bearing side chains of FKBP12 in the free and FK506-bound states using NMR relaxation-dispersion experiments. Tacrolimus 122-127 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 99-105 19361439-5 2009 In FK506-bound FKBP12, the backbone is confined to a single conformation, while conformational exchange prevails for many methyl groups. Tacrolimus 3-8 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 15-21 19201607-5 2009 Utilization of the optimized spacer enable the monolithic material bearing FK506 to identify not only FKBP12 but FKBP52, calcineurin A and calcineurin B at silver stained level, while that without spacer had failed. Tacrolimus 75-80 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 102-108 19112104-4 2009 Here we report that hensin secretion and matrix assembly were inhibited by the peptidylprolyl cis-trans isomerase (PPIase) inhibitors cyclosporin A (CsA) and a derivative of cyclosporin A with modifications in the d-Ser side chain (Cs9) but not by the calcineurin pathway inhibitor FK506. Tacrolimus 282-287 deleted in malignant brain tumors 1 protein Oryctolagus cuniculus 20-26 19201607-5 2009 Utilization of the optimized spacer enable the monolithic material bearing FK506 to identify not only FKBP12 but FKBP52, calcineurin A and calcineurin B at silver stained level, while that without spacer had failed. Tacrolimus 75-80 FKBP prolyl isomerase 4 Homo sapiens 113-119 19201607-5 2009 Utilization of the optimized spacer enable the monolithic material bearing FK506 to identify not only FKBP12 but FKBP52, calcineurin A and calcineurin B at silver stained level, while that without spacer had failed. Tacrolimus 75-80 protein phosphatase 3 regulatory subunit B, alpha Homo sapiens 139-152 19335686-2 2009 But the clinical efficacy of topical tacrolimus, a newly developed calcineurin inhibitor on seborrheic dermatitis, is not well investigated yet. Tacrolimus 37-47 calcineurin binding protein 1 Homo sapiens 67-88 19473564-0 2009 Potentiation of glucocorticoid receptor (GR)-mediated signaling by the immunosuppressant tacrolimus in rheumatoid synoviocytes. Tacrolimus 89-99 nuclear receptor subfamily 3 group C member 1 Homo sapiens 16-39 19473564-0 2009 Potentiation of glucocorticoid receptor (GR)-mediated signaling by the immunosuppressant tacrolimus in rheumatoid synoviocytes. Tacrolimus 89-99 nuclear receptor subfamily 3 group C member 1 Homo sapiens 41-43 19473564-2 2009 In this study, we investigated the effects of tacrolimus on glucocorticoid receptor (GR) signaling using rheumatoid fibroblast-like synoviocytes (RA-FLS). Tacrolimus 46-56 nuclear receptor subfamily 3 group C member 1 Homo sapiens 60-83 19473564-2 2009 In this study, we investigated the effects of tacrolimus on glucocorticoid receptor (GR) signaling using rheumatoid fibroblast-like synoviocytes (RA-FLS). Tacrolimus 46-56 nuclear receptor subfamily 3 group C member 1 Homo sapiens 85-87 19473564-8 2009 Tacrolimus treatment of RA-FLS results in potentiation of GR translocation to the nucleus even in the presence of a low concentration of Dex (10-9M). Tacrolimus 0-10 nuclear receptor subfamily 3 group C member 1 Homo sapiens 58-60 19473564-9 2009 GR-associated FKBP-51 decreased after tacrolimus treatment. Tacrolimus 38-48 nuclear receptor subfamily 3 group C member 1 Homo sapiens 0-2 19473564-10 2009 Furthermore, tacrolimus also decreased the IL-1Beta-induced DNA binding activity of p65, a subunit of NF-KappaB, in the presence of 10-9 M of Dex. Tacrolimus 13-23 interleukin 1 beta Homo sapiens 43-51 19473564-10 2009 Furthermore, tacrolimus also decreased the IL-1Beta-induced DNA binding activity of p65, a subunit of NF-KappaB, in the presence of 10-9 M of Dex. Tacrolimus 13-23 RELA proto-oncogene, NF-kB subunit Homo sapiens 84-87 19473564-10 2009 Furthermore, tacrolimus also decreased the IL-1Beta-induced DNA binding activity of p65, a subunit of NF-KappaB, in the presence of 10-9 M of Dex. Tacrolimus 13-23 nuclear factor kappa B subunit 1 Homo sapiens 102-111 19052845-7 2009 The IL-1 beta inducible ADAMTS9 expression was inhibited by NFAT inhibitors, FK506 and 11Arg (11R)-VIVIT. Tacrolimus 77-82 interleukin 1 beta Homo sapiens 4-13 19052845-7 2009 The IL-1 beta inducible ADAMTS9 expression was inhibited by NFAT inhibitors, FK506 and 11Arg (11R)-VIVIT. Tacrolimus 77-82 ADAM metallopeptidase with thrombospondin type 1 motif 9 Homo sapiens 24-31 19183931-7 2009 CYP3A4 inducers can lower levels of cyclosporine or tacrolimus so much that transplant rejection occurs, and CYP3A4 inhibitors can increase their levels, leading to nephrotoxicity. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 19041926-9 2009 Immunohistochemistry for phosphorylated 3"-5"-cyclic AMP response element binding protein (pCREB) revealed that unilateral infusion of FK506 resulted in an amplification of phosphorylated CREB in the olfactory bulb 40 min after training compared with saline-infused bulbs. Tacrolimus 135-140 cAMP responsive element binding protein 1 Homo sapiens 40-89 19041926-9 2009 Immunohistochemistry for phosphorylated 3"-5"-cyclic AMP response element binding protein (pCREB) revealed that unilateral infusion of FK506 resulted in an amplification of phosphorylated CREB in the olfactory bulb 40 min after training compared with saline-infused bulbs. Tacrolimus 135-140 cAMP responsive element binding protein 1 Homo sapiens 92-96 18976293-8 2009 Allospecific CD154 + T-helper-memory cells, not CD154 + Tc-memory, were inhibited by increasing Tacrolimus concentrations (p = 0.026). Tacrolimus 96-106 CD40 ligand Homo sapiens 13-18 19103144-7 2009 Second, specific protection from exogenous protease of FKBP12 by FK506 and Hsp90 fragments by radicicol were observed. Tacrolimus 65-70 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 55-61 19114031-7 2009 Phosphorylation status of osterix was augmented by treatment with phosphatase inhibitors, FK506 and calyculin A. Tacrolimus 90-95 Sp7 transcription factor Homo sapiens 26-33 19116363-5 2009 In addition, FGF-2-induced tumor growth and metastasis was abrogated by administration of either an immunosuppressant, FK506, or a COX-2 inhibitor. Tacrolimus 119-124 fibroblast growth factor 2 Mus musculus 13-18 19148552-3 2009 In the present study, we investigated the effects of tacrolimus (FK506) and cyclosporine A, well-known immunosuppressants, on the FGF-2-induced VEGF release in these cells. Tacrolimus 53-63 fibroblast growth factor 2 Mus musculus 130-135 19087230-5 2009 Furthermore, we investigated impact of gender, age, pre-transplant diagnosis and renal function, transplant type, early post-transplant dialysis, and tacrolimus trough levels on decline in eGFR using multivariate analysis. Tacrolimus 150-160 epidermal growth factor receptor Homo sapiens 189-193 18822327-0 2009 Novel double coated nanocapsules for intestinal delivery and enhanced oral bioavailability of tacrolimus, a P-gp substrate drug. Tacrolimus 94-104 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 108-112 18822327-1 2009 This study proposes a new concept of double coated nanocapsules to improve the oral bioavailability of a P-glycoprotein (P-gp) substrate drug, tacrolimus, without modulating the physiological activity of the P-gp pump. Tacrolimus 143-153 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 105-119 18822327-1 2009 This study proposes a new concept of double coated nanocapsules to improve the oral bioavailability of a P-glycoprotein (P-gp) substrate drug, tacrolimus, without modulating the physiological activity of the P-gp pump. Tacrolimus 143-153 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 121-125 19027753-0 2009 Immunosuppressant FK506 decreases the intracellular magnesium in the human osteoblast cell by inhibiting the ERK1/2 pathway. Tacrolimus 18-23 mitogen-activated protein kinase 3 Homo sapiens 109-115 19027753-6 2009 KEY FINDINGS: FK506 (0.1 microM) did not affect cell death in HOB cells after a 24 hour treatment but decreased the level of ERK 1/2 activation. Tacrolimus 14-19 mitogen-activated protein kinase 3 Homo sapiens 125-132 19027753-11 2009 These results suggest that the inhibition of ERK phosphorylation is an essential intermediate in the effects of FK506 on magnesium. Tacrolimus 112-117 mitogen-activated protein kinase 1 Homo sapiens 45-48 19027753-12 2009 Overall, FK506 causes bone disorders partly by decreasing [Mg2+]i accompanied by the inhibition of ERK 1/2. Tacrolimus 9-14 mitogen-activated protein kinase 3 Homo sapiens 99-106 19354332-1 2009 BACKGROUND: Tacrolimus, a calcineurin inhibitor, is an immunomodulating and anti-inflammatory drug that inhibits T-cell activation and production of cytokines. Tacrolimus 12-22 calcineurin binding protein 1 Homo sapiens 26-47 19101526-1 2009 The aim of this study was to evaluate the effect of tacrolimus on recombinant tissue-plasminogen activator (rt-PA)-induced hemorrhagic transformation, and to characterize its suppressive action for hemorrhage. Tacrolimus 52-62 plasminogen activator, tissue type Rattus norvegicus 78-106 19101526-8 2009 Combined treatment of rt-PA with tacrolimus reduced the rt-PA-induced extravasation of Evans blue and preserved CD31-positive cells in the ischemic hemisphere. Tacrolimus 33-43 platelet and endothelial cell adhesion molecule 1 Rattus norvegicus 112-116 19138532-4 2009 In contrast, FK506 and MPA selectively inhibited the secretion of IL-2 and IL-13, and MPA unexpectedly increased the production of IL-1beta. Tacrolimus 13-18 interleukin 2 Homo sapiens 66-70 19138532-4 2009 In contrast, FK506 and MPA selectively inhibited the secretion of IL-2 and IL-13, and MPA unexpectedly increased the production of IL-1beta. Tacrolimus 13-18 interleukin 13 Homo sapiens 75-80 18692599-3 2009 The aim of the present study was to investigate the effects of immunosuppressive drugs cyclosporin A (CsA), tacrolimus (FK-506) and pimecrolimus on NO production through iNOS pathway in activated macrophages and fibroblasts. Tacrolimus 108-118 nitric oxide synthase 2 Homo sapiens 170-174 18692599-3 2009 The aim of the present study was to investigate the effects of immunosuppressive drugs cyclosporin A (CsA), tacrolimus (FK-506) and pimecrolimus on NO production through iNOS pathway in activated macrophages and fibroblasts. Tacrolimus 120-126 nitric oxide synthase 2 Homo sapiens 170-174 18692599-4 2009 Calcineurin inhibitors (CsA, FK-506 and pimecrolimus) inhibited NO production and iNOS expression in a concentration-dependent manner, CsA being more potent than FK-506 and pimecrolimus. Tacrolimus 29-35 nitric oxide synthase 2 Homo sapiens 82-86 19148552-0 2009 Tacrolimus but not cyclosporine A enhances FGF-2-induced VEGF release in osteoblasts. Tacrolimus 0-10 fibroblast growth factor 2 Mus musculus 43-48 19148552-0 2009 Tacrolimus but not cyclosporine A enhances FGF-2-induced VEGF release in osteoblasts. Tacrolimus 0-10 vascular endothelial growth factor A Mus musculus 57-61 19148552-3 2009 In the present study, we investigated the effects of tacrolimus (FK506) and cyclosporine A, well-known immunosuppressants, on the FGF-2-induced VEGF release in these cells. Tacrolimus 53-63 vascular endothelial growth factor A Mus musculus 144-148 19148552-3 2009 In the present study, we investigated the effects of tacrolimus (FK506) and cyclosporine A, well-known immunosuppressants, on the FGF-2-induced VEGF release in these cells. Tacrolimus 65-70 fibroblast growth factor 2 Mus musculus 130-135 19148552-3 2009 In the present study, we investigated the effects of tacrolimus (FK506) and cyclosporine A, well-known immunosuppressants, on the FGF-2-induced VEGF release in these cells. Tacrolimus 65-70 vascular endothelial growth factor A Mus musculus 144-148 19148552-4 2009 Tacrolimus, but not cyclosporine A which alone had no effect on VEGF basal levels, significantly enhanced FGF-2-stimulated VEGF release. Tacrolimus 0-10 fibroblast growth factor 2 Mus musculus 106-111 19148552-4 2009 Tacrolimus, but not cyclosporine A which alone had no effect on VEGF basal levels, significantly enhanced FGF-2-stimulated VEGF release. Tacrolimus 0-10 vascular endothelial growth factor A Mus musculus 123-127 19148552-5 2009 Tacrolimus markedly enhanced FGF-2-induced phosphorylation of SAPK/JNK without affecting the phosphorylation of p44/p42 MAP or p38 MAP kinases. Tacrolimus 0-10 fibroblast growth factor 2 Mus musculus 29-34 19148552-6 2009 SP600125, a specific inhibitor of SAPK/JNK, reduced the amplification by tacrolimus of the FGF-2-induced VEGF release. Tacrolimus 73-83 fibroblast growth factor 2 Mus musculus 91-96 19148552-6 2009 SP600125, a specific inhibitor of SAPK/JNK, reduced the amplification by tacrolimus of the FGF-2-induced VEGF release. Tacrolimus 73-83 vascular endothelial growth factor A Mus musculus 105-109 19148552-7 2009 The FGF-2-induced phosphorylation of p70 S6 kinase was suppressed by tacrolimus. Tacrolimus 69-79 fibroblast growth factor 2 Mus musculus 4-9 19148552-8 2009 These results strongly suggest that tacrolimus enhances FGF-2-stimulated VEGF release via up-regulation of SAPK/JNK through modulating p70 S6 kinase in osteoblasts. Tacrolimus 36-46 fibroblast growth factor 2 Mus musculus 56-61 19148552-8 2009 These results strongly suggest that tacrolimus enhances FGF-2-stimulated VEGF release via up-regulation of SAPK/JNK through modulating p70 S6 kinase in osteoblasts. Tacrolimus 36-46 vascular endothelial growth factor A Mus musculus 73-77 19289993-0 2009 The effect of CYP3A5 and ABCB1 single nucleotide polymorphisms on tacrolimus dose requirements in Caucasian liver transplant patients. Tacrolimus 66-76 ATP binding cassette subfamily B member 1 Homo sapiens 25-30 19660176-7 2009 Tacrolimus reduced significantly all of them and restored baseline values of nucleosome and caspase-9 in both experiments and Bcl-2 and caspase-3 when IL-1 + gamma-IF was added. Tacrolimus 0-10 caspase 9 Homo sapiens 92-101 19289993-1 2009 BACKGROUND: Tacrolimus is a substrate of cytochrome P-450 (CYP) 3A enzyme and of the drug transporter ABCB1. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-66 19289993-1 2009 BACKGROUND: Tacrolimus is a substrate of cytochrome P-450 (CYP) 3A enzyme and of the drug transporter ABCB1. Tacrolimus 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 102-107 19289993-10 2009 CONCLUSIONS: Pharmacogenetic analysis of CYP3A5 in the donor could contribute to determine the appropriate initial dosage of tacrolimus in liver transplant patients. Tacrolimus 125-135 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 41-47 19500470-5 2009 In the present study, we show that 24-h exposure to FK506 or MMF impairs glucose-stimulated insulin secretion in human islets. Tacrolimus 52-57 insulin Homo sapiens 92-99 19500470-6 2009 FK506 had acute and direct effects on insulin exocytosis, whereas MMF did not. Tacrolimus 0-5 insulin Homo sapiens 38-45 18795918-6 2009 RESULTS: AD in APOC1 mice responded to topical treatment with tacrolimus or FP. Tacrolimus 62-72 apolipoprotein C-I Mus musculus 15-20 18795918-10 2009 CONCLUSIONS: AD in APOC1 mice responds to treatment with (diluted) tacrolimus or FP; treatment with FP cream, but not tacrolimus ointment, was associated with weight loss. Tacrolimus 67-77 apolipoprotein C-I Mus musculus 19-24 19660176-7 2009 Tacrolimus reduced significantly all of them and restored baseline values of nucleosome and caspase-9 in both experiments and Bcl-2 and caspase-3 when IL-1 + gamma-IF was added. Tacrolimus 0-10 BCL2 apoptosis regulator Homo sapiens 126-131 19660176-7 2009 Tacrolimus reduced significantly all of them and restored baseline values of nucleosome and caspase-9 in both experiments and Bcl-2 and caspase-3 when IL-1 + gamma-IF was added. Tacrolimus 0-10 caspase 3 Homo sapiens 136-145 19902988-6 2009 Both the haematocrit and PXR -25385C>T single nucleotide polymorphism (SNP) were identified as significant covariates for apparent oral clearance (CL/F) of tacrolimus, which allowed improvement of prediction accuracy. Tacrolimus 159-169 nuclear receptor subfamily 1 group I member 2 Homo sapiens 25-28 19728747-9 2009 Ciclosporin and tacrolimus have distinct pharmacokinetics, but both are metabolized by intestinal and hepatic CYP3A4/3A5 and transported across the cell membrane by P-glycoprotein. Tacrolimus 16-26 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 110-116 19728747-9 2009 Ciclosporin and tacrolimus have distinct pharmacokinetics, but both are metabolized by intestinal and hepatic CYP3A4/3A5 and transported across the cell membrane by P-glycoprotein. Tacrolimus 16-26 ATP binding cassette subfamily B member 1 Homo sapiens 165-179 19052693-0 2009 Phase II study of tacrolimus and methotrexate for prophylaxis of acute graft-versus-host disease after HLA-A, B, and DRB1 genotypically mismatched unrelated bone marrow transplantation among Japanese patients. Tacrolimus 18-28 major histocompatibility complex, class I, A Homo sapiens 103-111 19874240-9 2009 Compared to the control group, electron microscopy showed cytoplasm swelling and vacuolization, and marked decrease or absence of dense-core secretory granules in beta cells in rats with diabetes mellitus induced with FK506.Compared to the control group, expression of insulin receptor of hepatic cell decreased in rats of diabetes mellitus models induced with FK506 (P<0.05). Tacrolimus 218-223 insulin receptor Rattus norvegicus 269-285 19052693-0 2009 Phase II study of tacrolimus and methotrexate for prophylaxis of acute graft-versus-host disease after HLA-A, B, and DRB1 genotypically mismatched unrelated bone marrow transplantation among Japanese patients. Tacrolimus 18-28 major histocompatibility complex, class II, DR beta 1 Homo sapiens 117-121 19052693-11 2009 This study indicates tacrolimus and methotrexate can lower the risk of severe acute GVHD after HLA-A, B, or DRB1 genotypically 1 locus mismatched UR-BMT. Tacrolimus 21-31 major histocompatibility complex, class I, A Homo sapiens 95-103 19052693-11 2009 This study indicates tacrolimus and methotrexate can lower the risk of severe acute GVHD after HLA-A, B, or DRB1 genotypically 1 locus mismatched UR-BMT. Tacrolimus 21-31 major histocompatibility complex, class II, DR beta 1 Homo sapiens 108-112 19134532-12 2009 The rate of new-onset diabetes mellitus requiring insulin treatment was higher with tacrolimus. Tacrolimus 84-94 insulin Homo sapiens 50-57 18355214-0 2009 Localized pemphigoid (Brunsting-Perry type) with IgG antibody to BP180 NC16a domain resembling lupus erythematosus successfully treated with topical tacrolimus therapy. Tacrolimus 149-159 collagen type XVII alpha 1 chain Homo sapiens 65-70 19768190-5 2009 Tacrolimus, another calcineurin inhibitor widely used in transplantation, induced TGF-beta secretion by tumor cells and promoted metastases in the SCID- beige mice. Tacrolimus 0-10 transforming growth factor, beta 1 Mus musculus 82-90 19149927-0 2009 [Effect of tacrolimus on growth-associated protein-43 expression in the hippocampus of neonatal rats with hypoxic-ischemic brain damage]. Tacrolimus 11-21 growth associated protein 43 Rattus norvegicus 25-53 19652938-4 2009 The last mentioned immunological hypothesis has increasing importance, not at least based on the fact that the application of a topical calcineurin inhibitor (tacrolimus) improves the uremic pruritus. Tacrolimus 159-169 calcineurin binding protein 1 Homo sapiens 136-157 19149927-10 2009 The GAP-43 expression in the FK506 intervention group was significantly higher than that in the HIBD group 72 hrs and 7 days after HIBD. Tacrolimus 29-34 growth associated protein 43 Rattus norvegicus 4-10 19081378-1 2008 FK506-binding protein 12 (FKBP12) binds the immunosuppressant drugs FK506 and rapamycin and regulates several signaling pathways, including mammalian target of rapamycin (mTOR) signaling. Tacrolimus 0-5 FKBP prolyl isomerase 1A Homo sapiens 26-32 19081378-1 2008 FK506-binding protein 12 (FKBP12) binds the immunosuppressant drugs FK506 and rapamycin and regulates several signaling pathways, including mammalian target of rapamycin (mTOR) signaling. Tacrolimus 0-5 mechanistic target of rapamycin kinase Homo sapiens 140-169 19081378-1 2008 FK506-binding protein 12 (FKBP12) binds the immunosuppressant drugs FK506 and rapamycin and regulates several signaling pathways, including mammalian target of rapamycin (mTOR) signaling. Tacrolimus 0-5 mechanistic target of rapamycin kinase Homo sapiens 171-175 19067682-0 2008 Influence of CYP3A5 genetic polymorphism on tacrolimus daily dose requirements and acute rejection in renal graft recipients. Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 19067682-3 2008 Our objective was to determine the influence of CYP3A5 and ABCB1 genetic polymorphisms on tacrolimus daily requirements and on transplantation outcome. Tacrolimus 90-100 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 48-54 19067682-3 2008 Our objective was to determine the influence of CYP3A5 and ABCB1 genetic polymorphisms on tacrolimus daily requirements and on transplantation outcome. Tacrolimus 90-100 ATP binding cassette subfamily B member 1 Homo sapiens 59-64 19067682-6 2008 At 1-month post-transplantation, tacrolimus daily dose was higher for patients with CYP3A5*1/*1 genotype compared to CYP3A5*3/*3 genotype (0.26 +/- 0.03 versus 0.16 +/- 0.01 mg/kg/day, respectively, P < 0.0001). Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 84-90 19067682-6 2008 At 1-month post-transplantation, tacrolimus daily dose was higher for patients with CYP3A5*1/*1 genotype compared to CYP3A5*3/*3 genotype (0.26 +/- 0.03 versus 0.16 +/- 0.01 mg/kg/day, respectively, P < 0.0001). Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 117-123 19067682-11 2008 CYP3A5 genetic polymorphism appeared in our study to affect tacrolimus daily dose requirements and transplantation outcome. Tacrolimus 60-70 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 24459527-9 2008 TRPM6 is regulated at the transcriptional level by acid-base status, 17beta-estradiol, and both FK506 and cyclosporine. Tacrolimus 96-101 transient receptor potential cation channel subfamily M member 6 Homo sapiens 0-5 18760290-1 2008 FK1706, a derivative of FK506, is a non-immunosuppressive immunophilin ligand with significant neurotrophic activity mediated via FKBP-52 and the RAS/RAF/MAPK signaling pathway. Tacrolimus 24-29 FKBP prolyl isomerase 4 Rattus norvegicus 130-137 19114346-8 2008 In terms of pharmacogenomics, the major factors affecting tacrolimus concentration/dosage included MDR1 3435, MDR1 2677 and MDR1 1236 polymorphisms, which vastly varied between the patients early after the operation. Tacrolimus 58-68 ATP binding cassette subfamily B member 1 Homo sapiens 99-103 19114346-8 2008 In terms of pharmacogenomics, the major factors affecting tacrolimus concentration/dosage included MDR1 3435, MDR1 2677 and MDR1 1236 polymorphisms, which vastly varied between the patients early after the operation. Tacrolimus 58-68 ATP binding cassette subfamily B member 1 Homo sapiens 110-114 19114346-8 2008 In terms of pharmacogenomics, the major factors affecting tacrolimus concentration/dosage included MDR1 3435, MDR1 2677 and MDR1 1236 polymorphisms, which vastly varied between the patients early after the operation. Tacrolimus 58-68 ATP binding cassette subfamily B member 1 Homo sapiens 110-114 19114346-9 2008 Of these polymorphic sites, CYP3A5 produced only minor effects on tacrolimus concentration/dosage, and was not included as an active factor until the stable phase (over 1 year) following the transplantation; MDR1 3435 was found to be the predominant factor affecting tacrolimus metabolism in the stable phase. Tacrolimus 66-76 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 28-34 19005411-9 2008 Tacrolimus reduced the expression of troponin T type 2 and desmin during cardiomyogenic differentiation of MSC, whereas MPA decreased the deposition of calcified minerals during osteogenic differentiation. Tacrolimus 0-10 desmin Homo sapiens 37-65 19100409-1 2008 We have designed a protocol for ABO-incompatible kidney transplantations based on preoperative plasmapheresis with a tacrolimus/mycophenolate mofetil/methylprednisolone/basiliximab protocol using low-dose rituximab (200 mg/body) instead of splenectomy to prevent antibody-mediated acute rejection. Tacrolimus 117-127 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 32-35 19100412-0 2008 Different effect of cyclosporine and tacrolimus on renal expression of P-glycoprotein in human kidney transplantation. Tacrolimus 37-47 ATP binding cassette subfamily B member 1 Homo sapiens 71-85 19100412-1 2008 OBJECTIVE: To investigate the differential effects of cyclosporine (CsA) and tacrolimus (TAC) on renal expression of P-glycoprotein (P-gp) in a cohort of kidney transplant recipients. Tacrolimus 77-87 ATP binding cassette subfamily B member 1 Homo sapiens 117-131 19100412-1 2008 OBJECTIVE: To investigate the differential effects of cyclosporine (CsA) and tacrolimus (TAC) on renal expression of P-glycoprotein (P-gp) in a cohort of kidney transplant recipients. Tacrolimus 77-87 ATP binding cassette subfamily B member 1 Homo sapiens 133-137 19100412-1 2008 OBJECTIVE: To investigate the differential effects of cyclosporine (CsA) and tacrolimus (TAC) on renal expression of P-glycoprotein (P-gp) in a cohort of kidney transplant recipients. Tacrolimus 89-92 ATP binding cassette subfamily B member 1 Homo sapiens 117-131 19100412-1 2008 OBJECTIVE: To investigate the differential effects of cyclosporine (CsA) and tacrolimus (TAC) on renal expression of P-glycoprotein (P-gp) in a cohort of kidney transplant recipients. Tacrolimus 89-92 ATP binding cassette subfamily B member 1 Homo sapiens 133-137 19100455-8 2008 All 3 recipients were given FK506 (tacrolimus) regularly combined with periodic mycophenolate mofetil. Tacrolimus 28-33 paired box 5 Homo sapiens 0-5 19100455-8 2008 All 3 recipients were given FK506 (tacrolimus) regularly combined with periodic mycophenolate mofetil. Tacrolimus 35-45 paired box 5 Homo sapiens 0-5 19005401-1 2008 BACKGROUND: Bioavailability of tacrolimus (Tac) and cyclosporine is determined by cytochrome P450IIIA and by P-glycoprotein encoded by the CYP3A4/CYP3A5 and ABCB1 genes. Tacrolimus 31-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 139-145 19005401-1 2008 BACKGROUND: Bioavailability of tacrolimus (Tac) and cyclosporine is determined by cytochrome P450IIIA and by P-glycoprotein encoded by the CYP3A4/CYP3A5 and ABCB1 genes. Tacrolimus 31-41 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 146-152 18948815-10 2008 Multivariate analysis, including all patients (tacrolimus + cyclosporine) characterized higher exposure to immunosuppression (P = 0.01), alpha-fetoprotein levels (P = 0.001), tumor grading (P = 0.009), and microvascular invasion (P = 0.04) as independent predictors of HCC recurrence. Tacrolimus 47-57 alpha fetoprotein Homo sapiens 137-154 19005401-1 2008 BACKGROUND: Bioavailability of tacrolimus (Tac) and cyclosporine is determined by cytochrome P450IIIA and by P-glycoprotein encoded by the CYP3A4/CYP3A5 and ABCB1 genes. Tacrolimus 31-41 ATP binding cassette subfamily B member 1 Homo sapiens 157-162 19038099-7 2008 For tacrolimus, it has been demonstrated that the daily dose needed to achieve adequate trough levels are correlated with cytochrome P450 3A5 expression and therefore the presence of *1 allele. Tacrolimus 4-14 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 122-141 18568343-9 2008 When subjects were stratified by calcineurin inhibitor status, the UGT1A8*2 effect was only apparent in the tacrolimus group (p < 0.01). Tacrolimus 108-118 UDP glucuronosyltransferase family 1 member A8 Homo sapiens 67-73 18703031-9 2008 After treatment with FK506, Akt and Bad dephosphorylation was greatly reduced. Tacrolimus 21-26 AKT serine/threonine kinase 1 Rattus norvegicus 28-31 19010156-0 2008 Clinical relevance and prevalence of polymorphisms in CYP3A5 and MDR1 genes that encode tacrolimus biotransformation enzymes in liver transplant recipients. Tacrolimus 88-98 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 54-60 19010156-0 2008 Clinical relevance and prevalence of polymorphisms in CYP3A5 and MDR1 genes that encode tacrolimus biotransformation enzymes in liver transplant recipients. Tacrolimus 88-98 ATP binding cassette subfamily B member 1 Homo sapiens 65-69 19010156-1 2008 OBJECTIVES: To study the prevalence and clinical significance of polymorphisms in the CYP3A5 and MDR1 genes in liver transplant patients and their donors; to determine the relative importance of genes from the donor and the recipient; to assess the relationship of polymorphisms with the variability of concentration/dose of tacrolimus for optimization and individualization regimens. Tacrolimus 325-335 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 86-92 19010156-10 2008 CONCLUSIONS: For CYP3A5, the presence of the A allele appeared to be related to greater requirements for tacrolimus in the early days after transplantation. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 17-23 18832697-6 2008 This was evidenced by our findings that agents suppressing NFAT activity (e.g., cyclosporin A and FK506) enhanced TCR-mediated GITR expression, whereas agents enhancing NFAT activity (e.g., lithium chloride) suppressed TCR-mediated GITR up-regulation. Tacrolimus 98-103 TNF receptor superfamily member 18 Homo sapiens 127-131 18703143-9 2008 The insulin resistance level in tacrolimus-treated recipients was markedly higher than CsA-treated recipients, and there was a positive correlation between the blood concentration of tacrolimus and insulin resistance level. Tacrolimus 32-42 insulin Homo sapiens 4-11 18703143-9 2008 The insulin resistance level in tacrolimus-treated recipients was markedly higher than CsA-treated recipients, and there was a positive correlation between the blood concentration of tacrolimus and insulin resistance level. Tacrolimus 32-42 insulin Homo sapiens 198-205 18703143-9 2008 The insulin resistance level in tacrolimus-treated recipients was markedly higher than CsA-treated recipients, and there was a positive correlation between the blood concentration of tacrolimus and insulin resistance level. Tacrolimus 183-193 insulin Homo sapiens 4-11 18703143-9 2008 The insulin resistance level in tacrolimus-treated recipients was markedly higher than CsA-treated recipients, and there was a positive correlation between the blood concentration of tacrolimus and insulin resistance level. Tacrolimus 183-193 insulin Homo sapiens 198-205 18703143-12 2008 CONCLUSION: It is shown in the study that obesity or overweight, tacrolimus (especially when its blood concentration was high) were risk factors resulting in insulin resistance in kidney transplant recipients. Tacrolimus 65-75 insulin Homo sapiens 158-165 18704002-0 2008 Effects of CYP3A5 and MDR1 single nucleotide polymorphisms on drug interactions between tacrolimus and fluconazole in renal allograft recipients. Tacrolimus 88-98 ATP binding cassette subfamily B member 1 Homo sapiens 22-26 18926413-1 2008 BACKGROUND: The purpose of this study was to determine the utility of post-transplant serum soluble CD30 levels as a biomarker for the development of the bronchiolitis obliterans syndrome (BOS) after lung transplantation during a tacrolimus/mycophenolate mofetil-based regimen. Tacrolimus 230-240 TNF receptor superfamily member 8 Homo sapiens 100-104 18827354-0 2008 Effect of tacrolimus on activity and expression of P-glycoprotein and ATP-binding cassette transporter A5 (ABCA5) proteins in hematoencephalic barrier cells. Tacrolimus 10-20 ATP binding cassette subfamily B member 1 Homo sapiens 51-65 18827354-0 2008 Effect of tacrolimus on activity and expression of P-glycoprotein and ATP-binding cassette transporter A5 (ABCA5) proteins in hematoencephalic barrier cells. Tacrolimus 10-20 ATP binding cassette subfamily A member 5 Homo sapiens 107-112 18827354-4 2008 We investigated the effect of this therapeutic dose of tacrolimus on the expression and activity of the multidrug resistance protein 1 (MDR1 or Pgp, P-glycoprotein) and ATP-binding cassette transporters A5 (ABCA5) in human brain microvascular endothelial cells (HBMEC), derived from Blood-Brain Barrier (BBB) endothelium, these being the most predominantly expressed transcripts in these cells. Tacrolimus 55-65 ATP binding cassette subfamily B member 1 Homo sapiens 104-134 18827354-4 2008 We investigated the effect of this therapeutic dose of tacrolimus on the expression and activity of the multidrug resistance protein 1 (MDR1 or Pgp, P-glycoprotein) and ATP-binding cassette transporters A5 (ABCA5) in human brain microvascular endothelial cells (HBMEC), derived from Blood-Brain Barrier (BBB) endothelium, these being the most predominantly expressed transcripts in these cells. Tacrolimus 55-65 ATP binding cassette subfamily B member 1 Homo sapiens 136-140 18827354-4 2008 We investigated the effect of this therapeutic dose of tacrolimus on the expression and activity of the multidrug resistance protein 1 (MDR1 or Pgp, P-glycoprotein) and ATP-binding cassette transporters A5 (ABCA5) in human brain microvascular endothelial cells (HBMEC), derived from Blood-Brain Barrier (BBB) endothelium, these being the most predominantly expressed transcripts in these cells. Tacrolimus 55-65 phosphoglycolate phosphatase Homo sapiens 144-147 18827354-4 2008 We investigated the effect of this therapeutic dose of tacrolimus on the expression and activity of the multidrug resistance protein 1 (MDR1 or Pgp, P-glycoprotein) and ATP-binding cassette transporters A5 (ABCA5) in human brain microvascular endothelial cells (HBMEC), derived from Blood-Brain Barrier (BBB) endothelium, these being the most predominantly expressed transcripts in these cells. Tacrolimus 55-65 ATP binding cassette subfamily B member 1 Homo sapiens 149-163 18827354-4 2008 We investigated the effect of this therapeutic dose of tacrolimus on the expression and activity of the multidrug resistance protein 1 (MDR1 or Pgp, P-glycoprotein) and ATP-binding cassette transporters A5 (ABCA5) in human brain microvascular endothelial cells (HBMEC), derived from Blood-Brain Barrier (BBB) endothelium, these being the most predominantly expressed transcripts in these cells. Tacrolimus 55-65 ATP binding cassette subfamily A member 5 Homo sapiens 169-205 18827354-4 2008 We investigated the effect of this therapeutic dose of tacrolimus on the expression and activity of the multidrug resistance protein 1 (MDR1 or Pgp, P-glycoprotein) and ATP-binding cassette transporters A5 (ABCA5) in human brain microvascular endothelial cells (HBMEC), derived from Blood-Brain Barrier (BBB) endothelium, these being the most predominantly expressed transcripts in these cells. Tacrolimus 55-65 ATP binding cassette subfamily A member 5 Homo sapiens 207-212 18827354-5 2008 The expression and activity of MDR1 transporter decreased with 30 ng/ml tacrolimus. Tacrolimus 72-82 ATP binding cassette subfamily B member 1 Homo sapiens 31-35 18717725-11 2008 Tacrolimus is an immunosuppressant acting specifically in RyR antibody positive patients through enhancing RyR-related sarcoplasmic calcium release that in theory might be blocked by RyR antibodies, causing symptomatic relief in paraneoplastic MG. Tacrolimus 0-10 ryanodine receptor 2 Homo sapiens 58-61 18717725-11 2008 Tacrolimus is an immunosuppressant acting specifically in RyR antibody positive patients through enhancing RyR-related sarcoplasmic calcium release that in theory might be blocked by RyR antibodies, causing symptomatic relief in paraneoplastic MG. Tacrolimus 0-10 ryanodine receptor 2 Homo sapiens 107-110 18717725-11 2008 Tacrolimus is an immunosuppressant acting specifically in RyR antibody positive patients through enhancing RyR-related sarcoplasmic calcium release that in theory might be blocked by RyR antibodies, causing symptomatic relief in paraneoplastic MG. Tacrolimus 0-10 ryanodine receptor 2 Homo sapiens 107-110 18704002-2 2008 The aim of this study was to examine the influence of the CYP3A4, CYP3A5, and MDR1 single nucleotide polymorphisms on changes in tacrolimus exposure and dosing in renal allograft recipients treated with fluconazole. Tacrolimus 129-139 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 58-64 18704002-2 2008 The aim of this study was to examine the influence of the CYP3A4, CYP3A5, and MDR1 single nucleotide polymorphisms on changes in tacrolimus exposure and dosing in renal allograft recipients treated with fluconazole. Tacrolimus 129-139 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 66-72 18704002-2 2008 The aim of this study was to examine the influence of the CYP3A4, CYP3A5, and MDR1 single nucleotide polymorphisms on changes in tacrolimus exposure and dosing in renal allograft recipients treated with fluconazole. Tacrolimus 129-139 ATP binding cassette subfamily B member 1 Homo sapiens 78-82 18704002-6 2008 Homozygous CYP3A5*3 carriers experienced a significant decrease of weight-corrected tacrolimus dose requirements during fluconazole administration (54.7+/-23.7% from baseline, P<0.05) in contrast to heterozygous carriers of CYP3A5*1 (25.1+/-29.9%; P=0.07 between CYP3A5*3/*3 and CYP3A5*3/*1 genotypes). Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 11-17 18704002-8 2008 Significantly more CYP3A5*3/*3 carriers were exposed to tacrolimus dose-uncorrected trough blood tacrolimus concentration value greater than or equal to 15 ng/ml during administration of fluconazole compared with CYP3A5*3/*1 carriers (73.9 vs. 16.7%, P=0.01). Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 19-25 18704002-8 2008 Significantly more CYP3A5*3/*3 carriers were exposed to tacrolimus dose-uncorrected trough blood tacrolimus concentration value greater than or equal to 15 ng/ml during administration of fluconazole compared with CYP3A5*3/*1 carriers (73.9 vs. 16.7%, P=0.01). Tacrolimus 97-107 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 19-25 18704002-9 2008 CONCLUSION: In renal allograft recipients the CYP3A5*3/*1 genotype is associated with a reduced susceptibility for the inhibitory effects of fluconazole on tacrolimus metabolism, thereby identifying a genetic determinant of the clinical variability of CYP3A-mediated drug interactions. Tacrolimus 156-166 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 18929812-1 2008 OBJECTIVE: Tacrolimus, a potent calcineurin inhibitor, is a widely used immunosuppressant. Tacrolimus 11-21 calcineurin binding protein 1 Homo sapiens 32-53 18581211-1 2008 PURPOSE: To design a double-coated nanoparticulate delivery system of tacrolimus capable of overcoming the P-glycoprotein pump and CYP3A barriers without affecting their physiological activities. Tacrolimus 70-80 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 131-136 18929848-8 2008 FK506-induced cell death was confirmed by apoptosis characterized by nuclear fragmentation and caspase-3 protease activation. Tacrolimus 0-5 caspase 3 Homo sapiens 95-104 18929848-14 2008 In conclusion, FK506-induced cell death was apoptotic, characterized by nuclear fragmentation and caspase-3 activation. Tacrolimus 15-20 caspase 3 Homo sapiens 98-107 18929848-15 2008 FK506 induced G0/G1 phase cell cycle arrest via expression of CDK4 and cyclin D1. Tacrolimus 0-5 cyclin dependent kinase 4 Homo sapiens 62-66 18929848-15 2008 FK506 induced G0/G1 phase cell cycle arrest via expression of CDK4 and cyclin D1. Tacrolimus 0-5 cyclin D1 Homo sapiens 71-80 18845086-0 2008 [Regulatory function of tacrolimus and CsA on CD4/CD8 T lymphocyte subgroups and costimulators on them in allo-liver recipients]. Tacrolimus 24-34 CD4 molecule Homo sapiens 46-49 18845086-0 2008 [Regulatory function of tacrolimus and CsA on CD4/CD8 T lymphocyte subgroups and costimulators on them in allo-liver recipients]. Tacrolimus 24-34 CD8a molecule Homo sapiens 50-53 18845086-1 2008 AIM: To explore the regulatory function of FK506 and CsA on CD4/CD8 T lymphocyte subgroups and co-stimulators on them. Tacrolimus 43-48 CD4 molecule Homo sapiens 60-63 18845086-1 2008 AIM: To explore the regulatory function of FK506 and CsA on CD4/CD8 T lymphocyte subgroups and co-stimulators on them. Tacrolimus 43-48 CD8a molecule Homo sapiens 64-67 18845086-6 2008 Between two treatment group, the expression of CD4(+)T cells and the expression of CD28 and ICOS on CD8(+)T cells in CsA-treated group were much higher than those in FK506-treated group (P<0.05), and there was no significant difference between two treatment groups in other indexes. Tacrolimus 166-171 CD8a molecule Homo sapiens 100-103 18845086-9 2008 FK506 can not only inhibit the expression of positive co-stimulatory molecules CD28 and ICOS but also promote the expression of negative co-stimulatory molecule CD152, while CsA can exert its immunosuppressive effect mainly through promoting the expression of CD152. Tacrolimus 0-5 CD28 molecule Homo sapiens 79-83 18845086-9 2008 FK506 can not only inhibit the expression of positive co-stimulatory molecules CD28 and ICOS but also promote the expression of negative co-stimulatory molecule CD152, while CsA can exert its immunosuppressive effect mainly through promoting the expression of CD152. Tacrolimus 0-5 inducible T cell costimulator Homo sapiens 88-92 18845086-9 2008 FK506 can not only inhibit the expression of positive co-stimulatory molecules CD28 and ICOS but also promote the expression of negative co-stimulatory molecule CD152, while CsA can exert its immunosuppressive effect mainly through promoting the expression of CD152. Tacrolimus 0-5 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 161-166 18845086-9 2008 FK506 can not only inhibit the expression of positive co-stimulatory molecules CD28 and ICOS but also promote the expression of negative co-stimulatory molecule CD152, while CsA can exert its immunosuppressive effect mainly through promoting the expression of CD152. Tacrolimus 0-5 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 260-265 18582853-6 2008 Application to rats of two inhibitors of calcineurin (tacrolimus-FK506 and cyclosporin A) demonstrated that the mRNA levels of both the AChE catalytic subunit and ColQ in the extrajunctional regions of the soleus muscle are regulated by the calcineurin signaling pathway, but in a reciprocal way. Tacrolimus 54-64 acetylcholinesterase Rattus norvegicus 136-140 18582853-6 2008 Application to rats of two inhibitors of calcineurin (tacrolimus-FK506 and cyclosporin A) demonstrated that the mRNA levels of both the AChE catalytic subunit and ColQ in the extrajunctional regions of the soleus muscle are regulated by the calcineurin signaling pathway, but in a reciprocal way. Tacrolimus 54-64 collagen like tail subunit of asymmetric acetylcholinesterase Rattus norvegicus 163-167 18582853-6 2008 Application to rats of two inhibitors of calcineurin (tacrolimus-FK506 and cyclosporin A) demonstrated that the mRNA levels of both the AChE catalytic subunit and ColQ in the extrajunctional regions of the soleus muscle are regulated by the calcineurin signaling pathway, but in a reciprocal way. Tacrolimus 65-70 acetylcholinesterase Rattus norvegicus 136-140 18582853-6 2008 Application to rats of two inhibitors of calcineurin (tacrolimus-FK506 and cyclosporin A) demonstrated that the mRNA levels of both the AChE catalytic subunit and ColQ in the extrajunctional regions of the soleus muscle are regulated by the calcineurin signaling pathway, but in a reciprocal way. Tacrolimus 65-70 collagen like tail subunit of asymmetric acetylcholinesterase Rattus norvegicus 163-167 18556719-1 2008 Aprepitant (AP) is a known inhibitor of cytochrome P450 3A4 which may affect tacrolimus metabolism. Tacrolimus 77-87 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-59 18684975-2 2008 Herein we demonstrate that the CNI cyclosporin A and tacrolimus (FK506), independent of TGFbeta synthesis, rapidly activate TGFbeta/Smad signaling in cultured mesangial cells and in whole kidney samples from CNI-treated rats. Tacrolimus 53-63 transforming growth factor, beta 1 Rattus norvegicus 88-95 18790202-0 2008 Generic tacrolimus (Pan Graf) in renal transplantation: an experience of 155 recipients in India. Tacrolimus 8-18 Rho GTPase activating protein 26 Homo sapiens 24-28 18790202-2 2008 However, tacrolimus (Pan Graf) has only been available in India for the last 2 years. Tacrolimus 9-19 Rho GTPase activating protein 26 Homo sapiens 25-29 18684975-2 2008 Herein we demonstrate that the CNI cyclosporin A and tacrolimus (FK506), independent of TGFbeta synthesis, rapidly activate TGFbeta/Smad signaling in cultured mesangial cells and in whole kidney samples from CNI-treated rats. Tacrolimus 53-63 transforming growth factor, beta 1 Rattus norvegicus 124-131 18684975-2 2008 Herein we demonstrate that the CNI cyclosporin A and tacrolimus (FK506), independent of TGFbeta synthesis, rapidly activate TGFbeta/Smad signaling in cultured mesangial cells and in whole kidney samples from CNI-treated rats. Tacrolimus 53-63 SMAD family member 4 Rattus norvegicus 132-136 18684975-2 2008 Herein we demonstrate that the CNI cyclosporin A and tacrolimus (FK506), independent of TGFbeta synthesis, rapidly activate TGFbeta/Smad signaling in cultured mesangial cells and in whole kidney samples from CNI-treated rats. Tacrolimus 65-70 transforming growth factor, beta 1 Rattus norvegicus 124-131 18684975-2 2008 Herein we demonstrate that the CNI cyclosporin A and tacrolimus (FK506), independent of TGFbeta synthesis, rapidly activate TGFbeta/Smad signaling in cultured mesangial cells and in whole kidney samples from CNI-treated rats. Tacrolimus 65-70 SMAD family member 4 Rattus norvegicus 132-136 18342291-1 2008 BACKGROUND: Gingival crevicular fluid (GCF) levels of transforming growth factor-beta(1) (TGF-beta(1)) have been previously investigated in relation to the pathogenesis of cyclosporine-A (CsA)-induced gingival overgrowth (GO) but no clinical data are available regarding the GCF levels of TGF-beta(1) in patients treated with tacrolimus (Tac). Tacrolimus 326-336 transforming growth factor beta 1 Homo sapiens 54-88 18342291-1 2008 BACKGROUND: Gingival crevicular fluid (GCF) levels of transforming growth factor-beta(1) (TGF-beta(1)) have been previously investigated in relation to the pathogenesis of cyclosporine-A (CsA)-induced gingival overgrowth (GO) but no clinical data are available regarding the GCF levels of TGF-beta(1) in patients treated with tacrolimus (Tac). Tacrolimus 326-336 transforming growth factor beta 1 Homo sapiens 90-101 18349138-0 2008 FK506 can activate transforming growth factor-beta signalling in vascular smooth muscle cells and promote proliferation. Tacrolimus 0-5 transforming growth factor beta 1 Homo sapiens 19-50 18429967-8 2008 The CYP3A5 polymorphism may be associated with the time-dependent changes in the oral clearance of tacrolimus, suggesting that genotyping of this polymorphism is useful for determining the appropriate dose of tacrolimus in both the early and maintenance stages after renal transplantation. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 18429967-8 2008 The CYP3A5 polymorphism may be associated with the time-dependent changes in the oral clearance of tacrolimus, suggesting that genotyping of this polymorphism is useful for determining the appropriate dose of tacrolimus in both the early and maintenance stages after renal transplantation. Tacrolimus 209-219 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 18349138-10 2008 FK506 enhanced beta-catenin levels and activated the extracellular signal-regulated kinase, Akt, and mammalian target of rapamycin kinase, which are important effectors of proliferation. Tacrolimus 0-5 catenin beta 1 Homo sapiens 15-27 18349138-10 2008 FK506 enhanced beta-catenin levels and activated the extracellular signal-regulated kinase, Akt, and mammalian target of rapamycin kinase, which are important effectors of proliferation. Tacrolimus 0-5 AKT serine/threonine kinase 1 Homo sapiens 92-95 18349138-12 2008 We also demonstrate that FK506 activates the TGF-beta signal in VSMCs and that, through this mechanism, it stimulates cell proliferation. Tacrolimus 25-30 transforming growth factor beta 1 Homo sapiens 45-53 18656798-9 2008 Tacrolimus prophylaxis was associated with inhibition of recruitment of CD4+, CD8+ and interleukin-2R+ inflammatory cells into the allografts, suggesting a central role for interleukin-2 in the development of OAD. Tacrolimus 0-10 CD4 antigen Mus musculus 72-75 18562569-7 2008 Acid-base status, 17beta estradiol, and the immunosuppressive agents FK506 and cyclosporine affect plasma Mg(2+) levels by altering TRPM6 expression. Tacrolimus 69-74 transient receptor potential cation channel subfamily M member 6 Homo sapiens 132-137 18641302-5 2008 The ionomycin-mediated increase in HIF-1alpha protein levels was sensitive to the transcription inhibitor actinomycin D and to inhibitors of calcineurin, cyclosporin A (CsA), and FK506. Tacrolimus 179-184 hypoxia inducible factor 1 subunit alpha Homo sapiens 35-45 18641302-7 2008 The HIF1A promoter activity was significantly increased in ionomycin-activated mast cells, and the promoter activity could be inhibited by CsA and FK506. Tacrolimus 147-152 hypoxia inducible factor 1 subunit alpha Homo sapiens 4-9 18656798-9 2008 Tacrolimus prophylaxis was associated with inhibition of recruitment of CD4+, CD8+ and interleukin-2R+ inflammatory cells into the allografts, suggesting a central role for interleukin-2 in the development of OAD. Tacrolimus 0-10 interleukin 2 Mus musculus 87-100 18594053-5 2008 DATA SYNTHESIS: Corticosteroids share common metabolic and transporter pathways, the cytochrome P450 and P-glycoprotein (P-gp/ABCB1) systems, respectively, with cyclosporine, tacrolimus, and sirolimus. Tacrolimus 175-185 ATP binding cassette subfamily B member 1 Homo sapiens 105-119 18641556-7 2008 Coagulation factor V and whole/split liver graft were identified as covariates influencing tacrolimus clearance. Tacrolimus 91-101 coagulation factor V Homo sapiens 0-20 18577426-0 2008 Analysis of FK506-mediated protection in an organotypic model of spinal cord damage: heat shock protein 70 levels are modulated in microglial cells. Tacrolimus 12-17 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 85-106 18577426-4 2008 FK506 may exert neuroprotection via inhibition of calcineurin by binding the FKBP12, or by binding other immunophilins such as FKBP52, leading to modulation of heat shock proteins (Hsp) 90 and 70. Tacrolimus 0-5 FKBP prolyl isomerase 1A Rattus norvegicus 77-83 18577426-4 2008 FK506 may exert neuroprotection via inhibition of calcineurin by binding the FKBP12, or by binding other immunophilins such as FKBP52, leading to modulation of heat shock proteins (Hsp) 90 and 70. Tacrolimus 0-5 FKBP prolyl isomerase 4 Rattus norvegicus 127-133 18577426-7 2008 Hsp70 induction was restricted to microglial cells in spinal cord slices treated with either glutamate or FK506. Tacrolimus 106-111 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 0-5 18577426-10 2008 These observations suggest that FK506 might protect spinal cord tissue by targeting on microglial cells and that transient downregulation of Hsp70 on these cells after excitotoxicity is a relevant mechanism of action of FK506. Tacrolimus 220-225 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 141-146 18457657-2 2008 This study showed that stimulation with anti-CD3 mAb, PMA plus ionomycin, or an antigen increased the levels of GRP78 mRNA in primary T cells, which was inhibited by Ca(2+) chelators EGTA and BAPTA-AM and by an inhibitor of calcineurin FK506. Tacrolimus 236-241 heat shock protein 5 Mus musculus 112-117 18534252-7 2008 Injection of 12.5 nmol FK506 also significantly enhanced the phosphorylation of tau at Ser-262 (12E8 site), Ser-198, Ser-199, and/or Ser-202 (Tau-1 site) and Ser-396 and/or Ser-404 (PHF-1 site), without affecting total tau. Tacrolimus 23-28 PHD finger protein 1 Mus musculus 182-187 18622297-2 2008 We describe a case of a 47-year-old African American man with end-stage renal disease secondary to HIV-induced nephropathy who underwent a live unrelated (spouse) donor ABO blood group incompatible transplant using an intravenous immunoglobulin/plasmapheresis preconditioning regimen with interleukin-2 receptor antagonist induction along with tacrolimus and mycophenolate mofetil maintenance. Tacrolimus 344-354 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 169-172 18594053-5 2008 DATA SYNTHESIS: Corticosteroids share common metabolic and transporter pathways, the cytochrome P450 and P-glycoprotein (P-gp/ABCB1) systems, respectively, with cyclosporine, tacrolimus, and sirolimus. Tacrolimus 175-185 phosphoglycolate phosphatase Homo sapiens 121-125 18594053-5 2008 DATA SYNTHESIS: Corticosteroids share common metabolic and transporter pathways, the cytochrome P450 and P-glycoprotein (P-gp/ABCB1) systems, respectively, with cyclosporine, tacrolimus, and sirolimus. Tacrolimus 175-185 ATP binding cassette subfamily B member 1 Homo sapiens 126-131 18498403-0 2008 Lip-enhancing tattoo reaction resolving with topical tacrolimus. Tacrolimus 53-63 SMG1 nonsense mediated mRNA decay associated PI3K related kinase Homo sapiens 0-3 18341670-0 2008 Impact of CYP3A5 genetic polymorphism on pharmacokinetics of tacrolimus in healthy Japanese subjects. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 18465874-7 2008 Compared to the human FKBP12-FK506 complex reported earlier, the structure reveals structural differences in the beta5-beta6 segment that lines the FK506 binding site. Tacrolimus 29-34 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 22-28 18465874-8 2008 The presence in PfFKBD of Cys-106 and Ser-109 (substituting for His-87 and Ile-90, respectively, in human FKBP12), which are 4-5 A from the nearest atom of the FK506 compound, suggests possible routes for the rational design of analogues of FK506 with specific antiparasitic activity. Tacrolimus 160-165 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 106-112 18492692-2 2008 Recent findings of over-activation of the TGF-beta signal in FKBP12-knockout mouse prompted us to investigate whether FK506, the canonical ligand of FKBP, can activate the TGF-beta signal in chronic lymphocytic leukemia. Tacrolimus 118-123 transforming growth factor, beta 1 Mus musculus 172-180 18492692-11 2008 FK506 induced Smad2 phosphorylation and nuclear translocation. Tacrolimus 0-5 SMAD family member 2 Homo sapiens 14-19 18492692-15 2008 FK506 removed FKBP12 from its binding to the TGF-beta-receptor. Tacrolimus 0-5 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 14-20 18492692-15 2008 FK506 removed FKBP12 from its binding to the TGF-beta-receptor. Tacrolimus 0-5 transforming growth factor beta 1 Homo sapiens 45-53 18492692-17 2008 CONCLUSIONS: Our study shows that most chronic lymphocytic leukemia cells escape the homeostatic control of TGF-beta and that FK506 restores the TGF-beta signal in a proportion of non-responsive samples. Tacrolimus 126-131 transforming growth factor beta 1 Homo sapiens 145-153 18492692-18 2008 We demonstrated that FK506 activates TGF-beta receptor I kinase activity in chronic lymphocytic leukemia, which transduces apoptosis by a mitochondrial-dependent pathway. Tacrolimus 21-26 transforming growth factor beta 1 Homo sapiens 37-45 18547634-6 2008 RESULTS: Tacrolimus and TGF-beta1 act synergistically on the generation of LCs and the expression of CD40, CD80, CD86, CD83, and MHC II; stabilize TGF-beta receptor II expression; and decrease the stimulatory capacity of LCs toward T cells. Tacrolimus 9-19 CD40 molecule Homo sapiens 101-105 18547634-6 2008 RESULTS: Tacrolimus and TGF-beta1 act synergistically on the generation of LCs and the expression of CD40, CD80, CD86, CD83, and MHC II; stabilize TGF-beta receptor II expression; and decrease the stimulatory capacity of LCs toward T cells. Tacrolimus 9-19 CD80 molecule Homo sapiens 107-111 18547634-6 2008 RESULTS: Tacrolimus and TGF-beta1 act synergistically on the generation of LCs and the expression of CD40, CD80, CD86, CD83, and MHC II; stabilize TGF-beta receptor II expression; and decrease the stimulatory capacity of LCs toward T cells. Tacrolimus 9-19 CD86 molecule Homo sapiens 113-117 18547634-6 2008 RESULTS: Tacrolimus and TGF-beta1 act synergistically on the generation of LCs and the expression of CD40, CD80, CD86, CD83, and MHC II; stabilize TGF-beta receptor II expression; and decrease the stimulatory capacity of LCs toward T cells. Tacrolimus 9-19 CD83 molecule Homo sapiens 119-123 18346205-0 2008 FK506 binding protein 12 differentially accelerates fibril formation of wild type alpha-synuclein and its clinical mutants A30P or A53T. Tacrolimus 0-5 synuclein alpha Homo sapiens 82-97 18346205-2 2008 We have previously shown that aggregation of alpha-SYN in vitro is accelerated by addition of FK506 binding proteins (FKBP) and that this effect can be counteracted by FK506, a specific inhibitor of these enzymes. Tacrolimus 94-99 synuclein alpha Homo sapiens 45-54 18346205-2 2008 We have previously shown that aggregation of alpha-SYN in vitro is accelerated by addition of FK506 binding proteins (FKBP) and that this effect can be counteracted by FK506, a specific inhibitor of these enzymes. Tacrolimus 168-173 synuclein alpha Homo sapiens 45-54 18538359-2 2008 Chronic (>or=3h) treatment of cultured bovine adrenal chromaffin cells with cyclosporin A or FK506 decreased IRS-2 protein level by approximately 50% (IC(50)=200 or 10nM), without changing IRS-2 mRNA level, and insulin receptor, insulin-like growth factor-I (IGF-I) receptor, IRS-1, PI3K/PDK-1/Akt/GSK-3beta and ERK1/ERK2 protein levels. Tacrolimus 96-101 insulin receptor substrate 2 Bos taurus 112-117 18538359-2 2008 Chronic (>or=3h) treatment of cultured bovine adrenal chromaffin cells with cyclosporin A or FK506 decreased IRS-2 protein level by approximately 50% (IC(50)=200 or 10nM), without changing IRS-2 mRNA level, and insulin receptor, insulin-like growth factor-I (IGF-I) receptor, IRS-1, PI3K/PDK-1/Akt/GSK-3beta and ERK1/ERK2 protein levels. Tacrolimus 96-101 insulin receptor substrate 2 Bos taurus 192-197 18538359-2 2008 Chronic (>or=3h) treatment of cultured bovine adrenal chromaffin cells with cyclosporin A or FK506 decreased IRS-2 protein level by approximately 50% (IC(50)=200 or 10nM), without changing IRS-2 mRNA level, and insulin receptor, insulin-like growth factor-I (IGF-I) receptor, IRS-1, PI3K/PDK-1/Akt/GSK-3beta and ERK1/ERK2 protein levels. Tacrolimus 96-101 insulin like growth factor 1 Bos taurus 232-260 18538359-2 2008 Chronic (>or=3h) treatment of cultured bovine adrenal chromaffin cells with cyclosporin A or FK506 decreased IRS-2 protein level by approximately 50% (IC(50)=200 or 10nM), without changing IRS-2 mRNA level, and insulin receptor, insulin-like growth factor-I (IGF-I) receptor, IRS-1, PI3K/PDK-1/Akt/GSK-3beta and ERK1/ERK2 protein levels. Tacrolimus 96-101 insulin like growth factor 1 receptor Bos taurus 262-277 18538359-2 2008 Chronic (>or=3h) treatment of cultured bovine adrenal chromaffin cells with cyclosporin A or FK506 decreased IRS-2 protein level by approximately 50% (IC(50)=200 or 10nM), without changing IRS-2 mRNA level, and insulin receptor, insulin-like growth factor-I (IGF-I) receptor, IRS-1, PI3K/PDK-1/Akt/GSK-3beta and ERK1/ERK2 protein levels. Tacrolimus 96-101 insulin receptor substrate 1 Bos taurus 279-284 18538359-2 2008 Chronic (>or=3h) treatment of cultured bovine adrenal chromaffin cells with cyclosporin A or FK506 decreased IRS-2 protein level by approximately 50% (IC(50)=200 or 10nM), without changing IRS-2 mRNA level, and insulin receptor, insulin-like growth factor-I (IGF-I) receptor, IRS-1, PI3K/PDK-1/Akt/GSK-3beta and ERK1/ERK2 protein levels. Tacrolimus 96-101 pyruvate dehydrogenase kinase 1 Bos taurus 291-296 18538359-2 2008 Chronic (>or=3h) treatment of cultured bovine adrenal chromaffin cells with cyclosporin A or FK506 decreased IRS-2 protein level by approximately 50% (IC(50)=200 or 10nM), without changing IRS-2 mRNA level, and insulin receptor, insulin-like growth factor-I (IGF-I) receptor, IRS-1, PI3K/PDK-1/Akt/GSK-3beta and ERK1/ERK2 protein levels. Tacrolimus 96-101 AKT serine/threonine kinase 1 Bos taurus 297-300 18538359-2 2008 Chronic (>or=3h) treatment of cultured bovine adrenal chromaffin cells with cyclosporin A or FK506 decreased IRS-2 protein level by approximately 50% (IC(50)=200 or 10nM), without changing IRS-2 mRNA level, and insulin receptor, insulin-like growth factor-I (IGF-I) receptor, IRS-1, PI3K/PDK-1/Akt/GSK-3beta and ERK1/ERK2 protein levels. Tacrolimus 96-101 glycogen synthase kinase 3 beta Bos taurus 301-310 18538359-2 2008 Chronic (>or=3h) treatment of cultured bovine adrenal chromaffin cells with cyclosporin A or FK506 decreased IRS-2 protein level by approximately 50% (IC(50)=200 or 10nM), without changing IRS-2 mRNA level, and insulin receptor, insulin-like growth factor-I (IGF-I) receptor, IRS-1, PI3K/PDK-1/Akt/GSK-3beta and ERK1/ERK2 protein levels. Tacrolimus 96-101 mitogen-activated protein kinase 3 Bos taurus 315-319 18538359-2 2008 Chronic (>or=3h) treatment of cultured bovine adrenal chromaffin cells with cyclosporin A or FK506 decreased IRS-2 protein level by approximately 50% (IC(50)=200 or 10nM), without changing IRS-2 mRNA level, and insulin receptor, insulin-like growth factor-I (IGF-I) receptor, IRS-1, PI3K/PDK-1/Akt/GSK-3beta and ERK1/ERK2 protein levels. Tacrolimus 96-101 mitogen-activated protein kinase 1 Bos taurus 320-324 18538359-6 2008 Pulse-label followed by polyacrylamide gel electrophoresis revealed that cyclosporin A or FK506 accelerated IRS-2 degradation rate (t(1/2)) from >24 to approximately 4.2h, without altering IRS-2 synthesis. Tacrolimus 90-95 insulin receptor substrate 2 Bos taurus 108-113 18538359-6 2008 Pulse-label followed by polyacrylamide gel electrophoresis revealed that cyclosporin A or FK506 accelerated IRS-2 degradation rate (t(1/2)) from >24 to approximately 4.2h, without altering IRS-2 synthesis. Tacrolimus 90-95 insulin receptor substrate 2 Bos taurus 192-197 18538359-7 2008 IRS-2 reduction by cyclosporin A or FK506 was prevented by lactacystin (proteasome inhibitor), but not by calpeptin (calpain inhibitor) or leupeptin (lysosome inhibitor). Tacrolimus 36-41 insulin receptor substrate 2 Bos taurus 0-5 18538359-8 2008 Cyclosporin A or FK506 increased serine-phosphorylation and ubiquitination of IRS-2. Tacrolimus 17-22 insulin receptor substrate 2 Cricetulus griseus 78-83 18448283-9 2008 LPL activity in post-heparin normal human plasma was suppressed following the co-incubation with CsA, RAPA, FK-506 or MMF whereas HL activity remained unaffected. Tacrolimus 108-114 lipoprotein lipase Homo sapiens 0-3 18465874-8 2008 The presence in PfFKBD of Cys-106 and Ser-109 (substituting for His-87 and Ile-90, respectively, in human FKBP12), which are 4-5 A from the nearest atom of the FK506 compound, suggests possible routes for the rational design of analogues of FK506 with specific antiparasitic activity. Tacrolimus 241-246 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 106-112 18440706-0 2008 FK506-protective effects against trimethyltin neurotoxicity in rats: hippocampal expression analyses reveal the involvement of periarterial osteopontin. Tacrolimus 0-5 secreted phosphoprotein 1 Rattus norvegicus 140-151 18440706-3 2008 Histologically, TMT-induced neuronal damage was partially prevented by FK506 in the hippocampal CA1 region, but not in CA3. Tacrolimus 71-76 carbonic anhydrase 1 Rattus norvegicus 96-99 18440706-4 2008 FK506 treatment significantly reduced the number of apoptotic cells in CA1, but not in CA3, and also prevented induction of cognitive deficits by TMT. Tacrolimus 0-5 carbonic anhydrase 1 Rattus norvegicus 71-74 18440706-8 2008 Immunohistochemistry revealed that FK506 reduced osteopontin (OPN) induction by TMT in the periarterial area at 5 days postgavage. Tacrolimus 35-40 secreted phosphoprotein 1 Rattus norvegicus 49-60 18440706-8 2008 Immunohistochemistry revealed that FK506 reduced osteopontin (OPN) induction by TMT in the periarterial area at 5 days postgavage. Tacrolimus 35-40 secreted phosphoprotein 1 Rattus norvegicus 62-65 18465874-7 2008 Compared to the human FKBP12-FK506 complex reported earlier, the structure reveals structural differences in the beta5-beta6 segment that lines the FK506 binding site. Tacrolimus 148-153 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 22-28 17950843-7 2008 Under these conditions FK506, which displaces FKBP12 (to inhibit calcineurin) and rapamycin, which displaces FKBP12 (to inhibit mTOR), each increased the [Ca(2+)](c) rise evoked by caffeine. Tacrolimus 23-28 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 46-52 17950843-10 2008 Nor did inhibition of calcineurin by cypermethrin, okadaic acid or calcineurin inhibitory peptide block the FK506-induced increase in RyR-mediated Ca(2+) release. Tacrolimus 108-113 ryanodine receptor 2 Homo sapiens 134-137 18434567-9 2008 Preliminary experiments reveal that the commonly used immunosuppressive agent FK-506 can inhibit TRPC6 activity in vivo. Tacrolimus 78-84 transient receptor potential cation channel subfamily C member 6 Homo sapiens 97-102 18648179-12 2008 We changed the immunosuppressive agent to tacrolimus, which resulted in improvements in the ALT and AST levels; however, the total bilirubin level increased. Tacrolimus 42-52 solute carrier family 17 member 5 Homo sapiens 100-103 18221365-8 2008 Notably, addition of the calcineurin inhibitor FK-506 produced a more robust reduction in cell death in mutant huntingtin knock-in cells than it did in wild-type cells. Tacrolimus 47-53 huntingtin Mus musculus 111-121 18431361-3 2008 Lentiviral vectors have been engineered to produce a fusion protein between the furin-cleavable proinsulin and the self-dimerization mutant of FK506-binding protein to yield bioactive insulin in keratinocytes; this insulin is released as a response to exogenous administration of a small organic molecule, rapamycin. Tacrolimus 143-148 insulin Homo sapiens 184-191 18465778-4 2008 Both bisindolylmaleimide-I (BIM-I), a specific protein kinase C (PKC) inhibitor, and the immunosuppressant drug known as Tacrolimus (FK506), an IL-2 inhibitor, prevented mitogen-stimulated IL-2 production in Jurkat cells. Tacrolimus 121-131 interleukin 2 Homo sapiens 189-193 18466104-6 2008 To date, use of the CYP3A5 genotype to predict the appropriate initial dose of tacrolimus is the most promising option for individualization of drug therapy in organ transplantation. Tacrolimus 79-89 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 18465778-4 2008 Both bisindolylmaleimide-I (BIM-I), a specific protein kinase C (PKC) inhibitor, and the immunosuppressant drug known as Tacrolimus (FK506), an IL-2 inhibitor, prevented mitogen-stimulated IL-2 production in Jurkat cells. Tacrolimus 133-138 interleukin 2 Homo sapiens 189-193 18589121-5 2008 Tacrolimus did not affect COL-I, COL-III, or MMP gene expression, while LH2b and TGF-beta1 tended to be down-regulated after 1 mumol/L FK506. Tacrolimus 135-140 transforming growth factor beta 1 Homo sapiens 81-90 18589121-6 2008 Conversely, protein levels of MMP-1 (P = NS) and MMP-2 (P < .05 vs CT, 10 nmol/L, 100 nmol/L) were up-regulated after 1 mumol/L tacrolimus. Tacrolimus 131-141 matrix metallopeptidase 1 Homo sapiens 30-35 18589121-6 2008 Conversely, protein levels of MMP-1 (P = NS) and MMP-2 (P < .05 vs CT, 10 nmol/L, 100 nmol/L) were up-regulated after 1 mumol/L tacrolimus. Tacrolimus 131-141 matrix metallopeptidase 2 Homo sapiens 49-54 18589121-7 2008 Our findings confirmed that a high dose of tacrolimus does not induce interstitial collagen overexpression by gingival fibroblasts and induces up-regulation of MMPs protein levels. Tacrolimus 43-53 matrix metallopeptidase 1 Homo sapiens 160-164 18589137-0 2008 Regulatory effect of FK506 on CD152 and PD-1 in the liver allorecipients. Tacrolimus 21-26 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 30-35 18589137-0 2008 Regulatory effect of FK506 on CD152 and PD-1 in the liver allorecipients. Tacrolimus 21-26 MHC class I antigen 1 Sus scrofa 40-44 18589137-7 2008 CONCLUSION: FK506 up-regulated the expression of CD152 and PD-1 on the T-cell surface inhibiting proliferation and activation of effector T cells, favoring the survival of allorecipients. Tacrolimus 12-17 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 49-54 18589137-7 2008 CONCLUSION: FK506 up-regulated the expression of CD152 and PD-1 on the T-cell surface inhibiting proliferation and activation of effector T cells, favoring the survival of allorecipients. Tacrolimus 12-17 MHC class I antigen 1 Sus scrofa 59-63 18226502-2 2008 OBJECTIVE: We aimed to investigate (1) the UV transmission through tacrolimus ointment and (2) the impact of topical exposure to tacrolimus on the protein expression of thymine dimers (TD) and p53 in human skin. Tacrolimus 129-139 tumor protein p53 Homo sapiens 193-196 18252812-0 2008 Relation between mRNA expression level of multidrug resistance 1/ABCB1 in blood cells and required level of tacrolimus in pediatric living-donor liver transplantation. Tacrolimus 108-118 ATP binding cassette subfamily B member 1 Homo sapiens 42-64 18252812-0 2008 Relation between mRNA expression level of multidrug resistance 1/ABCB1 in blood cells and required level of tacrolimus in pediatric living-donor liver transplantation. Tacrolimus 108-118 ATP binding cassette subfamily B member 1 Homo sapiens 65-70 18252812-2 2008 In this study, we examined the significance of multidrug resistance 1 (MDR1) in the peripheral blood cells by comparing the trough concentration of tacrolimus with the occurrence of acute cellular rejection (ACR) in retrospectively collected pediatric living-donor liver transplant patients, who were enrolled after obtaining written informed consent. Tacrolimus 148-158 ATP binding cassette subfamily B member 1 Homo sapiens 47-69 18252812-2 2008 In this study, we examined the significance of multidrug resistance 1 (MDR1) in the peripheral blood cells by comparing the trough concentration of tacrolimus with the occurrence of acute cellular rejection (ACR) in retrospectively collected pediatric living-donor liver transplant patients, who were enrolled after obtaining written informed consent. Tacrolimus 148-158 ATP binding cassette subfamily B member 1 Homo sapiens 71-75 18252812-4 2008 The average trough concentration of tacrolimus during the 15-day postoperative period was significantly higher in the event-free patients than in those who experienced ACR (21 of 44 cases), and they had higher levels of blood MDR1 mRNA. Tacrolimus 36-46 ATP binding cassette subfamily B member 1 Homo sapiens 226-230 18252812-5 2008 In addition, the average trough concentration of tacrolimus significantly correlated with the logarithmically transformed MDR1 mRNA data from the blood cells in patients of both the event-free (r = 0.5406; P = 0.0077) and ACR (r = 0.4772; P = 0.0284). Tacrolimus 49-59 ATP binding cassette subfamily B member 1 Homo sapiens 122-126 18252812-7 2008 These results suggest that MDR1 in blood cells decreases the leukocytic concentration of tacrolimus, and it could be a useful marker to establish an individualized target concentration of tacrolimus to prevent ACR in pediatric patients after liver transplantation. Tacrolimus 89-99 ATP binding cassette subfamily B member 1 Homo sapiens 27-31 18252812-7 2008 These results suggest that MDR1 in blood cells decreases the leukocytic concentration of tacrolimus, and it could be a useful marker to establish an individualized target concentration of tacrolimus to prevent ACR in pediatric patients after liver transplantation. Tacrolimus 188-198 ATP binding cassette subfamily B member 1 Homo sapiens 27-31 18443548-0 2008 The effect of CYP3A5 polymorphisms on the pharmacokinetics of tacrolimus in adolescent kidney transplant recipients. Tacrolimus 62-72 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 18443548-1 2008 BACKGROUND: CYP3A5 gene polymorphism has been shown to influence tacrolimus (TAC) blood concentration and dose requirement in adult kidney transplant patients. Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 18408564-0 2008 Impact of MDR1 and CYP3A5 on the oral clearance of tacrolimus and tacrolimus-related renal dysfunction in adult living-donor liver transplant patients. Tacrolimus 51-61 ATP binding cassette subfamily B member 1 Homo sapiens 10-14 18408564-0 2008 Impact of MDR1 and CYP3A5 on the oral clearance of tacrolimus and tacrolimus-related renal dysfunction in adult living-donor liver transplant patients. Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 19-25 18408564-0 2008 Impact of MDR1 and CYP3A5 on the oral clearance of tacrolimus and tacrolimus-related renal dysfunction in adult living-donor liver transplant patients. Tacrolimus 66-76 ATP binding cassette subfamily B member 1 Homo sapiens 10-14 18408564-9 2008 CONCLUSION: These findings suggest that the CYP3A5*1 genotype as well as the MDR1 mRNA level in enterocytes contributes to interindividual variation in the CL/F of tacrolimus in adult recipients early after living-donor liver transplantation. Tacrolimus 164-174 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 44-50 18493655-4 2008 In support of this idea, we found that adding FKBP blocks binding of FK506 to the common cytochrome P(450) enzyme CYP3A4 in vitro. Tacrolimus 69-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-106 18493655-4 2008 In support of this idea, we found that adding FKBP blocks binding of FK506 to the common cytochrome P(450) enzyme CYP3A4 in vitro. Tacrolimus 69-74 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 114-120 18589174-3 2008 Frequencies of CYP3A5*1 versus *3 and MDR1-C3435T were correlated with tacrolimus (TAC) and cyclosporine (CSA) concentration-to-dose (C/D) ratios. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 18589174-3 2008 Frequencies of CYP3A5*1 versus *3 and MDR1-C3435T were correlated with tacrolimus (TAC) and cyclosporine (CSA) concentration-to-dose (C/D) ratios. Tacrolimus 71-81 ATP binding cassette subfamily B member 1 Homo sapiens 38-42 18355347-6 2008 RESULTS: The level of TGF-beta1 gene expression was insignificantly lower in the CsA-treated group compared with the tacrolimus group, and significantly lower in the group with GO compared with patients without GO. Tacrolimus 117-127 transforming growth factor beta 1 Homo sapiens 22-31 18355347-7 2008 In tacrolimus- and CsA-treated patients, but not in patients with GO, the level of TGF-beta1 gene expression was associated with functional phenotypes of TGF-beta1. Tacrolimus 3-13 transforming growth factor beta 1 Homo sapiens 83-92 18355347-7 2008 In tacrolimus- and CsA-treated patients, but not in patients with GO, the level of TGF-beta1 gene expression was associated with functional phenotypes of TGF-beta1. Tacrolimus 3-13 transforming growth factor beta 1 Homo sapiens 154-163 18295934-10 2008 This increase in mitochondrial BAD levels was matched by a decrease in cytochrome c. FK506 prevented the alterations of mitochondrial BAD and cytochrome c levels induced by A beta and PrP peptides. Tacrolimus 85-90 amyloid beta precursor protein Rattus norvegicus 173-179 18295934-10 2008 This increase in mitochondrial BAD levels was matched by a decrease in cytochrome c. FK506 prevented the alterations of mitochondrial BAD and cytochrome c levels induced by A beta and PrP peptides. Tacrolimus 85-90 prion protein Rattus norvegicus 184-187 18408564-9 2008 CONCLUSION: These findings suggest that the CYP3A5*1 genotype as well as the MDR1 mRNA level in enterocytes contributes to interindividual variation in the CL/F of tacrolimus in adult recipients early after living-donor liver transplantation. Tacrolimus 164-174 ATP binding cassette subfamily B member 1 Homo sapiens 77-81 18408564-10 2008 Furthermore, CYP3A5 in the kidney may play a protective role in the development of tacrolimus-related nephrotoxicity. Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 18379077-5 2008 These results suggest that topical tacrolimus exerts acute inhibitory effects on allergic and protease-activated receptor-2-mediated itching. Tacrolimus 35-45 coagulation factor II (thrombin) receptor-like 1 Mus musculus 94-123 18379077-6 2008 Though precise mechanisms remain unclear, the action on sensory neurons expressing protease-activated receptor-2 and transient receptor potential vanilloid-1 capsaicin receptor may be involved in the inhibitory effects of tacrolimus. Tacrolimus 222-232 coagulation factor II (thrombin) receptor-like 1 Mus musculus 83-176 17660216-9 2008 Tacrolimus, a P-gp inhibitor, restored IDLs in RA lymphocytes. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 14-18 18334918-1 2008 OBJECTIVE: Patients expressing the tacrolimus-metabolizing enzyme, cytochrome P450 (CYP) 3A5, require more tacrolimus to reach target concentrations. Tacrolimus 35-45 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-92 18334918-1 2008 OBJECTIVE: Patients expressing the tacrolimus-metabolizing enzyme, cytochrome P450 (CYP) 3A5, require more tacrolimus to reach target concentrations. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-92 18334918-7 2008 After day 3, the overall daily tacrolimus dose was 68% higher in CYP3A5 expressers (P<0.001). Tacrolimus 31-41 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 65-71 18334918-9 2008 CONCLUSION: We conclude that patients expressing CYP3A5 need more tacrolimus to reach target concentrations and have a lower tacrolimus exposure shortly after transplantation. Tacrolimus 66-76 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 49-55 18334918-9 2008 CONCLUSION: We conclude that patients expressing CYP3A5 need more tacrolimus to reach target concentrations and have a lower tacrolimus exposure shortly after transplantation. Tacrolimus 125-135 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 49-55 18242596-8 2008 In contrast, the calcineurin inhibitor tacrolimus (10 mg/kg) was effective only when administered during the period of ovalbumin aerosol exposure. Tacrolimus 39-49 serine (or cysteine) peptidase inhibitor, clade B, member 1, pseudogene Mus musculus 119-128 18482098-8 2008 FK506 and ascomycin inhibit signaling pathways in astrocytes and change the pattern of cytokine and neurotrophin gene expression. Tacrolimus 0-5 brain derived neurotrophic factor Homo sapiens 100-112 18480987-0 2008 Effect of FK506 on expression of hepatocyte growth factor in murine spinal cord following peripheral nerve injury. Tacrolimus 10-15 hepatocyte growth factor Mus musculus 33-57 18480987-1 2008 This study is to investigate the effect of FK506 on expression of hepatocyte growth factor (HGF) in rats" spinal cord following peripheral nerve injury and to elucidate the mechanisms for neuroprotective property of FK506. Tacrolimus 43-48 hepatocyte growth factor Rattus norvegicus 66-90 18480987-1 2008 This study is to investigate the effect of FK506 on expression of hepatocyte growth factor (HGF) in rats" spinal cord following peripheral nerve injury and to elucidate the mechanisms for neuroprotective property of FK506. Tacrolimus 43-48 hepatocyte growth factor Rattus norvegicus 92-95 18480987-1 2008 This study is to investigate the effect of FK506 on expression of hepatocyte growth factor (HGF) in rats" spinal cord following peripheral nerve injury and to elucidate the mechanisms for neuroprotective property of FK506. Tacrolimus 216-221 hepatocyte growth factor Rattus norvegicus 66-90 18480987-1 2008 This study is to investigate the effect of FK506 on expression of hepatocyte growth factor (HGF) in rats" spinal cord following peripheral nerve injury and to elucidate the mechanisms for neuroprotective property of FK506. Tacrolimus 216-221 hepatocyte growth factor Rattus norvegicus 92-95 18480987-9 2008 It is suggested that peripheral nerve injury does not result in up-regulation of the expression of HGF in spinal cord, while FK506 may induce high expression of endogenous HGF after injury thereby protecting neurons and promoting axonal outgrowth. Tacrolimus 125-130 hepatocyte growth factor Rattus norvegicus 172-175 18321198-9 2008 The rejected corneas in the FK506 nanosphere group showed significantly fewer CD4, CD8, CD68, CD79, vascular endothelial growth factor, ICAM, and tumor growth factor-beta(1)-positive cells than those in the other groups. Tacrolimus 28-33 Cd4 molecule Rattus norvegicus 78-81 18321198-9 2008 The rejected corneas in the FK506 nanosphere group showed significantly fewer CD4, CD8, CD68, CD79, vascular endothelial growth factor, ICAM, and tumor growth factor-beta(1)-positive cells than those in the other groups. Tacrolimus 28-33 Cd68 molecule Rattus norvegicus 88-92 18321198-9 2008 The rejected corneas in the FK506 nanosphere group showed significantly fewer CD4, CD8, CD68, CD79, vascular endothelial growth factor, ICAM, and tumor growth factor-beta(1)-positive cells than those in the other groups. Tacrolimus 28-33 intercellular adhesion molecule 1 Rattus norvegicus 136-140 18182482-6 2008 Moreover, tacrolimus (FK506), which had no effect on NCX1 protein expression, down-regulated NCX2 and NCX3 surface expression and transport activity without any significant effect on cell protein expression. Tacrolimus 10-20 solute carrier family 8 member A2 Homo sapiens 93-97 18182482-6 2008 Moreover, tacrolimus (FK506), which had no effect on NCX1 protein expression, down-regulated NCX2 and NCX3 surface expression and transport activity without any significant effect on cell protein expression. Tacrolimus 10-20 solute carrier family 8 member A3 Homo sapiens 102-106 18182482-6 2008 Moreover, tacrolimus (FK506), which had no effect on NCX1 protein expression, down-regulated NCX2 and NCX3 surface expression and transport activity without any significant effect on cell protein expression. Tacrolimus 22-27 solute carrier family 8 member A2 Homo sapiens 93-97 18182482-6 2008 Moreover, tacrolimus (FK506), which had no effect on NCX1 protein expression, down-regulated NCX2 and NCX3 surface expression and transport activity without any significant effect on cell protein expression. Tacrolimus 22-27 solute carrier family 8 member A3 Homo sapiens 102-106 18480504-1 2008 Tacrolimus, a calcineurin inhibitor, has become an increasingly valuable tool in the treatment of dermatological disorders during the last few years. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 14-35 18346634-12 2008 On the other hand, tacrolimus and dexamethasone, but not indomethacin, decreased TNF-alpha levels (P<0.01). Tacrolimus 19-29 tumor necrosis factor Mus musculus 81-90 18184875-9 2008 Application of cyclosporin A or FK506, inhibitors of PP2B, significantly decreased ROMK channels, and the effect of PP2B inhibitors was abolished by blocking p38 mitogen-activated protein kinase (MAPK) and ERK. Tacrolimus 32-37 potassium inwardly-rectifying channel, subfamily J, member 1 Mus musculus 83-87 18184875-9 2008 Application of cyclosporin A or FK506, inhibitors of PP2B, significantly decreased ROMK channels, and the effect of PP2B inhibitors was abolished by blocking p38 mitogen-activated protein kinase (MAPK) and ERK. Tacrolimus 32-37 mitogen-activated protein kinase 1 Mus musculus 196-200 18184875-9 2008 Application of cyclosporin A or FK506, inhibitors of PP2B, significantly decreased ROMK channels, and the effect of PP2B inhibitors was abolished by blocking p38 mitogen-activated protein kinase (MAPK) and ERK. Tacrolimus 32-37 mitogen-activated protein kinase 1 Mus musculus 206-209 18570909-7 2008 After binding to a specific immunophillin, cyclosporin and tacrolimus inhibit calcineurine, a serine/threonine phosphatase which plays a major role in cytokines transcription notably IL2 after T-cell activation. Tacrolimus 59-69 interleukin 2 Homo sapiens 183-186 17660216-14 2008 Measurement of P-gp expression on lymphocytes could be a potentially useful marker for assessing drug resistance in RA, and may be suitable for selecting infliximab or DMARDs including tacrolimus for RA treatment. Tacrolimus 185-195 ATP binding cassette subfamily B member 1 Homo sapiens 15-19 18306331-3 2008 Tacrolimus (Tac), but not cyclosporin A (CyA), had an inhibitory effect on IFN-alpha-induced double-stranded ribonucleic acid (RNA)-dependent protein kinase (PKR) in a dose-dependent manner. Tacrolimus 0-10 interferon alpha 1 Homo sapiens 75-84 18269661-7 2008 Tacrolimus patients had lower GCF MMP-8 levels than gingivitis (p<0.005), but levels similar to the healthy group. Tacrolimus 0-10 matrix metallopeptidase 8 Homo sapiens 34-39 18312164-2 2008 Over the last decade tacrolimus has become the calcineurin inhibitor of choice for the prevention of rejection in renal transplantation. Tacrolimus 21-31 calcineurin binding protein 1 Homo sapiens 47-68 18262659-3 2008 The culture study showed reduced IL-12, IL-17, IFN-gamma, GM-CSF, TNF-alpha and MIP-1beta, and elevated IL-10 in the PBMC from patients who received tacrolimus, which suggests inhibition of T cells and macrophages, and enhancement of type 1 regulatory T cells. Tacrolimus 149-159 interleukin 17A Homo sapiens 40-45 18262659-3 2008 The culture study showed reduced IL-12, IL-17, IFN-gamma, GM-CSF, TNF-alpha and MIP-1beta, and elevated IL-10 in the PBMC from patients who received tacrolimus, which suggests inhibition of T cells and macrophages, and enhancement of type 1 regulatory T cells. Tacrolimus 149-159 interferon gamma Homo sapiens 47-56 18262659-3 2008 The culture study showed reduced IL-12, IL-17, IFN-gamma, GM-CSF, TNF-alpha and MIP-1beta, and elevated IL-10 in the PBMC from patients who received tacrolimus, which suggests inhibition of T cells and macrophages, and enhancement of type 1 regulatory T cells. Tacrolimus 149-159 colony stimulating factor 2 Homo sapiens 58-64 18262659-3 2008 The culture study showed reduced IL-12, IL-17, IFN-gamma, GM-CSF, TNF-alpha and MIP-1beta, and elevated IL-10 in the PBMC from patients who received tacrolimus, which suggests inhibition of T cells and macrophages, and enhancement of type 1 regulatory T cells. Tacrolimus 149-159 tumor necrosis factor Homo sapiens 66-75 18262659-3 2008 The culture study showed reduced IL-12, IL-17, IFN-gamma, GM-CSF, TNF-alpha and MIP-1beta, and elevated IL-10 in the PBMC from patients who received tacrolimus, which suggests inhibition of T cells and macrophages, and enhancement of type 1 regulatory T cells. Tacrolimus 149-159 C-C motif chemokine ligand 3 Homo sapiens 80-89 18262659-3 2008 The culture study showed reduced IL-12, IL-17, IFN-gamma, GM-CSF, TNF-alpha and MIP-1beta, and elevated IL-10 in the PBMC from patients who received tacrolimus, which suggests inhibition of T cells and macrophages, and enhancement of type 1 regulatory T cells. Tacrolimus 149-159 interleukin 10 Homo sapiens 104-109 18306331-3 2008 Tacrolimus (Tac), but not cyclosporin A (CyA), had an inhibitory effect on IFN-alpha-induced double-stranded ribonucleic acid (RNA)-dependent protein kinase (PKR) in a dose-dependent manner. Tacrolimus 0-10 eukaryotic translation initiation factor 2 alpha kinase 2 Homo sapiens 158-161 18306331-3 2008 Tacrolimus (Tac), but not cyclosporin A (CyA), had an inhibitory effect on IFN-alpha-induced double-stranded ribonucleic acid (RNA)-dependent protein kinase (PKR) in a dose-dependent manner. Tacrolimus 0-3 interferon alpha 1 Homo sapiens 75-84 18306331-3 2008 Tacrolimus (Tac), but not cyclosporin A (CyA), had an inhibitory effect on IFN-alpha-induced double-stranded ribonucleic acid (RNA)-dependent protein kinase (PKR) in a dose-dependent manner. Tacrolimus 0-3 eukaryotic translation initiation factor 2 alpha kinase 2 Homo sapiens 158-161 18212618-2 2008 The most attractive candidate for a pharmacogenetic strategy is tacrolimus dosing based on the CYP3A5 genotype. Tacrolimus 64-74 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 95-101 18042735-7 2008 Cyclosporin A and tacrolimus, both potent inhibitors of the calcium/calmodulin-dependent phosphatase calcineurin, stimulated the enzymatic activity of cellular DLK in an in vitro kinase assay. Tacrolimus 18-28 mitogen-activated protein kinase kinase kinase 12 Mus musculus 160-163 18320068-0 2008 FK506 binding protein mediates glioma cell growth and sensitivity to rapamycin treatment by regulating NF-kappaB signaling pathway. Tacrolimus 0-5 nuclear factor kappa B subunit 1 Homo sapiens 103-112 17981040-7 2008 The use of tacrolimus resulted in improvement in clinical manifestations together with a reduction in HSC71 Ab titers in the "therapy-resistant group". Tacrolimus 11-21 heat shock protein family A (Hsp70) member 8 Homo sapiens 102-107 18311044-9 2008 Since IP was still active and serum KL-6 remained high, 3 mg/day of tacrolimus was added to control IP further and to reduce the dosage of PSL which was recognized as one of the aggravation factors of pneumomediastinum. Tacrolimus 68-78 mucin 1, cell surface associated Homo sapiens 36-40 18158124-10 2008 Therapy with tacrolimus was significantly associated with the expression of HLA-G and a better graft prognosis. Tacrolimus 13-23 major histocompatibility complex, class I, G Homo sapiens 76-81 18306108-5 2008 Here, we first identified the cDNA and protein structures of miniature pig HIF-1alpha, and next investigated the effects of cyclosporine and FK506 on HIF-1alpha expression in endothelial cells of miniature pig. Tacrolimus 141-146 hypoxia inducible factor 1 subunit alpha Sus scrofa 150-160 18463795-6 2008 Unlike RelB, which is always present in the mitochondrial fraction, NFATx appeared on the mitochondria in cells treated with ionomycin together with an immunosuppressant and inhibitor of calcineurin (FK506). Tacrolimus 200-205 nuclear factor of activated T cells 3 Homo sapiens 68-73 18721002-3 2008 Cinacalcet, as well as some immunosuppressants such as ciclosporin, tacrolimus and sirolimus, is partially metabolized by the cytochrome P450 3A enzymes (CYP3A). Tacrolimus 68-78 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 154-159 18261573-10 2008 Both CsA and FK506 caused mild osteoblastic proliferation and matrix mineralization activity, as reflected by increased osteocalcin and alkaline phosphatase levels in 22.6% and 12.5% of patients, respectively. Tacrolimus 13-18 bone gamma-carboxyglutamate protein Homo sapiens 120-131 17822876-4 2008 The aim of the current study was to examine the expression of heat-shock protein 70 (HSP70) in biopsy specimens from 11 OLP lesions before and after topical treatment with tacrolimus. Tacrolimus 172-182 heat shock protein family A (Hsp70) member 4 Homo sapiens 62-83 17822876-4 2008 The aim of the current study was to examine the expression of heat-shock protein 70 (HSP70) in biopsy specimens from 11 OLP lesions before and after topical treatment with tacrolimus. Tacrolimus 172-182 heat shock protein family A (Hsp70) member 4 Homo sapiens 85-90 17822876-7 2008 The moderate increase in HSP70 expression after treatment with tacrolimus was not significant. Tacrolimus 63-73 heat shock protein family A (Hsp70) member 4 Homo sapiens 25-30 17949992-7 2008 Moreover, depolarization of cultured DRG neurons induced de novo synthesis of CCR2 mRNA, which was blocked by the calcineurin inhibitors cyclosporin A and FK506. Tacrolimus 155-160 C-C motif chemokine receptor 2 Homo sapiens 78-82 17978097-5 2008 The effect of CCK was dependent on calcineurin, as these changes were blocked by immunosuppressants FK506 and CsA and by overexpression of the endogenous protein inhibitor CAIN. Tacrolimus 100-105 cholecystokinin Homo sapiens 14-17 18635947-2 2008 FK506-binding protein (FKBP) is one of two major immunophilins and most of FKBP family members bind FK506 and show peptidylprolyl cis/trans isomerase (PPIase) activity. Tacrolimus 0-5 peptidylprolyl isomerase like 3 Rattus norvegicus 115-149 18635947-2 2008 FK506-binding protein (FKBP) is one of two major immunophilins and most of FKBP family members bind FK506 and show peptidylprolyl cis/trans isomerase (PPIase) activity. Tacrolimus 0-5 peptidylprolyl isomerase like 3 Rattus norvegicus 151-157 18635947-8 2008 FK506 and its derivatives with no immunosuppressive activities bind to the conserved active sites of the canonical FKBP members such as FKBP12, which shows PPIase activity. Tacrolimus 0-5 FKBP prolyl isomerase 1A Rattus norvegicus 136-142 18635947-8 2008 FK506 and its derivatives with no immunosuppressive activities bind to the conserved active sites of the canonical FKBP members such as FKBP12, which shows PPIase activity. Tacrolimus 0-5 peptidylprolyl isomerase like 3 Rattus norvegicus 156-162 18162987-4 2007 One possible explanation for this finding is the prolonged pretransplantation exposure to tacrolimus in the ABO-incompatible group, resulting in tacrolimus-associated renal toxicity, which slows the reduction in plasma creatinine. Tacrolimus 90-100 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 108-111 18038971-6 2007 Compared to FK506, the fragment-based FKBP12 inhibitors developed herein possess significant advantages as drug candidates. Tacrolimus 12-17 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 38-44 18162987-4 2007 One possible explanation for this finding is the prolonged pretransplantation exposure to tacrolimus in the ABO-incompatible group, resulting in tacrolimus-associated renal toxicity, which slows the reduction in plasma creatinine. Tacrolimus 145-155 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 108-111 18162987-5 2007 In fact, the day before, and also immediately after, the transplantation (for the first 3-4 postoperative days), the tacrolimus 12-hr trough levels in the ABO-incompatible group were greater than in the ABO-compatible group. Tacrolimus 117-127 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 155-158 18031601-0 2007 Electrophysiological actions of cyclosporin A and tacrolimus on rat hippocampal CA1 pyramidal neurons. Tacrolimus 50-60 carbonic anhydrase 1 Rattus norvegicus 80-83 17495880-0 2007 CYP3A5 and CYP3A4 but not MDR1 single-nucleotide polymorphisms determine long-term tacrolimus disposition and drug-related nephrotoxicity in renal recipients. Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 17495880-5 2007 The lack of a time-related increase in dose-corrected tacrolimus exposure observed with the CYP3A4*1/CYP3A5*1 and CYP3A4*1B/CYP3A5*1 genotypes is associated with tacrolimus-related nephrotoxicity, possibly as a result of higher concentrations of toxic metabolites. Tacrolimus 54-64 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 92-98 17495880-0 2007 CYP3A5 and CYP3A4 but not MDR1 single-nucleotide polymorphisms determine long-term tacrolimus disposition and drug-related nephrotoxicity in renal recipients. Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 11-17 17495880-5 2007 The lack of a time-related increase in dose-corrected tacrolimus exposure observed with the CYP3A4*1/CYP3A5*1 and CYP3A4*1B/CYP3A5*1 genotypes is associated with tacrolimus-related nephrotoxicity, possibly as a result of higher concentrations of toxic metabolites. Tacrolimus 54-64 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 101-107 17495880-5 2007 The lack of a time-related increase in dose-corrected tacrolimus exposure observed with the CYP3A4*1/CYP3A5*1 and CYP3A4*1B/CYP3A5*1 genotypes is associated with tacrolimus-related nephrotoxicity, possibly as a result of higher concentrations of toxic metabolites. Tacrolimus 54-64 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 114-120 17495880-1 2007 The impact of CYP3A and MDR1 gene single-nucleotide polymorphisms on long-term tacrolimus disposition and drug-related toxicity has not been assessed. Tacrolimus 79-89 ATP binding cassette subfamily B member 1 Homo sapiens 24-28 17495880-5 2007 The lack of a time-related increase in dose-corrected tacrolimus exposure observed with the CYP3A4*1/CYP3A5*1 and CYP3A4*1B/CYP3A5*1 genotypes is associated with tacrolimus-related nephrotoxicity, possibly as a result of higher concentrations of toxic metabolites. Tacrolimus 54-64 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 124-130 17495880-5 2007 The lack of a time-related increase in dose-corrected tacrolimus exposure observed with the CYP3A4*1/CYP3A5*1 and CYP3A4*1B/CYP3A5*1 genotypes is associated with tacrolimus-related nephrotoxicity, possibly as a result of higher concentrations of toxic metabolites. Tacrolimus 162-172 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 114-120 17495880-4 2007 The CYP3A4*1/CYP3A5*1 and CYP3A4*1B/CYP3A5*1 genotypes were significantly more frequently associated with the development of biopsy-proven tacrolimus-related nephrotoxicity than the CYP3A4*1/ CYP3A5*3 genotype (37.5 vs 11.2%; P=0.03 and 42.8 vs 11.2%; P=0.02). Tacrolimus 139-149 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 4-10 17495880-4 2007 The CYP3A4*1/CYP3A5*1 and CYP3A4*1B/CYP3A5*1 genotypes were significantly more frequently associated with the development of biopsy-proven tacrolimus-related nephrotoxicity than the CYP3A4*1/ CYP3A5*3 genotype (37.5 vs 11.2%; P=0.03 and 42.8 vs 11.2%; P=0.02). Tacrolimus 139-149 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 17495880-4 2007 The CYP3A4*1/CYP3A5*1 and CYP3A4*1B/CYP3A5*1 genotypes were significantly more frequently associated with the development of biopsy-proven tacrolimus-related nephrotoxicity than the CYP3A4*1/ CYP3A5*3 genotype (37.5 vs 11.2%; P=0.03 and 42.8 vs 11.2%; P=0.02). Tacrolimus 139-149 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 26-32 17495880-4 2007 The CYP3A4*1/CYP3A5*1 and CYP3A4*1B/CYP3A5*1 genotypes were significantly more frequently associated with the development of biopsy-proven tacrolimus-related nephrotoxicity than the CYP3A4*1/ CYP3A5*3 genotype (37.5 vs 11.2%; P=0.03 and 42.8 vs 11.2%; P=0.02). Tacrolimus 139-149 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 17495880-4 2007 The CYP3A4*1/CYP3A5*1 and CYP3A4*1B/CYP3A5*1 genotypes were significantly more frequently associated with the development of biopsy-proven tacrolimus-related nephrotoxicity than the CYP3A4*1/ CYP3A5*3 genotype (37.5 vs 11.2%; P=0.03 and 42.8 vs 11.2%; P=0.02). Tacrolimus 139-149 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 26-32 17495880-4 2007 The CYP3A4*1/CYP3A5*1 and CYP3A4*1B/CYP3A5*1 genotypes were significantly more frequently associated with the development of biopsy-proven tacrolimus-related nephrotoxicity than the CYP3A4*1/ CYP3A5*3 genotype (37.5 vs 11.2%; P=0.03 and 42.8 vs 11.2%; P=0.02). Tacrolimus 139-149 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 17921453-6 2007 FK506 increased the amplitude and decreased the frequency of SOICR in HEK293 cells expressing RyR2 with or without FKBP12.6, indicating that the action of FK506 on SOICR is independent of FKBP12.6. Tacrolimus 0-5 ryanodine receptor 2 Homo sapiens 94-98 17785681-0 2007 FK506, a calcineurin inhibitor, prevents cadmium-induced testicular toxicity in mice. Tacrolimus 0-5 calcineurin binding protein 1 Mus musculus 9-30 18159131-2 2007 Since tacrolimus and PPIs share the CYP3A4 system for metabolism, pharmacokinetic interactions are anticipated when they are administered simultaneously. Tacrolimus 6-16 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 36-42 17888632-0 2007 FK506 induces interleukin-6 secretion from UVB irradiated cultured human keratinocytes via p38 mitogen-activated protein kinase pathway: implication on mechanisms of tacrolimus-induced skin irritation. Tacrolimus 0-5 interleukin 6 Homo sapiens 14-27 17888632-0 2007 FK506 induces interleukin-6 secretion from UVB irradiated cultured human keratinocytes via p38 mitogen-activated protein kinase pathway: implication on mechanisms of tacrolimus-induced skin irritation. Tacrolimus 0-5 mitogen-activated protein kinase 14 Homo sapiens 91-94 17888632-0 2007 FK506 induces interleukin-6 secretion from UVB irradiated cultured human keratinocytes via p38 mitogen-activated protein kinase pathway: implication on mechanisms of tacrolimus-induced skin irritation. Tacrolimus 166-176 interleukin 6 Homo sapiens 14-27 17921453-3 2007 Single channel recordings in lipid bilayers showed that FK506 treatment of recombinant RyR2 co-expressed with or without FKBP12.6 or native canine RyR2 did not induce long-lived subconductance states. Tacrolimus 56-61 ryanodine receptor 2 Canis lupus familiaris 87-91 17921453-6 2007 FK506 increased the amplitude and decreased the frequency of SOICR in HEK293 cells expressing RyR2 with or without FKBP12.6, indicating that the action of FK506 on SOICR is independent of FKBP12.6. Tacrolimus 0-5 FKBP prolyl isomerase 1B Homo sapiens 115-123 17921453-6 2007 FK506 increased the amplitude and decreased the frequency of SOICR in HEK293 cells expressing RyR2 with or without FKBP12.6, indicating that the action of FK506 on SOICR is independent of FKBP12.6. Tacrolimus 155-160 ryanodine receptor 2 Homo sapiens 94-98 17761160-3 2007 FK506 and cyclosporine A, calcineurin inhibitors, partially inhibited UTP-induced IL-6 mRNA expression and protein production. Tacrolimus 0-5 interleukin 6 Homo sapiens 82-86 17761160-4 2007 In addition, combined application of FK506 and PD98059 synergistically inhibited the UTP-induced IL-6 production. Tacrolimus 37-42 interleukin 6 Homo sapiens 97-101 17854353-1 2007 Tacrolimus ointment is a topical calcineurin inhibitor (TCI) that was developed specifically for the treatment of atopic dermatitis (AD). Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 33-54 17854353-1 2007 Tacrolimus ointment is a topical calcineurin inhibitor (TCI) that was developed specifically for the treatment of atopic dermatitis (AD). Tacrolimus 0-10 latexin Homo sapiens 56-59 17761346-2 2007 The importance of IL-2 down-regulation in preventing acute rejection in organ transplantation and the development of autoimmune diseases has been demonstrated by the therapeutic usefulness of the widely used immunosuppressants cyclosporine A and FK506. Tacrolimus 246-251 interleukin 2 Homo sapiens 18-22 17940132-0 2007 Upregulation of neurotrophic factor-related gene expression in retina with experimental autoimmune uveoretinitis by intravitreal injection of tacrolimus (FK506). Tacrolimus 142-152 neurotrophin 3 Rattus norvegicus 16-35 17940132-0 2007 Upregulation of neurotrophic factor-related gene expression in retina with experimental autoimmune uveoretinitis by intravitreal injection of tacrolimus (FK506). Tacrolimus 154-159 neurotrophin 3 Rattus norvegicus 16-35 17940132-1 2007 AIM: The current study was designed to determine whether intravitreal injection of tacrolimus (FK506) modulates the gene expression of neurotrophic factor-related molecules in the retina from eyes with induced experimental autoimmune uveoretinitis (EAU) in rats. Tacrolimus 83-93 neurotrophin 3 Rattus norvegicus 135-154 17940132-1 2007 AIM: The current study was designed to determine whether intravitreal injection of tacrolimus (FK506) modulates the gene expression of neurotrophic factor-related molecules in the retina from eyes with induced experimental autoimmune uveoretinitis (EAU) in rats. Tacrolimus 95-100 neurotrophin 3 Rattus norvegicus 135-154 17979656-7 2007 The maximal F(G) ratios for the 11 CYP3A4 substrates investigated ranged from 1.06-7.14 for alprazolam and tacrolimus, respectively. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-41 17978565-7 2007 In contrast, rats in the tacrolimus treatment group showed low serum insulin and high serum glucose levels. Tacrolimus 25-35 insulin Mesocricetus auratus 69-76 17880416-3 2007 RESULTS: Upon exposure to ciclosporin A (500 ng/mL) or tacrolimus (25 ng/mL) the number of cytokine expressing T cells was almost completely blocked in neonatal T cells while sirolimus (10 ng/mL) only inhibited intracytoplasmatic tumour necrosis factor alpha (TNF-alpha) expression (mean% positive cells; 4.0 +/- 2.1% vs. 1.09 +/- 0.6%, p = 0.003), but mildly stimulated the intracellular expression of interleukin (IL)-2 (24.4 +/- 6.5% vs. 28.1 +/- 7.1%, p = 0.041). Tacrolimus 55-65 tumor necrosis factor Homo sapiens 260-269 17880416-3 2007 RESULTS: Upon exposure to ciclosporin A (500 ng/mL) or tacrolimus (25 ng/mL) the number of cytokine expressing T cells was almost completely blocked in neonatal T cells while sirolimus (10 ng/mL) only inhibited intracytoplasmatic tumour necrosis factor alpha (TNF-alpha) expression (mean% positive cells; 4.0 +/- 2.1% vs. 1.09 +/- 0.6%, p = 0.003), but mildly stimulated the intracellular expression of interleukin (IL)-2 (24.4 +/- 6.5% vs. 28.1 +/- 7.1%, p = 0.041). Tacrolimus 55-65 interleukin 2 Homo sapiens 403-421 17880416-4 2007 In cord blood lymphocytes, the inhibitory effect of ciclosporin A and tacrolimus was dose-dependent (e.g. IL-2: control, 12.3 +/- 5.33%, ciclosporin A 5 ng/mL, 10.1 +/- 5.5%; 50 ng/mL, 7.1 +/- 4.7%; 500 ng/mL, 1.2 +/- 0.3%; tacrolimus 0.25 ng/mL, 9.3 +/- 4.9%; 2.5 ng/mL, 6.1 +/- 3.3%; 25 ng/mL, 1.0 +/- 0.6%), while the function of adult lymphocytes was only impaired at high doses of both compounds. Tacrolimus 70-80 interleukin 2 Homo sapiens 106-110 17880416-5 2007 In contrast, the number of cytokine expressing monocytes was not influenced by ciclosporin A and tacrolimus except for a minor decrease of TNF-alpha producing neonatal monocytes after addition of tacrolimus (17.9% vs. 13.9%, p = 0.031). Tacrolimus 196-206 tumor necrosis factor Homo sapiens 139-148 17652368-7 2007 Intrarenal injection of the RyR activator FK506 decreases RBF by 22% (P > 0.03). Tacrolimus 42-47 ryanodine receptor 2 Rattus norvegicus 28-31 17701362-4 2007 However, cytochrome c release and apoptosis can be restored by treatment with FK506. Tacrolimus 78-83 cytochrome c, somatic Homo sapiens 9-21 17675223-15 2007 We conclude that the present study did not confirm our earlier behavioral data, and suggest that FK506 is not effective in treating the behavioral outcomes of TGCI, despite its efficacy in reducing CA1, hippocampal damage. Tacrolimus 97-102 carbonic anhydrase 1 Rattus norvegicus 198-201 17522069-7 2007 This conclusion was based on the findings that inhibition of NFAT3 activation by either FK506 or NFAT3 siRNA dramatically down-regulated the TNF induction upon B[a]PDE exposure, and that knock-down of TNF by its specific siRNA also led to abrogation of B[a]PDE-induced cell transformation in Cl41 cells and their tumorigenicity in nude mice. Tacrolimus 88-93 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 4 Mus musculus 61-66 17522069-7 2007 This conclusion was based on the findings that inhibition of NFAT3 activation by either FK506 or NFAT3 siRNA dramatically down-regulated the TNF induction upon B[a]PDE exposure, and that knock-down of TNF by its specific siRNA also led to abrogation of B[a]PDE-induced cell transformation in Cl41 cells and their tumorigenicity in nude mice. Tacrolimus 88-93 tumor necrosis factor Mus musculus 141-144 17615153-0 2007 The immunophilin ligands cyclosporin A and FK506 suppress prostate cancer cell growth by androgen receptor-dependent and -independent mechanisms. Tacrolimus 43-48 androgen receptor Homo sapiens 89-106 17615153-4 2007 Therefore, we tested whether the immunophilin ligand FK506 affected AR activity in prostate cancer cell lines. Tacrolimus 53-58 androgen receptor Homo sapiens 68-70 17615153-8 2007 Interestingly, FK506 only inhibited LNCaP cells, suggesting a dependence on the AR for this effect. Tacrolimus 15-20 androgen receptor Homo sapiens 80-82 17615153-11 2007 Further studies in LNCaP cells revealed that CsA and FK506 were able to block or attenuate several stages of AR signaling, including hormone binding, nuclear translocation, and activity at several AR-responsive reporter and endogenous genes. Tacrolimus 53-58 androgen receptor Homo sapiens 109-111 17965516-7 2007 Recent reports on clinical pharmacokinetics indicate that dose levels of tacrolimus need to be adjusted in transplant patients with CYP3A5 polymorphism. Tacrolimus 73-83 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 132-138 17877381-1 2007 FKBP-12, a 12 kDa FK506-binding protein (neuroimmunophilin), acts as a receptor for the immunosuppressant drug FK506. Tacrolimus 18-23 FKBP prolyl isomerase 1A Rattus norvegicus 0-7 17878357-4 2007 We further found that IkappaB-alpha degradation, MAPK activation, and TNF-alpha production by FK506 were reduced in macrophages from mice deficient in MyD88, Toll/IL-1R domain-containing adaptor-inducing IFN-beta (TRIF), TLR2, or TLR4, whereas macrophages from TLR3-deficient or TLR9 mutant mice showed the same responses to FK506 as those of wild-type cells. Tacrolimus 94-99 tumor necrosis factor Mus musculus 70-79 17878357-4 2007 We further found that IkappaB-alpha degradation, MAPK activation, and TNF-alpha production by FK506 were reduced in macrophages from mice deficient in MyD88, Toll/IL-1R domain-containing adaptor-inducing IFN-beta (TRIF), TLR2, or TLR4, whereas macrophages from TLR3-deficient or TLR9 mutant mice showed the same responses to FK506 as those of wild-type cells. Tacrolimus 94-99 myeloid differentiation primary response gene 88 Mus musculus 151-156 17878357-4 2007 We further found that IkappaB-alpha degradation, MAPK activation, and TNF-alpha production by FK506 were reduced in macrophages from mice deficient in MyD88, Toll/IL-1R domain-containing adaptor-inducing IFN-beta (TRIF), TLR2, or TLR4, whereas macrophages from TLR3-deficient or TLR9 mutant mice showed the same responses to FK506 as those of wild-type cells. Tacrolimus 325-330 nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha Mus musculus 22-35 17878357-4 2007 We further found that IkappaB-alpha degradation, MAPK activation, and TNF-alpha production by FK506 were reduced in macrophages from mice deficient in MyD88, Toll/IL-1R domain-containing adaptor-inducing IFN-beta (TRIF), TLR2, or TLR4, whereas macrophages from TLR3-deficient or TLR9 mutant mice showed the same responses to FK506 as those of wild-type cells. Tacrolimus 325-330 myeloid differentiation primary response gene 88 Mus musculus 151-156 17666045-4 2007 Here we demonstrate that in mouse cortical neurons, inhibition of the Ca(2+)-dependent protein phosphatase calcineurin by FK506 or cyclosporine A blocks CREB-dependent gene expression induced by depolarization without inhibiting depolarization-induced Ca(2+) influx or CREB Ser-133 phosphorylation. Tacrolimus 122-127 cAMP responsive element binding protein 1 Mus musculus 153-157 17666045-4 2007 Here we demonstrate that in mouse cortical neurons, inhibition of the Ca(2+)-dependent protein phosphatase calcineurin by FK506 or cyclosporine A blocks CREB-dependent gene expression induced by depolarization without inhibiting depolarization-induced Ca(2+) influx or CREB Ser-133 phosphorylation. Tacrolimus 122-127 cAMP responsive element binding protein 1 Mus musculus 269-273 17875118-10 2007 CONCLUSION: The TBIL in patients and CYP3A5*3 genetic polymorphism in both donors and recipients contribute to the inter-individual variability of oral tacrolimus apparent clearance in Chinese adult liver transplant patients. Tacrolimus 152-162 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 17666045-6 2007 Stimulation of a CRE-luciferase reporter gene by depolarization was sensitive to FK506 throughout the entire time course of the transcriptional response, revealing that calcineurin activity is required to maintain CREB-dependent transcription. Tacrolimus 81-86 cAMP responsive element binding protein 1 Mus musculus 214-218 17367747-0 2007 Protective effects of Tacrolimus, a calcineurin inhibitor, in experimental periodontitis in rats. Tacrolimus 22-32 calcineurin binding protein 1 Rattus norvegicus 36-57 17885626-0 2007 1199G>A and 2677G>T/A polymorphisms of ABCB1 independently affect tacrolimus concentration in hepatic tissue after liver transplantation. Tacrolimus 72-82 ATP binding cassette subfamily B member 1 Homo sapiens 45-50 17367747-6 2007 Tacrolimus treatment in periodontitis-induced rats conferred protection against the inflammation-induced tissue and bone loss associated with periodontitis, through a mechanism involving IL-1beta, TNF-alpha and IL-6. Tacrolimus 0-10 interleukin 1 beta Rattus norvegicus 187-195 17367747-4 2007 RESULTS: Radiographic, enzymatic (myeloperoxidase) and histological analysis revealed that Tacrolimus reduced the severity of periodontitis. Tacrolimus 91-101 myeloperoxidase Rattus norvegicus 34-49 17367747-6 2007 Tacrolimus treatment in periodontitis-induced rats conferred protection against the inflammation-induced tissue and bone loss associated with periodontitis, through a mechanism involving IL-1beta, TNF-alpha and IL-6. Tacrolimus 0-10 tumor necrosis factor Rattus norvegicus 197-206 17367747-5 2007 More specifically, Tacrolimus suppressed the expression of serum interleukin (IL-1beta), tumour necrosis factor (TNF-alpha), IL-6, airpouch exudate PGE(2) and leukocytosis usually observed after the induction of periodontitis. Tacrolimus 19-29 interleukin 1 beta Rattus norvegicus 78-86 17367747-6 2007 Tacrolimus treatment in periodontitis-induced rats conferred protection against the inflammation-induced tissue and bone loss associated with periodontitis, through a mechanism involving IL-1beta, TNF-alpha and IL-6. Tacrolimus 0-10 interleukin 6 Rattus norvegicus 211-215 17367747-5 2007 More specifically, Tacrolimus suppressed the expression of serum interleukin (IL-1beta), tumour necrosis factor (TNF-alpha), IL-6, airpouch exudate PGE(2) and leukocytosis usually observed after the induction of periodontitis. Tacrolimus 19-29 tumor necrosis factor Rattus norvegicus 113-122 17367747-5 2007 More specifically, Tacrolimus suppressed the expression of serum interleukin (IL-1beta), tumour necrosis factor (TNF-alpha), IL-6, airpouch exudate PGE(2) and leukocytosis usually observed after the induction of periodontitis. Tacrolimus 19-29 interleukin 6 Rattus norvegicus 125-129 17385075-4 2007 Caspase-3 activation was accompanied by CsA- and FK506-induced cell death and inhibited by NGF. Tacrolimus 49-54 caspase 3 Rattus norvegicus 0-9 17588685-4 2007 Immunosuppressant FK506 inhibits Nur77 NBRE and NurRE binding activity but has no effect on thymocytes apoptosis, the subcellular localization of Nur77, or cytochrome c release. Tacrolimus 18-23 nuclear receptor subfamily 4 group A member 1 Homo sapiens 33-38 17662023-7 2007 Consistent with activation of the Fas-ligand promoter by FKHR dephosphorylation, Fas-ligand expression increased 2 days after ischemia/reperfusion, and treatment with the CaN inhibitor FK506 inhibited that expression. Tacrolimus 185-190 forkhead box O1 Mus musculus 57-61 17684059-7 2007 Pretreatment of PC12 cells with inhibitors of calcineurin (protein phosphatase 2B), cyclosporin A and FK506, prevented depolarization-induced nuclear translocation and tyrosine phosphorylation of PYK2. Tacrolimus 102-107 protein tyrosine kinase 2 beta Rattus norvegicus 196-200 17889118-3 2007 The purpose of this study was to provide a short overview of recent results obtained in the field of pharmacogenetics of tacrolimus and sirolimus, both substrates of the cytochrome P450 3A (CYP3A) enzymes and of the efflux pump P-glycoprotein, the product of the Multidrug Resistance-1 (MDR1) genes. Tacrolimus 121-131 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 170-188 17889118-3 2007 The purpose of this study was to provide a short overview of recent results obtained in the field of pharmacogenetics of tacrolimus and sirolimus, both substrates of the cytochrome P450 3A (CYP3A) enzymes and of the efflux pump P-glycoprotein, the product of the Multidrug Resistance-1 (MDR1) genes. Tacrolimus 121-131 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 190-195 17889118-3 2007 The purpose of this study was to provide a short overview of recent results obtained in the field of pharmacogenetics of tacrolimus and sirolimus, both substrates of the cytochrome P450 3A (CYP3A) enzymes and of the efflux pump P-glycoprotein, the product of the Multidrug Resistance-1 (MDR1) genes. Tacrolimus 121-131 ATP binding cassette subfamily B member 1 Homo sapiens 228-242 17889118-3 2007 The purpose of this study was to provide a short overview of recent results obtained in the field of pharmacogenetics of tacrolimus and sirolimus, both substrates of the cytochrome P450 3A (CYP3A) enzymes and of the efflux pump P-glycoprotein, the product of the Multidrug Resistance-1 (MDR1) genes. Tacrolimus 121-131 ATP binding cassette subfamily B member 1 Homo sapiens 263-285 17889118-3 2007 The purpose of this study was to provide a short overview of recent results obtained in the field of pharmacogenetics of tacrolimus and sirolimus, both substrates of the cytochrome P450 3A (CYP3A) enzymes and of the efflux pump P-glycoprotein, the product of the Multidrug Resistance-1 (MDR1) genes. Tacrolimus 121-131 ATP binding cassette subfamily B member 1 Homo sapiens 287-291 17889118-4 2007 A number of retrospective studies that demonstrated a link between the polymorphisms governing the CYP3A5 protein expression, with more conflicting results with the MDR1 gene polymorphisms, related to the daily dose necessary to achieve adequate blood tacrolimus levels. Tacrolimus 252-262 ATP binding cassette subfamily B member 1 Homo sapiens 165-169 17889153-11 2007 The logistic regression analysis (dependent variable: PTDM; independent variables: age, anti-CD25, tacrolimus vs cyclosporine) showed that treatment with anti-CD25 is an independent risk factor for PTDM (P = .041; OR 3.28; CI 95% 1.04-10.31). Tacrolimus 99-109 interleukin 2 receptor subunit alpha Homo sapiens 159-163 17889206-2 2007 The objective of this study was to prospectively assess the impact of conversion from cyclosporine (CsA) to tacrolimus on lung function in patients who developed BOS while receiving CsA-based immunosuppressive therapy. Tacrolimus 108-118 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 182-185 17854565-9 2007 The levels of CD4(+) T cell subset in the peripheral blood of the tacrolimus-treated and tacrolimus plus TGP-treated groups were (38.71+/-5.15)% and (32.43+/-4.39)% respectively 7 days after transplantation. Tacrolimus 66-76 Cd4 molecule Rattus norvegicus 14-17 17854565-9 2007 The levels of CD4(+) T cell subset in the peripheral blood of the tacrolimus-treated and tacrolimus plus TGP-treated groups were (38.71+/-5.15)% and (32.43+/-4.39)% respectively 7 days after transplantation. Tacrolimus 89-99 Cd4 molecule Rattus norvegicus 14-17 17660033-8 2007 The combination of tacrolimus and rapamycin limited the inhibitory effect of the latter drug on P70(S6K) activation. Tacrolimus 19-29 ribosomal protein S6 kinase B1 Homo sapiens 96-99 17854565-10 2007 The level of CD4(+) T cell subset in the tacrolimus plus TGP-treated group was lower than that in the tacrolimus-treated group (P<0.05). Tacrolimus 41-51 Cd4 molecule Rattus norvegicus 13-16 17854565-10 2007 The level of CD4(+) T cell subset in the tacrolimus plus TGP-treated group was lower than that in the tacrolimus-treated group (P<0.05). Tacrolimus 102-112 Cd4 molecule Rattus norvegicus 13-16 17568575-0 2007 Polymorphisms of tumor necrosis factor-alpha, interleukin-10, cytochrome P450 3A5 and ABCB1 in Chinese liver transplant patients treated with immunosuppressant tacrolimus. Tacrolimus 160-170 tumor necrosis factor Homo sapiens 17-44 17459670-12 2007 Preliminary experiments reveal the commonly used immunosuppressive agent FK-506 can inhibit TRPC6 activity in vivo. Tacrolimus 73-79 transient receptor potential cation channel subfamily C member 6 Homo sapiens 92-97 17391324-0 2007 Influence of the CYP3A5 and MDR1 genetic polymorphisms on the pharmacokinetics of tacrolimus in healthy Korean subjects. Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 17-23 17391324-0 2007 Influence of the CYP3A5 and MDR1 genetic polymorphisms on the pharmacokinetics of tacrolimus in healthy Korean subjects. Tacrolimus 82-92 ATP binding cassette subfamily B member 1 Homo sapiens 28-32 17612778-8 2007 This study shows that conversion from CsA to tacrolimus therapy leads to a reversal of the CsA-induced bone loss, which can probably be mediated by downregulation of IL-1beta, IL-6, and TNF-alpha production. Tacrolimus 45-55 interleukin 1 beta Rattus norvegicus 166-174 17612778-8 2007 This study shows that conversion from CsA to tacrolimus therapy leads to a reversal of the CsA-induced bone loss, which can probably be mediated by downregulation of IL-1beta, IL-6, and TNF-alpha production. Tacrolimus 45-55 interleukin 6 Rattus norvegicus 176-180 17612778-8 2007 This study shows that conversion from CsA to tacrolimus therapy leads to a reversal of the CsA-induced bone loss, which can probably be mediated by downregulation of IL-1beta, IL-6, and TNF-alpha production. Tacrolimus 45-55 tumor necrosis factor Rattus norvegicus 186-195 17630656-2 2007 The FKBPs are distinguished by their peptidyl-prolyl cis-trans isomerase (PPIase) activity and ability to bind the immunosuppressive drugs FK506 and rapamycin. Tacrolimus 139-144 peptidyl-prolyl cis-trans isomerase Bombyx mori 74-80 17391324-8 2007 CONCLUSIONS: This study shows that the CYP3A5*3 genetic polymorphisms may be associated with the individual difference in tacrolimus pharmacokinetics. Tacrolimus 122-132 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 39-45 17568575-0 2007 Polymorphisms of tumor necrosis factor-alpha, interleukin-10, cytochrome P450 3A5 and ABCB1 in Chinese liver transplant patients treated with immunosuppressant tacrolimus. Tacrolimus 160-170 ATP binding cassette subfamily B member 1 Homo sapiens 86-91 17568575-5 2007 RESULTS: The IL-10 G-1082A polymorphism in recipients was significantly associated with tacrolimus concentration/dose (C/D) ratios (IL-10-1082GG < GA < AA) within the first 3weeks posttransplantation (P < 0.05). Tacrolimus 88-98 interleukin 10 Homo sapiens 13-18 17568575-5 2007 RESULTS: The IL-10 G-1082A polymorphism in recipients was significantly associated with tacrolimus concentration/dose (C/D) ratios (IL-10-1082GG < GA < AA) within the first 3weeks posttransplantation (P < 0.05). Tacrolimus 88-98 interleukin 10 Homo sapiens 132-137 17568575-7 2007 CONCLUSIONS: The IL-10 G-1082A and CYP3A5()3 polymorphisms may influence the interindividual variability of tacrolimus pharmacokinetics in Chinese liver transplant patients. Tacrolimus 108-118 interleukin 10 Homo sapiens 17-22 17568575-7 2007 CONCLUSIONS: The IL-10 G-1082A and CYP3A5()3 polymorphisms may influence the interindividual variability of tacrolimus pharmacokinetics in Chinese liver transplant patients. Tacrolimus 108-118 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 35-41 17568575-0 2007 Polymorphisms of tumor necrosis factor-alpha, interleukin-10, cytochrome P450 3A5 and ABCB1 in Chinese liver transplant patients treated with immunosuppressant tacrolimus. Tacrolimus 160-170 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 62-81 17635182-0 2007 Tacrolimus pharmacokinetics and pharmacogenetics: influence of adenosine triphosphate-binding cassette B1 (ABCB1) and cytochrome (CYP) 3A polymorphisms. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 63-105 17635182-0 2007 Tacrolimus pharmacokinetics and pharmacogenetics: influence of adenosine triphosphate-binding cassette B1 (ABCB1) and cytochrome (CYP) 3A polymorphisms. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 107-112 17635182-2 2007 P-glycoprotein (P-gp), encoded by the adenosine triphosphate-binding cassette B1 (ABCB1) and the cytochrome (CYP) 3A4 and 3A5 enzymes appears to play a role in the tacrolimus metabolism. Tacrolimus 164-174 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 17635182-2 2007 P-glycoprotein (P-gp), encoded by the adenosine triphosphate-binding cassette B1 (ABCB1) and the cytochrome (CYP) 3A4 and 3A5 enzymes appears to play a role in the tacrolimus metabolism. Tacrolimus 164-174 ATP binding cassette subfamily B member 1 Homo sapiens 16-20 17635182-2 2007 P-glycoprotein (P-gp), encoded by the adenosine triphosphate-binding cassette B1 (ABCB1) and the cytochrome (CYP) 3A4 and 3A5 enzymes appears to play a role in the tacrolimus metabolism. Tacrolimus 164-174 ATP binding cassette subfamily B member 1 Homo sapiens 38-80 17635182-2 2007 P-glycoprotein (P-gp), encoded by the adenosine triphosphate-binding cassette B1 (ABCB1) and the cytochrome (CYP) 3A4 and 3A5 enzymes appears to play a role in the tacrolimus metabolism. Tacrolimus 164-174 ATP binding cassette subfamily B member 1 Homo sapiens 82-87 17635182-10 2007 We noticed that patients carrying a CYP3A5*1 allele require a twofold higher tacrolimus dose compared with homozygous carriers of the CYP3A5*3 variant allele to maintain the target dnAUC(0-12). Tacrolimus 77-87 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 17031644-1 2007 PURPOSE: We sought to determine the effects of the immunosuppressants, cyclosporin A (CsA), tacrolimus and sirolimus, on drug transport by the ATP-binding cassette proteins, P-glycoprotein (Pgp; ABCB1), multidrug resistance protein-1 (MRP-1; ABCC1) and breast cancer resistance protein (BCRP; ABCG2), and the major vault protein lung resistance protein (LRP). Tacrolimus 92-102 ATP binding cassette subfamily B member 1 Homo sapiens 174-188 17478757-1 2007 OBJECTIVES: FK506 Binding Protein 12 and its related isoform 12.6 (FKBP12/12.6) stabilize a closed state of intracellular Ca2+ release channels (ryanodine receptors [RyRs]), and in myocytes removal of FKBP12/12.6 from RyRs alters intracellular Ca2+ levels. Tacrolimus 12-17 FK506 binding protein 1a Mus musculus 67-73 17478757-1 2007 OBJECTIVES: FK506 Binding Protein 12 and its related isoform 12.6 (FKBP12/12.6) stabilize a closed state of intracellular Ca2+ release channels (ryanodine receptors [RyRs]), and in myocytes removal of FKBP12/12.6 from RyRs alters intracellular Ca2+ levels. Tacrolimus 12-17 FK506 binding protein 1a Mus musculus 201-207 17478757-2 2007 The immunosuppressive drugs rapamycin and FK506 bind and displace FKBP12/12.6 from RyRs, and can also cause endothelial dysfunction and hypertension. Tacrolimus 42-47 FK506 binding protein 1a Mus musculus 66-72 17478757-7 2007 In hypertensive FKBP12.6-/- mice, systolic blood pressures were further elevated after treatment with either rapamycin or FK506. Tacrolimus 122-127 FK506 binding protein 1b Mus musculus 16-24 17478757-9 2007 CONCLUSIONS: Complete removal of FKBP12 and 12.6 from endothelial RyRs induces an intracellular Ca2+ leak which may contribute to the pathogenesis of endothelial dysfunction and hypertension caused by rapamycin or FK506. Tacrolimus 214-219 FK506 binding protein 1a Mus musculus 33-39 17031644-1 2007 PURPOSE: We sought to determine the effects of the immunosuppressants, cyclosporin A (CsA), tacrolimus and sirolimus, on drug transport by the ATP-binding cassette proteins, P-glycoprotein (Pgp; ABCB1), multidrug resistance protein-1 (MRP-1; ABCC1) and breast cancer resistance protein (BCRP; ABCG2), and the major vault protein lung resistance protein (LRP). Tacrolimus 92-102 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 253-285 17031644-1 2007 PURPOSE: We sought to determine the effects of the immunosuppressants, cyclosporin A (CsA), tacrolimus and sirolimus, on drug transport by the ATP-binding cassette proteins, P-glycoprotein (Pgp; ABCB1), multidrug resistance protein-1 (MRP-1; ABCC1) and breast cancer resistance protein (BCRP; ABCG2), and the major vault protein lung resistance protein (LRP). Tacrolimus 92-102 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 287-291 17031644-1 2007 PURPOSE: We sought to determine the effects of the immunosuppressants, cyclosporin A (CsA), tacrolimus and sirolimus, on drug transport by the ATP-binding cassette proteins, P-glycoprotein (Pgp; ABCB1), multidrug resistance protein-1 (MRP-1; ABCC1) and breast cancer resistance protein (BCRP; ABCG2), and the major vault protein lung resistance protein (LRP). Tacrolimus 92-102 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 293-298 17683383-0 2007 Relationship between Kaposi"s varicelliform eruption in Japanese patients with atopic dermatitis treated with tacrolimus ointment and genetic polymorphisms in the IL-18 gene promoter region. Tacrolimus 110-120 interleukin 18 Homo sapiens 163-168 17683383-4 2007 IL-18 gene promoter polymorphisms were analyzed in 21 AD patients treated with tacrolimus ointment and in 100 healthy volunteers. Tacrolimus 79-89 interleukin 18 Homo sapiens 0-5 17683383-5 2007 Six AD patients with Kaposi"s varicelliform eruption during the treatment with tacrolimus ointment showed significantly higher frequency in G-to-C mutations at the IL-18 gene promoter region -137 compared with 15 AD patients without Kaposi"s varicelliform eruption. Tacrolimus 79-89 interleukin 18 Homo sapiens 164-169 17683383-7 2007 These results suggest that the onset of Kaposi"s varicelliform eruption following the treatment with tacrolimus ointment is associated with the mutation of G-to-C in the IL-18 gene promoter region -137, and that caution is required when using tacrolimus ointment for treating AD patients with this mutation. Tacrolimus 101-111 interleukin 18 Homo sapiens 170-175 17683383-7 2007 These results suggest that the onset of Kaposi"s varicelliform eruption following the treatment with tacrolimus ointment is associated with the mutation of G-to-C in the IL-18 gene promoter region -137, and that caution is required when using tacrolimus ointment for treating AD patients with this mutation. Tacrolimus 243-253 interleukin 18 Homo sapiens 170-175 17542013-7 2007 Severity of colitis as determined by myeloperoxidase activity was found to have linear correlation to changes in tacrolimus degradation (R2 = 0.8299). Tacrolimus 113-123 myeloperoxidase Rattus norvegicus 37-52 17591533-7 2007 A significant interaction of ACE inhibitors with cyclosporine in affecting LVMi change was shown by means of post hoc multiple regression analysis (P < 0.01; differences between cyclosporine and tacrolimus group, 13.3 +/- 3.9 g/m(2.7); 95% CI, 5.3 to 21.2; P < 0.01 in the ACE-inhibitor group; 3.7 +/- 4.2 g/m(2.7); 95% CI, -4.7 to 12.2; P = 0.4 in the control group). Tacrolimus 198-208 angiotensin I converting enzyme Homo sapiens 29-32 17031644-1 2007 PURPOSE: We sought to determine the effects of the immunosuppressants, cyclosporin A (CsA), tacrolimus and sirolimus, on drug transport by the ATP-binding cassette proteins, P-glycoprotein (Pgp; ABCB1), multidrug resistance protein-1 (MRP-1; ABCC1) and breast cancer resistance protein (BCRP; ABCG2), and the major vault protein lung resistance protein (LRP). Tacrolimus 92-102 major vault protein Homo sapiens 354-357 17031644-9 2007 CONCLUSIONS: CsA, tacrolimus and sirolimus modulate drug transport by Pgp, MRP-1 and BCRP and CsA and sirolimus modulate drug transport by LRP at concentrations that differ from immunosuppressive concentrations and maximum tolerated concentrations. Tacrolimus 18-28 ATP binding cassette subfamily B member 1 Homo sapiens 70-73 17031644-9 2007 CONCLUSIONS: CsA, tacrolimus and sirolimus modulate drug transport by Pgp, MRP-1 and BCRP and CsA and sirolimus modulate drug transport by LRP at concentrations that differ from immunosuppressive concentrations and maximum tolerated concentrations. Tacrolimus 18-28 ATP binding cassette subfamily B member 1 Homo sapiens 75-80 17031644-1 2007 PURPOSE: We sought to determine the effects of the immunosuppressants, cyclosporin A (CsA), tacrolimus and sirolimus, on drug transport by the ATP-binding cassette proteins, P-glycoprotein (Pgp; ABCB1), multidrug resistance protein-1 (MRP-1; ABCC1) and breast cancer resistance protein (BCRP; ABCG2), and the major vault protein lung resistance protein (LRP). Tacrolimus 92-102 ATP binding cassette subfamily B member 1 Homo sapiens 190-193 17031644-1 2007 PURPOSE: We sought to determine the effects of the immunosuppressants, cyclosporin A (CsA), tacrolimus and sirolimus, on drug transport by the ATP-binding cassette proteins, P-glycoprotein (Pgp; ABCB1), multidrug resistance protein-1 (MRP-1; ABCC1) and breast cancer resistance protein (BCRP; ABCG2), and the major vault protein lung resistance protein (LRP). Tacrolimus 92-102 ATP binding cassette subfamily B member 1 Homo sapiens 195-200 17031644-1 2007 PURPOSE: We sought to determine the effects of the immunosuppressants, cyclosporin A (CsA), tacrolimus and sirolimus, on drug transport by the ATP-binding cassette proteins, P-glycoprotein (Pgp; ABCB1), multidrug resistance protein-1 (MRP-1; ABCC1) and breast cancer resistance protein (BCRP; ABCG2), and the major vault protein lung resistance protein (LRP). Tacrolimus 92-102 ATP binding cassette subfamily B member 1 Homo sapiens 203-233 17031644-1 2007 PURPOSE: We sought to determine the effects of the immunosuppressants, cyclosporin A (CsA), tacrolimus and sirolimus, on drug transport by the ATP-binding cassette proteins, P-glycoprotein (Pgp; ABCB1), multidrug resistance protein-1 (MRP-1; ABCC1) and breast cancer resistance protein (BCRP; ABCG2), and the major vault protein lung resistance protein (LRP). Tacrolimus 92-102 ATP binding cassette subfamily B member 1 Homo sapiens 235-240 17031644-9 2007 CONCLUSIONS: CsA, tacrolimus and sirolimus modulate drug transport by Pgp, MRP-1 and BCRP and CsA and sirolimus modulate drug transport by LRP at concentrations that differ from immunosuppressive concentrations and maximum tolerated concentrations. Tacrolimus 18-28 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 85-89 17031644-1 2007 PURPOSE: We sought to determine the effects of the immunosuppressants, cyclosporin A (CsA), tacrolimus and sirolimus, on drug transport by the ATP-binding cassette proteins, P-glycoprotein (Pgp; ABCB1), multidrug resistance protein-1 (MRP-1; ABCC1) and breast cancer resistance protein (BCRP; ABCG2), and the major vault protein lung resistance protein (LRP). Tacrolimus 92-102 ATP binding cassette subfamily C member 1 Homo sapiens 242-247 17565322-5 2007 Interestingly, induction of IL-21 was highly dependent on IL-2 (as in the presence of anti-IL-2, anti-IL-2R alpha-chain, and the immunosuppressive drugs cyclosporine A, tacrolimus, and rapamycin) the transcription of IL-21 was almost completely inhibited, whereas in the presence of exogenous IL-2 the mRNA expression of IL-21 was even more upregulated. Tacrolimus 169-179 interleukin 21 Homo sapiens 28-33 16900300-0 2007 Tacrolimus as a reinforcement therapy for a patient with MPO-ANCA-associated diffuse alveolar hemorrhage. Tacrolimus 0-10 myeloperoxidase Homo sapiens 57-60 16900300-5 2007 Tacrolimus was employed for the maintenance therapy, and the oral prednisolone dosage could successfully be tapered without recurrence, along with the decrement of the titer of MPO-ANCA. Tacrolimus 0-10 myeloperoxidase Homo sapiens 177-180 17523132-1 2007 The effect of tacrolimus (FK506) on down-regulation of IL-2 production by T cells is considered to be mainly responsible for its strong suppression of immunological events. Tacrolimus 14-24 interleukin 2 Homo sapiens 55-59 17523132-1 2007 The effect of tacrolimus (FK506) on down-regulation of IL-2 production by T cells is considered to be mainly responsible for its strong suppression of immunological events. Tacrolimus 26-31 interleukin 2 Homo sapiens 55-59 17645716-1 2007 The mTOR (mammalian target of rapamycin) inhibitors sirolimus (SRL) and everolimus (EVL) are potent immunosuppressive agents, which allow reducing the dose of the nephrotoxic calcineurin inhibitors cyclosporin and tacrolimus (TAC) in solid organ transplant recipients. Tacrolimus 214-224 mechanistic target of rapamycin kinase Homo sapiens 4-8 17645716-1 2007 The mTOR (mammalian target of rapamycin) inhibitors sirolimus (SRL) and everolimus (EVL) are potent immunosuppressive agents, which allow reducing the dose of the nephrotoxic calcineurin inhibitors cyclosporin and tacrolimus (TAC) in solid organ transplant recipients. Tacrolimus 214-224 mechanistic target of rapamycin kinase Homo sapiens 10-39 17594193-2 2007 Tacrolimus inhibits the activation of an essential transcription factor for the transcription of cytokine genes in T cells leading to a decreased production of cytokines such as IL-2 and IFN-gamma. Tacrolimus 0-10 interleukin 2 Homo sapiens 178-182 17594193-2 2007 Tacrolimus inhibits the activation of an essential transcription factor for the transcription of cytokine genes in T cells leading to a decreased production of cytokines such as IL-2 and IFN-gamma. Tacrolimus 0-10 interferon gamma Homo sapiens 187-196 17554808-1 2007 FKBP-12 mediates the immunosuppressive actions of FK506 and rapamycin, and modulates the activities of the ryanodine, IP3 and type 1 TGF-ss receptors. Tacrolimus 50-55 peptidyl-prolyl cis-trans isomerase FKBP1A Oryctolagus cuniculus 0-7 18240909-4 2007 It has been recently shown that the CYP3A5*3 polymorphism is associated with pharmacokinetics of tacrolimus and sirolimus. Tacrolimus 97-107 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 17603267-6 2007 In this paper, we review the population pharmacokinetic and pharmacogenomic analysis of tacrolimus, focusing on an efflux transporter P-glycoprotein (multidrug resistance 1 [MDR1/ABCB1]) and drug-metabolizing enzymes cytochrome P450 (CYP) 3A4 and 3A5, and describe Bayesian forecasting to individualize the tacrolimus dose in de novo living-donor liver transplant recipients. Tacrolimus 88-98 ATP binding cassette subfamily B member 1 Homo sapiens 134-172 17603267-6 2007 In this paper, we review the population pharmacokinetic and pharmacogenomic analysis of tacrolimus, focusing on an efflux transporter P-glycoprotein (multidrug resistance 1 [MDR1/ABCB1]) and drug-metabolizing enzymes cytochrome P450 (CYP) 3A4 and 3A5, and describe Bayesian forecasting to individualize the tacrolimus dose in de novo living-donor liver transplant recipients. Tacrolimus 88-98 ATP binding cassette subfamily B member 1 Homo sapiens 174-178 17603267-6 2007 In this paper, we review the population pharmacokinetic and pharmacogenomic analysis of tacrolimus, focusing on an efflux transporter P-glycoprotein (multidrug resistance 1 [MDR1/ABCB1]) and drug-metabolizing enzymes cytochrome P450 (CYP) 3A4 and 3A5, and describe Bayesian forecasting to individualize the tacrolimus dose in de novo living-donor liver transplant recipients. Tacrolimus 88-98 ATP binding cassette subfamily B member 1 Homo sapiens 179-184 17603267-6 2007 In this paper, we review the population pharmacokinetic and pharmacogenomic analysis of tacrolimus, focusing on an efflux transporter P-glycoprotein (multidrug resistance 1 [MDR1/ABCB1]) and drug-metabolizing enzymes cytochrome P450 (CYP) 3A4 and 3A5, and describe Bayesian forecasting to individualize the tacrolimus dose in de novo living-donor liver transplant recipients. Tacrolimus 307-317 ATP binding cassette subfamily B member 1 Homo sapiens 174-178 17603267-6 2007 In this paper, we review the population pharmacokinetic and pharmacogenomic analysis of tacrolimus, focusing on an efflux transporter P-glycoprotein (multidrug resistance 1 [MDR1/ABCB1]) and drug-metabolizing enzymes cytochrome P450 (CYP) 3A4 and 3A5, and describe Bayesian forecasting to individualize the tacrolimus dose in de novo living-donor liver transplant recipients. Tacrolimus 307-317 ATP binding cassette subfamily B member 1 Homo sapiens 179-184 17313373-6 2007 Similarly, K201 was able to suppress spontaneous Ca2+ release in FK506-treated HEK-293 cells co-expressing RyR2 and FKBP12.6. Tacrolimus 65-70 ryanodine receptor 2 Homo sapiens 107-111 17313373-6 2007 Similarly, K201 was able to suppress spontaneous Ca2+ release in FK506-treated HEK-293 cells co-expressing RyR2 and FKBP12.6. Tacrolimus 65-70 FKBP prolyl isomerase 1B Homo sapiens 116-124 17565322-5 2007 Interestingly, induction of IL-21 was highly dependent on IL-2 (as in the presence of anti-IL-2, anti-IL-2R alpha-chain, and the immunosuppressive drugs cyclosporine A, tacrolimus, and rapamycin) the transcription of IL-21 was almost completely inhibited, whereas in the presence of exogenous IL-2 the mRNA expression of IL-21 was even more upregulated. Tacrolimus 169-179 interleukin 2 Homo sapiens 28-32 17565322-5 2007 Interestingly, induction of IL-21 was highly dependent on IL-2 (as in the presence of anti-IL-2, anti-IL-2R alpha-chain, and the immunosuppressive drugs cyclosporine A, tacrolimus, and rapamycin) the transcription of IL-21 was almost completely inhibited, whereas in the presence of exogenous IL-2 the mRNA expression of IL-21 was even more upregulated. Tacrolimus 169-179 interleukin 2 Homo sapiens 58-62 17565322-5 2007 Interestingly, induction of IL-21 was highly dependent on IL-2 (as in the presence of anti-IL-2, anti-IL-2R alpha-chain, and the immunosuppressive drugs cyclosporine A, tacrolimus, and rapamycin) the transcription of IL-21 was almost completely inhibited, whereas in the presence of exogenous IL-2 the mRNA expression of IL-21 was even more upregulated. Tacrolimus 169-179 interleukin 2 Homo sapiens 58-62 17551589-5 2007 METHODOLOGY/PRINCIPAL FINDINGS: Here, the role of Apis mellifera TOR (amTOR) in caste determination is examined by rapamycin/FK506 pharmacology and RNA interference (RNAi) gene knockdown. Tacrolimus 125-130 serine/threonine-protein kinase mTOR Apis mellifera 65-68 17551589-5 2007 METHODOLOGY/PRINCIPAL FINDINGS: Here, the role of Apis mellifera TOR (amTOR) in caste determination is examined by rapamycin/FK506 pharmacology and RNA interference (RNAi) gene knockdown. Tacrolimus 125-130 serine/threonine-protein kinase mTOR Apis mellifera 70-75 17551589-6 2007 We show that in queen-destined larvae, the TOR inhibitor rapamycin induces the development of worker characters that are blocked by the antagonist FK506. Tacrolimus 147-152 serine/threonine-protein kinase mTOR Apis mellifera 43-46 17482516-6 2007 However, the calcineurin inhibitors, tacrolimus and cyclosporine A (CsA), inhibited the IL-18-enhanced cytokine production and lymphocyte proliferation without any effect on the adhesion molecule expression. Tacrolimus 37-47 interleukin 18 Homo sapiens 88-93 17230494-10 2007 Mucosal Foxp3 mRNA levels and Foxp3+CD3+ cells were significantly reduced in transplant recipients using prednisone/azathioprine/tacrolimus compared with controls but no direct relationship between Foxp3 expression and 1 specific drug was detected. Tacrolimus 129-139 forkhead box P3 Homo sapiens 8-13 17613169-6 2007 Our results show that the BBH is critical for inhibition of CN by Cyp-CsA and FKBP-FK506. Tacrolimus 83-88 gamma-butyrobetaine hydroxylase 1 Homo sapiens 26-29 17388925-7 2007 RESULTS: Topical application of pimecrolimus and tacrolimus was followed by an initial release of substance P and calcitonin gene-related peptide from primary afferent nerve fibres in murine skin during the early inflammatory response. Tacrolimus 49-59 tachykinin 1 Mus musculus 98-109 17355970-4 2007 Using an approach with FK506, an inhibitor of Cn, and mRNA silencing by small interference RNA we show that mitochondrial stress-activated Cn is critical for increased GLUT 4 and IGF1R expression and activation. Tacrolimus 23-28 insulin-like growth factor I receptor Mus musculus 179-184 17377957-0 2007 Influence of rabeprazole and lansoprazole on the pharmacokinetics of tacrolimus in relation to CYP2C19, CYP3A5 and MDR1 polymorphisms in renal transplant recipients. Tacrolimus 69-79 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 104-110 17377957-0 2007 Influence of rabeprazole and lansoprazole on the pharmacokinetics of tacrolimus in relation to CYP2C19, CYP3A5 and MDR1 polymorphisms in renal transplant recipients. Tacrolimus 69-79 ATP binding cassette subfamily B member 1 Homo sapiens 115-119 17377957-1 2007 The objective of this study was to evaluate whether genetic polymorphisms of CYP2C19, CYP3A5 and MDR1 significantly impact the interaction between tacrolimus and rabeprazole or lansoprazole. Tacrolimus 147-157 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 77-84 17377957-1 2007 The objective of this study was to evaluate whether genetic polymorphisms of CYP2C19, CYP3A5 and MDR1 significantly impact the interaction between tacrolimus and rabeprazole or lansoprazole. Tacrolimus 147-157 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 86-92 17377957-1 2007 The objective of this study was to evaluate whether genetic polymorphisms of CYP2C19, CYP3A5 and MDR1 significantly impact the interaction between tacrolimus and rabeprazole or lansoprazole. Tacrolimus 147-157 ATP binding cassette subfamily B member 1 Homo sapiens 97-101 17377957-4 2007 The mean daily dose and the dose-adjusted area under the plasma concentration-time curves from 0 to 12 h (AUC(0-12)) of tacrolimus coadministered with rabeprazole or lansoprazole were the lowest and highest, respectively, in CYP2C19 poor metabolizers (PMs) having the CYP3A5*3/*3 genotype (0.084 and 0.112 mg/kg/day and 1.269 and 1.033 ng.h/ml/mg/kg, respectively). Tacrolimus 120-130 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 225-232 17377957-4 2007 The mean daily dose and the dose-adjusted area under the plasma concentration-time curves from 0 to 12 h (AUC(0-12)) of tacrolimus coadministered with rabeprazole or lansoprazole were the lowest and highest, respectively, in CYP2C19 poor metabolizers (PMs) having the CYP3A5*3/*3 genotype (0.084 and 0.112 mg/kg/day and 1.269 and 1.033 ng.h/ml/mg/kg, respectively). Tacrolimus 120-130 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 268-274 17377957-5 2007 On the other hand, the mean dose-adjusted AUC(0-12) of tacrolimus coadministered with rabeprazole or lansoprazole were the highest in CYP2C19 PMs having the MDR13435CC+CT genotype, but not significantly. Tacrolimus 55-65 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 134-141 17377957-6 2007 The present study indicates that there are significant interactions between tacrolimus and rabeprazole or lansoprazole in CYP2C19 PM renal transplant recipients bearing the CYP3A5*3/*3 genotypes. Tacrolimus 76-86 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 122-129 17377957-6 2007 The present study indicates that there are significant interactions between tacrolimus and rabeprazole or lansoprazole in CYP2C19 PM renal transplant recipients bearing the CYP3A5*3/*3 genotypes. Tacrolimus 76-86 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 173-179 17703430-7 2007 Immunohistochemical scores showed that expression of TGF-beta is up-regulated, and bone morphogenic protein (BMP-7) is down-regulated with FK506 toxicity. Tacrolimus 139-144 bone morphogenetic protein 7 Rattus norvegicus 109-114 17292639-0 2007 The immunosuppressant FK506 promotes development of the chondrogenic phenotype in human synovial stromal cells via modulation of the Smad signaling pathway. Tacrolimus 22-27 SMAD family member 1 Homo sapiens 133-137 17292639-4 2007 Furthermore, levels and intracellular location of phosphorylated Smad proteins related to BMP signaling and TGFbeta signaling were evaluated following exposure to FK506. Tacrolimus 163-168 SMAD family member 1 Homo sapiens 65-69 17292639-4 2007 Furthermore, levels and intracellular location of phosphorylated Smad proteins related to BMP signaling and TGFbeta signaling were evaluated following exposure to FK506. Tacrolimus 163-168 bone morphogenetic protein 2 Homo sapiens 90-93 17292639-6 2007 Additionally, FK506 further enhanced chondrogenesis of synovial stromal cells (SSCs) induced by BMP2 and TGFbeta(1), also in a dose-dependent manner. Tacrolimus 14-19 bone morphogenetic protein 2 Homo sapiens 96-100 17292639-6 2007 Additionally, FK506 further enhanced chondrogenesis of synovial stromal cells (SSCs) induced by BMP2 and TGFbeta(1), also in a dose-dependent manner. Tacrolimus 14-19 transforming growth factor beta 1 Homo sapiens 105-115 17292639-7 2007 Notably, phosphorylation of Smad1/5/8 and Smad3 was significantly increased by FK506. Tacrolimus 79-84 SMAD family member 1 Homo sapiens 28-37 17292639-7 2007 Notably, phosphorylation of Smad1/5/8 and Smad3 was significantly increased by FK506. Tacrolimus 79-84 SMAD family member 3 Homo sapiens 42-47 17292639-8 2007 Also, the ratio of nuclear translocation to cytoplasmic levels of phosphorylated Smad1/5/8 and Smad3 were increased following exposure of SSCs to FK506. Tacrolimus 146-151 SMAD family member 1 Homo sapiens 81-90 17292639-8 2007 Also, the ratio of nuclear translocation to cytoplasmic levels of phosphorylated Smad1/5/8 and Smad3 were increased following exposure of SSCs to FK506. Tacrolimus 146-151 SMAD family member 3 Homo sapiens 95-100 17292639-9 2007 Moreover, inhibition of Smad signaling significantly abrogated FK506-induced chondrogenic differentiation of SSCs. Tacrolimus 63-68 SMAD family member 1 Homo sapiens 24-28 17292639-10 2007 CONCLUSION: This study demonstrated that FK506 promotes chondrogenic differentiation of hSSCs likely via impact on Smad signaling pathways. Tacrolimus 41-46 SMAD family member 1 Homo sapiens 115-119 17355875-5 2007 In acutely dissociated CA1 pyramidal neurons of rat hippocampus, superfusion of FK506 (0.01-100 microM) selectively inhibited the delayed rectifier K(+) current (I(K)) with an IC(50) value of 13.2+/-4.9 microM. Tacrolimus 80-85 carbonic anhydrase 1 Rattus norvegicus 23-26 17347151-0 2007 Cyclosporin A and FK506 inhibit IL-12p40 production through the calmodulin/calmodulin-dependent protein kinase-activated phosphoinositide 3-kinase in lipopolysaccharide-stimulated human monocytic cells. Tacrolimus 18-23 interleukin 12b Mus musculus 32-40 17347151-0 2007 Cyclosporin A and FK506 inhibit IL-12p40 production through the calmodulin/calmodulin-dependent protein kinase-activated phosphoinositide 3-kinase in lipopolysaccharide-stimulated human monocytic cells. Tacrolimus 18-23 phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha Homo sapiens 121-146 17347151-2 2007 However, the mechanisms underlying the inhibitory effects of CyA and FK506 on the production of IL-12p40, a critical component of IL-12, remain unknown. Tacrolimus 69-74 interleukin 12b Mus musculus 96-104 17347151-9 2007 Importantly, both CyA and FK506 down-regulated LPS-induced IL-12p40 transcription by inhibiting CaM/CaMK-II-activated PI3K and their downstream transcription factors NFkappaB and AP-1 independent of the JNK pathway. Tacrolimus 26-31 toll-like receptor 4 Mus musculus 47-50 17347151-9 2007 Importantly, both CyA and FK506 down-regulated LPS-induced IL-12p40 transcription by inhibiting CaM/CaMK-II-activated PI3K and their downstream transcription factors NFkappaB and AP-1 independent of the JNK pathway. Tacrolimus 26-31 interleukin 12b Mus musculus 59-67 17347151-9 2007 Importantly, both CyA and FK506 down-regulated LPS-induced IL-12p40 transcription by inhibiting CaM/CaMK-II-activated PI3K and their downstream transcription factors NFkappaB and AP-1 independent of the JNK pathway. Tacrolimus 26-31 calcium/calmodulin dependent protein kinase II gamma Homo sapiens 100-107 17347151-9 2007 Importantly, both CyA and FK506 down-regulated LPS-induced IL-12p40 transcription by inhibiting CaM/CaMK-II-activated PI3K and their downstream transcription factors NFkappaB and AP-1 independent of the JNK pathway. Tacrolimus 26-31 nuclear factor kappa B subunit 1 Homo sapiens 166-174 17347151-9 2007 Importantly, both CyA and FK506 down-regulated LPS-induced IL-12p40 transcription by inhibiting CaM/CaMK-II-activated PI3K and their downstream transcription factors NFkappaB and AP-1 independent of the JNK pathway. Tacrolimus 26-31 mitogen-activated protein kinase 8 Homo sapiens 203-206 17320203-3 2007 The calpain inhibitor, calpeptin, and the calcineurin inhibitors, FK506 and cyclosporine A, inhibited acetylcholinesterase expression at both mRNA and protein levels and suppressed the activity of the human acetylcholinesterase promoter. Tacrolimus 66-71 acetylcholinesterase (Cartwright blood group) Homo sapiens 102-122 17307394-6 2007 Among MG patients, the mean ECCT was longer for patients with thymoma than for those without it (P=0.04), and was shorter for patients treated with FK506 (an immunosuppressant and also an enhancer of RyR related Ca(2+) release) than for those not receiving this treatment (p=0.04). Tacrolimus 148-153 ryanodine receptor 1 Homo sapiens 200-203 17524933-1 2007 UNLABELLED: Both tacrolimus (TAC) and sirolimus (SRL) bind to the same immunophilin FKBP12; however, their mechanisms of action are distinct. Tacrolimus 17-27 FKBP prolyl isomerase 1A Homo sapiens 71-90 17339317-9 2007 This study establishes that the secretory pathway FK506-binding PPIase enzymes are essential for normal nematode development, collagen biogenesis, and the formation of an intact exoskeleton under adverse physiological conditions. Tacrolimus 50-55 Peptidylprolyl isomerase Caenorhabditis elegans 64-70 17430486-0 2007 Influence of the CYP3A5 genotype on tacrolimus pharmacokinetics and pharmacodynamics in young kidney transplant recipients. Tacrolimus 36-46 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 17-23 17374625-8 2007 DATA SYNTHESIS: Elevated plasma concentrations and toxicities have been reported for a number of CYP3A substrates including amiodarone, carbamazepine, quinidine, tacrolimus, and cyclosporine when administered with metronidazole. Tacrolimus 162-172 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 97-102 17320203-3 2007 The calpain inhibitor, calpeptin, and the calcineurin inhibitors, FK506 and cyclosporine A, inhibited acetylcholinesterase expression at both mRNA and protein levels and suppressed the activity of the human acetylcholinesterase promoter. Tacrolimus 66-71 acetylcholinesterase (Cartwright blood group) Homo sapiens 207-227 17448782-0 2007 Bombesin protection against FK506 neurotoxicity. Tacrolimus 28-33 gastrin releasing peptide Homo sapiens 0-8 17196174-6 2007 The calcineurin inhibitors, tacrolimus (FK506) and cyclosporin A, had no effect on cell migration but completely blocked both cytokine production and the nuclear translocation of NFATc1 suggesting that Ca2+/calcineurin/NFAT is involved in CRTH2-dependent cytokine production but not chemotaxis. Tacrolimus 28-38 nuclear factor of activated T cells 1 Homo sapiens 179-185 17394163-1 2007 This study aims to investigate the potential role of endogenous interleukin (IL)-10 in long-term liver allograft survival induced by delayed immunosuppression (FK506 days 2-7). Tacrolimus 160-165 interleukin 10 Rattus norvegicus 64-83 17394163-4 2007 A transient upregulation of plasma IL-10 levels was detected in the nontreatment and FK506 treatment groups. Tacrolimus 85-90 interleukin 10 Rattus norvegicus 35-40 17394163-6 2007 Administration of IL-10-neutralizing antibody shortened the long-term isograft survival and FK506-induced indefinite allograft survival, particularly in the FK506 group. Tacrolimus 92-97 interleukin 10 Rattus norvegicus 18-23 17394163-6 2007 Administration of IL-10-neutralizing antibody shortened the long-term isograft survival and FK506-induced indefinite allograft survival, particularly in the FK506 group. Tacrolimus 157-162 interleukin 10 Rattus norvegicus 18-23 17536270-4 2007 vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) were detected in back skin in both the tacrolimus group and the minoxidil group, but not in the vaseline group. Tacrolimus 116-126 vascular endothelial growth factor A Mus musculus 0-34 17536270-4 2007 vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) were detected in back skin in both the tacrolimus group and the minoxidil group, but not in the vaseline group. Tacrolimus 116-126 vascular endothelial growth factor A Mus musculus 36-40 17536270-4 2007 vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) were detected in back skin in both the tacrolimus group and the minoxidil group, but not in the vaseline group. Tacrolimus 116-126 hepatocyte growth factor Mus musculus 46-70 17536270-4 2007 vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) were detected in back skin in both the tacrolimus group and the minoxidil group, but not in the vaseline group. Tacrolimus 116-126 hepatocyte growth factor Mus musculus 72-75 17536270-5 2007 CONCLUSION: Tacrolimus can promote the growth of hair by stimulating the hair follicle to enter anagen V in mice, which may be explained by the effects of VEGF and HGF. Tacrolimus 12-22 vascular endothelial growth factor A Mus musculus 155-159 17536270-5 2007 CONCLUSION: Tacrolimus can promote the growth of hair by stimulating the hair follicle to enter anagen V in mice, which may be explained by the effects of VEGF and HGF. Tacrolimus 12-22 hepatocyte growth factor Mus musculus 164-167 17445588-0 2007 Tacrolimus (Pan Graf) in live related renal transplantation: an initial experience of 101 recipients in India. Tacrolimus 0-10 Rho GTPase activating protein 26 Homo sapiens 16-20 17445588-3 2007 However, tacrolimus (Pan Graf, Panacea Biotec Ltd, India) has only been available in India for the last 2 years. Tacrolimus 9-19 Rho GTPase activating protein 26 Homo sapiens 25-29 17196174-6 2007 The calcineurin inhibitors, tacrolimus (FK506) and cyclosporin A, had no effect on cell migration but completely blocked both cytokine production and the nuclear translocation of NFATc1 suggesting that Ca2+/calcineurin/NFAT is involved in CRTH2-dependent cytokine production but not chemotaxis. Tacrolimus 40-45 nuclear factor of activated T cells 1 Homo sapiens 179-185 17000002-1 2007 Inhibition of the interleukin-2 (IL-2) pathway has potent immunosuppressive activity in humans as is evident from the broad therapeutic utility of cyclosporine, rapamycin, tacrolimus, and monoclonal antibodies blocking the high-affinity subunit of the IL-2 receptor (CD25). Tacrolimus 172-182 interleukin 2 Homo sapiens 18-31 17304209-7 2007 Inhibition of the expression of TNF-alpha and MMP9 by the anti-inflammatory reagent FK506 could cure the epidermal hyperplasia, suggesting a causal link between inflammation and epidermal hyperplasia. Tacrolimus 84-89 tumor necrosis factor Mus musculus 32-41 17304209-7 2007 Inhibition of the expression of TNF-alpha and MMP9 by the anti-inflammatory reagent FK506 could cure the epidermal hyperplasia, suggesting a causal link between inflammation and epidermal hyperplasia. Tacrolimus 84-89 matrix metallopeptidase 9 Mus musculus 46-50 17133350-8 2007 In addition to its inhibition of calcineurin activity, FK506 administration totally suppressed the exercise-induced IL-6 gene transcription, likely by an inhibition of p38 activation. Tacrolimus 55-60 interleukin 6 Rattus norvegicus 116-120 17133350-8 2007 In addition to its inhibition of calcineurin activity, FK506 administration totally suppressed the exercise-induced IL-6 gene transcription, likely by an inhibition of p38 activation. Tacrolimus 55-60 mitogen activated protein kinase 14 Rattus norvegicus 168-171 17000002-1 2007 Inhibition of the interleukin-2 (IL-2) pathway has potent immunosuppressive activity in humans as is evident from the broad therapeutic utility of cyclosporine, rapamycin, tacrolimus, and monoclonal antibodies blocking the high-affinity subunit of the IL-2 receptor (CD25). Tacrolimus 172-182 interleukin 2 Homo sapiens 33-37 17223872-6 2007 RESULTS: CD3+, CD4+ and CD8+ lymphocytes, and eosinophil and neutrophil granulocytes were significantly reduced in post-treatment tacrolimus specimens, while CD1a+ cells and mast cells were not. Tacrolimus 130-140 CD4 molecule Homo sapiens 15-18 17268072-5 2007 Immunohistochemical analysis showed that tacrolimus reduced the number of blood vessels positively stained for ICAM-1, E-selectin and P-selection. Tacrolimus 41-51 intercellular adhesion molecule 1 Rattus norvegicus 111-117 17268072-5 2007 Immunohistochemical analysis showed that tacrolimus reduced the number of blood vessels positively stained for ICAM-1, E-selectin and P-selection. Tacrolimus 41-51 selectin E Rattus norvegicus 119-129 17268068-0 2007 P-glycoprotein function in peripheral blood mononuclear cells of myasthenia gravis patients treated with tacrolimus. Tacrolimus 105-115 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 17268068-2 2007 A drug efflux pump P-glycoprotein (P-gp) actively transports FK506 out of target cells, thereby reducing their efficacy. Tacrolimus 61-66 ATP binding cassette subfamily B member 1 Homo sapiens 19-33 17268068-2 2007 A drug efflux pump P-glycoprotein (P-gp) actively transports FK506 out of target cells, thereby reducing their efficacy. Tacrolimus 61-66 ATP binding cassette subfamily B member 1 Homo sapiens 35-39 17268068-3 2007 We investigated the influence of FK506 therapy on the P-gp function of peripheral-blood mononuclear cells (PBMCs) in MG patients. Tacrolimus 33-38 ATP binding cassette subfamily B member 1 Homo sapiens 54-58 17268068-9 2007 The data raise the possibility that FK506 treatment attenuated P-gp function in the PBMCs of the MG patients. Tacrolimus 36-41 ATP binding cassette subfamily B member 1 Homo sapiens 63-67 16987901-9 2007 The gene expression of monocyte chemattractant protein-1 (MCP-1) and regulated upon activation, normal T cell expressed and secreted (RANTES) was markedly reduced in tacrolimus-treated eyes. Tacrolimus 166-176 C-C motif chemokine ligand 2 Rattus norvegicus 23-56 16987901-9 2007 The gene expression of monocyte chemattractant protein-1 (MCP-1) and regulated upon activation, normal T cell expressed and secreted (RANTES) was markedly reduced in tacrolimus-treated eyes. Tacrolimus 166-176 C-C motif chemokine ligand 2 Rattus norvegicus 58-63 16987901-9 2007 The gene expression of monocyte chemattractant protein-1 (MCP-1) and regulated upon activation, normal T cell expressed and secreted (RANTES) was markedly reduced in tacrolimus-treated eyes. Tacrolimus 166-176 C-C motif chemokine ligand 5 Rattus norvegicus 134-140 17223872-6 2007 RESULTS: CD3+, CD4+ and CD8+ lymphocytes, and eosinophil and neutrophil granulocytes were significantly reduced in post-treatment tacrolimus specimens, while CD1a+ cells and mast cells were not. Tacrolimus 130-140 CD8a molecule Homo sapiens 24-27 17223872-7 2007 The expression of cytokines and chemokine receptors tested, except for CXCR3, was diminished by tacrolimus treatment. Tacrolimus 96-106 C-X-C motif chemokine receptor 3 Homo sapiens 71-76 17192769-0 2007 CYP3A5 genotype markedly influences the pharmacokinetics of tacrolimus and sirolimus in kidney transplant recipients. Tacrolimus 60-70 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 17192769-6 2007 Therefore, CYP3A5 expressor status and not transporter variants is a main determinant of oral clearance, particularly for tacrolimus. Tacrolimus 122-132 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 11-17 17202418-1 2007 The effects of the immunosuppressants cyclosporin A (CsA) and tacrolimus (FK506) on the IL-1beta-induced matrix metalloproteinase-9 (MMP-9) were investigated. Tacrolimus 62-72 interleukin 1 beta Homo sapiens 88-96 16586042-0 2007 Tacrolimus and cyclosporine A inhibit human osteoclast formation via targeting the calcineurin-dependent NFAT pathway and an activation pathway for c-Jun or MITF in rheumatoid arthritis. Tacrolimus 0-10 Jun proto-oncogene, AP-1 transcription factor subunit Homo sapiens 148-153 16586042-0 2007 Tacrolimus and cyclosporine A inhibit human osteoclast formation via targeting the calcineurin-dependent NFAT pathway and an activation pathway for c-Jun or MITF in rheumatoid arthritis. Tacrolimus 0-10 melanocyte inducing transcription factor Homo sapiens 157-161 16586042-1 2007 In the present study, we aimed to determine whether tacrolimus (FK506) and cyclosporine A act directly on human osteoclast precursors obtained from patients with rheumatoid arthritis (RA) and influence monocyte-osteoclast differentiation induced by receptor activator of NF-kappaB ligand (RANKL) in vitro, the stage at which differentiation was affected and the manner in which tacrolimus or cyclosporine A affected the osteoclast signaling pathway. Tacrolimus 52-62 TNF superfamily member 11 Homo sapiens 249-287 16586042-1 2007 In the present study, we aimed to determine whether tacrolimus (FK506) and cyclosporine A act directly on human osteoclast precursors obtained from patients with rheumatoid arthritis (RA) and influence monocyte-osteoclast differentiation induced by receptor activator of NF-kappaB ligand (RANKL) in vitro, the stage at which differentiation was affected and the manner in which tacrolimus or cyclosporine A affected the osteoclast signaling pathway. Tacrolimus 52-62 TNF superfamily member 11 Homo sapiens 289-294 16586042-1 2007 In the present study, we aimed to determine whether tacrolimus (FK506) and cyclosporine A act directly on human osteoclast precursors obtained from patients with rheumatoid arthritis (RA) and influence monocyte-osteoclast differentiation induced by receptor activator of NF-kappaB ligand (RANKL) in vitro, the stage at which differentiation was affected and the manner in which tacrolimus or cyclosporine A affected the osteoclast signaling pathway. Tacrolimus 64-69 TNF superfamily member 11 Homo sapiens 249-287 16586042-1 2007 In the present study, we aimed to determine whether tacrolimus (FK506) and cyclosporine A act directly on human osteoclast precursors obtained from patients with rheumatoid arthritis (RA) and influence monocyte-osteoclast differentiation induced by receptor activator of NF-kappaB ligand (RANKL) in vitro, the stage at which differentiation was affected and the manner in which tacrolimus or cyclosporine A affected the osteoclast signaling pathway. Tacrolimus 64-69 TNF superfamily member 11 Homo sapiens 289-294 16586042-6 2007 Addition of tacrolimus or cyclosporine A resulted in a decrease in the number of TRAP-positive multinucleated cells (TRAP+ MNCs) and a decrease in the extent of lacunar resorption pit formation as compared to the control cultures; thus, human monocyte-osteoclast differentiation was more effectively inhibited at the late stage and addition of tacrolimus or cyclosporine A resulted in a decrease in the mRNA expression of NFATc1, c-Jun, and MITF at the late stage. Tacrolimus 12-22 nuclear factor of activated T cells 1 Homo sapiens 422-428 16586042-6 2007 Addition of tacrolimus or cyclosporine A resulted in a decrease in the number of TRAP-positive multinucleated cells (TRAP+ MNCs) and a decrease in the extent of lacunar resorption pit formation as compared to the control cultures; thus, human monocyte-osteoclast differentiation was more effectively inhibited at the late stage and addition of tacrolimus or cyclosporine A resulted in a decrease in the mRNA expression of NFATc1, c-Jun, and MITF at the late stage. Tacrolimus 12-22 Jun proto-oncogene, AP-1 transcription factor subunit Homo sapiens 430-435 16586042-6 2007 Addition of tacrolimus or cyclosporine A resulted in a decrease in the number of TRAP-positive multinucleated cells (TRAP+ MNCs) and a decrease in the extent of lacunar resorption pit formation as compared to the control cultures; thus, human monocyte-osteoclast differentiation was more effectively inhibited at the late stage and addition of tacrolimus or cyclosporine A resulted in a decrease in the mRNA expression of NFATc1, c-Jun, and MITF at the late stage. Tacrolimus 12-22 melanocyte inducing transcription factor Homo sapiens 441-445 16586042-7 2007 Our results suggest that tacrolimus or cyclosporine A acts directly on human osteoclast precursors in RA patients and exerts their immunosuppressive effects on human monocyte-osteoclast formation via targeting both the calcineurin-dependent NFAT pathway and activation pathway for c-Jun or MITF. Tacrolimus 25-35 Jun proto-oncogene, AP-1 transcription factor subunit Homo sapiens 281-286 16586042-7 2007 Our results suggest that tacrolimus or cyclosporine A acts directly on human osteoclast precursors in RA patients and exerts their immunosuppressive effects on human monocyte-osteoclast formation via targeting both the calcineurin-dependent NFAT pathway and activation pathway for c-Jun or MITF. Tacrolimus 25-35 melanocyte inducing transcription factor Homo sapiens 290-294 17202418-1 2007 The effects of the immunosuppressants cyclosporin A (CsA) and tacrolimus (FK506) on the IL-1beta-induced matrix metalloproteinase-9 (MMP-9) were investigated. Tacrolimus 62-72 matrix metallopeptidase 9 Homo sapiens 105-131 17202418-1 2007 The effects of the immunosuppressants cyclosporin A (CsA) and tacrolimus (FK506) on the IL-1beta-induced matrix metalloproteinase-9 (MMP-9) were investigated. Tacrolimus 62-72 matrix metallopeptidase 9 Homo sapiens 133-138 17202418-1 2007 The effects of the immunosuppressants cyclosporin A (CsA) and tacrolimus (FK506) on the IL-1beta-induced matrix metalloproteinase-9 (MMP-9) were investigated. Tacrolimus 74-79 interleukin 1 beta Homo sapiens 88-96 17202418-1 2007 The effects of the immunosuppressants cyclosporin A (CsA) and tacrolimus (FK506) on the IL-1beta-induced matrix metalloproteinase-9 (MMP-9) were investigated. Tacrolimus 74-79 matrix metallopeptidase 9 Homo sapiens 105-131 17202418-1 2007 The effects of the immunosuppressants cyclosporin A (CsA) and tacrolimus (FK506) on the IL-1beta-induced matrix metalloproteinase-9 (MMP-9) were investigated. Tacrolimus 74-79 matrix metallopeptidase 9 Homo sapiens 133-138 17158338-7 2007 In vitro and in vivo studies with losartan, small interfering RNA specific for calcineurin and FK506 demonstrated that Ang II-mediated sFlt-1 release was via Ang II type 1 receptor activation and calcineurin signaling, respectively. Tacrolimus 95-100 angiotensinogen Homo sapiens 119-125 17267565-8 2007 Treatment by tacrolimus and cyclosporin A, two inhibitors of calcineurin (but not by a related substance rapamycin, which does not inhibit calcineurin), increased the levels of AChE transcripts in the control soleus muscles and in tonically electrically stimulated soleus and EDL muscles, even to reach those in the control EDL muscles. Tacrolimus 13-23 acetylcholinesterase Rattus norvegicus 177-181 17161467-10 2007 Moreover PI3K/Akt inhibitor, NF-kappaB inhibitor, and FK506 significantly inhibited IL-15-induced IL-17 production in CD4+ T cells. Tacrolimus 54-59 interleukin 15 Homo sapiens 84-89 17161467-10 2007 Moreover PI3K/Akt inhibitor, NF-kappaB inhibitor, and FK506 significantly inhibited IL-15-induced IL-17 production in CD4+ T cells. Tacrolimus 54-59 interleukin 17A Homo sapiens 98-103 17055586-7 2007 It has recently been discovered that the cardio-protective action of the drug JTV519 can be attributed partly to its ability to stabilise the interaction between the RyR and the 12.6 kDa binding protein for the commonly used immunosuppressive drug FK506 (FKBP12.6, known as tacrolimus). Tacrolimus 274-284 ryanodine receptor 2 Homo sapiens 166-169 17307907-1 2007 FKBP52 is a member of the FK506-binding family of immunophilins and serves as a co-chaperone for steroid hormone nuclear receptors to govern appropriate hormone action in target tissues. Tacrolimus 26-31 FK506 binding protein 4 Mus musculus 0-6 17141216-1 2007 Calcineurin, the Ca2+/calmodulin-dependant serine/threonine phosphatase is the target for the immunosuppressant drugs FK506 and cyclosporine-A. Tacrolimus 118-123 calmodulin Gossypium hirsutum 22-32 17102134-7 2007 Treatment with the calcineurin inhibitors FK506 and cyclosporin A also blocked DREAM-mediated Kv4.2 channel trafficking and calcineurin de-phosphorylated GRK2-phosphorylated DREAM in vitro. Tacrolimus 42-47 potassium voltage-gated channel interacting protein 3 Homo sapiens 79-84 17102134-7 2007 Treatment with the calcineurin inhibitors FK506 and cyclosporin A also blocked DREAM-mediated Kv4.2 channel trafficking and calcineurin de-phosphorylated GRK2-phosphorylated DREAM in vitro. Tacrolimus 42-47 potassium voltage-gated channel subfamily D member 2 Homo sapiens 94-99 17102134-7 2007 Treatment with the calcineurin inhibitors FK506 and cyclosporin A also blocked DREAM-mediated Kv4.2 channel trafficking and calcineurin de-phosphorylated GRK2-phosphorylated DREAM in vitro. Tacrolimus 42-47 G protein-coupled receptor kinase 2 Homo sapiens 154-158 17102134-7 2007 Treatment with the calcineurin inhibitors FK506 and cyclosporin A also blocked DREAM-mediated Kv4.2 channel trafficking and calcineurin de-phosphorylated GRK2-phosphorylated DREAM in vitro. Tacrolimus 42-47 potassium voltage-gated channel interacting protein 3 Homo sapiens 174-179 17158338-7 2007 In vitro and in vivo studies with losartan, small interfering RNA specific for calcineurin and FK506 demonstrated that Ang II-mediated sFlt-1 release was via Ang II type 1 receptor activation and calcineurin signaling, respectively. Tacrolimus 95-100 FMS-like tyrosine kinase 1 Mus musculus 135-141 17225017-1 2007 Tacrolimus ointment is a topical calcineurin inhibitor for the treatment of atopic dermatitis. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 33-54 17198266-14 2006 Blocking both NF-kappaB and NFAT by DHMEQ and tacrolimus induces potent immunosuppression, which may become a new modality in controlling allograft rejection. Tacrolimus 46-56 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 14-23 17678714-9 2007 Strongest ICAM-1 expression coincided with sustained granulocyte accumulation at 72h which was suppressed by FK-506. Tacrolimus 109-115 intercellular adhesion molecule 1 Rattus norvegicus 10-16 17038582-4 2007 Here, we show that treatment of T cells with cyclosporin A, FK506, and dexamethasone, which are known to inhibit calcineurin and NF-kappaB, respectively, but not rapamycin, the inhibitor of mammalian target of rapamycin, selectively prevented TCR/CD3 relocalization into the IS, while relocalization of adhesion and cytoskeletal proteins as well as T cell/APC conjugate formation remained unaltered. Tacrolimus 60-65 nuclear factor kappa B subunit 1 Homo sapiens 129-138 17038582-4 2007 Here, we show that treatment of T cells with cyclosporin A, FK506, and dexamethasone, which are known to inhibit calcineurin and NF-kappaB, respectively, but not rapamycin, the inhibitor of mammalian target of rapamycin, selectively prevented TCR/CD3 relocalization into the IS, while relocalization of adhesion and cytoskeletal proteins as well as T cell/APC conjugate formation remained unaltered. Tacrolimus 60-65 mechanistic target of rapamycin kinase Homo sapiens 190-219 17260122-12 2007 On biochemical analysis, it was found that, compared to the control group, rapamycin reduced alkaline phosphatase activity and the calcium content of mesenchymal stem cells; FK506 increased alkaline phosphatase activity, calcium content, and osteocalcin content; and cyclosporine A had negligible effects. Tacrolimus 174-179 bone gamma-carboxyglutamate protein Homo sapiens 242-253 17202747-0 2007 Tacrolimus (FK506), an immunosuppressive agent, prevents indomethacin-induced small intestinal ulceration in the rat: inhibition of inducible nitric oxide synthase expression. Tacrolimus 0-10 nitric oxide synthase 2 Rattus norvegicus 132-163 17202747-0 2007 Tacrolimus (FK506), an immunosuppressive agent, prevents indomethacin-induced small intestinal ulceration in the rat: inhibition of inducible nitric oxide synthase expression. Tacrolimus 12-17 nitric oxide synthase 2 Rattus norvegicus 132-163 16838106-6 2007 Ang II-induced apoptosis was inhibited by CV-11974 (Candesartan; Ang II type 1 [AT1] receptor blocker), SK&F96365, and FK506 (calcineurin inhibitor). Tacrolimus 123-128 angiotensinogen Rattus norvegicus 0-6 17202802-3 2007 On the other hand, cyclosporine and tacrolimus competitively inhibited CYP3A4-mediated nifedipine oxidation activity, with inhibition constants (K(i)) of 1.42 and 0.36 muM, respectively. Tacrolimus 36-46 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 71-77 17217059-12 2007 Some pharmaceutical compounds, such as the immunosuppressant FK506, affect the Ca2+ balance by modulating TRPV5 gene expression. Tacrolimus 61-66 transient receptor potential cation channel, subfamily V, member 5 Mus musculus 106-111 17202747-8 2007 The increases in MPO activity and TBARS were also significantly attenuated by tacrolimus. Tacrolimus 78-88 myeloperoxidase Rattus norvegicus 17-20 17202747-9 2007 The expression of iNOS mRNA was markedly enhanced when examined 6 h after indomethacin administration, and this response was counteracted by tacrolimus. Tacrolimus 141-151 nitric oxide synthase 2 Rattus norvegicus 18-22 17275526-5 2007 RESULTS: After treatment with FK506, the proportion of CD4(+) cells significantly increased in the spleen at 12 hours, decreasing to control levels at 24 and 48 hours. Tacrolimus 30-35 Cd4 molecule Rattus norvegicus 55-58 17275526-8 2007 CONCLUSIONS: These results indicated that FK506 increased CD4(+) spleen lymphocyte activation in a short time, inducing a subsequent increase in CD4(+) cells subsets in peripheral blood. Tacrolimus 42-47 Cd4 molecule Rattus norvegicus 58-61 17275526-8 2007 CONCLUSIONS: These results indicated that FK506 increased CD4(+) spleen lymphocyte activation in a short time, inducing a subsequent increase in CD4(+) cells subsets in peripheral blood. Tacrolimus 42-47 Cd4 molecule Rattus norvegicus 145-148 17176100-2 2006 Inhibition of the protein phosphatase calcineurin (CaN), which has been implicated in the FK506-mediated blockade of T cell proliferation, was shown to involve a gain of function in the FKBP12/FK506 complex. Tacrolimus 193-198 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 186-192 17176100-5 2006 The different capacities of FK506/FKBP complexes to affect CaN activity are partially caused by substitutions corresponding to the amino acid side chains K34 and I90 of FKBP12. Tacrolimus 28-33 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 169-175 17176100-6 2006 Only the FK506 complexes of FKBP12, FKBP12.6, and FKBP51 showed high affinity to CaN; small interfering RNA against these FKBP allowed defining the contribution of individual FKBP in an NFAT reporter gene assay. Tacrolimus 9-14 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 28-34 17176100-6 2006 Only the FK506 complexes of FKBP12, FKBP12.6, and FKBP51 showed high affinity to CaN; small interfering RNA against these FKBP allowed defining the contribution of individual FKBP in an NFAT reporter gene assay. Tacrolimus 9-14 FKBP prolyl isomerase 1B Homo sapiens 36-44 17106006-7 2006 Studies have found that carriers of CYP3A5*1 consistently have higher clearance rates of tacrolimus than do CYP3A5*3 homozygotes. Tacrolimus 89-99 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 17106006-8 2006 The influences of CYP3A5 alleles on cyclosporine metabolism and the MDR1 C3435T polymorphism on tacrolimus metabolism remain controversial. Tacrolimus 96-106 ATP binding cassette subfamily B member 1 Homo sapiens 68-72 17106006-9 2006 CONCLUSION: For renal transplant recipients receiving tacrolimus as an immunosuppressant, practitioners can expect CYP3A5*1 carriers to have a tacrolimus clearance 25-45% greater than that of CYP3A5*3 homozygotes, with proportional dosing needs to maintain adequate immunosuppression. Tacrolimus 54-64 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 115-121 17106006-9 2006 CONCLUSION: For renal transplant recipients receiving tacrolimus as an immunosuppressant, practitioners can expect CYP3A5*1 carriers to have a tacrolimus clearance 25-45% greater than that of CYP3A5*3 homozygotes, with proportional dosing needs to maintain adequate immunosuppression. Tacrolimus 54-64 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 192-198 17106006-9 2006 CONCLUSION: For renal transplant recipients receiving tacrolimus as an immunosuppressant, practitioners can expect CYP3A5*1 carriers to have a tacrolimus clearance 25-45% greater than that of CYP3A5*3 homozygotes, with proportional dosing needs to maintain adequate immunosuppression. Tacrolimus 143-153 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 115-121 16982839-9 2006 In contrast, inhibitory effects on TLR-mediated chemokine expression of calcineurin blockade (by FK506) were greater for TLR2 than for TLR4 stimulation. Tacrolimus 97-102 toll like receptor 2 Homo sapiens 121-125 17359722-0 2006 [Effect of tacrolimus on apoptosis and expression of heat shock protein 70 after acute spinal cord injury in rats]. Tacrolimus 11-21 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 53-74 17359722-1 2006 OBJECTIVE: To investigate the effect of tacrolimus on expression of heat shock protein 70 (HSP 70) after spinal cord injuries (SCI) in rats and the relationship between expression of HSP 70 and apoptosis of neural cells. Tacrolimus 40-50 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 68-89 17359722-1 2006 OBJECTIVE: To investigate the effect of tacrolimus on expression of heat shock protein 70 (HSP 70) after spinal cord injuries (SCI) in rats and the relationship between expression of HSP 70 and apoptosis of neural cells. Tacrolimus 40-50 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 91-97 17359722-7 2006 RESULTS: Compared with the injury group, the expression of HSP 70 was significantly higher in the tacrolimus group, and the peak expression of HSP 70 mRNA and protein was respectively observed at 6, 24 h after SCI. Tacrolimus 98-108 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 59-65 17359722-8 2006 Caspase-3-positive or TUNEL-positive cells were significantly less in the tacrolimus group than in the injury group. Tacrolimus 74-84 caspase 3 Rattus norvegicus 0-9 17359722-10 2006 CONCLUSIONS: Tacrolimus may inhibit activity of Caspase-3, attenuate apoptosis of neural cells and ameliorate neurological function recovery after SCI by inducing high expression of HSP 70. Tacrolimus 13-23 caspase 3 Rattus norvegicus 48-57 17359722-10 2006 CONCLUSIONS: Tacrolimus may inhibit activity of Caspase-3, attenuate apoptosis of neural cells and ameliorate neurological function recovery after SCI by inducing high expression of HSP 70. Tacrolimus 13-23 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 182-188 16930148-7 2006 Upon dinucleotide-oligodeoxynucleotide (CpG-ODN) activation, tacrolimus pretreated PDCs showed a significant reduction in the surface expression of co-stimulatory molecules and human leucocyte antigen D-related (HLA-DR) and secreted reduced levels of tumour necrosis factor (TNF)-alpha. Tacrolimus 61-71 tumor necrosis factor Homo sapiens 251-285 16982839-9 2006 In contrast, inhibitory effects on TLR-mediated chemokine expression of calcineurin blockade (by FK506) were greater for TLR2 than for TLR4 stimulation. Tacrolimus 97-102 toll like receptor 4 Homo sapiens 135-139 17130675-1 2006 The aims of this study were to investigate the role of endothelin-1 in FK506-induced hypertension and vascular dysfunction of rats treated with the drug for 8 (short-term) or 30 (long-term) days and to measure malondialdehyde levels in the kidneys. Tacrolimus 71-76 endothelin 1 Rattus norvegicus 55-67 17023428-12 2006 Accordingly, induction by alkaline stress of the entire ENA1 promoter in a snf1 rim101 mutant in the presence of the calcineurin inhibitor FK506 is completely abolished. Tacrolimus 139-144 Na(+)/Li(+)-exporting P-type ATPase ENA1 Saccharomyces cerevisiae S288C 56-60 17023428-12 2006 Accordingly, induction by alkaline stress of the entire ENA1 promoter in a snf1 rim101 mutant in the presence of the calcineurin inhibitor FK506 is completely abolished. Tacrolimus 139-144 alkaline-responsive transcriptional regulator RIM101 Saccharomyces cerevisiae S288C 80-86 17149058-7 2006 Tacrolimus, cyclosporine, and interferon-alpha-2a should be used generally only if TNF inhibitors have failed as a result of their toxicities. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 83-86 17190370-1 2006 OBJECTIVE: Lansoprazole and tacrolimus are substrates of ATP binding cassette (ABC) transporters such as P-glycoprotein (ABCBI/multidrug resistance 1) and cytochrome P450 (CYP). Tacrolimus 28-38 ATP binding cassette subfamily B member 1 Homo sapiens 121-149 17190370-1 2006 OBJECTIVE: Lansoprazole and tacrolimus are substrates of ATP binding cassette (ABC) transporters such as P-glycoprotein (ABCBI/multidrug resistance 1) and cytochrome P450 (CYP). Tacrolimus 28-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 17190370-1 2006 OBJECTIVE: Lansoprazole and tacrolimus are substrates of ATP binding cassette (ABC) transporters such as P-glycoprotein (ABCBI/multidrug resistance 1) and cytochrome P450 (CYP). Tacrolimus 28-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-175 17190370-2 2006 The purpose of this study was to investigate the implication of the ABCB1 C3435Tpolymorphism on the pharmacokinetics of (R)-lansoprazole, the major enantiomer, in CYP2C19 extensive metabolizers (EMs) and on gastroesophageal symptoms in renal transplant recipients receiving tacrolimus. Tacrolimus 274-284 ATP binding cassette subfamily B member 1 Homo sapiens 68-73 17204907-8 2006 Inhibition of VSMC proliferation by both tacrolimus and sirolimus was associated with upregulation of the cell-cycle inhibitor p27. Tacrolimus 41-51 interferon alpha inducible protein 27 Homo sapiens 127-130 17130675-11 2006 Our results indicated that endothelin-1 plays a key role in the FK506-induced change in vascular reactivity to noradrenaline in renal vascular beds and drug-induced hypertension in the rats. Tacrolimus 64-69 endothelin 1 Rattus norvegicus 27-39 17067582-0 2006 Immunophilin-ligands FK506 and CsA inhibit HIF1alpha expression by a VHL- and ubiquitin-independent mechanism. Tacrolimus 21-26 hypoxia inducible factor 1 subunit alpha Homo sapiens 43-52 17175263-0 2006 Paradoxical response to tacrolimus assessed by interleukin-2 gene expression. Tacrolimus 24-34 interleukin 2 Homo sapiens 47-60 17175263-6 2006 RESULTS: IL-2 mRNA synthesis was suppressed by the presence of tacrolimus in most cases, compared with a control sample. Tacrolimus 63-73 interleukin 2 Homo sapiens 9-13 17175265-12 2006 Our discovery of IRI-induced up-regulation of genes associated with calcineurin and mTOR pathways are consistent with clinical observations that FK506 and Rapamycin can alter the course of DGF. Tacrolimus 145-150 mechanistic target of rapamycin kinase Homo sapiens 84-88 17011518-7 2006 We also found that the interaction was interfered, in a dose-dependent manner, by FK506, whose neuroprotective effect has been suggested to be correlated with its PPIase inhibitory activity. Tacrolimus 82-87 FKBP prolyl isomerase 5 Homo sapiens 163-169 17082615-2 2006 Previous studies have shown that the immunosuppressants cyclosporin A or FK506, which interact with the peptidyl-propyl isomerases cyclophilin A and FK506-binding protein (FKBP12), respectively, block cytokine expression. Tacrolimus 73-78 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 172-178 17067582-3 2006 We have evaluated the effect of the immunophilin ligands FK506 and cyclosporin A on HIF1alpha levels in different tumor cell lines. Tacrolimus 57-62 hypoxia inducible factor 1 subunit alpha Homo sapiens 84-93 17034537-8 2006 The increase of pigmentation was due to the result of the stimulatory action of FK506 on tyrosinase activity and its expression, which eventually led to melanin biosynthesis. Tacrolimus 80-85 tyrosinase Homo sapiens 89-99 17049058-1 2006 Genetic polymorphisms in biotransformation enzyme CYP3A5 (6986G > A, CYP3A5*3; 14690A > G, CYP3A5*6) and drug transporter ABCB1 (1236C > T; 2677G > T/A; 3435C > T) are known to influence tacrolimus (Tac) dose requirements and trough blood levels in stable transplant patients. Tacrolimus 202-212 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 50-56 17049058-1 2006 Genetic polymorphisms in biotransformation enzyme CYP3A5 (6986G > A, CYP3A5*3; 14690A > G, CYP3A5*6) and drug transporter ABCB1 (1236C > T; 2677G > T/A; 3435C > T) are known to influence tacrolimus (Tac) dose requirements and trough blood levels in stable transplant patients. Tacrolimus 202-212 ATP binding cassette subfamily B member 1 Homo sapiens 128-133 17040264-8 2006 Tacrolimus-treated specimens featured a significant reduction of the expression of ELAM-1, VCAM-1 and ICAM-1, while hydrocortisone-treated lesions did not. Tacrolimus 0-10 selectin E Homo sapiens 83-89 17040264-8 2006 Tacrolimus-treated specimens featured a significant reduction of the expression of ELAM-1, VCAM-1 and ICAM-1, while hydrocortisone-treated lesions did not. Tacrolimus 0-10 vascular cell adhesion molecule 1 Homo sapiens 91-97 17040264-8 2006 Tacrolimus-treated specimens featured a significant reduction of the expression of ELAM-1, VCAM-1 and ICAM-1, while hydrocortisone-treated lesions did not. Tacrolimus 0-10 intercellular adhesion molecule 1 Homo sapiens 102-108 17008059-11 2006 Immunosuppressive drugs such as dexamethasone (Dex), tacrolimus and cyclosporine (Cys) inhibited the CCL22 production by MoDCs in the AD patients. Tacrolimus 53-63 C-C motif chemokine ligand 22 Homo sapiens 101-106 17112846-0 2006 Tacrolimus dosing in Chinese renal transplant patients is related to MDR1 gene C3435T polymorphisms. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 69-73 17058250-8 2006 Using multiple linear regression models, the polynomial relationship between hematocrit and serum Epo values was similar to the control group in patients under tacrolimus, whereas Epo production was significantly reduced in patients under cyclosporine-based immunosuppression. Tacrolimus 160-170 erythropoietin Homo sapiens 98-101 17076660-8 2006 FK506 (1 muM) efficiently inhibits Glu-induced apoptosis of cultured astrocytes, DNA fragmentation and changes in mitochondrial DeltaPsi. Tacrolimus 0-5 latexin Homo sapiens 9-12 16877482-13 2006 CONCLUSION: Our results suggest that in patients with CAN and deteriorating allograft function, CsA-to-tacrolimus conversion or CsA minimization achieved comparable efficacies in retarding the decline of graft function. Tacrolimus 103-113 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 96-99 17112846-2 2006 P-glycoprotein (P-gp) plays an important role in the absorption metabolism of tacrolimus. Tacrolimus 78-88 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 17112846-2 2006 P-glycoprotein (P-gp) plays an important role in the absorption metabolism of tacrolimus. Tacrolimus 78-88 ATP binding cassette subfamily B member 1 Homo sapiens 16-20 17138058-6 2006 RESULTS: sCD30 and caspase-1 concentrations were non-significantly up-regulated in all analysed groups--with or without rejection signs or immunosuppressed with cyclosporine or especially tacrolimus. Tacrolimus 188-198 caspase 1 Homo sapiens 19-28 17112846-6 2006 The results showed a significant association between tacrolimus levels per dose mg/kg/d and MDR1 gene C3435T polymorphism (P < .05). Tacrolimus 53-63 ATP binding cassette subfamily B member 1 Homo sapiens 92-96 17138060-0 2006 TIRC7 is induced in rejected human kidneys and anti-TIRC7 mAb with FK506 prolongs survival of kidney allografts in rats. Tacrolimus 67-72 T cell immune regulator 1, ATPase H+ transporting V0 subunit a3 Homo sapiens 52-57 16996313-2 2006 The drugs form complexes with diverse FKBPs but apparently the FKBP52/FK506 complex was shown to be involved in the protection and regeneration of neurons. Tacrolimus 70-75 FKBP prolyl isomerase 4 Homo sapiens 63-69 17138060-5 2006 These results suggest a potential synergism of anti-TIRC7 mAb and FK506 action, which could be developed into a novel combination therapy in the clinic by lowering side effects of present CI treatment. Tacrolimus 66-71 T cell immune regulator 1, ATPase H+ transporting V0 subunit a3 Homo sapiens 52-57 16935431-10 2006 Tacrolimus also decreased caspase-3 and terminal deoxynucleotidyl transferase-mediated DNA nick-end labeling-positive cells, and reduced the size of infarct 24 h after reperfusion. Tacrolimus 0-10 caspase 3 Mus musculus 26-35 16985094-1 2006 OBJECTIVE: To review the literature on novel immunomodulators such as tumor necrosis factor alpha (TNF-alpha)- and interleukin (IL)-related agents, 6-thioguanine (6-TG), tacrolimus, and leflunomide, and antibiotics such as ornidazole, rifaximin, and ciprofloxacin for the treatment of Crohn"s disease. Tacrolimus 170-180 tumor necrosis factor Homo sapiens 99-108 17015051-10 2006 CONCLUSIONS: The enterocyte MDR1 mRNA level and the CYP3A5*1 allele in the graft liver contribute differently to the interindividual variability in the oral clearance of tacrolimus after living-donor liver transplantation. Tacrolimus 170-180 ATP binding cassette subfamily B member 1 Homo sapiens 28-32 17015051-10 2006 CONCLUSIONS: The enterocyte MDR1 mRNA level and the CYP3A5*1 allele in the graft liver contribute differently to the interindividual variability in the oral clearance of tacrolimus after living-donor liver transplantation. Tacrolimus 170-180 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 52-58 16844742-1 2006 The absolute (standard) binding free energy of eight FK506-related ligands to FKBP12 is calculated using free energy perturbation molecular dynamics (FEP/MD) simulations with explicit solvent. Tacrolimus 53-58 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 78-84 16996313-8 2006 It is proposed that binding of the FKBP52/FK506 complex to the membranes via the TPR motifs and its interaction with some membrane proteins could be in part responsible for some neuro-regeneration and neuro-protection of the brain during some ischaemia-induced stresses. Tacrolimus 42-47 FKBP prolyl isomerase 4 Homo sapiens 35-41 17166405-8 2006 IL-16 serum levels were significantly higher in patients affected by atopic dermatitis than in controls before and after treatment with tacrolimus ointment. Tacrolimus 136-146 interleukin 16 Homo sapiens 0-5 17098001-8 2006 Comparison of the treatment groups showed that the regimen associated with fever infections included an IL-2 receptor antagonist with tacrolimus, MMF, and S. Tacrolimus 134-144 interleukin 2 Homo sapiens 104-108 17039660-7 2006 Tacrolimus is an immunomodulatory macrolide that reduces the stimulatory capacity toward T cells and is therefore worth investigating as a treatment of CTCL. Tacrolimus 0-10 TSPY like 2 Homo sapiens 152-156 17039660-8 2006 Topical tacrolimus has been related to an unknown effect with the risk for secondary malignancies including CTCL. Tacrolimus 8-18 TSPY like 2 Homo sapiens 108-112 16880206-3 2006 Here, we report that Fyn, NFAT, and ERK signaling influence ICOS expression as various chemical inhibitors, such as PP2 that targets Src kinases, U0126 that targets MEK1/2, and cyclosporin A or FK506 that targets calcineurin and thereby affects NFAT, attenuate T cell receptor-mediated ICOS induction. Tacrolimus 194-199 Fyn proto-oncogene Mus musculus 21-24 16880206-3 2006 Here, we report that Fyn, NFAT, and ERK signaling influence ICOS expression as various chemical inhibitors, such as PP2 that targets Src kinases, U0126 that targets MEK1/2, and cyclosporin A or FK506 that targets calcineurin and thereby affects NFAT, attenuate T cell receptor-mediated ICOS induction. Tacrolimus 194-199 mitogen-activated protein kinase 1 Mus musculus 36-39 16880206-3 2006 Here, we report that Fyn, NFAT, and ERK signaling influence ICOS expression as various chemical inhibitors, such as PP2 that targets Src kinases, U0126 that targets MEK1/2, and cyclosporin A or FK506 that targets calcineurin and thereby affects NFAT, attenuate T cell receptor-mediated ICOS induction. Tacrolimus 194-199 inducible T cell co-stimulator Mus musculus 60-64 16880206-3 2006 Here, we report that Fyn, NFAT, and ERK signaling influence ICOS expression as various chemical inhibitors, such as PP2 that targets Src kinases, U0126 that targets MEK1/2, and cyclosporin A or FK506 that targets calcineurin and thereby affects NFAT, attenuate T cell receptor-mediated ICOS induction. Tacrolimus 194-199 neuropeptide Y receptor Y6 Mus musculus 116-119 16969296-0 2006 Multidrug resistance gene-1 (MDR-1) haplotypes have a minor influence on tacrolimus dose requirements. Tacrolimus 73-83 ATP binding cassette subfamily B member 1 Homo sapiens 29-34 16759707-5 2006 Both tacrolimus and cyclosporine are substrates of Pgp, CYP3A4 and CYP3A5, and therefore, these molecules are potential pharmacokinetic factors with which to establish personalized dosage regimens for these drugs. Tacrolimus 5-15 ATP binding cassette subfamily B member 1 Homo sapiens 51-54 16759707-5 2006 Both tacrolimus and cyclosporine are substrates of Pgp, CYP3A4 and CYP3A5, and therefore, these molecules are potential pharmacokinetic factors with which to establish personalized dosage regimens for these drugs. Tacrolimus 5-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 56-62 16759707-5 2006 Both tacrolimus and cyclosporine are substrates of Pgp, CYP3A4 and CYP3A5, and therefore, these molecules are potential pharmacokinetic factors with which to establish personalized dosage regimens for these drugs. Tacrolimus 5-15 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-73 16759707-7 2006 In living-donor liver transplant (LDLT) patients, the intestinal mRNA expression level of MDR1 and CYP3A5 genotyping both in the native intestine and in the grafted liver are suggested to be potential pharmacokinetic factors for adjusting initial dosage and predicting post-operative variation in the pharmacokinetics of tacrolimus. Tacrolimus 321-331 ATP binding cassette subfamily B member 1 Homo sapiens 90-94 16759707-7 2006 In living-donor liver transplant (LDLT) patients, the intestinal mRNA expression level of MDR1 and CYP3A5 genotyping both in the native intestine and in the grafted liver are suggested to be potential pharmacokinetic factors for adjusting initial dosage and predicting post-operative variation in the pharmacokinetics of tacrolimus. Tacrolimus 321-331 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 99-105 16914137-8 2006 Although tacrolimus reduced the increased expression of IFN-gamma and IL-4 mRNA, dexamethasone potently depressed that of IFN-gamma, IL-4 and IL-5 mRNA. Tacrolimus 9-19 interferon gamma Mus musculus 56-65 16914137-8 2006 Although tacrolimus reduced the increased expression of IFN-gamma and IL-4 mRNA, dexamethasone potently depressed that of IFN-gamma, IL-4 and IL-5 mRNA. Tacrolimus 9-19 interleukin 4 Mus musculus 70-74 17006323-8 2006 The conversion from CsA to tacrolimus resulted in a significant decrease in uric acid, total- and LDL-cholesterol, apolipoprotein B, creatinine, and homocysteine levels (all P<0.05). Tacrolimus 27-37 apolipoprotein B Homo sapiens 115-131 16906020-0 2006 Cyp3A4, Cyp3A5, and MDR-1 genetic influences on tacrolimus pharmacokinetics in renal transplant recipients. Tacrolimus 48-58 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 16619106-10 2006 Suppression of calcineurin activity by FK506 was associated with modest activation of ERK1/2. Tacrolimus 39-44 mitogen activated protein kinase 3 Rattus norvegicus 86-92 16404634-0 2006 Cyclosporin A, tacrolimus and sirolimus are potent inhibitors of the human breast cancer resistance protein (ABCG2) and reverse resistance to mitoxantrone and topotecan. Tacrolimus 15-25 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 75-107 16404634-0 2006 Cyclosporin A, tacrolimus and sirolimus are potent inhibitors of the human breast cancer resistance protein (ABCG2) and reverse resistance to mitoxantrone and topotecan. Tacrolimus 15-25 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 109-114 16404634-3 2006 Therefore, the aim of this study was to determine whether the immunosuppressants cyclosporin A, tacrolimus and sirolimus are inhibitors and/or substrates of BCRP. Tacrolimus 96-106 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 157-161 16404634-5 2006 RESULTS: Cyclosporin A, tacrolimus and sirolimus significantly inhibited BCRP-mediated efflux of pheophorbide A, mitoxantrone and BODIPY-prazosin. Tacrolimus 24-34 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 73-77 16990701-0 2006 Inhibition of arachidonic acid release by cytosolic phospholipase A2 is involved in the antiapoptotic effect of FK506 and cyclosporin a on astrocytes exposed to simulated ischemia in vitro. Tacrolimus 112-117 phospholipase A2 group IVA Rattus norvegicus 42-68 16990701-1 2006 In the present study, we investigated whether the protective effect of FK506 and cyclosporin A (CsA) against in vitro ischemic injury of astrocytes might be mediated through attenuation of cytosolic isoform of phospholipase A(2) (cPLA(2)) expression and activity as well as inhibition of arachidonic acid (AA) release. Tacrolimus 71-76 phospholipase A2 group IB Rattus norvegicus 210-228 16990701-1 2006 In the present study, we investigated whether the protective effect of FK506 and cyclosporin A (CsA) against in vitro ischemic injury of astrocytes might be mediated through attenuation of cytosolic isoform of phospholipase A(2) (cPLA(2)) expression and activity as well as inhibition of arachidonic acid (AA) release. Tacrolimus 71-76 phospholipase A2 group IVA Rattus norvegicus 230-237 16990701-3 2006 Obtained data suggest the cross-talk between the action of 0.25 - 10 microM CsA as well as 1 microM FK506 on calcineurin (CaN) and cPLA(2) in anti-apoptotic signal transduction pathways. Tacrolimus 100-105 phospholipase A2 group IVA Rattus norvegicus 131-138 16990701-6 2006 Our findings document a key role either for CaN or cPLA(2) expression attenuation and AA release inhibition in the antiapoptotic effect of FK506 and CsA in ischemic astrocytes. Tacrolimus 139-144 phospholipase A2 group IVA Rattus norvegicus 51-58 16969296-2 2006 Variability in tacrolimus absorption is influenced by P-gp activity which, in turn, is affected by single nucleotide polymorphisms (SNPs) within the multidrug resistance-1 gene (MDR-1). Tacrolimus 15-25 ATP binding cassette subfamily B member 1 Homo sapiens 54-58 16969296-2 2006 Variability in tacrolimus absorption is influenced by P-gp activity which, in turn, is affected by single nucleotide polymorphisms (SNPs) within the multidrug resistance-1 gene (MDR-1). Tacrolimus 15-25 ATP binding cassette subfamily B member 1 Homo sapiens 149-171 16969296-2 2006 Variability in tacrolimus absorption is influenced by P-gp activity which, in turn, is affected by single nucleotide polymorphisms (SNPs) within the multidrug resistance-1 gene (MDR-1). Tacrolimus 15-25 ATP binding cassette subfamily B member 1 Homo sapiens 178-183 16969296-3 2006 Tacrolimus dose requirements of 206 stable renal transplant patients were related to MDR-1 genotypes of SNPs C1236T, G2677T/A and C3435T, as well as haplotypes: C-G-C and T-T-T. Lower dose-normalized blood tacrolimus concentrations were achieved for: 2677-GG genotype patients, as compared to 2677-TT, and for 3435-CC patients as compared to 3435-TT patients. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 85-90 16969296-6 2006 Our data suggest that MDR-1 haplotypes have a relatively minor association with tacrolimus pharmacokinetics. Tacrolimus 80-90 ATP binding cassette subfamily B member 1 Homo sapiens 22-27 16404634-6 2006 The EC(50) values of cyclosporin A, tacrolimus and sirolimus for inhibition of BCRP-mediated pheophorbide A efflux were 4.3 +/- 1.9 microM, 3.6 +/- 1.8 microM and 1.9 +/- 0.4 microM, respectively. Tacrolimus 36-46 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 79-83 16404634-7 2006 Cyclosporin A, tacrolimus and sirolimus also effectively reversed resistance of HEK cells to topotecan and mitoxantrone conferred by BCRP. Tacrolimus 15-25 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 133-137 16911492-0 2006 Tacrolimus immunosuppression - an association with asymptomatic eosinophilia and elevated total and specific IgE levels. Tacrolimus 0-10 immunoglobulin heavy constant epsilon Homo sapiens 109-112 16911492-2 2006 An increased prevalence of food allergy noted specifically in children receiving tacrolimus immunosuppression supports the hypothesis that selective suppression of Th1 lymphocytes by the IL-2 inhibitor immunosuppressants CsA, and the more potent drug, tacrolimus , promotes Th2 lymphocytes and an allergic immune response. Tacrolimus 81-91 negative elongation factor complex member C/D Homo sapiens 164-167 16911492-2 2006 An increased prevalence of food allergy noted specifically in children receiving tacrolimus immunosuppression supports the hypothesis that selective suppression of Th1 lymphocytes by the IL-2 inhibitor immunosuppressants CsA, and the more potent drug, tacrolimus , promotes Th2 lymphocytes and an allergic immune response. Tacrolimus 81-91 interleukin 2 Homo sapiens 187-191 16911492-2 2006 An increased prevalence of food allergy noted specifically in children receiving tacrolimus immunosuppression supports the hypothesis that selective suppression of Th1 lymphocytes by the IL-2 inhibitor immunosuppressants CsA, and the more potent drug, tacrolimus , promotes Th2 lymphocytes and an allergic immune response. Tacrolimus 252-262 negative elongation factor complex member C/D Homo sapiens 164-167 16911492-2 2006 An increased prevalence of food allergy noted specifically in children receiving tacrolimus immunosuppression supports the hypothesis that selective suppression of Th1 lymphocytes by the IL-2 inhibitor immunosuppressants CsA, and the more potent drug, tacrolimus , promotes Th2 lymphocytes and an allergic immune response. Tacrolimus 252-262 interleukin 2 Homo sapiens 187-191 16911492-3 2006 This study was undertaken to characterize the IgE-mediated immune response, in CsA and tacrolimus-treated, post-OLT children. Tacrolimus 87-97 immunoglobulin heavy constant epsilon Homo sapiens 46-49 16906020-0 2006 Cyp3A4, Cyp3A5, and MDR-1 genetic influences on tacrolimus pharmacokinetics in renal transplant recipients. Tacrolimus 48-58 ATP binding cassette subfamily B member 1 Homo sapiens 20-25 16906020-2 2006 Cytochrome P450 3A4 (Cyp3A4) and Cyp3A5 are the most important contributors to tacrolimus metabolism while the P-glycoprotein pump (MDR-1) modulates its bioavailability. Tacrolimus 79-89 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-19 16906020-2 2006 Cytochrome P450 3A4 (Cyp3A4) and Cyp3A5 are the most important contributors to tacrolimus metabolism while the P-glycoprotein pump (MDR-1) modulates its bioavailability. Tacrolimus 79-89 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-27 16906020-2 2006 Cytochrome P450 3A4 (Cyp3A4) and Cyp3A5 are the most important contributors to tacrolimus metabolism while the P-glycoprotein pump (MDR-1) modulates its bioavailability. Tacrolimus 79-89 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 16906020-2 2006 Cytochrome P450 3A4 (Cyp3A4) and Cyp3A5 are the most important contributors to tacrolimus metabolism while the P-glycoprotein pump (MDR-1) modulates its bioavailability. Tacrolimus 79-89 ATP binding cassette subfamily B member 1 Homo sapiens 132-137 16906020-3 2006 The objective was to investigate the association between Cyp3A4, Cyp3A5, and MDR-1 polymorphisms and tacrolimus pharmacokinetics in the early period after renal transplantation. Tacrolimus 101-111 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 57-63 16906020-3 2006 The objective was to investigate the association between Cyp3A4, Cyp3A5, and MDR-1 polymorphisms and tacrolimus pharmacokinetics in the early period after renal transplantation. Tacrolimus 101-111 ATP binding cassette subfamily B member 1 Homo sapiens 77-82 16906020-7 2006 Patients who do not carry both Cyp3A5*3 alleles achieved lower mean dose-adjusted tacrolimus blood concentrations (p<0.001) and needed a longer time to reach the target concentration (10-12 ng/ml; p<0.001) compared to Cyp3A5*3 homozygotes. Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 31-37 16906020-8 2006 Patients with less than three copies of MDR-1 (T-129C, C3435T and G2677T) polymorphisms, associated with reduced expression of P-glycoprotein, had also lower dose-adjusted tacrolimus blood concentrations compared to patients having equal to or greater than three copies of MDR-1 genetic variants (P=0.003). Tacrolimus 172-182 ATP binding cassette subfamily B member 1 Homo sapiens 40-45 16906020-8 2006 Patients with less than three copies of MDR-1 (T-129C, C3435T and G2677T) polymorphisms, associated with reduced expression of P-glycoprotein, had also lower dose-adjusted tacrolimus blood concentrations compared to patients having equal to or greater than three copies of MDR-1 genetic variants (P=0.003). Tacrolimus 172-182 ATP binding cassette subfamily B member 1 Homo sapiens 127-141 16906020-8 2006 Patients with less than three copies of MDR-1 (T-129C, C3435T and G2677T) polymorphisms, associated with reduced expression of P-glycoprotein, had also lower dose-adjusted tacrolimus blood concentrations compared to patients having equal to or greater than three copies of MDR-1 genetic variants (P=0.003). Tacrolimus 172-182 ATP binding cassette subfamily B member 1 Homo sapiens 273-278 16906020-10 2006 CONCLUSION: The complete absence of Cyp3A5*3 allele and the accumulation of less than three copies of MDR-1 (T-129C, C3435T and G2677T) polymorphisms are associated with lower tacrolimus blood levels identifying these genotypes as markers for patients requiring higher tacrolimus doses. Tacrolimus 176-186 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 16906020-10 2006 CONCLUSION: The complete absence of Cyp3A5*3 allele and the accumulation of less than three copies of MDR-1 (T-129C, C3435T and G2677T) polymorphisms are associated with lower tacrolimus blood levels identifying these genotypes as markers for patients requiring higher tacrolimus doses. Tacrolimus 176-186 ATP binding cassette subfamily B member 1 Homo sapiens 102-107 16906020-10 2006 CONCLUSION: The complete absence of Cyp3A5*3 allele and the accumulation of less than three copies of MDR-1 (T-129C, C3435T and G2677T) polymorphisms are associated with lower tacrolimus blood levels identifying these genotypes as markers for patients requiring higher tacrolimus doses. Tacrolimus 269-279 ATP binding cassette subfamily B member 1 Homo sapiens 102-107 16892151-0 2006 [Effect of topical tacrolimus ointment on expression of Toll-like receptors 2 and 4 in lesional atopic dermatitis skin]. Tacrolimus 19-29 toll like receptor 2 Homo sapiens 56-83 16979989-0 2006 Tacrolimus (Pan Graf) as de novo therapy in renal transplant recipients in India. Tacrolimus 0-10 Rho GTPase activating protein 26 Homo sapiens 16-20 16980029-14 2006 RESULTS: CsA and FK506 stimulated gene expression and protein production of TGF-beta1, smad2, and smad3, but inhibited expression of smad7 both in VSMC and in the transplanted kidney. Tacrolimus 17-22 transforming growth factor, beta 1 Rattus norvegicus 76-85 16980029-14 2006 RESULTS: CsA and FK506 stimulated gene expression and protein production of TGF-beta1, smad2, and smad3, but inhibited expression of smad7 both in VSMC and in the transplanted kidney. Tacrolimus 17-22 SMAD family member 2 Rattus norvegicus 87-92 16980029-14 2006 RESULTS: CsA and FK506 stimulated gene expression and protein production of TGF-beta1, smad2, and smad3, but inhibited expression of smad7 both in VSMC and in the transplanted kidney. Tacrolimus 17-22 SMAD family member 3 Rattus norvegicus 98-103 16980029-14 2006 RESULTS: CsA and FK506 stimulated gene expression and protein production of TGF-beta1, smad2, and smad3, but inhibited expression of smad7 both in VSMC and in the transplanted kidney. Tacrolimus 17-22 SMAD family member 7 Rattus norvegicus 133-138 16980029-19 2006 CsA and FK506 can cause CAN, owing to up-regulated expression of smad2 and smad3, and down-regulation of smad7 expression. Tacrolimus 8-13 SMAD family member 2 Rattus norvegicus 65-70 16980029-19 2006 CsA and FK506 can cause CAN, owing to up-regulated expression of smad2 and smad3, and down-regulation of smad7 expression. Tacrolimus 8-13 SMAD family member 3 Rattus norvegicus 75-80 16980029-19 2006 CsA and FK506 can cause CAN, owing to up-regulated expression of smad2 and smad3, and down-regulation of smad7 expression. Tacrolimus 8-13 SMAD family member 7 Rattus norvegicus 105-110 16980036-0 2006 Comparison of expression of TGF-beta1, its receptors TGFbeta1R-I and TGFbeta1R-II in rat kidneys during chronic nephropathy induced by cyclosporine and tacrolimus. Tacrolimus 152-162 transforming growth factor, beta 1 Rattus norvegicus 28-37 16980036-8 2006 The results of immunohistochemistry and in situ hybridization showed that the expression of renal TGFbeta1, TR I, TR-II proteins and TR and TR II mRNA in the FK506 group were lower than those in the CsA groups. Tacrolimus 158-163 transforming growth factor, beta 1 Rattus norvegicus 98-106 16892151-1 2006 OBJECTIVE: To investigate the role of Toll-like receptor(TLR) 2 and TLR4 in pathogenesis of atopic dermatitis(AD) and the effect of topical tacrolimus ointment on expression of TLR2 and TLR4 in lesional AD skin. Tacrolimus 140-150 toll like receptor 2 Homo sapiens 177-181 16892151-1 2006 OBJECTIVE: To investigate the role of Toll-like receptor(TLR) 2 and TLR4 in pathogenesis of atopic dermatitis(AD) and the effect of topical tacrolimus ointment on expression of TLR2 and TLR4 in lesional AD skin. Tacrolimus 140-150 toll like receptor 4 Homo sapiens 186-190 16892151-5 2006 After using topical tacrolimus ointment for three weeks, TLR2 and TLR4 were expressed on basal and suprabasal keratinocytes with membranous and cytoplasmic staining pattern. Tacrolimus 20-30 toll like receptor 2 Homo sapiens 57-61 16892151-5 2006 After using topical tacrolimus ointment for three weeks, TLR2 and TLR4 were expressed on basal and suprabasal keratinocytes with membranous and cytoplasmic staining pattern. Tacrolimus 20-30 toll like receptor 4 Homo sapiens 66-70 16892151-7 2006 Topical tacrolimus may directly or indirectly inhibit or down-regulate TLR2 and TLR4 expression in KC and inhibit skin innate immuno-inflammatory response related to TLR-NFkappaB signal transduction and regulation in atopic dermatitis. Tacrolimus 8-18 toll like receptor 2 Homo sapiens 71-75 16892151-7 2006 Topical tacrolimus may directly or indirectly inhibit or down-regulate TLR2 and TLR4 expression in KC and inhibit skin innate immuno-inflammatory response related to TLR-NFkappaB signal transduction and regulation in atopic dermatitis. Tacrolimus 8-18 toll like receptor 4 Homo sapiens 80-84 16868484-9 2006 Whereas IFN-gamma and FK506 treatment lead to an enhanced intrapancreatic and systemic TNF-alpha protein release during the early course, IL-1beta concentrations were significantly reduced within the late intervals. Tacrolimus 22-27 tumor necrosis factor Rattus norvegicus 87-96 16871590-4 2006 Immunosuppressive drugs used were tacrolimus (a calcineurin inhibitor) and its synergistic partners, rapamycin (a regulator of the mammalian target of rapamycin [mTOR]) and mycophenolate mofetil (an inosine monophosphate dehydrogenase inhibitor). Tacrolimus 34-44 calcineurin binding protein 1 Homo sapiens 48-69 16911928-6 2006 RESULTS: The SNPs of CYP3A and P-gp are closely correlated to the large variations of cyclosporine and tacrolimus dosage between different patients, although conflicting results were obtained by some authors. Tacrolimus 103-113 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-26 16911928-6 2006 RESULTS: The SNPs of CYP3A and P-gp are closely correlated to the large variations of cyclosporine and tacrolimus dosage between different patients, although conflicting results were obtained by some authors. Tacrolimus 103-113 ATP binding cassette subfamily B member 1 Homo sapiens 31-35 16806841-8 2006 Tacrolimus also completely blocked the sensitization-associated increase of CD11c(+) antigen presenting cells (APC) in LN, whereas pimecrolimus showed significantly less inhibition. Tacrolimus 0-10 integrin subunit alpha X Homo sapiens 76-81 16860713-14 2006 In the same conditions, Tacrolimus significantly inhibited cell migration induced by either bradykinin, histamine or substance P (P < 0.05). Tacrolimus 24-34 tachykinin 1 Mus musculus 117-128 16842519-8 2006 The recovered T1/2 ratio (T1/2 ratio=T1/2 1-10 d/T1/2 30-40 d), this ratio being related to the recovery of hepatic function with ability of the graft to eliminate tacrolimus, showed great variability between patients, with values ranging from 1.08 to 4.21 (mean+/-SD, 1.95+/-0.81). Tacrolimus 164-174 interleukin 1 receptor like 1 Homo sapiens 49-61 16860713-2 2006 OBJECTIVES: This study evaluated whether the acute and systemic administration of Tacrolimus significantly interfered in leukocyte migration, exudation, myeloperoxidase and adenosine-deaminase and nitric oxide levels, as well as Interleukin-1 (IL-1beta) and tumor necrosis factor alpha (TNFalpha) levels in a mouse model of pleurisy in comparison to those obtained with dexamethasone. Tacrolimus 82-92 myeloperoxidase Mus musculus 153-168 16860713-2 2006 OBJECTIVES: This study evaluated whether the acute and systemic administration of Tacrolimus significantly interfered in leukocyte migration, exudation, myeloperoxidase and adenosine-deaminase and nitric oxide levels, as well as Interleukin-1 (IL-1beta) and tumor necrosis factor alpha (TNFalpha) levels in a mouse model of pleurisy in comparison to those obtained with dexamethasone. Tacrolimus 82-92 adenosine deaminase Mus musculus 173-192 16860713-2 2006 OBJECTIVES: This study evaluated whether the acute and systemic administration of Tacrolimus significantly interfered in leukocyte migration, exudation, myeloperoxidase and adenosine-deaminase and nitric oxide levels, as well as Interleukin-1 (IL-1beta) and tumor necrosis factor alpha (TNFalpha) levels in a mouse model of pleurisy in comparison to those obtained with dexamethasone. Tacrolimus 82-92 interleukin 1 beta Mus musculus 244-252 16860713-2 2006 OBJECTIVES: This study evaluated whether the acute and systemic administration of Tacrolimus significantly interfered in leukocyte migration, exudation, myeloperoxidase and adenosine-deaminase and nitric oxide levels, as well as Interleukin-1 (IL-1beta) and tumor necrosis factor alpha (TNFalpha) levels in a mouse model of pleurisy in comparison to those obtained with dexamethasone. Tacrolimus 82-92 tumor necrosis factor Mus musculus 258-285 16860713-2 2006 OBJECTIVES: This study evaluated whether the acute and systemic administration of Tacrolimus significantly interfered in leukocyte migration, exudation, myeloperoxidase and adenosine-deaminase and nitric oxide levels, as well as Interleukin-1 (IL-1beta) and tumor necrosis factor alpha (TNFalpha) levels in a mouse model of pleurisy in comparison to those obtained with dexamethasone. Tacrolimus 82-92 tumor necrosis factor Mus musculus 287-295 16542695-6 2006 Blocking NFAT activity using a number of reagents, such as Cyclosporin A, FK-506, or the NFAT-specific inhibitor known as VIVIT peptide, all markedly reduced IL-7-mediated induction of HIV replication in naive T cells. Tacrolimus 74-80 interleukin 7 Homo sapiens 158-162 17051976-9 2006 IFN-gamma production is inhibited by IL-4, IL-10, TGFbeta, glucocorticoids, cyclosporin A and FK506. Tacrolimus 94-99 interferon gamma Homo sapiens 0-9 16650863-8 2006 CONCLUSIONS: In our model it appears that a subtherapeutic dose of FK506 enhanced the tolerizing effect of NKT cells induced by oral tolerance, prolonging allograft survival by generating CD25+CD4+ CTLA4 T cells. Tacrolimus 67-72 interleukin 2 receptor, alpha chain Mus musculus 188-192 16786171-7 2006 These results indicate that two types of FK506-induced Ca2+ release mechanism operate in the endoplasmic reticulum of rat pancreatic acinar cells: a high-affinity mechanism of Ca2+ release, which involves activation of the ryanodine receptor, and a low-affinity mechanism of Ca2+ release, which involves activation of the IP3 receptor. Tacrolimus 41-46 inositol 1,4,5-trisphosphate receptor, type 3 Rattus norvegicus 322-334 16773453-5 2006 Tacrolimus treatment achieved a significant reduction of CD25+ cells. Tacrolimus 0-10 interleukin 2 receptor subunit alpha Homo sapiens 57-61 16773453-9 2006 Tacrolimus enhanced the production of TGF-beta, while hydrocortisone did not. Tacrolimus 0-10 transforming growth factor beta 1 Homo sapiens 38-46 16773453-10 2006 Restoration of TGF-beta-producing Treg cells may represent another important pharmacodynamic effect of tacrolimus on AD. Tacrolimus 103-113 transforming growth factor beta 1 Homo sapiens 15-23 16641133-3 2006 Here, we show that calcineurin antagonists such as cyclosporin A (CsA) or tacrolimus can markedly enhance the production of interferon-gamma (IFN-gamma) by human T cells. Tacrolimus 74-84 interferon gamma Homo sapiens 124-140 16641133-3 2006 Here, we show that calcineurin antagonists such as cyclosporin A (CsA) or tacrolimus can markedly enhance the production of interferon-gamma (IFN-gamma) by human T cells. Tacrolimus 74-84 interferon gamma Homo sapiens 142-151 16650863-8 2006 CONCLUSIONS: In our model it appears that a subtherapeutic dose of FK506 enhanced the tolerizing effect of NKT cells induced by oral tolerance, prolonging allograft survival by generating CD25+CD4+ CTLA4 T cells. Tacrolimus 67-72 cytotoxic T-lymphocyte-associated protein 4 Mus musculus 198-203 16569741-2 2006 OBJECTIVE: Our objective was to evaluate insulin sensitivity after islet transplantation performed under tacrolimus-based immunosuppression that used minimal steroids. Tacrolimus 105-115 insulin Homo sapiens 41-48 16792133-4 2006 Five patients with steroid-dependent minimal-change nephrotic syndrome, 1 patient with steroid-refractory minimal-change disease and 4 patients with steroid-refractory FSGS were started on tacrolimus at trough levels of 5 10 microg/l. Tacrolimus 189-199 actinin alpha 4 Homo sapiens 168-172 16689860-5 2006 Short-term topical treatment of the skin lesions with tacrolimus or pimecrolimus abolished the expression of cleaved caspase-3 in the spinous layer. Tacrolimus 54-64 caspase 3 Homo sapiens 117-126 16955241-1 2006 OBJECTIVE: To investigate whether FK506 (tacrolimus) can inhibit Fas- or A23187-induced interleukin (IL)-8 expression and cell death in A549 human alveolar epithelial cells, plus Fas-mediated acute lung injury in vivo. Tacrolimus 34-39 C-X-C motif chemokine ligand 8 Homo sapiens 88-106 16955241-1 2006 OBJECTIVE: To investigate whether FK506 (tacrolimus) can inhibit Fas- or A23187-induced interleukin (IL)-8 expression and cell death in A549 human alveolar epithelial cells, plus Fas-mediated acute lung injury in vivo. Tacrolimus 41-51 C-X-C motif chemokine ligand 8 Homo sapiens 88-106 16955241-9 2006 Tacrolimus inhibited A23187-induced IL-8 expression alone while it protected all Fas-mediated responses. Tacrolimus 0-10 chemokine (C-X-C motif) ligand 15 Mus musculus 36-40 16470176-0 2006 FK506 controls CD40L-induced systemic autoimmunity in mice. Tacrolimus 0-5 CD40 ligand Mus musculus 15-20 16470176-4 2006 Interestingly, FK506-treated CD40L tg mice showed significantly reduced autoimmune dermatitis scores and markedly decreased numbers of lesional infiltrating leukocytes. Tacrolimus 15-20 CD40 ligand Mus musculus 29-34 16470176-6 2006 Furthermore, FK506 suppressed the development of cytotoxic/autoreactive CD8(+) T cells as evidenced by the reduced expression of T cell activation markers in treated CD40L tg mice. Tacrolimus 13-18 CD40 ligand Mus musculus 166-171 16470176-7 2006 Importantly, FK506 induced a significant reduction in autoantibody titers in the serum of CD40L tg animals. Tacrolimus 13-18 CD40 ligand Mus musculus 90-95 16470176-8 2006 As CD40L tg mice develop nephritis leading to loss of renal function proteinuria was determined after FK506 injections. Tacrolimus 102-107 CD40 ligand Mus musculus 3-8 16470176-9 2006 Notably, FK506 treatment re-established renal function as indicated by significantly reduced uric protein concentrations of treated CD40L tg mice. Tacrolimus 9-14 CD40 ligand Mus musculus 132-137 16470176-10 2006 Together, these findings show the beneficial therapeutic effects of FK506 for the treatment of CD40L-induced autoimmunity. Tacrolimus 68-73 CD40 ligand Mus musculus 95-100 16720724-7 2006 FK506, a chemical ligand for FKBP12, which dissociates FKBP12 from the receptor, decreased the interaction between Smad7 and Smad ubiquitin regulatory factor 1 (Smurf1). Tacrolimus 0-5 FKBP prolyl isomerase 1A Homo sapiens 29-35 16720724-7 2006 FK506, a chemical ligand for FKBP12, which dissociates FKBP12 from the receptor, decreased the interaction between Smad7 and Smad ubiquitin regulatory factor 1 (Smurf1). Tacrolimus 0-5 FKBP prolyl isomerase 1A Homo sapiens 55-61 16720724-7 2006 FK506, a chemical ligand for FKBP12, which dissociates FKBP12 from the receptor, decreased the interaction between Smad7 and Smad ubiquitin regulatory factor 1 (Smurf1). Tacrolimus 0-5 SMAD family member 7 Homo sapiens 115-120 16720724-7 2006 FK506, a chemical ligand for FKBP12, which dissociates FKBP12 from the receptor, decreased the interaction between Smad7 and Smad ubiquitin regulatory factor 1 (Smurf1). Tacrolimus 0-5 SMAD specific E3 ubiquitin protein ligase 1 Homo sapiens 125-159 16720724-7 2006 FK506, a chemical ligand for FKBP12, which dissociates FKBP12 from the receptor, decreased the interaction between Smad7 and Smad ubiquitin regulatory factor 1 (Smurf1). Tacrolimus 0-5 SMAD specific E3 ubiquitin protein ligase 1 Homo sapiens 161-167 16720724-8 2006 FK506 also inhibited the ubiquitination of the type I receptor by Smurf1. Tacrolimus 0-5 SMAD specific E3 ubiquitin protein ligase 1 Homo sapiens 66-72 16859130-7 2006 FK506 obviously decreased LDH levels in the supernatant (P < 0.05) and attenuated IL-6 induced suppression of albumin synthesis (P < 0.01). Tacrolimus 0-5 interleukin 6 Homo sapiens 85-89 16574167-7 2006 Decreases in CyclinB2 and BIRC5 mRNA induced by FK506 or retinoic acid, both of which exert potentiation of NGF-induced neurite outgrowth effects but with different mechanisms, also correlated with their neurite outgrowth activities. Tacrolimus 48-53 cyclin B2 Homo sapiens 13-21 16574167-7 2006 Decreases in CyclinB2 and BIRC5 mRNA induced by FK506 or retinoic acid, both of which exert potentiation of NGF-induced neurite outgrowth effects but with different mechanisms, also correlated with their neurite outgrowth activities. Tacrolimus 48-53 baculoviral IAP repeat containing 5 Homo sapiens 26-31 16753004-0 2006 Influence of different allelic variants of the CYP3A and ABCB1 genes on the tacrolimus pharmacokinetic profile of Chinese renal transplant recipients. Tacrolimus 76-86 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-52 16753004-0 2006 Influence of different allelic variants of the CYP3A and ABCB1 genes on the tacrolimus pharmacokinetic profile of Chinese renal transplant recipients. Tacrolimus 76-86 ATP binding cassette subfamily B member 1 Homo sapiens 57-62 16753004-2 2006 The cytochrome P450 3A (CYP3A) and the ATP-binding cassette B1 (ABCB1) genes play an important role in the tacrolimus disposition. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 4-22 16753004-2 2006 The cytochrome P450 3A (CYP3A) and the ATP-binding cassette B1 (ABCB1) genes play an important role in the tacrolimus disposition. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-29 16753004-2 2006 The cytochrome P450 3A (CYP3A) and the ATP-binding cassette B1 (ABCB1) genes play an important role in the tacrolimus disposition. Tacrolimus 107-117 ATP binding cassette subfamily B member 1 Homo sapiens 39-62 16753004-2 2006 The cytochrome P450 3A (CYP3A) and the ATP-binding cassette B1 (ABCB1) genes play an important role in the tacrolimus disposition. Tacrolimus 107-117 ATP binding cassette subfamily B member 1 Homo sapiens 64-69 16753004-6 2006 Multiple regression analysis showed that the CYP3A5*3 polymorphism is the most significant independent variable and explained 35% of the dose requirement variability in relation to tacrolimus use. Tacrolimus 181-191 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 16753004-8 2006 In conclusion, the CYP3A5*3 polymorphism may be an important factor in determining the dose requirement for tacrolimus and genotyping can help determine the initial daily dose required by individual patients for adequate immunosuppression. Tacrolimus 108-118 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 19-25 16797284-0 2006 The effect of MDR1 (ABCB1) polymorphism on the pharmacokinetic of tacrolimus in Turkish renal transplant recipients. Tacrolimus 66-76 ATP binding cassette subfamily B member 1 Homo sapiens 14-18 16797284-0 2006 The effect of MDR1 (ABCB1) polymorphism on the pharmacokinetic of tacrolimus in Turkish renal transplant recipients. Tacrolimus 66-76 ATP binding cassette subfamily B member 1 Homo sapiens 20-25 16859130-10 2006 FK506 protected against the suppression of hepatic albumin synthesis caused by IL-6, suggesting its potential role in improving hypoalbuminaemia in immune glomerulonephritis. Tacrolimus 0-5 interleukin 6 Homo sapiens 79-83 16352746-6 2006 On the other hand, gene silencing of FKBP52 or administration of the FKBP52 blocker FK-506 enhances Ca(2+) influx through TRPV5. Tacrolimus 84-90 FKBP prolyl isomerase 4 Homo sapiens 69-75 16547004-1 2006 FK506 and FK506-derived inhibitors of the FK506-binding protein (FKBP)-type peptidylprolyl cis/trans-isomerases (PPIase) display potent neuroprotective and neuroregenerative properties in various neurodegeneration models, showing the importance of neuroimmunophilins as targets for the treatment of acute and chronic neurodegenerative diseases. Tacrolimus 0-5 FKBP prolyl isomerase family member 6 Rattus norvegicus 113-119 16547004-1 2006 FK506 and FK506-derived inhibitors of the FK506-binding protein (FKBP)-type peptidylprolyl cis/trans-isomerases (PPIase) display potent neuroprotective and neuroregenerative properties in various neurodegeneration models, showing the importance of neuroimmunophilins as targets for the treatment of acute and chronic neurodegenerative diseases. Tacrolimus 10-15 FKBP prolyl isomerase family member 6 Rattus norvegicus 113-119 16547004-1 2006 FK506 and FK506-derived inhibitors of the FK506-binding protein (FKBP)-type peptidylprolyl cis/trans-isomerases (PPIase) display potent neuroprotective and neuroregenerative properties in various neurodegeneration models, showing the importance of neuroimmunophilins as targets for the treatment of acute and chronic neurodegenerative diseases. Tacrolimus 10-15 FKBP prolyl isomerase family member 6 Rattus norvegicus 113-119 16926769-5 2006 There is conflicting evidence regarding the effect of specific immunosuppressant medications (eg, tacrolimus vs cyclosporine) on the risk of recurrent PBC. Tacrolimus 98-108 dihydrolipoamide S-acetyltransferase Homo sapiens 151-154 16352746-6 2006 On the other hand, gene silencing of FKBP52 or administration of the FKBP52 blocker FK-506 enhances Ca(2+) influx through TRPV5. Tacrolimus 84-90 transient receptor potential cation channel subfamily V member 5 Homo sapiens 122-127 29539165-4 2006 The aims of this study were to investigate transforming growth factor-beta1 (TGF-beta1) gene polymorphisms in renal transplant recipients treated with CsA or tacrolimus and to establish an association between these polymorphisms and TGF-beta1 plasma concentration and the incidence of GO. Tacrolimus 158-168 transforming growth factor beta 1 Homo sapiens 43-75 16501005-1 2006 Previous investigations of solid organ transplant patients treated with tacrolimus showed that individuals carrying a CYP3A5*1 allele have lower dose-adjusted trough blood concentrations compared with homozygous CYP3A5*3 individuals. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 212-218 16501005-2 2006 The objective of this investigation was to quantify the contribution of CYP3A5 to the hepatic and renal metabolic clearance of tacrolimus. Tacrolimus 127-137 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 72-78 16501005-9 2006 In vitro to in vivo scaling using both liver microsomes and recombinant enzymes yielded higher predicted in vivo tacrolimus clearances for patients with a CYP3A5*1/*3 genotype compared with those with a CYP3A5*3/*3 genotype. Tacrolimus 113-123 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 155-161 16501005-9 2006 In vitro to in vivo scaling using both liver microsomes and recombinant enzymes yielded higher predicted in vivo tacrolimus clearances for patients with a CYP3A5*1/*3 genotype compared with those with a CYP3A5*3/*3 genotype. Tacrolimus 113-123 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 203-209 16501005-11 2006 These data suggest that CYP3A5 contributes significantly to the metabolic clearance of tacrolimus in the liver and kidney. Tacrolimus 87-97 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-30 16671880-4 2006 The aims of this study were to investigate transforming growth factor-beta1 (TGF-beta1) gene polymorphisms in renal transplant recipients treated with CsA or tacrolimus and to establish an association between these polymorphisms and TGF-beta1 plasma concentration and the incidence of GO. Tacrolimus 158-168 transforming growth factor beta 1 Homo sapiens 43-75 16671880-4 2006 The aims of this study were to investigate transforming growth factor-beta1 (TGF-beta1) gene polymorphisms in renal transplant recipients treated with CsA or tacrolimus and to establish an association between these polymorphisms and TGF-beta1 plasma concentration and the incidence of GO. Tacrolimus 158-168 transforming growth factor beta 1 Homo sapiens 77-86 16671880-8 2006 TGF-beta1 plasma levels were estimated in 60 CsA- and 30 tacrolimus-treated patients. Tacrolimus 57-67 transforming growth factor beta 1 Homo sapiens 0-9 16501005-0 2006 Effect of CYP3A5 polymorphism on tacrolimus metabolic clearance in vitro. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 16501005-1 2006 Previous investigations of solid organ transplant patients treated with tacrolimus showed that individuals carrying a CYP3A5*1 allele have lower dose-adjusted trough blood concentrations compared with homozygous CYP3A5*3 individuals. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 118-124 16457995-1 2006 The significance of intestinal P-glycoprotein (P-gp) in determining the oral bioavailability of tacrolimus has been still controversial. Tacrolimus 96-106 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 31-45 16457995-1 2006 The significance of intestinal P-glycoprotein (P-gp) in determining the oral bioavailability of tacrolimus has been still controversial. Tacrolimus 96-106 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 47-51 16457995-2 2006 In this study, we reevaluated the interaction of tacrolimus with P-gp in the rat small intestine, by evaluating its absorption from the rat small intestine and its modulating effect on the absorption of known P-gp substrates (digoxin, methylprednisolone, and vinblastine). Tacrolimus 49-59 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 65-69 16457995-2 2006 In this study, we reevaluated the interaction of tacrolimus with P-gp in the rat small intestine, by evaluating its absorption from the rat small intestine and its modulating effect on the absorption of known P-gp substrates (digoxin, methylprednisolone, and vinblastine). Tacrolimus 49-59 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 209-213 16546704-5 2006 CsA and FK506 each dose-dependently inhibited antigen-induced histamine and beta-hexosaminidase secretion and the membrane repolarization phase, with similar IC50s for both actions, approximately 20 nM for CsA and approximately 2 nM for FK506. Tacrolimus 8-13 O-GlcNAcase Rattus norvegicus 76-95 16830109-7 2006 However, FK506 decreased LDH levels in the supernatant of cells (P < 0.05) and prevented the IL-6-induced suppression of albumin synthesis (P < 0.01) in a dose dependent manner. Tacrolimus 9-14 interleukin 6 Homo sapiens 96-100 16830109-11 2006 FK506 but not CSA protects against the suppression of hepatic albumin synthesis caused by IL-6. Tacrolimus 0-5 interleukin 6 Homo sapiens 90-94 16648664-2 2006 cADPR induced Ca2+ release in digitonin-permeabilized chromaffin cells and this was blocked by FK506 and rapamycin, ligands for FKBPs; 8Br-cADPR, a competitive antagonist for cADPR; and antibody for FKBP12/12.6, while it was enhanced by cyclosporin A. Tacrolimus 95-100 peptidyl-prolyl cis-trans isomerase FKBP1A Bos taurus 199-205 16648664-4 2006 FK506 binds to FKBP12.6 and removes it from RyRs, but cADPR did not affect the binding between FKBP12.6 and RyR. Tacrolimus 0-5 FKBP prolyl isomerase 1B Bos taurus 15-23 29539165-4 2006 The aims of this study were to investigate transforming growth factor-beta1 (TGF-beta1) gene polymorphisms in renal transplant recipients treated with CsA or tacrolimus and to establish an association between these polymorphisms and TGF-beta1 plasma concentration and the incidence of GO. Tacrolimus 158-168 transforming growth factor beta 1 Homo sapiens 77-86 29539165-8 2006 TGF-beta1 plasma levels were estimated in 60 CsA- and 30 tacrolimus-treated patients. Tacrolimus 57-67 transforming growth factor beta 1 Homo sapiens 0-9 16376427-0 2006 The usage of alternative splice sites in Mus musculus synaptotagmin-like 2 gene is modulated by cyclosporin A and FK506 in T-lymphocytes. Tacrolimus 114-119 synaptotagmin-like 2 Mus musculus 54-74 16628701-0 2006 Tacrolimus dose requirement in relation to donor and recipient ABCB1 and CYP3A5 gene polymorphisms in Chinese liver transplant patients. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 63-68 16628701-0 2006 Tacrolimus dose requirement in relation to donor and recipient ABCB1 and CYP3A5 gene polymorphisms in Chinese liver transplant patients. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 73-79 16628701-1 2006 The aim of this study was to investigate whether the heterogeneity in tacrolimus dose requirement is associated with ABCB1 and CYP3A5 gene polymorphisms in Chinese liver transplant patients during the first month after transplantation. Tacrolimus 70-80 ATP binding cassette subfamily B member 1 Homo sapiens 117-122 16628701-1 2006 The aim of this study was to investigate whether the heterogeneity in tacrolimus dose requirement is associated with ABCB1 and CYP3A5 gene polymorphisms in Chinese liver transplant patients during the first month after transplantation. Tacrolimus 70-80 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 127-133 16628701-7 2006 The tacrolimus C/D ratios were obviously lower in recipients carrying ABCB1 3435CC genotype. Tacrolimus 4-14 ATP binding cassette subfamily B member 1 Homo sapiens 70-75 16628701-10 2006 Analysis of the combination of recipients" ABCB1 and donors" CYP3A5 genotypes revealed that the tacrolimus C/D ratios were significantly lower in the ABCB1 3435CC-carrying recipients, regardless of donors" CYP3A5 genotype. Tacrolimus 96-106 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-67 16628701-10 2006 Analysis of the combination of recipients" ABCB1 and donors" CYP3A5 genotypes revealed that the tacrolimus C/D ratios were significantly lower in the ABCB1 3435CC-carrying recipients, regardless of donors" CYP3A5 genotype. Tacrolimus 96-106 ATP binding cassette subfamily B member 1 Homo sapiens 150-155 16628701-11 2006 In conclusion, our finding suggests that the recipients" ABCB1 and donors" CYP3A5 genotype affect the tacrolimus dose requirements. Tacrolimus 102-112 ATP binding cassette subfamily B member 1 Homo sapiens 57-62 16628701-11 2006 In conclusion, our finding suggests that the recipients" ABCB1 and donors" CYP3A5 genotype affect the tacrolimus dose requirements. Tacrolimus 102-112 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 75-81 16628701-12 2006 ABCB1 C3435T polymorphism is a major determinant of tacrolimus trough concentration in Chinese liver transplant recipients, and recipients with 3435CC genotype will require higher dose of tacrolimus. Tacrolimus 52-62 ATP binding cassette subfamily B member 1 Homo sapiens 0-5 16628701-12 2006 ABCB1 C3435T polymorphism is a major determinant of tacrolimus trough concentration in Chinese liver transplant recipients, and recipients with 3435CC genotype will require higher dose of tacrolimus. Tacrolimus 188-198 ATP binding cassette subfamily B member 1 Homo sapiens 0-5 16303143-0 2006 Immunosuppressant FK506 inhibits matrix metalloproteinase-9 induction in TNF-alpha-stimulated human hepatic stellate cells. Tacrolimus 18-23 matrix metallopeptidase 9 Homo sapiens 33-59 16303143-0 2006 Immunosuppressant FK506 inhibits matrix metalloproteinase-9 induction in TNF-alpha-stimulated human hepatic stellate cells. Tacrolimus 18-23 tumor necrosis factor Homo sapiens 73-82 16303143-5 2006 We also examined whether the immunosuppressant FK506 influences the MMPs expression in human HSCs. Tacrolimus 47-52 matrix metallopeptidase 9 Homo sapiens 68-72 16303143-6 2006 Human HSCs, LI90, were treated with TNF-alpha in the presence of FK506. Tacrolimus 65-70 tumor necrosis factor Homo sapiens 36-45 16303143-10 2006 FK506 suppressed this TNF-alpha-induced NK-kappaB activation, alone with pro-MMP-9 mRNA and protein induction, in HSC. Tacrolimus 0-5 tumor necrosis factor Homo sapiens 22-31 16303143-10 2006 FK506 suppressed this TNF-alpha-induced NK-kappaB activation, alone with pro-MMP-9 mRNA and protein induction, in HSC. Tacrolimus 0-5 matrix metallopeptidase 9 Homo sapiens 77-82 16303143-10 2006 FK506 suppressed this TNF-alpha-induced NK-kappaB activation, alone with pro-MMP-9 mRNA and protein induction, in HSC. Tacrolimus 0-5 fucosyltransferase 1 (H blood group) Homo sapiens 114-117 16303143-11 2006 TNF-alpha contributes to the perpetuation of liver fibrosis through MMP-9 production from HSCs and that FK506 inhibits the induction of MMP-9 through NF-kappaB pathway suggesting the anti-inflammatory properties of FK506. Tacrolimus 104-109 matrix metallopeptidase 9 Homo sapiens 136-141 16303143-11 2006 TNF-alpha contributes to the perpetuation of liver fibrosis through MMP-9 production from HSCs and that FK506 inhibits the induction of MMP-9 through NF-kappaB pathway suggesting the anti-inflammatory properties of FK506. Tacrolimus 215-220 matrix metallopeptidase 9 Homo sapiens 136-141 16680182-1 2006 The development by the Edmonton group of a sirolimus-based, steroid-free, low-tacrolimus regimen is a significant breakthrough that allows the rate of insulin independence after islet transplantation to increase from 13% to 80% at 1 year; however, the rate is reduced to 50% at 3 years, attributed to prolonged tacrolimus exposure. Tacrolimus 78-88 insulin Homo sapiens 151-158 16680182-1 2006 The development by the Edmonton group of a sirolimus-based, steroid-free, low-tacrolimus regimen is a significant breakthrough that allows the rate of insulin independence after islet transplantation to increase from 13% to 80% at 1 year; however, the rate is reduced to 50% at 3 years, attributed to prolonged tacrolimus exposure. Tacrolimus 311-321 insulin Homo sapiens 151-158 16866606-5 2006 Using these markers, an effect of CYP3A5 expression status has been demonstrated beyond doubt for therapies with the immunosuppressive drug tacrolimus. Tacrolimus 140-150 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 16704299-7 2006 Tacrolimus significantly upregulated the mRNA levels of insulin receptors and transforming growth factor-alpha (TGF-alpha), whereas rHuG-CSF did not. Tacrolimus 0-10 transforming growth factor alpha Rattus norvegicus 112-121 16490780-0 2006 Establishment of a cell model based on FKBP12 dimerization for screening of FK506-like neurotrophic small molecular compounds. Tacrolimus 76-81 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 39-45 16490780-7 2006 In this present study, the authors constructed a stable cell line, which underwent apoptosis upon treatment by AP20187, a wholly synthesized, cell-permeable dimeric FK506 derivative, based on FKBP12-mBax dimerization. Tacrolimus 165-170 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 192-198 16490780-8 2006 This AP20187-mediated apoptosis was rapidly reversed by the addition of an FKBP12-binding competitor molecule (FK506 or rapamycin), indicating that this cell line might be used to screen FK506 derivatives. Tacrolimus 111-116 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 75-81 16490780-8 2006 This AP20187-mediated apoptosis was rapidly reversed by the addition of an FKBP12-binding competitor molecule (FK506 or rapamycin), indicating that this cell line might be used to screen FK506 derivatives. Tacrolimus 187-192 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 75-81 16438932-13 2006 This PTH-stimulated ERK1/2 activation was inhibited by cyclosporine A and FK506. Tacrolimus 74-79 parathyroid hormone Mus musculus 5-8 16628088-7 2006 Treatment of mice with the calcineurin inhibitor FK506 totally blocked c-Jun NH2-terminal kinase activation, partially blocked the mammalian target of rapamycin pathway, and had no effect on extracellular signal-regulated kinase activation or the phosphorylation of eukaryotic initiation factor 4E. Tacrolimus 49-54 Jun proto-oncogene, AP-1 transcription factor subunit Homo sapiens 71-76 16628088-7 2006 Treatment of mice with the calcineurin inhibitor FK506 totally blocked c-Jun NH2-terminal kinase activation, partially blocked the mammalian target of rapamycin pathway, and had no effect on extracellular signal-regulated kinase activation or the phosphorylation of eukaryotic initiation factor 4E. Tacrolimus 49-54 eukaryotic translation initiation factor 4E Homo sapiens 266-297 16762204-10 2006 The concentration of FK506 in cornea was also higher in Group E than that in Group D. The expression of cytokines IL-2R alpha and MCP-1 mRNA was detected, but the expression of Fas and FasL mRNA was not detected in Group A. Tacrolimus 21-26 corticostatin-3 Oryctolagus cuniculus 130-135 16554484-0 2006 An analog of a dipeptide-like structure of FK506 increases glial cell line-derived neurotrophic factor expression through cAMP response element-binding protein activated by heat shock protein 90/Akt signaling pathway. Tacrolimus 43-48 glial cell derived neurotrophic factor Homo sapiens 59-102 16554484-0 2006 An analog of a dipeptide-like structure of FK506 increases glial cell line-derived neurotrophic factor expression through cAMP response element-binding protein activated by heat shock protein 90/Akt signaling pathway. Tacrolimus 43-48 cathelicidin antimicrobial peptide Homo sapiens 122-126 16554484-0 2006 An analog of a dipeptide-like structure of FK506 increases glial cell line-derived neurotrophic factor expression through cAMP response element-binding protein activated by heat shock protein 90/Akt signaling pathway. Tacrolimus 43-48 heat shock protein 90 alpha family class A member 1 Homo sapiens 173-194 16554484-0 2006 An analog of a dipeptide-like structure of FK506 increases glial cell line-derived neurotrophic factor expression through cAMP response element-binding protein activated by heat shock protein 90/Akt signaling pathway. Tacrolimus 43-48 AKT serine/threonine kinase 1 Homo sapiens 195-198 16554484-2 2006 We previously showed that leucine-isoleucine (Leu-Ile), an analog of a dipeptide-like structure of FK506 (tacrolimus), induces GDNF expression both in vivo and in vitro. Tacrolimus 99-104 glial cell derived neurotrophic factor Homo sapiens 127-131 16554484-2 2006 We previously showed that leucine-isoleucine (Leu-Ile), an analog of a dipeptide-like structure of FK506 (tacrolimus), induces GDNF expression both in vivo and in vitro. Tacrolimus 106-116 glial cell derived neurotrophic factor Homo sapiens 127-131 16438932-13 2006 This PTH-stimulated ERK1/2 activation was inhibited by cyclosporine A and FK506. Tacrolimus 74-79 mitogen-activated protein kinase 3 Mus musculus 20-26 16438932-10 2006 Cyclosporine A (10 microM) and FK506 (0.1 microM) did not affect the basal Mg(2+)uptake (140 +/- 16 and 142 +/- 14 nM/s, respectively), but they inhibited the PTH-stimulated Mg(2+) entry, decreasing it from 248+/-12 to 147 +/- 7 and 148 +/- 14 nM/s, respectively. Tacrolimus 31-36 parathyroid hormone Mus musculus 159-162 16549165-1 2006 BACKGROUND: Insulin resistance, a frequent prediabetic metabolic complication after renal transplantation, is generally linked to immunosuppressive drugs including corticosteroids, cyclosporine (CsA) or tacrolimus, as well as to age, cadaveric donors and ethnic factors. Tacrolimus 203-213 insulin Homo sapiens 12-19 16405911-2 2006 Removal of FKBP12 using FK506 or rapamycin causes an increased open probability and an increase in the frequency of sub-conductance states in RyR1. Tacrolimus 24-29 FKBP prolyl isomerase 1A Homo sapiens 11-17 16405911-2 2006 Removal of FKBP12 using FK506 or rapamycin causes an increased open probability and an increase in the frequency of sub-conductance states in RyR1. Tacrolimus 24-29 ryanodine receptor 1 Homo sapiens 142-146 16405911-7 2006 The orientation of RyR1-bound FKBP12, with part of its FK506 binding site facing towards RyR1, allows us to propose how FK506 is involved in the dissociation of FKBP12 from RyR1. Tacrolimus 55-60 ryanodine receptor 1 Homo sapiens 19-23 16405911-7 2006 The orientation of RyR1-bound FKBP12, with part of its FK506 binding site facing towards RyR1, allows us to propose how FK506 is involved in the dissociation of FKBP12 from RyR1. Tacrolimus 55-60 FKBP prolyl isomerase 1A Homo sapiens 30-36 16405911-7 2006 The orientation of RyR1-bound FKBP12, with part of its FK506 binding site facing towards RyR1, allows us to propose how FK506 is involved in the dissociation of FKBP12 from RyR1. Tacrolimus 55-60 FKBP prolyl isomerase 1A Homo sapiens 161-167 16405911-7 2006 The orientation of RyR1-bound FKBP12, with part of its FK506 binding site facing towards RyR1, allows us to propose how FK506 is involved in the dissociation of FKBP12 from RyR1. Tacrolimus 120-125 ryanodine receptor 1 Homo sapiens 19-23 16405911-7 2006 The orientation of RyR1-bound FKBP12, with part of its FK506 binding site facing towards RyR1, allows us to propose how FK506 is involved in the dissociation of FKBP12 from RyR1. Tacrolimus 120-125 FKBP prolyl isomerase 1A Homo sapiens 30-36 16405911-7 2006 The orientation of RyR1-bound FKBP12, with part of its FK506 binding site facing towards RyR1, allows us to propose how FK506 is involved in the dissociation of FKBP12 from RyR1. Tacrolimus 120-125 ryanodine receptor 1 Homo sapiens 89-93 16405911-7 2006 The orientation of RyR1-bound FKBP12, with part of its FK506 binding site facing towards RyR1, allows us to propose how FK506 is involved in the dissociation of FKBP12 from RyR1. Tacrolimus 120-125 FKBP prolyl isomerase 1A Homo sapiens 161-167 16405911-7 2006 The orientation of RyR1-bound FKBP12, with part of its FK506 binding site facing towards RyR1, allows us to propose how FK506 is involved in the dissociation of FKBP12 from RyR1. Tacrolimus 120-125 ryanodine receptor 1 Homo sapiens 89-93 16507844-6 2006 FK506 inhibited TGF-beta-induced collagen synthesis, and suppressed the expression of TGF-beta type I receptor (TbetaR-I) in TIG-3-20 cells. Tacrolimus 0-5 transforming growth factor beta 1 Homo sapiens 16-24 16507844-6 2006 FK506 inhibited TGF-beta-induced collagen synthesis, and suppressed the expression of TGF-beta type I receptor (TbetaR-I) in TIG-3-20 cells. Tacrolimus 0-5 transforming growth factor beta receptor 1 Homo sapiens 86-110 16507844-6 2006 FK506 inhibited TGF-beta-induced collagen synthesis, and suppressed the expression of TGF-beta type I receptor (TbetaR-I) in TIG-3-20 cells. Tacrolimus 0-5 transforming growth factor beta receptor 1 Homo sapiens 112-120 16507844-7 2006 Consistent with the in vitro findings, FK506 treatment starting on day 6 attenuated BLM-induced pulmonary fibrosis, in part, via reduced TbetaR-I expression. Tacrolimus 39-44 transforming growth factor beta receptor 1 Homo sapiens 137-145 16467444-2 2006 This study investigated whether tacrolimus, a novel calcineurin inhibitor, exerts fibrogenic effects to a similar extent. Tacrolimus 32-42 calcineurin binding protein 1 Homo sapiens 52-73 16604427-0 2006 Solution structure of the FK506-binding domain of human FKBP38. Tacrolimus 26-31 FKBP prolyl isomerase 8 Homo sapiens 56-62 16441773-0 2006 Effect of tacrolimus and partial hepatectomy on transthyretin metabolism in rats. Tacrolimus 10-20 transthyretin Rattus norvegicus 48-61 16441773-6 2006 After tacrolimus administration, TTR mRNA declined by 56% 12 h after the experiment started; however, after day 3, a rebound phenomenon occurred until day 7. Tacrolimus 6-16 transthyretin Rattus norvegicus 33-36 16441773-7 2006 Tacrolimus may facilitate serum TTR degradation, although production of TTR in the liver also increased. Tacrolimus 0-10 transthyretin Rattus norvegicus 32-35 16410343-0 2006 The aggregation of alpha-synuclein is stimulated by FK506 binding proteins as shown by fluorescence correlation spectroscopy. Tacrolimus 52-57 synuclein alpha Homo sapiens 19-34 16410343-2 2006 We have used fluorescence correlation spectroscopy (FCS) to study alpha-SYN aggregation in vitro and discovered that this process is clearly accelerated by addition of FK506 binding proteins (FKBPs). Tacrolimus 168-173 synuclein alpha Homo sapiens 66-75 16410343-3 2006 This effect was observed both with E. coli SlyD FKBP and with human FKBP12 and was counteracted by FK506, a specific inhibitor of FKBP. Tacrolimus 99-104 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 68-74 16549154-0 2006 Inhibition of the renin angiotensin system decreases fibrogenic cytokine expression in tacrolimus nephrotoxicity in rats. Tacrolimus 87-97 renin Rattus norvegicus 18-23 16549154-1 2006 The aim of our study was to investigate the influence of angiotensin-converting enzyme (ACE) inhibition and angiotensin II receptor blockage on the renal function by light microscopic and immunohistochemical findings in a rat model of tacrolimus nephrotoxicity. Tacrolimus 235-245 angiotensin I converting enzyme Rattus norvegicus 57-86 16549154-11 2006 In conclusion, the results of our study suggested that renin angiotensin inhibition down-regulates fibrogenic cytokine expression in rats displaying tacrolimus nephrotoxicity. Tacrolimus 149-159 renin Rattus norvegicus 55-60 16549155-0 2006 Transforming growth factor-beta1, vascular endothelial growth factor, and bone morphogenic protein-7 expression in tacrolimus-induced nephrotoxicity in rats. Tacrolimus 115-125 transforming growth factor, beta 1 Rattus norvegicus 0-32 16549155-0 2006 Transforming growth factor-beta1, vascular endothelial growth factor, and bone morphogenic protein-7 expression in tacrolimus-induced nephrotoxicity in rats. Tacrolimus 115-125 bone morphogenetic protein 7 Rattus norvegicus 74-100 16452687-0 2006 Inhibition of calcineurin by FK506 protects against polyglutamine-huntingtin toxicity through an increase of huntingtin phosphorylation at S421. Tacrolimus 29-34 huntingtin Rattus norvegicus 66-76 16445594-9 2006 Epigallocatechin-gallate and epigallocatechin suppressed a significant release of cytochrome c and activation of caspase-3 in FK506-treated LLC-PK1 cells. Tacrolimus 126-131 cytochrome c Sus scrofa 82-94 16445594-9 2006 Epigallocatechin-gallate and epigallocatechin suppressed a significant release of cytochrome c and activation of caspase-3 in FK506-treated LLC-PK1 cells. Tacrolimus 126-131 caspase 3 Sus scrofa 113-122 16452687-0 2006 Inhibition of calcineurin by FK506 protects against polyglutamine-huntingtin toxicity through an increase of huntingtin phosphorylation at S421. Tacrolimus 29-34 huntingtin Rattus norvegicus 109-119 16452687-6 2006 Inhibition of calcineurin activity, either by overexpression of the dominant-interfering form of CaN or by treatment with the specific inhibitor FK506, favors the phosphorylation of S421, restores the alteration in huntingtin S421 phosphorylation in HD neuronal cells, and prevents polyQ-mediated cell death of striatal neurons. Tacrolimus 145-150 huntingtin Rattus norvegicus 215-225 16452687-7 2006 Finally, we show that administration of FK506 to mice increases huntingtin S421 phosphorylation in brain. Tacrolimus 40-45 huntingtin Mus musculus 64-74 16418689-5 2006 Although CYP3A5 genotype is definitely associated with tacrolimus dosing, the only recommendation presently published is for an arbitrary doubling of the starting tacrolimus dose in CYP3A5 expressors. Tacrolimus 55-65 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 9-15 16418689-4 2006 The most extensive blood concentration/dose information available is on tacrolimus and its dosing related to CYP3A5 and ABCB1 gene polymorphisms. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 109-115 16444593-4 2006 In addition, similar treatment of cardiomyocytes with 10 nM FK506, a calcineurin inhibitor, could also attenuate isoproterenol-induced apoptosis. Tacrolimus 60-65 calcineurin binding protein 1 Homo sapiens 69-90 16424824-0 2006 Effect of intestinal CYP3A5 on postoperative tacrolimus trough levels in living-donor liver transplant recipients. Tacrolimus 45-55 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 21-27 16424824-1 2006 It has been reported that hepatic and intestinal cytochrome P450 (CYP) 3A4, CYP3A5 and P-glycoprotein affect the pharmacokinetics of tacrolimus, and that these proteins are associated with the large inter-individual variation in the pharmacokinetics of this drug. Tacrolimus 133-143 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 49-74 16424824-1 2006 It has been reported that hepatic and intestinal cytochrome P450 (CYP) 3A4, CYP3A5 and P-glycoprotein affect the pharmacokinetics of tacrolimus, and that these proteins are associated with the large inter-individual variation in the pharmacokinetics of this drug. Tacrolimus 133-143 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 76-82 16424824-1 2006 It has been reported that hepatic and intestinal cytochrome P450 (CYP) 3A4, CYP3A5 and P-glycoprotein affect the pharmacokinetics of tacrolimus, and that these proteins are associated with the large inter-individual variation in the pharmacokinetics of this drug. Tacrolimus 133-143 ATP binding cassette subfamily B member 1 Homo sapiens 87-101 16424824-2 2006 We previously showed that the concentration/dose ratio of tacrolimus tended to be lower in recipients of living-donor liver transplantation (LDLT) with a CYP3A5*1/*1-carrying graft. Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 154-160 16424824-7 2006 The tacrolimus concentration/dose ratio was lower in recipients with the CYP3A5*1/*1 and *1/*3 genotype (CYP3A5 expressors) compared to the CYP3A5*3/*3 genotype (non-expressors). Tacrolimus 4-14 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 73-79 16424824-7 2006 The tacrolimus concentration/dose ratio was lower in recipients with the CYP3A5*1/*1 and *1/*3 genotype (CYP3A5 expressors) compared to the CYP3A5*3/*3 genotype (non-expressors). Tacrolimus 4-14 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 105-111 16424824-7 2006 The tacrolimus concentration/dose ratio was lower in recipients with the CYP3A5*1/*1 and *1/*3 genotype (CYP3A5 expressors) compared to the CYP3A5*3/*3 genotype (non-expressors). Tacrolimus 4-14 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 105-111 16424824-8 2006 Amongst the combination of CYP3A5 genotypes between the graft liver and the native intestine, CYP3A5 expressors in both the graft liver and the native intestine had the lowest concentration/dose ratio of tacrolimus during 35 days after LDLT. Tacrolimus 204-214 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 94-100 16424824-9 2006 Patients with the intestinal CYP3A5*1 genotype tended to require a higher dose of tacrolimus compared to the other group with the same hepatic CYP3A5 genotype. Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 29-35 16424824-10 2006 These results indicate that intestinal CYP3A5, as well as hepatic CYP3A5, plays an important role in the first-pass effect of orally administered tacrolimus. Tacrolimus 146-156 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 39-45 16424824-10 2006 These results indicate that intestinal CYP3A5, as well as hepatic CYP3A5, plays an important role in the first-pass effect of orally administered tacrolimus. Tacrolimus 146-156 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 66-72 16418689-4 2006 The most extensive blood concentration/dose information available is on tacrolimus and its dosing related to CYP3A5 and ABCB1 gene polymorphisms. Tacrolimus 72-82 ATP binding cassette subfamily B member 1 Homo sapiens 120-125 16418689-5 2006 Although CYP3A5 genotype is definitely associated with tacrolimus dosing, the only recommendation presently published is for an arbitrary doubling of the starting tacrolimus dose in CYP3A5 expressors. Tacrolimus 163-173 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 182-188 16290149-2 2006 The novel affinity resins bearing FK506 (6a, 6b) captured specific binding protein, FKBP12, with a small amount of nonspecific binding proteins. Tacrolimus 34-39 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 84-90 16458528-4 2006 Here, we assessed the effect of FK506 on the expression of TGF-beta1, TbetaR-I, TbetaR-II, fibronectin (FN) and plasminogen activator inhibitor type-1 (PAI-1) in MCs. Tacrolimus 32-37 transforming growth factor beta 1 Homo sapiens 59-68 16458528-4 2006 Here, we assessed the effect of FK506 on the expression of TGF-beta1, TbetaR-I, TbetaR-II, fibronectin (FN) and plasminogen activator inhibitor type-1 (PAI-1) in MCs. Tacrolimus 32-37 transforming growth factor beta receptor 1 Homo sapiens 70-78 16458528-4 2006 Here, we assessed the effect of FK506 on the expression of TGF-beta1, TbetaR-I, TbetaR-II, fibronectin (FN) and plasminogen activator inhibitor type-1 (PAI-1) in MCs. Tacrolimus 32-37 transforming growth factor beta receptor 2 Homo sapiens 80-89 16458528-4 2006 Here, we assessed the effect of FK506 on the expression of TGF-beta1, TbetaR-I, TbetaR-II, fibronectin (FN) and plasminogen activator inhibitor type-1 (PAI-1) in MCs. Tacrolimus 32-37 fibronectin 1 Homo sapiens 91-102 16458528-4 2006 Here, we assessed the effect of FK506 on the expression of TGF-beta1, TbetaR-I, TbetaR-II, fibronectin (FN) and plasminogen activator inhibitor type-1 (PAI-1) in MCs. Tacrolimus 32-37 fibronectin 1 Homo sapiens 104-106 16458528-4 2006 Here, we assessed the effect of FK506 on the expression of TGF-beta1, TbetaR-I, TbetaR-II, fibronectin (FN) and plasminogen activator inhibitor type-1 (PAI-1) in MCs. Tacrolimus 32-37 serpin family E member 1 Homo sapiens 112-150 16458528-7 2006 Compared to untreated controls, FK506 stimulated TGF-beta1 mRNA (maximum at 8 h, 100 ng/mL: 2.13+/-0.15-fold, P<0.005) and protein expression (maximum at 96 h, 100 ng/mL: 1.96+/-0.29-fold, P<0.005). Tacrolimus 32-37 transforming growth factor beta 1 Homo sapiens 49-58 16458528-10 2006 These results indicate that, comparable to CsA, FK506 induced glomerulosclerosis is also due to a direct effect on mesangial matrix production, which is at least in part mediated via up-regulation of TGF-beta1 expression. Tacrolimus 48-53 transforming growth factor beta 1 Homo sapiens 200-209 16405733-11 2006 To this end, tacrolimus has been shown to have an impact on cancer signalling pathways such as the MAPK and the p53 pathway. Tacrolimus 13-23 tumor protein p53 Homo sapiens 112-115 16421475-0 2006 Influence of CYP3A5 gene polymorphisms of donor rather than recipient to tacrolimus individual dose requirement in liver transplantation. Tacrolimus 73-83 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 16637922-5 2006 The immunosuppressant (FK506), which enhances Ca(2+) release from the RyR, seems to have a symptomatic effect on MG patients with RyR antibodies. Tacrolimus 23-28 ryanodine receptor 1 Homo sapiens 70-73 16421475-2 2006 Tacrolimus is a substrate for CYP3A. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 30-35 16421475-3 2006 It has been conjectured that CYP3A5 polymorphism is associated with tacrolimus pharmacokinetic variations. Tacrolimus 68-78 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 29-35 16421475-4 2006 The objective of this study was to evaluate the contribution of polymorphisms of the donor and recipient CYP3A5 gene on tacrolimus disposition in liver transplantation. Tacrolimus 120-130 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 105-111 16421475-11 2006 CONCLUSION: The large interindividual variation of tacrolimus dose requirement is influenced by the metabolic activity of CYP3A5. Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 122-128 16671447-2 2006 We show that neuroprotection by 2 types of immunosuppressants, cyclosporin A (CsA) and tacrolimus (FK506), in a cryogenic brain injury model results from inhibition of calcineurin and protection from mitochondrial damage caused by formation of a mitochondrial permeability transition pore induced by cyclophilin D (CyPD), one of the prolyl cis/trans isomerase family members. Tacrolimus 87-97 peptidylprolyl isomerase D Rattus norvegicus 300-313 16671447-2 2006 We show that neuroprotection by 2 types of immunosuppressants, cyclosporin A (CsA) and tacrolimus (FK506), in a cryogenic brain injury model results from inhibition of calcineurin and protection from mitochondrial damage caused by formation of a mitochondrial permeability transition pore induced by cyclophilin D (CyPD), one of the prolyl cis/trans isomerase family members. Tacrolimus 87-97 peptidylprolyl isomerase D Rattus norvegicus 315-319 16671447-2 2006 We show that neuroprotection by 2 types of immunosuppressants, cyclosporin A (CsA) and tacrolimus (FK506), in a cryogenic brain injury model results from inhibition of calcineurin and protection from mitochondrial damage caused by formation of a mitochondrial permeability transition pore induced by cyclophilin D (CyPD), one of the prolyl cis/trans isomerase family members. Tacrolimus 99-104 peptidylprolyl isomerase D Rattus norvegicus 300-313 16671447-2 2006 We show that neuroprotection by 2 types of immunosuppressants, cyclosporin A (CsA) and tacrolimus (FK506), in a cryogenic brain injury model results from inhibition of calcineurin and protection from mitochondrial damage caused by formation of a mitochondrial permeability transition pore induced by cyclophilin D (CyPD), one of the prolyl cis/trans isomerase family members. Tacrolimus 99-104 peptidylprolyl isomerase D Rattus norvegicus 315-319 16637922-5 2006 The immunosuppressant (FK506), which enhances Ca(2+) release from the RyR, seems to have a symptomatic effect on MG patients with RyR antibodies. Tacrolimus 23-28 ryanodine receptor 1 Homo sapiens 130-133 16394527-2 2006 The values of total clearance (CL(tot)) of cyclosporin A, doxorubicin, tacrolimus and zonisamide in the CCl4-treated rats were decreased to about 1/2-1/3 of those in control rats. Tacrolimus 71-81 C-C motif chemokine ligand 4 Rattus norvegicus 104-108 16413244-6 2006 The mRNA expression level of MDR1 was inversely correlated with the tacrolimus concentration-oral dose ratio during the initial 4 days after surgery in patients with a graft-to-recipient weight ratio greater than 1.5 (r= -0.6798, P< .0001) and those with a graft-to-recipient weight ratio of less than 1.5 (r= -0.7180, P< .0001). Tacrolimus 68-78 ATP binding cassette subfamily B member 1 Homo sapiens 29-33 17032130-0 2006 The influence of genetic polymorphisms of cytochrome P450 3A5 and ABCB1 on starting dose- and weight-standardized tacrolimus trough concentrations after kidney transplantation in relation to renal function. Tacrolimus 114-124 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 42-61 17032130-0 2006 The influence of genetic polymorphisms of cytochrome P450 3A5 and ABCB1 on starting dose- and weight-standardized tacrolimus trough concentrations after kidney transplantation in relation to renal function. Tacrolimus 114-124 ATP binding cassette subfamily B member 1 Homo sapiens 66-71 17032130-1 2006 BACKGROUND: Cytochrome P450 3A5 (CYP3A5) and ABCB1 polymorphisms have been shown to influence tacrolimus (Tc) blood concentrations in the stable phase after organ transplantation. Tacrolimus 94-104 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-31 17032130-1 2006 BACKGROUND: Cytochrome P450 3A5 (CYP3A5) and ABCB1 polymorphisms have been shown to influence tacrolimus (Tc) blood concentrations in the stable phase after organ transplantation. Tacrolimus 94-104 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 17032130-1 2006 BACKGROUND: Cytochrome P450 3A5 (CYP3A5) and ABCB1 polymorphisms have been shown to influence tacrolimus (Tc) blood concentrations in the stable phase after organ transplantation. Tacrolimus 94-104 ATP binding cassette subfamily B member 1 Homo sapiens 45-50 17032130-1 2006 BACKGROUND: Cytochrome P450 3A5 (CYP3A5) and ABCB1 polymorphisms have been shown to influence tacrolimus (Tc) blood concentrations in the stable phase after organ transplantation. Tacrolimus 106-108 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-31 17032130-1 2006 BACKGROUND: Cytochrome P450 3A5 (CYP3A5) and ABCB1 polymorphisms have been shown to influence tacrolimus (Tc) blood concentrations in the stable phase after organ transplantation. Tacrolimus 106-108 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 17032130-1 2006 BACKGROUND: Cytochrome P450 3A5 (CYP3A5) and ABCB1 polymorphisms have been shown to influence tacrolimus (Tc) blood concentrations in the stable phase after organ transplantation. Tacrolimus 106-108 ATP binding cassette subfamily B member 1 Homo sapiens 45-50 17032130-3 2006 METHODS: We retrospectively analyzed data from a cohort of 59 kidney transplant recipients, in whom CYP3A5 (intron 3) and ABCB1 (exons 12, 21 and 26) genotypes were correlated to dose- and weight-standardized Tc trough concentrations obtained after initial Tc doses. Tacrolimus 209-211 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 100-106 17032130-5 2006 RESULTS: Dose- and weight-standardized Tc trough concentrations were lower in patients carrying the CYP3A5 *1 allele (p<0.01). Tacrolimus 39-41 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 100-106 17032130-7 2006 In a multivariate analysis, both the presence of at least one CYP3A5 *1 allele (p=0.006) and age at the time of transplantation (p=0.010) were significant independent variables affecting Tc trough blood concentrations standardized to the first dosages (model r2=0.23). Tacrolimus 187-189 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 62-68 16430309-11 2006 This may reflect faster turnover of tacrolimus by CYP3A5 than the other substrates. Tacrolimus 36-46 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 50-56 18689275-6 2006 The combination of daily plasmapheresis with fresh frozen plasma as a source of ADAMTS-13 and cyclosporine may be used as a rescue therapy in patients with FK506-induced TTP/HUS. Tacrolimus 156-161 ADAM metallopeptidase with thrombospondin type 1 motif 13 Homo sapiens 80-89 16263818-15 2006 Surprisingly, all APECED-related symptoms including candidiasis and autoantibody levels decreased, presumably due to the reinforced immunosuppression (tacrolimus, mycophenolate mofetil, prednisone). Tacrolimus 151-161 autoimmune regulator Homo sapiens 18-24 17190738-0 2006 FK506: an immunosuppressive agent preserving HIF-1 activity. Tacrolimus 0-5 hypoxia inducible factor 1 subunit alpha Homo sapiens 45-50 17190738-7 2006 FK506 could preserve HIF-1 activity in donor organs subjected to hypoxia. Tacrolimus 0-5 hypoxia inducible factor 1 subunit alpha Homo sapiens 21-26 16534238-8 2006 Creatinine clearance increased from 20 +/- 15 ml/min/1.73 m2 at the start of tacrolimus therapy to 37 +/- 29 ml/min/1.73 m2 and to 32 +/- 26 ml/min/1.73 m2 after 5 and 7 years. Tacrolimus 77-87 CD59 molecule (CD59 blood group) Homo sapiens 49-54 16410063-4 2006 In experimental animals, tacrolimus (FK506) was injected every day to inhibit the IL-2 production by helper T-cells. Tacrolimus 25-35 interleukin 2 Rattus norvegicus 82-86 16410063-4 2006 In experimental animals, tacrolimus (FK506) was injected every day to inhibit the IL-2 production by helper T-cells. Tacrolimus 37-42 interleukin 2 Rattus norvegicus 82-86 16289353-11 2006 CONCLUSION: The beneficial effects of tacrolimus during in vitro ischemia/reperfusion seem to indicate the restoration of a glutamate transporter-1-mediated activity and could be mediated by a FK506 binding protein 12 kDa pathway. Tacrolimus 38-48 solute carrier family 1 member 2 Rattus norvegicus 124-147 16431292-10 2006 CONCLUSION: This study demonstrates that ABCB1 haplotypes derived from three common polymorphisms are associated with tacrolimus dosing in lung transplant patients when eliminating the confounder CYP3A5 genotype. Tacrolimus 118-128 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 196-202 16507509-0 2006 Role of P-glycoprotein in the hepatic metabolism of tacrolimus. Tacrolimus 52-62 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 8-22 16431292-0 2006 Impact of ABCB1 (MDR1) haplotypes on tacrolimus dosing in adult lung transplant patients who are CYP3A5 *3/*3 non-expressors. Tacrolimus 37-47 ATP binding cassette subfamily B member 1 Homo sapiens 10-15 16431292-0 2006 Impact of ABCB1 (MDR1) haplotypes on tacrolimus dosing in adult lung transplant patients who are CYP3A5 *3/*3 non-expressors. Tacrolimus 37-47 ATP binding cassette subfamily B member 1 Homo sapiens 17-21 16431292-0 2006 Impact of ABCB1 (MDR1) haplotypes on tacrolimus dosing in adult lung transplant patients who are CYP3A5 *3/*3 non-expressors. Tacrolimus 37-47 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 97-103 16431292-1 2006 BACKGROUND: The influence of ABCB1 (MDR1) polymorphisms on tacrolimus dosing has been questioned in previous studies with contradictory findings, possibly due to the association between CYP3A5 polymorphisms and tacrolimus dosing. Tacrolimus 59-69 ATP binding cassette subfamily B member 1 Homo sapiens 29-34 16431292-1 2006 BACKGROUND: The influence of ABCB1 (MDR1) polymorphisms on tacrolimus dosing has been questioned in previous studies with contradictory findings, possibly due to the association between CYP3A5 polymorphisms and tacrolimus dosing. Tacrolimus 59-69 ATP binding cassette subfamily B member 1 Homo sapiens 36-40 16431292-1 2006 BACKGROUND: The influence of ABCB1 (MDR1) polymorphisms on tacrolimus dosing has been questioned in previous studies with contradictory findings, possibly due to the association between CYP3A5 polymorphisms and tacrolimus dosing. Tacrolimus 59-69 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 186-192 16431292-1 2006 BACKGROUND: The influence of ABCB1 (MDR1) polymorphisms on tacrolimus dosing has been questioned in previous studies with contradictory findings, possibly due to the association between CYP3A5 polymorphisms and tacrolimus dosing. Tacrolimus 211-221 ATP binding cassette subfamily B member 1 Homo sapiens 29-34 16431292-1 2006 BACKGROUND: The influence of ABCB1 (MDR1) polymorphisms on tacrolimus dosing has been questioned in previous studies with contradictory findings, possibly due to the association between CYP3A5 polymorphisms and tacrolimus dosing. Tacrolimus 211-221 ATP binding cassette subfamily B member 1 Homo sapiens 36-40 16431292-1 2006 BACKGROUND: The influence of ABCB1 (MDR1) polymorphisms on tacrolimus dosing has been questioned in previous studies with contradictory findings, possibly due to the association between CYP3A5 polymorphisms and tacrolimus dosing. Tacrolimus 211-221 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 186-192 16431292-2 2006 The objective of this study was to assess the effect of ABCB1 haplotypes from 3 distinct polymorphic sites on the tacrolimus level/dose [L/D] in lung transplant patients limited to CYP3A5 *3/*3 non-expressors. Tacrolimus 114-124 ATP binding cassette subfamily B member 1 Homo sapiens 56-61 16431292-10 2006 CONCLUSION: This study demonstrates that ABCB1 haplotypes derived from three common polymorphisms are associated with tacrolimus dosing in lung transplant patients when eliminating the confounder CYP3A5 genotype. Tacrolimus 118-128 ATP binding cassette subfamily B member 1 Homo sapiens 41-46 16507509-1 2006 The main objective was to determine the potential effect of P-glycoprotein (P-gp) modulation on hepatic metabolism of tacrolimus, a P-gp and cytochrome P450(CYP)3A4 substrate, and to investigate various potential factors that may contribute to the interaction between P-gp and CYP. Tacrolimus 118-128 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 60-74 16507509-1 2006 The main objective was to determine the potential effect of P-glycoprotein (P-gp) modulation on hepatic metabolism of tacrolimus, a P-gp and cytochrome P450(CYP)3A4 substrate, and to investigate various potential factors that may contribute to the interaction between P-gp and CYP. Tacrolimus 118-128 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 76-80 16507509-1 2006 The main objective was to determine the potential effect of P-glycoprotein (P-gp) modulation on hepatic metabolism of tacrolimus, a P-gp and cytochrome P450(CYP)3A4 substrate, and to investigate various potential factors that may contribute to the interaction between P-gp and CYP. Tacrolimus 118-128 cytochrome P450, family 3, subfamily a, polypeptide 23-polypeptide 1 Rattus norvegicus 157-160 16507509-1 2006 The main objective was to determine the potential effect of P-glycoprotein (P-gp) modulation on hepatic metabolism of tacrolimus, a P-gp and cytochrome P450(CYP)3A4 substrate, and to investigate various potential factors that may contribute to the interaction between P-gp and CYP. Tacrolimus 118-128 cytochrome P450, family 3, subfamily a, polypeptide 23-polypeptide 1 Rattus norvegicus 277-280 16368244-10 2005 Quantification of intratumor transcription levels using reverse transcription-real-time polymerase chain reaction showed recovery from tacrolimus-induced downregulation of interleukin-4 but not interferon-gamma levels. Tacrolimus 135-145 interleukin 4 Homo sapiens 172-185 16378079-0 2005 The immunosuppressive agent FK506 enhances the cytolytic activity of inhibitory natural killer cell receptor (CD94/NKG2A)-expressing CD8 T cells. Tacrolimus 28-33 killer cell lectin like receptor D1 Homo sapiens 110-114 16378079-0 2005 The immunosuppressive agent FK506 enhances the cytolytic activity of inhibitory natural killer cell receptor (CD94/NKG2A)-expressing CD8 T cells. Tacrolimus 28-33 killer cell lectin like receptor C1 Homo sapiens 115-120 16378079-1 2005 Tacrolimus (FK506) is a potent immunosuppressive agent that inhibit transcription of cytokines such as IL-2 in T cells. Tacrolimus 0-10 interleukin 2 Homo sapiens 103-107 16378079-1 2005 Tacrolimus (FK506) is a potent immunosuppressive agent that inhibit transcription of cytokines such as IL-2 in T cells. Tacrolimus 12-17 interleukin 2 Homo sapiens 103-107 16378079-4 2005 However, cytolytic activity levels of purified CD94-expressing cells from 7-day cultures with FK506 were much higher than those from 7-day cultures without FK506. Tacrolimus 94-99 killer cell lectin like receptor D1 Homo sapiens 47-51 16378079-4 2005 However, cytolytic activity levels of purified CD94-expressing cells from 7-day cultures with FK506 were much higher than those from 7-day cultures without FK506. Tacrolimus 156-161 killer cell lectin like receptor D1 Homo sapiens 47-51 16204252-5 2005 Both a calcineurin inhibitor, FK506, and a selective NMDA receptor antagonist, MK-801, inhibit the dephosphorylation of DAPK after in vitro ischemia. Tacrolimus 30-35 death associated protein kinase 1 Homo sapiens 120-124 16246307-0 2005 Mechanism of osteogenic induction by FK506 via BMP/Smad pathways. Tacrolimus 37-42 bone morphogenetic protein 1 Homo sapiens 47-50 16246307-0 2005 Mechanism of osteogenic induction by FK506 via BMP/Smad pathways. Tacrolimus 37-42 SMAD family member 6 Homo sapiens 51-55 16246307-1 2005 FK506 is an immunosuppressant that exerts effects by binding to FK506-binding protein 12 (FKBP12). Tacrolimus 0-5 FKBP prolyl isomerase 1A Homo sapiens 64-88 16246307-1 2005 FK506 is an immunosuppressant that exerts effects by binding to FK506-binding protein 12 (FKBP12). Tacrolimus 0-5 FKBP prolyl isomerase 1A Homo sapiens 90-96 16246307-3 2005 The present study initially showed that FK506 alone at a higher concentration (1muM) induced osteogenic differentiation of mesenchymal cell lines, which was suppressed by adenoviral introduction of Smad6. Tacrolimus 40-45 SMAD family member 6 Homo sapiens 198-203 16256941-4 2005 Treatment of CaM-TG mice with the calcineurin inhibitor FK506 (1mg/kg per day) prevented the increase in the heart-to-body weight ratio as well as that in cardiomyocyte width. Tacrolimus 56-61 calmodulin 2 Mus musculus 13-16 16246307-4 2005 FK506 rapidly activates the BMP-dependent Smads in the absence of BMPs, and the activation was blocked by Smad6. Tacrolimus 0-5 bone morphogenetic protein 1 Homo sapiens 28-31 16256941-5 2005 FK506 also inhibited the induction of fetal-type cardiac gene expression in CaM-TG mice. Tacrolimus 0-5 calmodulin 2 Mus musculus 76-79 16256941-6 2005 Overexpression of CaM in cultured rat cardiomyocytes activated the ANF gene promoter in a manner sensitive to FK506. Tacrolimus 110-115 calmodulin 1 Rattus norvegicus 18-21 16246307-4 2005 FK506 rapidly activates the BMP-dependent Smads in the absence of BMPs, and the activation was blocked by Smad6. Tacrolimus 0-5 SMAD family member 6 Homo sapiens 106-111 16246307-5 2005 Overexpression of FKBP12, which was reported to block the ligand-independent activation of BMP type I receptor A (BMPRIA), suppressed Smad signaling induced by FK506, but not that induced by BMP2. Tacrolimus 160-165 FKBP prolyl isomerase 1A Homo sapiens 18-24 16246307-5 2005 Overexpression of FKBP12, which was reported to block the ligand-independent activation of BMP type I receptor A (BMPRIA), suppressed Smad signaling induced by FK506, but not that induced by BMP2. Tacrolimus 160-165 bone morphogenetic protein 1 Homo sapiens 91-94 16246307-5 2005 Overexpression of FKBP12, which was reported to block the ligand-independent activation of BMP type I receptor A (BMPRIA), suppressed Smad signaling induced by FK506, but not that induced by BMP2. Tacrolimus 160-165 SMAD family member 6 Homo sapiens 134-138 16246307-6 2005 BMPRIA and FKBP12 bound to each other, and this binding was suppressed by FK506. Tacrolimus 74-79 FKBP prolyl isomerase 1A Homo sapiens 11-17 16246307-7 2005 These data suggest that FK506 promotes osteogenic differentiation by activating BMP receptors through interacting with FKBP12. Tacrolimus 24-29 bone morphogenetic protein 1 Homo sapiens 80-83 16246307-7 2005 These data suggest that FK506 promotes osteogenic differentiation by activating BMP receptors through interacting with FKBP12. Tacrolimus 24-29 FKBP prolyl isomerase 1A Homo sapiens 119-125 16256941-6 2005 Overexpression of CaM in cultured rat cardiomyocytes activated the ANF gene promoter in a manner sensitive to FK506. Tacrolimus 110-115 natriuretic peptide A Rattus norvegicus 67-70 16416180-0 2005 A higher dose requirement of tacrolimus in active Crohn"s disease may be related to a high intestinal P-glycoprotein content. Tacrolimus 29-39 ATP binding cassette subfamily B member 1 Homo sapiens 102-116 16409819-0 2005 [Relationship between MDR1 gene polymorphism and blood concentration of tacrolimus in renal transplant patients]. Tacrolimus 72-82 ATP binding cassette subfamily B member 1 Homo sapiens 22-26 16409819-1 2005 OBJECTIVE: To investigate the relationship between MDR1 exon 21 and exon 26 polymorphism and whole blood concentration of tacrolimus (FK506) in renal transplant patients. Tacrolimus 122-132 ATP binding cassette subfamily B member 1 Homo sapiens 51-55 16409819-1 2005 OBJECTIVE: To investigate the relationship between MDR1 exon 21 and exon 26 polymorphism and whole blood concentration of tacrolimus (FK506) in renal transplant patients. Tacrolimus 134-139 ATP binding cassette subfamily B member 1 Homo sapiens 51-55 16409819-9 2005 Three, six, and twelve months after the transplantation a significant correlation between the whole blood FK506 concentration per dose/day and MDR1 exon 21 and exon 26 genotypes. Tacrolimus 106-111 ATP binding cassette subfamily B member 1 Homo sapiens 143-147 16409819-14 2005 CONCLUSIONS: The MDR1 gene polymorphism is correlated with the whole blood concentration of FK506. Tacrolimus 92-97 ATP binding cassette subfamily B member 1 Homo sapiens 17-21 16400900-0 2005 Effect of FK506 on the interleukin 15-driven proliferation and apoptosis of anti-CD3-activated umbilical cord blood T cells. Tacrolimus 10-15 interleukin 15 Homo sapiens 23-37 16400900-2 2005 OBJECTIVE: To investigate the FK506 sensitivity of interleukin 15 (IL-15)- and IL-2-driven proliferation and apoptosis of anti-CD3-stimulated CB T cells compared with adult peripheral blood (APB) T cells. Tacrolimus 30-35 interleukin 15 Homo sapiens 51-65 16400900-2 2005 OBJECTIVE: To investigate the FK506 sensitivity of interleukin 15 (IL-15)- and IL-2-driven proliferation and apoptosis of anti-CD3-stimulated CB T cells compared with adult peripheral blood (APB) T cells. Tacrolimus 30-35 interleukin 15 Homo sapiens 67-72 16400900-2 2005 OBJECTIVE: To investigate the FK506 sensitivity of interleukin 15 (IL-15)- and IL-2-driven proliferation and apoptosis of anti-CD3-stimulated CB T cells compared with adult peripheral blood (APB) T cells. Tacrolimus 30-35 interleukin 2 Homo sapiens 79-83 16400900-5 2005 Although IL-15-driven proliferation evaluated by carboxyfluorescein diacetae succinimidyl ester staining revealed comparable sensitivity to FK506 in anti-CD3-stimulated CB and APB T cells, IL-15-driven CD25 up-regulation in CB T cells was more sensitive to FK506 inhibition than APB T cells. Tacrolimus 140-145 interleukin 15 Homo sapiens 9-14 16400900-5 2005 Although IL-15-driven proliferation evaluated by carboxyfluorescein diacetae succinimidyl ester staining revealed comparable sensitivity to FK506 in anti-CD3-stimulated CB and APB T cells, IL-15-driven CD25 up-regulation in CB T cells was more sensitive to FK506 inhibition than APB T cells. Tacrolimus 257-262 interleukin 15 Homo sapiens 189-194 16400900-7 2005 However, the FK506 sensitivity of anti-CD3-induced T-cell apoptosis was lost in IL-15-supplemented CB cultures (P = .51) but not in corresponding APB cultures (P = .002). Tacrolimus 13-18 interleukin 15 Homo sapiens 80-85 16400900-8 2005 The IL-15-enhanced Fas expression on CB T cells (CD95) was decreased by FK506, similar to that observed with adults. Tacrolimus 72-77 interleukin 15 Homo sapiens 4-9 16400900-8 2005 The IL-15-enhanced Fas expression on CB T cells (CD95) was decreased by FK506, similar to that observed with adults. Tacrolimus 72-77 Fas cell surface death receptor Homo sapiens 49-53 16400900-9 2005 CONCLUSIONS: We observed differential FK506 sensitivity of IL-15-driven CD25 up-regulation and apoptotic response comparing CB and APB T cells. Tacrolimus 38-43 interleukin 15 Homo sapiens 59-64 16400900-9 2005 CONCLUSIONS: We observed differential FK506 sensitivity of IL-15-driven CD25 up-regulation and apoptotic response comparing CB and APB T cells. Tacrolimus 38-43 interleukin 2 receptor subunit alpha Homo sapiens 72-76 16400900-10 2005 This finding suggests the potential therapeutic benefit of FK506 in ameliorating graft-vs-host disease by decreasing IL-15-driven donor T-cell proliferation without inhibiting associated activation-induced apoptosis during CB transplantation. Tacrolimus 59-64 interleukin 15 Homo sapiens 117-122 16416180-1 2005 Tacrolimus, a relatively new therapeutic option for patients with corticosteroid-refractory Crohn"s disease or ulcerative colitis, is a substrate for the apically directed efflux transporter P-glycoprotein (P-gp). Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 191-205 16416180-1 2005 Tacrolimus, a relatively new therapeutic option for patients with corticosteroid-refractory Crohn"s disease or ulcerative colitis, is a substrate for the apically directed efflux transporter P-glycoprotein (P-gp). Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 207-211 16416180-4 2005 Elevated intestinal P-gp could have resulted in decreased tacrolimus absorption, thereby leading to decreased blood concentration and decreased efficacy in this patient. Tacrolimus 58-68 ATP binding cassette subfamily B member 1 Homo sapiens 20-24 16417450-3 2005 Here, we report a patient with fulminant hepatic failure who received an ABO-incompatible liver transplantation who was treated with pre- and posttransplantation double-volume total plasma exchange, splenectomy, and triple immunosuppression (tacrolimus, mycophenolate mofetil, and prednisone) in July 2003. Tacrolimus 242-252 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 73-76 16316299-5 2005 Based on current evidence, the most promising strategy would be use of the cytochrome P450 3A5 expressor genotype to guide initial dosing with tacrolimus. Tacrolimus 143-153 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 75-94 16387090-20 2005 A two-point sampling method using C5 and C1.5 or three-point sampling method using C5, C1.5, and C3 might be the best abbreviated AUC for a cost-effective tacrolimus monitoring strategy. Tacrolimus 155-165 placenta associated 8 Homo sapiens 87-91 16278292-7 2005 However, FK506, which with FKBP12 inhibits calcineurin (but not mTOR), potentiated the IP3-evoked [Ca2+]c increase. Tacrolimus 9-14 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 27-33 16331555-0 2005 Impact of multidrug resistance 1 gene polymorphism on tacrolimus dose and concentration-to-dose ratio in Chinese liver transplantation recipients. Tacrolimus 54-64 ATP binding cassette subfamily B member 1 Homo sapiens 10-32 16331555-1 2005 OBJECTIVE: To investigate whether the polymorphism of multidrug resistance 1 gene (MDR1) in the donors and liver transplantation recipients was correlated with interindividual variation in tacrolimus dose requirement and concentration-to-dose ratio. Tacrolimus 189-199 ATP binding cassette subfamily B member 1 Homo sapiens 54-76 16331555-1 2005 OBJECTIVE: To investigate whether the polymorphism of multidrug resistance 1 gene (MDR1) in the donors and liver transplantation recipients was correlated with interindividual variation in tacrolimus dose requirement and concentration-to-dose ratio. Tacrolimus 189-199 ATP binding cassette subfamily B member 1 Homo sapiens 83-87 16331555-5 2005 Tacrolimus doses required to achieve target blood concentrations were higher in the patients with MDR1 CC genotype than in the CT or TT genotype patients, and the dose-adjusted trough levels were lower. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 98-102 16331555-7 2005 CONCLUSION: Tacrolimus dose requirement and dose-adjusted trough levels were correlated with MDR1 3435 (C-->T) polymorphism, and MDR1 3435 (C-->T) polymorphism analysis is helpful to individualize tacrolimus administration. Tacrolimus 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 93-97 16331555-7 2005 CONCLUSION: Tacrolimus dose requirement and dose-adjusted trough levels were correlated with MDR1 3435 (C-->T) polymorphism, and MDR1 3435 (C-->T) polymorphism analysis is helpful to individualize tacrolimus administration. Tacrolimus 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 132-136 16321613-10 2005 CD40L expression was reduced in tacrolimus-treated patients (median, 34 [interquartile range, 28-41] to 21 [interquartile range, 12-26] mean fluorescence intensity; P < .002) and cyclosporine-treated patients (median, 33 [interquartile range, 30-37] to 26 [interquartile range, 19-26] mean fluorescence intensity; P < .02). Tacrolimus 32-42 CD40 ligand Homo sapiens 0-5 16176796-6 2005 In this study, from our NMR studies and calcineurin assays in vitro, we demonstrate that the N-terminal fragment of FKBP38 which contains the FK-506 binding domain does not bind FK-506 at molecular level. Tacrolimus 142-148 FKBP prolyl isomerase 8 Homo sapiens 116-122 16083867-7 2005 Ryanodine (100 nM) and FK506 (10 microM), both of which promote spontaneous SR Ca(2+) release from ryanodine receptor (RyR2) during diastole, reproduced the effect of HF on I(K1) in normal cells but had no effect in HF cells. Tacrolimus 23-28 ryanodine receptor 2 Rattus norvegicus 119-123 16083867-9 2005 Rapamycin (10 microM), which removes FKBP binding proteins from RyR2 with no effect on calcineurin, mimicked the effect of FK506 on I(K1). Tacrolimus 123-128 ryanodine receptor 2 Rattus norvegicus 64-68 16321613-12 2005 MMP-9 decreased from 88 ng/mL (range, 49-135 ng/mL) to 57 ng/mL (range, 38-73 ng/mL) (P < .05) in tacrolimus-treated patients and from 79 ng/mL (range, 54-148 ng/mL) to 66 ng/mL (range, 41-97 ng/mL) (P < .01) in cyclosporine-treated patients. Tacrolimus 101-111 matrix metallopeptidase 9 Homo sapiens 0-5 16221205-10 2005 The 24 kD caspase-3, which proved to be an active caspase-3 subunit, was increased in I/R and CsA groups and deceased by tacrolimus, rapamycin, or MMF (P < 0.05), but not 32 kD precursor or 17 kD active caspase-3. Tacrolimus 121-131 caspase 3 Rattus norvegicus 10-19 16084678-8 2005 The present findings suggest that these two calcineurin inhibitors have a protective effect against acoustic injury of the cochlea, whereas the non-calcineurin inhibitor, rapamycin, not only has no effect against acoustic injury, but rather blocked the effect of FK506. Tacrolimus 263-268 calcineurin binding protein 1 Mus musculus 44-65 16221205-10 2005 The 24 kD caspase-3, which proved to be an active caspase-3 subunit, was increased in I/R and CsA groups and deceased by tacrolimus, rapamycin, or MMF (P < 0.05), but not 32 kD precursor or 17 kD active caspase-3. Tacrolimus 121-131 caspase 3 Rattus norvegicus 50-59 16221205-10 2005 The 24 kD caspase-3, which proved to be an active caspase-3 subunit, was increased in I/R and CsA groups and deceased by tacrolimus, rapamycin, or MMF (P < 0.05), but not 32 kD precursor or 17 kD active caspase-3. Tacrolimus 121-131 caspase 3 Rattus norvegicus 50-59 16235347-16 2005 Treating 100 recipients with tacrolimus instead of cyclosporin would avoid 12 suffering acute rejection, two losing their graft but cause an extra five to become insulin-requiring diabetics. Tacrolimus 29-39 insulin Homo sapiens 162-169 16386540-8 2005 RESULTS: The change to tacrolimus resulted in significant decreases in TC levels, 213 +/- 30 (B) versus 185 +/- 27 (12s) (P < .01); LDL, 129 +/- 24 (B) versus 104 +/- 14 (12s) (P = .002); and ApoB 98 +/- 15 (B) versus 85 +/- 10 (12s) (P < .01). Tacrolimus 23-33 apolipoprotein B Homo sapiens 195-199 16146556-0 2005 Influence of CYP3A5 and MDR1 polymorphisms on tacrolimus concentration in the early stage after renal transplantation. Tacrolimus 46-56 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 16221864-8 2005 Consequently, when calcineurin was inhibited by its inhibitor FK506 under the depolarizing condition, the CaMK-mediated increase in BDNF became a stimulatory signal, and the endogenous BDNF autocrine system was capable of upregulating NR2C mRNA via the common TrkB-ERK cascade. Tacrolimus 62-67 brain derived neurotrophic factor Mus musculus 132-136 16221864-8 2005 Consequently, when calcineurin was inhibited by its inhibitor FK506 under the depolarizing condition, the CaMK-mediated increase in BDNF became a stimulatory signal, and the endogenous BDNF autocrine system was capable of upregulating NR2C mRNA via the common TrkB-ERK cascade. Tacrolimus 62-67 brain derived neurotrophic factor Mus musculus 185-189 16221864-8 2005 Consequently, when calcineurin was inhibited by its inhibitor FK506 under the depolarizing condition, the CaMK-mediated increase in BDNF became a stimulatory signal, and the endogenous BDNF autocrine system was capable of upregulating NR2C mRNA via the common TrkB-ERK cascade. Tacrolimus 62-67 glutamate receptor, ionotropic, NMDA2C (epsilon 3) Mus musculus 235-239 16221864-8 2005 Consequently, when calcineurin was inhibited by its inhibitor FK506 under the depolarizing condition, the CaMK-mediated increase in BDNF became a stimulatory signal, and the endogenous BDNF autocrine system was capable of upregulating NR2C mRNA via the common TrkB-ERK cascade. Tacrolimus 62-67 neurotrophic tyrosine kinase, receptor, type 2 Mus musculus 260-264 16221864-8 2005 Consequently, when calcineurin was inhibited by its inhibitor FK506 under the depolarizing condition, the CaMK-mediated increase in BDNF became a stimulatory signal, and the endogenous BDNF autocrine system was capable of upregulating NR2C mRNA via the common TrkB-ERK cascade. Tacrolimus 62-67 mitogen-activated protein kinase 1 Mus musculus 265-268 15930144-5 2005 Pharmacological inhibition of calcineurin in mice with either cyclosporin A or FK506 resulted in a marked decrease in utrophin A expression at synaptic sites, whereas constitutive activation of calcineurin had the opposite effect. Tacrolimus 79-84 utrophin Mus musculus 118-126 16146556-2 2005 Cytochrome P450 (CYP) 3A and P-glycoprotein (P-gp, encoded by MDR1) play an important role in the absorption and metabolism of tacrolimus. Tacrolimus 127-137 ATP binding cassette subfamily B member 1 Homo sapiens 45-49 16146556-2 2005 Cytochrome P450 (CYP) 3A and P-glycoprotein (P-gp, encoded by MDR1) play an important role in the absorption and metabolism of tacrolimus. Tacrolimus 127-137 ATP binding cassette subfamily B member 1 Homo sapiens 62-66 16146556-7 2005 At 1 wk, 46% of the CYP3A5*1 allele carriers had a tacrolimus concentration lower than 5 ng/mL and 77% lower than 8 ng/mL, whereas 20% of the *3/*3 patients had a concentration higher than 20 ng/mL. Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 16146556-10 2005 CONCLUSION: CYP3A5*1/*3 polymorphisms are associated with tacrolimus pharmacokinetics and dose requirements in renal transplant recipients. Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 16249748-0 2005 Sirolimus and tacrolimus trough concentrations and dose requirements after kidney transplantation in relation to CYP3A5 and MDR1 polymorphisms and steroids. Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 113-119 16249748-0 2005 Sirolimus and tacrolimus trough concentrations and dose requirements after kidney transplantation in relation to CYP3A5 and MDR1 polymorphisms and steroids. Tacrolimus 14-24 ATP binding cassette subfamily B member 1 Homo sapiens 124-128 16249748-1 2005 BACKGROUND: CYP3A5 and MDR1 polymorphisms have been shown to influence tacrolimus blood concentrations and dose requirements. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 16249748-1 2005 BACKGROUND: CYP3A5 and MDR1 polymorphisms have been shown to influence tacrolimus blood concentrations and dose requirements. Tacrolimus 71-81 ATP binding cassette subfamily B member 1 Homo sapiens 23-27 16249748-7 2005 CYP3A5 (intron 3) and MDR1 (exons 12, 21, 26) genotypes were correlated to the adjusted trough concentrations and dose requirements for both sirolimus and tacrolimus. Tacrolimus 155-165 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 16249748-7 2005 CYP3A5 (intron 3) and MDR1 (exons 12, 21, 26) genotypes were correlated to the adjusted trough concentrations and dose requirements for both sirolimus and tacrolimus. Tacrolimus 155-165 ATP binding cassette subfamily B member 1 Homo sapiens 22-26 16249748-10 2005 In the subgroup of patients treated by tacrolimus and sirolimus, adjusted tacrolimus doses were higher in patients carrying at least one CYP3A5 *1 allele (median 0.083 vs. 0.035 mg/kg for CYP3A5*3/*3 patients, P<0.05). Tacrolimus 74-84 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 137-143 16249748-10 2005 In the subgroup of patients treated by tacrolimus and sirolimus, adjusted tacrolimus doses were higher in patients carrying at least one CYP3A5 *1 allele (median 0.083 vs. 0.035 mg/kg for CYP3A5*3/*3 patients, P<0.05). Tacrolimus 74-84 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 188-194 16249748-11 2005 Adjusted-prednisolone dose and CYP3A5 polymorphism explained up to 61% of the variability in tacrolimus dose requirements. Tacrolimus 93-103 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 31-37 16181452-5 2005 OBJECTIVES: To explore the effects of FK506 on human KCs in terms of TNF-alpha secretion and to investigate the regulatory pathway involved. Tacrolimus 38-43 tumor necrosis factor Homo sapiens 69-78 16181452-11 2005 RESULTS: Our results showed that FK506 dose-dependently downregulated the secretion of TNF-alpha from UVB-irradiated KCs. Tacrolimus 33-38 tumor necrosis factor Homo sapiens 87-96 16181452-15 2005 CONCLUSIONS: Our results indicate that FK506 inhibits TNF-alpha secretion in human KCs via direct regulation of NF-kappaB. Tacrolimus 39-44 tumor necrosis factor Homo sapiens 54-63 16146556-0 2005 Influence of CYP3A5 and MDR1 polymorphisms on tacrolimus concentration in the early stage after renal transplantation. Tacrolimus 46-56 ATP binding cassette subfamily B member 1 Homo sapiens 24-28 16146556-2 2005 Cytochrome P450 (CYP) 3A and P-glycoprotein (P-gp, encoded by MDR1) play an important role in the absorption and metabolism of tacrolimus. Tacrolimus 127-137 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-24 16146556-2 2005 Cytochrome P450 (CYP) 3A and P-glycoprotein (P-gp, encoded by MDR1) play an important role in the absorption and metabolism of tacrolimus. Tacrolimus 127-137 ATP binding cassette subfamily B member 1 Homo sapiens 29-43 16179256-2 2005 Here, we show that the FK506 binding protein Fpr3 prevents premature adaptation to damage and thus serves to maintain recombination checkpoint activity. Tacrolimus 23-28 formyl peptide receptor 3 Homo sapiens 45-49 16298573-8 2005 Those who developed PTDM and were taking tacrolimus were more likely to require insulin for blood glucose control (0.39 U/kg/24 hours vs 0 U/kg/24 hours; P = .05) compared to those not on tacrolimus. Tacrolimus 41-51 insulin Homo sapiens 80-87 16172265-2 2005 We examined the effect of rapamycin and FK-506 on tissue factor (TF) expression in human aortic endothelial cells (HAECs) and vascular smooth muscle cells (HAVSMCs). Tacrolimus 40-46 coagulation factor III, tissue factor Homo sapiens 50-63 16172265-2 2005 We examined the effect of rapamycin and FK-506 on tissue factor (TF) expression in human aortic endothelial cells (HAECs) and vascular smooth muscle cells (HAVSMCs). Tacrolimus 40-46 coagulation factor III, tissue factor Homo sapiens 65-67 16046394-5 2005 Here we demonstrate that the expression of OSCAR, but not that of TREM-2, is up-regulated during osteoclastogenesis and markedly suppressed by the calcineurin inhibitor FK506, suggesting that OSCAR is transcriptionally regulated by NFATc1. Tacrolimus 169-174 osteoclast associated Ig-like receptor Homo sapiens 43-48 16046394-5 2005 Here we demonstrate that the expression of OSCAR, but not that of TREM-2, is up-regulated during osteoclastogenesis and markedly suppressed by the calcineurin inhibitor FK506, suggesting that OSCAR is transcriptionally regulated by NFATc1. Tacrolimus 169-174 osteoclast associated Ig-like receptor Homo sapiens 192-197 16046394-5 2005 Here we demonstrate that the expression of OSCAR, but not that of TREM-2, is up-regulated during osteoclastogenesis and markedly suppressed by the calcineurin inhibitor FK506, suggesting that OSCAR is transcriptionally regulated by NFATc1. Tacrolimus 169-174 nuclear factor of activated T cells 1 Homo sapiens 232-238 16172265-10 2005 FK-506 antagonized the effect of rapamycin on thrombin-induced TF expression. Tacrolimus 0-6 coagulation factor II, thrombin Homo sapiens 46-54 16172265-10 2005 FK-506 antagonized the effect of rapamycin on thrombin-induced TF expression. Tacrolimus 0-6 coagulation factor III, tissue factor Homo sapiens 63-65 15994335-6 2005 Binding of FKBP12, calcineurin, and calmodulin to TRPC6 channels is blocked by the following: 1) inhibition of PKC; 2) mutation of the PKC phosphorylation site (Ser(7168/714)) in the channels; or 3) pretreatment with FK506 or rapamycin, immunosuppressants that directly bind FKBP12. Tacrolimus 217-222 FKBP prolyl isomerase 1A Rattus norvegicus 11-17 16055081-11 2005 Furthermore, Zn2+ and the immunosuppressant FK506 showed an additive inhibitory effect on ConA-induced IL-2 mRNA expression. Tacrolimus 44-49 interleukin 2 Homo sapiens 103-107 16041804-9 2005 GFAP and COX-2 reactivity was decreased in animals treated with FK 506 compared to that in animals given MP or saline, whereas IL-1 beta expression was similarly reduced in both FK 506- and MP-treated groups. Tacrolimus 64-70 glial fibrillary acidic protein Rattus norvegicus 0-4 15994335-6 2005 Binding of FKBP12, calcineurin, and calmodulin to TRPC6 channels is blocked by the following: 1) inhibition of PKC; 2) mutation of the PKC phosphorylation site (Ser(7168/714)) in the channels; or 3) pretreatment with FK506 or rapamycin, immunosuppressants that directly bind FKBP12. Tacrolimus 217-222 calmodulin 1 Rattus norvegicus 36-46 15994335-6 2005 Binding of FKBP12, calcineurin, and calmodulin to TRPC6 channels is blocked by the following: 1) inhibition of PKC; 2) mutation of the PKC phosphorylation site (Ser(7168/714)) in the channels; or 3) pretreatment with FK506 or rapamycin, immunosuppressants that directly bind FKBP12. Tacrolimus 217-222 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 50-55 16116327-10 2005 VCAM-1 blockade on ECs partially reversed cyclosporine-induced DC adhesion (P<0.001), whereas DC adhesion under tacrolimus exposure was significantly decreased by ICAM-1 (P<0.01) and PECAM-1 (P<0.001) blockade. Tacrolimus 115-125 intercellular adhesion molecule 1 Homo sapiens 166-172 16120063-9 2005 CONCLUSIONS: Our study supplies supportive clinical evidence that inhibition of the enterohepatic cycle in case of CsA co-administration explains some of the differences observed in PK of MMF when co-administered with either TACR or CsA. Tacrolimus 225-229 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 115-118 16120133-13 2005 The supernatant from FK506-treated KC culture showed a significant increase in MMP-9 activity. Tacrolimus 21-26 matrix metallopeptidase 9 Homo sapiens 79-84 16051191-0 2005 Nur77 nuclear import and its NBRE-binding activity in thymic lymphoma cells are regulated by different mechanisms sensitive to FK506 or HA1004. Tacrolimus 127-132 nuclear receptor subfamily 4 group A member 1 Homo sapiens 0-5 16051191-3 2005 We show that Nur77 could discriminate between calcium signals sensitive to FK506 and those sensitive to HA1004, as the inhibitors differentially regulate the kinetics of Nur77 nuclear import, and FK506, unlike HA1004, inhibits Nur77 DNA-binding activity. Tacrolimus 75-80 nuclear receptor subfamily 4 group A member 1 Homo sapiens 13-18 16051191-3 2005 We show that Nur77 could discriminate between calcium signals sensitive to FK506 and those sensitive to HA1004, as the inhibitors differentially regulate the kinetics of Nur77 nuclear import, and FK506, unlike HA1004, inhibits Nur77 DNA-binding activity. Tacrolimus 196-201 nuclear receptor subfamily 4 group A member 1 Homo sapiens 13-18 16009351-6 2005 These abnormal Ca2+ events and CK release were significantly prevented by Ca2+ handling drugs, tranilast, diltiazem, and FK506. Tacrolimus 121-126 carbonic anhydrase 2 Mus musculus 15-18 16054825-12 2005 The degree of donor leukocyte chimerism and the frequency of CD4+CD25+ T-cells were higher in the tacrolimus group, however, these differences were not statistically significant. Tacrolimus 98-108 CD4 molecule Homo sapiens 61-64 16054825-12 2005 The degree of donor leukocyte chimerism and the frequency of CD4+CD25+ T-cells were higher in the tacrolimus group, however, these differences were not statistically significant. Tacrolimus 98-108 interleukin 2 receptor subunit alpha Homo sapiens 65-69 16116327-9 2005 LFA-1 blockade on DCs significantly reduced cyclosporine- and tacrolimus-induced DC adhesion (P<0.001). Tacrolimus 62-72 integrin subunit alpha L Homo sapiens 0-5 16100295-1 2005 Tacrolimus is an approved immunosuppressive agent and a known substrate for CYP3A. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 76-81 15914462-4 2005 Both proline oxidase- and p53-induced activation of NFAT were sensitive to the calcineurin inhibitors cyclosporin A and FK-506, to scavengers of ROS, and to inhibitors of calcium mobilization. Tacrolimus 120-126 tumor protein p53 Homo sapiens 26-29 16143253-0 2005 Tacrolimus: in vitro effects on myelopoiesis, apoptosis, and CD11b expression. Tacrolimus 0-10 integrin subunit alpha M Homo sapiens 61-66 16143253-9 2005 RESULTS: Tacrolimus induced a dose-dependent enhancement of clonogenesis and survival of CD34+ cells at clinically relevant doses. Tacrolimus 9-19 CD34 molecule Homo sapiens 89-93 15975916-4 2005 This calcium-dependent transcription of DSCR1.4 was inhibited by the calcineurin inhibitors cyclosporin A and FK506. Tacrolimus 110-115 regulator of calcineurin 1 Homo sapiens 40-45 16015023-0 2005 FK506 (tacrolimus) inhibition of intracellular production and enhancement of interleukin 1alpha through glucocorticoid application to chemically treated human keratinocytes. Tacrolimus 0-5 interleukin 1 alpha Homo sapiens 77-95 16015023-0 2005 FK506 (tacrolimus) inhibition of intracellular production and enhancement of interleukin 1alpha through glucocorticoid application to chemically treated human keratinocytes. Tacrolimus 7-17 interleukin 1 alpha Homo sapiens 77-95 16015023-2 2005 A study was made to confirm whether immunosuppressants such as the macrocyclic lactone FK506 (tacrolimus) possibly serve to modulate glucocorticoid- stimulated IL-1alpha production from chemically treated KCs. Tacrolimus 87-92 interleukin 1 alpha Homo sapiens 160-169 16015023-2 2005 A study was made to confirm whether immunosuppressants such as the macrocyclic lactone FK506 (tacrolimus) possibly serve to modulate glucocorticoid- stimulated IL-1alpha production from chemically treated KCs. Tacrolimus 94-104 interleukin 1 alpha Homo sapiens 160-169 16015023-3 2005 METHODS: Using cultured human KCs, the effects of FK506 on the production of IL-1alpha treated with chemicals (trinitrobenzene sulfonic acid sodium salt, TNBS) plus hydrocortisone were analyzed by ELISA and mRNA expression of IL-1alpha using RT-PCR. Tacrolimus 50-55 interleukin 1 alpha Homo sapiens 77-86 16015023-7 2005 CONCLUSIONS: FK506may control the increase in IL-1alpha with glucocorticoid in KCs, suggesting FK506 to suppress harmful effects of glucocorticoids such as steroid rosacea. Tacrolimus 13-18 interleukin 1 alpha Homo sapiens 46-55 15937899-10 2005 We predict that the E60A mutation will result in a significantly reduced affinity of FKBP12 for its ligand FK506. Tacrolimus 107-112 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 85-91 15996247-8 2005 Pre-transplant donor-directed IFN-gamma ELISPOT assessment of anti-donor cellular immunity may function as a "cellular crossmatch" and independently correlates with renal allograft function in African Americans receiving tacrolimus- and sirolimus-based immunosuppression. Tacrolimus 221-231 interferon gamma Homo sapiens 30-39 16042540-5 2005 Inspired by strategies used in Nature and optimized over millions of years of evolution, we have created a bifunctional molecule [SLF (synthetic ligand for FK506-binding protein)-CR (Congo Red)] that is able to block Abeta (amyloid beta) aggregation by borrowing the surface and steric bulk of a cellular chaperone. Tacrolimus 156-161 amyloid beta precursor protein Homo sapiens 217-222 16042540-5 2005 Inspired by strategies used in Nature and optimized over millions of years of evolution, we have created a bifunctional molecule [SLF (synthetic ligand for FK506-binding protein)-CR (Congo Red)] that is able to block Abeta (amyloid beta) aggregation by borrowing the surface and steric bulk of a cellular chaperone. Tacrolimus 156-161 amyloid beta precursor protein Homo sapiens 224-236 21162213-9 2005 (3) FK506 reduced significantly production of CD40L in spontaneous and PMA/Ionomycin-induced PBMC of LN. Tacrolimus 4-9 CD40 ligand Homo sapiens 46-51 15951320-11 2005 The importance of CYP3A5 status for tacrolimus clearance is also dependent on the concomitant CYP3A4 activity. Tacrolimus 36-46 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 94-100 16109516-9 2005 Aminoguanidine and FK506 can inhibit the expression of iNOS and palliate acute rejection. Tacrolimus 19-24 nitric oxide synthase 2 Rattus norvegicus 55-59 15998421-0 2005 Transplant immunosuppressive agents in non-transplant chronic autoimmune hepatitis: the Canadian association for the study of liver (CASL) experience with mycophenolate mofetil and tacrolimus. Tacrolimus 181-191 neural precursor cell expressed, developmentally down-regulated 9 Homo sapiens 133-137 15998670-9 2005 Treatment of Fgl-2-/- xenografts with mycophenolate mofetil and tacrolimus, a clinically relevant immune suppression protocol, led to long-term graft acceptance. Tacrolimus 64-74 fibrinogen like 2 Homo sapiens 13-18 15937522-1 2005 Based on recent evidence that renal cyclooxygenase-2 (COX-2) gene expression is suppressed by immunosuppressive agents such as cyclosporin A (CsA), tacrolimus and dexamethasone, this study aimed to characterize the effect of the new immunosuppressant everolimus on COX-2 expression in the rat kidney. Tacrolimus 148-158 prostaglandin-endoperoxide synthase 2 Rattus norvegicus 36-52 15937522-1 2005 Based on recent evidence that renal cyclooxygenase-2 (COX-2) gene expression is suppressed by immunosuppressive agents such as cyclosporin A (CsA), tacrolimus and dexamethasone, this study aimed to characterize the effect of the new immunosuppressant everolimus on COX-2 expression in the rat kidney. Tacrolimus 148-158 prostaglandin-endoperoxide synthase 2 Rattus norvegicus 54-59 15937522-1 2005 Based on recent evidence that renal cyclooxygenase-2 (COX-2) gene expression is suppressed by immunosuppressive agents such as cyclosporin A (CsA), tacrolimus and dexamethasone, this study aimed to characterize the effect of the new immunosuppressant everolimus on COX-2 expression in the rat kidney. Tacrolimus 148-158 prostaglandin-endoperoxide synthase 2 Rattus norvegicus 265-270 15951320-0 2005 Contribution of CYP3A5 to the in vitro hepatic clearance of tacrolimus. Tacrolimus 60-70 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 16-22 15951320-1 2005 BACKGROUND: Tacrolimus is metabolized predominantly to 13-O-demethyltacrolimus in the liver and intestine by cytochrome P450 3A (CYP3A). Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 109-127 15951320-1 2005 BACKGROUND: Tacrolimus is metabolized predominantly to 13-O-demethyltacrolimus in the liver and intestine by cytochrome P450 3A (CYP3A). Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 129-134 15951320-2 2005 Patients with high concentrations of CYP3A5, a CYP3A isoenzyme polymorphically produced in these organs, require higher doses of tacrolimus, but the exact mechanism of this association is unknown. Tacrolimus 129-139 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 15951320-2 2005 Patients with high concentrations of CYP3A5, a CYP3A isoenzyme polymorphically produced in these organs, require higher doses of tacrolimus, but the exact mechanism of this association is unknown. Tacrolimus 129-139 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-42 15951320-3 2005 METHODS: cDNA-expressed CYP3A enzymes and a bank of human liver microsomes with known CYP3A4 and CYP3A5 content were used to investigate the contribution of CYP3A5 to the metabolism of tacrolimus to 13-O-demethyltacrolimus as quantified by liquid chromatography-tandem mass spectrometry. Tacrolimus 185-195 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 157-163 15951320-6 2005 Recombinant CYP3A5 metabolized tacrolimus with a catalytic efficiency (V(max)/K(m)) that was 64% higher than that of CYP3A4. Tacrolimus 31-41 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 15951320-7 2005 The contribution of CYP3A5 to 13-O-demethylation of tacrolimus in human liver microsomes varied from 1.5% to 40% (median, 18.8%). Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 15951320-10 2005 CONCLUSIONS: CYP3A5 affects metabolism of tacrolimus, thus explaining the association between CYP3A5 genotype and tacrolimus dosage. Tacrolimus 42-52 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 15951320-10 2005 CONCLUSIONS: CYP3A5 affects metabolism of tacrolimus, thus explaining the association between CYP3A5 genotype and tacrolimus dosage. Tacrolimus 42-52 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 94-100 15951320-10 2005 CONCLUSIONS: CYP3A5 affects metabolism of tacrolimus, thus explaining the association between CYP3A5 genotype and tacrolimus dosage. Tacrolimus 114-124 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 15951320-10 2005 CONCLUSIONS: CYP3A5 affects metabolism of tacrolimus, thus explaining the association between CYP3A5 genotype and tacrolimus dosage. Tacrolimus 114-124 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 94-100 15951320-11 2005 The importance of CYP3A5 status for tacrolimus clearance is also dependent on the concomitant CYP3A4 activity. Tacrolimus 36-46 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 18-24 15953564-6 2005 The stimulation by bupleuran 2IIc/PG-1 of cyclin D2 expression was significantly decreased by inhibitors, PI 3-kinase (LY294002 and Wortmannin), PLCgamma (U73122), PKC (H-7), receptor-operated calcium entry inhibitor (SK&F 96365), and calcineurin (FK506). Tacrolimus 252-257 cyclin D2 Mus musculus 42-51 16003241-7 2005 The CNI Cyclosporin and Tacrolimus, and alphaCD25 mAb inhibited in vitro induced FOXP3 gene transcription (range 70%-90%), whereas Rapa did not inhibit the induction. Tacrolimus 24-34 forkhead box P3 Homo sapiens 81-86 21162213-11 2005 Through inhibiting CaN activity, FK506 may prevent abnormal activation of CD40-CD40L costimulatory pathway in lupus nephritis. Tacrolimus 33-38 CD40 molecule Homo sapiens 74-78 21162213-11 2005 Through inhibiting CaN activity, FK506 may prevent abnormal activation of CD40-CD40L costimulatory pathway in lupus nephritis. Tacrolimus 33-38 CD40 ligand Homo sapiens 79-84 15955561-1 2005 FK506 and rapamycin are immunosuppressant drugs that disrupt the interaction of FK506-binding proteins (FKBPs) with ryanodine receptors (RyR1), which form homotetrameric Ca2+ release channels in the sarcoplasmic reticulum (SR) of skeletal muscle. Tacrolimus 0-5 ryanodine receptor 1 Homo sapiens 137-141 15955561-1 2005 FK506 and rapamycin are immunosuppressant drugs that disrupt the interaction of FK506-binding proteins (FKBPs) with ryanodine receptors (RyR1), which form homotetrameric Ca2+ release channels in the sarcoplasmic reticulum (SR) of skeletal muscle. Tacrolimus 80-85 ryanodine receptor 1 Homo sapiens 137-141 16267794-6 2005 Current therapy tends to reduce steroid treatment doses and, optimizing induction therapy with IL-2R inhibitors, using tacrolimus or mycophenolate or sirolimus. Tacrolimus 119-129 interleukin 2 receptor subunit alpha Homo sapiens 95-100 15879595-6 2005 TCR-induced IL-21 gene expression was inhibited by cyclosporin A and FK506. Tacrolimus 69-74 interleukin 21 Homo sapiens 12-17 15914333-8 2005 These results suggest that FK506 suppresses inflammation by inhibiting PGE(2) production from synovial cells through suppression of IL-1beta production from leukocytes. Tacrolimus 27-32 interleukin 1 beta Homo sapiens 132-140 16008701-15 2005 Topical tacrolimus is a promising alternative to topical CsA for treatment of KCS and may be beneficial in patients with less than optimal response to topical CsA. Tacrolimus 8-18 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 159-162 15982605-7 2005 There were no changes in high-density lipoprotein cholesterol and triglyceride levels, but apolipoprotein B therapy was reduced in tacrolimus-converted vs cyclosporine-maintained patients (p=0.0003). Tacrolimus 131-141 apolipoprotein B Homo sapiens 91-107 15982605-10 2005 CONCLUSIONS: Conversion from cyclosporine microemulsion- to tacrolimus-based immunoprophylaxis resulted in decreased cholesterol, apolipoprotein B, urea, creatinine, and uric acid without any clinically evident perturbation of glucose metabolism in stable heart transplant recipients with treated but persistent mild dyslipidemia. Tacrolimus 60-70 apolipoprotein B Homo sapiens 130-146 16113783-0 2005 Modulation of tissue factor expression by rapamycin and FK-506 in lipopolysaccharide-stimulated human mononuclear cells and serum-stimulated aortic smooth muscle cells. Tacrolimus 56-62 coagulation factor III, tissue factor Homo sapiens 14-27 16113783-4 2005 The aim of this study was to determine the effect of an immunosuppressant macrolide, rapamycin (Sirolimus), on the expression of TF and its inhibitor (TFPI) by monocytic cells (human blood mononuclear and THP-1 cells) and human aortic smooth muscle cells, in comparison with FK-506 and azithromycin. Tacrolimus 275-281 coagulation factor III, tissue factor Homo sapiens 129-131 16113783-4 2005 The aim of this study was to determine the effect of an immunosuppressant macrolide, rapamycin (Sirolimus), on the expression of TF and its inhibitor (TFPI) by monocytic cells (human blood mononuclear and THP-1 cells) and human aortic smooth muscle cells, in comparison with FK-506 and azithromycin. Tacrolimus 275-281 tissue factor pathway inhibitor Homo sapiens 151-155 16113783-5 2005 In monocytic cells, rapamycin and FK-506 inhibited LPS-induced TF activity, antigen and mRNA expression through a transcriptional mechanism involving NF-kappaB. Tacrolimus 34-40 coagulation factor III, tissue factor Homo sapiens 63-65 16113783-6 2005 In smooth muscle cells, rapamycin and azithromycin had no effect on serum-induced TF expression, while FK-506 increased serum-induced TF protein and mRNA expression. Tacrolimus 103-109 coagulation factor III, tissue factor Homo sapiens 134-136 16002550-0 2005 Short-term treatment with mycophenolic acid and tacrolimus is tolerogenic for INS-1 cell clone transplantation and the deleterious effects of the drugs are limited: in vivo and in vitro studies. Tacrolimus 48-58 insulin 1 Rattus norvegicus 78-83 16119834-6 2005 Plasma exchange and subsequent immunomodulating therapy with corticosteroids and tacrolimus showed a dramatic clinical improvement with a marked decline of MuSK Ab level in the serum. Tacrolimus 81-91 muscle associated receptor tyrosine kinase Homo sapiens 156-160 16182764-2 2005 Standard primary immunosuppressive therapy after orthotopic liver transplantation (OLT) is based on a calcineurin-inhibitor (CNI): cyclosporine or tacrolimus. Tacrolimus 147-157 calcineurin binding protein 1 Homo sapiens 125-128 15948978-10 2005 CONCLUSIONS: Topical tacrolimus reduces the number of eosinophils in tissue and suppresses the expression of eotaxin, CCR3, RANTES and IL-5 related to proliferation, recruitment, activation and survival of eosinophils. Tacrolimus 21-31 interleukin 5 Homo sapiens 135-139 15888024-0 2005 Low-dose FK506 blocks collar-induced atherosclerotic plaque development and stabilizes plaques in ApoE-/- mice. Tacrolimus 9-14 apolipoprotein E Mus musculus 98-102 15888024-2 2005 ApoE-/- mice were treated for 4 weeks with the immunosuppressive drug FK506 (0.05 mg/kg/day), yielding sustained blood levels (approximately 0.2 ng/mL) without systemic side effects. Tacrolimus 70-75 apolipoprotein E Mus musculus 0-4 15888024-10 2005 Low-dose FK506 inhibits collar-induced atherosclerotic plaque development and progression and induces more stable plaque phenotypes in ApoE-/- mice without any peripheral side effects. Tacrolimus 9-14 apolipoprotein E Mus musculus 135-139 15890490-0 2005 Differential effect of Cyclosporin A and FK506 on SPARC mRNA expression by human gingival fibroblasts. Tacrolimus 41-46 secreted protein acidic and cysteine rich Homo sapiens 50-55 15890490-6 2005 Our aim was, therefore, to analyze the effect of CsA and FK506 on SPARC gene expression. Tacrolimus 57-62 secreted protein acidic and cysteine rich Homo sapiens 66-71 15890490-7 2005 METHODS: Cultured human gingival fibroblasts were incubated with CsA, FK506 or with their vehicle (VH) for 24, 48 and 72 h. SPARC gene expression was determined by RT-PCR. Tacrolimus 70-75 secreted protein acidic and cysteine rich Homo sapiens 124-129 15948978-0 2005 Tacrolimus decreases the expression of eotaxin, CCR3, RANTES and interleukin-5 in atopic dermatitis. Tacrolimus 0-10 C-C motif chemokine ligand 11 Homo sapiens 39-46 15948978-0 2005 Tacrolimus decreases the expression of eotaxin, CCR3, RANTES and interleukin-5 in atopic dermatitis. Tacrolimus 0-10 C-C motif chemokine receptor 3 Homo sapiens 48-52 15948978-0 2005 Tacrolimus decreases the expression of eotaxin, CCR3, RANTES and interleukin-5 in atopic dermatitis. Tacrolimus 0-10 C-C motif chemokine ligand 5 Homo sapiens 54-60 15948978-0 2005 Tacrolimus decreases the expression of eotaxin, CCR3, RANTES and interleukin-5 in atopic dermatitis. Tacrolimus 0-10 interleukin 5 Homo sapiens 65-78 15939815-5 2005 Overexpression of atrial and brain natriuretic peptides, collagen, and fibronectin mRNAs in GCA-KO mice was also attenuated by FK506. Tacrolimus 127-132 fibronectin 1 Mus musculus 71-82 15821752-9 2005 ET-1 in the presence of FK506 did not restore calcineurin activity (P=0.1). Tacrolimus 24-29 endothelin 1 Homo sapiens 0-4 15948987-9 2005 Both RT-PCR and immunohistochemical staining of sections from ear biopsies demonstrated that nanocrystalline silver, tacrolimus and steroid significantly suppressed the expression of tumour necrosis factor (TNF)-alpha and interleukin (IL)-12. Tacrolimus 117-127 tumor necrosis factor Mus musculus 183-217 15948978-8 2005 RESULTS: After treatment with topical tacrolimus twice daily for 8 weeks, significant decreases were found in serum IL-5 levels, immunoreactive cell counts of eotaxin, IL-5, CCR3 and RANTES in AD skin, and tissue eosinophil counts. Tacrolimus 38-48 interleukin 5 Homo sapiens 116-120 15899924-2 2005 Immunosuppressive drugs, such as FK506 and cyclosporin A, block the priming of alloreactive CD4 T(h) cells and the subsequent induction of allospecific CD8 cytotoxic effector T cells and inhibit allograft rejection. Tacrolimus 33-38 CD4 molecule Homo sapiens 92-95 15948978-8 2005 RESULTS: After treatment with topical tacrolimus twice daily for 8 weeks, significant decreases were found in serum IL-5 levels, immunoreactive cell counts of eotaxin, IL-5, CCR3 and RANTES in AD skin, and tissue eosinophil counts. Tacrolimus 38-48 C-C motif chemokine ligand 11 Homo sapiens 159-166 15948978-8 2005 RESULTS: After treatment with topical tacrolimus twice daily for 8 weeks, significant decreases were found in serum IL-5 levels, immunoreactive cell counts of eotaxin, IL-5, CCR3 and RANTES in AD skin, and tissue eosinophil counts. Tacrolimus 38-48 interleukin 5 Homo sapiens 168-172 15948978-8 2005 RESULTS: After treatment with topical tacrolimus twice daily for 8 weeks, significant decreases were found in serum IL-5 levels, immunoreactive cell counts of eotaxin, IL-5, CCR3 and RANTES in AD skin, and tissue eosinophil counts. Tacrolimus 38-48 C-C motif chemokine receptor 3 Homo sapiens 174-178 15948978-8 2005 RESULTS: After treatment with topical tacrolimus twice daily for 8 weeks, significant decreases were found in serum IL-5 levels, immunoreactive cell counts of eotaxin, IL-5, CCR3 and RANTES in AD skin, and tissue eosinophil counts. Tacrolimus 38-48 C-C motif chemokine ligand 5 Homo sapiens 183-189 15948978-10 2005 CONCLUSIONS: Topical tacrolimus reduces the number of eosinophils in tissue and suppresses the expression of eotaxin, CCR3, RANTES and IL-5 related to proliferation, recruitment, activation and survival of eosinophils. Tacrolimus 21-31 C-C motif chemokine ligand 11 Homo sapiens 109-116 15948978-10 2005 CONCLUSIONS: Topical tacrolimus reduces the number of eosinophils in tissue and suppresses the expression of eotaxin, CCR3, RANTES and IL-5 related to proliferation, recruitment, activation and survival of eosinophils. Tacrolimus 21-31 C-C motif chemokine receptor 3 Homo sapiens 118-122 15948978-10 2005 CONCLUSIONS: Topical tacrolimus reduces the number of eosinophils in tissue and suppresses the expression of eotaxin, CCR3, RANTES and IL-5 related to proliferation, recruitment, activation and survival of eosinophils. Tacrolimus 21-31 C-C motif chemokine ligand 5 Homo sapiens 124-130 15829415-8 2005 It is worth noting that TNFR-Ig or prednisolone, which is effective for treatment of patients with severe-fulminant Crohn"s disease, markedly attenuated pathological clinical indices in this colitis model, whereas the immunosuppressive agents, azathioprine, tacrolimus, and cyclosporine A produced no significant effect. Tacrolimus 258-268 TNF receptor superfamily member 1A Homo sapiens 24-28 15899924-2 2005 Immunosuppressive drugs, such as FK506 and cyclosporin A, block the priming of alloreactive CD4 T(h) cells and the subsequent induction of allospecific CD8 cytotoxic effector T cells and inhibit allograft rejection. Tacrolimus 33-38 CD8a molecule Homo sapiens 152-155 15857611-8 2005 These results indicate that the induction of SNK mRNA by thapsigargin in SH-SY5Y cells is regulated by FK506 via an inhibition of calcineurin at the transcriptional stage, and the transcriptional regulation of the SNK gene by FK506 was well correlated with the protective effect of the compound against apoptosis. Tacrolimus 103-108 polo like kinase 2 Homo sapiens 45-48 15910387-1 2005 We encountered two cases of pediatric living-related liver transplant recipients who showed increases in blood concentration of cyclosporine or tacrolimus, a dual substrate for cytochrome P450 (CYP) 3A and P-glycoprotein (P-gp), during a diarrheal episode. Tacrolimus 144-154 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 177-201 15910387-1 2005 We encountered two cases of pediatric living-related liver transplant recipients who showed increases in blood concentration of cyclosporine or tacrolimus, a dual substrate for cytochrome P450 (CYP) 3A and P-glycoprotein (P-gp), during a diarrheal episode. Tacrolimus 144-154 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 206-220 15910387-1 2005 We encountered two cases of pediatric living-related liver transplant recipients who showed increases in blood concentration of cyclosporine or tacrolimus, a dual substrate for cytochrome P450 (CYP) 3A and P-glycoprotein (P-gp), during a diarrheal episode. Tacrolimus 144-154 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 222-226 15910387-8 2005 These findings suggest that the suppression of CYP3A and P-gp activities may be involved in the mechanism of elevated blood concentrations of cyclosporine and tacrolimus during enteritis-induced diarrhea. Tacrolimus 159-169 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 47-52 15910387-8 2005 These findings suggest that the suppression of CYP3A and P-gp activities may be involved in the mechanism of elevated blood concentrations of cyclosporine and tacrolimus during enteritis-induced diarrhea. Tacrolimus 159-169 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 57-61 15857611-0 2005 Protective effect of FK506 against apoptosis of SH-SY5Y cells correlates with regulation of the serum inducible kinase gene. Tacrolimus 21-26 polo like kinase 2 Homo sapiens 96-118 15857611-2 2005 The purpose of this paper was to investigate a possible correlation between the protective effect of FK506 against apoptosis and the regulation of the serum inducible kinase (SNK) and fibroblast growth factor inducible kinase (FNK) genes-which are polo-like kinases expressed abundantly in the brain by FK506. Tacrolimus 101-106 polo like kinase 2 Homo sapiens 151-173 15857611-2 2005 The purpose of this paper was to investigate a possible correlation between the protective effect of FK506 against apoptosis and the regulation of the serum inducible kinase (SNK) and fibroblast growth factor inducible kinase (FNK) genes-which are polo-like kinases expressed abundantly in the brain by FK506. Tacrolimus 101-106 polo like kinase 2 Homo sapiens 175-178 15857611-8 2005 These results indicate that the induction of SNK mRNA by thapsigargin in SH-SY5Y cells is regulated by FK506 via an inhibition of calcineurin at the transcriptional stage, and the transcriptional regulation of the SNK gene by FK506 was well correlated with the protective effect of the compound against apoptosis. Tacrolimus 226-231 polo like kinase 2 Homo sapiens 45-48 15857611-2 2005 The purpose of this paper was to investigate a possible correlation between the protective effect of FK506 against apoptosis and the regulation of the serum inducible kinase (SNK) and fibroblast growth factor inducible kinase (FNK) genes-which are polo-like kinases expressed abundantly in the brain by FK506. Tacrolimus 101-106 polo like kinase 3 Homo sapiens 227-230 15857611-8 2005 These results indicate that the induction of SNK mRNA by thapsigargin in SH-SY5Y cells is regulated by FK506 via an inhibition of calcineurin at the transcriptional stage, and the transcriptional regulation of the SNK gene by FK506 was well correlated with the protective effect of the compound against apoptosis. Tacrolimus 226-231 polo like kinase 2 Homo sapiens 214-217 15857611-2 2005 The purpose of this paper was to investigate a possible correlation between the protective effect of FK506 against apoptosis and the regulation of the serum inducible kinase (SNK) and fibroblast growth factor inducible kinase (FNK) genes-which are polo-like kinases expressed abundantly in the brain by FK506. Tacrolimus 303-308 polo like kinase 2 Homo sapiens 151-173 15860218-0 2005 FK506 suppresses the stimulation of matrix metalloproteinase 13 synthesis by interleukin-1beta in rheumatoid synovial fibroblasts. Tacrolimus 0-5 matrix metallopeptidase 13 Homo sapiens 36-63 15857611-2 2005 The purpose of this paper was to investigate a possible correlation between the protective effect of FK506 against apoptosis and the regulation of the serum inducible kinase (SNK) and fibroblast growth factor inducible kinase (FNK) genes-which are polo-like kinases expressed abundantly in the brain by FK506. Tacrolimus 303-308 polo like kinase 2 Homo sapiens 175-178 15857611-2 2005 The purpose of this paper was to investigate a possible correlation between the protective effect of FK506 against apoptosis and the regulation of the serum inducible kinase (SNK) and fibroblast growth factor inducible kinase (FNK) genes-which are polo-like kinases expressed abundantly in the brain by FK506. Tacrolimus 303-308 polo like kinase 3 Homo sapiens 227-230 15857611-4 2005 FK506 inhibited the increase in SNK mRNA but not FNK mRNA. Tacrolimus 0-5 polo like kinase 2 Homo sapiens 32-35 15857611-5 2005 Deletion analysis of the SNK promoter showed that the promoter site, which was regulated by thapsigargin and FK506 in a calcineurin-dependent manner, is a cAMP response element (CRE)/activating transcription factor (ATF)-like element located 84 base pairs (bp) proximal to the transcriptional initiation site. Tacrolimus 109-114 polo like kinase 2 Homo sapiens 25-28 15860218-0 2005 FK506 suppresses the stimulation of matrix metalloproteinase 13 synthesis by interleukin-1beta in rheumatoid synovial fibroblasts. Tacrolimus 0-5 interleukin 1 beta Homo sapiens 77-94 15857611-5 2005 Deletion analysis of the SNK promoter showed that the promoter site, which was regulated by thapsigargin and FK506 in a calcineurin-dependent manner, is a cAMP response element (CRE)/activating transcription factor (ATF)-like element located 84 base pairs (bp) proximal to the transcriptional initiation site. Tacrolimus 109-114 glial cell derived neurotrophic factor Homo sapiens 216-219 15860218-1 2005 The aim of this study was to determine whether FK506, which has been shown to be effective for the treatment of refractory RA, affects the synthesis of matrix metalloproteinases (MMPs) in rheumatoid synovial fibroblasts. Tacrolimus 47-52 matrix metallopeptidase 2 Homo sapiens 179-183 15860218-4 2005 In addition, synovial fibroblasts pretreated with FK506 were stimulated with IL-1beta for 10 min and cellular lysates were subjected to anti-phospho-specific mitogen-activated protein kinase (MAPK). Tacrolimus 50-55 interleukin 1 beta Homo sapiens 77-85 15860218-8 2005 FK506, however, significantly suppressed MMP-13 production from IL-1beta-stimulated synovial fibroblasts. Tacrolimus 0-5 matrix metallopeptidase 13 Homo sapiens 41-47 15860218-8 2005 FK506, however, significantly suppressed MMP-13 production from IL-1beta-stimulated synovial fibroblasts. Tacrolimus 0-5 interleukin 1 beta Homo sapiens 64-72 15860218-9 2005 FK506 also prevented IL-1beta-stimulated JNK activation and transcriptional activation of AP-1 in these cells. Tacrolimus 0-5 interleukin 1 beta Homo sapiens 21-29 15860218-9 2005 FK506 also prevented IL-1beta-stimulated JNK activation and transcriptional activation of AP-1 in these cells. Tacrolimus 0-5 mitogen-activated protein kinase 8 Homo sapiens 41-44 15860218-10 2005 Our results indicate that FK506 is capable of regulating MMP-13 synthesis via JNK pathway in rheumatoid synonvium. Tacrolimus 26-31 matrix metallopeptidase 13 Homo sapiens 57-63 15860218-10 2005 Our results indicate that FK506 is capable of regulating MMP-13 synthesis via JNK pathway in rheumatoid synonvium. Tacrolimus 26-31 mitogen-activated protein kinase 8 Homo sapiens 78-81 15716327-0 2005 Concentrations of cyclosporin A and FK506 that inhibit IL-2 induction in human T cells do not affect TGF-beta1 biosynthesis, whereas higher doses of cyclosporin A trigger apoptosis and release of preformed TGF-beta1. Tacrolimus 36-41 interleukin 2 Homo sapiens 55-59 15854665-5 2005 RESULTS: Tacrolimus reduced TGF-beta mRNA expression below control levels and treatment with pirfenidone at all doses did not alter this effect. Tacrolimus 9-19 transforming growth factor, beta 1 Rattus norvegicus 28-36 15854665-6 2005 Likewise, TIMP-1 mRNA expression was depressed by the addition of tacrolimus and pirfenidone caused a further decrease in expression. Tacrolimus 66-76 TIMP metallopeptidase inhibitor 1 Rattus norvegicus 10-16 15888147-6 2005 The addition of retinoids or corticosteroids to the cell culture inhibited the UVB-induced suppression of both ATP2A2 and ATP2C1 mRNA levels, and UVB-induced suppression of ATP2C1 mRNA was also inhibited by the addition of ciclosporin, tacrolimus and vitamin D(3). Tacrolimus 236-246 ATPase secretory pathway Ca2+ transporting 1 Homo sapiens 173-179 15862175-3 2005 We and others have shown that caspase 9 is cleaved in the retina in other injury models and we hypothesized that the neuroprotection observed with FK506 was mediated by interference with caspase 9 activation. Tacrolimus 147-152 caspase 9 Rattus norvegicus 30-39 15862175-3 2005 We and others have shown that caspase 9 is cleaved in the retina in other injury models and we hypothesized that the neuroprotection observed with FK506 was mediated by interference with caspase 9 activation. Tacrolimus 147-152 caspase 9 Rattus norvegicus 187-196 15862175-8 2005 Furthermore, the oral administration of FK506 5 mg kg(-1) day(-1) blocks the cleavage of caspase 9 at both time points. Tacrolimus 40-45 caspase 9 Rattus norvegicus 89-98 15862175-9 2005 These data suggest that caspase 9 activation may play an important role in retinal ganglion cell death following optic nerve crush and that the neuroprotection seen with FK506 may be mediated by interfering with the activation of caspase 9. Tacrolimus 170-175 caspase 9 Rattus norvegicus 230-239 15802130-0 2005 Equilibrium studies of a fluorescent tacrolimus binding to surfactant protein A. Tacrolimus 37-47 surfactant protein A1 Homo sapiens 59-79 15802130-2 2005 The objective of this study was to characterize the binding of FK506 to surfactant protein A (SP-A), an abundant lipoprotein found in the alveolar fluid that functions as part of the innate immune system in the lung. Tacrolimus 63-68 surfactant protein A1 Homo sapiens 72-92 15802130-2 2005 The objective of this study was to characterize the binding of FK506 to surfactant protein A (SP-A), an abundant lipoprotein found in the alveolar fluid that functions as part of the innate immune system in the lung. Tacrolimus 63-68 surfactant protein A1 Homo sapiens 94-98 15802130-4 2005 Using the fluorescence and anisotropy properties of DNS-FK, we demonstrated that tacrolimus avidly binds to SP-A with an apparent equilibrium association constant (K(app)) of 10(7)M(-1) and a Gibbs binding free energy of -40 kJ mol(-1)K(-1). Tacrolimus 81-91 surfactant protein A1 Homo sapiens 108-112 15376236-5 2005 By using either the CaM kinase II inhibitor KN-62, the calcineurin inhibitor FK 506 or the protein kinase A (PKA) inhibitor Rp-8-Br-cAMPS, the GIP-mediated Galphai2 mRNA increase was fully reversed. Tacrolimus 77-83 gastric inhibitory polypeptide Rattus norvegicus 143-146 15376236-5 2005 By using either the CaM kinase II inhibitor KN-62, the calcineurin inhibitor FK 506 or the protein kinase A (PKA) inhibitor Rp-8-Br-cAMPS, the GIP-mediated Galphai2 mRNA increase was fully reversed. Tacrolimus 77-83 G protein subunit alpha i2 Rattus norvegicus 156-164 15716327-1 2005 Cyclosporin A (CsA) and FK506 suppress T cell activation by inhibiting calcineurin and the calcineurin-dependent transcription factors nuclear factor of activated T cells (NFATc), which are central regulators of T cell function. Tacrolimus 24-29 nuclear factor of activated T cells 1 Homo sapiens 172-177 15716327-4 2005 In Jurkat T cells, the TGF-beta1 promoter was activated by calcineurin and NFATc and inhibited by CsA and FK506. Tacrolimus 106-111 transforming growth factor beta 1 Homo sapiens 23-32 15716327-7 2005 However, pretreatment of fresh lymphocytes with CsA or FK506 during primary TCR stimulation reduced their production of TGF-beta1 during secondary TCR activation. Tacrolimus 55-60 transforming growth factor beta 1 Homo sapiens 120-129 15716327-10 2005 Our results indicate that CsA and FK506 are not general inducers of TGF-beta1 biosynthesis but can cause different effects on TGF-beta1 depending on the cell type and concentrations used. Tacrolimus 34-39 transforming growth factor beta 1 Homo sapiens 68-77 15716327-10 2005 Our results indicate that CsA and FK506 are not general inducers of TGF-beta1 biosynthesis but can cause different effects on TGF-beta1 depending on the cell type and concentrations used. Tacrolimus 34-39 transforming growth factor beta 1 Homo sapiens 126-135 15919446-0 2005 Initial dosage adjustment for oral administration of tacrolimus using the intestinal MDR1 level in living-donor liver transplant recipients. Tacrolimus 53-63 ATP binding cassette subfamily B member 1 Homo sapiens 85-89 15919497-5 2005 After switching to tacrolimus, kidney function stabilized and even improved (creatinine: baseline after switching 2.9 mg/dL; delta(Tac) = -0.7 mg/dL; GFR: delta(Tac) = 14 mL/min). Tacrolimus 19-29 Rap guanine nucleotide exchange factor 5 Homo sapiens 150-153 15919446-3 2005 For 7 days postoperatively, good inverse correlation was found between the tacrolimus concentration/dose (C/D) ratio and the intestinal mRNA level of MDR1 (r = -0.776), but not of CYP3A4 (r = -0.096), in the 46 cases. Tacrolimus 75-85 ATP binding cassette subfamily B member 1 Homo sapiens 150-154 15919446-4 2005 After classifying the patients according to median of the intestinal MDR1 mRNA expression, the oral dose of tacrolimus in the high-MDR1 group was approximately twofold higher than in the low-MDR1 group (P < .001), whereas its trough level was similar between the two groups. Tacrolimus 108-118 ATP binding cassette subfamily B member 1 Homo sapiens 69-73 15919446-4 2005 After classifying the patients according to median of the intestinal MDR1 mRNA expression, the oral dose of tacrolimus in the high-MDR1 group was approximately twofold higher than in the low-MDR1 group (P < .001), whereas its trough level was similar between the two groups. Tacrolimus 108-118 ATP binding cassette subfamily B member 1 Homo sapiens 131-135 15919446-4 2005 After classifying the patients according to median of the intestinal MDR1 mRNA expression, the oral dose of tacrolimus in the high-MDR1 group was approximately twofold higher than in the low-MDR1 group (P < .001), whereas its trough level was similar between the two groups. Tacrolimus 108-118 ATP binding cassette subfamily B member 1 Homo sapiens 131-135 15919446-5 2005 In addition, the correlation between the intestinal MDR1 mRNA level and the tacrolimus C/D ratio was confirmed with a larger population (r = -0.645, n = 104). Tacrolimus 76-86 ATP binding cassette subfamily B member 1 Homo sapiens 52-56 15919446-6 2005 Using the regression line between the intestinal MDR1 mRNA level and tacrolimus C/D ratio, we could prospectively predict the individual C/D ratio of tacrolimus immediately after LDLT. Tacrolimus 69-79 ATP binding cassette subfamily B member 1 Homo sapiens 49-53 15919446-6 2005 Using the regression line between the intestinal MDR1 mRNA level and tacrolimus C/D ratio, we could prospectively predict the individual C/D ratio of tacrolimus immediately after LDLT. Tacrolimus 150-160 ATP binding cassette subfamily B member 1 Homo sapiens 49-53 15919508-0 2005 FK506 donor pretreatment improves intestinal graft microcirculation and morphology by concurrent inhibition of early NF-kappaB activation and augmented HSP72 synthesis. Tacrolimus 0-5 heat shock protein family A (Hsp70) member 1A Homo sapiens 152-157 15919508-15 2005 FK506 alleviated reperfusion injury by blocking NF-kappaB activation and ICAM-1 transcription, thus decreasing endothelial activation and improving the microcirculation. Tacrolimus 0-5 intercellular adhesion molecule 1 Homo sapiens 73-79 15919519-8 2005 CsA, MMF, and AS-ODNs inhibited MD-1 expression and lymphocyte proliferation, as well as decreased serum level of IL-2 and increased that of IL-10; FK506, treatment showed all the effects mentioned above but up-regulated the IL-10 level; SRL had no significant influence on either MD-1 expression or IL-2 and IL-10 level, although it equally suppressed the proliferation (P < .05 vs controls). Tacrolimus 148-153 interleukin 2 Mus musculus 114-118 15919519-8 2005 CsA, MMF, and AS-ODNs inhibited MD-1 expression and lymphocyte proliferation, as well as decreased serum level of IL-2 and increased that of IL-10; FK506, treatment showed all the effects mentioned above but up-regulated the IL-10 level; SRL had no significant influence on either MD-1 expression or IL-2 and IL-10 level, although it equally suppressed the proliferation (P < .05 vs controls). Tacrolimus 148-153 interleukin 10 Mus musculus 141-146 15919519-8 2005 CsA, MMF, and AS-ODNs inhibited MD-1 expression and lymphocyte proliferation, as well as decreased serum level of IL-2 and increased that of IL-10; FK506, treatment showed all the effects mentioned above but up-regulated the IL-10 level; SRL had no significant influence on either MD-1 expression or IL-2 and IL-10 level, although it equally suppressed the proliferation (P < .05 vs controls). Tacrolimus 148-153 interleukin 10 Mus musculus 225-230 15919446-1 2005 The role of intestinal P-glycoprotein (encoded by the MDR1/ABCB1 gene) and/or metabolic enzyme CYP3A4 for tacrolimus therapy was examined in recipients of living-donor liver transplantation (LDLT), under the hypothesis that these proteins are factors for pharmacokinetic variability. Tacrolimus 106-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 95-101 15919519-8 2005 CsA, MMF, and AS-ODNs inhibited MD-1 expression and lymphocyte proliferation, as well as decreased serum level of IL-2 and increased that of IL-10; FK506, treatment showed all the effects mentioned above but up-regulated the IL-10 level; SRL had no significant influence on either MD-1 expression or IL-2 and IL-10 level, although it equally suppressed the proliferation (P < .05 vs controls). Tacrolimus 148-153 interleukin 2 Mus musculus 300-304 15919519-8 2005 CsA, MMF, and AS-ODNs inhibited MD-1 expression and lymphocyte proliferation, as well as decreased serum level of IL-2 and increased that of IL-10; FK506, treatment showed all the effects mentioned above but up-regulated the IL-10 level; SRL had no significant influence on either MD-1 expression or IL-2 and IL-10 level, although it equally suppressed the proliferation (P < .05 vs controls). Tacrolimus 148-153 interleukin 10 Mus musculus 225-230 15818662-0 2005 Differential effects of the immunosuppressant FK-506 on human alpha2(I) collagen gene expression and transforming growth factor beta signaling in normal and scleroderma fibroblasts. Tacrolimus 46-52 collagen type I alpha 2 chain Homo sapiens 62-80 15919446-8 2005 This suggests that the intestinal mRNA level of MDR1 is a useful molecular marker for determination of the personalized oral dose of tacrolimus in recipients of LDLT immediately after surgery. Tacrolimus 133-143 ATP binding cassette subfamily B member 1 Homo sapiens 48-52 15919447-0 2005 Impact of CYP3A5 and MDR1(ABCB1) C3435T polymorphisms on the pharmacokinetics of tacrolimus in renal transplant recipients. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 15919447-0 2005 Impact of CYP3A5 and MDR1(ABCB1) C3435T polymorphisms on the pharmacokinetics of tacrolimus in renal transplant recipients. Tacrolimus 81-91 ATP binding cassette subfamily B member 1 Homo sapiens 21-25 15919447-0 2005 Impact of CYP3A5 and MDR1(ABCB1) C3435T polymorphisms on the pharmacokinetics of tacrolimus in renal transplant recipients. Tacrolimus 81-91 ATP binding cassette subfamily B member 1 Homo sapiens 26-31 15919447-1 2005 OBJECTIVE: The objective of this study was to assess the influence of CYP3A5 and MDR1 genetic polymorphisms on tacrolimus pharmacokinetics in Japanese renal transplant recipients. Tacrolimus 111-121 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 70-76 15919447-1 2005 OBJECTIVE: The objective of this study was to assess the influence of CYP3A5 and MDR1 genetic polymorphisms on tacrolimus pharmacokinetics in Japanese renal transplant recipients. Tacrolimus 111-121 ATP binding cassette subfamily B member 1 Homo sapiens 81-85 15919447-6 2005 The daily tacrolimus dose per body weight was significantly higher in CYP3A5*1 carriers than those of CYP3A5*3/*3 carriers (0.271 +/- 0.110 vs 0.150 +/- 0.056 mg/kg, P = .00016). Tacrolimus 10-20 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 70-76 15919447-6 2005 The daily tacrolimus dose per body weight was significantly higher in CYP3A5*1 carriers than those of CYP3A5*3/*3 carriers (0.271 +/- 0.110 vs 0.150 +/- 0.056 mg/kg, P = .00016). Tacrolimus 10-20 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 102-108 15919447-7 2005 In this study, a distinction was made between carriers of CYP3A5*1/*1+*1/*3 and CYP3A5*3/*3 to investigate the influence of the MDR1 C3435T mutation on tacrolimus pharmacokinetics. Tacrolimus 152-162 ATP binding cassette subfamily B member 1 Homo sapiens 128-132 15919447-9 2005 CONCLUSIONS: Renal transplant recipients who were CYP3A5*1 carriers required a higher dose of tacrolimus than CYP3A5*3/*3, indicating a significantly lower dose-adjusted AUC0-12 of tacrolimus. Tacrolimus 94-104 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 50-56 15919447-9 2005 CONCLUSIONS: Renal transplant recipients who were CYP3A5*1 carriers required a higher dose of tacrolimus than CYP3A5*3/*3, indicating a significantly lower dose-adjusted AUC0-12 of tacrolimus. Tacrolimus 181-191 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 50-56 15919458-6 2005 In this analysis, graft survival rate and medication usage patterns of antihyperlipidemics, antihypertensives, insulin, and oral hypoglycemics were observed for >5 years in patients receiving tacrolimus immunosuppression. Tacrolimus 195-205 insulin Homo sapiens 111-118 15919471-0 2005 Successful results after 5 years of tacrolimus therapy in ABO-incompatible kidney transplantation in Japan. Tacrolimus 36-46 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 58-61 15919471-3 2005 This study analyzed the results of 5 years of experience with tacrolimus in Japan, focusing on the efficacy of the drug in improving patient and graft survival in patients who underwent transplantation with ABO-incompatible kidney grafts. Tacrolimus 62-72 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 207-210 15919471-10 2005 The excellent outcome from this study demonstrates that even suboptimal ABO-incompatible donors can be used successfully as a source of kidneys when using tacrolimus as the immunosuppressive regimen. Tacrolimus 155-165 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 72-75 15919474-4 2005 We examined whether mouse peritoneal cavity CD5+ B-1a cells could be reduced by CsA/tacrolimus. Tacrolimus 84-94 CD5 antigen Mus musculus 44-47 15919474-6 2005 injection of various doses of CsA/tacrolimus decreased the percentage of PerC CD5+ B-1a cells and increased B-2 cells in a dose-dependent fashion. Tacrolimus 34-44 peroxisome proliferative activated receptor, gamma, coactivator 1 beta Mus musculus 73-77 15919474-6 2005 injection of various doses of CsA/tacrolimus decreased the percentage of PerC CD5+ B-1a cells and increased B-2 cells in a dose-dependent fashion. Tacrolimus 34-44 CD5 antigen Mus musculus 78-81 16476357-6 2005 MATERIAL AND METHODS: During the period from September 1, 2003 to April 30, 2004, an open study was carried out on 12 cases of vitiligo treated with topical tacrolimus 0.1 % twice a day. Tacrolimus 157-167 VAMAS6 Homo sapiens 127-135 16476357-15 2005 CONCLUSION: We believe that tacrolimus 0.1 % applied topically for a minimum of six months may be a valid alternative in the treatment of vitiligo in the facial area, especially the eyelids, where other therapeutic modes are not recommended because of the possible side effects. Tacrolimus 28-38 VAMAS6 Homo sapiens 138-146 15818662-5 2005 RESULTS: FK-506 did not affect the basal expression of type I procollagen protein or alpha2(I) collagen mRNA, but it significantly reduced the TGFbeta1-induced expression of type I procollagen protein and alpha2(I) collagen mRNA in normal fibroblasts. Tacrolimus 9-15 transforming growth factor beta 1 Homo sapiens 143-151 15818662-5 2005 RESULTS: FK-506 did not affect the basal expression of type I procollagen protein or alpha2(I) collagen mRNA, but it significantly reduced the TGFbeta1-induced expression of type I procollagen protein and alpha2(I) collagen mRNA in normal fibroblasts. Tacrolimus 9-15 collagen type I alpha 2 chain Homo sapiens 174-192 15818662-5 2005 RESULTS: FK-506 did not affect the basal expression of type I procollagen protein or alpha2(I) collagen mRNA, but it significantly reduced the TGFbeta1-induced expression of type I procollagen protein and alpha2(I) collagen mRNA in normal fibroblasts. Tacrolimus 9-15 collagen type I alpha 2 chain Homo sapiens 205-223 15818662-7 2005 In scleroderma fibroblasts, FK-506 significantly reduced the expression of type I procollagen protein and alpha2(I) collagen mRNA through posttranscriptional regulation, but not transcriptional regulation. Tacrolimus 28-34 collagen type I alpha 2 chain Homo sapiens 75-93 15818662-7 2005 In scleroderma fibroblasts, FK-506 significantly reduced the expression of type I procollagen protein and alpha2(I) collagen mRNA through posttranscriptional regulation, but not transcriptional regulation. Tacrolimus 28-34 collagen type I alpha 2 chain Homo sapiens 106-124 15818662-11 2005 CONCLUSION: FK-506 inhibits alpha2(I) collagen gene expression by reducing the stability of mRNA without exhibiting its activation effect on TGFbeta signaling in scleroderma fibroblasts. Tacrolimus 12-18 collagen type I alpha 2 chain Homo sapiens 28-46 15818662-1 2005 OBJECTIVE: To investigate the effects of FK-506 on the expression of the human alpha2(I) collagen gene and transforming growth factor beta (TGFbeta) signaling in normal and scleroderma fibroblasts. Tacrolimus 41-47 collagen type I alpha 2 chain Homo sapiens 79-97 15821130-10 2005 Cells deleted for CRZ1 or treated with the calcineurin inhibitor FK506 became hypersensitive to chitosan, supporting the notion that the Crz1p-controlled response offers protection against chitosan. Tacrolimus 65-70 DNA-binding transcription factor CRZ1 Saccharomyces cerevisiae S288C 137-142 15711594-3 2005 Cyclosporin A and tacrolimus also inhibit other calcineurin-dependent transcription factors including the ubiquitously expressed cAMP response element-binding protein (CREB). Tacrolimus 18-28 cAMP responsive element binding protein 1 Homo sapiens 129-166 15711594-3 2005 Cyclosporin A and tacrolimus also inhibit other calcineurin-dependent transcription factors including the ubiquitously expressed cAMP response element-binding protein (CREB). Tacrolimus 18-28 cAMP responsive element binding protein 1 Homo sapiens 168-172 15711594-5 2005 It was unknown at what step in CREB-mediated transcription cyclosporin A and tacrolimus interfere. Tacrolimus 77-87 cAMP responsive element binding protein 1 Homo sapiens 31-35 15711594-9 2005 The depolarization-induced transcriptional activity of the CBP C-terminus was enhanced by overexpression of calcineurin and was inhibited by cyclosporin A and tacrolimus in a concentration-dependent manner with IC50 values (10 and 1 nM, respectively) consistent with their known IC50 values for inhibition of calcineurin. Tacrolimus 159-169 CREB binding protein Homo sapiens 59-62 15711594-10 2005 These data suggest that, in contrast to NFAT, cyclosporin A and tacrolimus inhibit CREB transcriptional activity at the coactivator level. Tacrolimus 64-74 cAMP responsive element binding protein 1 Homo sapiens 83-87 15711594-0 2005 The immunosuppressive drugs cyclosporin A and tacrolimus inhibit membrane depolarization-induced CREB transcriptional activity at the coactivator level. Tacrolimus 46-56 cAMP responsive element binding protein 1 Homo sapiens 97-101 16392010-4 2005 OBJECTIVE: We report on a 51-year-old hepatitis C-positive man with corticosteroid refractory erosive lichen planus of the lip who had a rapid resolution of his lesions following a two-week course of topical 0.1% tacrolimus ointment. Tacrolimus 213-223 SMG1 nonsense mediated mRNA decay associated PI3K related kinase Homo sapiens 123-126 15746557-11 2005 FK506 aerosol markedly inhibited IL-5 mRNA expression in the lung. Tacrolimus 0-5 interleukin-5 Cavia porcellus 33-37 15746557-12 2005 In situ hybridization indicated that in the BALF IL-5 mRNA expression by lymphocyte-like cells was inhibited by FK506 aerosol. Tacrolimus 112-117 interleukin-5 Cavia porcellus 49-53 15778421-0 2005 Sequential analysis of tacrolimus dosing in adult lung transplant patients with ABCB1 haplotypes. Tacrolimus 23-33 ATP binding cassette subfamily B member 1 Homo sapiens 80-85 15778421-1 2005 The genetic polymorphisms in the ABCB1 gene, which encodes for the membrane pump, P-glycoprotein, have been previously demonstrated to have an association with tacrolimus dosing in organ transplant patients. Tacrolimus 160-170 ATP binding cassette subfamily B member 1 Homo sapiens 33-38 15778421-1 2005 The genetic polymorphisms in the ABCB1 gene, which encodes for the membrane pump, P-glycoprotein, have been previously demonstrated to have an association with tacrolimus dosing in organ transplant patients. Tacrolimus 160-170 ATP binding cassette subfamily B member 1 Homo sapiens 82-96 15778421-4 2005 Sequential analysis demonstrated that ABCB1 genotypes 00 and 01 had low tacrolimus [L/D] values at 1 and 3 months, but these values increased substantially at 6, 9, and 12 months after transplantation. Tacrolimus 72-82 ATP binding cassette subfamily B member 1 Homo sapiens 38-43 15778421-6 2005 Haplotype analysis also suggested that the homozygous for ABCB1 2677 variant allele had more of an impact on tacrolimus [L/D] in haplotype analysis than that of ABCB1 3435. Tacrolimus 109-119 ATP binding cassette subfamily B member 1 Homo sapiens 58-63 15732051-1 2005 Neurotrophic activity of neuroimmunophilin ligands (FK506 and its nonimmunosuppressant derivatives) has been assumed to be mediated by the FK506-binding protein-12 (FKBP-12). Tacrolimus 52-57 FKBP prolyl isomerase 1A Homo sapiens 139-163 15676204-3 2005 Mutant strain of Saccharomyces cerevisiae, named FAV20, sensitive to FK506 was constructed by disrupting VMA22 gene using the selectable marker kanMX4 which allowed detection of integration events. Tacrolimus 69-74 Vma22p Saccharomyces cerevisiae S288C 105-110 15732051-1 2005 Neurotrophic activity of neuroimmunophilin ligands (FK506 and its nonimmunosuppressant derivatives) has been assumed to be mediated by the FK506-binding protein-12 (FKBP-12). Tacrolimus 52-57 FKBP prolyl isomerase 1A Homo sapiens 165-172 15851377-0 2005 FK506 enhances triptolide-induced down-regulation of cyclooxygenase-2, inducible nitric oxide synthase as well as their products PGE2 and NO in TNF-alpha-stimulated synovial fibroblasts from rheumatoid arthritic patients. Tacrolimus 0-5 prostaglandin-endoperoxide synthase 2 Homo sapiens 53-69 15866630-2 2005 Our purpose was to assess the safety and efficacy of an immunosuppressive regimen using anti-IL-2R antibodies and MMF that allows delayed introduction of low-dose tacrolimus using elderly donors to elderly recipients. Tacrolimus 163-173 interleukin 2 receptor subunit alpha Homo sapiens 93-98 15773954-6 2005 An individualized approach to choice of calcineurin inhibitor, by which cyclosporine or tacrolimus are selected based on the patient"s particular risk profile, may thus help to reduce the toll of cardiovascular mortality among renal transplant recipients in the future. Tacrolimus 88-98 calcineurin binding protein 1 Homo sapiens 40-61 15851377-0 2005 FK506 enhances triptolide-induced down-regulation of cyclooxygenase-2, inducible nitric oxide synthase as well as their products PGE2 and NO in TNF-alpha-stimulated synovial fibroblasts from rheumatoid arthritic patients. Tacrolimus 0-5 nitric oxide synthase 2 Homo sapiens 71-102 15851377-10 2005 Combined treatment of FK506 and a lower concentration of TP (10 ng/ml) reduced both COX-2 and iNOS mRNA and protein expression, and correspondingly reduced PGE subset2 and NO produced by synovial fibroblasts. Tacrolimus 22-27 prostaglandin-endoperoxide synthase 2 Homo sapiens 84-89 15851377-10 2005 Combined treatment of FK506 and a lower concentration of TP (10 ng/ml) reduced both COX-2 and iNOS mRNA and protein expression, and correspondingly reduced PGE subset2 and NO produced by synovial fibroblasts. Tacrolimus 22-27 nitric oxide synthase 2 Homo sapiens 94-98 15851377-0 2005 FK506 enhances triptolide-induced down-regulation of cyclooxygenase-2, inducible nitric oxide synthase as well as their products PGE2 and NO in TNF-alpha-stimulated synovial fibroblasts from rheumatoid arthritic patients. Tacrolimus 0-5 tumor necrosis factor Homo sapiens 144-153 15851377-12 2005 NFkappaB activity in TNF-alpha-stimulated synovial cells was suppressed more profoundly by FK506 plus TP (10 ng/ml) than by TP (10 ng/ml) alone. Tacrolimus 91-96 nuclear factor kappa B subunit 1 Homo sapiens 0-8 15851377-1 2005 OBJECTIVE: To explore the effects of FK506 on the inhibition of cell proliferation and the expression of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS) and their products PGE subset2 and NO in TNF-alpha-stimulated human rheumatoid arthritis synovial fibroblasts (RASF) treated with triptolide (TP), and to study the mechanisms involved when combining FK506 and TP in RA therapy. Tacrolimus 37-42 prostaglandin-endoperoxide synthase 2 Homo sapiens 105-121 15851377-12 2005 NFkappaB activity in TNF-alpha-stimulated synovial cells was suppressed more profoundly by FK506 plus TP (10 ng/ml) than by TP (10 ng/ml) alone. Tacrolimus 91-96 tumor necrosis factor Homo sapiens 21-30 15851377-14 2005 CONCLUSION: FK506 enhanced TP-mediated down-regulation of COX-2 and iNOS as well as their products PGE subset2 and NO in human TNF-alpha-stimulated RASF by more profoundly suppressing the activity of NFkappaB. Tacrolimus 12-17 prostaglandin-endoperoxide synthase 2 Homo sapiens 58-63 15851377-1 2005 OBJECTIVE: To explore the effects of FK506 on the inhibition of cell proliferation and the expression of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS) and their products PGE subset2 and NO in TNF-alpha-stimulated human rheumatoid arthritis synovial fibroblasts (RASF) treated with triptolide (TP), and to study the mechanisms involved when combining FK506 and TP in RA therapy. Tacrolimus 37-42 prostaglandin-endoperoxide synthase 2 Homo sapiens 123-128 15851377-14 2005 CONCLUSION: FK506 enhanced TP-mediated down-regulation of COX-2 and iNOS as well as their products PGE subset2 and NO in human TNF-alpha-stimulated RASF by more profoundly suppressing the activity of NFkappaB. Tacrolimus 12-17 nitric oxide synthase 2 Homo sapiens 68-72 15851377-14 2005 CONCLUSION: FK506 enhanced TP-mediated down-regulation of COX-2 and iNOS as well as their products PGE subset2 and NO in human TNF-alpha-stimulated RASF by more profoundly suppressing the activity of NFkappaB. Tacrolimus 12-17 tumor necrosis factor Homo sapiens 127-136 15851377-1 2005 OBJECTIVE: To explore the effects of FK506 on the inhibition of cell proliferation and the expression of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS) and their products PGE subset2 and NO in TNF-alpha-stimulated human rheumatoid arthritis synovial fibroblasts (RASF) treated with triptolide (TP), and to study the mechanisms involved when combining FK506 and TP in RA therapy. Tacrolimus 37-42 nitric oxide synthase 2 Homo sapiens 131-162 15851377-14 2005 CONCLUSION: FK506 enhanced TP-mediated down-regulation of COX-2 and iNOS as well as their products PGE subset2 and NO in human TNF-alpha-stimulated RASF by more profoundly suppressing the activity of NFkappaB. Tacrolimus 12-17 nuclear factor kappa B subunit 1 Homo sapiens 200-208 15757646-7 2005 Inherent calcineurin inhibition by hFKBP38 would completely eliminate the need for FK506 in controlling many signal transduction pathways. Tacrolimus 83-88 FKBP prolyl isomerase 8 Homo sapiens 35-42 15748876-2 2005 We have demonstrated in vitro that the neuroimmunophilin, FK506 (10-100 nM), dose dependently and significantly restored the ROS production to the control level, increased the Bcl-2 protein level, partly inhibited the cytochrome C release from mitochondria and reduced the caspase-3 activation in SH-SY5Y cells. Tacrolimus 58-63 BCL2 apoptosis regulator Homo sapiens 176-181 15748876-2 2005 We have demonstrated in vitro that the neuroimmunophilin, FK506 (10-100 nM), dose dependently and significantly restored the ROS production to the control level, increased the Bcl-2 protein level, partly inhibited the cytochrome C release from mitochondria and reduced the caspase-3 activation in SH-SY5Y cells. Tacrolimus 58-63 caspase 3 Homo sapiens 273-282 15632146-5 2005 COX-2 expression and prostaglandin E2 (PGE2) production were induced upon pharmacological stimuli leading to NFAT activation and blunted by inhibition of calcineurin phosphatase with cyclosporin A or tacrolimus (FK506). Tacrolimus 200-210 prostaglandin-endoperoxide synthase 2 Homo sapiens 0-5 15632146-5 2005 COX-2 expression and prostaglandin E2 (PGE2) production were induced upon pharmacological stimuli leading to NFAT activation and blunted by inhibition of calcineurin phosphatase with cyclosporin A or tacrolimus (FK506). Tacrolimus 212-217 prostaglandin-endoperoxide synthase 2 Homo sapiens 0-5 15707993-7 2005 The mode of Pdr5p inhibition of enniatin was competitive against FK506, and its inhibitory activity was more potent with less toxicity than that of FK506. Tacrolimus 65-70 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 12-17 15707993-7 2005 The mode of Pdr5p inhibition of enniatin was competitive against FK506, and its inhibitory activity was more potent with less toxicity than that of FK506. Tacrolimus 148-153 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 12-17 15707993-8 2005 The enniatins showed similar inhibitory profile as FK506 against S1360 mutants (S1360A and S1360F) of Pdr5p. Tacrolimus 51-56 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 102-107 15745744-4 2005 When FK506-binding protein 12 (FKBP12) immobilized beads were used in this process, FK506 was efficiently purified in one step either from a mixture of chemical compounds or from fermented broth extract. Tacrolimus 5-10 FKBP prolyl isomerase 1A Homo sapiens 31-37 15932724-4 2005 RESULTS: Glucose-stimulated insulin secretion from human islet cells was significantly reduced after exposed to high concentrations of MMF and FK506 (both P < 0.05). Tacrolimus 143-148 insulin Homo sapiens 28-35 15932724-6 2005 CONCLUSION: (1) High concentrations of MMF and FK506 have deleterious effects on insulin secretion in human islet cells. Tacrolimus 47-52 insulin Homo sapiens 81-88 15770715-8 2005 RESULTS: Sirolimus alone or combined with tacrolimus inhibited the growth of both cell lines after 5 d by up to 35% in SK-Hep 1 cells, and by up to 68% in Hep 3B cells at 25 ng/mL. Tacrolimus 42-52 DNL-type zinc finger Homo sapiens 122-127 15770715-10 2005 We found an increase of apoptotic Hep 3B cells from 6 to 16%, and a G1-arrest in SK-Hep 1 cells with an increase of cells from 61 to 82%, when sirolimus and tacrolimus were combined. Tacrolimus 157-167 DNL-type zinc finger Homo sapiens 84-89 15728480-5 2005 Cyclosporin A and FK506, which target calcineurin and thereby inhibit TCR-mediated Ca(2+) signal pathways, block IL-6-mediated c-Maf expression. Tacrolimus 18-23 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 70-73 15728480-5 2005 Cyclosporin A and FK506, which target calcineurin and thereby inhibit TCR-mediated Ca(2+) signal pathways, block IL-6-mediated c-Maf expression. Tacrolimus 18-23 interleukin 6 Homo sapiens 113-117 15728480-5 2005 Cyclosporin A and FK506, which target calcineurin and thereby inhibit TCR-mediated Ca(2+) signal pathways, block IL-6-mediated c-Maf expression. Tacrolimus 18-23 MAF bZIP transcription factor Homo sapiens 127-132 15698457-0 2005 Impact of gastric acid suppressants on cytochrome P450 3A4 and P-glycoprotein: consequences for FK506 assimilation. Tacrolimus 96-101 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 39-58 15698457-0 2005 Impact of gastric acid suppressants on cytochrome P450 3A4 and P-glycoprotein: consequences for FK506 assimilation. Tacrolimus 96-101 ATP binding cassette subfamily B member 1 Homo sapiens 63-77 15698457-1 2005 BACKGROUND: Cytochrome P450 3A4 (CYP3A4) and P-glycoprotein (PGP) are important determinants of the oral bioavailability and clearance of tacrolimus. Tacrolimus 138-148 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 12-31 15698457-1 2005 BACKGROUND: Cytochrome P450 3A4 (CYP3A4) and P-glycoprotein (PGP) are important determinants of the oral bioavailability and clearance of tacrolimus. Tacrolimus 138-148 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 33-39 15698457-1 2005 BACKGROUND: Cytochrome P450 3A4 (CYP3A4) and P-glycoprotein (PGP) are important determinants of the oral bioavailability and clearance of tacrolimus. Tacrolimus 138-148 ATP binding cassette subfamily B member 1 Homo sapiens 45-59 15698457-1 2005 BACKGROUND: Cytochrome P450 3A4 (CYP3A4) and P-glycoprotein (PGP) are important determinants of the oral bioavailability and clearance of tacrolimus. Tacrolimus 138-148 ATP binding cassette subfamily B member 1 Homo sapiens 61-64 15882134-5 2005 This review describes polymorphisms of the genes coding for P-gp and CYPs, and focuses on the compounds cyclosporin and tacrolimus. Tacrolimus 120-130 ATP binding cassette subfamily B member 1 Homo sapiens 60-64 16895664-4 2005 Calcineurin inhibitors, cyclosporine and tacrolimus, are involved in tumor development through various mechanisms: they promote B-cell proliferation by increasing T lymphocyte IL6 secretion, decrease DNA repair ability and may be able to promote metastasis spreading by a direct cellular effect that is independent of their effect on the host"s immune cells. Tacrolimus 41-51 interleukin 6 Homo sapiens 176-179 15721171-11 2005 Pretreatment with FK506 (3 mg/kg) attenuated significantly the reduction of DAT and TH immunoreactivity after repeated administration of MAP. Tacrolimus 18-23 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 76-79 15729180-0 2005 Tacrolimus pharmacogenetics: the CYP3A5*1 allele predicts low dose-normalized tacrolimus blood concentrations in whites and South Asians. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 15729180-0 2005 Tacrolimus pharmacogenetics: the CYP3A5*1 allele predicts low dose-normalized tacrolimus blood concentrations in whites and South Asians. Tacrolimus 78-88 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 15729180-1 2005 Previously, we demonstrated that the dose-normalized tacrolimus blood concentration after renal transplantation was associated with a single nucleotide polymorphism (SNP) in the CYP3AP1 gene, probably through linkage with an SNP in the CYP3A5 gene. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 51, pseudogene Homo sapiens 178-185 15729180-1 2005 Previously, we demonstrated that the dose-normalized tacrolimus blood concentration after renal transplantation was associated with a single nucleotide polymorphism (SNP) in the CYP3AP1 gene, probably through linkage with an SNP in the CYP3A5 gene. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 236-242 15729180-4 2005 South Asian and white patients with at least one CYP3A5*1 allele achieved twofold lower dose-normalized tacrolimus blood concentrations compared with CYP3A5*3/*3 homozygotes, confirming our previous findings for the CYP3AP1 SNP. Tacrolimus 104-114 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 49-55 15729180-4 2005 South Asian and white patients with at least one CYP3A5*1 allele achieved twofold lower dose-normalized tacrolimus blood concentrations compared with CYP3A5*3/*3 homozygotes, confirming our previous findings for the CYP3AP1 SNP. Tacrolimus 104-114 cytochrome P450 family 3 subfamily A member 51, pseudogene Homo sapiens 216-223 15729180-6 2005 Determination of the CYP3A5*1/*3 genotype could be used to predict the tacrolimus dose requirement and, given incomplete linkage, would be better than determination of the CYP3AP1 genotype. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 21-27 15670575-0 2005 Transient up-regulation of P-glycoprotein reduces tacrolimus absorption after ischemia-reperfusion injury in rat ileum. Tacrolimus 50-60 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 27-41 15670575-8 2005 However, the P-glycoprotein level returned to normal at 48 h. The intra-individual variation in the absorptive rate of tacrolimus was suggested to be regulated by the morphological status of the intestinal epithelium and enterocyte expression level of P-glycoprotein. Tacrolimus 119-129 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 13-27 15670575-8 2005 However, the P-glycoprotein level returned to normal at 48 h. The intra-individual variation in the absorptive rate of tacrolimus was suggested to be regulated by the morphological status of the intestinal epithelium and enterocyte expression level of P-glycoprotein. Tacrolimus 119-129 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 252-266 15670575-9 2005 Therefore, the monitoring of the enterocyte P-glycoprotein level would provide useful information for determining the dosage of tacrolimus immediately after small bowl transplantation. Tacrolimus 128-138 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 44-58 15697228-0 2005 Differential control of glucocorticoid receptor hormone-binding function by tetratricopeptide repeat (TPR) proteins and the immunosuppressive ligand FK506. Tacrolimus 149-154 nuclear receptor subfamily 3 group C member 1 Homo sapiens 24-47 15697228-7 2005 FK506 was found to stimulate GR transactivity beyond the effect of this ligand on hormone retention. Tacrolimus 0-5 nuclear receptor subfamily 3 group C member 1 Homo sapiens 29-31 15471978-8 2005 FK-506 reversibly decreased the L-type Ca2+ current (ICaL) by 25%, although high-frequency-dependent facilitation of ICaL persisted, and decreased three distinct K+ currents: delayed rectifier K+ current (IK; >80%), transient outward K+ current (<20%), and inward rectifier K+ current (IK1; >40%). Tacrolimus 0-6 potassium calcium-activated channel subfamily N member 4 Rattus norvegicus 292-295 15697228-9 2005 By Scatchard analysis, FK506 was found to increase GR hormone-binding affinity while decreasing total binding sites for hormone. Tacrolimus 23-28 nuclear receptor subfamily 3 group C member 1 Homo sapiens 51-53 15713424-3 2005 FK1706, a derivative of FK506, showed similarly high affinity for two FKBP subtypes, FKBP-12 and FKBP-52, but inhibited T-cell proliferation and interleukin-2 cytokine production with much lower potency and efficacy than FK506. Tacrolimus 24-29 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 85-92 15713424-3 2005 FK1706, a derivative of FK506, showed similarly high affinity for two FKBP subtypes, FKBP-12 and FKBP-52, but inhibited T-cell proliferation and interleukin-2 cytokine production with much lower potency and efficacy than FK506. Tacrolimus 24-29 FKBP prolyl isomerase 4 Homo sapiens 97-104 15471978-11 2005 In conclusion, the effects of FK-506 on the cardiac AP are more complex than previously reported and include inhibitions of IK1 and ICaL. Tacrolimus 30-36 potassium calcium-activated channel subfamily N member 4 Rattus norvegicus 124-127 15479952-0 2005 Tacrolimus suppresses glucose-induced insulin release from pancreatic islets by reducing glucokinase activity. Tacrolimus 0-10 insulin Homo sapiens 38-45 15780950-7 2005 FK506 in the therapeutic range (25 nmol/L) suppressed expression of Cbfa1 and osterix mRNA. Tacrolimus 0-5 RUNX family transcription factor 2 Rattus norvegicus 68-73 15479952-0 2005 Tacrolimus suppresses glucose-induced insulin release from pancreatic islets by reducing glucokinase activity. Tacrolimus 0-10 glucokinase Homo sapiens 89-100 15479952-3 2005 We have investigated the metabolic effects of tacrolimus on insulin secretion at a concentration that does not influence insulin content. Tacrolimus 46-56 insulin Homo sapiens 60-67 15479952-4 2005 Twenty-four-hour exposure to 3 nM tacrolimus reduced high glucose (16.7 mM)-induced insulin secretion (control 2.14 +/- 0.08 vs. tacrolimus 1.75 +/- 0.02 ng.islet(-1).30 min(-1), P < 0.01) without affecting insulin content. Tacrolimus 34-44 insulin Homo sapiens 84-91 15479952-7 2005 However, insulin release from tacrolimus-treated islets was similar to that from control islets in the presence of 16.7 mM alpha-ketoisocaproate, a mitochondrial fuel. Tacrolimus 30-40 insulin Homo sapiens 9-16 15780950-1 2005 FK506 is a commonly used immunosuppressant that mediates its action by exclusively interacting with the cytosolic immunophilin, FK506 binding protein 12 (FKBP12). Tacrolimus 0-5 FKBP prolyl isomerase 1A Rattus norvegicus 128-152 15780950-1 2005 FK506 is a commonly used immunosuppressant that mediates its action by exclusively interacting with the cytosolic immunophilin, FK506 binding protein 12 (FKBP12). Tacrolimus 0-5 FKBP prolyl isomerase 1A Rattus norvegicus 154-160 15479952-8 2005 Glucokinase activity, which determines glycolytic velocity, was reduced by tacrolimus treatment (control 65.3 +/- 3.4 vs. tacrolimus 49.9 +/- 2.8 pmol.islet(-1).60 min(-1), P < 0.01), whereas hexokinase activity was not affected. Tacrolimus 75-85 glucokinase Homo sapiens 0-11 15479952-8 2005 Glucokinase activity, which determines glycolytic velocity, was reduced by tacrolimus treatment (control 65.3 +/- 3.4 vs. tacrolimus 49.9 +/- 2.8 pmol.islet(-1).60 min(-1), P < 0.01), whereas hexokinase activity was not affected. Tacrolimus 122-132 glucokinase Homo sapiens 0-11 15479952-9 2005 These results indicate that glucose-stimulated insulin release is decreased by chronic exposure to tacrolimus due to reduced ATP production and glycolysis derived from reduced glucokinase activity. Tacrolimus 99-109 insulin Homo sapiens 47-54 15479952-9 2005 These results indicate that glucose-stimulated insulin release is decreased by chronic exposure to tacrolimus due to reduced ATP production and glycolysis derived from reduced glucokinase activity. Tacrolimus 99-109 glucokinase Homo sapiens 176-187 15780950-3 2005 Therefore, in the present study we characterised FKBP12 in osteoblasts and investigated the role of FK506 in modulating osteoblast-specific transcription factors, core-binding factor alpha1 (Cbfa1) and osterix gene expression in ROS 17/2.8 cells. Tacrolimus 100-105 RUNX family transcription factor 2 Rattus norvegicus 163-189 15780950-7 2005 FK506 in the therapeutic range (25 nmol/L) suppressed expression of Cbfa1 and osterix mRNA. Tacrolimus 0-5 Sp7 transcription factor Rattus norvegicus 78-85 15677500-3 2005 In this study, we report that tacrolimus, sirolimus, and mycophenolic acid, when added to cultures of freshly isolated human islets, induce a downregulation of the synthesis and secretion of insulin. Tacrolimus 30-40 insulin Homo sapiens 191-198 15837449-3 2005 Evidence from large-scale analyses of registry databases has shown that the calcineurin inhibitor, tacrolimus, is associated with approximately a 50% increase in the risk of NODM compared to the microemulsion formulation of cyclosporine (CsA); but to date, little robust evidence is available from clinical trials. Tacrolimus 99-109 calcineurin binding protein 1 Homo sapiens 76-97 15783066-6 2005 RESULTS: The clinical activity score and myeloperoxidase activity decreased after the administration of all FK506-containing formulations. Tacrolimus 108-113 myeloperoxidase Rattus norvegicus 41-56 15627982-5 2005 This autoregulation involves the calcineurin signaling pathway, as rhTRX antagonizes the cyclosporine A (CsA)- and tacrolimus-mediated suppression of TRX gene expression. Tacrolimus 115-125 thioredoxin Homo sapiens 69-72 15694998-8 2005 The TGFbeta1 high-producer genotype had worse CrCl than intermediate and low producers at every time point, despite similar pretransplant CrCl (pretransplant = 120 +/- 53 vs 118 +/- 55 ml/mn/1.73 m2 [p = 0.8], 1 year = 92 +/- 38 vs 113 +/- 30 ml/mn/1.73 m2 [p = 0.03]) and similar tacrolimus levels. Tacrolimus 281-291 transforming growth factor beta 1 Homo sapiens 4-12 15691347-7 2005 CONCLUSIONS: These findings suggest that increased MMP-1 gene and protein expression may be important for regulating COL-I homeostasis in the gingival connective compartment of FK506-immunosuppressed subjects. Tacrolimus 177-182 matrix metallopeptidase 1 Homo sapiens 51-56 15598440-3 2005 Culture supernatant from peripheral blood mononuclear cells (PBMC) stimulated with anti-CD3 plus anti-CD2 antibodies effectively induced the expression of E-selectin, ICAM-1 and VCAM-1 on HMVEC, and treatment with FK506 down-regulated their expression. Tacrolimus 214-219 CD2 molecule Homo sapiens 102-105 15598440-0 2005 FK506 suppresses E-selectin, ICAM-1 and VCAM-1 expression on vascular endothelial cells by inhibiting tumor necrosis factor alpha secretion from peripheral blood mononuclear cells. Tacrolimus 0-5 selectin E Homo sapiens 17-27 15598440-3 2005 Culture supernatant from peripheral blood mononuclear cells (PBMC) stimulated with anti-CD3 plus anti-CD2 antibodies effectively induced the expression of E-selectin, ICAM-1 and VCAM-1 on HMVEC, and treatment with FK506 down-regulated their expression. Tacrolimus 214-219 selectin E Homo sapiens 155-165 15598440-0 2005 FK506 suppresses E-selectin, ICAM-1 and VCAM-1 expression on vascular endothelial cells by inhibiting tumor necrosis factor alpha secretion from peripheral blood mononuclear cells. Tacrolimus 0-5 intercellular adhesion molecule 1 Homo sapiens 29-35 15598440-0 2005 FK506 suppresses E-selectin, ICAM-1 and VCAM-1 expression on vascular endothelial cells by inhibiting tumor necrosis factor alpha secretion from peripheral blood mononuclear cells. Tacrolimus 0-5 vascular cell adhesion molecule 1 Homo sapiens 40-46 15598440-0 2005 FK506 suppresses E-selectin, ICAM-1 and VCAM-1 expression on vascular endothelial cells by inhibiting tumor necrosis factor alpha secretion from peripheral blood mononuclear cells. Tacrolimus 0-5 tumor necrosis factor Homo sapiens 102-129 15598440-1 2005 FK506 suppresses activation of T cells; however, it down-regulates E-selectin, ICAM-1 and VCAM-1 expression in inflamed tissues. Tacrolimus 0-5 selectin E Homo sapiens 67-77 15598440-3 2005 Culture supernatant from peripheral blood mononuclear cells (PBMC) stimulated with anti-CD3 plus anti-CD2 antibodies effectively induced the expression of E-selectin, ICAM-1 and VCAM-1 on HMVEC, and treatment with FK506 down-regulated their expression. Tacrolimus 214-219 intercellular adhesion molecule 1 Homo sapiens 167-173 15598440-3 2005 Culture supernatant from peripheral blood mononuclear cells (PBMC) stimulated with anti-CD3 plus anti-CD2 antibodies effectively induced the expression of E-selectin, ICAM-1 and VCAM-1 on HMVEC, and treatment with FK506 down-regulated their expression. Tacrolimus 214-219 vascular cell adhesion molecule 1 Homo sapiens 178-184 15598440-9 2005 These results indicate that FK506 suppresses migration of inflammatory cells through the inhibition of TNFalpha secretion from leukocytes. Tacrolimus 28-33 tumor necrosis factor Homo sapiens 103-111 15609388-4 2005 In recent years, PSC has been an area of active research worldwide with great interest in etiology, pathogenesis, diagnosis, and therapeutic options such as hydrophilic ursodeoxycholic acid and immunosuppressive agent tacrolimus. Tacrolimus 218-228 PSC Homo sapiens 17-20 15598440-1 2005 FK506 suppresses activation of T cells; however, it down-regulates E-selectin, ICAM-1 and VCAM-1 expression in inflamed tissues. Tacrolimus 0-5 intercellular adhesion molecule 1 Homo sapiens 79-85 15598440-1 2005 FK506 suppresses activation of T cells; however, it down-regulates E-selectin, ICAM-1 and VCAM-1 expression in inflamed tissues. Tacrolimus 0-5 vascular cell adhesion molecule 1 Homo sapiens 90-96 15548389-7 2004 30-40%) of caspase-3 activation by 5 microM FK506 and CsA was observed. Tacrolimus 44-49 caspase 3 Homo sapiens 11-20 15881424-3 2005 A 9-year-old boy with CN1 was prepared with plasmapheresis and immunosuppression with prednisolone and tacrolimus. Tacrolimus 103-113 5'-nucleotidase, cytosolic IA Homo sapiens 22-25 16038570-4 2005 Tacrolimus is primarily metabolised by cytochrome P450 (CYP) 3A enzymes in the gut wall and liver. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 39-63 16038570-5 2005 It is also a substrate for P-glycoprotein, which counter-transports diffused tacrolimus out of intestinal cells and back into the gut lumen. Tacrolimus 77-87 ATP binding cassette subfamily B member 1 Homo sapiens 27-41 16038570-6 2005 Age-associated alterations in CYP 3A and P-glycoprotein expression and/or activity, along with liver mass and body composition changes, would be expected to affect the pharmacokinetics of tacrolimus in the elderly. Tacrolimus 188-198 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 30-36 16038570-6 2005 Age-associated alterations in CYP 3A and P-glycoprotein expression and/or activity, along with liver mass and body composition changes, would be expected to affect the pharmacokinetics of tacrolimus in the elderly. Tacrolimus 188-198 ATP binding cassette subfamily B member 1 Homo sapiens 41-55 15390105-8 2005 In FK506-treated rats, a significant reduction of IL-1beta, IL-6, and TNF-alpha expression was observed 12 h after reperfusion. Tacrolimus 3-8 interleukin 1 beta Rattus norvegicus 50-58 15390105-8 2005 In FK506-treated rats, a significant reduction of IL-1beta, IL-6, and TNF-alpha expression was observed 12 h after reperfusion. Tacrolimus 3-8 interleukin 6 Rattus norvegicus 60-64 15390105-8 2005 In FK506-treated rats, a significant reduction of IL-1beta, IL-6, and TNF-alpha expression was observed 12 h after reperfusion. Tacrolimus 3-8 tumor necrosis factor Rattus norvegicus 70-79 15390105-9 2005 FK506 neuroprotection was associated with a significant downregulation of IL-1beta expression in astrocytes and microglia in the injured side. Tacrolimus 0-5 interleukin 1 beta Rattus norvegicus 74-82 15390105-10 2005 FK506 selectively decreased the levels of TNF-alpha, and IL-1beta mRNAs in astrocytes in vitro, with no effect on transforming growth factor-beta 1 (TGF-beta1) and IL-6 expression. Tacrolimus 0-5 tumor necrosis factor Rattus norvegicus 42-51 15390105-10 2005 FK506 selectively decreased the levels of TNF-alpha, and IL-1beta mRNAs in astrocytes in vitro, with no effect on transforming growth factor-beta 1 (TGF-beta1) and IL-6 expression. Tacrolimus 0-5 interleukin 1 beta Rattus norvegicus 57-65 15381148-1 2005 The large molecular-weight immunophilin, FKBP52, is a known target of the immunosuppressive drug FK506. Tacrolimus 97-102 FKBP prolyl isomerase 4 Homo sapiens 41-47 15381148-2 2005 FKBP52 exhibits peptidyl-prolyl cis-trans isomerase (PPIase) activity, which is inhibited by the binding of FK506--properties that it shares with the smaller but better-studied immunophilin, FKBP12. Tacrolimus 108-113 FKBP prolyl isomerase 4 Homo sapiens 0-6 15381148-2 2005 FKBP52 exhibits peptidyl-prolyl cis-trans isomerase (PPIase) activity, which is inhibited by the binding of FK506--properties that it shares with the smaller but better-studied immunophilin, FKBP12. Tacrolimus 108-113 FKBP prolyl isomerase 4 Homo sapiens 16-51 15381148-2 2005 FKBP52 exhibits peptidyl-prolyl cis-trans isomerase (PPIase) activity, which is inhibited by the binding of FK506--properties that it shares with the smaller but better-studied immunophilin, FKBP12. Tacrolimus 108-113 FKBP prolyl isomerase 4 Homo sapiens 53-59 16103732-0 2005 Tacrolimus and cyclosporinein vitro and in vivo induce osteopontin mRNA and protein expression in renal tissues. Tacrolimus 0-10 secreted phosphoprotein 1 Mus musculus 55-66 16103732-3 2005 In this study, using in vitro and in vivo models, we examined the effects of cyclosporine (CsA) and tacrolimus (TAC) on OPN mRNA and protein expression. Tacrolimus 100-110 secreted phosphoprotein 1 Mus musculus 120-123 15723604-4 2005 It has been recently shown that the CYP3A5*3 polymorphism is associated with both the pharmacokinetics and pharmacodynamic consequences of tacrolimus. Tacrolimus 139-149 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 16082080-2 2005 We hypothesized that combining low-dose FK506 with anti-CD40 Ligand (anti-CD40L mAb) would enhance regeneration through peripheral nerve allografts while preserving immune unresponsiveness. Tacrolimus 40-45 CD40 ligand Mus musculus 74-79 16082080-5 2005 RESULTS: Animals receiving low-dose FK506 with anti-CD40L mAb exhibited robust nerve regeneration comparable to the isograft and high-dose FK506 allograft groups. Tacrolimus 36-41 CD40 ligand Mus musculus 52-57 16082080-6 2005 Nerve density was significantly increased in the low-dose FK506 with anti-CD40L mAb group compared to animals receiving anti-CD40L mAb alone (p < 0.05). Tacrolimus 58-63 CD40 ligand Mus musculus 74-79 16082080-10 2005 CONCLUSION: When combined with anti-CD40L mAb, low-dose FK506 enhances nerve regeneration without disrupting immune unresponsiveness. Tacrolimus 56-61 CD40 ligand Mus musculus 36-41 15808571-10 2005 Tacrolimus alone decreased TGF-beta and TIMP-1 expression, suggesting some antifibrotic action; addition of pirfenidone had no further effect. Tacrolimus 0-10 transforming growth factor, beta 1 Rattus norvegicus 27-35 15808571-10 2005 Tacrolimus alone decreased TGF-beta and TIMP-1 expression, suggesting some antifibrotic action; addition of pirfenidone had no further effect. Tacrolimus 0-10 TIMP metallopeptidase inhibitor 1 Rattus norvegicus 40-46 15808578-11 2005 In conclusion, the mechanism of tacrolimus and sirolimus induced long-term allograft survival in primates relates to up-regulated FasL expression, NKT cells and dendritic cells, with downregulation of MLR sensitivity. Tacrolimus 32-42 Fas ligand Homo sapiens 130-134 15808586-0 2005 Genetic polymorphisms of CYP3A5 genes and concentration of the cyclosporine and tacrolimus. Tacrolimus 80-90 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 25-31 15808586-2 2005 Our objective was to determine the relationship between genetic polymorphisms of CYP3A5 with respect to interindividual variability in CsA and tacrolimus pharmacokinetics. Tacrolimus 143-153 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 81-87 15808586-6 2005 RESULTS: At 3, 6, and 12 months, the tacrolimus dose-adjusted trough levels (dose-adjusted C0) showed a statistically significant difference between the group of CYP3A5*3/*3 (n = 19) and the group of CYP3A5*1 allele carriers. Tacrolimus 37-47 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 162-168 15808586-6 2005 RESULTS: At 3, 6, and 12 months, the tacrolimus dose-adjusted trough levels (dose-adjusted C0) showed a statistically significant difference between the group of CYP3A5*3/*3 (n = 19) and the group of CYP3A5*1 allele carriers. Tacrolimus 37-47 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 200-206 15808586-9 2005 CONCLUSION: Patients with the CYP3A5*3/*3 genotype require less tacrolimus to reach target concentrations compared to those with the CYP3A5*1 allele. Tacrolimus 64-74 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 30-36 15548389-9 2004 In addition, FK506 significantly reduced not only caspase-8 but also caspase-9 activation, to a similar extent as CsA, thus suggesting a protective effect at the mitochondrial level of this drug, as has already been reported for CsA. Tacrolimus 13-18 caspase 8 Homo sapiens 50-59 15548389-9 2004 In addition, FK506 significantly reduced not only caspase-8 but also caspase-9 activation, to a similar extent as CsA, thus suggesting a protective effect at the mitochondrial level of this drug, as has already been reported for CsA. Tacrolimus 13-18 caspase 9 Homo sapiens 69-78 15383495-2 2004 Tacrolimus absorption from the gastrointestinal tract is to a large extent determined by the genotypic, phenotypic, and functional expression of P-glycoprotein and CYP3A in the gut wall and liver. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 145-159 15383495-2 2004 Tacrolimus absorption from the gastrointestinal tract is to a large extent determined by the genotypic, phenotypic, and functional expression of P-glycoprotein and CYP3A in the gut wall and liver. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 164-169 15571979-4 2004 FK506 decreased cytochrome c release and caspase-3-like activity induced by 3-NP, without changing the activities of other caspases. Tacrolimus 0-5 caspase-3-like Rattus norvegicus 41-55 15575024-4 2004 Administering tacrolimus to non-diabetic, dialysis patients was shown to result in a dose-related reduction in insulin secretion without altering insulin resistance. Tacrolimus 14-24 insulin Homo sapiens 111-118 15575024-6 2004 In a second study, corticosteroid withdrawal from tacrolimus-based immunosuppression reduced insulin resistance without changing insulin secretion. Tacrolimus 50-60 insulin Homo sapiens 93-100 15571979-7 2004 Our results also showed a decrease in mitochondrial Bax and an increase in mitochondrial Bcl-2 levels upon exposure to FK506 and 3-NP. Tacrolimus 119-124 BCL2 associated X, apoptosis regulator Rattus norvegicus 52-55 15575024-7 2004 Moreover, reducing tacrolimus blood levels by 30% within the therapeutic window increased both insulin and C-peptide secretion by 24 and 36%, respectively. Tacrolimus 19-29 insulin Homo sapiens 95-102 15575024-9 2004 A comparison of the effects of tacrolimus and ciclosporin on glucose metabolism revealed reduced insulin release with tacrolimus at week 3 post-transplant, but for the remainder of the 3 year follow-up there were no significant differences between the two treatment arms. Tacrolimus 31-41 insulin Homo sapiens 97-104 15571979-7 2004 Our results also showed a decrease in mitochondrial Bax and an increase in mitochondrial Bcl-2 levels upon exposure to FK506 and 3-NP. Tacrolimus 119-124 BCL2, apoptosis regulator Rattus norvegicus 89-94 15575024-9 2004 A comparison of the effects of tacrolimus and ciclosporin on glucose metabolism revealed reduced insulin release with tacrolimus at week 3 post-transplant, but for the remainder of the 3 year follow-up there were no significant differences between the two treatment arms. Tacrolimus 118-128 insulin Homo sapiens 97-104 15571979-9 2004 Altogether, the results suggest that FK506 neuroprotection against 3-NP-induced apoptosis is associated with the redistribution of Bcl-2 and Bax in the mitochondrial membrane. Tacrolimus 37-42 BCL2, apoptosis regulator Rattus norvegicus 131-136 15571979-9 2004 Altogether, the results suggest that FK506 neuroprotection against 3-NP-induced apoptosis is associated with the redistribution of Bcl-2 and Bax in the mitochondrial membrane. Tacrolimus 37-42 BCL2 associated X, apoptosis regulator Rattus norvegicus 141-144 15521904-1 2004 AIM: This retrospective study investigated the influence of MDR1 haplotypes derived from the polymorphisms 2677G > T (exon 21) and 3435C > T (exon 26) on the pharmacokinetics of the immunosuppressant drug tacrolimus in 73 renal transplant patients. Tacrolimus 211-221 ATP binding cassette subfamily B member 1 Homo sapiens 60-64 15570183-9 2004 In vitro, MPA at concentrations above the plasma therapeutic range was found to decrease the metabolism of tacrolimus, suggesting a possible competition for CYP3A. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 157-162 15621119-3 2004 Furthermore calcineurin inhibitor nephrotoxicity was exacerbated by rescue therapy with tacrolimus conversion. Tacrolimus 88-98 calcineurin binding protein 1 Homo sapiens 12-33 15488288-7 2004 These findings, taken together, indicate that FK506 application on inflamed skin may activate nociceptive C-fibers, which bear bradykinin receptors and capsaicin-sensitive heat transducer of TRP family, TRPV1. Tacrolimus 46-51 transient receptor potential cation channel, subfamily V, member 1 Rattus norvegicus 203-208 15523355-0 2004 Tacrolimus ointment promotes repigmentation of vitiligo in children: a review of 57 cases. Tacrolimus 0-10 VAMAS6 Homo sapiens 47-55 15523355-4 2004 OBJECTIVE: We sought to assess the efficacy of topical tacrolimus ointment in the treatment of pediatric vitiligo. Tacrolimus 55-65 VAMAS6 Homo sapiens 105-113 15523355-11 2004 CONCLUSIONS: Topical tacrolimus ointment is an effective alternative therapy for childhood vitiligo, particularly involving the head and neck. Tacrolimus 21-31 VAMAS6 Homo sapiens 91-99 15381199-2 2004 By using DNA array technology, we have previously demonstrated that IRF1 is significantly upregulated during acute rejection in rat heart allografts and is restored to isograft levels when recipients are treated with the immunosuppressants tacrolimus or cyclosporin A (CsA). Tacrolimus 240-250 interferon regulatory factor 1 Rattus norvegicus 68-72 15586415-0 2004 [Suppression of Tacrolimus on the expression of MMP-9 in rhHGF-stimulated ECV304 cell line]. Tacrolimus 16-26 matrix metallopeptidase 9 Homo sapiens 48-53 15555450-5 2004 FK506, CsA and DEX suppressed significantly the expressions of IL-18 mRNA and its protein (P<0.001) in LPS/PHA-stimulated whole blood cell from LN patients. Tacrolimus 0-5 interleukin 18 Homo sapiens 63-68 15555450-6 2004 The inhibitory effects of FK506, CsA and DEX on the IL-18 protein expression in LN patients were stronger than that in normal control group (P<0.01 or P<0.05). Tacrolimus 26-31 interleukin 18 Homo sapiens 52-57 15586415-1 2004 OBJECTIVE: To investigate the stimulating effect of recombinant human hepatocyte growth factor (rhHGF) on the expression of matrix metalloproteinase-9 (MMP-9) in ECV304 cells and the suppression effect of FK506. Tacrolimus 205-210 hepatocyte growth factor Homo sapiens 70-94 15586415-1 2004 OBJECTIVE: To investigate the stimulating effect of recombinant human hepatocyte growth factor (rhHGF) on the expression of matrix metalloproteinase-9 (MMP-9) in ECV304 cells and the suppression effect of FK506. Tacrolimus 205-210 matrix metallopeptidase 9 Homo sapiens 152-157 15586415-9 2004 Cell livability was 0.398764 +/-0.092476 (P<0.01) when 50 ng/ml FK506 was added to culture medium, and the expression level of MMP-9 was 14.61%. Tacrolimus 67-72 matrix metallopeptidase 9 Homo sapiens 130-135 15586415-12 2004 FK506 can suppress the proliferation of ECV304 cells and decrease the expression level of MMP-9. Tacrolimus 0-5 matrix metallopeptidase 9 Homo sapiens 90-95 15586415-13 2004 FK506 can inhibit the proliferation of ECV304 cells induced by rhHGF and decrease MMP-9 protein level. Tacrolimus 0-5 matrix metallopeptidase 9 Homo sapiens 82-87 15502717-0 2004 Influence of CYP3A5 and MDR1 (ABCB1) polymorphisms on the pharmacokinetics of tacrolimus in renal transplant recipients. Tacrolimus 78-88 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 15502717-10 2004 CONCLUSIONS: Kidney transplant recipients with the CYP3A5 *1 allele required a higher daily tacrolimus dose compared with those with the CYP3A5 *3/*3 genotype to maintain both the target trough level and AUC0-12, suggesting that this polymorphism is useful for determining the appropriate dose of tacrolimus. Tacrolimus 297-307 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 51-57 15502717-0 2004 Influence of CYP3A5 and MDR1 (ABCB1) polymorphisms on the pharmacokinetics of tacrolimus in renal transplant recipients. Tacrolimus 78-88 ATP binding cassette subfamily B member 1 Homo sapiens 24-28 15502717-0 2004 Influence of CYP3A5 and MDR1 (ABCB1) polymorphisms on the pharmacokinetics of tacrolimus in renal transplant recipients. Tacrolimus 78-88 ATP binding cassette subfamily B member 1 Homo sapiens 30-35 15502717-2 2004 Tacrolimus is a substrate for cytochrome P450 (CYP) 3A5 and p-glycoprotein encoded by CYP3A5 and MDR1 (ABCB1), respectively, having multiple single nucleotide polymorphisms. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 30-55 15502717-2 2004 Tacrolimus is a substrate for cytochrome P450 (CYP) 3A5 and p-glycoprotein encoded by CYP3A5 and MDR1 (ABCB1), respectively, having multiple single nucleotide polymorphisms. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 60-74 15502717-2 2004 Tacrolimus is a substrate for cytochrome P450 (CYP) 3A5 and p-glycoprotein encoded by CYP3A5 and MDR1 (ABCB1), respectively, having multiple single nucleotide polymorphisms. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 86-92 15502717-2 2004 Tacrolimus is a substrate for cytochrome P450 (CYP) 3A5 and p-glycoprotein encoded by CYP3A5 and MDR1 (ABCB1), respectively, having multiple single nucleotide polymorphisms. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 97-101 15502717-2 2004 Tacrolimus is a substrate for cytochrome P450 (CYP) 3A5 and p-glycoprotein encoded by CYP3A5 and MDR1 (ABCB1), respectively, having multiple single nucleotide polymorphisms. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 103-108 15502717-7 2004 RESULTS: The single tacrolimus dose per body weight was significantly higher in CYP3A5 *1 carriers than CYP3A5 *3/*3 carriers (0.143+/-0.050 vs. 0.078+/-0.031 mg/kg, P<0.001). Tacrolimus 20-30 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 80-86 15502717-7 2004 RESULTS: The single tacrolimus dose per body weight was significantly higher in CYP3A5 *1 carriers than CYP3A5 *3/*3 carriers (0.143+/-0.050 vs. 0.078+/-0.031 mg/kg, P<0.001). Tacrolimus 20-30 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 104-110 15502717-10 2004 CONCLUSIONS: Kidney transplant recipients with the CYP3A5 *1 allele required a higher daily tacrolimus dose compared with those with the CYP3A5 *3/*3 genotype to maintain both the target trough level and AUC0-12, suggesting that this polymorphism is useful for determining the appropriate dose of tacrolimus. Tacrolimus 92-102 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 51-57 15480330-11 2004 Interestingly, tacrolimus therapy was also associated with a reduction of CD8 + T cells expressing the T H 1 cytokine IFN-gamma. Tacrolimus 15-25 CD8a molecule Homo sapiens 74-77 15378610-1 2004 We investigated whether certain hydrophobic dipeptides, Leu-Ile, Leu-Pro, and Pro-Ile, which partially resemble the site on FK506 that binds to immunophilin, could stimulate glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) synthesis in cultured neurons and found only Leu-Ile to be an active dipeptide. Tacrolimus 124-129 glial cell line derived neurotrophic factor Mus musculus 174-217 15489928-2 2004 METHODS: Immunohistochemistry has been employed to study the expression of NF-kappaB in normal skin and lesional AD skin before and after using topical tacrolimus ointment. Tacrolimus 152-162 nuclear factor kappa B subunit 1 Homo sapiens 75-84 15489928-5 2004 After using topical tacrolimus ointment for three weeks , nuclear NF-kappaBp50 expressed on basal and suprabasal keratinocytes were lost and NF-kappaBp50 was expressed sparsely on basal keratinocytes cytoplasm or nuclear. Tacrolimus 20-30 nuclear factor kappa B subunit 1 Homo sapiens 66-78 15489928-5 2004 After using topical tacrolimus ointment for three weeks , nuclear NF-kappaBp50 expressed on basal and suprabasal keratinocytes were lost and NF-kappaBp50 was expressed sparsely on basal keratinocytes cytoplasm or nuclear. Tacrolimus 20-30 nuclear factor kappa B subunit 1 Homo sapiens 141-153 15489928-8 2004 Topical tacrolimus may directly or indirectly inhibit NF-kappaB nuclear expression in keratinocytes and inhibit skin innate immuno-inflammatory response in atopic dermatitis that related to NF-kappaB. Tacrolimus 8-18 nuclear factor kappa B subunit 1 Homo sapiens 54-63 15489928-8 2004 Topical tacrolimus may directly or indirectly inhibit NF-kappaB nuclear expression in keratinocytes and inhibit skin innate immuno-inflammatory response in atopic dermatitis that related to NF-kappaB. Tacrolimus 8-18 nuclear factor kappa B subunit 1 Homo sapiens 190-199 15365173-1 2004 Aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) is a member of the FK-506-binding protein family expressed specifically in retinal photoreceptors. Tacrolimus 80-86 aryl hydrocarbon receptor interacting protein like 1 Homo sapiens 0-52 15385801-9 2004 Interestingly, an additive inhibition between B1 and tacrolimus (FK506) was found in the CD69 expression stimulated by PMA/CD28 and PMA/ionomycin. Tacrolimus 53-63 Cd69 molecule Rattus norvegicus 89-93 15385801-9 2004 Interestingly, an additive inhibition between B1 and tacrolimus (FK506) was found in the CD69 expression stimulated by PMA/CD28 and PMA/ionomycin. Tacrolimus 53-63 Cd28 molecule Rattus norvegicus 123-127 15385801-9 2004 Interestingly, an additive inhibition between B1 and tacrolimus (FK506) was found in the CD69 expression stimulated by PMA/CD28 and PMA/ionomycin. Tacrolimus 65-70 Cd69 molecule Rattus norvegicus 89-93 15385801-9 2004 Interestingly, an additive inhibition between B1 and tacrolimus (FK506) was found in the CD69 expression stimulated by PMA/CD28 and PMA/ionomycin. Tacrolimus 65-70 Cd28 molecule Rattus norvegicus 123-127 15561265-7 2004 The effect of daily administration of FK506 on MGP mRNA levels in cryopreserved isografts and allografts after transplantation was also evaluated. Tacrolimus 38-43 matrix Gla protein Rattus norvegicus 47-50 15561265-10 2004 Daily administration of FK506 enhanced intragraft MGP mRNA (ninefold) in cryopreserved allografts (P < .01), but not in cryopreserved isografts. Tacrolimus 24-29 matrix Gla protein Rattus norvegicus 50-53 15365173-1 2004 Aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) is a member of the FK-506-binding protein family expressed specifically in retinal photoreceptors. Tacrolimus 80-86 aryl hydrocarbon receptor interacting protein like 1 Homo sapiens 54-59 15377358-0 2004 FK506 independently upregulates transforming growth factor beta and downregulates inducible nitric oxide synthase in cultured human keratinocytes: possible mechanisms of how tacrolimus ointment interacts with atopic skin. Tacrolimus 0-5 transforming growth factor beta 1 Homo sapiens 32-63 15340245-1 2004 The oral bioavailability of tacrolimus is low and varies considerably in humans due to first-pass metabolism by cytochrome P450 (CYP) 3A4 and the active efflux mediated by P-glycoprotein. Tacrolimus 28-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 112-137 15307840-0 2004 CYP3A4 and P-glycoprotein activity in healthy controls and transplant patients on cyclosporin vs. tacrolimus vs. sirolimus. Tacrolimus 98-108 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 15307840-0 2004 CYP3A4 and P-glycoprotein activity in healthy controls and transplant patients on cyclosporin vs. tacrolimus vs. sirolimus. Tacrolimus 98-108 ATP binding cassette subfamily B member 1 Homo sapiens 11-25 15307840-1 2004 This study aimed to determine the impact of maintenance immunosuppressive therapy with cyclosporin A (CsA), tacrolimus (FK506) and sirolimus (Rapa) on the in vivo activity of both intestinal and hepatic cytochrome P450 3A4 (CYP3A4) and P-glycoprotein (PGP) in renal transplant patients. Tacrolimus 120-125 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 203-222 15307840-1 2004 This study aimed to determine the impact of maintenance immunosuppressive therapy with cyclosporin A (CsA), tacrolimus (FK506) and sirolimus (Rapa) on the in vivo activity of both intestinal and hepatic cytochrome P450 3A4 (CYP3A4) and P-glycoprotein (PGP) in renal transplant patients. Tacrolimus 120-125 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 224-230 15307840-1 2004 This study aimed to determine the impact of maintenance immunosuppressive therapy with cyclosporin A (CsA), tacrolimus (FK506) and sirolimus (Rapa) on the in vivo activity of both intestinal and hepatic cytochrome P450 3A4 (CYP3A4) and P-glycoprotein (PGP) in renal transplant patients. Tacrolimus 120-125 ATP binding cassette subfamily B member 1 Homo sapiens 236-250 15307840-1 2004 This study aimed to determine the impact of maintenance immunosuppressive therapy with cyclosporin A (CsA), tacrolimus (FK506) and sirolimus (Rapa) on the in vivo activity of both intestinal and hepatic cytochrome P450 3A4 (CYP3A4) and P-glycoprotein (PGP) in renal transplant patients. Tacrolimus 120-125 ATP binding cassette subfamily B member 1 Homo sapiens 252-255 15307840-5 2004 A significant increase in intestinal CYP3A4 activity and a significant decrease in hepatic and intestinal PGP activity was seen in patients on CsA in comparison with those on FK506 or Rapa (p < 0.01). Tacrolimus 175-180 ATP binding cassette subfamily B member 1 Homo sapiens 106-109 15377358-0 2004 FK506 independently upregulates transforming growth factor beta and downregulates inducible nitric oxide synthase in cultured human keratinocytes: possible mechanisms of how tacrolimus ointment interacts with atopic skin. Tacrolimus 0-5 nitric oxide synthase 2 Homo sapiens 92-113 15377358-4 2004 OBJECTIVES: To investigate the direct effects of FK506 on KCs in terms of TGF-beta and inducible NOS (iNOS), and to explore the interactions between TGF-beta and iNOS in the KC system. Tacrolimus 49-54 transforming growth factor beta 1 Homo sapiens 74-82 15377358-4 2004 OBJECTIVES: To investigate the direct effects of FK506 on KCs in terms of TGF-beta and inducible NOS (iNOS), and to explore the interactions between TGF-beta and iNOS in the KC system. Tacrolimus 49-54 nitric oxide synthase 2 Homo sapiens 87-100 15377358-4 2004 OBJECTIVES: To investigate the direct effects of FK506 on KCs in terms of TGF-beta and inducible NOS (iNOS), and to explore the interactions between TGF-beta and iNOS in the KC system. Tacrolimus 49-54 nitric oxide synthase 2 Homo sapiens 102-106 15377358-6 2004 The changes in the KC system induced by FK506 were documented in terms of TGF-beta and iNOS using enzyme-linked immunosorbent assay and Western blotting techniques, respectively. Tacrolimus 40-45 transforming growth factor beta 1 Homo sapiens 74-82 15377358-6 2004 The changes in the KC system induced by FK506 were documented in terms of TGF-beta and iNOS using enzyme-linked immunosorbent assay and Western blotting techniques, respectively. Tacrolimus 40-45 nitric oxide synthase 2 Homo sapiens 87-91 15377358-9 2004 RESULTS: Our results showed that the release of TGF-beta was upregulated in FK506-treated KCs, particularly in the presence of TNF-alpha, while the expression of iNOS was downregulated. Tacrolimus 76-81 transforming growth factor beta 1 Homo sapiens 48-56 15377358-9 2004 RESULTS: Our results showed that the release of TGF-beta was upregulated in FK506-treated KCs, particularly in the presence of TNF-alpha, while the expression of iNOS was downregulated. Tacrolimus 76-81 tumor necrosis factor Homo sapiens 127-136 15377358-9 2004 RESULTS: Our results showed that the release of TGF-beta was upregulated in FK506-treated KCs, particularly in the presence of TNF-alpha, while the expression of iNOS was downregulated. Tacrolimus 76-81 nitric oxide synthase 2 Homo sapiens 162-166 15377358-11 2004 However, the addition of TNF-alpha did not further downregulate the expression of iNOS protein, suggesting that FK506 may regulate TGF-beta and iNOS through different pathways. Tacrolimus 112-117 transforming growth factor beta 1 Homo sapiens 131-139 15377358-11 2004 However, the addition of TNF-alpha did not further downregulate the expression of iNOS protein, suggesting that FK506 may regulate TGF-beta and iNOS through different pathways. Tacrolimus 112-117 nitric oxide synthase 2 Homo sapiens 144-148 15518758-12 2004 Tacrolimus shows large variability in bioavailability after oral administration, both due to intestinal metabolism by cytochrome P450 (CYP3A4) and active secretion from enterocyte into intestinal lumen by P-glycoprotein. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 135-141 15551000-9 2004 Therapeutic treatment with FK506 reduced the elevated level of IL-6, but not IL-1beta, in paw tissue. Tacrolimus 27-32 interleukin 6 Rattus norvegicus 63-67 15551000-13 2004 FK506 may improve arthritis in established stages of CIA, by reducing the elevated level of IL-6. Tacrolimus 0-5 interleukin 6 Rattus norvegicus 92-96 15518791-0 2004 Excellent outcome of ABO-incompatible living kidney transplantation under pretransplantation immunosuppression with tacrolimus, mycophenolate mofetil, and steroid. Tacrolimus 116-126 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 21-24 15199065-8 2004 The binding of FKBP12 and FKBP52 was specific and could be displaced by the immunosuppressant drug FK506, at concentrations of 0.5 and 10 microm, respectively. Tacrolimus 99-104 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 15-21 15284440-3 2004 Fused to the rapamycin-binding domain (FRB) of the kinase mammalian target of rapamycin and FK506-binding protein 12 (FKBP), respectively, the optimized FRB-N-terminal luciferase fragment (NLuc)/C-terminal luciferase fragment (CLuc)-FKBP luciferase complementation imaging (LCI) pair reconstituted luciferase activity in cells upon single-site binding of rapamycin in an FK506-competitive manner. Tacrolimus 92-97 FKBP prolyl isomerase 1A Homo sapiens 118-122 15199065-12 2004 Receptor-mediated activation of TRPC6, stably expressed in HEK cells, was significantly inhibited by FK506, which also disrupted interaction between TRPC6 and the endogenous immunophilin found in HEK cells. Tacrolimus 101-106 transient receptor potential cation channel subfamily C member 6 Homo sapiens 32-37 15199065-12 2004 Receptor-mediated activation of TRPC6, stably expressed in HEK cells, was significantly inhibited by FK506, which also disrupted interaction between TRPC6 and the endogenous immunophilin found in HEK cells. Tacrolimus 101-106 transient receptor potential cation channel subfamily C member 6 Homo sapiens 149-154 15199065-8 2004 The binding of FKBP12 and FKBP52 was specific and could be displaced by the immunosuppressant drug FK506, at concentrations of 0.5 and 10 microm, respectively. Tacrolimus 99-104 FKBP prolyl isomerase 4 Homo sapiens 26-32 15207740-8 2004 Treatment with CsA or FK506 produced nuclear fragmentation and disruption of the multinucleated osteoclasts and an increase in caspase-3 activity. Tacrolimus 22-27 caspase 3 Mus musculus 127-136 15316356-6 2004 It has been also demonstrated that there is a link between the polymorphisms of the cytochrome P450 3A5, 3A4 and the multidrug resistance-1 (MDR1) genes, and the daily dose necessary to achieve adequate blood tacrolimus levels. Tacrolimus 209-219 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 84-103 15316356-6 2004 It has been also demonstrated that there is a link between the polymorphisms of the cytochrome P450 3A5, 3A4 and the multidrug resistance-1 (MDR1) genes, and the daily dose necessary to achieve adequate blood tacrolimus levels. Tacrolimus 209-219 ATP binding cassette subfamily B member 1 Homo sapiens 117-139 15316356-6 2004 It has been also demonstrated that there is a link between the polymorphisms of the cytochrome P450 3A5, 3A4 and the multidrug resistance-1 (MDR1) genes, and the daily dose necessary to achieve adequate blood tacrolimus levels. Tacrolimus 209-219 ATP binding cassette subfamily B member 1 Homo sapiens 141-145 15316360-0 2004 Acute nephrotoxicity of tacrolimus and sirolimus in renal isografts: differential intragraft expression of transforming growth factor-beta1 and alpha-smooth muscle actin. Tacrolimus 24-34 transforming growth factor, beta 1 Rattus norvegicus 107-139 15316360-0 2004 Acute nephrotoxicity of tacrolimus and sirolimus in renal isografts: differential intragraft expression of transforming growth factor-beta1 and alpha-smooth muscle actin. Tacrolimus 24-34 actin gamma 2, smooth muscle Rattus norvegicus 144-169 15044154-2 2004 The immunosuppressant FK506 inhibits the Ca(2+)-calmodulin-dependent phosphatase calcineurin in pancreatic acinar cells and blocks pancreatic growth induced by chronic CCK treatment. Tacrolimus 22-27 cholecystokinin Rattus norvegicus 168-171 15044154-5 2004 The immunosuppressant FK506 dose-dependently inhibited CCK-stimulated protein synthesis over the same concentration range that blocked calcineurin activity, as assessed by dephosphorylation of the calcineurin substrate calcium-regulated heat-stable protein of 24 kDa. Tacrolimus 22-27 cholecystokinin Rattus norvegicus 55-58 15197524-1 2004 OBJECTIVE: The aim of this study was to (a) quantify the gene expression of some cytochromes P(450) (CYP), especially CYP3A4, in serial biopsies from liver grafts the first year after orthoptic liver transplantation (OLT) and (b) study the relationship between hepatic CYP3A4 gene expression and plasma levels of cyclosporine and tacrolimus. Tacrolimus 330-340 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 118-124 15197524-7 2004 The gene expression of CYP3A4 was related to the plasma concentration of cyclosporine and tacrolimus, i.e. low mRNA concentrations corresponded to high serum concentration levels and vice versa. Tacrolimus 90-100 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 23-29 15197524-11 2004 Low CYP3A4 gene expression was related to high plasma levels of cyclosporine and tacrolimus and vice versa. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 4-10 15285851-1 2004 The aim of this study was to investigate the effects of the proton pump inhibitors (PPIs), lansoprazole and rabeprazole, on tacrolimus pharmacokinetics in healthy volunteers with mutations in the cytochrome P450 (CYP) 2C19 gene (CYP2C19). Tacrolimus 124-134 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 196-222 15285851-7 2004 Large individual variation was observed in the effects of lansoprazole on tacrolimus AUC0-8 owing to CYP2C19 genotype status. Tacrolimus 74-84 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 101-108 15285851-8 2004 The percent change for tacrolimus AUC0-8 in subjects with and without CYP2C19 mutant alleles was 81% and 29%, respectively. Tacrolimus 23-33 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 70-77 15285851-10 2004 These observations suggest that drug interaction between tacrolimus and lansoprazole occurs in subjects with higher lansoprazole blood concentrations corresponding to CYP2C19 genetic status. Tacrolimus 57-67 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 167-174 15260130-4 2004 However, pre-treatment with FK506 and V10367 significantly prevented any increase in this ratio or p53 mRNA expression. Tacrolimus 28-33 tumor protein p53 Homo sapiens 99-102 15260130-9 2004 In U251 cells, FK506, GPI1046 and V10367 had GSH-activating and NTF-activating effects. Tacrolimus 15-20 neurotrophin 3 Homo sapiens 64-67 15212998-6 2004 Tacrolimus also significantly decreased microglial activation at 8 h and inflammatory markers (cytokine-induced neutrophil chemoattractant and myeloperoxidase [MPO] activity) at 24 h after reperfusion in the ischemic cortex but not in the caudate putamen. Tacrolimus 0-10 myeloperoxidase Rattus norvegicus 143-158 15212998-6 2004 Tacrolimus also significantly decreased microglial activation at 8 h and inflammatory markers (cytokine-induced neutrophil chemoattractant and myeloperoxidase [MPO] activity) at 24 h after reperfusion in the ischemic cortex but not in the caudate putamen. Tacrolimus 0-10 myeloperoxidase Rattus norvegicus 160-163 15133031-6 2004 The interaction between FKBP52 and Atox1 was observed in both glutathione S-transferase pull-down experiments and when proteins were ectopically expressed in human embryonic kidney (HEK) 293T cells and was sensitive to FK506. Tacrolimus 219-224 FKBP prolyl isomerase 4 Homo sapiens 24-30 15133031-6 2004 The interaction between FKBP52 and Atox1 was observed in both glutathione S-transferase pull-down experiments and when proteins were ectopically expressed in human embryonic kidney (HEK) 293T cells and was sensitive to FK506. Tacrolimus 219-224 antioxidant 1 copper chaperone Homo sapiens 35-40 15207740-2 2004 Cyclosporin A (CsA), cyclosporin B (CsB), cyclosporin H (CsH), and FK506 all inhibited receptor activator of NFkappaB ligand (RANKL)-stimulated tartrate-resistant acid phosphatase (TRAP) activity and generation of TRAP+ multinucleated cells in the cultures. Tacrolimus 67-72 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 87-124 15207740-2 2004 Cyclosporin A (CsA), cyclosporin B (CsB), cyclosporin H (CsH), and FK506 all inhibited receptor activator of NFkappaB ligand (RANKL)-stimulated tartrate-resistant acid phosphatase (TRAP) activity and generation of TRAP+ multinucleated cells in the cultures. Tacrolimus 67-72 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 126-131 15207740-2 2004 Cyclosporin A (CsA), cyclosporin B (CsB), cyclosporin H (CsH), and FK506 all inhibited receptor activator of NFkappaB ligand (RANKL)-stimulated tartrate-resistant acid phosphatase (TRAP) activity and generation of TRAP+ multinucleated cells in the cultures. Tacrolimus 67-72 acid phosphatase 5, tartrate resistant Mus musculus 144-179 15207740-2 2004 Cyclosporin A (CsA), cyclosporin B (CsB), cyclosporin H (CsH), and FK506 all inhibited receptor activator of NFkappaB ligand (RANKL)-stimulated tartrate-resistant acid phosphatase (TRAP) activity and generation of TRAP+ multinucleated cells in the cultures. Tacrolimus 67-72 acid phosphatase 5, tartrate resistant Mus musculus 181-185 15207740-2 2004 Cyclosporin A (CsA), cyclosporin B (CsB), cyclosporin H (CsH), and FK506 all inhibited receptor activator of NFkappaB ligand (RANKL)-stimulated tartrate-resistant acid phosphatase (TRAP) activity and generation of TRAP+ multinucleated cells in the cultures. Tacrolimus 67-72 acid phosphatase 5, tartrate resistant Mus musculus 214-218 15207740-7 2004 When CsA or FK506 were added for 1 day to cultures in which osteoclasts had already formed, the numbers of TRAP+ osteoclasts decreased. Tacrolimus 12-17 acid phosphatase 5, tartrate resistant Mus musculus 107-111 15207740-9 2004 The apoptosis inhibitor z-VAD partially prevented the inhibitory effects of CsA and FK506 on the survival of TRAP+ multinucleated cells in the cultures and also preserved the normal osteoclast morphology. Tacrolimus 84-89 acid phosphatase 5, tartrate resistant Mus musculus 109-113 15241357-8 2004 RESULTS: Tacrolimus and steroid ointment induced a selective depletion of IDECs from the epidermis and reduced the expression of the costimulatory molecules CD80 and CD86. Tacrolimus 9-19 CD80 molecule Homo sapiens 157-161 15241357-8 2004 RESULTS: Tacrolimus and steroid ointment induced a selective depletion of IDECs from the epidermis and reduced the expression of the costimulatory molecules CD80 and CD86. Tacrolimus 9-19 CD86 molecule Homo sapiens 166-170 15243524-18 2004 Suppression of TNF-alpha after topical tacrolimus application may be associated with repigmentation of vitiligo. Tacrolimus 39-49 tumor necrosis factor Homo sapiens 15-24 15226679-0 2004 CYP3A5*1-carrying graft liver reduces the concentration/oral dose ratio of tacrolimus in recipients of living-donor liver transplantation. Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 15263820-5 2004 Moreover, VEGF-stimulated induction of DSCR1 was blocked by anti-VEGF receptor-2 monoclonal antibody (mAb), or the specific calcineurin inhibitors cyclosporin A and FK506. Tacrolimus 165-170 vascular endothelial growth factor A Homo sapiens 10-14 15263820-5 2004 Moreover, VEGF-stimulated induction of DSCR1 was blocked by anti-VEGF receptor-2 monoclonal antibody (mAb), or the specific calcineurin inhibitors cyclosporin A and FK506. Tacrolimus 165-170 regulator of calcineurin 1 Homo sapiens 39-44 15226679-6 2004 The mRNA level of CYP3A5 was significantly reduced by the *3/*3 genotype, and the tacrolimus C/D ratio was decreased in recipients engrafted with partial liver carrying CYP3A5*1/*1 genotype. Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 169-175 15226679-7 2004 An analysis of the combination of intestinal MDR1 level and liver CYP3A5 genotype revealed that the tacrolimus C/D ratio was lower in the group with higher MDR1 levels regardless of CYP3A5 genotype during postoperative week 1. Tacrolimus 100-110 ATP binding cassette subfamily B member 1 Homo sapiens 45-49 15226679-7 2004 An analysis of the combination of intestinal MDR1 level and liver CYP3A5 genotype revealed that the tacrolimus C/D ratio was lower in the group with higher MDR1 levels regardless of CYP3A5 genotype during postoperative week 1. Tacrolimus 100-110 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 66-72 15226679-7 2004 An analysis of the combination of intestinal MDR1 level and liver CYP3A5 genotype revealed that the tacrolimus C/D ratio was lower in the group with higher MDR1 levels regardless of CYP3A5 genotype during postoperative week 1. Tacrolimus 100-110 ATP binding cassette subfamily B member 1 Homo sapiens 156-160 15226679-8 2004 CONCLUSIONS: These results indicate that in recipients of LDLT, the pharmacokinetics of tacrolimus is influenced by flux via P-glycoprotein in the intestine during the first week; after that, it is mostly the hepatic metabolism that contributes to the excretion of tacrolimus, and carriers of the CYP3A5*1/*1 genotype require a high dose of tacrolimus to achieve the target concentration. Tacrolimus 88-98 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 297-303 15147425-1 2004 Previously, we reported that, at 3 months after renal transplantation, individuals with CYP3AP1 genotype CYP3AP1*1 (linked to CYP3A5*1 and strongly associated with expression of CYP3A5) required twofold higher doses of tacrolimus to achieve target blood concentrations than individuals with the genotype CYP3AP1*3/*3 (CYP3A5 nonexpressors). Tacrolimus 219-229 cytochrome P450 family 3 subfamily A member 51, pseudogene Homo sapiens 88-95 15223905-7 2004 Most of the patients received anti-interleukin (IL)-2 receptor monoclonal antibody induction and tacrolimus-based maintenance immunosuppression, which resulted in a low incidence of aRx. Tacrolimus 97-107 aristaless related homeobox Homo sapiens 182-185 15147425-1 2004 Previously, we reported that, at 3 months after renal transplantation, individuals with CYP3AP1 genotype CYP3AP1*1 (linked to CYP3A5*1 and strongly associated with expression of CYP3A5) required twofold higher doses of tacrolimus to achieve target blood concentrations than individuals with the genotype CYP3AP1*3/*3 (CYP3A5 nonexpressors). Tacrolimus 219-229 cytochrome P450 family 3 subfamily A member 51, pseudogene Homo sapiens 105-112 15147425-1 2004 Previously, we reported that, at 3 months after renal transplantation, individuals with CYP3AP1 genotype CYP3AP1*1 (linked to CYP3A5*1 and strongly associated with expression of CYP3A5) required twofold higher doses of tacrolimus to achieve target blood concentrations than individuals with the genotype CYP3AP1*3/*3 (CYP3A5 nonexpressors). Tacrolimus 219-229 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 126-132 15147425-1 2004 Previously, we reported that, at 3 months after renal transplantation, individuals with CYP3AP1 genotype CYP3AP1*1 (linked to CYP3A5*1 and strongly associated with expression of CYP3A5) required twofold higher doses of tacrolimus to achieve target blood concentrations than individuals with the genotype CYP3AP1*3/*3 (CYP3A5 nonexpressors). Tacrolimus 219-229 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 178-184 15147425-1 2004 Previously, we reported that, at 3 months after renal transplantation, individuals with CYP3AP1 genotype CYP3AP1*1 (linked to CYP3A5*1 and strongly associated with expression of CYP3A5) required twofold higher doses of tacrolimus to achieve target blood concentrations than individuals with the genotype CYP3AP1*3/*3 (CYP3A5 nonexpressors). Tacrolimus 219-229 cytochrome P450 family 3 subfamily A member 51, pseudogene Homo sapiens 105-112 15147425-1 2004 Previously, we reported that, at 3 months after renal transplantation, individuals with CYP3AP1 genotype CYP3AP1*1 (linked to CYP3A5*1 and strongly associated with expression of CYP3A5) required twofold higher doses of tacrolimus to achieve target blood concentrations than individuals with the genotype CYP3AP1*3/*3 (CYP3A5 nonexpressors). Tacrolimus 219-229 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 178-184 15147425-3 2004 Patients with CYP3AP1*1/*3 or *1/*1 had lower mean tacrolimus concentrations during the first week (Median 13.5 vs. 18.5 microg/L, p < 0.0001) with significant delay in achieving target concentrations (15-20 microg/L during week 1, then 10-15 microg/L). Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 51, pseudogene Homo sapiens 14-21 15147425-4 2004 More CYP3AP1*3/*3 patients had tacrolimus concentrations above target during the first week (73.6% vs. 35.8%, p = 0.003). Tacrolimus 31-41 cytochrome P450 family 3 subfamily A member 51, pseudogene Homo sapiens 5-12 15147425-6 2004 In conclusion, an initial dosing regimen for tacrolimus based on knowledge of the CYP3AP1 genotype and subsequently guided by concentration measurements has the potential to increase the proportion of patients achieving target blood concentrations early after transplantation. Tacrolimus 45-55 cytochrome P450 family 3 subfamily A member 51, pseudogene Homo sapiens 82-89 15149578-7 2004 However, tacrolimus, a potent calcineurin inhibitor and inhibitor of interleukin-2 expression, has shown efficacy in Crohn"s disease, albeit at the cost of substantial potential toxicity. Tacrolimus 9-19 interleukin 2 Homo sapiens 69-82 15214906-9 2004 One-year treatment with tacrolimus ointment was associated with an increase in collagen synthesis; the median increase in combined procollagen propeptide levels was 272 micro g L-1 (+ 140.9%, P < 0.001) and was accompanied by a significant increase in skin thickness. Tacrolimus 24-34 immunoglobulin kappa variable 1-16 Homo sapiens 177-180 15214910-2 2004 We report for the first time two patients with ACS which was successfully treated with topical tacrolimus 0.1% ointment. Tacrolimus 95-105 1-aminocyclopropane-1-carboxylate synthase homolog (inactive) Homo sapiens 47-50 15153522-8 2004 As a downstream of calpain, calcineurin was activated and the inhibition of calcineurin activation by FK506 diminished insulinoma cell death by IFN-gamma/TNF-alpha. Tacrolimus 102-107 interferon gamma Mus musculus 144-153 15499186-4 2004 Since it has been reported that tacrolimus absorption is regulated mainly by Cytochrome P-450 (CYP) mediated metabolism in the jejunum, but by P-glycoprotein (P-gp) mediated efflux in the ileum, these factors might contribute to the changes in intestinal absorption of tacrolimus in rat of acute renal failure. Tacrolimus 32-42 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 77-93 15499186-4 2004 Since it has been reported that tacrolimus absorption is regulated mainly by Cytochrome P-450 (CYP) mediated metabolism in the jejunum, but by P-glycoprotein (P-gp) mediated efflux in the ileum, these factors might contribute to the changes in intestinal absorption of tacrolimus in rat of acute renal failure. Tacrolimus 32-42 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 95-98 15499186-4 2004 Since it has been reported that tacrolimus absorption is regulated mainly by Cytochrome P-450 (CYP) mediated metabolism in the jejunum, but by P-glycoprotein (P-gp) mediated efflux in the ileum, these factors might contribute to the changes in intestinal absorption of tacrolimus in rat of acute renal failure. Tacrolimus 32-42 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 143-157 15499186-4 2004 Since it has been reported that tacrolimus absorption is regulated mainly by Cytochrome P-450 (CYP) mediated metabolism in the jejunum, but by P-glycoprotein (P-gp) mediated efflux in the ileum, these factors might contribute to the changes in intestinal absorption of tacrolimus in rat of acute renal failure. Tacrolimus 32-42 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 159-163 15499186-4 2004 Since it has been reported that tacrolimus absorption is regulated mainly by Cytochrome P-450 (CYP) mediated metabolism in the jejunum, but by P-glycoprotein (P-gp) mediated efflux in the ileum, these factors might contribute to the changes in intestinal absorption of tacrolimus in rat of acute renal failure. Tacrolimus 269-279 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 159-163 15194145-8 2004 Tacrolimus, CsA and Dex individually suppressed TARC production by PBMCs from AD patients which were co-cultured with DME for 7 days. Tacrolimus 0-10 C-C motif chemokine ligand 17 Homo sapiens 48-52 15153522-8 2004 As a downstream of calpain, calcineurin was activated and the inhibition of calcineurin activation by FK506 diminished insulinoma cell death by IFN-gamma/TNF-alpha. Tacrolimus 102-107 tumor necrosis factor Mus musculus 154-163 15153522-9 2004 BAD phosphorylation was decreased by IFN-gamma/TNF-alpha because of the increased calcineurin activity, which was reversed by FK506. Tacrolimus 126-131 interferon gamma Mus musculus 37-46 15153522-9 2004 BAD phosphorylation was decreased by IFN-gamma/TNF-alpha because of the increased calcineurin activity, which was reversed by FK506. Tacrolimus 126-131 tumor necrosis factor Mus musculus 47-56 15159550-1 2004 FK506-binding protein 52 (FKBP52), which binds FK506 and possesses peptidylprolyl isomerase activity, is an important immunophilin involved in the heterocomplex of steroid receptors with heat-shock protein 90. Tacrolimus 0-5 FKBP prolyl isomerase 4 Homo sapiens 26-32 15162468-0 2004 Significant enhancement by anti-ICOS antibody of suboptimal tacrolimus immunosuppression in rat liver transplantation. Tacrolimus 60-70 inducible T-cell co-stimulator Rattus norvegicus 32-36 15162468-4 2004 However, in this article, we report that anti-rat ICOS antibody markedly enhanced the immunosuppressive activity of a suboptimal dose of tacrolimus (FK506). Tacrolimus 137-147 inducible T-cell co-stimulator Rattus norvegicus 50-54 15162468-4 2004 However, in this article, we report that anti-rat ICOS antibody markedly enhanced the immunosuppressive activity of a suboptimal dose of tacrolimus (FK506). Tacrolimus 149-154 inducible T-cell co-stimulator Rattus norvegicus 50-54 15047861-7 2004 These phenotypes observed in apm1 mutations were accentuated upon temperature up-shift and FK506 treatment. Tacrolimus 91-96 zinc finger and BTB domain containing 7C Homo sapiens 29-33 14684381-9 2004 In summary, 1) CCK released by chronic camostat feeding induces pancreatic growth in mice; 2) this growth is blocked by treatment with both CsA and FK506, indicating a role for CN; 3) CCK stimulation also increases CN protein. Tacrolimus 148-153 cholecystokinin Mus musculus 15-18 15251325-1 2004 Tacrolimus-induced toxicity is considered a dose-related side effect largely due to a direct action of this potent calcineurin inhibitor on its targets including the kidney and the pancreas. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 115-136 15159506-3 2004 In a preliminary open trial with FK506, immunosuppressant and enhancer of RyR-related sarcoplasmic calcium release, the authors observed the sustained benefits in anti-RyR-positive MG patients. Tacrolimus 33-38 ryanodine receptor 1 Homo sapiens 168-171 15144206-6 2004 We demonstrate that interactions of the type I receptor for TGFbeta with FKBP12 and the epidermal growth factor receptor (EGFR) with p85 are effectively disrupted by FK506 and EGFR kinase inhibitor AG1478, respectively. Tacrolimus 166-171 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 73-79 15144206-6 2004 We demonstrate that interactions of the type I receptor for TGFbeta with FKBP12 and the epidermal growth factor receptor (EGFR) with p85 are effectively disrupted by FK506 and EGFR kinase inhibitor AG1478, respectively. Tacrolimus 166-171 epidermal growth factor receptor Homo sapiens 88-120 15144206-6 2004 We demonstrate that interactions of the type I receptor for TGFbeta with FKBP12 and the epidermal growth factor receptor (EGFR) with p85 are effectively disrupted by FK506 and EGFR kinase inhibitor AG1478, respectively. Tacrolimus 166-171 epidermal growth factor receptor Homo sapiens 122-126 15144206-6 2004 We demonstrate that interactions of the type I receptor for TGFbeta with FKBP12 and the epidermal growth factor receptor (EGFR) with p85 are effectively disrupted by FK506 and EGFR kinase inhibitor AG1478, respectively. Tacrolimus 166-171 phosphoinositide-3-kinase regulatory subunit 2 Homo sapiens 133-136 15146068-0 2004 Identification of Ald6p as the target of a class of small-molecule suppressors of FK506 and their use in network dissection. Tacrolimus 82-87 aldehyde dehydrogenase (NADP(+)) ALD6 Saccharomyces cerevisiae S288C 18-23 15146068-9 2004 Furthermore, growth improvement by the SFKs on high NaCl plus FK506 was shown to require GPD1, which encodes an NADH-dependent glycerol-3-phosphate dehydrogenase that is important for the production of glycerol in response to osmotic stress. Tacrolimus 62-67 glycerol-3-phosphate dehydrogenase (NAD(+)) GPD1 Saccharomyces cerevisiae S288C 89-93 15126071-8 2004 The enhancement of IL-4 production by OP was blocked by FK506, a calcineurin inhibitor, but not by the estrogen receptor (ER) antagonist ICI 182780. Tacrolimus 56-61 interleukin 4 Mus musculus 19-23 15126071-9 2004 FK506 inhibited the NF-AT-DNA binding activity and IL-4 gene promoter activity enhanced by OP in a dose-dependent manner. Tacrolimus 0-5 interleukin 4 Mus musculus 51-55 14684381-9 2004 In summary, 1) CCK released by chronic camostat feeding induces pancreatic growth in mice; 2) this growth is blocked by treatment with both CsA and FK506, indicating a role for CN; 3) CCK stimulation also increases CN protein. Tacrolimus 148-153 cholecystokinin Mus musculus 184-187 15081597-6 2004 In a separate study, subcutaneous injections of FK506 (2 or 10 mg/kg) for 2 weeks markedly increased heat shock protein-70 (Hsp-70) immunostaining in sensory neurons, motor neurons, Purkinje cells, and other regions of the brain (in particular, the amygdala) from nonintoxicated and AC-intoxicated rats compared to controls. Tacrolimus 48-53 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 101-122 15010519-0 2004 Lansoprazole-tacrolimus interaction in Japanese transplant recipient with CYP2C19 polymorphism. Tacrolimus 13-23 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 74-81 15010519-9 2004 Since tacrolimus is also metabolized by CYP3A4, the blood concentration of tacrolimus in this patient who had a CYP2C19 gene mutation may have been elevated by decreased hepatic elimination of lansoprazole. Tacrolimus 6-16 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-46 15010519-9 2004 Since tacrolimus is also metabolized by CYP3A4, the blood concentration of tacrolimus in this patient who had a CYP2C19 gene mutation may have been elevated by decreased hepatic elimination of lansoprazole. Tacrolimus 6-16 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 112-119 15010519-9 2004 Since tacrolimus is also metabolized by CYP3A4, the blood concentration of tacrolimus in this patient who had a CYP2C19 gene mutation may have been elevated by decreased hepatic elimination of lansoprazole. Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-46 15010519-9 2004 Since tacrolimus is also metabolized by CYP3A4, the blood concentration of tacrolimus in this patient who had a CYP2C19 gene mutation may have been elevated by decreased hepatic elimination of lansoprazole. Tacrolimus 75-85 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 112-119 15115509-3 2004 Topical tacrolimus may inhibit local activation of T lymphocytes through altered expression of cytokines such as interleukin-1, -4 and -5, tumour necrosis factor-alpha and interferon-gamma. Tacrolimus 8-18 interleukin 1 alpha Homo sapiens 113-167 15115509-3 2004 Topical tacrolimus may inhibit local activation of T lymphocytes through altered expression of cytokines such as interleukin-1, -4 and -5, tumour necrosis factor-alpha and interferon-gamma. Tacrolimus 8-18 interferon gamma Homo sapiens 172-188 15081597-8 2004 Thus, the ability of FK506 to increase Hsp-70 expression may underlie its neuroprotective action. Tacrolimus 21-26 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 39-45 15497458-1 2004 FKBP1B belongs to immunophilins superfamily and functions as a cytosolic receptor protein of FK506. Tacrolimus 93-98 FKBP prolyl isomerase 1B Homo sapiens 0-6 15497458-2 2004 The role of FKBP1B in the immunosuppressive pathway of FK506 is well established. Tacrolimus 55-60 FKBP prolyl isomerase 1B Homo sapiens 12-18 15081597-6 2004 In a separate study, subcutaneous injections of FK506 (2 or 10 mg/kg) for 2 weeks markedly increased heat shock protein-70 (Hsp-70) immunostaining in sensory neurons, motor neurons, Purkinje cells, and other regions of the brain (in particular, the amygdala) from nonintoxicated and AC-intoxicated rats compared to controls. Tacrolimus 48-53 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 124-130 15118362-8 2004 In addition, this SAM-induced increase in RyR/Ca(2+) channel activity was blocked by 30 microM ryanodine and by FK506 (100 microM), a ligand for the RyR accessory protein. Tacrolimus 112-117 ryanodine receptor 1 Homo sapiens 42-45 15153659-4 2004 FK506, a specific calcineurin inhibitor, inhibited the enhancing effect of adenosine on the NGF-induced neurite outgrowth and increased the duration of p38 MAP kinase phosphorylation without affecting ERK phosphorylation. Tacrolimus 0-5 nerve growth factor Rattus norvegicus 92-95 15153659-4 2004 FK506, a specific calcineurin inhibitor, inhibited the enhancing effect of adenosine on the NGF-induced neurite outgrowth and increased the duration of p38 MAP kinase phosphorylation without affecting ERK phosphorylation. Tacrolimus 0-5 mitogen activated protein kinase 14 Rattus norvegicus 152-155 15118362-8 2004 In addition, this SAM-induced increase in RyR/Ca(2+) channel activity was blocked by 30 microM ryanodine and by FK506 (100 microM), a ligand for the RyR accessory protein. Tacrolimus 112-117 ryanodine receptor 1 Homo sapiens 149-152 15067199-0 2004 FK506 requires stimulation of the extracellular signal-regulated kinase 1/2 and the steroid receptor chaperone protein p23 for neurite elongation. Tacrolimus 0-5 mitogen-activated protein kinase 1 Homo sapiens 34-100 15067199-0 2004 FK506 requires stimulation of the extracellular signal-regulated kinase 1/2 and the steroid receptor chaperone protein p23 for neurite elongation. Tacrolimus 0-5 prostaglandin E synthase 3 Homo sapiens 119-122 15067199-3 2004 Here, we tested the involvement of the ERK and p23 in neurite elongation by FK506 in human SH-SY5Y cells. Tacrolimus 76-81 mitogen-activated protein kinase 1 Homo sapiens 39-42 15067199-4 2004 FK506 (10 nM) increased ERK1/2 phosphorylation at 12 and 24 h, eliciting a 3.5-fold increase at 24 h, which was inhibited in a concentration-dependent manner by an antibody (JJ3) to recombinant human p23. Tacrolimus 0-5 mitogen-activated protein kinase 3 Homo sapiens 24-30 15067199-4 2004 FK506 (10 nM) increased ERK1/2 phosphorylation at 12 and 24 h, eliciting a 3.5-fold increase at 24 h, which was inhibited in a concentration-dependent manner by an antibody (JJ3) to recombinant human p23. Tacrolimus 0-5 prostaglandin E synthase 3 Homo sapiens 200-203 15067199-5 2004 Neurite elongation by FK506 (10 nM), determined by measuring neurite lengths at 96 and 168 h, was completely blocked by the mitogen-activated protein kinase inhibitor PD 098059 (10 microM) and prevented, in a concentration-dependent fashion, by the p23 antibody. Tacrolimus 22-27 prostaglandin E synthase 3 Homo sapiens 249-252 15067199-6 2004 Taken together, the results demonstrate the functional role for ERK and p23 in the neurite elongation activity of FK506 and reveal a novel signal transduction pathway involving p23 activation of ERK. Tacrolimus 114-119 mitogen-activated protein kinase 1 Homo sapiens 64-67 15067199-6 2004 Taken together, the results demonstrate the functional role for ERK and p23 in the neurite elongation activity of FK506 and reveal a novel signal transduction pathway involving p23 activation of ERK. Tacrolimus 114-119 prostaglandin E synthase 3 Homo sapiens 72-75 15067199-6 2004 Taken together, the results demonstrate the functional role for ERK and p23 in the neurite elongation activity of FK506 and reveal a novel signal transduction pathway involving p23 activation of ERK. Tacrolimus 114-119 prostaglandin E synthase 3 Homo sapiens 177-180 15067199-6 2004 Taken together, the results demonstrate the functional role for ERK and p23 in the neurite elongation activity of FK506 and reveal a novel signal transduction pathway involving p23 activation of ERK. Tacrolimus 114-119 mitogen-activated protein kinase 1 Homo sapiens 195-198 14981085-7 2004 Cyclosporin A and FK506, specific inhibitors of the calcium/calmodulin-dependent protein phosphatase (calcineurin), completely eliminated TRESK activation. Tacrolimus 18-23 potassium channel, two pore domain subfamily K, member 18 L homeolog Xenopus laevis 138-143 15607500-9 2004 FK506 (1-1000 ng/ml) did not affect cSMC proliferation alone, although a > or =250-fold excess of FK506 over Rapa completely reversed the inhibitory effect of Rapa, confirming that these two agents share a common intracellular receptor, the FK506-binding protein (FKBP). Tacrolimus 101-106 transcriptional regulating factor 1 Homo sapiens 162-166 15114099-0 2004 Effect of tacrolimus on the expression of macrophage scavenger and nuclear hormone receptors in THP-1-derived human macrophages. Tacrolimus 10-20 GLI family zinc finger 2 Homo sapiens 96-101 15114099-3 2004 This work evaluated the effects of clinically relevant tacrolimus concentrations on the expression of the MSR genes CD36 and CD68, SR-A and SR-BII, lectin-like oxidized low-density lipoprotein receptor-1, the nuclear hormone receptors peroxisome proliferator-activated receptor (PPAR)gamma and liver-X-receptor-alpha, and the cholesterol efflux pump ABCA1 in the in vitro human THP-1 macrophage model. Tacrolimus 55-65 CD68 molecule Homo sapiens 125-129 15114099-12 2004 CONCLUSIONS: Tacrolimus seems to regulate MSRs, nuclear hormone receptors, and ABCA1 in THP-1 macrophages. Tacrolimus 13-23 ATP binding cassette subfamily A member 1 Homo sapiens 79-84 15114099-12 2004 CONCLUSIONS: Tacrolimus seems to regulate MSRs, nuclear hormone receptors, and ABCA1 in THP-1 macrophages. Tacrolimus 13-23 GLI family zinc finger 2 Homo sapiens 88-93 14970329-7 2004 Immunosuppressive drugs known to prevent T cell deletion in vivo, such as cyclosporin A or FK506, blocked Bim up-regulation and rescued T cells from death receptor-independent AICD, whereas rapamycin, which allows the development of stable immunological tolerance, did not exhibit these activities. Tacrolimus 91-96 BCL2 like 11 Homo sapiens 106-109 15036951-4 2004 FK506-induced [Ca(2+)](i) increase was completely blocked by the RyR antagonist ruthenium red and ryanodine, but not the IP(3)R antagonist heparin. Tacrolimus 0-5 ryanodine receptor 1, skeletal muscle Mus musculus 65-68 15060513-0 2004 Tacrolimus therapy according to mucosal MDR1 levels in small-bowel transplant recipients. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 40-44 15060513-7 2004 The mRNA levels of MDR1, but not CYP3A4, correlated well with the concentration/oral dose ratio and the oral dosage of tacrolimus. Tacrolimus 119-129 ATP binding cassette subfamily B member 1 Homo sapiens 19-23 15060513-8 2004 The good progress after transplantation in both cases suggested that monitoring the change in expression of MDR1 mRNA in the graft intestine might be helpful for understanding the pharmacokinetic profile and determining when to change the route of tacrolimus administration in small-bowel transplant recipients. Tacrolimus 248-258 ATP binding cassette subfamily B member 1 Homo sapiens 108-112 15066193-4 2004 In the present study, we demonstrate that calcineurin inhibitors (cyclosporine A, FK506, and FK520), but not non-calcineurin inhibitors (rapamycin and GPI1046) that bind the same intracellular receptor as that for FK506, induce edema and gut coiling disruption and exhibit teratogenesis in the kidney, heart, gut, liver, and somitic tissue during Xenopus development. Tacrolimus 82-87 protein phosphatase 3, catalytic subunit, alpha isozyme L homeolog Xenopus laevis 42-53 15066193-4 2004 In the present study, we demonstrate that calcineurin inhibitors (cyclosporine A, FK506, and FK520), but not non-calcineurin inhibitors (rapamycin and GPI1046) that bind the same intracellular receptor as that for FK506, induce edema and gut coiling disruption and exhibit teratogenesis in the kidney, heart, gut, liver, and somitic tissue during Xenopus development. Tacrolimus 214-219 protein phosphatase 3, catalytic subunit, alpha isozyme L homeolog Xenopus laevis 42-53 15052412-0 2004 FK506 sensitizes mammalian cells to high osmolarity by modulating p38 MAP kinase activation. Tacrolimus 0-5 mitogen-activated protein kinase 14 Homo sapiens 66-69 15003790-0 2004 Expression of osteoclast differentiation factor and osteoclastogenesis inhibitory factor in rat osteoporosis induced by immunosuppressant FK506. Tacrolimus 138-143 TNF superfamily member 11 Rattus norvegicus 14-47 15003790-13 2004 The results of this study support the hypothesis that FK506-mediated osteoporosis occurs by action of the drug on osteoclasts, promoting expression of ODF messenger ribonucleic acid (mRNA) and thus prompting osteoclast differentiation and maturation. Tacrolimus 54-59 TNF superfamily member 11 Rattus norvegicus 151-154 15001192-1 2004 BACKGROUND: Calcineurin inhibitor drugs (cyclosporine and tacrolimus) given to renal transplant recipients to prevent rejection are associated with an increased incidence of hypertension. Tacrolimus 58-68 calcineurin binding protein 1 Homo sapiens 12-33 15052412-5 2004 FK506, but not CsA, regulated p38 activation by osmotic stress, and decreased viability in osmostressed cells. Tacrolimus 0-5 mitogen-activated protein kinase 14 Homo sapiens 30-33 15052412-6 2004 In addition, FK506 treatment strongly increased the phosphorylation of the eukaryotic initiation factor-2alpha (eIF-2alpha) subunit. Tacrolimus 13-18 eukaryotic translation initiation factor 2A Homo sapiens 112-122 15052412-7 2004 eIF-2alpha phosphorylation, p38 inhibition and cell lethality were relieved by addition of excess amino acids to the medium, suggesting that amino acid availability mediated FK506 toxicity. Tacrolimus 174-179 eukaryotic translation initiation factor 2A Homo sapiens 0-10 14970260-10 2004 Our findings show for the first time that the central binding site for the 12 kDa FK506-binding protein of type-3 ryanodine receptor, encompassing the critical valine proline motif, plays a crucial role in the modulation of the Ca2+ release properties of the type-3 ryanodine receptor channel, including the regulation of both global Ca2+ responses and spontaneous Ca2+ sparks. Tacrolimus 82-87 ryanodine receptor 3 Homo sapiens 107-132 15083887-0 2004 Rapid inhibitory effect of tacrolimus on T cell migration by suppressing CD29-related functions. Tacrolimus 27-37 integrin subunit beta 1 Homo sapiens 73-77 15083887-1 2004 OBJECTIVE: To clarify the direct effect of Tacrolimus (FK506) on T cell function in relation to CD29. Tacrolimus 43-53 integrin subunit beta 1 Homo sapiens 96-100 15083887-1 2004 OBJECTIVE: To clarify the direct effect of Tacrolimus (FK506) on T cell function in relation to CD29. Tacrolimus 55-60 integrin subunit beta 1 Homo sapiens 96-100 15083887-6 2004 RESULTS: The ability of H9 cells and PHA-activated T cells incubated with Tacrolimus for 2 hours (hrs) to migrate and to adhere to fibronectin was significantly suppressed. Tacrolimus 74-84 fibronectin 1 Homo sapiens 131-142 15083887-8 2004 Tacrolimus showed slight but significant reduction of cell surface expression of CD29 within 4 hrs, but CD3, CD11a, CD25, CD26 and CD44 were not affected. Tacrolimus 0-10 integrin subunit beta 1 Homo sapiens 81-85 15083887-10 2004 Intracellular tyrosin-phosphorylation induced by CD29 stimulation was also inhibited by, Tacrolimus in H9 cells. Tacrolimus 89-99 integrin subunit beta 1 Homo sapiens 49-53 15083887-11 2004 CONCLUSION: Tacrolimus appeared to have transient early phase inhibitory effects on CD29-related function that may be associated with T cell migration. Tacrolimus 12-22 integrin subunit beta 1 Homo sapiens 84-88 14978156-6 2004 FK506 significantly decreased the renal mRNA expression of TRPV5 (62 +/- 7% relative to controls), calbindin-D(28K) (9 +/- 1%), and TRPM6 (52 +/- 8%), as determined by real-time quantitative PCR analysis. Tacrolimus 0-5 transient receptor potential cation channel, subfamily V, member 5 Rattus norvegicus 59-64 14978156-6 2004 FK506 significantly decreased the renal mRNA expression of TRPV5 (62 +/- 7% relative to controls), calbindin-D(28K) (9 +/- 1%), and TRPM6 (52 +/- 8%), as determined by real-time quantitative PCR analysis. Tacrolimus 0-5 transient receptor potential cation channel, subfamily M, member 6 Rattus norvegicus 132-137 14978156-7 2004 Furthermore, semiquantitative immunohistochemistry showed reduced renal protein abundance of TRPV5 (24 +/- 5%) and calbindin-D(28K) (29 +/- 4%), altogether suggesting that downregulation of these transport proteins is responsible for the FK506-induced Ca(2+) and Mg(2+) wasting. Tacrolimus 238-243 transient receptor potential cation channel, subfamily V, member 5 Rattus norvegicus 93-98 14970260-10 2004 Our findings show for the first time that the central binding site for the 12 kDa FK506-binding protein of type-3 ryanodine receptor, encompassing the critical valine proline motif, plays a crucial role in the modulation of the Ca2+ release properties of the type-3 ryanodine receptor channel, including the regulation of both global Ca2+ responses and spontaneous Ca2+ sparks. Tacrolimus 82-87 ryanodine receptor 3 Homo sapiens 259-284 14767008-0 2004 In vitro treatment of dendritic cells with tacrolimus: impaired T-cell activation and IP-10 expression. Tacrolimus 43-53 C-X-C motif chemokine ligand 10 Homo sapiens 86-91 14767008-5 2004 Although interleukin (IL)-12 production of DC was impaired, they did not promote Th2 development as T cells activated by tacrolimus-treated DC produced less interferon (IFN)-gamma, IL-4 and IL-10. Tacrolimus 121-131 interferon gamma Homo sapiens 157-179 14767008-5 2004 Although interleukin (IL)-12 production of DC was impaired, they did not promote Th2 development as T cells activated by tacrolimus-treated DC produced less interferon (IFN)-gamma, IL-4 and IL-10. Tacrolimus 121-131 interleukin 4 Homo sapiens 181-185 14767008-5 2004 Although interleukin (IL)-12 production of DC was impaired, they did not promote Th2 development as T cells activated by tacrolimus-treated DC produced less interferon (IFN)-gamma, IL-4 and IL-10. Tacrolimus 121-131 interleukin 10 Homo sapiens 190-195 14767008-7 2004 In addition, tacrolimus-treated DC produced less IP-10 (CXCL10), which is known to be involved in allograft rejection. Tacrolimus 13-23 C-X-C motif chemokine ligand 10 Homo sapiens 49-54 14767008-7 2004 In addition, tacrolimus-treated DC produced less IP-10 (CXCL10), which is known to be involved in allograft rejection. Tacrolimus 13-23 C-X-C motif chemokine ligand 10 Homo sapiens 56-62 14767008-9 2004 CONCLUSIONS: Tacrolimus treatment reduces the ability of DC to stimulate T cells and the impaired production of DC-derived IP-10 (CXCL10) and IL-12 might play a role in the immunosuppressive action of tacrolimus. Tacrolimus 13-23 C-X-C motif chemokine ligand 10 Homo sapiens 130-136 14767008-9 2004 CONCLUSIONS: Tacrolimus treatment reduces the ability of DC to stimulate T cells and the impaired production of DC-derived IP-10 (CXCL10) and IL-12 might play a role in the immunosuppressive action of tacrolimus. Tacrolimus 201-211 C-X-C motif chemokine ligand 10 Homo sapiens 123-128 14767008-9 2004 CONCLUSIONS: Tacrolimus treatment reduces the ability of DC to stimulate T cells and the impaired production of DC-derived IP-10 (CXCL10) and IL-12 might play a role in the immunosuppressive action of tacrolimus. Tacrolimus 201-211 C-X-C motif chemokine ligand 10 Homo sapiens 130-136 15167702-4 2004 The objective of this study was to investigate the effect of CYP3A5 and MDR1 (ABCB1) polymorphisms on cyclosporine and tacrolimus dose requirements and trough blood concentrations in stable transplant patients. Tacrolimus 119-129 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-67 15006473-11 2004 FK506 also significantly reduced the extent of hippocampal CA1 cell loss; however, this effect did not correlate with behavior. Tacrolimus 0-5 carbonic anhydrase 1 Rattus norvegicus 59-62 15167702-4 2004 The objective of this study was to investigate the effect of CYP3A5 and MDR1 (ABCB1) polymorphisms on cyclosporine and tacrolimus dose requirements and trough blood concentrations in stable transplant patients. Tacrolimus 119-129 ATP binding cassette subfamily B member 1 Homo sapiens 78-83 15167702-7 2004 Dose-adjusted trough concentrations were three-fold and 1.6-fold higher in CYP3A5*3/*3 patients than in CYP3A5*1/*3 patients for tacrolimus and cyclosporine, respectively. Tacrolimus 129-139 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 75-81 15167702-8 2004 In the case of tacrolimus, the difference was even more striking when considering CYP3A5*1/*1 patients showing dose-adjusted trough concentrations 5.8-fold lower than CYP3A5*3/*3 patients. Tacrolimus 15-25 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 82-88 15167702-8 2004 In the case of tacrolimus, the difference was even more striking when considering CYP3A5*1/*1 patients showing dose-adjusted trough concentrations 5.8-fold lower than CYP3A5*3/*3 patients. Tacrolimus 15-25 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 167-173 15167702-10 2004 Multiple regression analyses showed that CYP3A5*1/*3 polymorphism explained up to 45% of the variability in dose requirement in relation to tacrolimus use. Tacrolimus 140-150 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 41-47 15041369-2 2004 The development by the Edmonton group of a sirolimus-based, steroid-free, low-tacrolimus regimen was a significant breakthrough that allowed the rate of insulin independence after islet transplantation to increase from 13% to 80% at 1 year. Tacrolimus 78-88 insulin Homo sapiens 153-160 15041369-3 2004 Drawbacks include the side effects of sirolimus and the reduction in insulin independence to 50% at 3 years, the latter being attributed to prolonged tacrolimus exposure. Tacrolimus 150-160 insulin Homo sapiens 69-76 15077060-3 2004 FKBP12 is the mediator of immunosuppressive action of FK506. Tacrolimus 54-59 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 0-6 14960307-1 2004 FK506 binding protein 23 from mouse (mFKBP23) is a peptidyl-prolyl cis-trans isomerase (PPIase) from the endoplasmic reticulum (ER), which consists of an N-terminal PPIase domain and a C-terminal domain with Ca(2+) binding sites. Tacrolimus 0-5 FK506 binding protein 7 Mus musculus 37-44 15108766-0 2004 Activation of nuclear factor-kappa B and macrophage invasion in cyclosporin A-and tacrolimus-treated renal transplants. Tacrolimus 82-92 nuclear factor kappa B subunit 1 Homo sapiens 14-36 14746888-3 2004 In the present study, FK506 is shown to potentiate mitogenic effects of the neurotrophic factor, neurturin, on normal adult rat adrenal chromaffin cells in culture. Tacrolimus 22-27 neurotrophin 3 Rattus norvegicus 76-95 14746888-3 2004 In the present study, FK506 is shown to potentiate mitogenic effects of the neurotrophic factor, neurturin, on normal adult rat adrenal chromaffin cells in culture. Tacrolimus 22-27 neurturin Rattus norvegicus 97-106 14769211-0 2004 Effects of tacrolimus on infection of Friend murine leukemia virus to Fv-4 gene heterozygous mice. Tacrolimus 11-21 Friend virus susceptibility 4 Mus musculus 70-74 14769211-9 2004 Compared to the virus-inoculated control, the Friend MuLV-sensitivity of tacrolimus-treated BALB/c mice and the Friend MuLV-resistance of tacrolimus-treated Fv-4 mice were the same as the controls, but only F1 mice became the symptoms and viral proliferation after both treatments. Tacrolimus 138-148 Friend virus susceptibility 4 Mus musculus 157-161 14966413-8 2004 FK506 reduced the expansion of donor CD8+ and, to a lesser extent, CD4+ T cells in the spleen and inhibited donor anti-host T-cell proliferative and cytotoxic responses. Tacrolimus 0-5 CD8a molecule Homo sapiens 37-40 14966413-8 2004 FK506 reduced the expansion of donor CD8+ and, to a lesser extent, CD4+ T cells in the spleen and inhibited donor anti-host T-cell proliferative and cytotoxic responses. Tacrolimus 0-5 CD4 molecule Homo sapiens 67-70 14966413-10 2004 CONCLUSIONS: FK506 inhibits both GvHD and GvL activity when given at clinical doses by inhibiting donor T-cell expansion, donor anti-host T-cell reactivity, and Th1 immune responses. Tacrolimus 13-18 negative elongation factor complex member C/D Homo sapiens 161-164 15108766-1 2004 This retrospective study was designed to compare the efficacy of cyclosporin A (CyA) and tacrolimus (FK506) on chronic rejection (CR) associated with nuclear factor-kappa B (NF-kappaB) activation and macrophage invasion. Tacrolimus 89-99 nuclear factor kappa B subunit 1 Homo sapiens 150-172 15108766-1 2004 This retrospective study was designed to compare the efficacy of cyclosporin A (CyA) and tacrolimus (FK506) on chronic rejection (CR) associated with nuclear factor-kappa B (NF-kappaB) activation and macrophage invasion. Tacrolimus 89-99 nuclear factor kappa B subunit 1 Homo sapiens 174-183 15108766-1 2004 This retrospective study was designed to compare the efficacy of cyclosporin A (CyA) and tacrolimus (FK506) on chronic rejection (CR) associated with nuclear factor-kappa B (NF-kappaB) activation and macrophage invasion. Tacrolimus 101-106 nuclear factor kappa B subunit 1 Homo sapiens 150-172 15108766-1 2004 This retrospective study was designed to compare the efficacy of cyclosporin A (CyA) and tacrolimus (FK506) on chronic rejection (CR) associated with nuclear factor-kappa B (NF-kappaB) activation and macrophage invasion. Tacrolimus 101-106 nuclear factor kappa B subunit 1 Homo sapiens 174-183 14747421-1 2004 Tacrolimus is a potent immunosuppressive agent used in lung transplantation and is a substrate for both P-glycoprotein (P-gp, encoded by the gene MDR1) and cytochrome (CYP) P4503A. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 104-118 14747421-1 2004 Tacrolimus is a potent immunosuppressive agent used in lung transplantation and is a substrate for both P-glycoprotein (P-gp, encoded by the gene MDR1) and cytochrome (CYP) P4503A. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 120-124 14747421-1 2004 Tacrolimus is a potent immunosuppressive agent used in lung transplantation and is a substrate for both P-glycoprotein (P-gp, encoded by the gene MDR1) and cytochrome (CYP) P4503A. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 146-150 14747421-2 2004 A previous study by the authors identified a correlation between the tacrolimus blood level per dose with CYP3A5 and MDR1 gene polymorphisms in pediatric heart transplant patients. Tacrolimus 69-79 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 106-112 15000258-6 2004 RESULTS: FK506 and CsA at concentrations of 1-10 ng/mL inhibited the growth of both HLE and HuH-7 and those immunosuppressors at concentrations over 100 ng/mL exhibited cytotoxicity on these cells. Tacrolimus 9-14 MIR7-3 host gene Homo sapiens 92-97 14747421-2 2004 A previous study by the authors identified a correlation between the tacrolimus blood level per dose with CYP3A5 and MDR1 gene polymorphisms in pediatric heart transplant patients. Tacrolimus 69-79 ATP binding cassette subfamily B member 1 Homo sapiens 117-121 14747421-11 2004 This study confirms the relationship of CYP3A5 polymorphisms to tacrolimus dosing in organ transplant patients. Tacrolimus 64-74 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 40-46 14747421-12 2004 CYP3A5 expressor genotypes required a larger tacrolimus dose to achieve the same blood levels than the CYP3A5 nonexpressors at all time points during the first posttransplant year. Tacrolimus 45-55 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 14747421-14 2004 The authors therefore conclude that tacrolimus dosing in adult lung transplant patients is associated with CYP3A5 gene polymorphisms. Tacrolimus 36-46 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 107-113 15000258-9 2004 The production of ICAM-1 in HLE and HuH-7 was suppressed by FK506 at concentrations of 1-10 ng/mL after treatment for 3-5 days but the effect was not significant in the initial phase at days 1-2 and the last phase at days 5-6. Tacrolimus 60-65 intercellular adhesion molecule 1 Homo sapiens 18-24 15000258-9 2004 The production of ICAM-1 in HLE and HuH-7 was suppressed by FK506 at concentrations of 1-10 ng/mL after treatment for 3-5 days but the effect was not significant in the initial phase at days 1-2 and the last phase at days 5-6. Tacrolimus 60-65 MIR7-3 host gene Homo sapiens 36-41 14709642-2 2004 Previous investigations have demonstrated the occurrence of tacrolimus-resistant production of interleukin-2 (IL-2) in vitro, which may explain in part why rejection episodes are still a frequent problem despite attainment of therapeutic blood concentrations and HLA matching. Tacrolimus 60-70 interleukin 2 Homo sapiens 95-108 14968553-6 2004 On the other hand, unlike FKBP12, FKBP38 inhibits serine/threonine phosphatase calcineurin in an FK506-independent manner. Tacrolimus 97-102 FKBP prolyl isomerase 8 Homo sapiens 34-40 14709642-2 2004 Previous investigations have demonstrated the occurrence of tacrolimus-resistant production of interleukin-2 (IL-2) in vitro, which may explain in part why rejection episodes are still a frequent problem despite attainment of therapeutic blood concentrations and HLA matching. Tacrolimus 60-70 interleukin 2 Homo sapiens 110-114 14709642-4 2004 METHODS: We investigated the immunosuppressive effects of tacrolimus on anti-CD3/anti-CD28 T-cell costimulation in a human whole-blood assay, analyzing T-cell proliferation, activation marker expression (CD25, CD69), IL-2 protein expression, and cytokine mRNA expression in vitro (n = 11 healthy individuals). Tacrolimus 58-68 CD28 molecule Homo sapiens 86-90 14709642-8 2004 Whole-blood samples from 3 of 11 healthy individuals demonstrated marked suppression of IL-2 mRNA expression (>50%) when tacrolimus was administered in vitro. Tacrolimus 124-134 interleukin 2 Homo sapiens 88-92 14709642-10 2004 Two individuals responded conversely, indicating that differences in the in vitro response to tacrolimus and CsA among individuals may be attributable to potential heterogeneity in the involvement of the CD28 pathway. Tacrolimus 94-104 CD28 molecule Homo sapiens 204-208 14709642-11 2004 Kinetic profiles of IL-2 mRNA expression also revealed individually distinct degrees of calcineurin inhibitor sensitivity in patients undergoing tacrolimus or CsA monotherapy before living-donor kidney transplantation. Tacrolimus 145-155 interleukin 2 Homo sapiens 20-24 15244495-7 2004 Factors reported to influence the pharmacokinetics of tacrolimus include the patient group studied, hepatic dysfunction, hepatitis C status, time after transplantation, patient age, donor liver characteristics, recipient race, haematocrit and albumin concentrations, diurnal rhythm, food administration, corticosteroid dosage, diarrhoea and cytochrome P450 (CYP) isoenzyme and P-glycoprotein expression. Tacrolimus 54-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 341-356 15244495-7 2004 Factors reported to influence the pharmacokinetics of tacrolimus include the patient group studied, hepatic dysfunction, hepatitis C status, time after transplantation, patient age, donor liver characteristics, recipient race, haematocrit and albumin concentrations, diurnal rhythm, food administration, corticosteroid dosage, diarrhoea and cytochrome P450 (CYP) isoenzyme and P-glycoprotein expression. Tacrolimus 54-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 358-361 15244495-7 2004 Factors reported to influence the pharmacokinetics of tacrolimus include the patient group studied, hepatic dysfunction, hepatitis C status, time after transplantation, patient age, donor liver characteristics, recipient race, haematocrit and albumin concentrations, diurnal rhythm, food administration, corticosteroid dosage, diarrhoea and cytochrome P450 (CYP) isoenzyme and P-glycoprotein expression. Tacrolimus 54-64 ATP binding cassette subfamily B member 1 Homo sapiens 377-391 15350151-5 2004 Drugs most prominently affected and contraindicated for concomitant use with St John"s wort are metabolised via both CYP3A4 and P-glycoprotein pathways, including HIV protease inhibitors, HIV non-nucleoside reverse transcriptase inhibitors (only CYP3A4), the immunosuppressants ciclosporin and tacrolimus, and the antineoplastic agents irinotecan and imatinib mesylate. Tacrolimus 294-304 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 117-123 15490265-7 2004 The level of serum osteocalcin in the FK506-treated group at week 1 of administration was significantly higher than the control. Tacrolimus 38-43 bone gamma-carboxyglutamate protein Rattus norvegicus 19-30 14766396-1 2004 FKBP12 as an immunophilin that binds to two well-known immunosuppressive macrolides, FK506 and rapamycin, has attracted immense attention and its role in mediating the immunosuppressive functions of these macrolides has been extensively studied. Tacrolimus 85-90 FKBP prolyl isomerase 1A Homo sapiens 0-6 14726632-7 2004 Topical application of FK506 treatment reduced the elevated level of both IL-4 and IFN-gamma in the skin, but did not decrease the expansion of the Th2 population in the LNs. Tacrolimus 23-28 interleukin 4 Mus musculus 74-78 14726632-7 2004 Topical application of FK506 treatment reduced the elevated level of both IL-4 and IFN-gamma in the skin, but did not decrease the expansion of the Th2 population in the LNs. Tacrolimus 23-28 interferon gamma Mus musculus 83-92 15490265-9 2004 Of the various bone resorption factors tested, the level of serum parathyroid hormone (PTH) in the FK506-treated group was significantly higher than the control at week 3 of administration. Tacrolimus 99-104 parathyroid hormone Rattus norvegicus 66-85 14675044-5 2004 FK506-induced cell death was examined in the presence of the hydrogen peroxide scavenger, catalase, or a scavenger of hydroxyl radicals, sodium benzoate. Tacrolimus 0-5 catalase Homo sapiens 90-98 14754647-3 2004 A topical formulation of tacrolimus, a macrolide calcineurin inhibitor, has recently been developed. Tacrolimus 25-35 calcineurin binding protein 1 Homo sapiens 49-70 14675044-14 2004 CONCLUSION: Catalase is uniquely required in cellular protection against FK506 cytotoxicity, which suggests an important role for hydrogen peroxide in the cellular actions of FK506. Tacrolimus 73-78 catalase Homo sapiens 12-20 14675044-6 2004 As a control, FK506-induced cell death was also measured in the presence of superoxide anion inhibitor, 4,5-dihydroxy-1,2-benzene disulfonic acid (Tiron), TEMPO, or overexpressed human manganese superoxide dismutase (MnSOD). Tacrolimus 14-19 superoxide dismutase 2 Homo sapiens 185-215 14675044-14 2004 CONCLUSION: Catalase is uniquely required in cellular protection against FK506 cytotoxicity, which suggests an important role for hydrogen peroxide in the cellular actions of FK506. Tacrolimus 175-180 catalase Homo sapiens 12-20 14675044-6 2004 As a control, FK506-induced cell death was also measured in the presence of superoxide anion inhibitor, 4,5-dihydroxy-1,2-benzene disulfonic acid (Tiron), TEMPO, or overexpressed human manganese superoxide dismutase (MnSOD). Tacrolimus 14-19 superoxide dismutase 2 Homo sapiens 217-222 14675044-9 2004 Abrogation of FK506-mediated ROS by catalase and N-acetylcysteine blunted FK506-induced cell death. Tacrolimus 14-19 catalase Homo sapiens 36-44 14675044-10 2004 Furthermore, overexpression of catalase, sodium benzoate, and deferoxamine inhibited the cytotoxic effect of FK506. Tacrolimus 109-114 catalase Homo sapiens 31-39 14675044-12 2004 In fact, TEMPO or expression of MnSOD enhanced the effect of FK506. Tacrolimus 61-66 superoxide dismutase 2 Homo sapiens 32-37 15782552-7 2004 The newer calcineurin inhibitor tacrolimus is already widely used in the transplantation medicine while its effectiveness in clinical nephrology has to be tested. Tacrolimus 32-42 calcineurin binding protein 1 Homo sapiens 10-31 15047987-4 2004 CsA (at concentrations of 10-50 microM), FK506 (at all used concentrations) and rapamycin (in dose-dependent manner) significantly attenuated IL-1beta release after 24 h exposure to ischemic conditions. Tacrolimus 41-46 interleukin 1 beta Rattus norvegicus 142-150 15047987-6 2004 Moreover, significant decrease in IL-2 secretion at 0.25, 0.5, 1 and 50 microM CsA and FK506 at concentrations of 100 and 1000 nM were observed. Tacrolimus 87-92 interleukin 2 Rattus norvegicus 34-38 15619640-1 2004 Calcineurin-inhibitor induced pain syndrome (CIPS) is a newly described entity with a characteristic feature of sudden onset of severe lower limb pain, and high levels of cyclosporine or tacrolimus may be involved in the pathogenesis. Tacrolimus 187-197 calcineurin binding protein 1 Homo sapiens 0-21 14632538-7 2003 DISCUSSION: Tacrolimus is metabolized in the liver via CYP3A4. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 55-61 15163112-0 2004 Predominant inhibition of Th1 cytokines in New Zealand black/white F1 mice treated with FK506. Tacrolimus 88-93 negative elongation factor complex member C/D, Th1l Mus musculus 26-29 15163112-6 2004 In contrast, expression of mRNA for interleukin (IL)-2, and interferon (IFN)-gamma was suppressed, while that for IL-4 and IL-10 was not suppressed in the group treated with 10 mg/kg of FK506. Tacrolimus 186-191 interferon gamma Mus musculus 60-82 15163112-6 2004 In contrast, expression of mRNA for interleukin (IL)-2, and interferon (IFN)-gamma was suppressed, while that for IL-4 and IL-10 was not suppressed in the group treated with 10 mg/kg of FK506. Tacrolimus 186-191 interleukin 4 Mus musculus 114-118 15163112-6 2004 In contrast, expression of mRNA for interleukin (IL)-2, and interferon (IFN)-gamma was suppressed, while that for IL-4 and IL-10 was not suppressed in the group treated with 10 mg/kg of FK506. Tacrolimus 186-191 interleukin 10 Mus musculus 123-128 15163112-7 2004 The serum levels of IgG-class anti-DNA antibodies, which had been elevated before the treatment, were suppressed--especially in the IgG2a subclass--and the deposition of IgG2a and IgG2b in the glomeruli was reduced in the group treated with 10 mg/kg/day of FK506 compared with the other groups. Tacrolimus 257-262 immunoglobulin heavy variable V1-9 Mus musculus 170-175 15163112-7 2004 The serum levels of IgG-class anti-DNA antibodies, which had been elevated before the treatment, were suppressed--especially in the IgG2a subclass--and the deposition of IgG2a and IgG2b in the glomeruli was reduced in the group treated with 10 mg/kg/day of FK506 compared with the other groups. Tacrolimus 257-262 immunoglobulin heavy constant gamma 2B Mus musculus 180-185 15163112-8 2004 These findings suggest that an improvement in the lupus nephritis of 6-month-old B/WF1 mice induced by FK506 might be associated with a predominant inhibition of Th1 cytokine. Tacrolimus 103-108 TP53 regulated inhibitor of apoptosis 1 Mus musculus 83-86 15163112-8 2004 These findings suggest that an improvement in the lupus nephritis of 6-month-old B/WF1 mice induced by FK506 might be associated with a predominant inhibition of Th1 cytokine. Tacrolimus 103-108 negative elongation factor complex member C/D, Th1l Mus musculus 162-165 14532276-2 2003 Abnormally high phosphorylation levels (hyperphosphorylation) at Ser-2843 in RyR1 and Ser-2809 in RyR2 and dissociation of FK506-binding proteins from the receptors have been implicated as one of the causes of altered calcium homeostasis observed during human heart failure. Tacrolimus 123-128 ryanodine receptor 1 Homo sapiens 77-81 14673984-10 2003 CONCLUSION: Tacrolimus, at dosages of both 2 mg/day and 3 mg/day, is efficacious and safe as monotherapy for patients with active RA, but treatment with the 3-mg dose of tacrolimus resulted in generally better ACR response rates. Tacrolimus 12-22 acrosin Homo sapiens 210-213 14673984-10 2003 CONCLUSION: Tacrolimus, at dosages of both 2 mg/day and 3 mg/day, is efficacious and safe as monotherapy for patients with active RA, but treatment with the 3-mg dose of tacrolimus resulted in generally better ACR response rates. Tacrolimus 170-180 acrosin Homo sapiens 210-213 14991082-1 2003 OBJECTIVE AND DESIGN: To determine the effect of FK506 (tacrolimus) on paw inflammation, TNF-alpha expression in joint, and bone and cartilage destruction in type II collagen-induced arthritis (CIA) model in rats. Tacrolimus 56-66 tumor necrosis factor Rattus norvegicus 89-98 15320493-2 2003 Immunophilin-binding agents like FK506 are known to inactivate neuronal nitric oxide synthase (nNOS) by inhibiting calcineurin and decrease the production of nitric oxide. Tacrolimus 33-38 nitric oxide synthase 1, neuronal Mus musculus 63-93 14991082-10 2003 FK506 (3.2 mg/kg) markedly reduced TNF-alpha expression. Tacrolimus 0-5 tumor necrosis factor Rattus norvegicus 35-44 15320493-2 2003 Immunophilin-binding agents like FK506 are known to inactivate neuronal nitric oxide synthase (nNOS) by inhibiting calcineurin and decrease the production of nitric oxide. Tacrolimus 33-38 nitric oxide synthase 1, neuronal Mus musculus 95-99 15320493-6 2003 Nitric oxide synthase (NOS) inhibitor L-NAME significantly and dose dependently (10-40 mg/kg, ip) potentiated the FK506 (0.5 mg/kg)-induced antinociception. Tacrolimus 114-119 nitric oxide synthase 1, neuronal Mus musculus 0-21 14991082-14 2003 CONCLUSION: These results show that FK506 is effective in suppressing inflammation, TNF-alpha expression in joint, and damage to bone and cartilage in rat CIA, and may be useful in the treatment of rheumatoid arthritis. Tacrolimus 36-41 tumor necrosis factor Rattus norvegicus 84-93 14632799-8 2003 A significant reduction in the CD4/CD8 ratio was found in axillary and inguinal lymph nodes in tacrolimus-treated mice, supporting the presumption that the immunosuppressive effect of the drug was responsible for its effect in promoting tumorigenesis. Tacrolimus 95-105 CD4 antigen Mus musculus 31-34 14678762-4 2003 Inhibition of PP2A with okadiac acid produced enhanced and more lasting Erk1/2 phosphorylation after BK stimulation, whereas FK506, an inhibitor of PP2B and FK-binding protein, inhibited the BK-stimulated Erk1/2 phosphorylation. Tacrolimus 125-130 mitogen-activated protein kinase 3 Homo sapiens 205-211 14870897-1 2003 Azole antifungals inhibit the metabolism of tacrolimus mediated by CYP3A4. Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 67-73 14614095-5 2003 H2O2-dependent LTP in tg-SOD was blocked by the protein phosphatase calcineurin inhibitor FK506, but not by rapamycin, an FK-binding protein 12 (FKBP12) inhibitor or by 1-(5-isoquinolinesulfonyl)-2-methylpiperazine (H7), a serine-kinase inhibitor. Tacrolimus 90-95 superoxide dismutase 1, soluble Mus musculus 25-28 14614095-10 2003 Both young tg-SOD and aged control mice displayed altered protein phosphatase activity, compared with that of young controls; moreover, FK506 inhibited LTP in old tg-SOD as well as in old wt mice treated with H2O2. Tacrolimus 136-141 superoxide dismutase 1, soluble Mus musculus 14-17 14614095-10 2003 Both young tg-SOD and aged control mice displayed altered protein phosphatase activity, compared with that of young controls; moreover, FK506 inhibited LTP in old tg-SOD as well as in old wt mice treated with H2O2. Tacrolimus 136-141 superoxide dismutase 1, soluble Mus musculus 166-169 12958044-4 2003 We thus investigated whether synthesis of IL-8 from primary human aortic smooth muscle cells is influenced by CsA and FK506. Tacrolimus 118-123 C-X-C motif chemokine ligand 8 Homo sapiens 42-46 14559856-0 2003 FK506 potentiates NGF-induced neurite outgrowth via the Ras/Raf/MAP kinase pathway. Tacrolimus 0-5 zinc fingers and homeoboxes 2 Homo sapiens 60-63 14559856-4 2003 Inhibitors of Ras (lovastatin), Raf (GW5074), or MAP kinase (PD98059 and U0126) blocked FK506 activity, as did inhibitors of phospholipase C (U73122) and phosphatidylinositol 3" kinase (LY294002). Tacrolimus 88-93 zinc fingers and homeoboxes 2 Homo sapiens 32-35 14559856-6 2003 These data suggest that FK506 potentiates neurite outgrowth through the Ras/Raf/MAP kinase signaling pathway downstream of phospholipase C and phosphatidylinositol 3" kinase. Tacrolimus 24-29 zinc fingers and homeoboxes 2 Homo sapiens 76-79 14713297-4 2003 In the present study, Abeta peptide-triggered ER calcium release in primary cortical neurons in culture is modulated by Xestospongin C, 2-aminoethoxydiphenyl borate or FK506. Tacrolimus 168-173 amyloid beta precursor protein Homo sapiens 22-27 14623295-2 2003 The crystal structures of CN and its complexes with FKBP12-FK506 and cyclophilin-cyclosporin showed that the two structurally unrelated immunophilins-immunosuppressants bind to a common composite surface made up of the residues from both catalytic subunit and regulatory subunit of CN. Tacrolimus 59-64 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 52-58 14623295-4 2003 However, the binding pattern of FKBP12-FK506 such as hydrogen bonding is significantly different from that of CyPA-CsA. Tacrolimus 39-44 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 32-38 14581219-4 2003 In comparing FKBP12-rapamycin complex and FKBP12-FK506 complex as well as FKBP12-GPI-1046 solution structure with these new complexes, significant volume and surface area effects and obvious contact changes were detected which are expected to cause their different binding energies-showing these two novel ligands will become more effective neuron regeneration drugs than GPI-1046, which is currently undergoing phase II clinical trail as a neurotrophic drug. Tacrolimus 49-54 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 42-48 14581219-4 2003 In comparing FKBP12-rapamycin complex and FKBP12-FK506 complex as well as FKBP12-GPI-1046 solution structure with these new complexes, significant volume and surface area effects and obvious contact changes were detected which are expected to cause their different binding energies-showing these two novel ligands will become more effective neuron regeneration drugs than GPI-1046, which is currently undergoing phase II clinical trail as a neurotrophic drug. Tacrolimus 49-54 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 42-48 14564197-9 2003 Two polymorphisms (C3435T and G2677[A/T]) of the MDR-1 gene have been shown to influence the bioavailability and toxicity of tacrolimus and cyclosporin. Tacrolimus 125-135 ATP binding cassette subfamily B member 1 Homo sapiens 49-54 14570757-2 2003 Livers from groups of rats were perfused in a recirculatory manner following a bolus dose of tacrolimus (100 microg), a substrate for P-glycoprotein (P-gp) and CYP3A, or with felodipine (200 microg), a substrate only for CYP3A. Tacrolimus 93-103 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 134-148 14625173-7 2003 The dose of the insulin of the FK506 group (34.35 U/d, 14.09 U/d) was not significantly different from that in the CsA group (28.15 U/d, 13.05 U/d) at the first month and 1 year after the operation (P>0.05). Tacrolimus 31-36 insulin Homo sapiens 16-23 14570757-2 2003 Livers from groups of rats were perfused in a recirculatory manner following a bolus dose of tacrolimus (100 microg), a substrate for P-glycoprotein (P-gp) and CYP3A, or with felodipine (200 microg), a substrate only for CYP3A. Tacrolimus 93-103 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 150-154 14570757-14 2003 It is concluded that GG918 increased the hepatic exposure of tacrolimus by inhibiting the canalicular P-gp transport, whereas GG918 has no effect on hepatic disposition of felodipine. Tacrolimus 61-71 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 102-106 14555288-10 2003 Treatment with LPS or CM increased iNOS expression in hepatocyte culture, which was inhibited by L-NAME, FK 506 or AG. Tacrolimus 105-111 nitric oxide synthase 2 Rattus norvegicus 35-39 14565935-5 2003 Thus, whereas CsA and FK506 strongly enhanced TCR- and phorbol myristate acetate-induced LAT expression in T cells, rapamycin effectively inhibited activation-induced LAT expression. Tacrolimus 22-27 linker for activation of T cells Homo sapiens 89-92 14565935-9 2003 Given the important role of LAT in initiating T cell activation, our data suggests that the effects of rapamycin, CsA and FK506 on T cell activation involve regulating early T cell signaling. Tacrolimus 122-127 linker for activation of T cells Homo sapiens 28-31 14579277-4 2003 Only very high doses of cyclosporin A and FK506 inhibited macrophage proliferation induced by growth factors, such as M-CSF, granulocyte-macrophage (GM)-CSF or IL-3. Tacrolimus 42-47 colony stimulating factor 1 Homo sapiens 118-123 14579277-4 2003 Only very high doses of cyclosporin A and FK506 inhibited macrophage proliferation induced by growth factors, such as M-CSF, granulocyte-macrophage (GM)-CSF or IL-3. Tacrolimus 42-47 colony stimulating factor 2 Homo sapiens 125-156 14579277-4 2003 Only very high doses of cyclosporin A and FK506 inhibited macrophage proliferation induced by growth factors, such as M-CSF, granulocyte-macrophage (GM)-CSF or IL-3. Tacrolimus 42-47 interleukin 3 Homo sapiens 160-164 14530907-7 2003 Additional inhibitors of phalloidin uptake mediated by SLC21A6 included the immunosuppressive drugs cyclosporin A, FK506, and rapamycin, whereas alpha-amanitin was only a weak inhibitor. Tacrolimus 115-120 solute carrier organic anion transporter family member 1B1 Homo sapiens 55-62 14551375-1 2003 BACKGROUND: Tacrolimus is an immunosuppressive drug that is a substrate of cytochrome P450 3A (CYP3A) enzymes and P-glycoprotein (P-gp). Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 75-93 14578760-2 2003 Tacrolimus is a substrate of cytochrome p450 (CYP), of subfamily CYP3A. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-70 14551375-1 2003 BACKGROUND: Tacrolimus is an immunosuppressive drug that is a substrate of cytochrome P450 3A (CYP3A) enzymes and P-glycoprotein (P-gp). Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 95-100 14551375-1 2003 BACKGROUND: Tacrolimus is an immunosuppressive drug that is a substrate of cytochrome P450 3A (CYP3A) enzymes and P-glycoprotein (P-gp). Tacrolimus 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 114-128 14551375-1 2003 BACKGROUND: Tacrolimus is an immunosuppressive drug that is a substrate of cytochrome P450 3A (CYP3A) enzymes and P-glycoprotein (P-gp). Tacrolimus 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 130-134 14578760-10 2003 CONCLUSIONS: Determination of CYP3A5 genotype is predictive of the dose of tacrolimus in renal transplant recipients and may help to determine the initial daily dose needed by individual patients for adequate immunosuppression without excess nephrotoxicity. Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 30-36 14578760-6 2003 We postulated that CYP3A5 polymorphism is associated with tacrolimus pharmacokinetic variations. Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 19-25 14578760-7 2003 METHODS: CYP3A5 genotype was evaluated in 80 renal transplant recipients and correlated with the daily tacrolimus dose and concentration-to-dose ratio. Tacrolimus 103-113 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 9-15 14550820-7 2003 RESULTS: CsA and FK 506 reduced the presence graft-infiltrating leukocytes (CD4, CD8, CD11a, CD18) in the PVS of intra- and epicardial arteries when compared with control animals. Tacrolimus 17-23 Cd4 molecule Rattus norvegicus 76-79 14555476-4 2003 In contrast, the increased ENA1 expression in a ppz1 mutant is mediated solely by an intact calcineurin/Crz1 signaling pathway, on the basis that (i) this effect maps to a single region that contains the CDRE and (ii) it is blocked by the calcineurin inhibitor FK506, as well as by deletion of the CNB1 or CRZ1 gene. Tacrolimus 261-266 Na(+)/Li(+)-exporting P-type ATPase ENA1 Saccharomyces cerevisiae S288C 27-31 14555476-4 2003 In contrast, the increased ENA1 expression in a ppz1 mutant is mediated solely by an intact calcineurin/Crz1 signaling pathway, on the basis that (i) this effect maps to a single region that contains the CDRE and (ii) it is blocked by the calcineurin inhibitor FK506, as well as by deletion of the CNB1 or CRZ1 gene. Tacrolimus 261-266 salt homeostasis regulator Saccharomyces cerevisiae S288C 48-52 14519526-7 2003 CyA (5 nM and 20 nM), which inhibits both PP2B and PPIase activity, causes permanent memory loss from 60 min post-training when injected into the left hemisphere, an effect also observed following administration of FK506 (20 nM), which also inhibits PP2B and PPIase activity, and [MeVal(4)]CyA (5 nM), which inhibits PPIase activity but not PP2B activity. Tacrolimus 215-220 FK506 binding protein 5 Gallus gallus 51-57 14519526-7 2003 CyA (5 nM and 20 nM), which inhibits both PP2B and PPIase activity, causes permanent memory loss from 60 min post-training when injected into the left hemisphere, an effect also observed following administration of FK506 (20 nM), which also inhibits PP2B and PPIase activity, and [MeVal(4)]CyA (5 nM), which inhibits PPIase activity but not PP2B activity. Tacrolimus 215-220 FK506 binding protein 5 Gallus gallus 259-265 14519526-7 2003 CyA (5 nM and 20 nM), which inhibits both PP2B and PPIase activity, causes permanent memory loss from 60 min post-training when injected into the left hemisphere, an effect also observed following administration of FK506 (20 nM), which also inhibits PP2B and PPIase activity, and [MeVal(4)]CyA (5 nM), which inhibits PPIase activity but not PP2B activity. Tacrolimus 215-220 FK506 binding protein 5 Gallus gallus 259-265 14557767-0 2003 Insulin independence after conversion to tacrolimus and sirolimus-based immunosuppression in islet-kidney recipients. Tacrolimus 41-51 insulin Homo sapiens 0-7 14550820-7 2003 RESULTS: CsA and FK 506 reduced the presence graft-infiltrating leukocytes (CD4, CD8, CD11a, CD18) in the PVS of intra- and epicardial arteries when compared with control animals. Tacrolimus 17-23 CD8a molecule Homo sapiens 81-84 14550820-7 2003 RESULTS: CsA and FK 506 reduced the presence graft-infiltrating leukocytes (CD4, CD8, CD11a, CD18) in the PVS of intra- and epicardial arteries when compared with control animals. Tacrolimus 17-23 integrin subunit alpha L Homo sapiens 86-91 14550820-7 2003 RESULTS: CsA and FK 506 reduced the presence graft-infiltrating leukocytes (CD4, CD8, CD11a, CD18) in the PVS of intra- and epicardial arteries when compared with control animals. Tacrolimus 17-23 integrin subunit beta 2 Rattus norvegicus 93-97 12813040-0 2003 The immunosuppressant FK506 uncovers a positive regulatory cross-talk between the Hog1p and Gcn2p pathways. Tacrolimus 22-27 mitogen-activated protein kinase HOG1 Saccharomyces cerevisiae S288C 82-87 14528517-8 2003 FK506 caused a marked reduction of TNF-a and IL-1beta levels in paw tissue even in short-term (3-day) therapeutic treatment. Tacrolimus 0-5 tumor necrosis factor Rattus norvegicus 35-40 14528517-8 2003 FK506 caused a marked reduction of TNF-a and IL-1beta levels in paw tissue even in short-term (3-day) therapeutic treatment. Tacrolimus 0-5 interleukin 1 beta Rattus norvegicus 45-53 14528517-12 2003 CONCLUSION: FK506 is more effective than MTX in reducing elevated levels of inflammatory cytokines TNF-a, IL-1beta, and IL-6 in established stages of AIA. Tacrolimus 12-17 tumor necrosis factor Rattus norvegicus 99-104 14528517-12 2003 CONCLUSION: FK506 is more effective than MTX in reducing elevated levels of inflammatory cytokines TNF-a, IL-1beta, and IL-6 in established stages of AIA. Tacrolimus 12-17 interleukin 1 beta Rattus norvegicus 106-114 14528517-12 2003 CONCLUSION: FK506 is more effective than MTX in reducing elevated levels of inflammatory cytokines TNF-a, IL-1beta, and IL-6 in established stages of AIA. Tacrolimus 12-17 interleukin 6 Rattus norvegicus 120-124 12796489-2 2003 We originally identified N4WBP5A as an unknown expressed sequence tag (AA770150) represented in a cDNA microarray analysis that was up-regulated upon activation of T cells and inhibited by cell treatment with the calcineurin phosphatase inhibitors, cyclosporine (CsA) and tacrolimus (FK506). Tacrolimus 272-282 Nedd4 family interacting protein 2 Homo sapiens 25-32 12796489-2 2003 We originally identified N4WBP5A as an unknown expressed sequence tag (AA770150) represented in a cDNA microarray analysis that was up-regulated upon activation of T cells and inhibited by cell treatment with the calcineurin phosphatase inhibitors, cyclosporine (CsA) and tacrolimus (FK506). Tacrolimus 284-289 Nedd4 family interacting protein 2 Homo sapiens 25-32 12813040-0 2003 The immunosuppressant FK506 uncovers a positive regulatory cross-talk between the Hog1p and Gcn2p pathways. Tacrolimus 22-27 serine/threonine-protein kinase GCN2 Saccharomyces cerevisiae S288C 92-97 12813040-8 2003 Mutation of the GCN3 and -4 genes partially alleviated FK506 toxicity, suggesting that activation of the GCN pathway by FK506 was also involved in osmotic tolerance. Tacrolimus 55-60 translation initiation factor eIF2B subunit alpha Saccharomyces cerevisiae S288C 16-27 12813040-8 2003 Mutation of the GCN3 and -4 genes partially alleviated FK506 toxicity, suggesting that activation of the GCN pathway by FK506 was also involved in osmotic tolerance. Tacrolimus 120-125 translation initiation factor eIF2B subunit alpha Saccharomyces cerevisiae S288C 16-27 12813040-9 2003 FK506 enhanced osmotic stress-dependent Hog1p kinase phosphorylation that was not accompanied by induction of a Hog1p-dependent reporter. Tacrolimus 0-5 mitogen-activated protein kinase HOG1 Saccharomyces cerevisiae S288C 40-45 12813040-10 2003 Interestingly, deletion of the GCN2 gene suppressed FK506-dependent Hog1p hyperphosphorylation and restored Hog1p-dependent reporter activity. Tacrolimus 52-57 serine/threonine-protein kinase GCN2 Saccharomyces cerevisiae S288C 31-35 12813040-10 2003 Interestingly, deletion of the GCN2 gene suppressed FK506-dependent Hog1p hyperphosphorylation and restored Hog1p-dependent reporter activity. Tacrolimus 52-57 mitogen-activated protein kinase HOG1 Saccharomyces cerevisiae S288C 68-73 12813040-11 2003 Conversely, deletion of the HOG1 gene impaired FK506-dependent activation of Gcn2p kinase and translation of a GCN4-LacZ reporter, highlighting functional cross-talk between the Gcn2p and Hog1p protein kinases. Tacrolimus 47-52 mitogen-activated protein kinase HOG1 Saccharomyces cerevisiae S288C 28-32 12813040-11 2003 Conversely, deletion of the HOG1 gene impaired FK506-dependent activation of Gcn2p kinase and translation of a GCN4-LacZ reporter, highlighting functional cross-talk between the Gcn2p and Hog1p protein kinases. Tacrolimus 47-52 serine/threonine-protein kinase GCN2 Saccharomyces cerevisiae S288C 77-82 12813040-11 2003 Conversely, deletion of the HOG1 gene impaired FK506-dependent activation of Gcn2p kinase and translation of a GCN4-LacZ reporter, highlighting functional cross-talk between the Gcn2p and Hog1p protein kinases. Tacrolimus 47-52 amino acid starvation-responsive transcription factor GCN4 Saccharomyces cerevisiae S288C 111-115 12813040-11 2003 Conversely, deletion of the HOG1 gene impaired FK506-dependent activation of Gcn2p kinase and translation of a GCN4-LacZ reporter, highlighting functional cross-talk between the Gcn2p and Hog1p protein kinases. Tacrolimus 47-52 serine/threonine-protein kinase GCN2 Saccharomyces cerevisiae S288C 178-183 12813040-11 2003 Conversely, deletion of the HOG1 gene impaired FK506-dependent activation of Gcn2p kinase and translation of a GCN4-LacZ reporter, highlighting functional cross-talk between the Gcn2p and Hog1p protein kinases. Tacrolimus 47-52 mitogen-activated protein kinase HOG1 Saccharomyces cerevisiae S288C 188-193 12813040-12 2003 Taken together, these data demonstrate that both FK506-induced amino acid starvation and activation of the GCN pathway contribute to cell sensitivity to osmotic stress and reveal a positive regulatory loop between the Hog1p and Gcn2p pathways. Tacrolimus 49-54 mitogen-activated protein kinase HOG1 Saccharomyces cerevisiae S288C 218-223 12813040-12 2003 Taken together, these data demonstrate that both FK506-induced amino acid starvation and activation of the GCN pathway contribute to cell sensitivity to osmotic stress and reveal a positive regulatory loop between the Hog1p and Gcn2p pathways. Tacrolimus 49-54 serine/threonine-protein kinase GCN2 Saccharomyces cerevisiae S288C 228-233 12813040-13 2003 Given the conserved nature of Gcn2p and Hog1p pathways, this mechanism of FK506 toxicity could be relevant to the non-therapeutic effects of FK506 therapy. Tacrolimus 74-79 serine/threonine-protein kinase GCN2 Saccharomyces cerevisiae S288C 30-35 12813040-13 2003 Given the conserved nature of Gcn2p and Hog1p pathways, this mechanism of FK506 toxicity could be relevant to the non-therapeutic effects of FK506 therapy. Tacrolimus 74-79 mitogen-activated protein kinase HOG1 Saccharomyces cerevisiae S288C 40-45 12813040-13 2003 Given the conserved nature of Gcn2p and Hog1p pathways, this mechanism of FK506 toxicity could be relevant to the non-therapeutic effects of FK506 therapy. Tacrolimus 141-146 serine/threonine-protein kinase GCN2 Saccharomyces cerevisiae S288C 30-35 12813040-13 2003 Given the conserved nature of Gcn2p and Hog1p pathways, this mechanism of FK506 toxicity could be relevant to the non-therapeutic effects of FK506 therapy. Tacrolimus 141-146 mitogen-activated protein kinase HOG1 Saccharomyces cerevisiae S288C 40-45 12966368-0 2003 Genetic polymorphisms of the CYP3A4, CYP3A5, and MDR-1 genes and pharmacokinetics of the calcineurin inhibitors cyclosporine and tacrolimus. Tacrolimus 129-139 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-35 12966368-0 2003 Genetic polymorphisms of the CYP3A4, CYP3A5, and MDR-1 genes and pharmacokinetics of the calcineurin inhibitors cyclosporine and tacrolimus. Tacrolimus 129-139 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 12966368-10 2003 CONCLUSION: As a group, patients with the CYP3A5*3/*3 genotype require less tacrolimus to reach target predose concentrations compared with CYP3A5*1 allele carriers, whereas CYP3A4*1B carriers require more tacrolimus to reach target trough concentrations compared with CYP3A4*1 homozygotes. Tacrolimus 206-216 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 174-180 12966368-0 2003 Genetic polymorphisms of the CYP3A4, CYP3A5, and MDR-1 genes and pharmacokinetics of the calcineurin inhibitors cyclosporine and tacrolimus. Tacrolimus 129-139 ATP binding cassette subfamily B member 1 Homo sapiens 49-54 12966368-3 2003 OBJECTIVE: Our objective was to determine the role of genetic polymorphisms in CYP3A4, CYP3A5, and MDR-1 with respect to interindividual variability in cyclosporine and tacrolimus pharmacokinetics. Tacrolimus 169-179 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 79-85 12966368-3 2003 OBJECTIVE: Our objective was to determine the role of genetic polymorphisms in CYP3A4, CYP3A5, and MDR-1 with respect to interindividual variability in cyclosporine and tacrolimus pharmacokinetics. Tacrolimus 169-179 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 87-93 12966368-3 2003 OBJECTIVE: Our objective was to determine the role of genetic polymorphisms in CYP3A4, CYP3A5, and MDR-1 with respect to interindividual variability in cyclosporine and tacrolimus pharmacokinetics. Tacrolimus 169-179 ATP binding cassette subfamily B member 1 Homo sapiens 99-104 12966368-6 2003 RESULTS: Tacrolimus dose-adjusted trough levels were higher in CYP3A5*3/*3 patients (n = 45) than in *1/*3 plus *1/*1 patients (n = 17), as follows: median and range, 94 (34-398) ng/mL per mg/kg versus 61 (37-163) ng/mL per mg/kg (P <.0001, Mann-Whitney test). Tacrolimus 9-19 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 63-69 12966368-7 2003 CYP3A4*1B allele carriers (n = 10) had lower tacrolimus dose-adjusted trough levels compared with those in patients with the wild-type (*1/*1) genotype (n = 54): median and range, 57 (40-163) ng/mL per mg/kg versus 89 (34-398) ng/mL per mg/kg) (P =.003, Mann-Whitney test). Tacrolimus 45-55 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 12966368-10 2003 CONCLUSION: As a group, patients with the CYP3A5*3/*3 genotype require less tacrolimus to reach target predose concentrations compared with CYP3A5*1 allele carriers, whereas CYP3A4*1B carriers require more tacrolimus to reach target trough concentrations compared with CYP3A4*1 homozygotes. Tacrolimus 76-86 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 42-48 12966368-10 2003 CONCLUSION: As a group, patients with the CYP3A5*3/*3 genotype require less tacrolimus to reach target predose concentrations compared with CYP3A5*1 allele carriers, whereas CYP3A4*1B carriers require more tacrolimus to reach target trough concentrations compared with CYP3A4*1 homozygotes. Tacrolimus 76-86 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 269-275 12966368-10 2003 CONCLUSION: As a group, patients with the CYP3A5*3/*3 genotype require less tacrolimus to reach target predose concentrations compared with CYP3A5*1 allele carriers, whereas CYP3A4*1B carriers require more tacrolimus to reach target trough concentrations compared with CYP3A4*1 homozygotes. Tacrolimus 206-216 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 42-48 12783880-4 2003 In particular, calmodulin antagonists, FK506, and cyclosporin, immunosuppressants that inhibit the calcium-dependent phosphatase calcineurin, suppress ERK1/2 activation by both glucose and GLP-1. Tacrolimus 39-44 mitogen-activated protein kinase 3 Homo sapiens 151-157 12871656-8 2003 FK506 completely inhibited caspase-3 activation and apoptosis caused by TG in a concentration-dependent manner, but not that caused by TM, Eto, and STS. Tacrolimus 0-5 caspase 3 Homo sapiens 27-36 12871656-12 2003 Thus, the suppressive effects of FK506 on cell death are specific to SH-SY5Y cells treated with TG and are caused by the inhibition of calcineurin and subsequent suppression of caspase-3 activation. Tacrolimus 33-38 caspase 3 Homo sapiens 177-186 12836095-1 2003 Tacrolimus (TAC), a widely used nephrotoxic calcineurin inhibitor, is associated with renal vasoconstriction possibly through adenosine receptor activation. Tacrolimus 0-10 calcineurin binding protein 1 Rattus norvegicus 44-65 12836095-1 2003 Tacrolimus (TAC), a widely used nephrotoxic calcineurin inhibitor, is associated with renal vasoconstriction possibly through adenosine receptor activation. Tacrolimus 12-15 calcineurin binding protein 1 Rattus norvegicus 44-65 12967638-9 2003 Cyclosporin A and FK506, the calcineurin inhibitors, significantly inhibited the increases in both c-fos expression and protein synthesis. Tacrolimus 18-23 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 99-104 14623177-0 2003 FK 506 and aminoguanidine suppress iNOS induction in orthotopic corneal allografts and prolong graft survival in mice. Tacrolimus 0-6 nitric oxide synthase 2, inducible Mus musculus 35-39 14623177-8 2003 The treatment with FK 506 resulted in the inhibition of iNOS expression to a high degree in the rejected corneas. Tacrolimus 19-25 nitric oxide synthase 2, inducible Mus musculus 56-60 12783880-4 2003 In particular, calmodulin antagonists, FK506, and cyclosporin, immunosuppressants that inhibit the calcium-dependent phosphatase calcineurin, suppress ERK1/2 activation by both glucose and GLP-1. Tacrolimus 39-44 glucagon like peptide 1 receptor Homo sapiens 189-194 12935893-7 2003 The specificity of the effect of rapamycin was confirmed by the use of the structural analog FK506, which did not have a significant effect on chemotaxis but effectively rescued rapamycin-induced p70S6K inhibition. Tacrolimus 93-98 ribosomal protein S6 kinase B1 Homo sapiens 196-202 12883227-8 2003 Dose reductions of tacrolimus in both study groups were reflected by concordant decreasing pharmacokinetic exposure parameters, which illustrates the common clinical practice of reducing the dose of calcineurin inhibitor as time elapses after transplantation. Tacrolimus 19-29 calcineurin binding protein 1 Homo sapiens 199-220 12923450-0 2003 Tacrolimus enhances transforming growth factor-beta1 expression and promotes tumor progression. Tacrolimus 0-10 hemoglobin, beta adult major chain Mus musculus 46-52 12923450-4 2003 We also determined whether tacrolimus induces TGF-beta(1) expression. Tacrolimus 27-37 transforming growth factor, beta 1 Mus musculus 46-57 12923450-5 2003 Spleens from tacrolimus-treated mice were analyzed for level of expression of TGF-beta(1) mRNA with the use of competitive-quantitative polymerase chain reaction assay, and circulating levels of TGF-beta(1) protein were measured with the use of an enzyme-linked immunosorbent assay. Tacrolimus 13-23 transforming growth factor, beta 1 Mus musculus 78-89 12923450-5 2003 Spleens from tacrolimus-treated mice were analyzed for level of expression of TGF-beta(1) mRNA with the use of competitive-quantitative polymerase chain reaction assay, and circulating levels of TGF-beta(1) protein were measured with the use of an enzyme-linked immunosorbent assay. Tacrolimus 13-23 transforming growth factor, beta 1 Mus musculus 195-206 12923450-7 2003 Treatment with 4 mg/kg but not 2 mg/kg of tacrolimus resulted in a significant increase in the levels of expression of TGF-beta(1) mRNA and circulating levels of TGF-beta(1) protein. Tacrolimus 42-52 transforming growth factor, beta 1 Mus musculus 119-130 12923450-7 2003 Treatment with 4 mg/kg but not 2 mg/kg of tacrolimus resulted in a significant increase in the levels of expression of TGF-beta(1) mRNA and circulating levels of TGF-beta(1) protein. Tacrolimus 42-52 transforming growth factor, beta 1 Mus musculus 162-173 12923450-8 2003 CONCLUSIONS: Tacrolimus has a dose-dependent effect on tumor progression and TGF-beta(1) expression, and tacrolimus-induced TGF-beta(1) overexpression may be a pathogenetic mechanism in tumor progression. Tacrolimus 13-23 transforming growth factor, beta 1 Mus musculus 77-88 12923450-8 2003 CONCLUSIONS: Tacrolimus has a dose-dependent effect on tumor progression and TGF-beta(1) expression, and tacrolimus-induced TGF-beta(1) overexpression may be a pathogenetic mechanism in tumor progression. Tacrolimus 105-115 transforming growth factor, beta 1 Mus musculus 124-135 12884468-3 2003 These two cases demonstrate a clear-cut therapeutic response of chronic, topical corticosteroid-resistant annular erythema to topical tacrolimus ointment 0.1% BID. Tacrolimus 134-144 BH3 interacting domain death agonist Homo sapiens 159-162 12740386-4 2003 The immunosuppressant FK506 failed to inhibit p70S6K activation, but was able to rescue the rapamycin-induced downshift, pointing to a role for the mammalian target of rapamycin (mTOR) kinase. Tacrolimus 22-27 mechanistic target of rapamycin kinase Homo sapiens 148-177 12962785-1 2003 Employing tacrolimus (Tac) for routine immunosuppression in renal transplantation (RT), produced an incidence of new-onset, insulin-treated, diabetes mellitus (newDM) as high as 20%. Tacrolimus 10-20 insulin Homo sapiens 124-131 12740386-4 2003 The immunosuppressant FK506 failed to inhibit p70S6K activation, but was able to rescue the rapamycin-induced downshift, pointing to a role for the mammalian target of rapamycin (mTOR) kinase. Tacrolimus 22-27 mechanistic target of rapamycin kinase Homo sapiens 179-183 12818377-5 2003 The Pgp-mediated tacrolimus transport was the highest on day 1 and the lowest on day 11. Tacrolimus 17-27 ATP binding cassette subfamily B member 1 Homo sapiens 4-7 12892836-9 2003 In hypoxia-reoxygenation-subjected hepatocytes, tacrolimus reduced JNK1/SAPK1 and caspase 3 activation. Tacrolimus 48-58 caspase 3 Rattus norvegicus 82-91 12819249-15 2003 In conclusion, conversion from cyclosporine to tacrolimus in stable renal transplant patients has a beneficial effect on renal function, BP, serum concentration and atherogenic properties of serum lipids, and fibrinogen. Tacrolimus 47-57 fibrinogen beta chain Homo sapiens 209-219 12839866-20 2003 FK506 reduced IL-1beta and COX-2 mRNA expression and PGE(2) level in arthritic paws. Tacrolimus 0-5 interleukin 1 beta Rattus norvegicus 14-22 12839866-20 2003 FK506 reduced IL-1beta and COX-2 mRNA expression and PGE(2) level in arthritic paws. Tacrolimus 0-5 cytochrome c oxidase II, mitochondrial Rattus norvegicus 27-32 14523907-2 2003 The CaN inhibitor drugs, such as cyclosporin A (CyA), carried by cyclophillin, and tacrolimus, carried by FK-binding protein-12 (FKBP-12), inhibit the binding with the regulatory subunit CaNB. Tacrolimus 83-93 FKBP prolyl isomerase 1A Homo sapiens 106-127 14523907-2 2003 The CaN inhibitor drugs, such as cyclosporin A (CyA), carried by cyclophillin, and tacrolimus, carried by FK-binding protein-12 (FKBP-12), inhibit the binding with the regulatory subunit CaNB. Tacrolimus 83-93 FKBP prolyl isomerase 1A Homo sapiens 129-136 12819250-0 2003 Association of the multidrug resistance-1 gene single-nucleotide polymorphisms with the tacrolimus dose requirements in renal transplant recipients. Tacrolimus 88-98 ATP binding cassette subfamily B member 1 Homo sapiens 19-41 12819250-1 2003 The immunosuppressive drug tacrolimus, whose pharmacokinetic characteristics display large interindividual variations, is a substrate for P-glycoprotein (P-gp), the product of the multidrug resistance-1 (MDR1) gene. Tacrolimus 27-37 ATP binding cassette subfamily B member 1 Homo sapiens 138-152 12819250-1 2003 The immunosuppressive drug tacrolimus, whose pharmacokinetic characteristics display large interindividual variations, is a substrate for P-glycoprotein (P-gp), the product of the multidrug resistance-1 (MDR1) gene. Tacrolimus 27-37 ATP binding cassette subfamily B member 1 Homo sapiens 154-158 12819250-1 2003 The immunosuppressive drug tacrolimus, whose pharmacokinetic characteristics display large interindividual variations, is a substrate for P-glycoprotein (P-gp), the product of the multidrug resistance-1 (MDR1) gene. Tacrolimus 27-37 ATP binding cassette subfamily B member 1 Homo sapiens 180-202 12819250-1 2003 The immunosuppressive drug tacrolimus, whose pharmacokinetic characteristics display large interindividual variations, is a substrate for P-glycoprotein (P-gp), the product of the multidrug resistance-1 (MDR1) gene. Tacrolimus 27-37 ATP binding cassette subfamily B member 1 Homo sapiens 204-208 12819250-3 2003 Because P-gp is known to control tacrolimus intestinal absorption, it was postulated that these polymorphisms are associated with tacrolimus pharmacokinetic variations in renal transplant recipients. Tacrolimus 33-43 ATP binding cassette subfamily B member 1 Homo sapiens 8-12 12819250-3 2003 Because P-gp is known to control tacrolimus intestinal absorption, it was postulated that these polymorphisms are associated with tacrolimus pharmacokinetic variations in renal transplant recipients. Tacrolimus 130-140 ATP binding cassette subfamily B member 1 Homo sapiens 8-12 12819250-10 2003 Genotype monitoring of the MDR1 gene reliably predicts the optimal dose of tacrolimus in renal transplant recipients and may predict the initial daily dose needed by individual patients to obtain adequate immunosuppression. Tacrolimus 75-85 ATP binding cassette subfamily B member 1 Homo sapiens 27-31 12835079-13 2003 Cyclosporine and Tacrolimus, collectively known as calcineurin inhibitors, seems to act on the IL-2 by inhibiting its production thus leading to a decrease in the proliferation of the activated lymphocyte. Tacrolimus 17-27 interleukin 2 Homo sapiens 95-99 12676880-10 2003 From these findings, it is concluded that the site-dependent differences in P-gp and/or P450 activity could be the prime cause of large intra- and interindividual variability in the oral absorption of tacrolimus. Tacrolimus 201-211 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 76-80 12826242-4 2003 Pretreatment with dexamethasone or tacrolimus significantly suppressed long-duration scratching in NC/Nga mice but did not suppress short-duration scratching induced by ovalbumin active cutaneous anaphylaxis in BALB/c mice and in ICR mice subcutaneously injected with histamine. Tacrolimus 35-45 reticulon 4 Mus musculus 102-105 12817066-8 2003 Even fewer data are available on the success rate of the use of tacrolimus in resistant forms of FSGS in adults. Tacrolimus 64-74 actinin alpha 4 Homo sapiens 97-101 12837242-3 2003 The FK506 binding protein (FKBP12.6) stabilizes RyR2, preventing aberrant activation of the channel during the resting phase of the cardiac cycle. Tacrolimus 4-9 FKBP prolyl isomerase 1B Homo sapiens 27-35 12826147-2 2003 Ketoconazole increases tacrolimus bioavailability by inhibiting cytochrome P450 3A4 and glycoprotein-p. Tacrolimus 23-33 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 64-83 12837242-3 2003 The FK506 binding protein (FKBP12.6) stabilizes RyR2, preventing aberrant activation of the channel during the resting phase of the cardiac cycle. Tacrolimus 4-9 ryanodine receptor 2 Homo sapiens 48-52 12767900-1 2003 We previously reported synergistic induction of apoptosis by IFN-gamma plus either cyclosporin A (CsA) or tacrolimus (FK506) in gastric carcinoma cells. Tacrolimus 118-123 interferon gamma Homo sapiens 61-70 12767900-8 2003 Activation of NF-kappa B occurred in response to IFN-gamma, and which was markedly inhibited by either CsA or FK506. Tacrolimus 110-115 nuclear factor kappa B subunit 1 Homo sapiens 14-24 12767900-8 2003 Activation of NF-kappa B occurred in response to IFN-gamma, and which was markedly inhibited by either CsA or FK506. Tacrolimus 110-115 interferon gamma Homo sapiens 49-58 12808614-7 2003 There was a higher incidence of insulin resistance in the tacrolimus group (post-transplant diabetes mellitus, glucose tolerance testing) but this was not statistically significant. Tacrolimus 58-68 insulin Homo sapiens 32-39 12746298-1 2003 FKBP51 and FKBP52 are large molecular weight FK506-binding immunophilins that have diverse biochemical functions. Tacrolimus 45-50 FKBP prolyl isomerase 4 Homo sapiens 11-17 12773514-5 2003 Downstream signaling routes involved mitogen-activated protein kinase (MAPK) kinase (MEK)1/extracellular signal-regulated kinase (ERK), p38 MAPK, and calcineurin, as MICA expression was prevented by U0126, SB202190, cyclosporin A, and FK506. Tacrolimus 235-240 MHC class I polypeptide-related sequence A Homo sapiens 166-170 12761338-0 2003 Inhibition of human insulin gene transcription by the immunosuppressive drugs cyclosporin A and tacrolimus in primary, mature islets of transgenic mice. Tacrolimus 96-106 insulin Homo sapiens 20-27 12761338-4 2003 Previous studies have shown that tacrolimus can inhibit insulin gene transcription at high concentrations in tumor cell lines. Tacrolimus 33-43 insulin Homo sapiens 56-63 12761338-8 2003 After stimulation with glucose, human insulin promoter-mediated gene expression was inhibited in normal, mature islet cells by both tacrolimus and cyclosporin A to a large extent (approximately 70%) and with high potency at concentrations that are known to inhibit calcineurin phosphatase activity (IC50 values of 1 and 35 nM, respectively). Tacrolimus 132-142 insulin Homo sapiens 38-45 12761338-10 2003 The high potency of cyclosporin A and tacrolimus in normal islets suggests that inhibition of insulin gene transcription by cyclosporin A and tacrolimus is clinically important and is one mechanism of the diabetogenic effect of these immunosuppressive drugs. Tacrolimus 38-48 insulin Homo sapiens 94-101 12761338-10 2003 The high potency of cyclosporin A and tacrolimus in normal islets suggests that inhibition of insulin gene transcription by cyclosporin A and tacrolimus is clinically important and is one mechanism of the diabetogenic effect of these immunosuppressive drugs. Tacrolimus 142-152 insulin Homo sapiens 94-101 14703094-8 2003 FK506 prolonged allograft survival (6.5 vs 31 days), and suppressed activation of myocardial MAPKs (ERK: 66% in LV and 67% in SEP, p38MAPK: 62% in LV and 72% in SEP), AP-1 (24% in LV and 18% in SEP), and NF-kappaB (41% in LV and 20% in SEP) (the mean value of activities in the control group was represented as 100%). Tacrolimus 0-5 Eph receptor B1 Rattus norvegicus 100-103 14703094-8 2003 FK506 prolonged allograft survival (6.5 vs 31 days), and suppressed activation of myocardial MAPKs (ERK: 66% in LV and 67% in SEP, p38MAPK: 62% in LV and 72% in SEP), AP-1 (24% in LV and 18% in SEP), and NF-kappaB (41% in LV and 20% in SEP) (the mean value of activities in the control group was represented as 100%). Tacrolimus 0-5 Jun proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 167-171 12769796-9 2003 FK506 has been shown to interfere with the apoptotic pathway of neuronal cells, including inhibiting JNK activity, cytochrome c release, caspase 3 activation, and CD95 ligand expression. Tacrolimus 0-5 cytochrome c, somatic Homo sapiens 115-127 12769796-9 2003 FK506 has been shown to interfere with the apoptotic pathway of neuronal cells, including inhibiting JNK activity, cytochrome c release, caspase 3 activation, and CD95 ligand expression. Tacrolimus 0-5 caspase 3 Homo sapiens 137-146 12769796-9 2003 FK506 has been shown to interfere with the apoptotic pathway of neuronal cells, including inhibiting JNK activity, cytochrome c release, caspase 3 activation, and CD95 ligand expression. Tacrolimus 0-5 Fas cell surface death receptor Homo sapiens 163-167 12780674-0 2003 The use of an anti-CD25 monoclonal antibody and mycophenolate mofetil enables the use of a low-dose tacrolimus and early withdrawal of steroids in renal transplant recipients. Tacrolimus 100-110 interleukin 2 receptor subunit alpha Homo sapiens 19-23 12780674-2 2003 We examined the hypothesis that using anti-CD25 monoclonal antibody induction and mycophenolate mofetil (MMF) would allow the lowering of target pre-dose blood concentrations of tacrolimus immediately after transplantation and subsequently stopping steroids at 5 months. Tacrolimus 178-188 interleukin 2 receptor subunit alpha Homo sapiens 43-47 12780674-11 2003 CONCLUSION: A regimen consisting of anti-CD25 monoclonal antibody induction and MMF allows the safe and efficient use of low-target pre-dose trough concentrations of tacrolimus and enables the early discontinuation of steroids. Tacrolimus 166-176 interleukin 2 receptor subunit alpha Homo sapiens 41-45 12942158-3 2003 The inhibition of calcineurin activation by cyclosporin A and FK-506 blocks T-cell receptor-mediated production of interleukin-2 (IL-2), a growth factor critical for T-cell proliferation. Tacrolimus 62-68 interleukin 2 Homo sapiens 115-128 12942158-3 2003 The inhibition of calcineurin activation by cyclosporin A and FK-506 blocks T-cell receptor-mediated production of interleukin-2 (IL-2), a growth factor critical for T-cell proliferation. Tacrolimus 62-68 interleukin 2 Homo sapiens 130-134 12782134-1 2003 FKBP52 is a widely expressed FK506-binding immunophilin that possesses peptidylprolyl isomerase activity and a tetratricopeptide repeat involved in protein-protein interaction. Tacrolimus 29-34 FKBP prolyl isomerase 4 Homo sapiens 0-6 12787363-6 2003 Phosphorylation of Mpk1 is induced in response to perturbations of cell wall biosynthesis by the antifungal drugs nikkomycin Z (a chitin synthase inhibitor), caspofungin (a beta-1,3-glucan synthase inhibitor), or FK506 (a calcineurin inhibitor), and mutants lacking Mpk1 display enhanced sensitivity to nikkomycin Z and caspofungin. Tacrolimus 213-218 mitogen-activated serine/threonine-protein kinase SLT2 Saccharomyces cerevisiae S288C 19-23 12826230-0 2003 Insulin-like growth factor-I enhances choleretic action of FK506 in rats. Tacrolimus 59-64 insulin-like growth factor 1 Rattus norvegicus 0-28 12826230-4 2003 Recently, we demonstrated in rats that exogenous treatment with insulin-like growth factor I (IGF-I) results in an increase in bile flow and also that FK506 has the potential to increase hepatic production of IGF-I. Tacrolimus 151-156 insulin-like growth factor 1 Rattus norvegicus 64-92 12826230-4 2003 Recently, we demonstrated in rats that exogenous treatment with insulin-like growth factor I (IGF-I) results in an increase in bile flow and also that FK506 has the potential to increase hepatic production of IGF-I. Tacrolimus 151-156 insulin-like growth factor 1 Rattus norvegicus 94-99 12826230-4 2003 Recently, we demonstrated in rats that exogenous treatment with insulin-like growth factor I (IGF-I) results in an increase in bile flow and also that FK506 has the potential to increase hepatic production of IGF-I. Tacrolimus 151-156 insulin-like growth factor 1 Rattus norvegicus 209-214 12826230-5 2003 However, circulating levels of IGF-I in FK506-treated rats were only 30% higher than in nontreated rats. Tacrolimus 40-45 insulin-like growth factor 1 Rattus norvegicus 31-36 12826230-8 2003 Overall, this study demonstrated that IGF-I treatment enhanced the choleretic action of FK506, providing potential clinical utility for combination therapy using these two drugs, in treatment after liver transplantation. Tacrolimus 88-93 insulin-like growth factor 1 Rattus norvegicus 38-43 12826211-0 2003 Monocyte chemoattractant protein-1 levels in bronchoalveolar lavage fluid of lung-transplanted patients treated with tacrolimus as rescue treatment for refractory acute rejection. Tacrolimus 117-127 C-C motif chemokine ligand 2 Homo sapiens 0-34 12826211-5 2003 In contrast, monocyte chemoattractant protein-1 (MCP-1) levels showed a significant and sustained decrease in BAL-F during tacrolimus therapy. Tacrolimus 123-133 C-C motif chemokine ligand 2 Homo sapiens 13-47 12826211-5 2003 In contrast, monocyte chemoattractant protein-1 (MCP-1) levels showed a significant and sustained decrease in BAL-F during tacrolimus therapy. Tacrolimus 123-133 C-C motif chemokine ligand 2 Homo sapiens 49-54 12826211-7 2003 CONCLUSIONS: These findings suggest that the clinical and functional stabilization of patients observed after conversion to a tacrolimus based regimen, may be due, at least in part, to the induced down-regulation of MCP-1 production. Tacrolimus 126-136 C-C motif chemokine ligand 2 Homo sapiens 216-221 12914753-9 2003 Most interestingly, the calcineurin inhibitor, FK506, consistently inhibited CD3/CD28-induced CXCR6 down-regulation. Tacrolimus 47-52 calcineurin binding protein 1 Homo sapiens 24-45 12732617-3 2003 Phosphorylated IkappaBbeta is a substrate for Cn phosphatase, which was inhibited by FK506 and RII peptide. Tacrolimus 85-90 NFKB inhibitor beta Homo sapiens 15-26 12732617-4 2003 Chemical cross-linking and coimmunoprecipitation show that NFkappaB/Rel factor-bound IkappaBbeta forms a ternary complex with Cn under in vitro and in vivo conditions that was sensitive to FK506. Tacrolimus 189-194 nuclear factor kappa B subunit 1 Homo sapiens 59-67 12732617-4 2003 Chemical cross-linking and coimmunoprecipitation show that NFkappaB/Rel factor-bound IkappaBbeta forms a ternary complex with Cn under in vitro and in vivo conditions that was sensitive to FK506. Tacrolimus 189-194 NFKB inhibitor beta Homo sapiens 85-96 12777858-10 2003 The type of baseline calcineurin inhibitor (tacrolimus) and the age at OLT (beyond 6 years) were significantly associated with a better graft survival. Tacrolimus 44-54 calcineurin binding protein 1 Homo sapiens 21-42 12914753-9 2003 Most interestingly, the calcineurin inhibitor, FK506, consistently inhibited CD3/CD28-induced CXCR6 down-regulation. Tacrolimus 47-52 CD28 molecule Homo sapiens 81-85 12914753-9 2003 Most interestingly, the calcineurin inhibitor, FK506, consistently inhibited CD3/CD28-induced CXCR6 down-regulation. Tacrolimus 47-52 C-X-C motif chemokine receptor 6 Homo sapiens 94-99 12914753-10 2003 FK506 also blocked the decrease of CXCR6 expression caused by ionomycin, whereas staurosporine or PP2 had no effect on this decrease. Tacrolimus 0-5 C-X-C motif chemokine receptor 6 Homo sapiens 35-40 12698107-7 2003 In cyclosporine- or FK506-treated mice, T-cell proliferation was suppressed in the CD4 subset but not in the CD8 subset. Tacrolimus 20-25 CD4 antigen Mus musculus 83-86 12709020-8 2003 The enhancement of IL-4 production by BPA or NP was significantly reduced by nitrendipine, a blocker of Ca2+ influx, and by FK506, a calcineurin inhibitor. Tacrolimus 124-129 interleukin 4 Mus musculus 19-23 12709020-9 2003 FK506 inhibited the NF-AT-DNA binding activity and IL-4 gene promoter activity enhanced by BPA or NP. Tacrolimus 0-5 interleukin 4 Mus musculus 51-55 12742481-1 2003 Calcineurin inhibitor drugs (CNI), primarily cyclosporine then tacrolimus, have been the centerpieces of maintenance immunosuppression for kidney transplantation since their introduction in the 1980s. Tacrolimus 63-73 calcineurin binding protein 1 Homo sapiens 0-21 12706270-2 2003 Both FK506 (an immunosuppressive IPL) and GPI1046 (a non-immunosuppressive IPL) significantly increased glial cell line-derived neurotrophic factor (GDNF) content in the substantia nigra. Tacrolimus 5-10 glial cell line derived neurotrophic factor Mus musculus 104-147 12706270-2 2003 Both FK506 (an immunosuppressive IPL) and GPI1046 (a non-immunosuppressive IPL) significantly increased glial cell line-derived neurotrophic factor (GDNF) content in the substantia nigra. Tacrolimus 5-10 glial cell line derived neurotrophic factor Mus musculus 149-153 12706270-3 2003 In addition, FK506 increased striatal brain-derived neurotrophic factor (BDNF) content significantly. Tacrolimus 13-18 brain derived neurotrophic factor Mus musculus 38-71 12706270-3 2003 In addition, FK506 increased striatal brain-derived neurotrophic factor (BDNF) content significantly. Tacrolimus 13-18 brain derived neurotrophic factor Mus musculus 73-77 12570836-6 2003 Disappointingly, CSA and the other molecules as FK506, sharing the capacity to inhibit calcineurin, should be administered for all patient life, as tolerance to alloantigens is not achieved by these molecules. Tacrolimus 48-53 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 17-20 12706476-1 2003 Immunophilin ligands, cyclosporine A and FK506 (tacrolimus), besides their immunosuppressive action, have several effects on different neural functions, such as modulation of the release of many neurotransmitters, the reduction of nitric oxide (NO) production by the inhibition of dephosphorylation of neuronal nitric oxide synthase (nNOS) and the alteration of the expression of certain genes. Tacrolimus 41-46 nitric oxide synthase, brain Cavia porcellus 302-332 12706476-1 2003 Immunophilin ligands, cyclosporine A and FK506 (tacrolimus), besides their immunosuppressive action, have several effects on different neural functions, such as modulation of the release of many neurotransmitters, the reduction of nitric oxide (NO) production by the inhibition of dephosphorylation of neuronal nitric oxide synthase (nNOS) and the alteration of the expression of certain genes. Tacrolimus 41-46 nitric oxide synthase, brain Cavia porcellus 334-338 12706476-1 2003 Immunophilin ligands, cyclosporine A and FK506 (tacrolimus), besides their immunosuppressive action, have several effects on different neural functions, such as modulation of the release of many neurotransmitters, the reduction of nitric oxide (NO) production by the inhibition of dephosphorylation of neuronal nitric oxide synthase (nNOS) and the alteration of the expression of certain genes. Tacrolimus 48-58 nitric oxide synthase, brain Cavia porcellus 302-332 12706476-1 2003 Immunophilin ligands, cyclosporine A and FK506 (tacrolimus), besides their immunosuppressive action, have several effects on different neural functions, such as modulation of the release of many neurotransmitters, the reduction of nitric oxide (NO) production by the inhibition of dephosphorylation of neuronal nitric oxide synthase (nNOS) and the alteration of the expression of certain genes. Tacrolimus 48-58 nitric oxide synthase, brain Cavia porcellus 334-338 12700828-8 2003 In contrast, the repeated administration of FK506 combining iv plus ip administration reduced CA1 cell death on all stereotaxic planes both 7 and 30 days post-ischemia (experiment 2; P<=0.01). Tacrolimus 44-49 carbonic anhydrase 1 Rattus norvegicus 94-97 12694072-0 2003 Tacrolimus dosing in pediatric heart transplant patients is related to CYP3A5 and MDR1 gene polymorphisms. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 71-77 12694072-0 2003 Tacrolimus dosing in pediatric heart transplant patients is related to CYP3A5 and MDR1 gene polymorphisms. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 82-86 12694072-1 2003 Tacrolimus is a substrate for P-glycoprotein (P-gp) and cytochrome (CYP) P4503A. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 30-44 12694072-1 2003 Tacrolimus is a substrate for P-glycoprotein (P-gp) and cytochrome (CYP) P4503A. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 46-50 12694072-1 2003 Tacrolimus is a substrate for P-glycoprotein (P-gp) and cytochrome (CYP) P4503A. Tacrolimus 0-10 peptidylprolyl isomerase G Homo sapiens 68-71 12694072-2 2003 P-gp is encoded by the multiple drug resistance gene MDR1 and CYP3A is the major enzyme responsible for tacrolimus metabolism. Tacrolimus 104-114 ATP binding cassette subfamily B member 1 Homo sapiens 0-4 12694072-2 2003 P-gp is encoded by the multiple drug resistance gene MDR1 and CYP3A is the major enzyme responsible for tacrolimus metabolism. Tacrolimus 104-114 ATP binding cassette subfamily B member 1 Homo sapiens 53-57 12694072-2 2003 P-gp is encoded by the multiple drug resistance gene MDR1 and CYP3A is the major enzyme responsible for tacrolimus metabolism. Tacrolimus 104-114 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-67 12694072-4 2003 The objective of this study was to evaluate whether the MDR1 exon21 and exon26 polymorphisms and the CYP3A5 polymorphism are associated with tacrolimus disposition in pediatric heart transplant patients. Tacrolimus 141-151 ATP binding cassette subfamily B member 1 Homo sapiens 56-60 12694072-4 2003 The objective of this study was to evaluate whether the MDR1 exon21 and exon26 polymorphisms and the CYP3A5 polymorphism are associated with tacrolimus disposition in pediatric heart transplant patients. Tacrolimus 141-151 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 101-107 12694072-5 2003 At 3, 6 and 12 months post transplantation, a significant difference in tacrolimus blood level per dose/kg/day was found between the CYP3A5 *1/*3 (CYP3A5 expressor) vs. *3/*3 (nonexpressor) genotypes with the *1/*3 patients requiring a larger tacrolimus dose to maintain the same blood concentration. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 133-139 12694072-5 2003 At 3, 6 and 12 months post transplantation, a significant difference in tacrolimus blood level per dose/kg/day was found between the CYP3A5 *1/*3 (CYP3A5 expressor) vs. *3/*3 (nonexpressor) genotypes with the *1/*3 patients requiring a larger tacrolimus dose to maintain the same blood concentration. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 147-153 12694072-7 2003 We conclude that specific genotypes of MDR1 and CYP3A5 in pediatric heart transplant patients require larger tacrolimus doses to maintain their tacrolimus blood concentration, and that this information could be used prospectively to manage patient"s immunosuppressive therapy. Tacrolimus 109-119 ATP binding cassette subfamily B member 1 Homo sapiens 39-43 12694072-7 2003 We conclude that specific genotypes of MDR1 and CYP3A5 in pediatric heart transplant patients require larger tacrolimus doses to maintain their tacrolimus blood concentration, and that this information could be used prospectively to manage patient"s immunosuppressive therapy. Tacrolimus 109-119 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 48-54 12694072-7 2003 We conclude that specific genotypes of MDR1 and CYP3A5 in pediatric heart transplant patients require larger tacrolimus doses to maintain their tacrolimus blood concentration, and that this information could be used prospectively to manage patient"s immunosuppressive therapy. Tacrolimus 144-154 ATP binding cassette subfamily B member 1 Homo sapiens 39-43 12694072-7 2003 We conclude that specific genotypes of MDR1 and CYP3A5 in pediatric heart transplant patients require larger tacrolimus doses to maintain their tacrolimus blood concentration, and that this information could be used prospectively to manage patient"s immunosuppressive therapy. Tacrolimus 144-154 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 48-54 12646197-0 2003 The immunosuppressive agent tacrolimus induces p21WAF/CIP1WAF1/CIP1 via TGF-beta secretion. Tacrolimus 28-38 cyclin dependent kinase inhibitor 1A Homo sapiens 54-58 12646197-0 2003 The immunosuppressive agent tacrolimus induces p21WAF/CIP1WAF1/CIP1 via TGF-beta secretion. Tacrolimus 28-38 transforming growth factor beta 1 Homo sapiens 72-80 12646197-7 2003 Using anti-TGF-beta antibody, we studied if induction of p21WAF/CIP1 by tacrolimus is dependent on TGF-beta. Tacrolimus 72-82 cyclin dependent kinase inhibitor 1A Homo sapiens 64-68 12646197-9 2003 The induction of p21WAF/CIP1 expression by tacrolimus was dependent on TGF-beta since a neutralizing anti-TGF-beta antibody inhibited induction of p21WAF/CIP1in A-549 cells. Tacrolimus 43-53 cyclin dependent kinase inhibitor 1A Homo sapiens 24-28 12633892-0 2003 Hippocampal CA1 neuron survival and cytosolic FKBP12, the 12 kDa FK506-binding protein, after ischemia and tacrolimus treatment in gerbils. Tacrolimus 107-117 carbonic anhydrase 1 Homo sapiens 12-15 12646197-9 2003 The induction of p21WAF/CIP1 expression by tacrolimus was dependent on TGF-beta since a neutralizing anti-TGF-beta antibody inhibited induction of p21WAF/CIP1in A-549 cells. Tacrolimus 43-53 transforming growth factor beta 1 Homo sapiens 71-79 12646197-9 2003 The induction of p21WAF/CIP1 expression by tacrolimus was dependent on TGF-beta since a neutralizing anti-TGF-beta antibody inhibited induction of p21WAF/CIP1in A-549 cells. Tacrolimus 43-53 transforming growth factor beta 1 Homo sapiens 106-114 12646197-9 2003 The induction of p21WAF/CIP1 expression by tacrolimus was dependent on TGF-beta since a neutralizing anti-TGF-beta antibody inhibited induction of p21WAF/CIP1in A-549 cells. Tacrolimus 43-53 cyclin dependent kinase inhibitor 1A Homo sapiens 154-158 12604780-2 2003 We have previously shown that FAP48-FKBP complexes are dissociated by FK506 and rapamycin, suggesting that FAP48 is an endogenous ligand of FKBP. Tacrolimus 70-75 glomulin, FKBP associated protein Homo sapiens 30-35 12604780-2 2003 We have previously shown that FAP48-FKBP complexes are dissociated by FK506 and rapamycin, suggesting that FAP48 is an endogenous ligand of FKBP. Tacrolimus 70-75 glomulin, FKBP associated protein Homo sapiens 107-112 12606707-6 2003 Binding of BIG1, BIG2, and ARF to cell membranes in vitro was increased by guanosine 5"-[gamma-thio]triphosphate, and further increases were induced by FK506. Tacrolimus 152-157 ADP ribosylation factor guanine nucleotide exchange factor 1 Homo sapiens 11-15 12604780-4 2003 We report that overexpression of FAP48 results in the inhibition of cellular proliferation as does the exposure of Jurkat T cells to FK506. Tacrolimus 133-138 glomulin, FKBP associated protein Homo sapiens 33-38 12604780-7 2003 However, contrary to FK506, which blocks IL2 synthesis, we observed that FAP48-FKBP complexes increase IL2 production, thus revealing a previously uncharacterized aspect of the immunosuppressive mechanism of FK506. Tacrolimus 208-213 glomulin, FKBP associated protein Homo sapiens 73-78 12606707-6 2003 Binding of BIG1, BIG2, and ARF to cell membranes in vitro was increased by guanosine 5"-[gamma-thio]triphosphate, and further increases were induced by FK506. Tacrolimus 152-157 ADP ribosylation factor guanine nucleotide exchange factor 2 Homo sapiens 17-21 12604780-7 2003 However, contrary to FK506, which blocks IL2 synthesis, we observed that FAP48-FKBP complexes increase IL2 production, thus revealing a previously uncharacterized aspect of the immunosuppressive mechanism of FK506. Tacrolimus 208-213 interleukin 2 Homo sapiens 103-106 12606707-7 2003 Incubation of Jurkat T cells with FK506 increased binding of BIG1, BIG2, and ARF to Golgi and other membranes in a time- and concentration-dependent manner, without effects on clathrin or gamma-adaptin binding. Tacrolimus 34-39 ADP ribosylation factor guanine nucleotide exchange factor 1 Homo sapiens 61-65 12606707-7 2003 Incubation of Jurkat T cells with FK506 increased binding of BIG1, BIG2, and ARF to Golgi and other membranes in a time- and concentration-dependent manner, without effects on clathrin or gamma-adaptin binding. Tacrolimus 34-39 ADP ribosylation factor guanine nucleotide exchange factor 2 Homo sapiens 67-71 12606707-0 2003 Interaction of FK506-binding protein 13 with brefeldin A-inhibited guanine nucleotide-exchange protein 1 (BIG1): effects of FK506. Tacrolimus 15-20 ADP ribosylation factor guanine nucleotide exchange factor 1 Homo sapiens 45-104 12606707-8 2003 Binding of BIG1, BIG2, and ARF to membranes was also increased by L-732,531, an agonist structurally related to FK506, but was not increased by a related antagonist, L-685,818, nor by cyclosporin A or rapamycin. Tacrolimus 112-117 ADP ribosylation factor guanine nucleotide exchange factor 1 Homo sapiens 11-15 12606707-8 2003 Binding of BIG1, BIG2, and ARF to membranes was also increased by L-732,531, an agonist structurally related to FK506, but was not increased by a related antagonist, L-685,818, nor by cyclosporin A or rapamycin. Tacrolimus 112-117 ADP ribosylation factor guanine nucleotide exchange factor 2 Homo sapiens 17-21 12606707-0 2003 Interaction of FK506-binding protein 13 with brefeldin A-inhibited guanine nucleotide-exchange protein 1 (BIG1): effects of FK506. Tacrolimus 15-20 ADP ribosylation factor guanine nucleotide exchange factor 1 Homo sapiens 106-110 12642403-7 2003 FK506, but not V-10,367, inhibited the cytochrome c release out of the mitochondria and the caspase 3 activation, while both molecules inhibited the cleavage of Poly-(ADP-ribose)-polymerase (Parp) and prevented the expression of p53. Tacrolimus 0-5 caspase 3 Rattus norvegicus 92-101 12820430-0 2003 FK506 restores sensitivity of thymic lymphomas to calcium-mediated apoptosis and the inducible expression of Fas ligand. Tacrolimus 0-5 Fas ligand (TNF superfamily, member 6) Mus musculus 109-119 12820430-6 2003 Sensitivity to ionomycin-induced apoptosis could be restored by FK506 treatment, which also abolished the abrogation of induction of the FasL expression. Tacrolimus 64-69 Fas ligand (TNF superfamily, member 6) Mus musculus 137-141 12642403-7 2003 FK506, but not V-10,367, inhibited the cytochrome c release out of the mitochondria and the caspase 3 activation, while both molecules inhibited the cleavage of Poly-(ADP-ribose)-polymerase (Parp) and prevented the expression of p53. Tacrolimus 0-5 poly (ADP-ribose) polymerase 1 Rattus norvegicus 191-195 12642403-7 2003 FK506, but not V-10,367, inhibited the cytochrome c release out of the mitochondria and the caspase 3 activation, while both molecules inhibited the cleavage of Poly-(ADP-ribose)-polymerase (Parp) and prevented the expression of p53. Tacrolimus 0-5 Wistar clone pR53P1 p53 pseudogene Rattus norvegicus 229-232 12642403-8 2003 (3) FK506 and V-10,367 rapidly induced the expression of Hsp70 and Hsp27, but not Hsp90. Tacrolimus 4-9 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 57-62 12642403-8 2003 (3) FK506 and V-10,367 rapidly induced the expression of Hsp70 and Hsp27, but not Hsp90. Tacrolimus 4-9 heat shock protein family B (small) member 1 Rattus norvegicus 67-72 12632182-3 2003 This review focuses on the pharmacological modulation of the various functionally important sub-domains of the IP(3)R, including the IP(3)-binding domain, calmodulin-binding sites, adenine nucleotide-binding sites and the sites for interaction for FK506-binding proteins and other regulators. Tacrolimus 248-253 inositol 1,4,5-trisphosphate receptor type 3 Homo sapiens 111-117 12733828-9 2003 We also observed a similar pattern of largely nucleoplasmic Ca2+ mobilization upon exposure of cells to the immunosuppressant drug FK506 (tacrolimus), which binds to the RyR-associated immunophillin-binding proteins FKBP12 and FKBP12.6. Tacrolimus 131-136 LOC100009439 Oryctolagus cuniculus 170-173 12733828-9 2003 We also observed a similar pattern of largely nucleoplasmic Ca2+ mobilization upon exposure of cells to the immunosuppressant drug FK506 (tacrolimus), which binds to the RyR-associated immunophillin-binding proteins FKBP12 and FKBP12.6. Tacrolimus 131-136 peptidyl-prolyl cis-trans isomerase FKBP1A Oryctolagus cuniculus 216-222 12733828-9 2003 We also observed a similar pattern of largely nucleoplasmic Ca2+ mobilization upon exposure of cells to the immunosuppressant drug FK506 (tacrolimus), which binds to the RyR-associated immunophillin-binding proteins FKBP12 and FKBP12.6. Tacrolimus 131-136 peptidyl-prolyl cis-trans isomerase FKBP1B Oryctolagus cuniculus 227-235 12733828-9 2003 We also observed a similar pattern of largely nucleoplasmic Ca2+ mobilization upon exposure of cells to the immunosuppressant drug FK506 (tacrolimus), which binds to the RyR-associated immunophillin-binding proteins FKBP12 and FKBP12.6. Tacrolimus 138-148 LOC100009439 Oryctolagus cuniculus 170-173 12733828-9 2003 We also observed a similar pattern of largely nucleoplasmic Ca2+ mobilization upon exposure of cells to the immunosuppressant drug FK506 (tacrolimus), which binds to the RyR-associated immunophillin-binding proteins FKBP12 and FKBP12.6. Tacrolimus 138-148 peptidyl-prolyl cis-trans isomerase FKBP1A Oryctolagus cuniculus 216-222 12733828-9 2003 We also observed a similar pattern of largely nucleoplasmic Ca2+ mobilization upon exposure of cells to the immunosuppressant drug FK506 (tacrolimus), which binds to the RyR-associated immunophillin-binding proteins FKBP12 and FKBP12.6. Tacrolimus 138-148 peptidyl-prolyl cis-trans isomerase FKBP1B Oryctolagus cuniculus 227-235 12733828-11 2003 This suggested FK506 exerts both activatory and inhibitory effects upon RyR channels. Tacrolimus 15-20 LOC100009439 Oryctolagus cuniculus 72-75 12808349-10 2003 Daily administration of 0.5 mg/kg FK506 (an immunosuppressant) induced intragraft REG mRNA both in cryopreserved isografts and allografts (p < 0.01). Tacrolimus 34-39 regenerating family member 1 alpha Rattus norvegicus 82-85 12644894-0 2003 Regulation of human insulin gene transcription by the immunosuppressive drugs cyclosporin A and tacrolimus at concentrations that inhibit calcineurin activity and involving the transcription factor CREB. Tacrolimus 96-106 insulin Homo sapiens 20-27 12644894-0 2003 Regulation of human insulin gene transcription by the immunosuppressive drugs cyclosporin A and tacrolimus at concentrations that inhibit calcineurin activity and involving the transcription factor CREB. Tacrolimus 96-106 cAMP responsive element binding protein 1 Homo sapiens 198-202 12644894-3 2003 In a single study, tacrolimus (100 nM) inhibited human insulin gene transcription in the beta-cell line HIT. Tacrolimus 19-29 insulin Homo sapiens 55-62 12644894-4 2003 Using transfections of a human insulin-reporter gene into HIT cells, the present study shows that this inhibition is seen only at high concentrations of tacrolimus and is not caused by cyclosporin A. Tacrolimus 153-163 insulin Homo sapiens 31-38 12644894-5 2003 However, after stimulation by the major second messengers in the regulation of the insulin gene, cAMP and depolarization-induced calcium influx, both tacrolimus and cyclosporin A inhibited human insulin gene transcription in a concentration-dependent manner with IC(50) values of 1 nM and 30 nM, respectively. Tacrolimus 150-160 insulin Homo sapiens 83-90 12644894-5 2003 However, after stimulation by the major second messengers in the regulation of the insulin gene, cAMP and depolarization-induced calcium influx, both tacrolimus and cyclosporin A inhibited human insulin gene transcription in a concentration-dependent manner with IC(50) values of 1 nM and 30 nM, respectively. Tacrolimus 150-160 insulin Homo sapiens 195-202 12644894-7 2003 These data demonstrate for the first time that cAMP- and calcium-induced activity of the human insulin gene is mediated by CREB and blocked by both tacrolimus and cyclosporin A at concentrations that inhibit calcineurin phosphatase activity. Tacrolimus 148-158 insulin Homo sapiens 95-102 12644894-8 2003 Since also the immunosuppressive effects of cyclosporin A and tacrolimus are thought to be secondary to inhibition of calcineurin, the present study suggests that inhibition of human insulin gene transcription by the immunosuppressants is clinically important and may contribute to their diabetogenic effect. Tacrolimus 62-72 insulin Homo sapiens 183-190 12773967-6 2003 The IL-2R mAbs have been used with a variety of maintenance immunosuppression regimens double therapy with cyclosporine and prednisone, triple therapy with cyclosporine, azathioprine and prednisone and with newer regimens such as cyclosporine or tacrolimus, mycophenolate mofetil (MMF) and prednisone, and most recently with sirolimus, MMF and prednisone. Tacrolimus 246-256 interleukin 2 receptor subunit alpha Homo sapiens 4-9 12621554-0 2003 [Enhancement by FK506 of triptolide-induced inhibition of expression of COX-2 and iNOS in human rheumatoid arthritis synovial fibroblasts]. Tacrolimus 16-21 prostaglandin-endoperoxide synthase 2 Homo sapiens 72-77 12621554-10 2003 NF-kappa B activity in TNF alpha-stimulated synovial cells was suppressed more profoundly by FK506 plus TP (10 microg/L) treatment than those with TP (10 microg/L) alone. Tacrolimus 93-98 nuclear factor kappa B subunit 1 Homo sapiens 0-10 12621554-0 2003 [Enhancement by FK506 of triptolide-induced inhibition of expression of COX-2 and iNOS in human rheumatoid arthritis synovial fibroblasts]. Tacrolimus 16-21 nitric oxide synthase 2 Homo sapiens 82-86 12621554-10 2003 NF-kappa B activity in TNF alpha-stimulated synovial cells was suppressed more profoundly by FK506 plus TP (10 microg/L) treatment than those with TP (10 microg/L) alone. Tacrolimus 93-98 tumor necrosis factor Homo sapiens 23-32 12621554-1 2003 To explore the effects of FK506 on the inhibition by triptolide (TP) of cell proliferation and expressions of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS) and their inducing products PGE2, NO in human rheumatoid arthritis synovial fibroblasts (RASF), and to study the mechanisms of combination of FK506 and TP in RA therapy, RASF used in the experiments were obtained from synovial tissue of patients with RA and were cultured. Tacrolimus 26-31 nitric oxide synthase 2 Homo sapiens 136-167 12621554-12 2003 In conclusion, FK506 enhanced TP-mediated down-regulation of COX-2, iNOS and their inducing products PGE2, NO in human RASF by suppressing the activity of NF-kappa B. Tacrolimus 15-20 prostaglandin-endoperoxide synthase 2 Homo sapiens 61-66 12621554-12 2003 In conclusion, FK506 enhanced TP-mediated down-regulation of COX-2, iNOS and their inducing products PGE2, NO in human RASF by suppressing the activity of NF-kappa B. Tacrolimus 15-20 nitric oxide synthase 2 Homo sapiens 68-72 12621554-8 2003 Combined treatment of FK506 and a lower concentration of TP (10 microg/L) down-regulated COX-2 and iNOS mRNA and protein expression, and their inducing products PGE2 and NO of synovial fibroblasts. Tacrolimus 22-27 prostaglandin-endoperoxide synthase 2 Homo sapiens 89-94 12621554-12 2003 In conclusion, FK506 enhanced TP-mediated down-regulation of COX-2, iNOS and their inducing products PGE2, NO in human RASF by suppressing the activity of NF-kappa B. Tacrolimus 15-20 nuclear factor kappa B subunit 1 Homo sapiens 155-165 12621554-8 2003 Combined treatment of FK506 and a lower concentration of TP (10 microg/L) down-regulated COX-2 and iNOS mRNA and protein expression, and their inducing products PGE2 and NO of synovial fibroblasts. Tacrolimus 22-27 nitric oxide synthase 2 Homo sapiens 99-103 14753416-0 2003 Calcineurin and cyclophilin D are differential targets of neuroprotection by immunosuppressants CsA and FK506 in ischemic brain damage. Tacrolimus 104-109 peptidylprolyl isomerase F Rattus norvegicus 16-29 12548506-12 2003 Based on the derived model, a 70-kg patient with an aspartate aminotransferase (AST) level less than 70 U/L would require a tacrolimus dose of 4.7 mg twice daily to achieve a steady-state trough concentration of 10 ng/mL. Tacrolimus 124-134 solute carrier family 17 member 5 Homo sapiens 52-78 12548506-12 2003 Based on the derived model, a 70-kg patient with an aspartate aminotransferase (AST) level less than 70 U/L would require a tacrolimus dose of 4.7 mg twice daily to achieve a steady-state trough concentration of 10 ng/mL. Tacrolimus 124-134 solute carrier family 17 member 5 Homo sapiens 80-83 12548534-0 2003 Effects of FK506 on myasthenia gravis patients with high interleukin-2 productivity in peripheral blood mononuclear cells. Tacrolimus 11-16 interleukin 2 Homo sapiens 57-70 12559954-7 2003 A correlative inhibition of JNK activation by immunosuppressive drug FK 506 induced ERK activation and MKP-1 expression, and suppressed RINm5F cell death. Tacrolimus 69-75 mitogen-activated protein kinase 8 Rattus norvegicus 28-31 12668156-1 2003 Pretransplant treatment of recipients with recombinant human granulocyte colony-stimulating factor (rhG-CSF, 250 microg/kg/day s.c. for 5 days) facilitates heart allograft acceptance in tacrolimus-treated rat recipients. Tacrolimus 186-196 colony stimulating factor 3 Homo sapiens 61-98 12926794-5 2003 Circulating IL-16 levels decreased significantly in patients with atopic dermatitis after topical treatment with corticosteroids or tacrolimus. Tacrolimus 132-142 interleukin 16 Homo sapiens 12-17 12605126-0 2003 Facilitation of tacrolimus-induced heart-allograft acceptability by pretransplant host treatment with granulocyte colony-stimulating factor: interleukin-12-restricted suppression of intragraft monokine mRNA expression. Tacrolimus 16-26 colony stimulating factor 3 Homo sapiens 102-139 12552015-3 2003 We have documented that a cellular protein that binds the immunosuppressant drug FK506, termed the FK506-binding protein (FKBP52), interacts with the single-stranded D sequence within the AAV inverted terminal repeats, inhibits viral second-strand DNA synthesis, and consequently limits high-efficiency transgene expression (K. Qing, J. Hansen, K. A. Weigel-Kelley, M. Tan, S. Zhou, and A. Srivastava, J. Tacrolimus 81-86 FKBP prolyl isomerase 4 Homo sapiens 122-128 12676530-4 2003 FK506 inhibits Erk and PI-3K/Akt signaling, two crucial pro-survival pathways. Tacrolimus 0-5 Eph receptor B1 Rattus norvegicus 15-18 12676530-4 2003 FK506 inhibits Erk and PI-3K/Akt signaling, two crucial pro-survival pathways. Tacrolimus 0-5 AKT serine/threonine kinase 1 Rattus norvegicus 29-32 12676530-5 2003 The levels of phosphorylated Akt and p42/44 Erk decline in few hours after FK506 addition. Tacrolimus 75-80 AKT serine/threonine kinase 1 Rattus norvegicus 29-32 12676530-5 2003 The levels of phosphorylated Akt and p42/44 Erk decline in few hours after FK506 addition. Tacrolimus 75-80 Eph receptor B1 Rattus norvegicus 44-47 12676530-6 2003 Furthermore, in FK506-treated astrocyte cultures the levels of mRNA encoding PDGF, bFGF, and CNTF decreased. Tacrolimus 16-21 fibroblast growth factor 2 Rattus norvegicus 83-87 12676530-6 2003 Furthermore, in FK506-treated astrocyte cultures the levels of mRNA encoding PDGF, bFGF, and CNTF decreased. Tacrolimus 16-21 ciliary neurotrophic factor Rattus norvegicus 93-97 12676530-7 2003 Downregulation of growth factor expression by FK506 may play a role in the inhibition of mitogenic/hypertrophic responses. Tacrolimus 46-51 myotrophin Rattus norvegicus 18-31 12676530-9 2003 Interestingly, expression of BDNF increased in a dose-dependent manner in FK506-treated astrocytes. Tacrolimus 74-79 brain-derived neurotrophic factor Rattus norvegicus 29-33 12676530-10 2003 Upregulation of BDNF mRNA and protein level in astrocytes exposed to FK506 may underlie neuroprotective action of FK506. Tacrolimus 69-74 brain-derived neurotrophic factor Rattus norvegicus 16-20 12676530-10 2003 Upregulation of BDNF mRNA and protein level in astrocytes exposed to FK506 may underlie neuroprotective action of FK506. Tacrolimus 114-119 brain-derived neurotrophic factor Rattus norvegicus 16-20 12548123-11 2003 CONCLUSION: Three years of tacrolimus therapy demonstrated excellent efficacy and safety in more than 1,000 renal transplant recipients, including recipients of living ABO-incompatible and non-heart-beating donor grafts, with favorable graft function maintained. Tacrolimus 27-37 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 168-171 12559954-7 2003 A correlative inhibition of JNK activation by immunosuppressive drug FK 506 induced ERK activation and MKP-1 expression, and suppressed RINm5F cell death. Tacrolimus 69-75 Eph receptor B1 Rattus norvegicus 84-87 12559954-7 2003 A correlative inhibition of JNK activation by immunosuppressive drug FK 506 induced ERK activation and MKP-1 expression, and suppressed RINm5F cell death. Tacrolimus 69-75 dual specificity phosphatase 1 Rattus norvegicus 103-108 12499534-2 2003 The N-terminal domain of FKBP52 (FKBP52-N; residues 1-140) is responsible for peptidyl-prolyl isomerase activity and binding of FK506. Tacrolimus 128-133 FKBP prolyl isomerase 4 Homo sapiens 25-31 12499534-2 2003 The N-terminal domain of FKBP52 (FKBP52-N; residues 1-140) is responsible for peptidyl-prolyl isomerase activity and binding of FK506. Tacrolimus 128-133 FKBP prolyl isomerase 4 Homo sapiens 33-39 12499534-5 2003 FKBP52-N is able to bind FK506 in a similar way to FKBP12. Tacrolimus 25-30 FKBP prolyl isomerase 4 Homo sapiens 0-6 12499534-7 2003 The Pro120 change corresponding to Gly89 in FKBP12 limits the conformational adaptation between the loop (residues 108-127) and FK506 and decreases the FK506 affinity, while the Lys121 substitution corresponding to Ile90 of FKBP12 destroys a key interaction between FKBP52-N and calcineurin. Tacrolimus 128-133 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 44-50 12499534-7 2003 The Pro120 change corresponding to Gly89 in FKBP12 limits the conformational adaptation between the loop (residues 108-127) and FK506 and decreases the FK506 affinity, while the Lys121 substitution corresponding to Ile90 of FKBP12 destroys a key interaction between FKBP52-N and calcineurin. Tacrolimus 152-157 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 44-50 14753416-5 2003 Both CsA and FK506 are specific inhibitors of immunophilins, (CsA inhibits cyclophilins, FK506 inhibits FKBPs), and of calcineurin, a type 2B Ser/Thr phosphatase that is abundant in the central nervous system. Tacrolimus 13-18 peptidylprolyl isomerase F Rattus norvegicus 75-87 12871167-4 2003 However, recent studies indicate that both CsA and FK506 can block activation of JNK and p38 signaling pathways during T cell activation. Tacrolimus 51-56 mitogen-activated protein kinase 14 Homo sapiens 89-92 12502928-1 2003 FKBP12 is a cytosolic FK506 binding protein that interacts with calcineurin and thereby mediates the immunosuppressive effects of FK506. Tacrolimus 22-27 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 0-6 12401518-5 2003 We also found that this stimulating effect on Akt phosphorylation by PKC inhibitors was not the result of phosphatase inhibition, since treatment with PP2A, PP2B and tyrosine phosphatase inhibitors (okadaic acid, FK506 and sodium orthovanadate, respectively) had no effect. Tacrolimus 213-218 AKT serine/threonine kinase 1 Homo sapiens 46-49 12401518-5 2003 We also found that this stimulating effect on Akt phosphorylation by PKC inhibitors was not the result of phosphatase inhibition, since treatment with PP2A, PP2B and tyrosine phosphatase inhibitors (okadaic acid, FK506 and sodium orthovanadate, respectively) had no effect. Tacrolimus 213-218 protein kinase C beta Homo sapiens 69-72 12401523-0 2003 Modulation of the electrophoretic mobility of the linker for activation of T cells (LAT) by the calcineurin inhibitors CsA and FK506: LAT is a potential substrate for PKC and calcineurin signaling pathways. Tacrolimus 127-132 linker for activation of T cells Homo sapiens 84-87 12401523-0 2003 Modulation of the electrophoretic mobility of the linker for activation of T cells (LAT) by the calcineurin inhibitors CsA and FK506: LAT is a potential substrate for PKC and calcineurin signaling pathways. Tacrolimus 127-132 linker for activation of T cells Homo sapiens 134-137 12401523-4 2003 We show that TCR ligation in the presence of FK506 or CsA induced rapid modifications in LAT that modulate the electrophoretic mobility of the molecule in SDS-PAGE. Tacrolimus 45-50 linker for activation of T cells Homo sapiens 89-92 12401523-5 2003 Calcineurin, a target for CsA and FK506, dephosphorylated LAT in vitro and restored its electrophoretic mobility. Tacrolimus 34-39 linker for activation of T cells Homo sapiens 58-61 12871167-5 2003 CsA and FK506, thus, have two distinct mechanisms of action; one is the inhibition of the protein phosphatase activity of calcineurin, leading to the blockade of the nuclear translocation of NFAT transcription factors, and the other is the suppression of JNK and p38 activation pathways. Tacrolimus 8-13 mitogen-activated protein kinase 14 Homo sapiens 263-266 12871170-2 2003 The 565 kDa RyR protein forms a tetrameric channel that is part of a macromolecular signaling complex that also includes four FK506 binding proteins (FKBPs). Tacrolimus 126-131 ryanodine receptor 1 Homo sapiens 12-15 12594826-6 2003 The effect of NF-ATc2 was efficiently blocked with FK506, which prevented its nuclear translocation through inactivation of calcineurin. Tacrolimus 51-56 nuclear factor of activated T cells 2 Homo sapiens 14-21 12887261-15 2003 In the current immunosuppressive regimens, a calcineurin inhibitor, either tacrolimus or cyclosporin, is the essential basic standard immunosuppressant. Tacrolimus 75-85 calcineurin binding protein 1 Homo sapiens 45-66 12594826-7 2003 In addition, TR3 induction during BCR cross-linking was reduced by FK506 and the VIVIT peptide, which is a highly selective inhibitor for NF-AT. Tacrolimus 67-72 TNF receptor superfamily member 25 Homo sapiens 13-16 12469222-0 2003 Renin mRNA expression and renal dysfunction in tacrolimus-induced acute nephrotoxicity. Tacrolimus 47-57 renin Rattus norvegicus 0-5 15035807-3 2003 FK506 (tacrolimus), an immunosuppressant, is known to reduce the activation of microglia in vitro and affect the expression of various cytokines like interleukin-1, interleukin-6 and tumor necrosis factor. Tacrolimus 0-5 interleukin 6 Rattus norvegicus 165-178 15035807-3 2003 FK506 (tacrolimus), an immunosuppressant, is known to reduce the activation of microglia in vitro and affect the expression of various cytokines like interleukin-1, interleukin-6 and tumor necrosis factor. Tacrolimus 7-17 interleukin 6 Rattus norvegicus 165-178 15035807-11 2003 The myeloperoxidase enzyme level was significantly increased in carrageenan-treated animals, which was significantly reversed by FK506 treatment. Tacrolimus 129-134 myeloperoxidase Rattus norvegicus 4-19 12469222-4 2003 The purpose of this study was to elucidate the role of renin-angiotensin system (RAS) in tacrolimus-induced acute nephrotoxicity. Tacrolimus 89-99 renin Rattus norvegicus 55-60 12469222-5 2003 We examined the renal mRNA levels of renin in order to elucidate the relationship between plasma renin activity (PRA) and tacrolimus-induced renal dysfunction. Tacrolimus 122-132 renin Rattus norvegicus 37-42 12469222-5 2003 We examined the renal mRNA levels of renin in order to elucidate the relationship between plasma renin activity (PRA) and tacrolimus-induced renal dysfunction. Tacrolimus 122-132 renin Rattus norvegicus 97-102 12469222-8 2003 Renin mRNA levels in the renal cortex in tacrolimus treated rats significantly increased when compared to the vehicle-treated rats. Tacrolimus 41-51 renin Rattus norvegicus 0-5 12490779-4 2002 METHODS: The dose-normalized blood concentrations of tacrolimus at 3 months after renal transplantation were related to CYP3AP1 and multiple drug resistance (MDR)-1 genotypes determined by polymerase chain reaction followed by restriction fragment length polymorphism analysis. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 51, pseudogene Homo sapiens 120-127 12472797-12 2003 FK506 up-regulated renal cortical gene expression of chemoattractant proteins, monocyte chemoattractant protein-1 (MCP-1) and osteopontin. Tacrolimus 0-5 C-C motif chemokine ligand 2 Rattus norvegicus 79-113 12472797-12 2003 FK506 up-regulated renal cortical gene expression of chemoattractant proteins, monocyte chemoattractant protein-1 (MCP-1) and osteopontin. Tacrolimus 0-5 C-C motif chemokine ligand 2 Rattus norvegicus 115-120 12472797-12 2003 FK506 up-regulated renal cortical gene expression of chemoattractant proteins, monocyte chemoattractant protein-1 (MCP-1) and osteopontin. Tacrolimus 0-5 secreted phosphoprotein 1 Rattus norvegicus 126-137 12510191-2 2003 Here we show that mitochondrial FK506-binding protein 38 (FKBP38), unlike FKBP12, binds to and inhibits calcineurin in the absence of the immunosuppressant FK506, suggesting that FKBP38 is an inherent inhibitor of this phosphatase. Tacrolimus 32-37 FKBP prolyl isomerase 8 Homo sapiens 58-64 12510191-2 2003 Here we show that mitochondrial FK506-binding protein 38 (FKBP38), unlike FKBP12, binds to and inhibits calcineurin in the absence of the immunosuppressant FK506, suggesting that FKBP38 is an inherent inhibitor of this phosphatase. Tacrolimus 32-37 FKBP prolyl isomerase 8 Homo sapiens 179-185 12698322-2 2003 Peptide bond cis/trans isomerases as cyclophilins, Fk506-binding proteins, parvulins, and bacterial hsp70 generally assist in the interconversion of the polypeptide substrate cis/trans isomers, and rate acceleration is the dominating mechanism of action in cells. Tacrolimus 51-56 heat shock protein family A (Hsp70) member 4 Homo sapiens 100-105 12507579-7 2003 We show that BMP-2 engineered allogeneic MSC can repair critical bone defects to the same degree as rats treated with BMP-2 engineered autologous MSC, if the allogeneic group receives short-term treatment with immunosuppressant FK506. Tacrolimus 228-233 bone morphogenetic protein 2 Rattus norvegicus 13-18 12490609-2 2003 FK506 binds to and reverses effects of FKBP on RyR, thus increasing RyR sensitivity to Ca2+, decreasing RyR cooperativity, and increasing RyR open probability. Tacrolimus 0-5 LOC100009439 Oryctolagus cuniculus 47-50 12490609-2 2003 FK506 binds to and reverses effects of FKBP on RyR, thus increasing RyR sensitivity to Ca2+, decreasing RyR cooperativity, and increasing RyR open probability. Tacrolimus 0-5 LOC100009439 Oryctolagus cuniculus 68-71 12490609-2 2003 FK506 binds to and reverses effects of FKBP on RyR, thus increasing RyR sensitivity to Ca2+, decreasing RyR cooperativity, and increasing RyR open probability. Tacrolimus 0-5 LOC100009439 Oryctolagus cuniculus 68-71 12490609-2 2003 FK506 binds to and reverses effects of FKBP on RyR, thus increasing RyR sensitivity to Ca2+, decreasing RyR cooperativity, and increasing RyR open probability. Tacrolimus 0-5 LOC100009439 Oryctolagus cuniculus 68-71 12490609-11 2003 These results indicate that alteration of the functions of RyR by FK506-mediated dissociation of FKBP from RyR has different species-dependent effects on SR Ca2+ load and thus [Ca2+]i transients. Tacrolimus 66-71 LOC100009439 Oryctolagus cuniculus 59-62 12490609-11 2003 These results indicate that alteration of the functions of RyR by FK506-mediated dissociation of FKBP from RyR has different species-dependent effects on SR Ca2+ load and thus [Ca2+]i transients. Tacrolimus 66-71 LOC100009439 Oryctolagus cuniculus 107-110 12490609-12 2003 This difference may result from the fact that [Na+]i is low in rabbit myocytes, allowing extrusion by Na+/Ca2+ exchange of Ca2+ released by FK506-induced dissociation of FKBP12.6 from SR RyR. Tacrolimus 140-145 peptidyl-prolyl cis-trans isomerase FKBP1B Oryctolagus cuniculus 170-178 12490609-12 2003 This difference may result from the fact that [Na+]i is low in rabbit myocytes, allowing extrusion by Na+/Ca2+ exchange of Ca2+ released by FK506-induced dissociation of FKBP12.6 from SR RyR. Tacrolimus 140-145 LOC100009439 Oryctolagus cuniculus 187-190 12927209-5 2003 After 90 min of transient middle cerebral artery occlusion in male rats the expression of FKBP12, 52 and 65 was analyzed by Western blot in FK506-treated and control animals and the peptidyl-prolyl cis/trans isomerase activity was determined. Tacrolimus 140-145 FKBP prolyl isomerase 1A Rattus norvegicus 90-96 12927209-12 2003 c-Jun phosphorylation in neurons of the peri-infarct area and Fas-L expression was reduced by FK506 treatment whereas ATF-2 expression was preserved. Tacrolimus 94-99 Fas ligand Rattus norvegicus 62-67 12927209-14 2003 It was shown for the first time that neuroprotection by FK506 also included the suppression of the cerebral peptidyl-prolyl cis/trans isomerase activity of FKBP in vivo whereas the expression levels of FKBP12, 52 and 65 following ischemia changed slightly and FK506 treatment does not suppress the expression patterns. Tacrolimus 56-61 FKBP prolyl isomerase 1A Rattus norvegicus 202-208 12856882-4 2003 This phosphorylation was maximal 2 h after treatment with either agent and declined to basal levels by 24 h. Inhibitor studies revealed that both UVB and the BMT diol phosphorylate BRCA1 through the FK506-binding protein-FKBP rapamycin-associated binding protein pathway, but the BMT diol also led to phosphorylation of BRCA1 through casein kinase II. Tacrolimus 199-204 BRCA1 DNA repair associated Homo sapiens 181-186 12856882-4 2003 This phosphorylation was maximal 2 h after treatment with either agent and declined to basal levels by 24 h. Inhibitor studies revealed that both UVB and the BMT diol phosphorylate BRCA1 through the FK506-binding protein-FKBP rapamycin-associated binding protein pathway, but the BMT diol also led to phosphorylation of BRCA1 through casein kinase II. Tacrolimus 199-204 BRCA1 DNA repair associated Homo sapiens 320-325 12490779-4 2002 METHODS: The dose-normalized blood concentrations of tacrolimus at 3 months after renal transplantation were related to CYP3AP1 and multiple drug resistance (MDR)-1 genotypes determined by polymerase chain reaction followed by restriction fragment length polymorphism analysis. Tacrolimus 53-63 ATP binding cassette subfamily B member 1 Homo sapiens 132-164 12490779-5 2002 RESULTS: We found that a single nucleotide polymorphism in the CYP3AP1 pseudogene (A/G(44)) that previously has been noted to be more common in African Americans and strongly associated with hepatic CYP3A5 activity correlated well with the tacrolimus dose requirement. Tacrolimus 240-250 cytochrome P450 family 3 subfamily A member 51, pseudogene Homo sapiens 63-70 12490779-5 2002 RESULTS: We found that a single nucleotide polymorphism in the CYP3AP1 pseudogene (A/G(44)) that previously has been noted to be more common in African Americans and strongly associated with hepatic CYP3A5 activity correlated well with the tacrolimus dose requirement. Tacrolimus 240-250 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 199-205 12490779-7 2002 CONCLUSIONS: The CYP3AP1 genotype is a major factor in determining the dose requirement for tacrolimus, and genotyping may be of value in planning patient-specific drug dosing. Tacrolimus 92-102 cytochrome P450 family 3 subfamily A member 51, pseudogene Homo sapiens 17-24 12486498-3 2002 Basal insulin release after exposure to cytokines and FK506 was significantly higher than in control islets. Tacrolimus 54-59 insulin Homo sapiens 6-13 12427484-0 2002 Effect of FK506 on osteoinduction by recombinant human bone morphogenetic protein-2. Tacrolimus 10-15 bone morphogenetic protein 2 Homo sapiens 55-83 12427484-5 2002 In this study, we evaluated the influence of FK506 on osteoinduction by recombinant human bone morphogenetic protein-2 (rhBMP-2) using atelopeptide type I collagen as a carrier. Tacrolimus 45-50 bone morphogenetic protein 2 Homo sapiens 90-118 12450577-4 2002 Topical administration of FK506 (tacrolimus hydrate) dramatically suppressed ear swelling and epidermal hyperplasia as well as the increase in interferon-gamma expression. Tacrolimus 26-31 interferon gamma Rattus norvegicus 143-159 12450577-4 2002 Topical administration of FK506 (tacrolimus hydrate) dramatically suppressed ear swelling and epidermal hyperplasia as well as the increase in interferon-gamma expression. Tacrolimus 33-43 interferon gamma Rattus norvegicus 143-159 12486498-8 2002 FK506 was, among those tested, the immunosuppressive drug that most profoundly altered the normal insulin secretory pattern of human beta-cells, whereas CsA was the only inhibiting insulin gene expression. Tacrolimus 0-5 insulin Homo sapiens 98-105 12685503-1 2002 Tacrolimus, a potent immunosuppressive drug, is known to be metabolized predominantly in the liver by cytochrome P450 3A (CYP3A). Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 102-120 12496747-8 2002 On the basis of the derived model, a patient with normal AST (20 U/L) or high AST (200 U/L) concentrations 7 days after commencement of therapy would require a tacrolimus dose of 4.6 mg or 4.0 mg, respectively, to achieve a steady-state trough concentration of 10 ng/mL. Tacrolimus 160-170 solute carrier family 17 member 5 Homo sapiens 78-81 14561179-4 2002 Cyclosporine and tacrolimus are calcineurin inhibitors that impair the transcription of interleukin 2, reduce the expression of cytokines, and diminish T lymphocyte proliferation. Tacrolimus 17-27 interleukin 2 Homo sapiens 88-101 12685503-1 2002 Tacrolimus, a potent immunosuppressive drug, is known to be metabolized predominantly in the liver by cytochrome P450 3A (CYP3A). Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 122-127 12685503-5 2002 Tacrolimus had no effect on any CYP at concentrations below 1 microM, while at higher concentrations it had a mild inhibitory effect on CYP3A4 and 3A5. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 136-142 14599350-1 2002 FKBP12 is best known as the target of the widely used immunosuppressive drug FK506 but may also play a role in neuronal survival. Tacrolimus 77-82 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 0-6 12494287-1 2002 To evaluate whether FK506 and other immunophilin ligands may have potential therapeutic efficacy for erectile function preservation after penile nerve injury, we demonstrated localizations of the immunophilin FK506 binding protein 12 (FKBP 12) in intact and injured rat penile nerves and correlated these findings with localizations of neuronal nitric oxide synthase (nNOS), which neuronally forms nitric oxide for mediation of penile erection, in response to systemically administered FK506. Tacrolimus 20-25 FKBP prolyl isomerase 1A Rattus norvegicus 235-242 12494287-1 2002 To evaluate whether FK506 and other immunophilin ligands may have potential therapeutic efficacy for erectile function preservation after penile nerve injury, we demonstrated localizations of the immunophilin FK506 binding protein 12 (FKBP 12) in intact and injured rat penile nerves and correlated these findings with localizations of neuronal nitric oxide synthase (nNOS), which neuronally forms nitric oxide for mediation of penile erection, in response to systemically administered FK506. Tacrolimus 209-214 FKBP prolyl isomerase 1A Rattus norvegicus 235-242 12427154-0 2002 Expression of TGF-beta and fibrogenic genes in transplant recipients with tacrolimus and cyclosporine nephrotoxicity. Tacrolimus 74-84 transforming growth factor beta 1 Homo sapiens 14-22 12451243-0 2002 Lowered blood concentration of tacrolimus and its recovery with changes in expression of CYP3A and P-glycoprotein after high-dose steroid therapy. Tacrolimus 31-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 89-94 12453218-9 2002 In contrast, part of the alkali-induced expression of PHO89 was maintained in pho4 or pho2 cells, but was fully abolished in a crz1 strain or in the presence of FK506. Tacrolimus 161-166 Pho89p Saccharomyces cerevisiae S288C 54-59 12593072-7 2002 Tacrolimus may alleviate rejection partly by inhibiting p38 mitogen-activated protein kinase. Tacrolimus 0-10 mitogen activated protein kinase 14 Rattus norvegicus 56-59 12451243-0 2002 Lowered blood concentration of tacrolimus and its recovery with changes in expression of CYP3A and P-glycoprotein after high-dose steroid therapy. Tacrolimus 31-41 ATP binding cassette subfamily B member 1 Homo sapiens 99-113 12451243-10 2002 CONCLUSION: Our results indicate that the decrease in the blood FK506 concentration caused by high-dose steroid therapy is a consequence of the induction of P-glycoprotein and CYP3A in the liver and intestine, and these changes were reversed within 2 weeks after cessation of steroid therapy. Tacrolimus 64-69 ATP binding cassette subfamily B member 1 Homo sapiens 157-171 12451243-10 2002 CONCLUSION: Our results indicate that the decrease in the blood FK506 concentration caused by high-dose steroid therapy is a consequence of the induction of P-glycoprotein and CYP3A in the liver and intestine, and these changes were reversed within 2 weeks after cessation of steroid therapy. Tacrolimus 64-69 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 176-181 12530540-7 2002 A correlative inhibition of JNK activation by the immunosuppressive drug FK506, as well as a selective inhibition of p38 MAPK activation by SB 203580, significantly suppressed procaspase-3 processing and the extent of amylin-induced cell death. Tacrolimus 73-78 mitogen-activated protein kinase 8 Homo sapiens 28-31 12228224-5 2002 Caspase-3 activation induced by thapsigargin was blocked by increasing the phosphorylation of Ser-9-GSK3beta with insulin-like growth factor-1 or with the phosphatase inhibitors okadaic acid and calyculin A, but the calcineurin inhibitors FK506 and cyclosporin A were ineffective. Tacrolimus 239-244 caspase 3 Homo sapiens 0-9 12228224-5 2002 Caspase-3 activation induced by thapsigargin was blocked by increasing the phosphorylation of Ser-9-GSK3beta with insulin-like growth factor-1 or with the phosphatase inhibitors okadaic acid and calyculin A, but the calcineurin inhibitors FK506 and cyclosporin A were ineffective. Tacrolimus 239-244 glycogen synthase kinase 3 beta Homo sapiens 100-108 12408978-1 2002 NFAT (nuclear factor of activated T cells) plays a pivotal role in inducible gene transcription during the immune response and functions as a major target for immunosuppressive drugs such as cyclosporin A and FK-506. Tacrolimus 209-215 nuclear factor of activated T cells 2 Homo sapiens 0-4 12530540-8 2002 Moreover, simultaneous pretreatment with both FK506 and SB 203580, or with the caspase-3 inhibitor Ac-DEVD-CHO alone, almost completely abolished procaspase-3 processing and cell death. Tacrolimus 46-51 caspase 3 Homo sapiens 146-158 12409674-6 2002 RESULTS: Cyclosporine, tacrolimus, and anti-IL-2R monoclonal antibody therapy abrogated the effect of a single-dose protocol of anti-CD154 therapy. Tacrolimus 23-33 CD40 ligand Mus musculus 133-138 12530540-7 2002 A correlative inhibition of JNK activation by the immunosuppressive drug FK506, as well as a selective inhibition of p38 MAPK activation by SB 203580, significantly suppressed procaspase-3 processing and the extent of amylin-induced cell death. Tacrolimus 73-78 caspase 3 Homo sapiens 176-188 12530540-7 2002 A correlative inhibition of JNK activation by the immunosuppressive drug FK506, as well as a selective inhibition of p38 MAPK activation by SB 203580, significantly suppressed procaspase-3 processing and the extent of amylin-induced cell death. Tacrolimus 73-78 islet amyloid polypeptide Homo sapiens 218-224 12195013-6 2002 This enhancement was nearly abolished by blocking the nuclear translocation of NFATc by using the calcineurin inhibitor FK506. Tacrolimus 120-125 nuclear factor of activated T cells 1 Homo sapiens 79-84 12414688-6 2002 This effect was similar to those of FK506 and rapamycin, two drugs known to cause dissociation of FKBP12 from RyR. Tacrolimus 36-41 FK506 binding protein 1a Mus musculus 98-104 12414688-6 2002 This effect was similar to those of FK506 and rapamycin, two drugs known to cause dissociation of FKBP12 from RyR. Tacrolimus 36-41 ryanodine receptor 1, skeletal muscle Mus musculus 110-113 12202955-7 2002 FK506 had no effect on cell proliferation or TRAP activity at concentrations up to 2000 ng/ml; however, like CsA, 1000 ng/ml FK506 inhibited TRAP+MNC formation and the expression of TRAP and CTR mRNAs. Tacrolimus 125-130 acid phosphatase 5, tartrate resistant Mus musculus 141-145 12202955-7 2002 FK506 had no effect on cell proliferation or TRAP activity at concentrations up to 2000 ng/ml; however, like CsA, 1000 ng/ml FK506 inhibited TRAP+MNC formation and the expression of TRAP and CTR mRNAs. Tacrolimus 125-130 acid phosphatase 5, tartrate resistant Mus musculus 141-145 12202955-7 2002 FK506 had no effect on cell proliferation or TRAP activity at concentrations up to 2000 ng/ml; however, like CsA, 1000 ng/ml FK506 inhibited TRAP+MNC formation and the expression of TRAP and CTR mRNAs. Tacrolimus 125-130 calcitonin receptor Mus musculus 191-194 12387959-12 2002 Tacrolimus should be the first choice of calcineurin inhibitor for patients receiving their first liver graft. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 41-62 12386643-7 2002 When stimulated with 2-micromol/L thrombin receptor-activating peptide, CD62 expression in platelets from patients treated with azathioprine (63% +/- 17%; P <.05), cyclosporine (51% +/- 23%; P <.05), and tacrolimus (50% +/- 22%; P <.05) was elevated compared with control subjects (33% +/- 19%). Tacrolimus 210-220 selectin P Homo sapiens 72-76 12386643-13 2002 However, the "newer" immunosuppressive agents tacrolimus and mycophenolate mofetil seemed to have fewer unfavorable effects on platelet CD62 expression and PAC1 expression and aggregation. Tacrolimus 46-56 selectin P Homo sapiens 136-140 12386643-13 2002 However, the "newer" immunosuppressive agents tacrolimus and mycophenolate mofetil seemed to have fewer unfavorable effects on platelet CD62 expression and PAC1 expression and aggregation. Tacrolimus 46-56 ADCYAP receptor type I Homo sapiens 156-160 12207006-2 2002 Data regarding the role of cyclosporine (CsA) and FK-506 in AMT have shown that CsA is less effective than FK-506. Tacrolimus 50-56 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 80-83 12368659-8 2002 In conclusion, tacrolimus, used in combination with t-PA, augmented therapeutic efficacy on brain damage associated with focal ischemia and extended the therapeutic time window compared to single-drug treatments. Tacrolimus 15-25 plasminogen activator, tissue type Rattus norvegicus 52-56 12234299-9 2002 Thirdly, triptolide (4 to 8 ng/mL), CsA (500 to 2500 ng/mL) and FK506 (1250 ng/mL) inhibited up-regulation of CD40 and B7h mRNA, the effect on B7h and CD40 mRNA expression by CsA and FK506 being greater than that on C3 mRNA expression. Tacrolimus 64-69 CD40 molecule Homo sapiens 110-114 12234299-9 2002 Thirdly, triptolide (4 to 8 ng/mL), CsA (500 to 2500 ng/mL) and FK506 (1250 ng/mL) inhibited up-regulation of CD40 and B7h mRNA, the effect on B7h and CD40 mRNA expression by CsA and FK506 being greater than that on C3 mRNA expression. Tacrolimus 64-69 CD274 molecule Homo sapiens 119-122 12234299-9 2002 Thirdly, triptolide (4 to 8 ng/mL), CsA (500 to 2500 ng/mL) and FK506 (1250 ng/mL) inhibited up-regulation of CD40 and B7h mRNA, the effect on B7h and CD40 mRNA expression by CsA and FK506 being greater than that on C3 mRNA expression. Tacrolimus 64-69 CD274 molecule Homo sapiens 143-146 12234299-9 2002 Thirdly, triptolide (4 to 8 ng/mL), CsA (500 to 2500 ng/mL) and FK506 (1250 ng/mL) inhibited up-regulation of CD40 and B7h mRNA, the effect on B7h and CD40 mRNA expression by CsA and FK506 being greater than that on C3 mRNA expression. Tacrolimus 64-69 CD40 molecule Homo sapiens 151-155 12234299-9 2002 Thirdly, triptolide (4 to 8 ng/mL), CsA (500 to 2500 ng/mL) and FK506 (1250 ng/mL) inhibited up-regulation of CD40 and B7h mRNA, the effect on B7h and CD40 mRNA expression by CsA and FK506 being greater than that on C3 mRNA expression. Tacrolimus 183-188 CD274 molecule Homo sapiens 119-122 12218175-1 2002 Calcineurin, a Ca2+/calmodulin-dependent protein phosphatase, is the common target for two immunophilin-immunosuppressant complexes, cyclophilin A-cyclosporin A (CyPA-CsA) and FKBP-FK506. Tacrolimus 181-186 peptidylprolyl isomerase A Homo sapiens 162-166 12218175-4 2002 The CyPA-CsA complex binds to a composite surface formed by the catalytic and regulatory subunits of CN, where the complex of FK506 and its binding protein FKBP also binds. Tacrolimus 126-131 peptidylprolyl isomerase A Homo sapiens 4-8 12270545-5 2002 Conversely, we show that CsA and FK506 completely inhibit SEA-induced IL-10 protein production and mRNA expression. Tacrolimus 33-38 interleukin 10 Homo sapiens 70-75 12381413-9 2002 Stills others such as the immunosuppressants cyclosporine A and FK506/tacrolimus or erythropoietin have inconsistent effects on eNOS. Tacrolimus 64-69 nitric oxide synthase 3 Homo sapiens 128-132 12381413-9 2002 Stills others such as the immunosuppressants cyclosporine A and FK506/tacrolimus or erythropoietin have inconsistent effects on eNOS. Tacrolimus 70-80 nitric oxide synthase 3 Homo sapiens 128-132 12431607-0 2002 Drug interaction of tacrolimus and proton pump inhibitors in renal transplant recipients with CYP2C19 gene mutation. Tacrolimus 20-30 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 94-101 12135707-4 2002 Preincubation of pNFAT-TA-Luc-transfected cells with cyclosporine A (0.1 microM) and FK506 (0.01 microM) abrogated the gal-1-induced expression of the reporter luciferase (Luc). Tacrolimus 85-90 galectin 1 Homo sapiens 119-124 12502894-2 2002 When the immunosuppressant drug FK506 and nerve growth factor (NGF) were introduced to the PC12m3 cells, the frequency of neurite outgrowth increased approximately 40-fold for NGF alone. Tacrolimus 32-37 nerve growth factor Rattus norvegicus 176-179 12352326-7 2002 At baseline, in cyclosporine-and tacrolimus-treated patients, p22 and TGF-beta mRNA were similarly increased in comparison with normotensive healthy controls (0.90 +/- 0.05 d.u. Tacrolimus 33-43 calcineurin like EF-hand protein 1 Homo sapiens 62-65 12352326-7 2002 At baseline, in cyclosporine-and tacrolimus-treated patients, p22 and TGF-beta mRNA were similarly increased in comparison with normotensive healthy controls (0.90 +/- 0.05 d.u. Tacrolimus 33-43 transforming growth factor beta 1 Homo sapiens 70-78 12352326-9 2002 Endothelial NOS mRNA was increased in cyclosporine-and tacrolimus-treated patients in comparison with controls (0.92 +/- 0.09, 0.96 +/- 0.04, and 0.37 +/- 0.05 respectively; p < 0.001), whereas no difference was found between patients and controls in HO-1 mRNA. Tacrolimus 55-65 nitric oxide synthase 3 Homo sapiens 0-15 12352326-9 2002 Endothelial NOS mRNA was increased in cyclosporine-and tacrolimus-treated patients in comparison with controls (0.92 +/- 0.09, 0.96 +/- 0.04, and 0.37 +/- 0.05 respectively; p < 0.001), whereas no difference was found between patients and controls in HO-1 mRNA. Tacrolimus 55-65 heme oxygenase 1 Homo sapiens 254-258 12389075-7 2002 Unsurprisingly, both cyclosporin A and tacrolimus significantly inhibited IL-2 and IL-2Rbeta at both time points. Tacrolimus 39-49 interleukin 2 Rattus norvegicus 74-78 12389075-7 2002 Unsurprisingly, both cyclosporin A and tacrolimus significantly inhibited IL-2 and IL-2Rbeta at both time points. Tacrolimus 39-49 interleukin 2 receptor subunit beta Rattus norvegicus 83-92 12084723-7 2002 NaCl-induced ENA1 expression was inhibited by EGTA, cch1Delta mutation, and FK506, indicating that the [Ca(2+)](cyt) transient activates calcineurin signaling to mediate ion homeostasis and salt tolerance. Tacrolimus 76-81 Na(+)/Li(+)-exporting P-type ATPase ENA1 Saccharomyces cerevisiae S288C 13-17 12196452-0 2002 A case showing complete insulin independence after severe diabetic ketoacidosis associated with tacrolimus treatment. Tacrolimus 96-106 insulin Homo sapiens 24-31 12198647-1 2002 This report examines the effect of FK506 pretreatment on liver insulin receptor expression in partially (70%) hepatectomized rats. Tacrolimus 35-40 insulin receptor Rattus norvegicus 63-79 12429226-0 2002 FK506 treatment inhibits caspase-3 activation and promotes oligodendroglial survival following traumatic spinal cord injury. Tacrolimus 0-5 caspase 3 Rattus norvegicus 25-34 12429226-1 2002 The focus of this study is to examine the ability of FK506, an immunosuppressant that inhibits calcineurin activation, to limit caspase-3 activation in oligodendroglia following spinal cord injury (SCI). Tacrolimus 53-58 caspase 3 Rattus norvegicus 128-137 12429226-5 2002 However, our findings suggest that treatment with FK506, but not rapamycin reduces the number of oligodendroglia expressing activated caspase-3 and increases the number of surviving oligodendroglia in dorsal white matter. Tacrolimus 50-55 caspase 3 Rattus norvegicus 134-143 12198647-2 2002 FK506 pretreatment led to an increased insulin receptor number 24 hours after hepatectomy, detected by means of insulin binding and cross-linking procedures. Tacrolimus 0-5 insulin receptor Rattus norvegicus 39-55 12198647-5 2002 The results show that FK506 pretreatment elicits an increase in the amount of insulin receptor alpha-subunits as measured by Western blot. Tacrolimus 22-27 insulin receptor Rattus norvegicus 78-94 12198647-7 2002 Moreover, in FK506-pretreated rat hepatocytes, obtained from remnant livers 24 hours after partial hepatectomy (PH), the increase in insulin receptor number was associated with improved sensitivity to the hormone. Tacrolimus 13-18 insulin receptor Rattus norvegicus 133-149 12198647-9 2002 In conclusion, our findings suggest that FK506 pretreatment induces insulin receptor expression in regenerating rat liver and promotes liver regeneration in hepatectomized rats. Tacrolimus 41-46 insulin receptor Rattus norvegicus 68-84 12231370-7 2002 Exposure to tacrolimus and rapamycin caused severe impairment of relaxations to serotonin and a lesser one to bradykinin. Tacrolimus 12-22 kininogen 1 Homo sapiens 110-120 12183690-1 2002 The polyketides FK506 (tacrolimus) and FK520 (ascomycin) are potent immunosuppressants that function by inhibiting calcineurin phosphatase through formation of an FKBP12-FK506/520-calcineurin ternary complex. Tacrolimus 16-21 FKBP prolyl isomerase 1A Rattus norvegicus 163-169 12169765-0 2002 Effect of FK506 on neurotransmitter content and expression of GAP-43 in neurotoxin-lesioned peripheral sensory and sympathetic neurons. Tacrolimus 10-15 growth associated protein 43 Rattus norvegicus 62-68 12183690-3 2002 Based on the crystal structure of the FKBP12-FK506-calcineurin complex, we deduced that the 13- and 15-methoxy groups of FK506 or FK520 are important for inhibition of calcineurin phosphatase but not for binding to FKBP12. Tacrolimus 45-50 FKBP prolyl isomerase 1A Rattus norvegicus 38-44 12183690-3 2002 Based on the crystal structure of the FKBP12-FK506-calcineurin complex, we deduced that the 13- and 15-methoxy groups of FK506 or FK520 are important for inhibition of calcineurin phosphatase but not for binding to FKBP12. Tacrolimus 45-50 FKBP prolyl isomerase 1A Rattus norvegicus 215-221 12183690-3 2002 Based on the crystal structure of the FKBP12-FK506-calcineurin complex, we deduced that the 13- and 15-methoxy groups of FK506 or FK520 are important for inhibition of calcineurin phosphatase but not for binding to FKBP12. Tacrolimus 121-126 FKBP prolyl isomerase 1A Rattus norvegicus 38-44 12183690-3 2002 Based on the crystal structure of the FKBP12-FK506-calcineurin complex, we deduced that the 13- and 15-methoxy groups of FK506 or FK520 are important for inhibition of calcineurin phosphatase but not for binding to FKBP12. Tacrolimus 121-126 FKBP prolyl isomerase 1A Rattus norvegicus 215-221 12183690-1 2002 The polyketides FK506 (tacrolimus) and FK520 (ascomycin) are potent immunosuppressants that function by inhibiting calcineurin phosphatase through formation of an FKBP12-FK506/520-calcineurin ternary complex. Tacrolimus 23-33 FKBP prolyl isomerase 1A Rattus norvegicus 163-169 12169765-9 2002 The expression of a marker protein for growth processes in cells of sympathetic or sensory ganglia, growth-associated protein-43, was significantly increased by the FK506 treatment. Tacrolimus 165-170 growth associated protein 43 Rattus norvegicus 100-128 12352921-0 2002 Neurotoxicity induced by tacrolimus after liver transplantation: relation to genetic polymorphisms of the ABCB1 (MDR1) gene. Tacrolimus 25-35 ATP binding cassette subfamily B member 1 Homo sapiens 106-111 12352921-0 2002 Neurotoxicity induced by tacrolimus after liver transplantation: relation to genetic polymorphisms of the ABCB1 (MDR1) gene. Tacrolimus 25-35 ATP binding cassette subfamily B member 1 Homo sapiens 113-117 12352921-1 2002 BACKGROUND: Tacrolimus is a substrate of P-glycoprotein (PGP) encoded by the multidrug resistant (MDR)1 gene (ABCB1). Tacrolimus 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 41-55 12352921-1 2002 BACKGROUND: Tacrolimus is a substrate of P-glycoprotein (PGP) encoded by the multidrug resistant (MDR)1 gene (ABCB1). Tacrolimus 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 57-60 12352921-1 2002 BACKGROUND: Tacrolimus is a substrate of P-glycoprotein (PGP) encoded by the multidrug resistant (MDR)1 gene (ABCB1). Tacrolimus 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 98-103 12352921-1 2002 BACKGROUND: Tacrolimus is a substrate of P-glycoprotein (PGP) encoded by the multidrug resistant (MDR)1 gene (ABCB1). Tacrolimus 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 110-115 12352921-2 2002 PGP, a multidrug efflux pump, restricts the distribution of tacrolimus in the brain. Tacrolimus 60-70 ATP binding cassette subfamily B member 1 Homo sapiens 0-3 12352921-3 2002 In this study, we investigate the correlation of ABCB1 gene polymorphism with tacrolimus-induced neurotoxicity in patients after liver transplantation. Tacrolimus 78-88 ATP binding cassette subfamily B member 1 Homo sapiens 49-54 12352921-7 2002 CONCLUSION: It is indicated that blood concentrations, liver function, graft weight, and polymorphism in the ABCB1 gene are important factors in tacrolimus-induced neurotoxicity. Tacrolimus 145-155 ATP binding cassette subfamily B member 1 Homo sapiens 109-114 12358793-5 2002 Through the use of two other immunosuppressors, FK506, which also inhibits calcineurin, and rapamycin, which does not, two different mechanisms of choline uptake inhibition were uncovered. Tacrolimus 48-53 protein phosphatase 3, catalytic subunit, alpha isozyme L homeolog Xenopus laevis 75-86 12193059-1 2002 Several studies have demonstrated that endothelin-1 (ET-1) plays an important pathophysiological role in ischaemic renal failure and drug-induced renal injury, such as cyclosporine A (CsA)- and tacrolimus-associated nephrotoxicity. Tacrolimus 194-204 endothelin 1 Homo sapiens 39-51 12193059-1 2002 Several studies have demonstrated that endothelin-1 (ET-1) plays an important pathophysiological role in ischaemic renal failure and drug-induced renal injury, such as cyclosporine A (CsA)- and tacrolimus-associated nephrotoxicity. Tacrolimus 194-204 endothelin 1 Homo sapiens 53-57 12228764-0 2002 Cyclosporin A and FK506 decrease adenosine kinase activity and adenosine uptake in T-lymphocytes. Tacrolimus 18-23 adenosine kinase Homo sapiens 33-49 12228764-4 2002 The immunosuppressive drugs cyclosporin A (CsA) and FK506 inhibited both adenosine uptake and AK activity in a concentration-dependent manner. Tacrolimus 52-57 adenosine kinase Homo sapiens 94-96 12228764-8 2002 These data demonstrate that CsA and FK506 enhance adenosine concentrations in T-lymphocytes by way of a mechanism that involves AK inhibition. Tacrolimus 36-41 adenosine kinase Homo sapiens 128-130 12358793-8 2002 Choline uptake by oocytes expressing tCHT1 was inhibited by all three immunosuppressors and also by microinjection of the specific calcineurin autoinhibitory domain A457-481, indicating that the phosphatase calcineurin regulates CHT1 activity and could be the common target of cyclosporin and FK506. Tacrolimus 293-298 protein phosphatase 3, catalytic subunit, alpha isozyme L homeolog Xenopus laevis 131-142 12358793-8 2002 Choline uptake by oocytes expressing tCHT1 was inhibited by all three immunosuppressors and also by microinjection of the specific calcineurin autoinhibitory domain A457-481, indicating that the phosphatase calcineurin regulates CHT1 activity and could be the common target of cyclosporin and FK506. Tacrolimus 293-298 solute carrier family 5 (sodium/choline cotransporter), member 7 L homeolog Xenopus laevis 38-42 12172213-2 2002 In the present study, we analyzed whether genetic polymorphism of the MDR1 had some influence on the intestinal expression levels of Pgp and CYP3A4 and the tacrolimus concentration/dose ratio over the first postoperative days in recipients of living-donor liver transplantation (LDLT). Tacrolimus 156-166 ATP binding cassette subfamily B member 1 Homo sapiens 70-74 12136004-5 2002 We screened for mutations that confer sensitivity to the calcineurin inhibitor FK506 and to a high concentration of chloride ion and isolated a mutant, cis2-1/myp2-c2, which contains a novel allele of the myp2(+)/myo3(+) gene that encodes a type 2 myosin heavy chain. Tacrolimus 79-84 Myopia, high grade, autosomal dominant 1 Homo sapiens 159-163 12144947-0 2002 Calcineurin inhibitors, cyclosporin A and tacrolimus inhibit expression of inducible nitric oxide synthase in colon epithelial and macrophage cell lines. Tacrolimus 42-52 nitric oxide synthase 2 Homo sapiens 75-106 12144947-4 2002 Cyclosporin A and FK-506 inhibited iNOS expression, and subsequent NO production, in a dose-dependent manner at therapeutically achievable drug concentrations in both cell lines. Tacrolimus 18-24 nitric oxide synthase 2 Homo sapiens 35-39 12144947-9 2002 Two inhibitors of phosphatase calcineurin, cyclosporin A and FK-506, inhibited iNOS expression and NO production in human T84 colon epithelial cells and in murine J774 macrophages by an NF-kappaB independent manner. Tacrolimus 61-67 nitric oxide synthase 2 Homo sapiens 79-83 12135494-7 2002 At low concentrations of FKBP12/FK506, CaN containing the alpha isoform is more sensitive to inhibition than CaN containing the beta isoform using the phosphopeptide substrates. Tacrolimus 32-37 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 25-31 12135494-8 2002 Higher concentrations of FKBP12/FK506 are required for maximal inhibition of beta CaN using PO4-DARPP-32(20-38) as substrate. Tacrolimus 32-37 protein phosphatase 1 regulatory inhibitor subunit 1B Homo sapiens 96-104 12091117-1 2002 BACKGROUND AND OBJECTIVES: FK506 (tacrolimus) is a potent immunosuppressive agent that inhibits interleukin-2 (IL-2) and interferon-g production by CD4+ cells. Tacrolimus 27-32 interleukin 2 Homo sapiens 96-109 12091117-1 2002 BACKGROUND AND OBJECTIVES: FK506 (tacrolimus) is a potent immunosuppressive agent that inhibits interleukin-2 (IL-2) and interferon-g production by CD4+ cells. Tacrolimus 27-32 interleukin 2 Homo sapiens 111-115 12091117-1 2002 BACKGROUND AND OBJECTIVES: FK506 (tacrolimus) is a potent immunosuppressive agent that inhibits interleukin-2 (IL-2) and interferon-g production by CD4+ cells. Tacrolimus 34-44 interleukin 2 Homo sapiens 96-109 12091117-1 2002 BACKGROUND AND OBJECTIVES: FK506 (tacrolimus) is a potent immunosuppressive agent that inhibits interleukin-2 (IL-2) and interferon-g production by CD4+ cells. Tacrolimus 34-44 interleukin 2 Homo sapiens 111-115 12091117-10 2002 We found a decrease in CD1a median fluorescence intensity (MFI) and an increase in percentage of CD86-positive cells with lengthy exposure (6 days) to FK506 at 5000 ng/mL. Tacrolimus 151-156 CD86 molecule Homo sapiens 97-101 12091117-11 2002 In the sequential study, 5000 ng/mL FK506 before LPS addition resulted in a significant decrease in CD1a MFI and in the percentage of cells co-expressing CD83 and CD86. Tacrolimus 36-41 CD1a molecule Homo sapiens 100-104 12091117-11 2002 In the sequential study, 5000 ng/mL FK506 before LPS addition resulted in a significant decrease in CD1a MFI and in the percentage of cells co-expressing CD83 and CD86. Tacrolimus 36-41 CD83 molecule Homo sapiens 154-158 12091117-11 2002 In the sequential study, 5000 ng/mL FK506 before LPS addition resulted in a significant decrease in CD1a MFI and in the percentage of cells co-expressing CD83 and CD86. Tacrolimus 36-41 CD86 molecule Homo sapiens 163-167 12176404-0 2002 Tacrolimus treats ongoing allograft rejection by inhibiting interleukin-10 mediated functional cytotoxic cell infiltration. Tacrolimus 0-10 interleukin 10 Homo sapiens 60-74 12176502-0 2002 Successful results after 3 years" tacrolimus immunosuppression in ABO-incompatible kidney transplantation recipients in Japan. Tacrolimus 34-44 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 66-69 12136004-5 2002 We screened for mutations that confer sensitivity to the calcineurin inhibitor FK506 and to a high concentration of chloride ion and isolated a mutant, cis2-1/myp2-c2, which contains a novel allele of the myp2(+)/myo3(+) gene that encodes a type 2 myosin heavy chain. Tacrolimus 79-84 Myopia, high grade, autosomal dominant 1 Homo sapiens 205-209 12138894-4 2002 FK506, an immunosupressive agent and calcineurin inhibitor, also partially rescued the anti-IgM antibody-induced death of MBC-1 cells. Tacrolimus 0-5 coiled-coil domain containing 112 Homo sapiens 122-127 12100723-9 2002 FK506, a calcineurin inhibitor, and nifedipine, a calcium channel inhibitor, inhibited not only the expression of TNF-alpha and CD40L, but also the up-regulation of CD80 on irradiated A20-HL cells. Tacrolimus 0-5 calcineurin binding protein 1 Mus musculus 9-30 12100723-9 2002 FK506, a calcineurin inhibitor, and nifedipine, a calcium channel inhibitor, inhibited not only the expression of TNF-alpha and CD40L, but also the up-regulation of CD80 on irradiated A20-HL cells. Tacrolimus 0-5 tumor necrosis factor Mus musculus 114-123 12100723-9 2002 FK506, a calcineurin inhibitor, and nifedipine, a calcium channel inhibitor, inhibited not only the expression of TNF-alpha and CD40L, but also the up-regulation of CD80 on irradiated A20-HL cells. Tacrolimus 0-5 CD40 ligand Mus musculus 128-133 12100723-9 2002 FK506, a calcineurin inhibitor, and nifedipine, a calcium channel inhibitor, inhibited not only the expression of TNF-alpha and CD40L, but also the up-regulation of CD80 on irradiated A20-HL cells. Tacrolimus 0-5 CD80 antigen Mus musculus 165-169 12007565-2 2002 Activation of lacZ expression, driven by the GAL4 promoter, has been demonstrated for precedented compound-protein interactions between FK506 and FK506 binding protein 12 (FKBP12) and also between methotrexate (MTX) and dihydrofolate reductase (DHFR). Tacrolimus 136-141 galactose-responsive transcription factor GAL4 Saccharomyces cerevisiae S288C 45-49 12085006-0 2002 Tacrolimus and cyclosporine differ in their capacity to overcome ongoing allograft rejection as a result of their differential abilities to inhibit interleukin-10 production. Tacrolimus 0-10 interleukin 10 Rattus norvegicus 148-162 12032184-2 2002 The aim of this study was to investigate in vitro the possible interaction of tacrolimus and sirolimus with motilin receptors in the rabbit antrum and duodenum. Tacrolimus 78-88 promotilin Oryctolagus cuniculus 108-115 12032184-6 2002 Displacement by tacrolimus of (125)I-nle(13)-porcine motilin bound to its receptor was tested with crude homogenates of the smooth-muscle layer of the rabbit antrum. Tacrolimus 16-26 promotilin Oryctolagus cuniculus 53-60 12032184-13 2002 Tacrolimus weakly displaced motilin bound to its receptor. Tacrolimus 0-10 promotilin Oryctolagus cuniculus 28-35 12032184-16 2002 Tacrolimus did not activate the neural motilin receptor of the rabbit gastric antrum and had low affinity for the smooth-muscle motilin receptor. Tacrolimus 0-10 promotilin Oryctolagus cuniculus 128-135 12042657-5 2002 RESULTS: Lymph nodes from patients with or without CI cyclosporine (CsA) or FK506 (FK) treatment showed comparable CD154 expression, which was present on the cell surface of T cells. Tacrolimus 76-81 CD40 ligand Homo sapiens 115-120 12125507-4 2002 Although the exact mechanism of action of tacrolimus ointment in atopic dermatitis is unknown, tacrolimus is known to inhibit up-regulation of cytokine production following T cell activation and to decrease Fc epsilon RI expression on dendritic antigen-presenting cells in skin. Tacrolimus 95-105 Fc epsilon receptor Ia Homo sapiens 207-220 12085006-11 2002 CONCLUSIONS: Inhibition of IL-10 production is a critical factor in the ability of tacrolimus to reverse ongoing allograft rejection. Tacrolimus 83-93 interleukin 10 Rattus norvegicus 27-32 12085010-0 2002 FKBP12 is the only FK506 binding protein mediating T-cell inhibition by the immunosuppressant FK506. Tacrolimus 19-24 FK506 binding protein 1a Mus musculus 0-6 12085010-0 2002 FKBP12 is the only FK506 binding protein mediating T-cell inhibition by the immunosuppressant FK506. Tacrolimus 94-99 FK506 binding protein 1a Mus musculus 0-6 12085010-7 2002 RESULTS: We found that growth inhibition induced by FK506 is abolished in FKBP12-deficient cells but not in FKBP12.6-deficient cells. Tacrolimus 52-57 FK506 binding protein 1a Mus musculus 74-80 12036304-6 2002 The exon-intron organization of Fkbp10 displays a pattern of repetition that reflects the coding sequence of the four PPIase, or FK506-binding, domains present in the mature protein. Tacrolimus 129-134 FK506 binding protein 10 Mus musculus 32-38 11988144-6 2002 In T lymphocytes, these drugs act by inhibiting the action of calmodulin, a vital enzyme in the activation chain of these cells that ends in the production of interleukin 2 and other proinflammatory cytokines.The accumulated evidence of the various publications seems to indicate that tacrolimus is a safe and effective treatment of atopic dermatitis in patients aged 2 years or more. Tacrolimus 285-295 calmodulin 1 Homo sapiens 62-72 12115224-7 2002 The suppressive action of CSA on VEGF synthesis was calcineurin dependent, as evidenced by a comparable inhibition by FK-506. Tacrolimus 118-124 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 26-29 12007565-2 2002 Activation of lacZ expression, driven by the GAL4 promoter, has been demonstrated for precedented compound-protein interactions between FK506 and FK506 binding protein 12 (FKBP12) and also between methotrexate (MTX) and dihydrofolate reductase (DHFR). Tacrolimus 136-141 dihydrofolate reductase Saccharomyces cerevisiae S288C 245-249 11985515-7 2002 CsA and FK506 even prevented AICD by down-modulation of CD95L. Tacrolimus 8-13 Fas ligand Homo sapiens 56-61 11956176-0 2002 Cyclosporine a and FK506 inhibit transcriptional activity of the human mineralocorticoid receptor: a cell-based model to investigate partial aldosterone resistance in kidney transplantation. Tacrolimus 19-24 nuclear receptor subfamily 3 group C member 2 Homo sapiens 71-97 11956176-3 2002 Upon CsA and FK506 administration, hMR mRNA levels and aldosterone binding in M cells remained unchanged (maximum number of sites, approximately 80 fmol/mg protein; K(d) = approximately 1 nM). Tacrolimus 13-18 mannose receptor C-type 1 Homo sapiens 35-38 11956176-6 2002 In contrast, aldosterone-stimulated hMR transcriptional activity was reduced to 53 +/- 11.2% (P < 0.03) after pretreatment of M cells for 3 d with CsA and to 71 +/- 9.6% (P < 0.05) after pretreatment with FK506. Tacrolimus 211-216 mannose receptor C-type 1 Homo sapiens 36-39 11956176-6 2002 In contrast, aldosterone-stimulated hMR transcriptional activity was reduced to 53 +/- 11.2% (P < 0.03) after pretreatment of M cells for 3 d with CsA and to 71 +/- 9.6% (P < 0.05) after pretreatment with FK506. Tacrolimus 211-216 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 150-153 12030571-7 2002 Tacrolimus and dexamethasone decreased IL-6, iNOS and MCP-1. Tacrolimus 0-10 interleukin 6 Rattus norvegicus 39-43 11969419-3 2002 From the study of glucocorticoid receptor (GR).hsp90.immunophilin complexes in mammalian cells, there is considerable evidence that both hsp90 and the FK506-binding immunophilin FKBP52 play a role in receptor movement from the cytoplasm to the nucleus. Tacrolimus 151-156 nuclear receptor subfamily 3 group C member 1 Homo sapiens 18-41 11969419-3 2002 From the study of glucocorticoid receptor (GR).hsp90.immunophilin complexes in mammalian cells, there is considerable evidence that both hsp90 and the FK506-binding immunophilin FKBP52 play a role in receptor movement from the cytoplasm to the nucleus. Tacrolimus 151-156 nuclear receptor subfamily 3 group C member 1 Homo sapiens 43-45 11969419-3 2002 From the study of glucocorticoid receptor (GR).hsp90.immunophilin complexes in mammalian cells, there is considerable evidence that both hsp90 and the FK506-binding immunophilin FKBP52 play a role in receptor movement from the cytoplasm to the nucleus. Tacrolimus 151-156 heat shock protein 90 alpha family class A member 1 Homo sapiens 47-52 12030571-7 2002 Tacrolimus and dexamethasone decreased IL-6, iNOS and MCP-1. Tacrolimus 0-10 nitric oxide synthase 2 Rattus norvegicus 45-49 11893565-6 2002 This FK-506-induced activation of RyR/Ca(2+) release channels was abolished by pretreatment with anti-FKBP12 antibody. Tacrolimus 5-11 peptidyl-prolyl cis-trans isomerase FKBP1A Bos taurus 102-108 12030571-7 2002 Tacrolimus and dexamethasone decreased IL-6, iNOS and MCP-1. Tacrolimus 0-10 C-C motif chemokine ligand 2 Rattus norvegicus 54-59 11893565-9 2002 Addition of anti-FKBP12 antibody also completely blocked cADPR-induced activation of these channels, and removal of FKBP12.6 by preincubation with FK-506 and subsequent gradient centrifugation abolished cADPR-induced increase in the NP(O) of RyR/Ca(2+) release channels. Tacrolimus 147-153 FKBP prolyl isomerase 1B Bos taurus 116-124 11941319-10 2002 IL-16 expression in LCs in patients with AD correlated with the number of infiltrating CD4(+)cells (r =.72, P =.0017) and was completely downregulated parallel to the clinical response of AD lesions to topical treatment with FK506. Tacrolimus 225-230 interleukin 16 Homo sapiens 0-5 11959794-18 2002 In difference to PBMCs alpha-CD3/alpha-CD28 stimulated IL-13 synthesis was effectively inhibited by CsA, FK-506 and U0126. Tacrolimus 105-111 interleukin 13 Homo sapiens 55-60 11959794-7 2002 Cyclosporine (CsA) and FK-506 inhibited IL-13 synthesis, when cells were stimulated by TPA/ionomycin. Tacrolimus 23-29 interleukin 13 Homo sapiens 40-45 11930095-5 2002 Anti-CD20 therapy was associated with withdrawal of tacrolimus or ciclosporine therapy in all patients. Tacrolimus 52-62 keratin 20 Homo sapiens 5-9 11986604-8 2002 The protective effects of cyclosporine and tacrolimus treatment before reperfusion correlated with 42% and 43% reductions in tissue polymorphonuclear leukocyte (myeloperoxidase) content (P <.008) and marked reductions in bronchoalveolar lavage leukocyte accumulation (P <.01). Tacrolimus 43-53 myeloperoxidase Rattus norvegicus 161-176 11872668-4 2002 Significantly, the actions of both GLP-1 and forskolin were abolished by the selective Ca(2+)/calmodulin-dependent phosphatase 2B (calcineurin) inhibitor, FK506, as well as by the chelation of intracellular Ca(2+) by BAPTA (bis(2-aminophenoxy)ethane-N,N,N",N"-tetraacetate). Tacrolimus 155-160 glucagon Rattus norvegicus 35-40 11918741-5 2002 RESULTS: FK506 treatment with 1.0 and 0.3 mg/kg body weight (BW) daily from day 1 to day 4 significantly reduced the glomerular expression of mRNA for interferon-gamma (IFN-gamma; 1.0 mg/kg BW FK506, 32.4% to the placebo group, P < 0.01) and IL-2 (55.6%, P < 0.01) on day 5. Tacrolimus 9-14 interferon gamma Rattus norvegicus 151-167 11918741-5 2002 RESULTS: FK506 treatment with 1.0 and 0.3 mg/kg body weight (BW) daily from day 1 to day 4 significantly reduced the glomerular expression of mRNA for interferon-gamma (IFN-gamma; 1.0 mg/kg BW FK506, 32.4% to the placebo group, P < 0.01) and IL-2 (55.6%, P < 0.01) on day 5. Tacrolimus 9-14 interferon gamma Rattus norvegicus 169-178 11918741-5 2002 RESULTS: FK506 treatment with 1.0 and 0.3 mg/kg body weight (BW) daily from day 1 to day 4 significantly reduced the glomerular expression of mRNA for interferon-gamma (IFN-gamma; 1.0 mg/kg BW FK506, 32.4% to the placebo group, P < 0.01) and IL-2 (55.6%, P < 0.01) on day 5. Tacrolimus 9-14 interleukin 2 Rattus norvegicus 245-249 11918741-5 2002 RESULTS: FK506 treatment with 1.0 and 0.3 mg/kg body weight (BW) daily from day 1 to day 4 significantly reduced the glomerular expression of mRNA for interferon-gamma (IFN-gamma; 1.0 mg/kg BW FK506, 32.4% to the placebo group, P < 0.01) and IL-2 (55.6%, P < 0.01) on day 5. Tacrolimus 193-198 interferon gamma Rattus norvegicus 151-167 11918741-5 2002 RESULTS: FK506 treatment with 1.0 and 0.3 mg/kg body weight (BW) daily from day 1 to day 4 significantly reduced the glomerular expression of mRNA for interferon-gamma (IFN-gamma; 1.0 mg/kg BW FK506, 32.4% to the placebo group, P < 0.01) and IL-2 (55.6%, P < 0.01) on day 5. Tacrolimus 193-198 interferon gamma Rattus norvegicus 169-178 11918741-7 2002 Although no side effects were detected in rats with 0.3 mg/kg BW of FK506 treatment from day +1, the treatment also ameliorated proteinuria (day 14, 3.7 +/- 0.89 vs. 19.8 +/- 12.3 mg/100 g BW/day P < 0.05) and glomerular alterations [total cell number, 63.1 +/- 3.1 vs. 80.2 +/- 7.4, P < 0.01; matrix expansion, 0.90 +/- 0.30 vs. 1.34 +/- 0.27, P < 0.05; alpha-smooth muscle actin (alphaSMA) expression; 1.20 +/- 0.12 vs. 1.96 +/- 0.29, P < 0.01] on day 14. Tacrolimus 68-73 actin gamma 2, smooth muscle Rattus norvegicus 364-389 11918741-7 2002 Although no side effects were detected in rats with 0.3 mg/kg BW of FK506 treatment from day +1, the treatment also ameliorated proteinuria (day 14, 3.7 +/- 0.89 vs. 19.8 +/- 12.3 mg/100 g BW/day P < 0.05) and glomerular alterations [total cell number, 63.1 +/- 3.1 vs. 80.2 +/- 7.4, P < 0.01; matrix expansion, 0.90 +/- 0.30 vs. 1.34 +/- 0.27, P < 0.05; alpha-smooth muscle actin (alphaSMA) expression; 1.20 +/- 0.12 vs. 1.96 +/- 0.29, P < 0.01] on day 14. Tacrolimus 68-73 actin gamma 2, smooth muscle Rattus norvegicus 391-399 11923712-0 2002 Tacrolimus-induced nephrotoxicity unmasked by induction of the CYP3A4 system with St John"s wort. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 63-69 11907581-1 2002 FK506 binding proteins 12 and 12.6 (FKBP12 and FKBP12.6) are intracellular receptors for the immunosuppressant drug FK506 (ref. Tacrolimus 0-5 FK506 binding protein 1a Mus musculus 36-42 11907581-1 2002 FK506 binding proteins 12 and 12.6 (FKBP12 and FKBP12.6) are intracellular receptors for the immunosuppressant drug FK506 (ref. Tacrolimus 0-5 FK506 binding protein 1b Mus musculus 47-55 11907581-1 2002 FK506 binding proteins 12 and 12.6 (FKBP12 and FKBP12.6) are intracellular receptors for the immunosuppressant drug FK506 (ref. Tacrolimus 116-121 FK506 binding protein 1a Mus musculus 36-42 11907581-1 2002 FK506 binding proteins 12 and 12.6 (FKBP12 and FKBP12.6) are intracellular receptors for the immunosuppressant drug FK506 (ref. Tacrolimus 116-121 FK506 binding protein 1b Mus musculus 47-55 12137728-7 2002 Experimental evidence suggests that tacrolimus inhibits T-lymphocytes activation by binding to an intracellular protein, FKBP-12. Tacrolimus 36-46 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 121-128 11897999-10 2002 NFAT2 protein accumulated in the nuclei of basophils activated for 1 hour with anti-IgE, and this was inhibited with the addition of FK506. Tacrolimus 133-138 nuclear factor of activated T cells 1 Homo sapiens 0-5 11890899-1 2002 FK506 (Tacrolimus) and cyclosporin A exert their immunosuppressive effects via a common mechanism, calcineurin inhibition, after binding to intracellular proteins termed immunophilins: FK506-binding protein (FKBP) and cyclophilin. Tacrolimus 0-5 FK506 binding protein 1a Mus musculus 185-206 11865967-1 2002 Cyclosporine and tacrolimus are substrates and potent inhibitors of the multidrug transporter, P-glycoprotein, in vitro. Tacrolimus 17-27 ATP binding cassette subfamily B member 1 Homo sapiens 95-109 11865967-6 2002 However, patients who were started on tacrolimus or cyclosporine had an initial decline in expression of P-gp on CD4 T cells. Tacrolimus 38-48 phosphoglycolate phosphatase Homo sapiens 105-109 11865967-6 2002 However, patients who were started on tacrolimus or cyclosporine had an initial decline in expression of P-gp on CD4 T cells. Tacrolimus 38-48 CD4 molecule Homo sapiens 113-116 11987039-4 2002 The mode of action of Fk506 is similar to that of cyclosporin A, i.e. it exerts an inhibitory effect on transcription of interleukin 2 in T lymphocytes. Tacrolimus 22-27 interleukin 2 Homo sapiens 121-134 11920757-0 2002 Tacrolimus is a class II low-solubility high-permeability drug: the effect of P-glycoprotein efflux on regional permeability of tacrolimus in rats. Tacrolimus 0-10 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 78-92 11920757-0 2002 Tacrolimus is a class II low-solubility high-permeability drug: the effect of P-glycoprotein efflux on regional permeability of tacrolimus in rats. Tacrolimus 128-138 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 78-92 11920757-1 2002 The objective of this study is to investigate the role of P-glycoprotein (P-gp), a membrane efflux pump associated with multidrug resistance (MDR) and a known substrate for tacrolimus, in determining the regional intestinal permeability of tacrolimus in rats. Tacrolimus 173-183 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 58-72 11920757-1 2002 The objective of this study is to investigate the role of P-glycoprotein (P-gp), a membrane efflux pump associated with multidrug resistance (MDR) and a known substrate for tacrolimus, in determining the regional intestinal permeability of tacrolimus in rats. Tacrolimus 173-183 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 74-78 11920757-1 2002 The objective of this study is to investigate the role of P-glycoprotein (P-gp), a membrane efflux pump associated with multidrug resistance (MDR) and a known substrate for tacrolimus, in determining the regional intestinal permeability of tacrolimus in rats. Tacrolimus 240-250 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 58-72 11920757-1 2002 The objective of this study is to investigate the role of P-glycoprotein (P-gp), a membrane efflux pump associated with multidrug resistance (MDR) and a known substrate for tacrolimus, in determining the regional intestinal permeability of tacrolimus in rats. Tacrolimus 240-250 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 74-78 11920757-5 2002 The results suggest that systemic absorption of tacrolimus from the gastrointestinal tract could be significantly affected by P-gp efflux mechanisms. Tacrolimus 48-58 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 126-130 11920757-6 2002 It is also possible that differences in P-gp function at various intestinal sites in a subject or at a given intestinal site in various subjects could lead to large intra- and interindividual variability in bioavailability of tacrolimus following oral administration. Tacrolimus 226-236 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 40-44 11890899-1 2002 FK506 (Tacrolimus) and cyclosporin A exert their immunosuppressive effects via a common mechanism, calcineurin inhibition, after binding to intracellular proteins termed immunophilins: FK506-binding protein (FKBP) and cyclophilin. Tacrolimus 0-5 FK506 binding protein 1a Mus musculus 208-212 11890899-1 2002 FK506 (Tacrolimus) and cyclosporin A exert their immunosuppressive effects via a common mechanism, calcineurin inhibition, after binding to intracellular proteins termed immunophilins: FK506-binding protein (FKBP) and cyclophilin. Tacrolimus 7-17 FK506 binding protein 1a Mus musculus 185-206 11890899-1 2002 FK506 (Tacrolimus) and cyclosporin A exert their immunosuppressive effects via a common mechanism, calcineurin inhibition, after binding to intracellular proteins termed immunophilins: FK506-binding protein (FKBP) and cyclophilin. Tacrolimus 7-17 FK506 binding protein 1a Mus musculus 208-212 11890899-4 2002 Rapamycin, an immunosuppressant that binds to FKBP, antagonized the effect of FK506. Tacrolimus 78-83 FK506 binding protein 1a Mus musculus 46-50 11890899-6 2002 Taken together, these results suggest that FK506 induces chondrogenic differentiation of ATDC5 cells via a calcineurin-independent mechanism, after binding to FKBP. Tacrolimus 43-48 FK506 binding protein 1a Mus musculus 159-163 11852053-4 2002 The RyR activators caffeine, FK506, ryanodine and 4-chloro-m-cresol mobilized Ca(2+) in DC, and responses to 4-chloro-m-cresol were inhibited by dantrolene. Tacrolimus 29-34 ryanodine receptor 1, skeletal muscle Mus musculus 4-7 11931080-14 2002 CONCLUSION: Topical FK506 treatment suppressed EC in BN rats, possibly by inhibition of IL-4 in the conjunctiva. Tacrolimus 20-25 interleukin 4 Rattus norvegicus 88-92 11935154-9 2002 Islet grafts from sirolimus plus tacrolimus-treated mice expressed significantly decreased mRNA contents of Th1-type cytokines (IFN- gamma and IL-2) and the highest ratio of TGF- beta1/IFN- gamma mRNA. Tacrolimus 33-43 negative elongation factor complex member C/D, Th1l Mus musculus 108-111 11935154-9 2002 Islet grafts from sirolimus plus tacrolimus-treated mice expressed significantly decreased mRNA contents of Th1-type cytokines (IFN- gamma and IL-2) and the highest ratio of TGF- beta1/IFN- gamma mRNA. Tacrolimus 33-43 interferon gamma Mus musculus 128-138 11935154-9 2002 Islet grafts from sirolimus plus tacrolimus-treated mice expressed significantly decreased mRNA contents of Th1-type cytokines (IFN- gamma and IL-2) and the highest ratio of TGF- beta1/IFN- gamma mRNA. Tacrolimus 33-43 interleukin 2 Mus musculus 143-147 11935154-9 2002 Islet grafts from sirolimus plus tacrolimus-treated mice expressed significantly decreased mRNA contents of Th1-type cytokines (IFN- gamma and IL-2) and the highest ratio of TGF- beta1/IFN- gamma mRNA. Tacrolimus 33-43 transforming growth factor, beta 1 Mus musculus 174-177 11935154-9 2002 Islet grafts from sirolimus plus tacrolimus-treated mice expressed significantly decreased mRNA contents of Th1-type cytokines (IFN- gamma and IL-2) and the highest ratio of TGF- beta1/IFN- gamma mRNA. Tacrolimus 33-43 hemoglobin, beta adult major chain Mus musculus 179-195 11935154-10 2002 CONCLUSION/INTERPRETATION: These findings suggest that combination therapy with sirolimus and tacrolimus prevent autoimmune beta-cell destruction by upregulating expression of the immunoregulatory cytokine, TGF- beta1 and reducing Th1 cytokines (IFN- gamma and IL-2) expressed in the islets. Tacrolimus 94-104 transforming growth factor, beta 1 Mus musculus 207-217 11935154-10 2002 CONCLUSION/INTERPRETATION: These findings suggest that combination therapy with sirolimus and tacrolimus prevent autoimmune beta-cell destruction by upregulating expression of the immunoregulatory cytokine, TGF- beta1 and reducing Th1 cytokines (IFN- gamma and IL-2) expressed in the islets. Tacrolimus 94-104 negative elongation factor complex member C/D, Th1l Mus musculus 231-234 11935154-10 2002 CONCLUSION/INTERPRETATION: These findings suggest that combination therapy with sirolimus and tacrolimus prevent autoimmune beta-cell destruction by upregulating expression of the immunoregulatory cytokine, TGF- beta1 and reducing Th1 cytokines (IFN- gamma and IL-2) expressed in the islets. Tacrolimus 94-104 interferon gamma Mus musculus 246-256 11935154-10 2002 CONCLUSION/INTERPRETATION: These findings suggest that combination therapy with sirolimus and tacrolimus prevent autoimmune beta-cell destruction by upregulating expression of the immunoregulatory cytokine, TGF- beta1 and reducing Th1 cytokines (IFN- gamma and IL-2) expressed in the islets. Tacrolimus 94-104 interleukin 2 Mus musculus 261-265 11896462-4 2002 In this approach, the Akt PH domain has been replaced with the rapamycin (and FK506)-binding domain, FKBP12, to make F3-DeltaPH.Akt. Tacrolimus 78-83 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 101-107 11896462-4 2002 In this approach, the Akt PH domain has been replaced with the rapamycin (and FK506)-binding domain, FKBP12, to make F3-DeltaPH.Akt. Tacrolimus 78-83 AKT serine/threonine kinase 1 Homo sapiens 128-131 11884930-7 2002 Insulin secretion from Wistar rat islets was reduced by 0% and 48% after exposure to mycophenolate (P<0.001) and 20% and 31% after exposure to tacrolimus (P<0.05). Tacrolimus 146-156 insulin Mesocricetus auratus 0-7 11694517-1 2002 The immunosuppressive agents cyclosporin A (CsA) and tacrolimus (FK506) bind to unrelated intracellular immunophilin receptors, cyclophilin (CyP) and FK506-binding protein (FKBP), respectively. Tacrolimus 53-63 peptidylprolyl isomerase G Homo sapiens 128-139 11694517-1 2002 The immunosuppressive agents cyclosporin A (CsA) and tacrolimus (FK506) bind to unrelated intracellular immunophilin receptors, cyclophilin (CyP) and FK506-binding protein (FKBP), respectively. Tacrolimus 53-63 peptidylprolyl isomerase G Homo sapiens 141-144 11694517-1 2002 The immunosuppressive agents cyclosporin A (CsA) and tacrolimus (FK506) bind to unrelated intracellular immunophilin receptors, cyclophilin (CyP) and FK506-binding protein (FKBP), respectively. Tacrolimus 65-70 peptidylprolyl isomerase G Homo sapiens 128-139 11694517-1 2002 The immunosuppressive agents cyclosporin A (CsA) and tacrolimus (FK506) bind to unrelated intracellular immunophilin receptors, cyclophilin (CyP) and FK506-binding protein (FKBP), respectively. Tacrolimus 65-70 peptidylprolyl isomerase G Homo sapiens 141-144 11807435-4 2002 Pimecrolimus (ASM 981), a newer calcineurin inhibitor closely related to tacrolimus, is also being developed for atopic dermatitis therapy. Tacrolimus 73-83 calcineurin binding protein 1 Homo sapiens 32-53 11849412-16 2002 In particular, the use of cyclosporine compared to tacrolimus was associated with a tenfold increase in TGF-beta mRNA (TGF-beta mRNA at 6 months, CsA vs. Tac, 3 +/- 0.3 vs. 2 +/- 0.3 log copies, P < 0.05), interstitial fibrosis (Vvi at 6 months, CsA vs. Tac, 33 +/- 4% vs. 24 +/- 2%, P < 0.05). Tacrolimus 51-61 transforming growth factor beta 1 Homo sapiens 104-112 11849412-16 2002 In particular, the use of cyclosporine compared to tacrolimus was associated with a tenfold increase in TGF-beta mRNA (TGF-beta mRNA at 6 months, CsA vs. Tac, 3 +/- 0.3 vs. 2 +/- 0.3 log copies, P < 0.05), interstitial fibrosis (Vvi at 6 months, CsA vs. Tac, 33 +/- 4% vs. 24 +/- 2%, P < 0.05). Tacrolimus 51-61 transforming growth factor beta 1 Homo sapiens 119-127 11805719-1 2002 The intracellular class of proteins that bind the interleukin-2 suppressing drugs (cyclosporin, tacrolimus, and sirolimus) are called immunophilins. Tacrolimus 96-106 interleukin 2 Homo sapiens 50-63 11772413-6 2002 The effects of FKBP12 were reversed by FK506. Tacrolimus 39-44 FKBP prolyl isomerase 1A Rattus norvegicus 15-21 11792863-9 2002 The inhibition by rapamycin was blocked by FK506, which competitively inhibits those effects of rapamycin that are mediated by inhibition of mTOR. Tacrolimus 43-48 mechanistic target of rapamycin kinase Homo sapiens 141-145 11848304-5 2002 Tacrolimus enhanced renal glutathione reductase (GSH-R) activities and glutathione (GSH) and depressed catalase (CAT) activities. Tacrolimus 0-10 glutathione-disulfide reductase Rattus norvegicus 26-47 11848304-5 2002 Tacrolimus enhanced renal glutathione reductase (GSH-R) activities and glutathione (GSH) and depressed catalase (CAT) activities. Tacrolimus 0-10 catalase Rattus norvegicus 113-116 11848304-9 2002 These findings suggest that FTS is useful for the prevention of tacrolimus-induced nephrotoxicity, and that the increase of renal CAT activity in the defense mechanism of FTS might be important for cell protection against active oxygen species. Tacrolimus 64-74 AKT interacting protein Rattus norvegicus 28-31 12372041-1 2002 Tacrolimus inhibits lymphocyte responses by blocking calcium-dependent signalling pathways important in IL-2 generation. Tacrolimus 0-10 interleukin 2 Homo sapiens 104-108 14720063-5 2002 Tacrolimus, a new calcineurin inhibitor, is increasingly employed as the primary immunosuppressive agent in lung transplant recipients. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 18-39 11841938-1 2002 We used olefin metathesis to synthesize C40 derivatives of FK506 and measured their ability, when complexed to FKBP12, to inhibit calcineurin"s phosphatase activity. Tacrolimus 59-64 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 111-117 12142035-3 2002 The calcineurin inhibitor FK506 at 1nM inhibited the development of thymocytes to either lineage, but 0.3nM FK506 significantly switched the development from the CD4 cell fate to the CD8 cell fate. Tacrolimus 108-113 CD4 molecule Homo sapiens 162-165 12142035-3 2002 The calcineurin inhibitor FK506 at 1nM inhibited the development of thymocytes to either lineage, but 0.3nM FK506 significantly switched the development from the CD4 cell fate to the CD8 cell fate. Tacrolimus 108-113 CD8a molecule Homo sapiens 183-186 12190331-8 2002 Interaction of other drugs with P-glycoprotein may change tacrolimus tissue distribution and modify its toxicity and immunosuppressive activity. Tacrolimus 58-68 ATP binding cassette subfamily B member 1 Homo sapiens 32-46 12190331-11 2002 In the near future, progress can be expected from studies evaluating potential pharmacokinetic interactions caused by herbal preparations and food components, the exact biochemical mechanism underlying tacrolimus toxicity, and the potential of inhibition of CYP3A and P-glycoprotein to improve oral bioavailability and to decrease intraindividual variability of tacrolimus pharmacokinetics. Tacrolimus 362-372 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 258-263 11779223-7 2002 FK506 was found to not only stimulate osteoblastic differentiation, but also to enhance BMP-4 induced osteoblastic differentiation. Tacrolimus 0-5 bone morphogenetic protein 4 Rattus norvegicus 88-93 12190331-2 2002 As a substrate of cytochrome P450 (CYP) 3A enzymes and P-glycoprotein, tacrolimus interacts with several other drugs used in transplantation medicine, which also are known CYP3A and/or P-glycoprotein inhibitors and/or inducers. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 18-42 12190331-2 2002 As a substrate of cytochrome P450 (CYP) 3A enzymes and P-glycoprotein, tacrolimus interacts with several other drugs used in transplantation medicine, which also are known CYP3A and/or P-glycoprotein inhibitors and/or inducers. Tacrolimus 71-81 ATP binding cassette subfamily B member 1 Homo sapiens 55-69 12190331-2 2002 As a substrate of cytochrome P450 (CYP) 3A enzymes and P-glycoprotein, tacrolimus interacts with several other drugs used in transplantation medicine, which also are known CYP3A and/or P-glycoprotein inhibitors and/or inducers. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 172-177 12190331-2 2002 As a substrate of cytochrome P450 (CYP) 3A enzymes and P-glycoprotein, tacrolimus interacts with several other drugs used in transplantation medicine, which also are known CYP3A and/or P-glycoprotein inhibitors and/or inducers. Tacrolimus 71-81 ATP binding cassette subfamily B member 1 Homo sapiens 185-199 12190331-3 2002 In clinical studies, CYP3A/P-glycoprotein inhibitors and inducers primarily affect oral bioavailability of tacrolimus rather than its clearance, indicating a key role of intestinal P-glycoprotein and CYP3A. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-26 12190331-3 2002 In clinical studies, CYP3A/P-glycoprotein inhibitors and inducers primarily affect oral bioavailability of tacrolimus rather than its clearance, indicating a key role of intestinal P-glycoprotein and CYP3A. Tacrolimus 107-117 ATP binding cassette subfamily B member 1 Homo sapiens 27-41 12190331-3 2002 In clinical studies, CYP3A/P-glycoprotein inhibitors and inducers primarily affect oral bioavailability of tacrolimus rather than its clearance, indicating a key role of intestinal P-glycoprotein and CYP3A. Tacrolimus 107-117 ATP binding cassette subfamily B member 1 Homo sapiens 181-195 12190331-3 2002 In clinical studies, CYP3A/P-glycoprotein inhibitors and inducers primarily affect oral bioavailability of tacrolimus rather than its clearance, indicating a key role of intestinal P-glycoprotein and CYP3A. Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 200-205 12190331-7 2002 P-glycoprotein regulates distribution of tacrolimus through the blood-brain barrier into the brain as well as distribution into lymphocytes. Tacrolimus 41-51 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 12372041-3 2002 We reasoned therefore that the absence of IL-2R should permit lower doses of tacrolimus and thereby less toxicity. Tacrolimus 77-87 interleukin 2 receptor subunit alpha Homo sapiens 42-47 12167066-4 2002 Oral bioavailability of tacrolimus can be increased by concomitant administration of inhibitors of either CYP3A or P-glycoprotein. Tacrolimus 24-34 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 106-111 12167066-4 2002 Oral bioavailability of tacrolimus can be increased by concomitant administration of inhibitors of either CYP3A or P-glycoprotein. Tacrolimus 24-34 ATP binding cassette subfamily B member 1 Homo sapiens 115-129 12372972-3 2002 We here present a case of severe alloimmune hemolytic anemia due to ABO minor incompatibility after renal transplantation in a patient treated with tacrolimus. Tacrolimus 148-158 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 68-71 11752040-10 2002 The only significant difference between patients who received tacrolimus and those who received cyclosporine was in pancreatic secretion capacity at week 3 after transplantation, when the increment of C-peptide secretion was 57% lower and the increment of insulin secretion was 48% lower for patients receiving tacrolimus. Tacrolimus 62-72 insulin Homo sapiens 201-210 12497749-1 2002 AIM: Hepatic metabolism of sildenafil uses the same metabolic pathway as the calcineurin inhibitors (cyclosporine/tacrolimus), through the CYP3A4 isoenzyme. Tacrolimus 114-124 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 139-145 11812963-11 2002 Tacrolimus (100 microg/kg, administered 2 minutes after bleeding was discontinued) inhibited the loss of IkappaBalpha protein from the cytoplasm and prevented NF-kappaB binding activity in the nucleus. Tacrolimus 0-10 NFKB inhibitor alpha Rattus norvegicus 105-117 11808341-7 2002 In addition, the intestinal P-glycoprotein was suggested to act as an absorptive barrier for tacrolimus in recipients of liver and small bowel transplantation. Tacrolimus 93-103 ATP binding cassette subfamily B member 1 Homo sapiens 28-42 11812963-12 2002 Moreover, tacrolimus increased survival time (118 +/- 7 minutes; P <.01) and survival rate (vehicle = 0 and tacrolimus = 90% 240 minutes after bleeding was discontinued), reverted the marked hypotension, decreased liver messenger RNA for TNF-alpha reduced plasma TNF-alpha (35 +/- 6 pg/mL), and restored the hyporeactivity to phenylephrine to control values. Tacrolimus 10-20 tumor necrosis factor Rattus norvegicus 241-250 11812963-12 2002 Moreover, tacrolimus increased survival time (118 +/- 7 minutes; P <.01) and survival rate (vehicle = 0 and tacrolimus = 90% 240 minutes after bleeding was discontinued), reverted the marked hypotension, decreased liver messenger RNA for TNF-alpha reduced plasma TNF-alpha (35 +/- 6 pg/mL), and restored the hyporeactivity to phenylephrine to control values. Tacrolimus 10-20 tumor necrosis factor Rattus norvegicus 266-275 11744119-6 2001 Using immunofluorescent laser scanning confocal microscopy we demonstrate that in human fetal brain cultures FK506 significantly increases cell numbers, including neurons, and the expression of the neuronal marker MAP-2. Tacrolimus 109-114 microtubule associated protein 2 Homo sapiens 214-219 11744119-8 2001 Interestingly, in combination with brain-derived neurotrophic factor, FK506 induces a prominent decrease in glial fibrillary acidic protein expression, which indicates an inhibitory effect on astrogliosis in vitro. Tacrolimus 70-75 brain derived neurotrophic factor Homo sapiens 35-68 11740384-5 2001 were used to induce ischemic tolerance in Sprague-Dawley rats, and the induction of heat shock protein (hsp) 70 by CsA or FK506 was evaluated overtime. Tacrolimus 122-127 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 84-111 11718743-8 2001 The FK506-treated group stained more weakly for ICAM-1 than the untreated EAM group. Tacrolimus 4-9 intercellular adhesion molecule 1 Mus musculus 48-54 11762554-1 2001 Immunosuppressive agents such as cyclosporine, tacrolimus, sirolimus, and corticosteroids are substrates for the transmembrane multidrug resistance pump P-glycoprotein (P-gp). Tacrolimus 47-57 ATP binding cassette subfamily B member 1 Homo sapiens 153-167 11773940-5 2001 Treatment with calcineurin inhibitor FK506 (0.5 or 1.0 mg/kg/day) from the age of four to eight weeks attenuated cardiac hypertrophy without beneficially affecting cardiac function. Tacrolimus 37-42 calcineurin binding protein 1 Mus musculus 15-36 11740384-6 2001 Rats were pretreated with CsA or FK506 6 hr before I/R injury when hsp70 was maximally expressed, and were killed 24 hr later. Tacrolimus 33-38 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 67-72 11740384-11 2001 CONCLUSIONS: Pretreatment with low-dose CsA or FK506 prevents subsequent I/R injury, and this effect may be related to the induction of hsp70. Tacrolimus 47-52 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 136-141 11743719-10 2001 dao-1, dao-8 and dao-9 are homologs of the FK506 binding proteins that interact with the mammalian insulin pathway. Tacrolimus 43-48 amine oxidase copper containing 1 Homo sapiens 0-5 11743719-10 2001 dao-1, dao-8 and dao-9 are homologs of the FK506 binding proteins that interact with the mammalian insulin pathway. Tacrolimus 43-48 insulin Homo sapiens 99-106 11762554-1 2001 Immunosuppressive agents such as cyclosporine, tacrolimus, sirolimus, and corticosteroids are substrates for the transmembrane multidrug resistance pump P-glycoprotein (P-gp). Tacrolimus 47-57 ATP binding cassette subfamily B member 1 Homo sapiens 169-173 11733621-1 2001 BACKGROUND: Endothelin-1 (ET-1) is a potent vasoconstrictive peptide which plays an important pathophysiological role in ischaemic renal failure and drug-induced renal injury such as cyclosporin A (CsA)- and tacrolimus-associated nephrotoxicity. Tacrolimus 208-218 endothelin 1 Homo sapiens 12-24 11733621-1 2001 BACKGROUND: Endothelin-1 (ET-1) is a potent vasoconstrictive peptide which plays an important pathophysiological role in ischaemic renal failure and drug-induced renal injury such as cyclosporin A (CsA)- and tacrolimus-associated nephrotoxicity. Tacrolimus 208-218 endothelin 1 Homo sapiens 26-30 11673895-3 2001 The fusion protein was immobilized in avidin-coated multiwell plates, and varying concentrations of test ligands were allowed to compete with [3H]FK506 for FKBP12 sites on the plate. Tacrolimus 146-151 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 156-162 11844153-5 2001 Finally, potential therapeutic strategies are discussed to modify NF-kappaB activity with certain immunosuppression medications, including cyclosporine, tacrolimus, and glucocorticoids. Tacrolimus 153-163 nuclear factor kappa B subunit 1 Homo sapiens 66-75 11696436-7 2001 Lptn mRNA was found to be up-regulated on stimulation with phorbol-12-myristate-13-acetate and concanavalin A in T cells isolated from peripheral blood, which could be prevented by dexamethasone, cyclosporine A, and FK506. Tacrolimus 216-221 X-C motif chemokine ligand 1 Homo sapiens 0-4 11606756-3 2001 Here, we report that the up-regulation of slow myosin heavy chain (MyHC) and a MyHC-slow promoter induced by slow motor neurons in regenerating rat soleus muscle is prevented by the calcineurin inhibitors cyclosporin A (CsA), FK506, and the calcineurin inhibitory protein domain from cain/cabin-1. Tacrolimus 226-231 myosin heavy chain 13 Rattus norvegicus 67-71 11606756-3 2001 Here, we report that the up-regulation of slow myosin heavy chain (MyHC) and a MyHC-slow promoter induced by slow motor neurons in regenerating rat soleus muscle is prevented by the calcineurin inhibitors cyclosporin A (CsA), FK506, and the calcineurin inhibitory protein domain from cain/cabin-1. Tacrolimus 226-231 myosin heavy chain 13 Rattus norvegicus 79-83 11606756-6 2001 Calcineurin is also involved in the maintenance of the slow muscle fiber gene program because in the adult soleus muscle, cain causes a switch from MyHC-slow to fast-type MyHC-2X and MyHC-2B gene expression, and the activity of the MyHC-slow promoter is inhibited by CsA and FK506. Tacrolimus 275-280 calcineurin binding protein 1 Rattus norvegicus 122-126 11703081-0 2001 Overexpression of the FK506-binding immunophilin FKBP51 is the common cause of glucocorticoid resistance in three New World primates. Tacrolimus 22-27 FK506 binding protein 5 Mus musculus 49-55 11719731-1 2001 BACKGROUND: Interindividual variation in the pharmacokinetics of the immunosuppressive agents cyclosporine (INN, ciclosporin) and tacrolimus may result from differences in the activity of cytochrome P4503A (CYP3A). Tacrolimus 130-140 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 188-194 11719731-1 2001 BACKGROUND: Interindividual variation in the pharmacokinetics of the immunosuppressive agents cyclosporine (INN, ciclosporin) and tacrolimus may result from differences in the activity of cytochrome P4503A (CYP3A). Tacrolimus 130-140 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 207-212 11583721-6 2001 In the CsA group, there were significant increases in cholesterol contents in very-low-density lipoprotein (VLDL), LDL2 and HDL2 fractions, whereas, in the tacrolimus group, cholesterol content was increased in VLDL and HDL2 fractions. Tacrolimus 156-166 junctophilin 3 Homo sapiens 124-128 11514552-8 2001 Addition of FK506 prior to immunoprecipitation resulted in a temperature-dependent dissociation of dFKBP59 and TRPL. Tacrolimus 12-17 FK506-binding protein 59kD Drosophila melanogaster 99-106 11514552-8 2001 Addition of FK506 prior to immunoprecipitation resulted in a temperature-dependent dissociation of dFKBP59 and TRPL. Tacrolimus 12-17 transient receptor potential-like Drosophila melanogaster 111-115 11606920-8 2001 OBJECTIVE: Based on this clinical observation and the observation that rapamycin, a chemically similar compound to tacrolimus, is known to inhibit signaling from the gli-1 oncogene, we examined keloids and scars for expression of Gli-1 protein. Tacrolimus 115-125 GLI family zinc finger 1 Homo sapiens 166-171 11583721-6 2001 In the CsA group, there were significant increases in cholesterol contents in very-low-density lipoprotein (VLDL), LDL2 and HDL2 fractions, whereas, in the tacrolimus group, cholesterol content was increased in VLDL and HDL2 fractions. Tacrolimus 156-166 junctophilin 3 Homo sapiens 220-224 11758940-7 2001 Some physicochemical data suggested that the Pdr5-specific inhibitor was different from an immunosuppressant, FK506, reported as the only inhibitor of Pdr5 in vivo. Tacrolimus 110-115 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 151-155 11516757-5 2001 Immunosuppressants acting via inhibition of interleukin-2 synthesis, such as CSA or FK506, inhibited the production of GFP in a dose-dependent manner. Tacrolimus 84-89 interleukin 2 Homo sapiens 44-57 11680570-3 2001 In this study, we performed a combination spleen/pancreas transplantation using high-dose tacrolimus in a high-responder rat combination of DA (RT1a) to LEW (RT1) and induced permanent survival in the few recipient rats. Tacrolimus 90-100 RT1 class I, locus A Rattus norvegicus 144-148 11605779-1 2001 Like cyclosporine (CsA), tacrolimus acts through the inhibition of renal phosphatase calcineurin. Tacrolimus 25-35 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 19-22 11544316-6 2001 Both FK506 and rapamycin efficiently inhibited proliferation of p18-null T cells. Tacrolimus 5-10 cyclin dependent kinase inhibitor 2C Mus musculus 64-67 11557049-2 2001 FKBP12 demonstrated a very tight, high affinity association with skeletal muscle microsomes, which was displaced by FK506. Tacrolimus 116-121 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 0-6 11522944-4 2001 We tested FK506 in a transgenic mouse model expressing mutated SOD1 for potential beneficial effects. Tacrolimus 10-15 superoxide dismutase 1, soluble Mus musculus 63-67 11504680-4 2001 Pretreatment with 100 ng/ml of FK506 (a PP2B inhibitor) but not with the PP1 and PP2A inhibitors calyculin A or okadaic acid potentiated the thrombin-induced increase in PKC phosphotransferase activity. Tacrolimus 31-36 coagulation factor II, thrombin Bos taurus 141-149 11504680-4 2001 Pretreatment with 100 ng/ml of FK506 (a PP2B inhibitor) but not with the PP1 and PP2A inhibitors calyculin A or okadaic acid potentiated the thrombin-induced increase in PKC phosphotransferase activity. Tacrolimus 31-36 protein kinase C alpha Bos taurus 170-173 11504680-5 2001 FK506 also potentiated thrombin-induced PKC-alpha but not PKC-beta phosphorylation. Tacrolimus 0-5 coagulation factor II, thrombin Bos taurus 23-31 11504680-5 2001 FK506 also potentiated thrombin-induced PKC-alpha but not PKC-beta phosphorylation. Tacrolimus 0-5 protein kinase C alpha Bos taurus 40-49 11504680-6 2001 FK506 but not calyculin A or okadaic acid inhibited recovery from the thrombin-induced decrease in transendothelial resistance. Tacrolimus 0-5 coagulation factor II, thrombin Bos taurus 70-78 11504680-8 2001 Downregulation of PKC with phorbol 12-myristate 13-acetate rescued the FK506-mediated inhibition of recovery, which was consistent with the finding that the thrombin-induced phosphorylation of PKC-alpha was reduced during the recovery phase. Tacrolimus 71-76 protein kinase C alpha Bos taurus 18-21 11504680-8 2001 Downregulation of PKC with phorbol 12-myristate 13-acetate rescued the FK506-mediated inhibition of recovery, which was consistent with the finding that the thrombin-induced phosphorylation of PKC-alpha was reduced during the recovery phase. Tacrolimus 71-76 coagulation factor II, thrombin Bos taurus 157-165 11504680-8 2001 Downregulation of PKC with phorbol 12-myristate 13-acetate rescued the FK506-mediated inhibition of recovery, which was consistent with the finding that the thrombin-induced phosphorylation of PKC-alpha was reduced during the recovery phase. Tacrolimus 71-76 protein kinase C alpha Bos taurus 193-202 11513333-4 2001 However, it has been recently observed that drugs like cyclosporin A, FK506, leflunomide, mycophenolate mofetil, pentoxifylline, and linomide can directly modulate cytokine and/or LPS-induced NO production in various cell types in vitro, probably by interfering with iNOS gene transcription or catalytic activity of iNOS enzyme. Tacrolimus 70-75 nitric oxide synthase 2 Homo sapiens 267-271 11513333-4 2001 However, it has been recently observed that drugs like cyclosporin A, FK506, leflunomide, mycophenolate mofetil, pentoxifylline, and linomide can directly modulate cytokine and/or LPS-induced NO production in various cell types in vitro, probably by interfering with iNOS gene transcription or catalytic activity of iNOS enzyme. Tacrolimus 70-75 nitric oxide synthase 2 Homo sapiens 316-320 11525945-5 2001 Mice treated successfully with FK506 had reduced perifollicular infiltrates of CD4+ and CD8+ cells and a decreased expression of MHC class I and II and ICAM-1 on hair follicle epithelium, compared to control mice. Tacrolimus 31-36 intercellular adhesion molecule 1 Mus musculus 152-158 11520640-1 2001 Immunophilin FK binding protein-12 (FKBP-12), the soluble receptor for the immunosuppressant drug FK506, is involved in a number of neuronal activities including increased nerve regeneration in the peripheral nervous system and enhanced recovery in animal models of neurodegenerative diseases. Tacrolimus 98-103 FKBP prolyl isomerase 1A Homo sapiens 36-43 11557049-4 2001 Furthermore, of the proteins solubilised from skeletal muscle, cardiac muscle and brain microsomes, only skeletal muscle RyR1 bound to an FKBP12-glutathione-S-transferase fusion protein, in a high affinity FK506 displaceable manner. Tacrolimus 206-211 ryanodine receptor 1 Homo sapiens 121-125 11557049-4 2001 Furthermore, of the proteins solubilised from skeletal muscle, cardiac muscle and brain microsomes, only skeletal muscle RyR1 bound to an FKBP12-glutathione-S-transferase fusion protein, in a high affinity FK506 displaceable manner. Tacrolimus 206-211 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 138-144 11523700-10 2001 Decreased production of IL-12 and tumor necrosis factor alpha, but not of IL-10, is likely to contribute to the impaired accessory-cell function of tacrolimus- and CsA-treated DCs. Tacrolimus 148-158 tumor necrosis factor Homo sapiens 34-61 11502273-0 2001 FK506 promotes adenosine release from endothelial cells via inhibition of adenosine kinase. Tacrolimus 0-5 adenosine kinase Homo sapiens 74-90 11502273-3 2001 Since inhibition of adenosine kinase promotes an increase in endogenous adenosine release, we tested a hypothesis that FK506 induces adenosine release via inhibition of adenosine kinase activity. Tacrolimus 119-124 adenosine kinase Homo sapiens 20-36 11502273-3 2001 Since inhibition of adenosine kinase promotes an increase in endogenous adenosine release, we tested a hypothesis that FK506 induces adenosine release via inhibition of adenosine kinase activity. Tacrolimus 119-124 adenosine kinase Homo sapiens 169-185 11502273-7 2001 These observations indicate that FK506 inhibits in situ adenosine kinase activity in endothelial cells. Tacrolimus 33-38 adenosine kinase Homo sapiens 56-72 11502273-9 2001 In conclusion, FK506 promotes adenosine release from endothelial cells by a novel mechanism involving inhibition of adenosine kinase activity associated with the membrane. Tacrolimus 15-20 adenosine kinase Homo sapiens 116-132 11387321-6 2001 The treatment of FK506, a specific inhibitor of calcineurin, abolishes the salt-tolerant phenotype of the pmr1 mutant. Tacrolimus 17-22 Ca(2+)/Mn(2+)-transporting P-type ATPase PMR1 Saccharomyces cerevisiae S288C 106-110 11520309-5 2001 Glucose-stimulated plasma immunoreactive insulin concentrations in the tacrolimus-treatment group were significantly higher than in the cyclosporin group (p < 0.05) and the controls (p < 0.001). Tacrolimus 71-81 insulin Homo sapiens 41-48 11520309-7 2001 The raised insulin concentrations with normal or increased blood glucose concentrations after renal transplantation suggests that insulin resistance was more marked in patients receiving tacrolimus-based immunosuppression. Tacrolimus 187-197 insulin Homo sapiens 11-18 11520309-7 2001 The raised insulin concentrations with normal or increased blood glucose concentrations after renal transplantation suggests that insulin resistance was more marked in patients receiving tacrolimus-based immunosuppression. Tacrolimus 187-197 insulin Homo sapiens 130-137 11477316-0 2001 Quantitation of immunosuppression by tacrolimus using flow cytometric analysis of interleukin-2 and interferon-gamma inhibition in CD8(-) and CD8(+) peripheral blood T cells. Tacrolimus 37-47 interleukin 2 Homo sapiens 82-95 11406149-5 2001 The expression of CD8alpha in mitogen-stimulated CD4(+) cells was blocked completely by calcineurin inhibitors (cyclosporine A and FK-506), and partially by rapamycin and SDZ-RAD. Tacrolimus 131-137 CD8a molecule Rattus norvegicus 18-26 11406149-5 2001 The expression of CD8alpha in mitogen-stimulated CD4(+) cells was blocked completely by calcineurin inhibitors (cyclosporine A and FK-506), and partially by rapamycin and SDZ-RAD. Tacrolimus 131-137 Cd4 molecule Rattus norvegicus 49-52 11477316-0 2001 Quantitation of immunosuppression by tacrolimus using flow cytometric analysis of interleukin-2 and interferon-gamma inhibition in CD8(-) and CD8(+) peripheral blood T cells. Tacrolimus 37-47 interferon gamma Homo sapiens 100-116 11477316-0 2001 Quantitation of immunosuppression by tacrolimus using flow cytometric analysis of interleukin-2 and interferon-gamma inhibition in CD8(-) and CD8(+) peripheral blood T cells. Tacrolimus 37-47 CD8a molecule Homo sapiens 131-134 11477316-0 2001 Quantitation of immunosuppression by tacrolimus using flow cytometric analysis of interleukin-2 and interferon-gamma inhibition in CD8(-) and CD8(+) peripheral blood T cells. Tacrolimus 37-47 CD8a molecule Homo sapiens 142-145 11477316-1 2001 The authors have determined the frequency of intracellular interleukin-2 (IL-2) and interferon-gamma (IFN-gamma) synthesis by T-cell subsets in whole blood (WB) and isolated lymphocytes in 16 transplant recipients treated with tacrolimus and 10 control patients who were not transplant recipients. Tacrolimus 227-237 interleukin 2 Homo sapiens 59-72 11512052-5 2001 In addition, Western blotting analysis of intragraft cell infiltrates showed a reduction in the expression of activated p38MAP kinase in allografts treated with FK506. Tacrolimus 161-166 mitogen activated protein kinase 14 Rattus norvegicus 120-133 11477316-1 2001 The authors have determined the frequency of intracellular interleukin-2 (IL-2) and interferon-gamma (IFN-gamma) synthesis by T-cell subsets in whole blood (WB) and isolated lymphocytes in 16 transplant recipients treated with tacrolimus and 10 control patients who were not transplant recipients. Tacrolimus 227-237 interferon gamma Homo sapiens 84-100 11477316-1 2001 The authors have determined the frequency of intracellular interleukin-2 (IL-2) and interferon-gamma (IFN-gamma) synthesis by T-cell subsets in whole blood (WB) and isolated lymphocytes in 16 transplant recipients treated with tacrolimus and 10 control patients who were not transplant recipients. Tacrolimus 227-237 interferon gamma Homo sapiens 102-111 11512052-6 2001 We conclude that intragraft cell infiltrate expression of activated p38MAP kinase is an important marker of acute rejection in this animal model of small bowel transplantation, and that FK506 is an effective immunosuppressant, in this situation, that may act in part by preventing the activation of p38MAP kinase. Tacrolimus 186-191 mitogen activated protein kinase 14 Rattus norvegicus 299-312 11477316-6 2001 Adding tacrolimus (10 ng/mL) to lymphocyte cultures inhibited (90%) IL-2 production in isolated T cells but not in the whole-blood assay. Tacrolimus 7-17 interleukin 2 Homo sapiens 68-72 11477316-7 2001 The dose of tacrolimus required for a 50% inhibition of IL-2 release in T cells was 10-fold higher in cultures with RBC than without. Tacrolimus 12-22 interleukin 2 Homo sapiens 56-60 11477316-8 2001 Peripheral blood mononuclear cells (PBMC) isolated from tacrolimus-treated whole blood (WB) showed less IL-2 inhibition than did lymphocytes in the WB. Tacrolimus 56-66 interleukin 2 Homo sapiens 104-108 11418477-9 2001 Rapa exerts its apoptotic effect via a reversible binding to the cytosolic receptor protein FKBP-12, as demonstrated in competition experiments with FK506, which is structurally related to Rapa. Tacrolimus 149-154 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 92-99 11335722-1 2001 FKBP12 is a ubiquitous and a highly conserved prolyl isomerase that binds the immunosuppressive drugs FK506 and rapamycin. Tacrolimus 102-107 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 0-6 11469900-9 2001 However, the decreases in PEPT1 and SGLT1 expression (both mRNA and protein) were partially prevented by tacrolimus treatment. Tacrolimus 105-115 solute carrier family 15 member 1 Rattus norvegicus 26-31 11469900-9 2001 However, the decreases in PEPT1 and SGLT1 expression (both mRNA and protein) were partially prevented by tacrolimus treatment. Tacrolimus 105-115 solute carrier family 5 member 1 Rattus norvegicus 36-41 11437371-4 2001 Ascomycin, an FK506 analogue, was identified for the first time as a drug which can disrupt the FKBP12-RyR1 complex. Tacrolimus 14-19 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 96-102 11437371-4 2001 Ascomycin, an FK506 analogue, was identified for the first time as a drug which can disrupt the FKBP12-RyR1 complex. Tacrolimus 14-19 ryanodine receptor 1 Homo sapiens 103-107 11311446-3 2001 We used FK506 and three different monoperoxovanadium complexes (mpVs) as pharmacological tools for manipulation of MKP-1 expression. Tacrolimus 8-13 dual specificity phosphatase 1 Rattus norvegicus 115-120 11472421-9 2001 Finally, a suppressive effect of FK506 on Bcl-2 production but not on Bax production was confirmed by Western blotting. Tacrolimus 33-38 B cell leukemia/lymphoma 2 Mus musculus 42-47 11472421-10 2001 This unique effect of FK506 on the augmentation of T cell apoptosis is probably one of the mechanisms explaining its beneficial effect on aly autoimmune pancreatitis. Tacrolimus 22-27 mitogen-activated protein kinase kinase kinase 14 Mus musculus 138-141 11515543-8 2001 In addition, accumulation of rhodamine 6G in the cells was detected as a result of Pdr5 inhibition by FK506. Tacrolimus 102-107 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 83-87 11311446-5 2001 On the other hand, FK 506 has transient effect on ERK activation. Tacrolimus 19-25 Eph receptor B1 Rattus norvegicus 50-53 11311446-7 2001 The concomitant alterations observed in intensities and duration of phospho-ERKs and phospho-JNKs signals suggest that monoperoxovanadium complexes in combination with FK 506 enhance survival of PC12 cells by an induction of MKP-1 expression. Tacrolimus 168-174 dual specificity phosphatase 1 Rattus norvegicus 225-230 11407316-6 2001 In a comparative study with steroids (alclometasone dipropionate and betamethason valerate) in anti-CD3/CD2 system, tacrolimus and both steroids inhibited Th1 cytokines (IL-2, IFN-gamma), Th2 cytokines (IL-4, IL-5) and IL-3, GM-CSF (produced by both Th1 and Th2). Tacrolimus 116-126 negative elongation factor complex member C/D Homo sapiens 155-158 11428741-3 2001 OBJECTIVES: We compared the effects of tacrolimus on interleukin (IL)-5 and tumor necrosis factor-alpha (TNF-alpha) production and chemical mediator release from excised human lung tissue with those of steroids. Tacrolimus 39-49 interleukin 5 Homo sapiens 53-71 11428741-3 2001 OBJECTIVES: We compared the effects of tacrolimus on interleukin (IL)-5 and tumor necrosis factor-alpha (TNF-alpha) production and chemical mediator release from excised human lung tissue with those of steroids. Tacrolimus 39-49 tumor necrosis factor Homo sapiens 76-103 11428741-14 2001 Tacrolimus and dexamethasone significantly inhibited TNF-alpha and IL-5 protein production and mRNA expression. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 53-62 11428741-14 2001 Tacrolimus and dexamethasone significantly inhibited TNF-alpha and IL-5 protein production and mRNA expression. Tacrolimus 0-10 interleukin 5 Homo sapiens 67-71 11519126-4 2001 We have examined whether the expression levels of the intestinal absorptive barriers, MDR1 gene product P-glycoprotein and cytochrome P450 IIIA4(CYP3A4), correlate with the trough levels of orally administered tacrolimus in a recipient of small bowel transplant for 4 months. Tacrolimus 210-220 ATP binding cassette subfamily B member 1 Homo sapiens 86-90 11519126-4 2001 We have examined whether the expression levels of the intestinal absorptive barriers, MDR1 gene product P-glycoprotein and cytochrome P450 IIIA4(CYP3A4), correlate with the trough levels of orally administered tacrolimus in a recipient of small bowel transplant for 4 months. Tacrolimus 210-220 ATP binding cassette subfamily B member 1 Homo sapiens 104-118 11519126-6 2001 The tacrolimus concentration/dose ratio correlated well with the mRNA expression level of MDR1, but not CYP3A4. Tacrolimus 4-14 ATP binding cassette subfamily B member 1 Homo sapiens 90-94 11519126-7 2001 Intestinal P-glycoprotein rather than CYP3A4 is a good probe to predict the intraindividual variation in the tacrolimus pharmacokinetics. Tacrolimus 109-119 ATP binding cassette subfamily B member 1 Homo sapiens 11-25 11407316-6 2001 In a comparative study with steroids (alclometasone dipropionate and betamethason valerate) in anti-CD3/CD2 system, tacrolimus and both steroids inhibited Th1 cytokines (IL-2, IFN-gamma), Th2 cytokines (IL-4, IL-5) and IL-3, GM-CSF (produced by both Th1 and Th2). Tacrolimus 116-126 interleukin 2 Homo sapiens 170-174 11407316-6 2001 In a comparative study with steroids (alclometasone dipropionate and betamethason valerate) in anti-CD3/CD2 system, tacrolimus and both steroids inhibited Th1 cytokines (IL-2, IFN-gamma), Th2 cytokines (IL-4, IL-5) and IL-3, GM-CSF (produced by both Th1 and Th2). Tacrolimus 116-126 interferon gamma Homo sapiens 176-185 11407316-6 2001 In a comparative study with steroids (alclometasone dipropionate and betamethason valerate) in anti-CD3/CD2 system, tacrolimus and both steroids inhibited Th1 cytokines (IL-2, IFN-gamma), Th2 cytokines (IL-4, IL-5) and IL-3, GM-CSF (produced by both Th1 and Th2). Tacrolimus 116-126 interleukin 4 Homo sapiens 203-207 11407316-6 2001 In a comparative study with steroids (alclometasone dipropionate and betamethason valerate) in anti-CD3/CD2 system, tacrolimus and both steroids inhibited Th1 cytokines (IL-2, IFN-gamma), Th2 cytokines (IL-4, IL-5) and IL-3, GM-CSF (produced by both Th1 and Th2). Tacrolimus 116-126 interleukin 5 Homo sapiens 209-213 11407316-6 2001 In a comparative study with steroids (alclometasone dipropionate and betamethason valerate) in anti-CD3/CD2 system, tacrolimus and both steroids inhibited Th1 cytokines (IL-2, IFN-gamma), Th2 cytokines (IL-4, IL-5) and IL-3, GM-CSF (produced by both Th1 and Th2). Tacrolimus 116-126 interleukin 3 Homo sapiens 219-223 11407316-6 2001 In a comparative study with steroids (alclometasone dipropionate and betamethason valerate) in anti-CD3/CD2 system, tacrolimus and both steroids inhibited Th1 cytokines (IL-2, IFN-gamma), Th2 cytokines (IL-4, IL-5) and IL-3, GM-CSF (produced by both Th1 and Th2). Tacrolimus 116-126 colony stimulating factor 2 Homo sapiens 225-231 11407316-6 2001 In a comparative study with steroids (alclometasone dipropionate and betamethason valerate) in anti-CD3/CD2 system, tacrolimus and both steroids inhibited Th1 cytokines (IL-2, IFN-gamma), Th2 cytokines (IL-4, IL-5) and IL-3, GM-CSF (produced by both Th1 and Th2). Tacrolimus 116-126 negative elongation factor complex member C/D Homo sapiens 250-253 11451170-0 2001 Identification of cyclosporine A and tacrolimus glucuronidation in human liver and the gastrointestinal tract by a differentially expressed UDP-glucuronosyltransferase: UGT2B7. Tacrolimus 37-47 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 169-175 11369912-7 2001 RESULTS: Pruritus, tenderness, and erythema were resolved in all 3 patients after 7 to 10 consecutive days" use of tacrolimus 0.075% ointment in conjunction with avoidance of topical steroids, caffeine, spicy food, alcohol, hot fluids, and fluoride. Tacrolimus 115-125 alcohol dehydrogenase iron containing 1 Homo sapiens 224-227 11369912-8 2001 CONCLUSION: This preliminary study demonstrates that tacrolimus 0.075% ointment may be effective for patients with steroid-induced rosacea, when combined with avoidance of topical steroid use, as well as avoidance of other agents known to aggravate rosacea (caffeine, spicy foods, alcohol, hot fluids, and fluoride). Tacrolimus 53-63 alcohol dehydrogenase iron containing 1 Homo sapiens 290-293 11451170-7 2001 Analyses using recombinant UDPglucuronosyltransferases identified UGT2B7 as a human UDP-glucuronosyltransferase with specific activity toward cyclosporine A and tacrolimus. Tacrolimus 161-171 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 66-72 11437083-5 2001 Traumatic axonal injury (TAI), detected via an antibody against beta-amyloid precursor protein (APP), a specific marker of axonal injury, was significantly reduced at 24 hr postinjury in Sprague-Dawley rats receiving intravenous FK506 (2 mg/kg; n = 5) 30 min prior to injury compared to vehicle controls (n = 3). Tacrolimus 229-234 amyloid beta precursor protein Rattus norvegicus 64-94 11353864-3 2001 RyR3 was purified from detergent-solubilized transfected cells by affinity chromatography using 12.6-kDa FK506-binding protein in the form of a GST fusion as the affinity ligand. Tacrolimus 105-110 ryanodine receptor 3 Homo sapiens 0-4 11397967-1 2001 BACKGROUND: Tacrolimus, a substrate of CYP3A, has low and variable bioavailability similar to cyclosporine. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 39-44 11411802-11 2001 FK506 enhanced the induction of Hsp70, but CsA again had no effect on Hsp70 induction. Tacrolimus 0-5 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 32-37 11408029-0 2001 FK506 (tacrolimus) increases rat alpha1-acid glycoprotein expression in liver and primary cultured hepatocytes. Tacrolimus 0-5 orosomucoid 1 Rattus norvegicus 33-57 11408029-0 2001 FK506 (tacrolimus) increases rat alpha1-acid glycoprotein expression in liver and primary cultured hepatocytes. Tacrolimus 7-17 orosomucoid 1 Rattus norvegicus 33-57 11408029-1 2001 FK506 (tacrolimus) (10 mg/kg, s.c., 5 days) increased rat alpha1-acid glycoprotein (AGP) in serum and AGP mRNA in liver. Tacrolimus 0-5 orosomucoid 1 Rattus norvegicus 58-82 11408029-1 2001 FK506 (tacrolimus) (10 mg/kg, s.c., 5 days) increased rat alpha1-acid glycoprotein (AGP) in serum and AGP mRNA in liver. Tacrolimus 0-5 orosomucoid 1 Rattus norvegicus 84-87 11408029-1 2001 FK506 (tacrolimus) (10 mg/kg, s.c., 5 days) increased rat alpha1-acid glycoprotein (AGP) in serum and AGP mRNA in liver. Tacrolimus 0-5 orosomucoid 1 Rattus norvegicus 102-105 11408029-1 2001 FK506 (tacrolimus) (10 mg/kg, s.c., 5 days) increased rat alpha1-acid glycoprotein (AGP) in serum and AGP mRNA in liver. Tacrolimus 7-17 orosomucoid 1 Rattus norvegicus 58-82 11408029-1 2001 FK506 (tacrolimus) (10 mg/kg, s.c., 5 days) increased rat alpha1-acid glycoprotein (AGP) in serum and AGP mRNA in liver. Tacrolimus 7-17 orosomucoid 1 Rattus norvegicus 84-87 11408029-1 2001 FK506 (tacrolimus) (10 mg/kg, s.c., 5 days) increased rat alpha1-acid glycoprotein (AGP) in serum and AGP mRNA in liver. Tacrolimus 7-17 orosomucoid 1 Rattus norvegicus 102-105 11408029-2 2001 FK506 potentiated the dexamethasone-increased AGP expression in primary cultured hepatocytes. Tacrolimus 0-5 orosomucoid 1 Rattus norvegicus 46-49 11408029-3 2001 In the luciferase promoter assay, FK506 potentiated the dexamethasone-increased promoter activity of the AGP gene in cultured rat hepatocytes, although FK506 alone had no effect on its activity. Tacrolimus 34-39 orosomucoid 1 Rattus norvegicus 105-108 11408029-3 2001 In the luciferase promoter assay, FK506 potentiated the dexamethasone-increased promoter activity of the AGP gene in cultured rat hepatocytes, although FK506 alone had no effect on its activity. Tacrolimus 152-157 orosomucoid 1 Rattus norvegicus 105-108 11408029-5 2001 These results indicated that FK506 causes the transcriptional induction of AGP, at least in part, via a glucocorticoid-mediated mechanism. Tacrolimus 29-34 orosomucoid 1 Rattus norvegicus 75-78 11397967-2 2001 Co-administration of ketoconazole, potent inhibitor of gut and hepatic CYP3A, has been shown to increase tacrolimus bioavailability in healthy volunteers. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 71-76 11278965-9 2001 The calcineurin blockers CsA and FK506 also prevented the PDGF-induced NFAT4 nuclear localization. Tacrolimus 33-38 nuclear factor of activated T cells 3 Homo sapiens 71-76 11341768-6 2001 FK506 also reduced the expression of myosin, induced a more elongated cell morphology, and impaired network formation in cultured chick embryonic cardiomyocytes. Tacrolimus 0-5 myosin, heavy chain 15 Gallus gallus 37-43 11371998-5 2001 RESULTS: The mRNA expression level of MDR1 (r = -0.776), but not CYP3A4 (r = -0.094), was inversely related to the concentration/dose ratio of tacrolimus. Tacrolimus 143-153 ATP binding cassette subfamily B member 1 Homo sapiens 38-42 11348636-0 2001 Induction of choleresis by immunosuppressant FK506 through stimulation of insulin-like growth factor-I production in the liver of rats. Tacrolimus 45-50 insulin-like growth factor 1 Rattus norvegicus 74-102 11348636-4 2001 A significant increase in plasma levels of IGF-I was observed in rats 30 min after a single intravenous administration of FK506. Tacrolimus 122-127 insulin-like growth factor 1 Rattus norvegicus 43-48 11348636-5 2001 Oral treatment of rats with FK506 for 1 week also resulted in an increase in both plasma and hepatic levels of IGF-I. Tacrolimus 28-33 insulin-like growth factor 1 Rattus norvegicus 111-116 11348636-6 2001 Overall, this study showed that FK506 treatment increased bile flow and also induced an increase in the plasma and hepatic levels of IGF-I in rats, suggesting that a stimulation of hepatic IGF-I production by FK506 may contribute to its choleretic profile. Tacrolimus 32-37 insulin-like growth factor 1 Rattus norvegicus 133-138 11348636-6 2001 Overall, this study showed that FK506 treatment increased bile flow and also induced an increase in the plasma and hepatic levels of IGF-I in rats, suggesting that a stimulation of hepatic IGF-I production by FK506 may contribute to its choleretic profile. Tacrolimus 32-37 insulin-like growth factor 1 Rattus norvegicus 189-194 11348636-6 2001 Overall, this study showed that FK506 treatment increased bile flow and also induced an increase in the plasma and hepatic levels of IGF-I in rats, suggesting that a stimulation of hepatic IGF-I production by FK506 may contribute to its choleretic profile. Tacrolimus 209-214 insulin-like growth factor 1 Rattus norvegicus 133-138 11348636-6 2001 Overall, this study showed that FK506 treatment increased bile flow and also induced an increase in the plasma and hepatic levels of IGF-I in rats, suggesting that a stimulation of hepatic IGF-I production by FK506 may contribute to its choleretic profile. Tacrolimus 209-214 insulin-like growth factor 1 Rattus norvegicus 189-194 11371998-8 2001 CONCLUSIONS: Intestinal MDR1 is not only a good probe with which to predict the interindividual variation in tacrolimus pharmacokinetics after LDLT but also a powerful prognostic indicator for the outcome of LDLT. Tacrolimus 109-119 ATP binding cassette subfamily B member 1 Homo sapiens 24-28 11303042-0 2001 Contribution of P-glycoprotein to the enhancing effects of dimethyl-beta-cyclodextrin on oral bioavailability of tacrolimus. Tacrolimus 113-123 ATP binding cassette subfamily B member 1 Homo sapiens 16-30 11359520-1 2001 Immunosuppressant drugs, like FK506, and nonimmunosuppressant compounds like, GPI1046 and L685818, are immunophilin ligands that specifically bind to immunophilins, like FK506 binding protein 12 (FKBP12). Tacrolimus 30-35 FKBP prolyl isomerase 1A Rattus norvegicus 170-194 11359520-1 2001 Immunosuppressant drugs, like FK506, and nonimmunosuppressant compounds like, GPI1046 and L685818, are immunophilin ligands that specifically bind to immunophilins, like FK506 binding protein 12 (FKBP12). Tacrolimus 30-35 FKBP prolyl isomerase 1A Rattus norvegicus 196-202 11354410-9 2001 Although CSF levels of IL-6 and TNFalpha were completely suppressed by tacrolimus at all time points and at both concentrations, CSF levels of glutamate and hypoxanthine, as well as edema formation, were only marginally influenced. Tacrolimus 71-81 interleukin 6 Rattus norvegicus 23-27 11303042-9 2001 These results suggested that the enhancing effect of DM-beta-CyD on the oral bioavailability of tacrolimus is due not only to its solubilizing effect but also, at least in part, to its inhibitory effect on the P-gp-mediated efflux of tacrolimus from intestinal epithelial cells. Tacrolimus 96-106 ATP binding cassette subfamily B member 1 Homo sapiens 210-214 11303042-9 2001 These results suggested that the enhancing effect of DM-beta-CyD on the oral bioavailability of tacrolimus is due not only to its solubilizing effect but also, at least in part, to its inhibitory effect on the P-gp-mediated efflux of tacrolimus from intestinal epithelial cells. Tacrolimus 234-244 ATP binding cassette subfamily B member 1 Homo sapiens 210-214 11465431-0 2001 Effects of tacrolimus and cyclosporin A on peptide transporter PEPT1 in Caco-2 cells. Tacrolimus 11-21 solute carrier family 15 member 1 Homo sapiens 63-68 11392610-8 2001 FK506, a specific inhibitor of calcineurin, inhibited A23187- and thapsigargin-induced IL-8 mRNA expression in a dose dependent manner. Tacrolimus 0-5 C-X-C motif chemokine ligand 8 Homo sapiens 87-91 11357886-3 2001 FK506 potently inhibited IL-6 production from PBMC stimulated with anti-CD3 and anti-CD28 monoclonal antibody (anti-CD3/CD28). Tacrolimus 0-5 interleukin 6 Homo sapiens 25-29 11357886-3 2001 FK506 potently inhibited IL-6 production from PBMC stimulated with anti-CD3 and anti-CD28 monoclonal antibody (anti-CD3/CD28). Tacrolimus 0-5 CD28 molecule Homo sapiens 85-89 11357886-3 2001 FK506 potently inhibited IL-6 production from PBMC stimulated with anti-CD3 and anti-CD28 monoclonal antibody (anti-CD3/CD28). Tacrolimus 0-5 CD28 molecule Homo sapiens 120-124 11357886-12 2001 The present study suggests that FK506 is the most effective among the four agents for the suppression of IL-6 production and IL-6-mediated autoantibody production in T cell activation related autoimmune diseases such as RA. Tacrolimus 32-37 interleukin 6 Homo sapiens 105-109 11377534-0 2001 Effect of tacrolimus on the gene expression of renin and endothelin in the rat kidney. Tacrolimus 10-20 renin Rattus norvegicus 47-52 11357886-0 2001 Effects of FK506 and other immunosuppressive anti-rheumatic agents on T cell activation mediated IL-6 and IgM production in vitro. Tacrolimus 11-16 interleukin 6 Homo sapiens 97-101 11104762-10 2001 The immunoreactivity for phosphorylated tau at Ser-320 increased in the presence of a phosphatase inhibitor, FK506 treatment, which means that calcineurin (protein phosphatase 2B) may be involved in dephosphorylating tau at Ser-320 site. Tacrolimus 109-114 microtubule-associated protein tau Cricetulus griseus 40-43 11392178-3 2001 Phenytoin, an anti-convulsant agent, can induce the Cytochrome P450 enzyme in the liver, which metabolizes Tacrolimus. Tacrolimus 107-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 11357886-12 2001 The present study suggests that FK506 is the most effective among the four agents for the suppression of IL-6 production and IL-6-mediated autoantibody production in T cell activation related autoimmune diseases such as RA. Tacrolimus 32-37 interleukin 6 Homo sapiens 125-129 11328549-13 2001 Both ABO donor-recipient mismatch patients were started on tacrolimus/MMF as primary therapy and had no significant episodes of rejection. Tacrolimus 59-69 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 5-8 11104762-10 2001 The immunoreactivity for phosphorylated tau at Ser-320 increased in the presence of a phosphatase inhibitor, FK506 treatment, which means that calcineurin (protein phosphatase 2B) may be involved in dephosphorylating tau at Ser-320 site. Tacrolimus 109-114 microtubule-associated protein tau Cricetulus griseus 217-220 11150862-8 2001 Interleukin (IL)-2 and IL-2 receptor synthesis was upregulated in control animals and inhibited by tacrolimus treatment. Tacrolimus 99-109 interleukin 2 Rattus norvegicus 0-18 11237798-0 2001 CD8+ cytotoxic lymphocyte responses against cytomegalovirus after liver transplantation: correlation with time from transplant to receipt of tacrolimus. Tacrolimus 141-151 CD8a molecule Homo sapiens 0-3 11237798-4 2001 There was an inverse correlation among levels of the calcineurin inhibitor, tacrolimus, and ppUL83 CD8(+) CTL frequencies (r=-.31; P=.005), which is consistent with the presence of a large proportion (70%) of activated (CD38(+)) ppUL83 CD8(+) CTL within the population of HLA class I tetramer-positive cells. Tacrolimus 76-86 CD8a molecule Homo sapiens 99-102 11164950-7 2001 Binding of FAP48 to FKBPs is inhibited by the macrolide FK506 indicating that the binding sites on the immunophilins coincide with the binding site for FK506. Tacrolimus 56-61 glomulin, FKBP associated protein Homo sapiens 11-16 11164950-7 2001 Binding of FAP48 to FKBPs is inhibited by the macrolide FK506 indicating that the binding sites on the immunophilins coincide with the binding site for FK506. Tacrolimus 152-157 glomulin, FKBP associated protein Homo sapiens 11-16 11171588-12 2001 FK-506, an inhibitor of FKBP12/IP(3) receptor interactions, had no effect on the rise in basal Ca(2+) but blocked the inhibitory effects of increased basal Ca(2+) and ACh on Ca(2+) transients. Tacrolimus 0-6 FK506 binding protein 1a Mus musculus 24-30 11171588-12 2001 FK-506, an inhibitor of FKBP12/IP(3) receptor interactions, had no effect on the rise in basal Ca(2+) but blocked the inhibitory effects of increased basal Ca(2+) and ACh on Ca(2+) transients. Tacrolimus 0-6 inositol 1,4,5-triphosphate receptor 3 Mus musculus 31-45 11292296-13 2001 Plasmapheresis and tacrolimus-mycophenolate mofetil rescue reversed rejection in 9 of 10 recipients with refractory AHR. Tacrolimus 19-29 aryl hydrocarbon receptor Homo sapiens 116-119 11150862-8 2001 Interleukin (IL)-2 and IL-2 receptor synthesis was upregulated in control animals and inhibited by tacrolimus treatment. Tacrolimus 99-109 interleukin 2 Rattus norvegicus 23-27 11181807-13 2001 Insulin sensitivity correlated negatively with tacrolimus trough level. Tacrolimus 47-57 insulin Homo sapiens 0-7 11181807-15 2001 During tacrolimus administration, k(G) decreased in almost all patients as a result of a diminished insulin secretion response to a glucose load, whereas insulin resistance did not change. Tacrolimus 7-17 insulin Homo sapiens 100-107 11226255-1 2001 FKBP12, the 12-kDa FK506-binding protein, is a ubiquitous abundant protein that acts as a receptor for the immunosuppressant drug FK506, binds tightly to intracellular calcium release channels and to the transforming growth factor beta (TGF-beta) type I receptor. Tacrolimus 19-24 FK506 binding protein 1a Mus musculus 0-6 11240954-3 2001 OBJECTIVE: The present study focused on the effects of topical tacrolimus treatment on epidermal CD1a+/FcepsilonRI+ DC populations in lesional AD. Tacrolimus 63-73 CD1a molecule Homo sapiens 97-101 11240954-3 2001 OBJECTIVE: The present study focused on the effects of topical tacrolimus treatment on epidermal CD1a+/FcepsilonRI+ DC populations in lesional AD. Tacrolimus 63-73 Fc epsilon receptor Ia Homo sapiens 103-114 11240954-8 2001 Finally, topical tacrolimus led to a progressive decrease in the IDEC population within the pool of CD1a+ epidermal DCs and also to a decrease in their CD36 expression, which is indicative of lower local inflammation. Tacrolimus 17-27 CD1a molecule Homo sapiens 100-104 11240954-9 2001 CONCLUSION: Epidermal CD1a+ DCs may represent a target for topical tacrolimus in the treatment of AD. Tacrolimus 67-77 CD1a molecule Homo sapiens 22-26 11251806-5 2001 We show that the FKR1-1 mutation, which confers dominant FK506 resistance, results from a 6 bp duplication generating a two-amino-acid insertion in the latch region of calcineurin B. Tacrolimus 57-62 protein phosphatase 3 regulatory subunit B, alpha Homo sapiens 168-181 11239516-1 2001 OBJECTIVES: Little is known about the effect of ischemia/reperfusion with xenogenic blood on function and gene expression of CYP3A4, the enzyme largely responsible for the metabolism of the immunosuppressants Cyclosporin A (CsA) and Tacrolimus. Tacrolimus 233-243 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 125-131 11322650-1 2001 The modulatory effect of FK 506 and cyclosporin A (CsA) on the expression of inducible nitric oxide synthase (iNOS) in macrophages and mechanisms of their action were analysed. Tacrolimus 25-31 nitric oxide synthase 2 Rattus norvegicus 77-108 11322650-1 2001 The modulatory effect of FK 506 and cyclosporin A (CsA) on the expression of inducible nitric oxide synthase (iNOS) in macrophages and mechanisms of their action were analysed. Tacrolimus 25-31 nitric oxide synthase 2 Rattus norvegicus 110-114 11255695-5 2001 Treatment of DS rats with the calcineurin inhibitor FK506 (0.1 or 0.01 mg/kg every second day) from the age of 6 weeks to 12 weeks inhibited the activation of calcineurin in the heart in a dose-dependent manner and attenuated the development of load-induced cardiac hypertrophy and fibrosis without change of hemodynamic parameters. Tacrolimus 52-57 calcineurin binding protein 1 Rattus norvegicus 30-51 11174203-7 2001 Tacrolimus inhibited the expression of IL-2R (CD25) and of the costimulatory molecules CD80 (B7.1) and CD40. Tacrolimus 0-10 interleukin 2 receptor subunit alpha Homo sapiens 39-44 11174203-7 2001 Tacrolimus inhibited the expression of IL-2R (CD25) and of the costimulatory molecules CD80 (B7.1) and CD40. Tacrolimus 0-10 interleukin 2 receptor subunit alpha Homo sapiens 46-50 11174203-7 2001 Tacrolimus inhibited the expression of IL-2R (CD25) and of the costimulatory molecules CD80 (B7.1) and CD40. Tacrolimus 0-10 CD80 molecule Homo sapiens 87-91 11174203-7 2001 Tacrolimus inhibited the expression of IL-2R (CD25) and of the costimulatory molecules CD80 (B7.1) and CD40. Tacrolimus 0-10 CD40 molecule Homo sapiens 103-107 11164708-6 2001 Tacrolimus (FK-506), at a concentration of 10(-8) M, significantly inhibited DME-induced IL-13 production from PBMCs. Tacrolimus 0-10 interleukin 13 Homo sapiens 89-94 11164708-6 2001 Tacrolimus (FK-506), at a concentration of 10(-8) M, significantly inhibited DME-induced IL-13 production from PBMCs. Tacrolimus 12-18 interleukin 13 Homo sapiens 89-94 11172433-6 2001 Activation of CD4+ cells was suppressed significantly in the FK506-treated group on postoperative day 7 compared with the untreated group (29.4% +/- 3.55% v 52.83% +/- 11.9%; P <.01). Tacrolimus 61-66 Cd4 molecule Rattus norvegicus 14-17 11172433-8 2001 Interestingly, at late acute rejection, activated CD4+ and CD8+ T cells remained at almost the same low levels as those on postoperative day 7 in the FK506-treated group. Tacrolimus 150-155 Cd4 molecule Rattus norvegicus 50-53 11169223-6 2001 In contrast, cyclosporine A and tacrolimus inhibited the tumour necrosis factor (TNF)-alpha production in response to LPS, but not to PepG and LTA. Tacrolimus 32-42 tumor necrosis factor Homo sapiens 57-91 11054412-11 2001 12-kDa FK506-binding protein (FKBP12), however, could not be responsible for it, because FK506 treatment did not eliminate the suppression, in contrast to marked removal of 12-kDa FK506-binding protein from alpha-RyR. Tacrolimus 7-12 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 30-36 11266931-0 2001 IL-10: a tacrolimus-specific cytotoxic mediator in ongoing allograft rejection. Tacrolimus 9-19 interleukin 10 Homo sapiens 0-5 11714196-4 2001 FK506, in a concentration-dependent fashion, inhibited T-cell proliferation and steady-state mRNA expression of IL-2 and IL-7; half-maximal suppression was obtained at 10(-7) to 5 x 10(-8) M. We tested whether FK506 antiproliferative effect could be overcome with exogenously reconstituted rIL-2 and/or rIL-7. Tacrolimus 0-5 interleukin 2 Homo sapiens 112-116 11258666-4 2001 cADPR interacts with FK506, an immunosuppressant, at FKBP12.6, FK506-binding-protein, and calcineurin, or ryanodine receptors. Tacrolimus 21-26 FK506 binding protein 1b Mus musculus 53-61 11213069-15 2001 Insulin dependence in our population rarely resolved, even after lowering tacrolimus and steroid doses. Tacrolimus 74-84 insulin Homo sapiens 0-7 11714196-4 2001 FK506, in a concentration-dependent fashion, inhibited T-cell proliferation and steady-state mRNA expression of IL-2 and IL-7; half-maximal suppression was obtained at 10(-7) to 5 x 10(-8) M. We tested whether FK506 antiproliferative effect could be overcome with exogenously reconstituted rIL-2 and/or rIL-7. Tacrolimus 0-5 interleukin 7 Homo sapiens 121-125 11714196-4 2001 FK506, in a concentration-dependent fashion, inhibited T-cell proliferation and steady-state mRNA expression of IL-2 and IL-7; half-maximal suppression was obtained at 10(-7) to 5 x 10(-8) M. We tested whether FK506 antiproliferative effect could be overcome with exogenously reconstituted rIL-2 and/or rIL-7. Tacrolimus 0-5 interleukin 2 Rattus norvegicus 290-295 11714196-4 2001 FK506, in a concentration-dependent fashion, inhibited T-cell proliferation and steady-state mRNA expression of IL-2 and IL-7; half-maximal suppression was obtained at 10(-7) to 5 x 10(-8) M. We tested whether FK506 antiproliferative effect could be overcome with exogenously reconstituted rIL-2 and/or rIL-7. Tacrolimus 0-5 interleukin 7 Rattus norvegicus 303-308 11714196-8 2001 FK506 inhibited T-cell activation stimulated by rIL-2 and by rIL-7, individually and in combination. Tacrolimus 0-5 interleukin 2 Rattus norvegicus 48-53 11714196-8 2001 FK506 inhibited T-cell activation stimulated by rIL-2 and by rIL-7, individually and in combination. Tacrolimus 0-5 interleukin 7 Rattus norvegicus 61-66 11208908-5 2001 Pre-incubation of the cells with ryanodine or FK506 completely eliminated PCB95 responses, suggesting a primary action on the FKPP12/RyR-sensitive store. Tacrolimus 46-51 ryanodine receptor 2 Rattus norvegicus 133-136 11368293-13 2001 The terminal elimination half-life (t1/2beta) of tacrolimus is approximately 12 hours (with a range of 3.5 to 40.5 hours). Tacrolimus 49-59 interleukin 1 receptor like 1 Homo sapiens 36-44 11385231-0 2001 Interferon-gamma-induced RANTES production by human keratinocytes is enhanced by IL-1beta, TNF-alpha, IL-4 and IL-13 and is inhibited by dexamethasone and tacrolimus. Tacrolimus 155-165 C-C motif chemokine ligand 5 Homo sapiens 25-31 11549851-2 2001 We compared the acute toxic and antiproliferative effects as well as the effects on the transport activity of Na(+)/K(+)-ATPase and Na(+)/K(+)/2Cl(-) cotransporter of CsA and FK506 in an established cell line of distal/collecting tubule origin (MDCK cells). Tacrolimus 175-180 albumin Canis lupus familiaris 167-170 11549851-16 2001 CONCLUSIONS: Both CsA and FK506 showed acute toxicity in MDCK cells in vitro with the effects of FK506 being less pronounced. Tacrolimus 97-102 albumin Canis lupus familiaris 18-21 11549851-17 2001 CsA and FK506 had different effects on the in vivo transport rates of the Na(+)/K(+)-ATPase and the Na(+)/K(+)/2Cl(-) cotransporter; CsA inhibited the activity of the Na(+)/K(+)-ATPase and the Na(+)/K(+)/2Cl(-) cotransporter whereas FK506 stimulated the activity of Na(+)/K(+)/2Cl(-) cotransporter. Tacrolimus 8-13 albumin Canis lupus familiaris 133-136 11549851-17 2001 CsA and FK506 had different effects on the in vivo transport rates of the Na(+)/K(+)-ATPase and the Na(+)/K(+)/2Cl(-) cotransporter; CsA inhibited the activity of the Na(+)/K(+)-ATPase and the Na(+)/K(+)/2Cl(-) cotransporter whereas FK506 stimulated the activity of Na(+)/K(+)/2Cl(-) cotransporter. Tacrolimus 233-238 albumin Canis lupus familiaris 0-3 11549851-17 2001 CsA and FK506 had different effects on the in vivo transport rates of the Na(+)/K(+)-ATPase and the Na(+)/K(+)/2Cl(-) cotransporter; CsA inhibited the activity of the Na(+)/K(+)-ATPase and the Na(+)/K(+)/2Cl(-) cotransporter whereas FK506 stimulated the activity of Na(+)/K(+)/2Cl(-) cotransporter. Tacrolimus 233-238 albumin Canis lupus familiaris 133-136 11455576-0 2001 Tacrolimus, but not cyclosporine A, significantly increases expression of ICAM-1 and IFN-gamma in the NOD mouse. Tacrolimus 0-10 intercellular adhesion molecule 1 Mus musculus 74-80 11455576-0 2001 Tacrolimus, but not cyclosporine A, significantly increases expression of ICAM-1 and IFN-gamma in the NOD mouse. Tacrolimus 0-10 interferon gamma Mus musculus 85-94 11455576-1 2001 We studied the alterations of cytokines and ICAM-1 expression in the NOD mouse pancreas produced by the administration of Cyclosporine A (CY) and Tacrolimus (TA), two widely used immunosuppressive drugs. Tacrolimus 146-156 intercellular adhesion molecule 1 Mus musculus 44-50 11455576-4 2001 On the other hand, TA-treated animals had infiltrated islets containing numerous dendritic cells, adhesion molecule overexpression, increased IFN-gamma and ICAM-1 mRNA transcripts, and interestingly, high levels of circulating ICAM-1. Tacrolimus 19-21 interferon gamma Mus musculus 142-151 11455576-4 2001 On the other hand, TA-treated animals had infiltrated islets containing numerous dendritic cells, adhesion molecule overexpression, increased IFN-gamma and ICAM-1 mRNA transcripts, and interestingly, high levels of circulating ICAM-1. Tacrolimus 19-21 intercellular adhesion molecule 1 Mus musculus 156-162 11455576-4 2001 On the other hand, TA-treated animals had infiltrated islets containing numerous dendritic cells, adhesion molecule overexpression, increased IFN-gamma and ICAM-1 mRNA transcripts, and interestingly, high levels of circulating ICAM-1. Tacrolimus 19-21 intercellular adhesion molecule 1 Mus musculus 227-233 11563819-6 2001 Tacrolimus treatment added with donor specific BMT down-regulated IL-12 and IFN-gamma transcript, resulted in a significant prolongation of intestinal allograft survival. Tacrolimus 0-10 interferon gamma Homo sapiens 76-85 11104682-7 2000 Furthermore, mobilization of calcium by A23187 and thapsigargin blocked the TNFalpha-mediated induction of RGS16, which was reversed by EGTA and by the immunosuppressants FK506 and cyclosporin A, suggesting that the calcineurin/NF-AT (nuclear factor of activated T cells) pathway may repress the up-regulation process. Tacrolimus 171-176 tumor necrosis factor Homo sapiens 76-84 11176043-1 2001 Tacrolimus, available for intravenous, oral and now topical administration, is a potent immunosuppressive agent with the ability to block the production of Interleukin-2 (IL-2) and inhibit T-cell proliferation. Tacrolimus 0-10 interleukin 2 Homo sapiens 156-169 11176043-1 2001 Tacrolimus, available for intravenous, oral and now topical administration, is a potent immunosuppressive agent with the ability to block the production of Interleukin-2 (IL-2) and inhibit T-cell proliferation. Tacrolimus 0-10 interleukin 2 Homo sapiens 171-175 11336351-6 2001 The potency of inhibition of P-gp was cyclosporine > tacrolimus > quinidine > verapamil > vinblastine. Tacrolimus 56-66 ATP binding cassette subfamily B member 1 Homo sapiens 29-33 11263551-8 2001 The Calcineurin-inhibitor Induced Pain Syndrome (CIPS) is a rare but severe side effect of cyclosporine or tacrolimus and is accurately diagnosed by its typical presentation, magnetic resonance imaging and bone scans. Tacrolimus 107-117 calcineurin binding protein 1 Homo sapiens 4-25 11137126-7 2000 Daily intramuscular treatment of GB piglets with 1mg/kg of FK506 from birth for 4 weeks resulted in lowered (P<0.05) in vitro secretion of interferon-gamma and interleukin-8. Tacrolimus 59-64 interferon gamma Homo sapiens 142-158 11137126-7 2000 Daily intramuscular treatment of GB piglets with 1mg/kg of FK506 from birth for 4 weeks resulted in lowered (P<0.05) in vitro secretion of interferon-gamma and interleukin-8. Tacrolimus 59-64 C-X-C motif chemokine ligand 8 Homo sapiens 163-176 11137126-9 2000 The depletions of mononuclear cells and low levels of interferon-gamma and interleukin-8 in piglets treated with FK506 were accompanied by lower proportion of CD3+, CD2+CD4+ and CD2+CD8+ T-cell phenotypes in peripheral blood but not in thymus and mesenteric lymph nodes. Tacrolimus 113-118 interferon gamma Homo sapiens 54-70 11137126-9 2000 The depletions of mononuclear cells and low levels of interferon-gamma and interleukin-8 in piglets treated with FK506 were accompanied by lower proportion of CD3+, CD2+CD4+ and CD2+CD8+ T-cell phenotypes in peripheral blood but not in thymus and mesenteric lymph nodes. Tacrolimus 113-118 C-X-C motif chemokine ligand 8 Homo sapiens 75-88 11104682-7 2000 Furthermore, mobilization of calcium by A23187 and thapsigargin blocked the TNFalpha-mediated induction of RGS16, which was reversed by EGTA and by the immunosuppressants FK506 and cyclosporin A, suggesting that the calcineurin/NF-AT (nuclear factor of activated T cells) pathway may repress the up-regulation process. Tacrolimus 171-176 regulator of G protein signaling 16 Homo sapiens 107-112 11326544-9 2000 According to the result using whole cell study with a patch pipette, including FK-506, an antagonist of calcineurin, the induction site of this LTD is presynaptic, which defers from conventional LTD. We have also discussed the involvement of murine protein tyrosine phosphatase (MPTP) in LTD induction in the hippocampal CA1 region by using an MPTP delta knockout mouse. Tacrolimus 79-85 carbonic anhydrase 1 Mus musculus 321-324 11078832-5 2000 By contrast, the immunosuppresant drugs cyclosporin A and FK506 abolished the effects of calcium elevation on HSF-1 activation. Tacrolimus 58-63 heat shock transcription factor 1 Homo sapiens 110-115 11062058-4 2000 Physical interactions between the RyR and calcineurin were found in the CSMF in the presence of added 100 microM Ca(2+); however, the interactions were interrupted in the presence of 20 mM EGTA, 1 microM rapamycin or 1 microM FK506, suggesting that the interaction is Ca(2+)-dependent, and is mediated by FKBP12.6. Tacrolimus 226-231 ryanodine receptor 2 Rattus norvegicus 34-37 11144010-5 2000 Thyroxin is a potent activator of the cytochrome P-450- CYP 3A enzyme system, which is crucial for tacrolimus metabolism. Tacrolimus 99-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 11144010-8 2000 In conclusion, it is important to consider thyroid function when prescribing medications with a narrow therapeutic range, which are metabolized by the cytochrome P-450 system such as tacrolimus, and the possible devastating effect of impaired drug metabolism during hypothyroidism. Tacrolimus 183-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 11258483-2 2000 Here we show that FK506-FKBP complex suppresses the activation of JNK and p38 pathways at a level upstream of mitogen-activated protein kinase (MAPK) kinase kinase (MAPKK-K) besides the calcineurin-NFAT pathway. Tacrolimus 18-23 mitogen-activated protein kinase 14 Homo sapiens 74-77 11258483-5 2000 We further demonstrate that co-expression of a constitutively active NFAT and a constitutively active MEKK1 renders the interleukin-2 promoter in Jurkat T lymphocytes resistant to CsA and FK506, whereas Jurkat cells expressing a constitutively active NFAT alone are still sensitive to CsA or FK506. Tacrolimus 188-193 mitogen-activated protein kinase kinase kinase 1 Homo sapiens 102-107 11258483-5 2000 We further demonstrate that co-expression of a constitutively active NFAT and a constitutively active MEKK1 renders the interleukin-2 promoter in Jurkat T lymphocytes resistant to CsA and FK506, whereas Jurkat cells expressing a constitutively active NFAT alone are still sensitive to CsA or FK506. Tacrolimus 188-193 interleukin 2 Homo sapiens 120-133 11258483-5 2000 We further demonstrate that co-expression of a constitutively active NFAT and a constitutively active MEKK1 renders the interleukin-2 promoter in Jurkat T lymphocytes resistant to CsA and FK506, whereas Jurkat cells expressing a constitutively active NFAT alone are still sensitive to CsA or FK506. Tacrolimus 292-297 mitogen-activated protein kinase kinase kinase 1 Homo sapiens 102-107 11258483-5 2000 We further demonstrate that co-expression of a constitutively active NFAT and a constitutively active MEKK1 renders the interleukin-2 promoter in Jurkat T lymphocytes resistant to CsA and FK506, whereas Jurkat cells expressing a constitutively active NFAT alone are still sensitive to CsA or FK506. Tacrolimus 292-297 interleukin 2 Homo sapiens 120-133 11258483-6 2000 Therefore, CsA and FK506 exert their immunosuppressive effects through targeting both the calcineurin-dependent NFAT pathway and calcineurin-independent activation pathway for JNK and p38. Tacrolimus 19-24 mitogen-activated protein kinase 14 Homo sapiens 184-187 11046013-6 2000 The induction of NFATL1 protein, as well as NFATL1-dependent transcription, is inhibited by cyclosporin A and FK506, and expression of constitutively active calcineurin induces NFATL1-dependent transcription. Tacrolimus 110-115 nuclear factor of activated T cells 5 Homo sapiens 17-23 11046013-6 2000 The induction of NFATL1 protein, as well as NFATL1-dependent transcription, is inhibited by cyclosporin A and FK506, and expression of constitutively active calcineurin induces NFATL1-dependent transcription. Tacrolimus 110-115 nuclear factor of activated T cells 5 Homo sapiens 44-50 11046013-6 2000 The induction of NFATL1 protein, as well as NFATL1-dependent transcription, is inhibited by cyclosporin A and FK506, and expression of constitutively active calcineurin induces NFATL1-dependent transcription. Tacrolimus 110-115 nuclear factor of activated T cells 5 Homo sapiens 44-50 11084058-12 2000 Preincubation with tacrolimus augmented (P <.05) the production of IL-10 in patients (n = 18), but not controls (n = 6). Tacrolimus 19-29 interleukin 10 Homo sapiens 70-75 11093160-9 2000 In untransformed human peripheral blood T lymphocytes, these phosphatases function through a cyclosporin A/FK506-resistant co-stimulatory signaling pathway which is common for the accessory receptors CD2 and CD28. Tacrolimus 107-112 CD2 molecule Homo sapiens 200-203 11093160-9 2000 In untransformed human peripheral blood T lymphocytes, these phosphatases function through a cyclosporin A/FK506-resistant co-stimulatory signaling pathway which is common for the accessory receptors CD2 and CD28. Tacrolimus 107-112 CD28 molecule Homo sapiens 208-212 11121130-0 2000 Suppressive effects of cyclosporin A and FK-506 on superoxide generation in human polymorphonuclear leukocytes primed by tumor necrosis factor alpha. Tacrolimus 41-47 tumor necrosis factor Homo sapiens 121-148 11029284-8 2000 Synergistic action of ethanol and H(2)O(2) was also observed on the purified RyR1 channel, which was free from FK506 binding protein (FKBP12). Tacrolimus 111-116 ryanodine receptor 1 Oryctolagus cuniculus 77-81 11091247-6 2000 RESULTS: The expression of messenger RNA (mRNA) for collagen III and TIMP-1 was significantly higher in patients receiving cyclosporin therapy than in those having tacrolimus (P < 0.01); this finding was accounted for by differences in the biopsy material at 1 week. Tacrolimus 164-174 TIMP metallopeptidase inhibitor 1 Homo sapiens 69-75 11120255-0 2000 FK 506 significantly improves transferred insulin gene expression in total pancreatectomized dogs. Tacrolimus 0-6 insulin Canis lupus familiaris 42-49 11131636-2 2000 The apoptosis inducing effect of interferon-gamma and its modulation by cyclosporin-A or tacrolimus (FK-506) were investigated in in vitro and ex vivo experiments. Tacrolimus 89-99 interferon gamma Homo sapiens 33-49 11119903-0 2000 ABO-incompatible living donor kidney transplantation under tacrolimus immunosuppression. Tacrolimus 59-69 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 0-3 11032725-4 2000 Binding of tacrolimus to two commercial sources of ubiquitin, a bovine product, and a recombinant human ubiquitin, was also demonstrated after HPLC purification of the purchased preparations. Tacrolimus 11-21 ubiquitin Bos taurus 51-60 11032725-4 2000 Binding of tacrolimus to two commercial sources of ubiquitin, a bovine product, and a recombinant human ubiquitin, was also demonstrated after HPLC purification of the purchased preparations. Tacrolimus 11-21 ubiquitin Bos taurus 104-113 11034951-5 2000 Treatment of DS rats with the calcineurin inhibitor FK506 (0.1 or 0.01 mg/kg twice daily) from the age of 6 weeks to 12 weeks inhibited the activation of calcineurin in the heart in a dose-dependent manner and attenuated the development of load-induced cardiac hypertrophy and fibrosis without change of hemodynamic parameters. Tacrolimus 52-57 calcineurin binding protein 1 Rattus norvegicus 30-51 11061569-1 2000 Tacrolimus (FK-506) and cyclosporin A (CsA) are calcineurin antagonists used widely as T-cell immunosuppressants; however, their relative efficacy on the production of interleukin-18 (IL-18) remains undefined. Tacrolimus 0-10 interleukin 18 Homo sapiens 168-182 11061569-1 2000 Tacrolimus (FK-506) and cyclosporin A (CsA) are calcineurin antagonists used widely as T-cell immunosuppressants; however, their relative efficacy on the production of interleukin-18 (IL-18) remains undefined. Tacrolimus 0-10 interleukin 18 Homo sapiens 184-189 11061569-1 2000 Tacrolimus (FK-506) and cyclosporin A (CsA) are calcineurin antagonists used widely as T-cell immunosuppressants; however, their relative efficacy on the production of interleukin-18 (IL-18) remains undefined. Tacrolimus 12-18 interleukin 18 Homo sapiens 168-182 11061569-1 2000 Tacrolimus (FK-506) and cyclosporin A (CsA) are calcineurin antagonists used widely as T-cell immunosuppressants; however, their relative efficacy on the production of interleukin-18 (IL-18) remains undefined. Tacrolimus 12-18 interleukin 18 Homo sapiens 184-189 11117573-8 2000 The serum TNF-alpha concentration was lower in the FK 506-treated group (1,419 +/- 957 pg/ml) than in the saline group (9205 +/- 2215) (P < 0.01). Tacrolimus 51-57 tumor necrosis factor Rattus norvegicus 10-19 11003356-8 2000 Plasma IGF-I levels were significantly elevated in FK506-treated rats but not in CsAtreated rats. Tacrolimus 51-56 insulin-like growth factor 1 Rattus norvegicus 7-12 11042265-5 2000 We studied two calcineurin inhibitors, CsA and FK506, and found that each potently inhibited TNF cytotoxicity. Tacrolimus 47-52 tumor necrosis factor-like Rattus norvegicus 93-96 11007881-3 2000 Pretreatment of animals with FK506, a potent inhibitor of calcineurin activity, or MK801, an NMDA glutamate receptor antagonist, blocked BAD dephosphorylation and abolished activation of the caspase-3 apoptotic cascade. Tacrolimus 29-34 caspase 3 Homo sapiens 191-200 11007866-0 2000 Tacrolimus/itraconazole interactions: a case report of ABO-incompatible living-related renal transplantation. Tacrolimus 0-10 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 55-58 10973631-6 2000 FK506 and cyclosporin A suppressed the chemotactic activity of the supernatant in parallel to the suppression of interleukin-8 production by peripheral blood mononuclear cells. Tacrolimus 0-5 C-X-C motif chemokine ligand 8 Homo sapiens 113-126 10973631-8 2000 These studies indicate that FK506 may exert a beneficial effect on human inflammatory diseases by suppressing neutrophil chemotaxis secondary to inhibition of chemoattractant (for example, interleukin-8) production by leukocytes. Tacrolimus 28-33 C-X-C motif chemokine ligand 8 Homo sapiens 189-202 11131636-2 2000 The apoptosis inducing effect of interferon-gamma and its modulation by cyclosporin-A or tacrolimus (FK-506) were investigated in in vitro and ex vivo experiments. Tacrolimus 101-107 interferon gamma Homo sapiens 33-49 11053632-0 2000 Switch from cyclosporine A to tacrolimus in renal transplant recipients: impact on Th1, Th2, and monokine responses. Tacrolimus 30-40 negative elongation factor complex member C/D Homo sapiens 83-86 10972232-2 2000 The mechanism of action of cyclosporine and tacrolimus has been explained on the basis of their inhibition of interleukin-2 (IL-2), and induction of transforming growth factor-beta (TGF-beta). Tacrolimus 44-54 interleukin 2 Homo sapiens 110-123 11045663-9 2000 The optimal doses of CsA and tacrolimus had similar inhibitory effects on Th1 type cytokine IL-2 and interferon [INF]-gamma), inflammatory cytokine (IL-1beta and tumor necrosis factor [TNF]-alpha), and cytotoxic factor (granzyme B and perforin) mRNA expression. Tacrolimus 29-39 interleukin 1 beta Rattus norvegicus 149-157 11045663-9 2000 The optimal doses of CsA and tacrolimus had similar inhibitory effects on Th1 type cytokine IL-2 and interferon [INF]-gamma), inflammatory cytokine (IL-1beta and tumor necrosis factor [TNF]-alpha), and cytotoxic factor (granzyme B and perforin) mRNA expression. Tacrolimus 29-39 tumor necrosis factor Rattus norvegicus 185-195 11045663-9 2000 The optimal doses of CsA and tacrolimus had similar inhibitory effects on Th1 type cytokine IL-2 and interferon [INF]-gamma), inflammatory cytokine (IL-1beta and tumor necrosis factor [TNF]-alpha), and cytotoxic factor (granzyme B and perforin) mRNA expression. Tacrolimus 29-39 granzyme B Rattus norvegicus 220-230 11045663-11 2000 Whereas IL-4 expression was not affected by tacrolimus and was enhanced by CsA, IL-10 expression was more significantly suppressed by tacrolimus than CsA. Tacrolimus 134-144 interleukin 10 Rattus norvegicus 80-85 11053632-6 2000 Treatment with Tacr resulted in a decreased expression of costimulatory ligands and adhesion molecules (T cells: CD40L, p < 0.05; CD28 and CD54, p < or = 0.01; B cells: CD25, p = 0.05; CD40, p < 0.001; monocytes: CD40, p < 0.05), which coincided with decreased PHA-stimulated T cell IL-2 responses (398 +/- 153 versus 43 +/- 15 pg/ml, p < 0.05), impaired CD4 helper activity (117% +/- 22% versus 73% +/- 19%, p < 0.05) and increased CD4 suppressor activity (-120% +/- 28% versus -18% +/- 27%, p = 0.02). Tacrolimus 15-19 CD40 ligand Homo sapiens 113-118 11053632-6 2000 Treatment with Tacr resulted in a decreased expression of costimulatory ligands and adhesion molecules (T cells: CD40L, p < 0.05; CD28 and CD54, p < or = 0.01; B cells: CD25, p = 0.05; CD40, p < 0.001; monocytes: CD40, p < 0.05), which coincided with decreased PHA-stimulated T cell IL-2 responses (398 +/- 153 versus 43 +/- 15 pg/ml, p < 0.05), impaired CD4 helper activity (117% +/- 22% versus 73% +/- 19%, p < 0.05) and increased CD4 suppressor activity (-120% +/- 28% versus -18% +/- 27%, p = 0.02). Tacrolimus 15-19 CD28 molecule Homo sapiens 133-137 11053632-6 2000 Treatment with Tacr resulted in a decreased expression of costimulatory ligands and adhesion molecules (T cells: CD40L, p < 0.05; CD28 and CD54, p < or = 0.01; B cells: CD25, p = 0.05; CD40, p < 0.001; monocytes: CD40, p < 0.05), which coincided with decreased PHA-stimulated T cell IL-2 responses (398 +/- 153 versus 43 +/- 15 pg/ml, p < 0.05), impaired CD4 helper activity (117% +/- 22% versus 73% +/- 19%, p < 0.05) and increased CD4 suppressor activity (-120% +/- 28% versus -18% +/- 27%, p = 0.02). Tacrolimus 15-19 intercellular adhesion molecule 1 Homo sapiens 142-146 11053632-6 2000 Treatment with Tacr resulted in a decreased expression of costimulatory ligands and adhesion molecules (T cells: CD40L, p < 0.05; CD28 and CD54, p < or = 0.01; B cells: CD25, p = 0.05; CD40, p < 0.001; monocytes: CD40, p < 0.05), which coincided with decreased PHA-stimulated T cell IL-2 responses (398 +/- 153 versus 43 +/- 15 pg/ml, p < 0.05), impaired CD4 helper activity (117% +/- 22% versus 73% +/- 19%, p < 0.05) and increased CD4 suppressor activity (-120% +/- 28% versus -18% +/- 27%, p = 0.02). Tacrolimus 15-19 interleukin 2 receptor subunit alpha Homo sapiens 175-179 11053632-6 2000 Treatment with Tacr resulted in a decreased expression of costimulatory ligands and adhesion molecules (T cells: CD40L, p < 0.05; CD28 and CD54, p < or = 0.01; B cells: CD25, p = 0.05; CD40, p < 0.001; monocytes: CD40, p < 0.05), which coincided with decreased PHA-stimulated T cell IL-2 responses (398 +/- 153 versus 43 +/- 15 pg/ml, p < 0.05), impaired CD4 helper activity (117% +/- 22% versus 73% +/- 19%, p < 0.05) and increased CD4 suppressor activity (-120% +/- 28% versus -18% +/- 27%, p = 0.02). Tacrolimus 15-19 CD40 molecule Homo sapiens 113-117 11053632-6 2000 Treatment with Tacr resulted in a decreased expression of costimulatory ligands and adhesion molecules (T cells: CD40L, p < 0.05; CD28 and CD54, p < or = 0.01; B cells: CD25, p = 0.05; CD40, p < 0.001; monocytes: CD40, p < 0.05), which coincided with decreased PHA-stimulated T cell IL-2 responses (398 +/- 153 versus 43 +/- 15 pg/ml, p < 0.05), impaired CD4 helper activity (117% +/- 22% versus 73% +/- 19%, p < 0.05) and increased CD4 suppressor activity (-120% +/- 28% versus -18% +/- 27%, p = 0.02). Tacrolimus 15-19 CD40 molecule Homo sapiens 191-195 11053632-6 2000 Treatment with Tacr resulted in a decreased expression of costimulatory ligands and adhesion molecules (T cells: CD40L, p < 0.05; CD28 and CD54, p < or = 0.01; B cells: CD25, p = 0.05; CD40, p < 0.001; monocytes: CD40, p < 0.05), which coincided with decreased PHA-stimulated T cell IL-2 responses (398 +/- 153 versus 43 +/- 15 pg/ml, p < 0.05), impaired CD4 helper activity (117% +/- 22% versus 73% +/- 19%, p < 0.05) and increased CD4 suppressor activity (-120% +/- 28% versus -18% +/- 27%, p = 0.02). Tacrolimus 15-19 interleukin 2 Homo sapiens 295-299 11053632-6 2000 Treatment with Tacr resulted in a decreased expression of costimulatory ligands and adhesion molecules (T cells: CD40L, p < 0.05; CD28 and CD54, p < or = 0.01; B cells: CD25, p = 0.05; CD40, p < 0.001; monocytes: CD40, p < 0.05), which coincided with decreased PHA-stimulated T cell IL-2 responses (398 +/- 153 versus 43 +/- 15 pg/ml, p < 0.05), impaired CD4 helper activity (117% +/- 22% versus 73% +/- 19%, p < 0.05) and increased CD4 suppressor activity (-120% +/- 28% versus -18% +/- 27%, p = 0.02). Tacrolimus 15-19 CD4 molecule Homo sapiens 113-116 11053632-6 2000 Treatment with Tacr resulted in a decreased expression of costimulatory ligands and adhesion molecules (T cells: CD40L, p < 0.05; CD28 and CD54, p < or = 0.01; B cells: CD25, p = 0.05; CD40, p < 0.001; monocytes: CD40, p < 0.05), which coincided with decreased PHA-stimulated T cell IL-2 responses (398 +/- 153 versus 43 +/- 15 pg/ml, p < 0.05), impaired CD4 helper activity (117% +/- 22% versus 73% +/- 19%, p < 0.05) and increased CD4 suppressor activity (-120% +/- 28% versus -18% +/- 27%, p = 0.02). Tacrolimus 15-19 CD4 molecule Homo sapiens 191-194 11053632-8 2000 Our data show that switching of immunosuppressive therapy from CsA to tacrolimus results in suppression of costimulatory ligands, adhesion molecules, Th1 responses and CD4 helper activity. Tacrolimus 70-80 negative elongation factor complex member C/D Homo sapiens 150-153 11053632-8 2000 Our data show that switching of immunosuppressive therapy from CsA to tacrolimus results in suppression of costimulatory ligands, adhesion molecules, Th1 responses and CD4 helper activity. Tacrolimus 70-80 CD4 molecule Homo sapiens 168-171 10972232-2 2000 The mechanism of action of cyclosporine and tacrolimus has been explained on the basis of their inhibition of interleukin-2 (IL-2), and induction of transforming growth factor-beta (TGF-beta). Tacrolimus 44-54 transforming growth factor beta 1 Homo sapiens 149-180 10972232-2 2000 The mechanism of action of cyclosporine and tacrolimus has been explained on the basis of their inhibition of interleukin-2 (IL-2), and induction of transforming growth factor-beta (TGF-beta). Tacrolimus 44-54 transforming growth factor beta 1 Homo sapiens 182-190 10972232-5 2000 RESULTS: In this study, we report that rapamycin in combination with either tacrolimus or cyclosporine significantly inhibited the lymphocyte proliferation, IL-2 expression, and induced TGF-beta, compared with these drugs alone. Tacrolimus 76-86 transforming growth factor beta 1 Homo sapiens 186-194 10949194-6 2000 The modifying effect of cyclosporine (CsA) and FK506 on CD40L gene expression was further tested in vitro in CD4+ T lymphocytes separated from pokeweed mitogen- (PWM) activated peripheral blood lymphocytes (PBL) preparations. Tacrolimus 47-52 CD40 ligand Homo sapiens 56-61 10949194-9 2000 After in vitro activation, CD40L gene expression increased by approximately 4-fold and the addition of CsA or FK506 diminished CD40L gene expression to a base level. Tacrolimus 110-115 CD40 ligand Homo sapiens 127-132 10949194-10 2000 CONCLUSION: Peripheral CD4+ T cell CD40L gene expression increases significantly in acute rejection and CAN and may serve as a non-invasive method to monitor allograft function and determine the biological response to CsA and FK506. Tacrolimus 226-231 CD40 ligand Homo sapiens 35-40 10928971-0 2000 FK506 potently inhibits T cell activation induced TNF-alpha and IL-1beta production in vitro by human peripheral blood mononuclear cells. Tacrolimus 0-5 tumor necrosis factor Homo sapiens 50-59 10961655-1 2000 We compared in rats with 6-hydroxydopamine (6-OHDA)-induced hemiparkinsonism the content of tumor necrosis factor (TNF)-alpha in the nigrostriatal dopaminergic region of the control side with that of the 6-OHDA-injected experimental side, and explored the effects of 6-OHDA injection combined with the immunosuppressant FK506 treatment (0.5 or 4 mg/kg per day for 2 weeks). Tacrolimus 320-325 tumor necrosis factor Rattus norvegicus 92-125 10961655-2 2000 The ratios of the concentration of TNF-alpha in the striatum and substantia nigra on the 6-OHDA injection side to that on the control side in the 6-OHDA hemiparkinsonism rats were significantly higher than those in the control rats without 6-OHDA treatment, whereas those in the rats treated with 6-OHDA and FK506 were not significantly different from those in the control rats. Tacrolimus 308-313 tumor necrosis factor Rattus norvegicus 35-44 10961655-3 2000 Thus FK506 attenuated increased TNF-alpha level in the nigrostriatal dopaminergic region injured by 6-OHDA treatment. Tacrolimus 5-10 tumor necrosis factor Rattus norvegicus 32-41 10928971-0 2000 FK506 potently inhibits T cell activation induced TNF-alpha and IL-1beta production in vitro by human peripheral blood mononuclear cells. Tacrolimus 0-5 interleukin 1 beta Homo sapiens 64-72 10928971-1 2000 The aim of this study was to elucidate the in vitro inhibitory potency of FK506 on production of the inflammatory cytokines, tumour necrosis factor (TNF)-alpha and interleukin (IL)-1beta, with a view to assessing this immunosuppressive agent as a potential anti-rheumatic drug. Tacrolimus 74-79 tumor necrosis factor Homo sapiens 125-159 10928971-1 2000 The aim of this study was to elucidate the in vitro inhibitory potency of FK506 on production of the inflammatory cytokines, tumour necrosis factor (TNF)-alpha and interleukin (IL)-1beta, with a view to assessing this immunosuppressive agent as a potential anti-rheumatic drug. Tacrolimus 74-79 interleukin 1 beta Homo sapiens 164-186 10928971-4 2000 FK506 inhibited anti-CD3/CD28 induced TNF-alpha and IL-1beta production at concentrations less than 1 ng ml(-1). Tacrolimus 0-5 CD28 molecule Homo sapiens 25-29 10928971-4 2000 FK506 inhibited anti-CD3/CD28 induced TNF-alpha and IL-1beta production at concentrations less than 1 ng ml(-1). Tacrolimus 0-5 tumor necrosis factor Homo sapiens 38-47 10928971-4 2000 FK506 inhibited anti-CD3/CD28 induced TNF-alpha and IL-1beta production at concentrations less than 1 ng ml(-1). Tacrolimus 0-5 interleukin 1 beta Homo sapiens 52-60 10928971-5 2000 Flow cytometric analysis of intracellular TNF-alpha and IL-1beta positive cells showed that FK506 potently suppresses inflammatory cytokine production from CD14+ monocytes as well as from T cells. Tacrolimus 92-97 tumor necrosis factor Homo sapiens 42-51 10928971-5 2000 Flow cytometric analysis of intracellular TNF-alpha and IL-1beta positive cells showed that FK506 potently suppresses inflammatory cytokine production from CD14+ monocytes as well as from T cells. Tacrolimus 92-97 interleukin 1 beta Homo sapiens 56-64 10928971-5 2000 Flow cytometric analysis of intracellular TNF-alpha and IL-1beta positive cells showed that FK506 potently suppresses inflammatory cytokine production from CD14+ monocytes as well as from T cells. Tacrolimus 92-97 CD14 molecule Homo sapiens 156-160 10928971-7 2000 FK506 and CsA, but not DEX, specifically inhibited anti-CD3/CD28 induced inflammatory cytokine production without affecting the lipopolysaccaride (LPS) induced effect. Tacrolimus 0-5 CD28 molecule Homo sapiens 60-64 11030448-0 2000 Differential transcriptional regulation of endothelin-1 by immunosuppressants FK506 and cyclosporin A. Tacrolimus 78-83 endothelin 1 Rattus norvegicus 43-55 10899116-3 2000 We show that CBP1 binds to calcineurin in vitro and in vivo, and FKBP12-FK506 inhibits CBP1 binding to calcineurin. Tacrolimus 72-77 serpin family H member 1 Homo sapiens 87-91 10945321-0 2000 Effect of intestinal P-glycoprotein on daily tacrolimus trough level in a living-donor small bowel recipient. Tacrolimus 45-55 ATP binding cassette subfamily B member 1 Homo sapiens 21-35 10945321-1 2000 We have examined whether the expression levels of the intestinal absorptive barriers, MDR1 gene product P-glycoprotein and cytochrome P450 IIIA4 (CYP3A4), correlate with the trough levels of orally administered tacrolimus in a recipient of small bowel transplant for 4 months. Tacrolimus 211-221 ATP binding cassette subfamily B member 1 Homo sapiens 86-90 10945321-1 2000 We have examined whether the expression levels of the intestinal absorptive barriers, MDR1 gene product P-glycoprotein and cytochrome P450 IIIA4 (CYP3A4), correlate with the trough levels of orally administered tacrolimus in a recipient of small bowel transplant for 4 months. Tacrolimus 211-221 ATP binding cassette subfamily B member 1 Homo sapiens 104-118 10945321-5 2000 The tacrolimus concentration/dose ratio correlated well with the mRNA expression level of MDR1, but not CYP3A4. Tacrolimus 4-14 ATP binding cassette subfamily B member 1 Homo sapiens 90-94 10945321-6 2000 These results suggested that intestinal P-glycoprotein rather than CYP3A4 is a good probe to predict the intraindividual variation in the tacrolimus pharmacokinetics during immunosuppressant therapy after small bowel transplantation. Tacrolimus 138-148 ATP binding cassette subfamily B member 1 Homo sapiens 40-54 11030448-6 2000 Likewise, luciferase assays demonstrate stronger ET-1 promoter-dependent stimulation of the reporter gene by CsA than by FK506. Tacrolimus 121-126 endothelin 1 Rattus norvegicus 49-53 11030448-9 2000 These observations demonstrate that calcineurin antagonists FK506 and CsA differ in quality to induce transcription of prepro ET-1 in HUVEC via calcium-dependent nuclear signalling events. Tacrolimus 60-65 endothelin 1 Rattus norvegicus 126-130 10931176-2 2000 A singular FK506-like binding domain (FKBD) has about 12 kDa and occurs in the form of archetypal FKBP-12 and as a part of different proteins ranging in size from 13 to 135 kDa. Tacrolimus 11-16 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 98-105 10931176-7 2000 These variations at the sequence positions which are crucial for binding the immunosuppressive macrolide FK506 and peptidyl-prolyl cis/trans isomerase (PPIase) activity are small. Tacrolimus 105-110 FKBP prolyl isomerase like Homo sapiens 152-158 10884573-0 2000 Rapamycin and FK506 induce long-term potentiation by pairing stimulation via an intracellular Ca(2+) signaling mechanism in rat hippocampal CA1 neurons. Tacrolimus 14-19 carbonic anhydrase 1 Rattus norvegicus 140-143 18034554-3 2000 Tacrolimus is a macrolide lactone that inhibits the signal transduction of interleukin-2 (IL-2) via calcineurin inhibition. Tacrolimus 0-10 interleukin 2 Homo sapiens 75-88 18034554-3 2000 Tacrolimus is a macrolide lactone that inhibits the signal transduction of interleukin-2 (IL-2) via calcineurin inhibition. Tacrolimus 0-10 interleukin 2 Homo sapiens 90-94 10947839-6 2000 Inhibition of PP-2B (calcineurin) with cyclosporin A or FK506 reduced Cx43 dephosphorylation and junctional uncoupling seen after hypoxia. Tacrolimus 56-61 gap junction protein alpha 1 Homo sapiens 70-74 10861270-9 2000 However, early withdrawal of FK506 reversed the immunosuppressive effect of anti-LFA-1 treatment. Tacrolimus 29-34 integrin alpha L Mus musculus 81-86 11030448-1 2000 Calcineurin antagonists FK506 and CsA, administered to treat organ allograft rejection, exert specific effects on renal vasoconstriction and nephrotoxicity, possibly due to endogenous vasoconstrictor release such as ET-1. Tacrolimus 24-29 endothelin 1 Rattus norvegicus 216-220 11030448-2 2000 We investigated contribution of FK506 and CsA on regulation of prepro ET-1 gene transcription in HUVEC. Tacrolimus 32-37 endothelin 1 Rattus norvegicus 70-74 11030448-5 2000 Northern blot analysis shows differential induction of prepro ET-1 mRNA in favour of CsA over FK506. Tacrolimus 94-99 endothelin 1 Rattus norvegicus 62-66 10887335-5 2000 RESULTS: Dexamethasone, FK506, and cyclosporin A suppressed the production of IL-2, IL-4, and IL-5 by human helper T cells, which shows a similar concentration-response relationship in each case. Tacrolimus 24-29 interleukin 2 Homo sapiens 78-82 10887335-5 2000 RESULTS: Dexamethasone, FK506, and cyclosporin A suppressed the production of IL-2, IL-4, and IL-5 by human helper T cells, which shows a similar concentration-response relationship in each case. Tacrolimus 24-29 interleukin 4 Homo sapiens 84-88 10887335-5 2000 RESULTS: Dexamethasone, FK506, and cyclosporin A suppressed the production of IL-2, IL-4, and IL-5 by human helper T cells, which shows a similar concentration-response relationship in each case. Tacrolimus 24-29 interleukin 5 Homo sapiens 94-98 10846105-10 2000 The mice with the Fv-4(r/r) genotype treated with FK506 still showed resistance, but the mice with the Fv-4(r/-) genotype became highly sensitive to F-MuLV infection. Tacrolimus 50-55 Friend virus susceptibility 4 Mus musculus 18-22 10861056-0 2000 Microchimerism, donor dendritic cells, and alloimmune reactivity in recipients of Flt3 ligand-mobilized hemopoietic cells: modulation by tacrolimus. Tacrolimus 137-147 fms related receptor tyrosine kinase 3 ligand Homo sapiens 82-93 10861056-9 2000 In vitro, tacrolimus suppressed GM-CSF-stimulated growth of myeloid DC from normal BM much more effectively than from FL-BM without affecting MHC class II or costimulatory molecule expression. Tacrolimus 10-20 colony stimulating factor 2 Homo sapiens 32-38 10846105-12 2000 The Fv-4 gene product was also detected on the cells from the FK506-treated mice as well as on those from untreated mice. Tacrolimus 62-67 Friend virus susceptibility 4 Mus musculus 4-8 10839803-4 2000 An inhibitor of calcineurin activation, FK506, inhibited the in vitro anti-TCR-induced Th2 cell generation in a dose-dependent manner. Tacrolimus 40-45 T cell receptor alpha variable 6-3 Mus musculus 75-78 10856121-1 2000 The immunophilin FKBP12 associates with intracellular Ca2+ channels and this interaction can be disrupted by the immunosuppressant FK506. Tacrolimus 131-136 FKBP prolyl isomerase 1A Homo sapiens 4-23 10856121-10 2000 FK506 (10 microM) did not affect IP3-induced Ca2+ release in permeabilized SH-SY5Y and A7r5 cells, but enhanced caffeine-induced Ca2+ release via the ryanodine receptor (RyR) in differentiated BC3H1 cells. Tacrolimus 0-5 ryanodine receptor 2 Rattus norvegicus 150-168 10856121-10 2000 FK506 (10 microM) did not affect IP3-induced Ca2+ release in permeabilized SH-SY5Y and A7r5 cells, but enhanced caffeine-induced Ca2+ release via the ryanodine receptor (RyR) in differentiated BC3H1 cells. Tacrolimus 0-5 ryanodine receptor 2 Rattus norvegicus 170-173 10856121-11 2000 In conclusion, FK506 inhibited active Ca2+ uptake by the SERCA Ca2+ pump; in addition, FK506 enhanced intracellular Ca2+ release through the RyR, but it had no direct effect on IP3-induced Ca2+ release. Tacrolimus 87-92 ryanodine receptor 1 Homo sapiens 141-144 10870125-0 2000 Tacrolimus (FK 506) induced thrombotic thrombocytopenic purpura after ABO mismatched second liver transplantation: salvage with plasmapheresis and prostacyclin. Tacrolimus 12-18 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 70-73 10870125-0 2000 Tacrolimus (FK 506) induced thrombotic thrombocytopenic purpura after ABO mismatched second liver transplantation: salvage with plasmapheresis and prostacyclin. Tacrolimus 0-10 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 70-73 10870125-1 2000 We report the course of thrombotic thrombocytopenic purpura (TTP) in a patient receiving tacrolimus (FK506) immunosuppression for an ABO mismatched second liver graft. Tacrolimus 89-99 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 133-136 10870125-1 2000 We report the course of thrombotic thrombocytopenic purpura (TTP) in a patient receiving tacrolimus (FK506) immunosuppression for an ABO mismatched second liver graft. Tacrolimus 101-106 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 133-136 10851085-4 2000 Furthermore, we demonstrated that the inhibition of the phosphatase activity of calcineurin with FK506 and cyclosporin A resulted in an overall increase in cdk4 kinase activity, suggesting that the phosphatase activity of calcineurin was inhibitory to the kinase activity of cdk4. Tacrolimus 97-102 cyclin dependent kinase 4 Homo sapiens 156-160 10831643-10 2000 There was a significant reduction of the insulin first-phase concentrations, both after 1 min and after 3 min in the FK506 group compared with the CsA group (112+/-17 vs 237+/-57 microU/ml, P=0.034). Tacrolimus 117-122 insulin Homo sapiens 41-48 10831643-11 2000 In patients with repetitive IVGTTs, glucose constant decay and insulin production improved after lowering FK506 whole-blood trough levels. Tacrolimus 106-111 insulin Homo sapiens 63-70 10830164-2 2000 The channel is a tetramer comprised of four type 2 RyR polypeptides (RyR2) and four FK506 binding proteins (FKBP12.6). Tacrolimus 84-89 FKBP prolyl isomerase 1B Homo sapiens 108-116 12548975-3 2000 However, their analog, the immunosupressant FK-506, can inhibit the proliferation of fibroblast PBL1 without interfering with the activities of P90RSK, P70S6K and MAPK. Tacrolimus 44-50 ribosomal protein S6 kinase A1 Homo sapiens 144-150 12548975-3 2000 However, their analog, the immunosupressant FK-506, can inhibit the proliferation of fibroblast PBL1 without interfering with the activities of P90RSK, P70S6K and MAPK. Tacrolimus 44-50 ribosomal protein S6 kinase B1 Homo sapiens 152-158 10809730-7 2000 Drug resistance of sit4 mutants was shown to be mediated by ABC transporters as efflux of the anionic fluorescent dye rhodamine 6G, a substrate for the Pdr5-type pump, is markedly increased in sit4 mutants in an energy-dependent and FK506-sensitive manner. Tacrolimus 233-238 type 2A-related serine/threonine-protein phosphatase SIT4 Saccharomyces cerevisiae S288C 19-23 10809730-7 2000 Drug resistance of sit4 mutants was shown to be mediated by ABC transporters as efflux of the anionic fluorescent dye rhodamine 6G, a substrate for the Pdr5-type pump, is markedly increased in sit4 mutants in an energy-dependent and FK506-sensitive manner. Tacrolimus 233-238 type 2A-related serine/threonine-protein phosphatase SIT4 Saccharomyces cerevisiae S288C 193-197 10779414-0 2000 Potentiation of CD3-induced expression of the linker for activation of T cells (LAT) by the calcineurin inhibitors cyclosporin A and FK506. Tacrolimus 133-138 linker for activation of T cells Homo sapiens 80-83 10775131-5 2000 Rapamycin, an inhibitor of the mammalian target of rapamycin (mTOR), and wortmannin, a phosphatidylinositol 3-kinase inhibitor, blocked flow-induced pp70(S6k) activation; FK-506, a rapamycin analog with minimal mTOR inhibitory activity, and PD-98059, an inhibitor of the flow-sensitive mitogen-activated protein kinase pathway, had no effect. Tacrolimus 171-177 mechanistic target of rapamycin kinase Homo sapiens 31-60 10775131-5 2000 Rapamycin, an inhibitor of the mammalian target of rapamycin (mTOR), and wortmannin, a phosphatidylinositol 3-kinase inhibitor, blocked flow-induced pp70(S6k) activation; FK-506, a rapamycin analog with minimal mTOR inhibitory activity, and PD-98059, an inhibitor of the flow-sensitive mitogen-activated protein kinase pathway, had no effect. Tacrolimus 171-177 mechanistic target of rapamycin kinase Homo sapiens 62-66 10781282-3 2000 The immunosuppressant drugs FK506 (tacrolimus) and rapamycin can promote dissociation of FK506 binding protein from the ryanodine receptor 1 and by this mechanism increase sensitivity of ryanodine receptor 1 to agonists such as caffeine. Tacrolimus 28-33 ryanodine receptor 1 Homo sapiens 120-140 10781282-3 2000 The immunosuppressant drugs FK506 (tacrolimus) and rapamycin can promote dissociation of FK506 binding protein from the ryanodine receptor 1 and by this mechanism increase sensitivity of ryanodine receptor 1 to agonists such as caffeine. Tacrolimus 28-33 ryanodine receptor 1 Homo sapiens 187-207 10781282-3 2000 The immunosuppressant drugs FK506 (tacrolimus) and rapamycin can promote dissociation of FK506 binding protein from the ryanodine receptor 1 and by this mechanism increase sensitivity of ryanodine receptor 1 to agonists such as caffeine. Tacrolimus 35-45 ryanodine receptor 1 Homo sapiens 120-140 10781282-3 2000 The immunosuppressant drugs FK506 (tacrolimus) and rapamycin can promote dissociation of FK506 binding protein from the ryanodine receptor 1 and by this mechanism increase sensitivity of ryanodine receptor 1 to agonists such as caffeine. Tacrolimus 35-45 ryanodine receptor 1 Homo sapiens 187-207 10779414-6 2000 Strikingly, the calcineurin inhibitors cyclosporin A (CsA) and FK506 strongly potentiated TCR-induced LAT expression, suggesting that the activation of calcineurin following TCR ligation negatively regulates LAT expression. Tacrolimus 63-68 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 90-93 10779414-6 2000 Strikingly, the calcineurin inhibitors cyclosporin A (CsA) and FK506 strongly potentiated TCR-induced LAT expression, suggesting that the activation of calcineurin following TCR ligation negatively regulates LAT expression. Tacrolimus 63-68 linker for activation of T cells Homo sapiens 102-105 10779414-6 2000 Strikingly, the calcineurin inhibitors cyclosporin A (CsA) and FK506 strongly potentiated TCR-induced LAT expression, suggesting that the activation of calcineurin following TCR ligation negatively regulates LAT expression. Tacrolimus 63-68 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 174-177 10779414-6 2000 Strikingly, the calcineurin inhibitors cyclosporin A (CsA) and FK506 strongly potentiated TCR-induced LAT expression, suggesting that the activation of calcineurin following TCR ligation negatively regulates LAT expression. Tacrolimus 63-68 linker for activation of T cells Homo sapiens 208-211 10779414-8 2000 CsA and FK506 blocked the Ca(++ )ionophores" inhibitory effect on LAT expression. Tacrolimus 8-13 linker for activation of T cells Homo sapiens 66-69 10779414-9 2000 Notably, CsA and FK506 preferentially up-regulated TCR-induced LAT expression; under the same conditions, these compounds did not increase the expression of 14 other molecules that previously had been implicated in T-cell activation. Tacrolimus 17-22 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 51-54 10779414-9 2000 Notably, CsA and FK506 preferentially up-regulated TCR-induced LAT expression; under the same conditions, these compounds did not increase the expression of 14 other molecules that previously had been implicated in T-cell activation. Tacrolimus 17-22 linker for activation of T cells Homo sapiens 63-66 10779414-11 2000 Furthermore, the potentiation of TCR-induced LAT expression by CsA and FK506 suggests that the action of these agents involves up-regulating the cellular level of critical signaling molecules. Tacrolimus 71-76 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 33-36 10779414-11 2000 Furthermore, the potentiation of TCR-induced LAT expression by CsA and FK506 suggests that the action of these agents involves up-regulating the cellular level of critical signaling molecules. Tacrolimus 71-76 linker for activation of T cells Homo sapiens 45-48 10816008-4 2000 The crystal structures of PP1c and PP2B (calcineurin) heterotetramer calcineurinA x calcineurinB x FKBP x FK506 have been determined. Tacrolimus 106-111 protein phosphatase 1 catalytic subunit gamma Homo sapiens 26-30 15602799-1 2000 P-glycoprotein (P-gp) is a membrane efflux pump increasing the transport of drugs such as tacrolimus out of the cells. Tacrolimus 90-100 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 15602799-1 2000 P-glycoprotein (P-gp) is a membrane efflux pump increasing the transport of drugs such as tacrolimus out of the cells. Tacrolimus 90-100 ATP binding cassette subfamily B member 1 Homo sapiens 16-20 15602799-0 2000 Low levels of lymphocyte MDR1 gene expression during early renal transplantation in patients treated with tacrolimus. Tacrolimus 106-116 ATP binding cassette subfamily B member 1 Homo sapiens 25-29 15602799-2 2000 The aim of the study was to determine the kinetics of lymphocyte P-gp expression in patients treated with tacrolimus during the first 3 months following renal transplantation. Tacrolimus 106-116 ATP binding cassette subfamily B member 1 Homo sapiens 65-69 10884973-0 2000 Effects of tacrolimus hydrate (FK-506) on the expression of tissue factor in THP-1 human monocyte cell line. Tacrolimus 11-21 coagulation factor III, tissue factor Homo sapiens 60-73 10796896-8 2000 Increased staining intensity and frequency of NF-kappaB-positive cells were similarly observed in HCV-positive allografts obtained from patients under tacrolimus- compared with cyclosporine-based immunosuppression. Tacrolimus 151-161 nuclear factor kappa B subunit 1 Homo sapiens 49-55 10884973-0 2000 Effects of tacrolimus hydrate (FK-506) on the expression of tissue factor in THP-1 human monocyte cell line. Tacrolimus 31-37 coagulation factor III, tissue factor Homo sapiens 60-73 10878284-11 2000 Cyclosporine and FK-506 (Tacrolimus) inhibit the phosphatase activity of calcineurin, thereby suppressing the production of IL-2 and other cytokines. Tacrolimus 17-23 interleukin 2 Homo sapiens 124-128 10878284-11 2000 Cyclosporine and FK-506 (Tacrolimus) inhibit the phosphatase activity of calcineurin, thereby suppressing the production of IL-2 and other cytokines. Tacrolimus 25-35 interleukin 2 Homo sapiens 124-128 10884973-4 2000 FK-506 exerted a concentration-dependent inhibitory effect on LPS (10 micrograms/ml) induction of procoagulant activity, identified as TF activity as judged from immunostaining of TF antigen and by functional characterization with the use of coagulation factor VII-deficient plasma and an antibody against human TF. Tacrolimus 0-6 coagulation factor III, tissue factor Homo sapiens 135-137 10884973-4 2000 FK-506 exerted a concentration-dependent inhibitory effect on LPS (10 micrograms/ml) induction of procoagulant activity, identified as TF activity as judged from immunostaining of TF antigen and by functional characterization with the use of coagulation factor VII-deficient plasma and an antibody against human TF. Tacrolimus 0-6 coagulation factor III, tissue factor Homo sapiens 180-182 10884973-4 2000 FK-506 exerted a concentration-dependent inhibitory effect on LPS (10 micrograms/ml) induction of procoagulant activity, identified as TF activity as judged from immunostaining of TF antigen and by functional characterization with the use of coagulation factor VII-deficient plasma and an antibody against human TF. Tacrolimus 0-6 coagulation factor III, tissue factor Homo sapiens 180-182 10884973-5 2000 In addition, the reverse transcription polymerase chain reaction demonstrated reduced expression of TF mRNA in LPS-stimulated THP-1 cells exposed to FK-506. Tacrolimus 149-155 coagulation factor III, tissue factor Homo sapiens 100-102 10884973-6 2000 Thus, FK-506 acts favorably not only as a direct immunomodulating agent but also as an alleviator of local activation of the coagulation cascade contributing to transplant arteriopathy through modulation of monocyte expression of TF. Tacrolimus 6-12 coagulation factor III, tissue factor Homo sapiens 230-232 10845669-8 2000 The effect of FK506 treatment on NK1.1+ LAL was tested by cell-sorting and cytotoxicity assay. Tacrolimus 14-19 killer cell lectin-like receptor subfamily B member 1C Mus musculus 33-38 10845669-10 2000 RESULTS: Both FK506 treatment and oral tolerance induced a significant increase in NK1.1+ LAL number and cytotoxicity function. Tacrolimus 14-19 killer cell lectin-like receptor subfamily B member 1C Mus musculus 83-88 10845669-13 2000 CONCLUSIONS: This study shows for the first time that immune tolerance induced by both oral administration of an antigen and by FK506 treatment may be mediated via enhancement of NK1.1+ LAL. Tacrolimus 128-133 killer cell lectin-like receptor subfamily B member 1C Mus musculus 179-184 10836388-0 2000 Comparison between tacrolimus and cyclosporine as immunosuppressive agents compatible with tolerance induction by CD4/CD8 blockade. Tacrolimus 19-29 CD4 antigen Mus musculus 114-117 10767777-3 2000 SDZ-RAD has a molecular mass of 957.57 Da (C53H83NO14) and shares the same common intracellular receptor as tacrolimus, the FK-506 binding protein (FKBP-12). Tacrolimus 108-118 FKBP prolyl isomerase like Sus scrofa 124-146 10798763-6 2000 FK506 and MMF were also able to upregulate IL-8, although the effect was less dramatic than observed for CsA. Tacrolimus 0-5 C-X-C motif chemokine ligand 8 Homo sapiens 43-47 10798763-6 2000 FK506 and MMF were also able to upregulate IL-8, although the effect was less dramatic than observed for CsA. Tacrolimus 0-5 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 105-108 10798785-0 2000 STAT 6 up-regulation by FK506 in the presence of interleukin-4. Tacrolimus 24-29 signal transducer and activator of transcription 6 Mus musculus 0-6 10798785-0 2000 STAT 6 up-regulation by FK506 in the presence of interleukin-4. Tacrolimus 24-29 interleukin 4 Mus musculus 49-62 10798785-5 2000 We ask if FK506 biases STAT activation toward a TH2-type response. Tacrolimus 10-15 heart and neural crest derivatives expressed 2 Mus musculus 48-51 10798785-7 2000 RESULTS: Interleukin-4 specifically induced activation of STAT6, and pretreatment with FK506 enhanced this activity. Tacrolimus 87-92 signal transducer and activator of transcription 6 Mus musculus 58-63 10798785-9 2000 CONCLUSION: FK506 protects against allograft rejection by inhibiting interleukin-2 production. Tacrolimus 12-17 interleukin 2 Mus musculus 69-82 10798785-10 2000 Such protection may be enhanced by FK506-mediated up-regulation of STAT6 activity. Tacrolimus 35-40 signal transducer and activator of transcription 6 Mus musculus 67-72 10752948-5 2000 By gavage, animals were given 5 mg/kg per day of the FKBP12 ligand FK506 in sterile phosphate-buffered saline (PBS) or in PBS alone. Tacrolimus 67-72 FKBP prolyl isomerase 1A Rattus norvegicus 53-59 10752948-11 2000 In addition, the FKBP12 ligand FK506 confers neuroprotection on RGCs after optic nerve crush. Tacrolimus 31-36 FKBP prolyl isomerase 1A Rattus norvegicus 17-23 10755567-0 2000 TGF-beta expression in renal transplant biopsies: a comparative study between cyclosporin-A and tacrolimus. Tacrolimus 96-106 transforming growth factor beta 1 Homo sapiens 0-8 10755567-4 2000 The current study was designed to measure the expression of TGF-beta(b) in renal transplant biopsy specimens from patients undergoing immunosuppressive therapy with either CsA or tacrolimus (FK506). Tacrolimus 179-189 transforming growth factor beta 1 Homo sapiens 60-68 10755567-4 2000 The current study was designed to measure the expression of TGF-beta(b) in renal transplant biopsy specimens from patients undergoing immunosuppressive therapy with either CsA or tacrolimus (FK506). Tacrolimus 191-196 transforming growth factor beta 1 Homo sapiens 60-68 10710358-7 2000 Both FK-506 and rapamycin are known to target the immunophilin FKBP12. Tacrolimus 5-11 FK506 binding protein 1a Mus musculus 50-69 10685002-7 2000 FK506 as well as steroids inhibited TNF-alpha and IL-4 production by RBL-1 cells. Tacrolimus 0-5 tumor necrosis factor Rattus norvegicus 36-45 10685002-7 2000 FK506 as well as steroids inhibited TNF-alpha and IL-4 production by RBL-1 cells. Tacrolimus 0-5 interleukin 4 Rattus norvegicus 50-54 10789681-0 2000 Involvement of FK506-sensitive and insensitive granule exocytosis pathways in perforin-dependent target cell lysis mediated by a CD8+ CTL clone. Tacrolimus 15-20 CD8a molecule Homo sapiens 129-132 10789681-1 2000 The release of granzyme A and B through granule exocytosis by CD8+ CTL clone OE4 upon T cell receptor (TCR) activation was blocked by FK506 in a dose-dependent manner (IC50 = 3 nM), whereas a significant granzyme release was still detectable even in the presence of excess FK506. Tacrolimus 134-139 granzyme A Homo sapiens 15-31 10789681-1 2000 The release of granzyme A and B through granule exocytosis by CD8+ CTL clone OE4 upon T cell receptor (TCR) activation was blocked by FK506 in a dose-dependent manner (IC50 = 3 nM), whereas a significant granzyme release was still detectable even in the presence of excess FK506. Tacrolimus 134-139 CD8a molecule Homo sapiens 62-65 10789681-1 2000 The release of granzyme A and B through granule exocytosis by CD8+ CTL clone OE4 upon T cell receptor (TCR) activation was blocked by FK506 in a dose-dependent manner (IC50 = 3 nM), whereas a significant granzyme release was still detectable even in the presence of excess FK506. Tacrolimus 134-139 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 86-101 10789681-1 2000 The release of granzyme A and B through granule exocytosis by CD8+ CTL clone OE4 upon T cell receptor (TCR) activation was blocked by FK506 in a dose-dependent manner (IC50 = 3 nM), whereas a significant granzyme release was still detectable even in the presence of excess FK506. Tacrolimus 134-139 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 103-106 10789681-1 2000 The release of granzyme A and B through granule exocytosis by CD8+ CTL clone OE4 upon T cell receptor (TCR) activation was blocked by FK506 in a dose-dependent manner (IC50 = 3 nM), whereas a significant granzyme release was still detectable even in the presence of excess FK506. Tacrolimus 273-278 granzyme A Homo sapiens 15-31 10789681-1 2000 The release of granzyme A and B through granule exocytosis by CD8+ CTL clone OE4 upon T cell receptor (TCR) activation was blocked by FK506 in a dose-dependent manner (IC50 = 3 nM), whereas a significant granzyme release was still detectable even in the presence of excess FK506. Tacrolimus 273-278 CD8a molecule Homo sapiens 62-65 10789681-1 2000 The release of granzyme A and B through granule exocytosis by CD8+ CTL clone OE4 upon T cell receptor (TCR) activation was blocked by FK506 in a dose-dependent manner (IC50 = 3 nM), whereas a significant granzyme release was still detectable even in the presence of excess FK506. Tacrolimus 273-278 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 86-101 10789681-2 2000 In contrast, the production of IFN-gamma was highly sensitive to FK506 (IC50 = 0.01 nM) and could be completely blocked by FK506. Tacrolimus 65-70 interferon gamma Homo sapiens 31-40 10789681-2 2000 In contrast, the production of IFN-gamma was highly sensitive to FK506 (IC50 = 0.01 nM) and could be completely blocked by FK506. Tacrolimus 123-128 interferon gamma Homo sapiens 31-40 10757347-7 2000 FK506 and anti-CD4 antibody administration decreased utrophin expression in adenovirus-injected mdx muscles and prevented the dystrophic phenotype from being mitigated, suggesting that an immune reaction is involved in utrophin upregulation. Tacrolimus 0-5 utrophin Mus musculus 53-61 10757347-7 2000 FK506 and anti-CD4 antibody administration decreased utrophin expression in adenovirus-injected mdx muscles and prevented the dystrophic phenotype from being mitigated, suggesting that an immune reaction is involved in utrophin upregulation. Tacrolimus 0-5 utrophin Mus musculus 219-227 10731442-3 2000 This study investigated whether cardioprotection induced by tacrolimus in myocardial ischaemia-reperfusion (MI/R) injury might be due to inhibition of the nuclear factor kappa B (NF- kappaB) that in turn causes reduced cardiac ICAM-1 expression and blunted polymorphonuclear leukocyte accumulation. Tacrolimus 60-70 intercellular adhesion molecule 1 Rattus norvegicus 227-233 10731442-11 2000 The present data suggest that tacrolimus blocks the early activation of the transcription factor NF- kappaB, suppresses ICAM-1 gene activation, reduces leukocyte accumulation and protects against myocardial ischaemia-reperfusion injury. Tacrolimus 30-40 intercellular adhesion molecule 1 Rattus norvegicus 120-126 10712705-1 2000 We have previously shown that a S1360F mutation in transmembrane domain 10 (TMD10) of the Pdr5p ABC transporter modulates substrate specificity and simultaneously leads to a loss of FK506 inhibition. Tacrolimus 182-187 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 90-95 10712705-5 2000 Most remarkably, however, the S1360A mutation increases FK506 inhibitor susceptibility, because Pdr5p-S1360A is hypersensitive to FK506 inhibition when compared with either wild-type Pdr5p or the non-responsive S1360F variant. Tacrolimus 56-61 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 96-101 10712705-5 2000 Most remarkably, however, the S1360A mutation increases FK506 inhibitor susceptibility, because Pdr5p-S1360A is hypersensitive to FK506 inhibition when compared with either wild-type Pdr5p or the non-responsive S1360F variant. Tacrolimus 56-61 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 183-188 10712705-5 2000 Most remarkably, however, the S1360A mutation increases FK506 inhibitor susceptibility, because Pdr5p-S1360A is hypersensitive to FK506 inhibition when compared with either wild-type Pdr5p or the non-responsive S1360F variant. Tacrolimus 130-135 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 96-101 10708116-2 2000 As a result, serious adverse effects, such as nephrotoxicity and neurotoxicity, have been observed under CsA/tacrolimus therapy. Tacrolimus 109-119 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 105-108 10688206-2 2000 FK506 and cyclosporin are immunosuppressants which block three antigen-receptor signalling pathways (NFAT, NFkappaB and JNK), through inhibition of calcineurin, and inhibit mature lymphocyte proliferation to antigen. Tacrolimus 0-5 nuclear factor kappa B subunit 1 Homo sapiens 107-115 10688206-2 2000 FK506 and cyclosporin are immunosuppressants which block three antigen-receptor signalling pathways (NFAT, NFkappaB and JNK), through inhibition of calcineurin, and inhibit mature lymphocyte proliferation to antigen. Tacrolimus 0-5 mitogen-activated protein kinase 8 Homo sapiens 120-123 10652370-4 2000 The immunosuppressant calcineurin inhibitor FK506 inhibited the down-regulation of the PMCA4CII at both the protein and the mRNA level but did not affect the changes of the other PMCA pumps. Tacrolimus 44-49 ATPase plasma membrane Ca2+ transporting 4 Homo sapiens 87-92 10751036-1 2000 PURPOSE: To study the contribution of P-glycoprotein (P-gp) to the oral absorption of a substrate, tacrolimus, by comparing the extent and rate of bioavailability in normal and mdr1a knockout mice. Tacrolimus 99-109 phosphoglycolate phosphatase Mus musculus 38-52 10701683-0 2000 FK506 augments glucocorticoid-mediated cyclooxygenase-2 down-regulation in human rheumatoid synovial fibroblasts. Tacrolimus 0-5 prostaglandin-endoperoxide synthase 2 Homo sapiens 39-55 10701683-3 2000 We examined the regulation of COX-2 mRNA and protein expression in response to both glucocorticoids (GC) and FK506 using rheumatoid synovial fibroblasts. Tacrolimus 109-114 mitochondrially encoded cytochrome c oxidase II Homo sapiens 30-35 10701683-4 2000 Combined treatment of FK506 and a low concentration of dexamethasone (DEX) (10(-9) M) down-regulated synovial COX-2 mRNA and protein expression. Tacrolimus 22-27 mitochondrially encoded cytochrome c oxidase II Homo sapiens 110-115 10701683-6 2000 Immunocytochemical studies showed that pretreatment with FK506 enhanced the nuclear translocation of the glucocorticoid receptor (GR) in synovial fibroblasts in the presence of low concentrations of DEX (10(-9) M). Tacrolimus 57-62 nuclear receptor subfamily 3 group C member 1 Homo sapiens 105-128 10701683-6 2000 Immunocytochemical studies showed that pretreatment with FK506 enhanced the nuclear translocation of the glucocorticoid receptor (GR) in synovial fibroblasts in the presence of low concentrations of DEX (10(-9) M). Tacrolimus 57-62 nuclear receptor subfamily 3 group C member 1 Homo sapiens 130-132 10701683-9 2000 Electrophoretic mobility shift assay demonstrated that DNA-binding activity of NF-KB was suppressed more profoundly by FK506 plus DEX (10(-9) M) treatment with those of DEX (10(-9)M) alone in IL-1beta-stimulated synovial cells. Tacrolimus 119-124 interleukin 1 beta Homo sapiens 192-200 10701683-10 2000 Our results indicated that FK506-induced potentiation of GR-mediated repression of synovial COX-2 gene transcription is the result of increased translocation of GR to the nucleus and subsequent repression of NF-kappaB transactivation. Tacrolimus 27-32 mitochondrially encoded cytochrome c oxidase II Homo sapiens 92-97 10701683-11 2000 Our results also suggest that FK506 may exert anti-inflammatory effects in the rheumatoid synovium by potentiating GR-mediated signal transduction. Tacrolimus 30-35 nuclear receptor subfamily 3 group C member 1 Homo sapiens 115-117 10731062-3 2000 As CYP3A4 enzymes and P-gp are present at differing concentrations throughout the gastrointestinal tract, the bioavailability of tacrolimus may be influenced by changes in gastrointestinal transit time in addition to changes in hepatic metabolism. Tacrolimus 129-139 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 3-9 10731062-3 2000 As CYP3A4 enzymes and P-gp are present at differing concentrations throughout the gastrointestinal tract, the bioavailability of tacrolimus may be influenced by changes in gastrointestinal transit time in addition to changes in hepatic metabolism. Tacrolimus 129-139 ATP binding cassette subfamily B member 1 Homo sapiens 22-26 10751036-1 2000 PURPOSE: To study the contribution of P-glycoprotein (P-gp) to the oral absorption of a substrate, tacrolimus, by comparing the extent and rate of bioavailability in normal and mdr1a knockout mice. Tacrolimus 99-109 phosphoglycolate phosphatase Mus musculus 54-58 10751036-14 2000 The clearance data also suggest a very dominant role of P-glycoprotein in controlling the rate of hepatic metabolism of tacrolimus in normal mice, and P-glycoprotein may serve as an effective efflux pump for direct transport of metabolites formed in hepatocytes into the blood circulation. Tacrolimus 120-130 phosphoglycolate phosphatase Mus musculus 56-70 11193162-5 2000 We now show that neuroimmunophilin ligands (like cyclosporin A or FK-506) and nonsteroidal antiinflammatory agents (NSAIDs), including cyclooxygenase (COX)-2 inhibitors, can also prevent APP overexpression and the overproduction of amyloidogenic peptides. Tacrolimus 66-72 mitochondrially encoded cytochrome c oxidase II Homo sapiens 135-157 10623799-2 2000 IL-4 mRNA, constitutively present in basophils, was increased after stimulation by gp120 and was inhibited cyclosporin A and tacrolimus. Tacrolimus 125-135 interleukin 4 Homo sapiens 0-4 10989953-11 2000 Although the interpretation of our metabolic data in patients with concomitant liver disease and prednisolone therapy has limitations, we suggest insulin resistance caused by treatment with FK506. Tacrolimus 190-195 insulin Homo sapiens 146-153 10632096-7 2000 Tacrolimus also reduced the number of activated cdr2-specific CTLs in the peripheral blood, but did not lead to tumor recurrence. Tacrolimus 0-10 cerebellar degeneration related protein 2 Homo sapiens 48-52 10632096-8 2000 We suggest that activated cdr2-specific CTLs in the CSF contribute to Purkinje degeneration in PCD, and that tacrolimus therapy may benefit patients with paraneoplastic neurological disease and other T cell-mediated autoimmune neurological disorders. Tacrolimus 109-119 cerebellar degeneration related protein 2 Homo sapiens 26-30 10729997-4 2000 The percentage of CD8+CD57+ cells in the patients receiving tacrolimus and developing acute graft-versus-host disease (GVHD) (grade I, 20.1% +/- 10.6%; grade II-IV, 13.2% +/- 6.3%) was significantly higher than in the patients receiving CsA and developing acute GVHD (grade I, 10.7% +/- 5.2%; grade II-IV, 7.7% +/- 4.0%) (grade I, P = .0036; grade II-IV, P = .0255). Tacrolimus 60-70 CD8a molecule Homo sapiens 18-21 11019782-3 2000 Stable IL-3 transcripts in the PB-3c-derived tumour cell line V2D1 decayed in response to CsA and FK506, but not in response to SB202190. Tacrolimus 98-103 interleukin 3 Mus musculus 7-11 10729997-8 2000 These results suggest that, compared with CsA, tacrolimus administered prophylactically induces more CD8+CD57+ lymphocytes when acute GVHD occurs and accelerates the recovery from acute GVHD more rapidly. Tacrolimus 47-57 CD8a molecule Homo sapiens 101-104 10729997-8 2000 These results suggest that, compared with CsA, tacrolimus administered prophylactically induces more CD8+CD57+ lymphocytes when acute GVHD occurs and accelerates the recovery from acute GVHD more rapidly. Tacrolimus 47-57 beta-1,3-glucuronyltransferase 1 Homo sapiens 105-109 10778917-0 2000 Differential anti-inflammatory effects of immunosuppressive drugs: cyclosporin, rapamycin and FK-506 on inducible nitric oxide synthase, nitric oxide, cyclooxygenase-2 and PGE2 production. Tacrolimus 94-100 prostaglandin-endoperoxide synthase 2 Mus musculus 151-167 10729997-5 2000 The absolute number of CD8+CD57+ cells in the patients with grade II-IV acute GVHD was also significantly higher in the tacrolimus group compared with the CsA group. Tacrolimus 120-130 CD8a molecule Homo sapiens 23-26 10729997-5 2000 The absolute number of CD8+CD57+ cells in the patients with grade II-IV acute GVHD was also significantly higher in the tacrolimus group compared with the CsA group. Tacrolimus 120-130 beta-1,3-glucuronyltransferase 1 Homo sapiens 27-31 10648038-0 2000 FK506 attenuates developing and established joint inflammation and suppresses interleukin 6 and nitric oxide expression in bacterial cell wall induced polyarthritis. Tacrolimus 0-5 interleukin 6 Rattus norvegicus 78-91 10731632-7 2000 Immunosuppressants FK506 and cyclosporin A were able to induce partial nuclear translocation of GR, suggesting that potentiation of glucocorticoid action by these compounds may also proceed through enhanced GR nuclear transfer. Tacrolimus 19-24 nuclear receptor subfamily 3 group C member 1 Homo sapiens 96-98 10731632-7 2000 Immunosuppressants FK506 and cyclosporin A were able to induce partial nuclear translocation of GR, suggesting that potentiation of glucocorticoid action by these compounds may also proceed through enhanced GR nuclear transfer. Tacrolimus 19-24 nuclear receptor subfamily 3 group C member 1 Homo sapiens 207-209 10648038-9 2000 The elevated expression of IL-6 and NO characteristic of chronic joint inflammation in the serum and in joint tissue was significantly reduced by FK506 treatment. Tacrolimus 146-151 interleukin 6 Rattus norvegicus 27-31 11041285-10 2000 A strong immunostaining for FKBP-12, a tacrolimus-binding protein, was observed in the medulla of the kidneys of rats treated with tacrolimus either with or without furosemide. Tacrolimus 39-49 FKBP prolyl isomerase 1A Rattus norvegicus 28-35 10670442-0 2000 The immunophilin ligand FK506, but not GPI-1046, protects against neuronal death and inhibits c-Jun expression in the substantia nigra pars compacta following transection of the rat medial forebrain bundle. Tacrolimus 24-29 Jun proto-oncogene, AP-1 transcription factor subunit Homo sapiens 94-99 10670442-6 2000 In contrast, application of FK506 resulted in survival of 46%, 44% and 28% of the axotomized nigral neurons, and the majority of these surviving neurons showed continuous expression of tyrosine hydroxylase, the pacemaker enzyme for dopamine synthesis. Tacrolimus 28-33 tyrosine hydroxylase Homo sapiens 185-205 10670442-7 2000 Moreover, FK506 significantly reduced the expression of the inducible transcription factor c-Jun and its N-terminal phosphorylation and prevented the axotomy-induced suppression of the constitutive transcription factor ATF-2 in neurons of the substantia nigra and mammillary body. Tacrolimus 10-15 Jun proto-oncogene, AP-1 transcription factor subunit Homo sapiens 91-96 10670442-7 2000 Moreover, FK506 significantly reduced the expression of the inducible transcription factor c-Jun and its N-terminal phosphorylation and prevented the axotomy-induced suppression of the constitutive transcription factor ATF-2 in neurons of the substantia nigra and mammillary body. Tacrolimus 10-15 activating transcription factor 2 Homo sapiens 219-224 10670442-11 2000 Our data provide the first evidence that FK506 acts as a neuroprotective molecule that rescues axotomized otherwise degenerating central intrinsic neurons in the adult mammalian brain by mechanisms that interfere with the transcriptional program of the axotomy-induced cell body response, such as activating transcription factor-2 suppression and c-Jun expression and phosphorylation. Tacrolimus 41-46 Jun proto-oncogene, AP-1 transcription factor subunit Homo sapiens 347-352 10601253-1 1999 FKBP52 is a high molecular mass immunophilin possessing peptidylprolyl isomerase (PPIase) activity that is inhibited by the immunosuppressant drug FK506. Tacrolimus 147-152 FKBP prolyl isomerase 4 Homo sapiens 0-6 12671210-15 2000 FK 506 promotes axonal regeneration through binding to FKBP-12. Tacrolimus 0-6 FKBP prolyl isomerase 1A Rattus norvegicus 55-62 11112017-4 2000 The current study was designed to measure the expression of TGF-beta in renal transplant biopsies from patients immunosuppressed with either CyA or tacrolimus. Tacrolimus 148-158 transforming growth factor beta 1 Homo sapiens 60-68 10601253-1 1999 FKBP52 is a high molecular mass immunophilin possessing peptidylprolyl isomerase (PPIase) activity that is inhibited by the immunosuppressant drug FK506. Tacrolimus 147-152 FKBP prolyl isomerase 4 Homo sapiens 82-88 10601253-1 1999 FKBP52 is a high molecular mass immunophilin possessing peptidylprolyl isomerase (PPIase) activity that is inhibited by the immunosuppressant drug FK506. Tacrolimus 147-152 FKBP prolyl isomerase 4 Homo sapiens 56-80 10586059-11 1999 Cyclosporin A and FK506, but not KN-62, blocked Ca2+ ionophore-mediated inhibition of Pyk2 expression, implicating calcineurin in down-regulating Pyk2 expression. Tacrolimus 18-23 protein tyrosine kinase 2 beta Homo sapiens 86-90 10593895-2 1999 Calcineurin is the target of the immunosuppressive drugs cyclosporin A and FK506 complexed with their cytoplasmic receptors cyclophilin and FKBP12, respectively. Tacrolimus 75-80 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 140-146 10585867-8 1999 One major protein of the first group, p42, had a rapid increase in synthesis that decreased by 8 h. Its synthesis was strongly enhanced by FK506, but reduced by rapamycin after ionomycin+PMA stimulation. Tacrolimus 139-144 cyclin dependent kinase 20 Homo sapiens 38-41 10585867-11 1999 mRNA levels of both gamma- and beta-actin were found to be enhanced with FK506 treatment suggesting that regulation of actin was at a transcriptional or post-transcriptional level. Tacrolimus 73-78 POTE ankyrin domain family member F Homo sapiens 20-41 10586059-11 1999 Cyclosporin A and FK506, but not KN-62, blocked Ca2+ ionophore-mediated inhibition of Pyk2 expression, implicating calcineurin in down-regulating Pyk2 expression. Tacrolimus 18-23 protein tyrosine kinase 2 beta Homo sapiens 146-150 10594646-7 1999 Ca2 + reperfusion injury induced by Ca2 + depletion or H2O2 exposure was blocked by the iron chelator 1, 10-phenanthroline, the NF-kappaB inhibitor pyrrolidinedithiocarbamate and the calcineurin inhibitor FK506. Tacrolimus 205-210 carbonic anhydrase 2 Homo sapiens 0-3 10574930-0 1999 Immunosuppressant FK506 activates NF-kappaB through the proteasome-mediated degradation of IkappaBalpha. Tacrolimus 18-23 NFKB inhibitor alpha Homo sapiens 91-103 10574930-2 1999 The immunosuppressant FK506 activates NF-kappaB through IkappaBalpha degradation in nonlymphoid cells. Tacrolimus 22-27 NFKB inhibitor alpha Homo sapiens 56-68 10574930-3 1999 In the present study, we analyzed mechanisms by which FK506 induces IkappaBalpha degradation. Tacrolimus 54-59 NFKB inhibitor alpha Homo sapiens 68-80 10574930-4 1999 We found that FK506 induces the degradation of both IkappaBalpha and IkappaBbeta and that the time courses of the FK506-induced degradation are quite different from degradation induced by interleukin 1 (IL-1). Tacrolimus 14-19 NFKB inhibitor alpha Homo sapiens 52-64 10574930-4 1999 We found that FK506 induces the degradation of both IkappaBalpha and IkappaBbeta and that the time courses of the FK506-induced degradation are quite different from degradation induced by interleukin 1 (IL-1). Tacrolimus 14-19 NFKB inhibitor beta Homo sapiens 69-80 10574930-4 1999 We found that FK506 induces the degradation of both IkappaBalpha and IkappaBbeta and that the time courses of the FK506-induced degradation are quite different from degradation induced by interleukin 1 (IL-1). Tacrolimus 114-119 NFKB inhibitor alpha Homo sapiens 52-64 10574930-5 1999 Despite this difference, FK506-induced IkappaBalpha degradation was dependent on the N-terminal Ser-32 and Ser-36 phosphorylation sites and was mediated by proteasomes, as is the case for IL-1-induced IkappaBalpha degradation. Tacrolimus 25-30 NFKB inhibitor alpha Homo sapiens 39-51 10574930-5 1999 Despite this difference, FK506-induced IkappaBalpha degradation was dependent on the N-terminal Ser-32 and Ser-36 phosphorylation sites and was mediated by proteasomes, as is the case for IL-1-induced IkappaBalpha degradation. Tacrolimus 25-30 interleukin 1 alpha Homo sapiens 188-192 10574930-5 1999 Despite this difference, FK506-induced IkappaBalpha degradation was dependent on the N-terminal Ser-32 and Ser-36 phosphorylation sites and was mediated by proteasomes, as is the case for IL-1-induced IkappaBalpha degradation. Tacrolimus 25-30 NFKB inhibitor alpha Homo sapiens 201-213 10574930-6 1999 We further showed that FK506 induces weak and slow phosphorylation of IkappaBalpha at Ser-32. Tacrolimus 23-28 NFKB inhibitor alpha Homo sapiens 70-82 10574930-8 1999 These results therefore indicate that FK506 and IL-1 utilize similar but distinct mechanisms to induce the phosphorylation and degradation of IkappaBalpha. Tacrolimus 38-43 NFKB inhibitor alpha Homo sapiens 142-154 10574997-1 1999 It has been reported that immunosuppressant cyclosporin A or FK506 binds to immunophilins in the cell and that these immunophilins make a complex with molecular chaperones HSP70 or HSP90. Tacrolimus 61-66 heat shock protein family A (Hsp70) member 4 Homo sapiens 172-177 10574997-1 1999 It has been reported that immunosuppressant cyclosporin A or FK506 binds to immunophilins in the cell and that these immunophilins make a complex with molecular chaperones HSP70 or HSP90. Tacrolimus 61-66 heat shock protein 90 alpha family class A member 1 Homo sapiens 181-186 10594646-7 1999 Ca2 + reperfusion injury induced by Ca2 + depletion or H2O2 exposure was blocked by the iron chelator 1, 10-phenanthroline, the NF-kappaB inhibitor pyrrolidinedithiocarbamate and the calcineurin inhibitor FK506. Tacrolimus 205-210 carbonic anhydrase 2 Homo sapiens 36-39 10594746-10 1999 Cyclosporin A (CsA) and FK506 slightly inhibited CD40 and B7-1 expression on Langerhans cells, but not B7-2. Tacrolimus 24-29 CD40 molecule Homo sapiens 49-53 10594746-10 1999 Cyclosporin A (CsA) and FK506 slightly inhibited CD40 and B7-1 expression on Langerhans cells, but not B7-2. Tacrolimus 24-29 CD80 molecule Homo sapiens 58-62 10543746-6 1999 On the other hand, FK506, which is also a macrolide derivative, inhibited transcriptional activation of both NF-kappaB and NFAT more strongly than EM did. Tacrolimus 19-24 nuclear factor kappa B subunit 1 Homo sapiens 109-118 10551814-7 1999 These actions of B10 were produced only on the cytoplasmic face of the channel, were selectively eliminated by pretreatment of channels with FK506 or rapamycin, and were reconstituted by human recombinant FKBP12. Tacrolimus 141-146 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 205-211 10573073-0 1999 Microangiopathy in kidney and simultaneous pancreas/kidney recipients treated with tacrolimus: evidence of endothelin and cytokine involvement. Tacrolimus 83-93 TNF receptor superfamily member 8 Homo sapiens 122-130 10589954-2 1999 Previous studies have shown that stimulation of T cells via CD28 or phorbol myristate acetate (PMA) activation is highly resistant to inhibition by cyclosporine A (CsA) and tacrolimus (FK506), as is the response of T cells to phytohemmaglutinin in the presence of endothelial cells. Tacrolimus 173-183 CD28 molecule Homo sapiens 60-64 10589954-2 1999 Previous studies have shown that stimulation of T cells via CD28 or phorbol myristate acetate (PMA) activation is highly resistant to inhibition by cyclosporine A (CsA) and tacrolimus (FK506), as is the response of T cells to phytohemmaglutinin in the presence of endothelial cells. Tacrolimus 185-190 CD28 molecule Homo sapiens 60-64 10589954-3 1999 We have investigated the inhibitory effects of CsA and FK506 on the direct response of human CD4+ T cells to HUVEC and PAEC and the effect of adding B7-1 transfectants. Tacrolimus 55-60 CD4 molecule Homo sapiens 93-96 10543746-8 1999 Expression of calcineurin did not render transactivation of NF-kappaB in T cells more resistant to EM, while the inhibitory effect of FK506 on transactivation of NF-kappaB was attenuated. Tacrolimus 134-139 nuclear factor kappa B subunit 1 Homo sapiens 162-171 10521511-10 1999 The identification of MEF2 as a novel target of calcineurin may provide in part a biochemical explanation for the therapeutic and toxic effects of immunosuppressants CsA and FK506. Tacrolimus 174-179 myocyte enhancer factor 2A Homo sapiens 22-26 10592469-9 1999 IL-8 production was significantly inhibited by N-acetyl-L-cysteine, FK506 and MG-132, inhibitors of NF-kappaB activation and translocation. Tacrolimus 68-73 C-X-C motif chemokine ligand 8 Homo sapiens 0-4 10592469-9 1999 IL-8 production was significantly inhibited by N-acetyl-L-cysteine, FK506 and MG-132, inhibitors of NF-kappaB activation and translocation. Tacrolimus 68-73 nuclear factor kappa B subunit 1 Homo sapiens 100-109 10555752-7 1999 Growth of tumour cells was also inhibited by the immunosuppressive drugs, cyclosporin A (CsA), FK506 and rapamycin (RPA), known to interfere with the IL-2 pathway in lymphocytes. Tacrolimus 95-100 interleukin 2 Homo sapiens 150-154 10531391-5 1999 Myoblasts and myotubes of C2-C12 cells express similar amounts of cyclophilin A and FKBP12, immunophilins known to be intracellular-binding targets for CsA and tacrolimus, respectively. Tacrolimus 160-170 peptidylprolyl isomerase A Mus musculus 66-79 10531391-5 1999 Myoblasts and myotubes of C2-C12 cells express similar amounts of cyclophilin A and FKBP12, immunophilins known to be intracellular-binding targets for CsA and tacrolimus, respectively. Tacrolimus 160-170 FK506 binding protein 1a Mus musculus 84-90 10578312-0 1999 Efficacy of tacrolimus in ABO-incompatible kidney transplantation: clinicopathological aspect of humoral rejection. Tacrolimus 12-22 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 26-29 10529362-6 1999 Moreover, the LTB(4)R antagonist exerted an additive inhibitory effect to FK506 on T cell proliferation. Tacrolimus 74-79 leukotriene B4 receptor Homo sapiens 14-21 10521511-4 1999 The specific inhibitors of calcineurin cyclosporin A (CsA) and FK506 could overcome KCl-dependent MEF2A hypophosphorylation. Tacrolimus 63-68 myocyte enhancer factor 2A Homo sapiens 98-103 10508685-6 1999 The approach has been established by isolating a full-length gene clone of FKBP12 (FK506-binding protein) from a human brain cDNA library using a biotinylated FK506 probe molecule. Tacrolimus 83-88 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 75-81 10521511-7 1999 Consistent with this, CsA/FK506 also inhibited MEF2-dependent reporter gene expression. Tacrolimus 26-31 myocyte enhancer factor 2A Homo sapiens 47-51 10532544-0 1999 FK506 markedly enhances apoptosis of antigen-stimulated peripheral T cells by down-regulation of Bcl-xL. Tacrolimus 0-5 BCL2-like 1 Mus musculus 97-103 10532544-8 1999 As Bcl-2-related proteins are involved in apoptotic process, we also evaluated their role by examining the expression of Bcl-2, Bcl-X(L), and Bax on SEB-FK506-treated murine splenic T cells. Tacrolimus 153-158 B cell leukemia/lymphoma 2 Mus musculus 3-8 10532544-8 1999 As Bcl-2-related proteins are involved in apoptotic process, we also evaluated their role by examining the expression of Bcl-2, Bcl-X(L), and Bax on SEB-FK506-treated murine splenic T cells. Tacrolimus 153-158 B cell leukemia/lymphoma 2 Mus musculus 121-126 10532544-8 1999 As Bcl-2-related proteins are involved in apoptotic process, we also evaluated their role by examining the expression of Bcl-2, Bcl-X(L), and Bax on SEB-FK506-treated murine splenic T cells. Tacrolimus 153-158 BCL2-associated X protein Mus musculus 142-145 10532544-10 1999 In contrast, the expression of Bcl-x(L) on Vgamma8+ T cells, which was markedly induced by SEB, was abrogated by FK506. Tacrolimus 113-118 BCL2-like 1 Mus musculus 31-39 10532544-11 1999 CONCLUSIONS: Our findings indicate FK506-induced enhancement of apoptosis of activated T cells is mediated by down-regulation of Bcl-X(L) expression on these cells. Tacrolimus 35-40 BCL2-like 1 Mus musculus 129-137 10516933-4 1999 The objective of this review is to discuss the effects of P-gp modulation on the pharmacokinetics and the pharmacodynamics of immunosuppressive agents such as cyclosporine, tacrolimus, sirolimus, and corticosteroids. Tacrolimus 173-183 ATP binding cassette subfamily B member 1 Homo sapiens 58-62 10515221-10 1999 When we compared the frequencies of cytokine-expressing cells in FK506- and CsA-treated patients, we found that the frequency of IL-2-expressing T cells was significantly lower with FK506 (10.9+/-1.61%) than with CsA (16.3 +/- 1.8%; p = 0.03), whereas the frequencies of the other cytokine-expressing cells were not statistically different between the two groups. Tacrolimus 182-187 interleukin 2 Homo sapiens 129-133 10515221-11 1999 In conclusion, our study clearly demonstrated that studied ex vivo, FK506 and CsA decrease the frequencies of cells expressing IL-2, IL-4 and IL-2/IFN-gamma in vivo but do not affect those expressing IFN-gamma. Tacrolimus 68-73 interleukin 2 Homo sapiens 127-131 10515221-11 1999 In conclusion, our study clearly demonstrated that studied ex vivo, FK506 and CsA decrease the frequencies of cells expressing IL-2, IL-4 and IL-2/IFN-gamma in vivo but do not affect those expressing IFN-gamma. Tacrolimus 68-73 interleukin 4 Homo sapiens 133-137 10515221-11 1999 In conclusion, our study clearly demonstrated that studied ex vivo, FK506 and CsA decrease the frequencies of cells expressing IL-2, IL-4 and IL-2/IFN-gamma in vivo but do not affect those expressing IFN-gamma. Tacrolimus 68-73 interleukin 2 Homo sapiens 142-146 10515221-11 1999 In conclusion, our study clearly demonstrated that studied ex vivo, FK506 and CsA decrease the frequencies of cells expressing IL-2, IL-4 and IL-2/IFN-gamma in vivo but do not affect those expressing IFN-gamma. Tacrolimus 68-73 interferon gamma Homo sapiens 147-156 10515221-12 1999 Meanwhile, the frequency of IL-2-producing T cells was more affected with FK506 than with CsA and was negatively correlated with the CsA trough level. Tacrolimus 74-79 interleukin 2 Homo sapiens 28-32 10515221-13 1999 Finally, our results regarding IL-2 might explain to some extent the higher efficiency of FK506 in vivo than CsA. Tacrolimus 90-95 interleukin 2 Homo sapiens 31-35 10563471-7 1999 Furthermore we could show that IL-4, IFN-gamma, FK506, loratadine and UVA enhance the mRNA levels of the IL-10R1 in vitro in normal cultured keratinocytes. Tacrolimus 48-53 interleukin 10 receptor subunit alpha Homo sapiens 105-112 10563471-9 1999 CONCLUSIONS: Our results demonstrate for the first time that IL-10 receptors may have a role in the pathogenesis of atopic eczema and its upregulation by FK506 and UVA could explain the therapeutic efficacy of these agents. Tacrolimus 154-159 interleukin 10 Homo sapiens 61-66 10582320-8 1999 Thus, the inhibitory action of CsA and FK506 under in vitro conditions should occur before the step of acyl group reduction and the effect is likely to be attributable to the inhibition of calcineurin in the signal transduction cascade mechanism of PBAN, in B. mori. Tacrolimus 39-44 PBAN-type neuropeptides Bombyx mori 249-253 10491310-4 1999 Here, we show that CyA or FK506 downregulates FasL expression on endothelial cells with accompanying decrease in the cytotoxicity toward Fas-bearing cells. Tacrolimus 26-31 Fas ligand Homo sapiens 46-50 10510443-3 1999 Cyclosporin A and FK506 (at 1 microM) also significantly inhibited nitrite production induced by recombinant murine interferon-gamma (rIFNgamma) and recombinant murine interleukin-1beta (rIL-1beta) in J774 and VSMC, respectively. Tacrolimus 18-23 interferon gamma Mus musculus 116-132 10510443-3 1999 Cyclosporin A and FK506 (at 1 microM) also significantly inhibited nitrite production induced by recombinant murine interferon-gamma (rIFNgamma) and recombinant murine interleukin-1beta (rIL-1beta) in J774 and VSMC, respectively. Tacrolimus 18-23 interleukin 1 beta Mus musculus 168-185 10510443-3 1999 Cyclosporin A and FK506 (at 1 microM) also significantly inhibited nitrite production induced by recombinant murine interferon-gamma (rIFNgamma) and recombinant murine interleukin-1beta (rIL-1beta) in J774 and VSMC, respectively. Tacrolimus 18-23 interleukin 1 beta Rattus norvegicus 187-196 10463953-8 1999 In the presence of suboptimal amounts of S100betabeta, FK506, cyclosporin A, and cypermethrin (but not rapamycin) also increased NF-kappaB activity, as measured by immunofluorescence of cells stained with antibody to the active subunit (p65) and by immunoblotting of nuclear extracts. Tacrolimus 55-60 synaptotagmin 1 Gallus gallus 237-240 10491144-7 1999 FK506, another immunosuppressant that targets calcineurin, also inhibits pol beta upregulation, while rapamycin competes with FK506 action. Tacrolimus 0-5 DNA polymerase beta Homo sapiens 73-81 10468022-0 1999 P-glycoprotein-dependent disposition kinetics of tacrolimus: studies in mdr1a knockout mice. Tacrolimus 49-59 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 72-77 10480422-0 1999 Blood levels of TGFbeta1 in liver transplant recipients receiving either tacrolimus or micro-emulsified cyclosporine. Tacrolimus 73-83 transforming growth factor beta 1 Homo sapiens 16-24 10480422-3 1999 To determine whether tacrolimus (FK506) similarly increases TGFbeta1 we have measured TGFbeta levels in blood samples from liver graft recipients who were of known TGFbeta1-responder status. Tacrolimus 33-38 transforming growth factor beta 1 Homo sapiens 60-68 10480422-3 1999 To determine whether tacrolimus (FK506) similarly increases TGFbeta1 we have measured TGFbeta levels in blood samples from liver graft recipients who were of known TGFbeta1-responder status. Tacrolimus 33-38 transforming growth factor beta 1 Homo sapiens 60-67 10566134-15 1999 FK 506 promotes axonal regeneration through binding to FKBP-12, thus activating GAP-43 (growth associated protein) and the TGF beta 1-pathway (transforming growth factor). Tacrolimus 0-6 FKBP prolyl isomerase 1A Rattus norvegicus 55-62 10566134-15 1999 FK 506 promotes axonal regeneration through binding to FKBP-12, thus activating GAP-43 (growth associated protein) and the TGF beta 1-pathway (transforming growth factor). Tacrolimus 0-6 growth associated protein 43 Rattus norvegicus 80-86 10566134-15 1999 FK 506 promotes axonal regeneration through binding to FKBP-12, thus activating GAP-43 (growth associated protein) and the TGF beta 1-pathway (transforming growth factor). Tacrolimus 0-6 transforming growth factor, beta 1 Rattus norvegicus 123-133 10486154-7 1999 FK506, an inhibitor of NFAT activation, inhibited IFN-gamma production by IL-18 without any effect on the NF-kappaB activation. Tacrolimus 0-5 interferon gamma Mus musculus 50-59 10486154-7 1999 FK506, an inhibitor of NFAT activation, inhibited IFN-gamma production by IL-18 without any effect on the NF-kappaB activation. Tacrolimus 0-5 interleukin 18 Mus musculus 74-79 10461545-4 1999 Regardless of the underlying mechanism involved, the FKBP-52 antibody data reveal that it should be possible to design, based on the structure of FK506, non-FKBP-12-binding (nonimmunosuppressant) compounds selective for FKBP-52 and test these new libraries for their ability to augment nerve regeneration. Tacrolimus 146-151 FKBP prolyl isomerase 4 Homo sapiens 53-60 10461545-4 1999 Regardless of the underlying mechanism involved, the FKBP-52 antibody data reveal that it should be possible to design, based on the structure of FK506, non-FKBP-12-binding (nonimmunosuppressant) compounds selective for FKBP-52 and test these new libraries for their ability to augment nerve regeneration. Tacrolimus 146-151 FKBP prolyl isomerase 4 Homo sapiens 220-227 10440408-5 1999 We hypothesize that the protease inhibitors" competition for binding to cytochrome P450 isoenzyme system CYP3A induced extreme prolongation of tacrolimus metabolism. Tacrolimus 143-153 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-110 10450987-1 1999 The synthesis of C32-O-arylethyl ether derivatives of ascomycin that possess equivalent immunosuppressant activity but reduced toxicity, compared to FK-506, is described. Tacrolimus 149-155 chemokine like factor Homo sapiens 17-20 10465413-3 1999 Two classes of immunophilin have been identified with cyclophilins (CyP"s) being proteins specifically binding CsA and FKBPs specifically binding FK506. Tacrolimus 146-151 peptidylprolyl isomerase G Homo sapiens 68-71 10401019-7 1999 Those 40 cases in which the biopsy showed evidence of CSA or tacrolimus nephrotoxicity had a significantly (P < 0.01) greater SMA level in the corresponding urine samples (0.089 +/- 0.126 microgram/mL; mean +/- SD) than the 49 cases without toxicity (0.018 +/- 0.027 microgram/mL) or 6 control subjects (0.003 +/- 0.007 microgram/mL), although there was considerable overlap of SMA values among these groups. Tacrolimus 61-71 survival of motor neuron 1, telomeric Homo sapiens 129-132 10401019-7 1999 Those 40 cases in which the biopsy showed evidence of CSA or tacrolimus nephrotoxicity had a significantly (P < 0.01) greater SMA level in the corresponding urine samples (0.089 +/- 0.126 microgram/mL; mean +/- SD) than the 49 cases without toxicity (0.018 +/- 0.027 microgram/mL) or 6 control subjects (0.003 +/- 0.007 microgram/mL), although there was considerable overlap of SMA values among these groups. Tacrolimus 61-71 survival of motor neuron 1, telomeric Homo sapiens 381-384 10401019-8 1999 The greatest SMA levels were seen in patients with CSA or tacrolimus nephrotoxicity that was likely to be relatively acute, namely those with thrombotic microangiopathy and those without previous biopsy evidence of toxicity. Tacrolimus 58-68 survival of motor neuron 1, telomeric Homo sapiens 13-16 10465413-6 1999 While the crystal structure of the FK506-FKBP12-CN complex has been reported, no structure for a CsA-CyP CN complex has been determined. Tacrolimus 35-40 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 41-47 10465413-8 1999 The first stage of constructing this model consisted of using conformational comparison of CsA and FK506, GRID and GROUP analysis and restrained molecular dynamics to dock CsA into the FK506 binding site of the FK506-FKBP12-CN structure. Tacrolimus 99-104 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 217-223 10465413-8 1999 The first stage of constructing this model consisted of using conformational comparison of CsA and FK506, GRID and GROUP analysis and restrained molecular dynamics to dock CsA into the FK506 binding site of the FK506-FKBP12-CN structure. Tacrolimus 185-190 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 172-175 10465413-8 1999 The first stage of constructing this model consisted of using conformational comparison of CsA and FK506, GRID and GROUP analysis and restrained molecular dynamics to dock CsA into the FK506 binding site of the FK506-FKBP12-CN structure. Tacrolimus 185-190 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 217-223 10465413-8 1999 The first stage of constructing this model consisted of using conformational comparison of CsA and FK506, GRID and GROUP analysis and restrained molecular dynamics to dock CsA into the FK506 binding site of the FK506-FKBP12-CN structure. Tacrolimus 185-190 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 172-175 10465413-8 1999 The first stage of constructing this model consisted of using conformational comparison of CsA and FK506, GRID and GROUP analysis and restrained molecular dynamics to dock CsA into the FK506 binding site of the FK506-FKBP12-CN structure. Tacrolimus 185-190 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 217-223 10367710-0 1999 FK506 and cyclosporin A inhibit stem cell factor-dependent cell proliferation/survival, while inducing upregulation of c-kit expression in cells of the mast cell line MC/9. Tacrolimus 0-5 kit ligand Mus musculus 32-48 10410178-9 1999 DISCUSSION: Tacrolimus is known to be a substrate for P-glycoprotein and metabolized by CYP3A. Tacrolimus 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 54-68 10410178-9 1999 DISCUSSION: Tacrolimus is known to be a substrate for P-glycoprotein and metabolized by CYP3A. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-93 10406194-8 1999 FK506 markedly inhibited the nitric oxide formation, inducible nitric oxide synthase protein synthesis and inducible nitric oxide synthase mRNA expression induced by interleukin 1beta, but cyclosporin A had no effects. Tacrolimus 0-5 nitric oxide synthase 2 Rattus norvegicus 53-84 10406194-8 1999 FK506 markedly inhibited the nitric oxide formation, inducible nitric oxide synthase protein synthesis and inducible nitric oxide synthase mRNA expression induced by interleukin 1beta, but cyclosporin A had no effects. Tacrolimus 0-5 nitric oxide synthase 2 Rattus norvegicus 107-138 10406194-8 1999 FK506 markedly inhibited the nitric oxide formation, inducible nitric oxide synthase protein synthesis and inducible nitric oxide synthase mRNA expression induced by interleukin 1beta, but cyclosporin A had no effects. Tacrolimus 0-5 interleukin 1 beta Rattus norvegicus 166-183 10406194-9 1999 Furthermore, FK506 inhibited nuclear factor-kappaB activation and decreased mRNA levels of the p50/p65 subunits of nuclear factor-kappaB. Tacrolimus 13-18 synaptotagmin 1 Rattus norvegicus 99-102 10406194-10 1999 CONCLUSIONS: These results demonstrate that FK506, but not cyclosporin A, inhibits the induction of inducible nitric oxide synthase expression during nuclear factor-kappaB activation. Tacrolimus 44-49 nitric oxide synthase 2 Rattus norvegicus 100-131 10367710-6 1999 FK506 and CsA inhibited the SCF-dependent, but not the IL-3 dependent, stimulatory effects on MC/9 cell proliferation/survival. Tacrolimus 0-5 kit ligand Mus musculus 28-31 10367710-8 1999 FK506 and CsA inhibited the SCF-dependent rescue effect from apoptosis. Tacrolimus 0-5 kit ligand Mus musculus 28-31 10367710-10 1999 However, immunoblot and reverse transcriptase-polymerase chain reaction (RT-PCR) analyses indicated that c-kit protein and c-kit mRNA transcripts were increased following the FK506 and CsA treatments in the presence of IL-3. Tacrolimus 175-180 KIT proto-oncogene receptor tyrosine kinase Mus musculus 105-110 10367710-10 1999 However, immunoblot and reverse transcriptase-polymerase chain reaction (RT-PCR) analyses indicated that c-kit protein and c-kit mRNA transcripts were increased following the FK506 and CsA treatments in the presence of IL-3. Tacrolimus 175-180 KIT proto-oncogene receptor tyrosine kinase Mus musculus 123-128 10367710-10 1999 However, immunoblot and reverse transcriptase-polymerase chain reaction (RT-PCR) analyses indicated that c-kit protein and c-kit mRNA transcripts were increased following the FK506 and CsA treatments in the presence of IL-3. Tacrolimus 175-180 interleukin 3 Mus musculus 219-223 10367710-11 1999 In addition, MC/9 cells pretreated with FK506 or CsA showed an increased adhesiveness to NIH/3T3 cells that express membrane-bound SCF. Tacrolimus 40-45 kit ligand Mus musculus 131-134 10361256-2 1999 The mechanism of this inhibition is investigated using FK506, which competes with rapamycin for binding to their common target FK506-binding protein (FKBP)12. Tacrolimus 55-60 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 150-157 10406194-0 1999 Immunosuppressant FK506 inhibits inducible nitric oxide synthase gene expression at a step of NF-kappaB activation in rat hepatocytes. Tacrolimus 18-23 nitric oxide synthase 2 Rattus norvegicus 33-64 10406194-3 1999 Studies were performed to determine whether the immunosuppressants FK506 and cyclosporin A directly influence gene expression of inducible nitric oxide synthase by interleukin 1beta in hepatocytes. Tacrolimus 67-72 nitric oxide synthase 2 Rattus norvegicus 129-160 10406194-3 1999 Studies were performed to determine whether the immunosuppressants FK506 and cyclosporin A directly influence gene expression of inducible nitric oxide synthase by interleukin 1beta in hepatocytes. Tacrolimus 67-72 interleukin 1 beta Rattus norvegicus 164-181 10336507-1 1999 The neurotrophic property of the immunosuppressant drug FK506 (tacrolimus) is believed to depend on the 12-kDa FK506-binding protein (FKBP-12). Tacrolimus 56-61 FKBP prolyl isomerase 1A Homo sapiens 134-141 10336507-1 1999 The neurotrophic property of the immunosuppressant drug FK506 (tacrolimus) is believed to depend on the 12-kDa FK506-binding protein (FKBP-12). Tacrolimus 63-73 FKBP prolyl isomerase 1A Homo sapiens 134-141 10336507-2 1999 Here, we show that FK506 maintains its neurotrophic activity in primary hippocampal cell cultures from FKBP-12 knockout mice. Tacrolimus 19-24 FK506 binding protein 1a Mus musculus 103-110 10336507-3 1999 In human neuroblastoma SH-SY5Y cells, the neurotrophic action of FK506 (10 pM to 10 nM) is completely prevented by the addition of a monoclonal antibody (50-100 nM) to the immunophilin FKBP-52 (also known as FKBP-59 or heat shock protein 56), a component of mature steroid receptor complexes. Tacrolimus 65-70 FKBP prolyl isomerase 4 Homo sapiens 185-192 10336507-3 1999 In human neuroblastoma SH-SY5Y cells, the neurotrophic action of FK506 (10 pM to 10 nM) is completely prevented by the addition of a monoclonal antibody (50-100 nM) to the immunophilin FKBP-52 (also known as FKBP-59 or heat shock protein 56), a component of mature steroid receptor complexes. Tacrolimus 65-70 FKBP prolyl isomerase 4 Homo sapiens 208-215 10347253-3 1999 Because inhibition of CREB/CRE-directed transcription by cyclosporin A and FK506 has previously been observed by using synthetic minienhancers, reporter fusion genes were constructed to examine the effect of cyclosporin A and FK506 on the transcriptional activity of CRE-containing natural promoters. Tacrolimus 75-80 cAMP responsive element binding protein 1 Rattus norvegicus 22-26 10234013-6 1999 FK506 also abolished phosphorylation, but not expression, of the c-Jun transcription factor involved in the transcriptional control of CD95 ligand. Tacrolimus 0-5 Fas (TNF receptor superfamily member 6) Mus musculus 135-139 10367710-13 1999 These results indicate that FK506 and CsA, while inducing c-kit of MC/9 cells, selectively inhibit the SCF-dependent stimulatory effects on MC/9 cell proliferation/survival by a mechanism independent of, or at point(s) distal to, the c-kit-MAP kinase pathway. Tacrolimus 28-33 KIT proto-oncogene receptor tyrosine kinase Mus musculus 58-63 10367710-13 1999 These results indicate that FK506 and CsA, while inducing c-kit of MC/9 cells, selectively inhibit the SCF-dependent stimulatory effects on MC/9 cell proliferation/survival by a mechanism independent of, or at point(s) distal to, the c-kit-MAP kinase pathway. Tacrolimus 28-33 kit ligand Mus musculus 103-106 10367710-13 1999 These results indicate that FK506 and CsA, while inducing c-kit of MC/9 cells, selectively inhibit the SCF-dependent stimulatory effects on MC/9 cell proliferation/survival by a mechanism independent of, or at point(s) distal to, the c-kit-MAP kinase pathway. Tacrolimus 28-33 KIT proto-oncogene receptor tyrosine kinase Mus musculus 234-239 10329481-0 1999 A potent immunosuppressant FK506 inhibits IL-8 expression in human eosinophils. Tacrolimus 27-32 C-X-C motif chemokine ligand 8 Homo sapiens 42-46 10224298-6 1999 The immunosuppressant FK506 inhibited the release of ceramide, expression of CD95-L and apoptosis in an in vitro and in vivo model for ischemia/reperfusion. Tacrolimus 22-27 Fas ligand Rattus norvegicus 77-83 10385251-3 1999 Tacrolimus, formerly known as FK506, is a macrolide antibiotic, that blocks the transcription of several proinflammatory cytokines including TNF-alpha. Tacrolimus 0-10 tumor necrosis factor Rattus norvegicus 141-150 10385251-3 1999 Tacrolimus, formerly known as FK506, is a macrolide antibiotic, that blocks the transcription of several proinflammatory cytokines including TNF-alpha. Tacrolimus 30-35 tumor necrosis factor Rattus norvegicus 141-150 10385251-12 1999 Tacrolimus (100 microg kg(-1), 5 min after splanchnic arteries occlusion) increased survival rate (SAO + Tacrolimus = 100% at 4 h of reperfusion), reverted the marked hypotension, reduced serum TNF-alpha (15+/-3 U ml(-1)), ameliorated leukopenia, reduced ileal MPO (0.9+/-0.01 U g(-1) tissue), restored to control values the hyporeactivity to PE. Tacrolimus 0-10 tumor necrosis factor Rattus norvegicus 194-203 10385251-12 1999 Tacrolimus (100 microg kg(-1), 5 min after splanchnic arteries occlusion) increased survival rate (SAO + Tacrolimus = 100% at 4 h of reperfusion), reverted the marked hypotension, reduced serum TNF-alpha (15+/-3 U ml(-1)), ameliorated leukopenia, reduced ileal MPO (0.9+/-0.01 U g(-1) tissue), restored to control values the hyporeactivity to PE. Tacrolimus 0-10 myeloperoxidase Rattus norvegicus 261-264 10416209-2 1999 FK506 is complexed with an intracellular binding protein FKBP-12. Tacrolimus 0-5 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 57-64 10329481-3 1999 In the present study, we evaluated if FK506 had any effect on the production of IL-8, one of the important chemokines for a variety of inflammatory cells, from human peripheral eosinophils. Tacrolimus 38-43 C-X-C motif chemokine ligand 8 Homo sapiens 80-84 10329481-5 1999 FK506 showed a dose-dependent suppressive effect on IL-8 production by eosinophils stimulated with calcium ionophore, but showed no effect on unstimulated cells. Tacrolimus 0-5 C-X-C motif chemokine ligand 8 Homo sapiens 52-56 10329481-6 1999 Evaluation of IL-8 mRNA levels by reverse transcription and polymerase chain reaction demonstrated that FK506 suppressed IL-8 gene expression only in activated eosinophils. Tacrolimus 104-109 C-X-C motif chemokine ligand 8 Homo sapiens 14-18 10329481-6 1999 Evaluation of IL-8 mRNA levels by reverse transcription and polymerase chain reaction demonstrated that FK506 suppressed IL-8 gene expression only in activated eosinophils. Tacrolimus 104-109 C-X-C motif chemokine ligand 8 Homo sapiens 121-125 10329481-7 1999 FK506 further showed a suppressive effect on eotaxin and MCP-1 release from eosinophils. Tacrolimus 0-5 C-C motif chemokine ligand 11 Homo sapiens 45-52 10329481-7 1999 FK506 further showed a suppressive effect on eotaxin and MCP-1 release from eosinophils. Tacrolimus 0-5 C-C motif chemokine ligand 2 Homo sapiens 57-62 10077670-5 1999 Calcineurin antagonists such as cyclosporin A and FK506, as well as nonimmunosuppressant analogs of cyclosporin A, significantly enhanced SODV148G- and SODA4V-induced cell death. Tacrolimus 50-55 superoxide dismutase 1 Homo sapiens 138-146 10205001-15 1999 CsA and FK506 reduced the release of cytochrome c to the cytosol and the activation of caspase-3 in cells treated with nitric oxide donors. Tacrolimus 8-13 caspase 3 Homo sapiens 87-96 10051602-1 1999 FKBP52 (FKBP59, FKBP4) is a "macro" immunophilin that, although sharing high structural and functional homologies in its amino-terminal domain with FKBP12 (FKBP1), does not have immunosuppressant activity when complexed with FK506, unlike FKBP12. Tacrolimus 225-230 FKBP prolyl isomerase 4 Homo sapiens 0-6 10051602-1 1999 FKBP52 (FKBP59, FKBP4) is a "macro" immunophilin that, although sharing high structural and functional homologies in its amino-terminal domain with FKBP12 (FKBP1), does not have immunosuppressant activity when complexed with FK506, unlike FKBP12. Tacrolimus 225-230 FKBP prolyl isomerase 4 Homo sapiens 8-14 10051602-1 1999 FKBP52 (FKBP59, FKBP4) is a "macro" immunophilin that, although sharing high structural and functional homologies in its amino-terminal domain with FKBP12 (FKBP1), does not have immunosuppressant activity when complexed with FK506, unlike FKBP12. Tacrolimus 225-230 FKBP prolyl isomerase 4 Homo sapiens 16-21 10051602-8 1999 Whereas the binding of calcineurin to FKBP12 is potentiated by FK506, the specific association of PAHX and FKBP52 is maintained in the presence of FK506. Tacrolimus 63-68 FKBP prolyl isomerase 1A Homo sapiens 38-44 10051602-8 1999 Whereas the binding of calcineurin to FKBP12 is potentiated by FK506, the specific association of PAHX and FKBP52 is maintained in the presence of FK506. Tacrolimus 147-152 phytanoyl-CoA 2-hydroxylase Homo sapiens 98-102 10051602-8 1999 Whereas the binding of calcineurin to FKBP12 is potentiated by FK506, the specific association of PAHX and FKBP52 is maintained in the presence of FK506. Tacrolimus 147-152 FKBP prolyl isomerase 4 Homo sapiens 107-113 10205001-7 1999 Moreover, incubation of activated macrophages with CsA and FK506 contributed to maintain higher levels of Bcl-2 than in LPS/IFN-gamma treated cells. Tacrolimus 59-64 BCL2 apoptosis regulator Homo sapiens 106-111 10205001-7 1999 Moreover, incubation of activated macrophages with CsA and FK506 contributed to maintain higher levels of Bcl-2 than in LPS/IFN-gamma treated cells. Tacrolimus 59-64 interferon gamma Homo sapiens 124-133 10375080-9 1999 Both Th1 and Th2 cytokine expression was suppressed in FK506-treated mice. Tacrolimus 55-60 negative elongation factor complex member C/D, Th1l Mus musculus 5-8 10205001-12 1999 Both CsA and FK506 inhibited pathways leading to caspase-3 activation. Tacrolimus 13-18 caspase 3 Homo sapiens 49-58 10205001-15 1999 CsA and FK506 reduced the release of cytochrome c to the cytosol and the activation of caspase-3 in cells treated with nitric oxide donors. Tacrolimus 8-13 cytochrome c, somatic Homo sapiens 37-49 10049913-1 1999 The immunosuppressive drugs FK506 and rapamycin bind to the cellular protein FKBP12, and the resulting FKBP12-drug complexes inhibit signal transduction. Tacrolimus 28-33 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 77-83 10049913-1 1999 The immunosuppressive drugs FK506 and rapamycin bind to the cellular protein FKBP12, and the resulting FKBP12-drug complexes inhibit signal transduction. Tacrolimus 28-33 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 103-109 10049913-9 1999 Because HMG1/2 proteins are conserved from yeast to humans, our findings suggest that FKBP12-HMG1/2 interactions could represent the first conserved function of FKBP12 other than mediating FK506 and rapamycin actions. Tacrolimus 189-194 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 86-92 10049913-9 1999 Because HMG1/2 proteins are conserved from yeast to humans, our findings suggest that FKBP12-HMG1/2 interactions could represent the first conserved function of FKBP12 other than mediating FK506 and rapamycin actions. Tacrolimus 189-194 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 161-167 10027932-9 1999 FK506, another immunosuppressant, increased SMIT activity, but only in isotonic conditions. Tacrolimus 0-5 LOC100856716 Canis lupus familiaris 44-48 10375080-9 1999 Both Th1 and Th2 cytokine expression was suppressed in FK506-treated mice. Tacrolimus 55-60 heart and neural crest derivatives expressed 2 Mus musculus 13-16 10071036-0 1999 Tacrolimus induces increased expression of transforming growth factor-beta1 in mammalian lymphoid as well as nonlymphoid cells. Tacrolimus 0-10 transforming growth factor beta 1 Homo sapiens 43-75 10026208-6 1999 A mutational analysis of G2 function and nuclear protein binding as well as the effect of FK506 indicate that calcium responsiveness is conferred to the G2 element by NFATp functionally interacting with HNF-3beta binding to a closely associated site. Tacrolimus 90-95 nuclear factor of activated T cells 2 Homo sapiens 167-172 10026208-6 1999 A mutational analysis of G2 function and nuclear protein binding as well as the effect of FK506 indicate that calcium responsiveness is conferred to the G2 element by NFATp functionally interacting with HNF-3beta binding to a closely associated site. Tacrolimus 90-95 forkhead box A2 Homo sapiens 203-212 10071036-2 1999 In view of similarities between tacrolimus and CsA with respect to immunosuppressive mechanisms, we determined whether tacrolimus, in a fashion similar to CsA, induces TGF-beta1 hyperexpression in mammalian cells. Tacrolimus 119-129 transforming growth factor beta 1 Homo sapiens 168-177 10083347-0 1999 Inhibition of iNOS induction by FK506, but not by cyclosporine, in rat hepatocytes. Tacrolimus 32-37 nitric oxide synthase 2 Rattus norvegicus 14-18 10355966-4 1999 In MRL-Ipr(cg) mice, an immunosuppressive agent, FK506, improved the serological abnormalities (decreased levels of anti-double-stranded DNA antibody of IgG2a and IgG3 subclasses) and proteinuria. Tacrolimus 49-54 immunoglobulin heavy variable V1-9 Mus musculus 153-158 10355966-4 1999 In MRL-Ipr(cg) mice, an immunosuppressive agent, FK506, improved the serological abnormalities (decreased levels of anti-double-stranded DNA antibody of IgG2a and IgG3 subclasses) and proteinuria. Tacrolimus 49-54 Immunoglobulin heavy constant gamma 3 Mus musculus 163-167 9918571-3 1999 Drugs binding to FKBP12 and inhibiting calcineurin, such as FK506 and SDZ ASM 981, dose dependently reduced the infarct volumes, determined 48 h after MCAO by both magnetic resonance imaging and triphenyltetrazolium chloride staining but only in the transient MCAO model. Tacrolimus 60-65 FKBP prolyl isomerase 1A Rattus norvegicus 17-23 10051052-2 1999 This finding was accompanied by a corresponding reduction of the inactive glucuronide metabolite of MPA (MPAG) in patients, suggesting that tacrolimus may effect the conversion of MPA to MPAG by the enzyme UDP-glucuronosyltransferase (UDPGT). Tacrolimus 140-150 UDP glucuronosyltransferase family 1 member A4 Homo sapiens 206-233 10051052-2 1999 This finding was accompanied by a corresponding reduction of the inactive glucuronide metabolite of MPA (MPAG) in patients, suggesting that tacrolimus may effect the conversion of MPA to MPAG by the enzyme UDP-glucuronosyltransferase (UDPGT). Tacrolimus 140-150 UDP glucuronosyltransferase family 1 member A4 Homo sapiens 235-240 10051052-8 1999 The addition of clinically relevant concentrations of CsA (200-1,000 ng/mL) or tacrolimus (10-25 ng/mL) resulted in a dose-dependent inhibition of the UDPGT enzyme by both agents with tacrolimus, which was approximately 60-fold more efficient as an inhibitor. Tacrolimus 79-89 UDP glucuronosyltransferase family 1 member A4 Homo sapiens 151-156 10051052-8 1999 The addition of clinically relevant concentrations of CsA (200-1,000 ng/mL) or tacrolimus (10-25 ng/mL) resulted in a dose-dependent inhibition of the UDPGT enzyme by both agents with tacrolimus, which was approximately 60-fold more efficient as an inhibitor. Tacrolimus 184-194 UDP glucuronosyltransferase family 1 member A4 Homo sapiens 151-156 10051052-9 1999 The calculated inhibition constants (KI) of tacrolimus and CsA for the purified UDPGT were 27.3+/-5.6 ng/ml and 2,518+/-1473 ng/ml. Tacrolimus 44-54 UDP glucuronosyltransferase family 1 member A4 Homo sapiens 80-85 10051052-12 1999 This finding suggested that the significantly more efficient inhibition of UDPGT by tacrolimus may occur by a more complicated mechanism that is yet to be determined. Tacrolimus 84-94 UDP glucuronosyltransferase family 1 member A4 Homo sapiens 75-80 10083538-0 1999 Inhibition of IL-10 by FK 506 may be responsible for overcoming ongoing allograft rejection in the rat. Tacrolimus 23-29 interleukin 10 Rattus norvegicus 14-19 9890631-8 1999 Immunosuppressing the mice with FK506 to decrease the infiltration of activated T-cells in infected nerves lengthened beta-galactosidase activity to 8 weeks, the longest time point examined. Tacrolimus 32-37 galactosidase, beta 1 Mus musculus 118-136 10433096-8 1999 The autoantibodies inhibited pentamerization of FKBP12 with FK506, calcineurin, calmodulin, and Ca2+ in vitro. Tacrolimus 60-65 FKBP prolyl isomerase 1A Homo sapiens 48-54 10470441-3 1999 It results from these investigations that the molecular mechanism of the CLA action is the same as that of cyclosporin A and FK-506 compound, i.e. it consists in formation of the complex with cyclophilin and inhibition--in this form--of the phosphatase activity of calcineurin. Tacrolimus 125-131 selectin P ligand Homo sapiens 73-76 9884362-3 1999 The IC50 of FK506 in inhibiting IL-2 production in whole blood and isolated PBMC stimulated with PMA and ionomycin measured 1.2 and 0.04 nM, respectively. Tacrolimus 12-17 IL2 Sus scrofa 32-36 9884362-7 1999 For pigs dosed with 0.15 mg/kg, 50% recovery was observed at 38 h and 100% at 72 h. Blood concentrations of FK506 at 50 and 100% recovery of IL-2 production measured 10.8 and 2.2 nM for pigs dosed with 0.05 mg/kg and 6.1 and 1.1 nM for pigs dosed with 0.15 mg/kg, respectively. Tacrolimus 108-113 IL2 Sus scrofa 141-145 9884362-8 1999 These concentrations are severalfold higher than predicted from the IC50 of FK506 for inhibiting IL-2 production in the whole blood assay. Tacrolimus 76-81 IL2 Sus scrofa 97-101 9931093-8 1999 FK 506 decreased eNOS activity and the levels of eNOS mRNA in the aorta (48% and 55%, respectively). Tacrolimus 0-6 nitric oxide synthase 3 Rattus norvegicus 17-21 9931093-8 1999 FK 506 decreased eNOS activity and the levels of eNOS mRNA in the aorta (48% and 55%, respectively). Tacrolimus 0-6 nitric oxide synthase 3 Rattus norvegicus 49-53 9931093-11 1999 These results indicate that FK 506 may increase blood pressure not only by increasing ET-1 production but also by decreasing NO synthesis in the vasculature. Tacrolimus 28-34 endothelin 1 Rattus norvegicus 86-90 9987705-7 1999 Rifampin appears to induce both intestinal and hepatic metabolism of tacrolimus, most likely through induction of CYP3A and P-glycoprotein in the liver and small bowel. Tacrolimus 69-79 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 114-119 9889794-0 1999 Suppressed endothelin-1 production by FK506 and cyclosporin A in ischemia/reperfusion of rat small intestine. Tacrolimus 38-43 endothelin 1 Rattus norvegicus 11-23 9987705-7 1999 Rifampin appears to induce both intestinal and hepatic metabolism of tacrolimus, most likely through induction of CYP3A and P-glycoprotein in the liver and small bowel. Tacrolimus 69-79 ATP binding cassette subfamily B member 1 Homo sapiens 124-138 10067059-7 1999 RFP induces cytochrome P450 3A in the liver to decrease the activities of many drugs, such as cyclosporin A, tacrolimus, protease inhibitor, itraconazole, and clarithromycin. Tacrolimus 109-119 tripartite motif containing 27 Homo sapiens 0-3 9858612-4 1999 FK506, another calcineurin inhibitor, shared the effect. Tacrolimus 0-5 calcineurin binding protein 1 Homo sapiens 15-36 9889794-6 1999 FK506 suppressed ET-1 expression (27.3% +/- 5.2%, 4.1 +/- 2.2 pg/mL), maintained microcirculation (96% +/- 16%), and blood pressure, reduced histologic damage, and improved survival. Tacrolimus 0-5 endothelin 1 Rattus norvegicus 17-21 9889794-10 1999 CONCLUSIONS: FK506 and CsA, particularly the former, maintain microcirculation and protect the tissue from I/R injury by suppressing the production and release of ET-1. Tacrolimus 13-18 endothelin 1 Rattus norvegicus 163-167 9877176-3 1998 The stretch-induced increase in c-src activity was inhibited by FK506, a specific inhibitor for calcineurin, by Gd3+, a blocker for stretch activated channels, and by the extracellular Ca2+ depletion suggesting the involvement of SA channels. Tacrolimus 64-69 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 32-37 10080403-0 1999 Effect of hepatocyte growth factor on tacrolimus-induced nephrotoxicity in spontaneously hypertensive rats. Tacrolimus 38-48 hepatocyte growth factor Rattus norvegicus 10-34 10080403-2 1999 We investigated the effect of hepatocyte growth factor (HGF) on tacrolimus-induced nephrotoxicity in spontaneously hypertensive rats (SHR). Tacrolimus 64-74 hepatocyte growth factor Rattus norvegicus 30-54 10080403-2 1999 We investigated the effect of hepatocyte growth factor (HGF) on tacrolimus-induced nephrotoxicity in spontaneously hypertensive rats (SHR). Tacrolimus 64-74 hepatocyte growth factor Rattus norvegicus 56-59 10080403-5 1999 HGF significantly inhibited the tacrolimus-induced increase in the serum creatinine (SCr) level (P < 0.05). Tacrolimus 32-42 hepatocyte growth factor Rattus norvegicus 0-3 10080403-6 1999 HGF also prevented the tacrolimus-induced loss in body weight. Tacrolimus 23-33 hepatocyte growth factor Rattus norvegicus 0-3 10080403-7 1999 The bromodeoxyuridine (BrdU) index was significantly higher in kidney specimens from the tacrolimus + HGF group. Tacrolimus 89-99 hepatocyte growth factor Rattus norvegicus 102-105 10080403-8 1999 These findings suggest that HGF induces the regeneration of renal tubular cells and suppresses tacrolimus-induced renal toxicity in SHR. Tacrolimus 95-105 hepatocyte growth factor Rattus norvegicus 28-31 9869086-14 1998 CONCLUSIONS: Our findings suggest that a therapeutic approach combining PE and tacrolimus-mycophenolate mofetil rescue has the potential to improve the outcome of AHR. Tacrolimus 79-89 aryl hydrocarbon receptor Homo sapiens 163-166 10363590-8 1999 The percentage of detectable serum IL-2 level was 45% in the allograft control group, but was undetectable in groups treated with the most effective dose of tacrolimus or CyA at days 3 and 6 after grafting. Tacrolimus 157-167 interleukin 2 Rattus norvegicus 35-39 9878113-0 1998 Distinctive calcineurin-dependent (FK506-sensitive) mechanisms regulate the production of the CC chemokines macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, and RANTES vs IL-2 and TNF-alpha by activated human T cells. Tacrolimus 35-40 C-C motif chemokine ligand 3 Homo sapiens 108-152 9878113-0 1998 Distinctive calcineurin-dependent (FK506-sensitive) mechanisms regulate the production of the CC chemokines macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, and RANTES vs IL-2 and TNF-alpha by activated human T cells. Tacrolimus 35-40 C-C motif chemokine ligand 4 Homo sapiens 154-163 9878113-0 1998 Distinctive calcineurin-dependent (FK506-sensitive) mechanisms regulate the production of the CC chemokines macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, and RANTES vs IL-2 and TNF-alpha by activated human T cells. Tacrolimus 35-40 C-C motif chemokine ligand 5 Homo sapiens 169-175 9878113-0 1998 Distinctive calcineurin-dependent (FK506-sensitive) mechanisms regulate the production of the CC chemokines macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, and RANTES vs IL-2 and TNF-alpha by activated human T cells. Tacrolimus 35-40 interleukin 2 Homo sapiens 179-183 9878113-0 1998 Distinctive calcineurin-dependent (FK506-sensitive) mechanisms regulate the production of the CC chemokines macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, and RANTES vs IL-2 and TNF-alpha by activated human T cells. Tacrolimus 35-40 tumor necrosis factor Homo sapiens 188-197 9878113-4 1998 With both modes of activation, FK506 inhibited RANTES production only partially and late during a 3-day culture, whereas it suppressed both MIP-1alpha and MIP-1beta production throughout the culture. Tacrolimus 31-36 C-C motif chemokine ligand 5 Homo sapiens 47-53 9878113-4 1998 With both modes of activation, FK506 inhibited RANTES production only partially and late during a 3-day culture, whereas it suppressed both MIP-1alpha and MIP-1beta production throughout the culture. Tacrolimus 31-36 C-C motif chemokine ligand 4 Homo sapiens 155-164 9878113-5 1998 However, FK506 inhibition was more pronounced on MIP-1beta than MIP-1alpha, especially in CD3/CD28-activated T cells. Tacrolimus 9-14 C-C motif chemokine ligand 4 Homo sapiens 49-58 9878113-5 1998 However, FK506 inhibition was more pronounced on MIP-1beta than MIP-1alpha, especially in CD3/CD28-activated T cells. Tacrolimus 9-14 C-C motif chemokine ligand 3 Homo sapiens 64-74 9878113-5 1998 However, FK506 inhibition was more pronounced on MIP-1beta than MIP-1alpha, especially in CD3/CD28-activated T cells. Tacrolimus 9-14 CD28 molecule Homo sapiens 94-98 9878113-6 1998 Surprisingly, FK506 also significantly reduced MIP-1beta induction by PMA alone. Tacrolimus 14-19 C-C motif chemokine ligand 4 Homo sapiens 47-56 9878202-0 1998 Tacrolimus (FK506) increases neuronal expression of GAP-43 and improves functional recovery after spinal cord injury in rats. Tacrolimus 0-10 growth associated protein 43 Rattus norvegicus 52-58 9843498-5 1998 Overexpression of NPR1 inhibits growth and induces the degradation of TAT2, whereas loss of NPR1 confers resistance to rapamycin and to FK506, an inhibitor of amino acid import. Tacrolimus 136-141 serine/threonine protein kinase NPR1 Saccharomyces cerevisiae S288C 92-96 9884067-4 1998 Cyclosporin A and FK506 caused a concentration-dependent contraction in guinea-pig isolated bronchus, which was significantly attenuated by NK-1 and NK-2 receptor antagonists. Tacrolimus 18-23 substance-K receptor Cavia porcellus 149-162 9878202-0 1998 Tacrolimus (FK506) increases neuronal expression of GAP-43 and improves functional recovery after spinal cord injury in rats. Tacrolimus 12-17 growth associated protein 43 Rattus norvegicus 52-58 9878202-3 1998 In injured animals receiving tacrolimus, the number of neurons expressing GAP-43 mRNA and protein approximately doubled compared to that in injured animals receiving vehicle alone. Tacrolimus 29-39 growth associated protein 43 Rattus norvegicus 74-80 9839278-0 1998 CsA and FK506 up-regulate eNOS expression: role of reactive oxygen species and AP-1. Tacrolimus 8-13 nitric oxide synthase 3 Bos taurus 26-30 9839278-0 1998 CsA and FK506 up-regulate eNOS expression: role of reactive oxygen species and AP-1. Tacrolimus 8-13 Jun proto-oncogene, AP-1 transcription factor subunit Bos taurus 79-83 9839278-1 1998 Cyclosporine A (CsA) and FK506 increase endothelial nitric oxide synthase (eNOS) mRNA expression in cultured bovine aortic endothelial cells (BAEC). Tacrolimus 25-30 nitric oxide synthase 3 Bos taurus 40-73 9839278-1 1998 Cyclosporine A (CsA) and FK506 increase endothelial nitric oxide synthase (eNOS) mRNA expression in cultured bovine aortic endothelial cells (BAEC). Tacrolimus 25-30 nitric oxide synthase 3 Bos taurus 75-79 9865315-0 1998 In vitro transcription and translation of the tumour suppressor protein P53: qualitative and quantitative effects of FK506 and rapamycin. Tacrolimus 117-122 tumor protein p53 Homo sapiens 72-75 9870481-3 1998 We have previously demonstrated an enhancement of agonist-mediated platelet activation by cyclosporine and tacrolimus which was associated with increased phosphorylation of two intracellular platelet proteins, p20 and p40. Tacrolimus 107-117 tubulin polymerization promoting protein family member 3 Homo sapiens 210-213 9870481-3 1998 We have previously demonstrated an enhancement of agonist-mediated platelet activation by cyclosporine and tacrolimus which was associated with increased phosphorylation of two intracellular platelet proteins, p20 and p40. Tacrolimus 107-117 interleukin 9 Homo sapiens 218-221 9831660-10 1998 In this TcFK, 9 out of 15 amino acid residues forming the FK506 binding pocket of human FKBP12 were found. Tacrolimus 58-63 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 88-94 10342736-9 1998 A striking increase in donor MHC class II+ cells was noted in both the spleen and BM of the BM + tacrolimus-treated group compared to either the BM alone, or BM + FL-treated groups. Tacrolimus 97-107 fms related receptor tyrosine kinase 3 ligand Homo sapiens 163-165 10342736-10 1998 Addition of FL treatment to BM + tacrolimus led to a further increase in donor cells in spleen (three-fold at 10 days, and two-fold at 21 days). Tacrolimus 33-43 fms related receptor tyrosine kinase 3 ligand Homo sapiens 12-14 10342736-12 1998 PCR analysis at this time revealed enhanced donor DNA in the BM + FL + tacrolimus group compared to that in the BM + tacrolimus group. Tacrolimus 71-81 fms related receptor tyrosine kinase 3 ligand Homo sapiens 66-68 9824778-9 1998 Cyclosporine and tacrolimus prevent NF-kappa B activation by inhibiting the action of calcineurin, a phosphatase that indirectly induces I kappa B degradation. Tacrolimus 17-27 nuclear factor kappa B subunit 1 Homo sapiens 36-46 9825815-5 1998 RESULTS: With the daily dose of tacrolimus increased by a median 1.89-fold (range: 1.2-5), alanine aminotransferase, bilirubin, and gamma-glutamyltranspeptidase levels rapidly reached normal values within the first month. Tacrolimus 32-42 inactive glutathione hydrolase 2 Homo sapiens 132-160 9865597-6 1998 IL-4 and MCP-I release was inhibited by treatment with the immunosuppressant FK-506 and actinomycin D. Tacrolimus 77-83 interleukin 4 Rattus norvegicus 0-4 9865597-7 1998 Therefore, in our system IL-4 and MCP-1 release involves Ca2(+)-dependent and FK-506-sensitive signaling pathways. Tacrolimus 78-84 interleukin 4 Rattus norvegicus 25-29 9865597-7 1998 Therefore, in our system IL-4 and MCP-1 release involves Ca2(+)-dependent and FK-506-sensitive signaling pathways. Tacrolimus 78-84 mast cell protease 1-like 1 Rattus norvegicus 34-39 9820545-6 1998 IL-4 mRNA, constitutively present in basophils, was increased after stimulation by pFv and was inhibited by cyclosporin A and tacrolimus. Tacrolimus 126-136 interleukin 4 Homo sapiens 0-4 9844139-10 1998 CsA and tacrolimus (0.5 to 5000 microgram/liter) induced a significant, dose-dependent increase of ET-1 and ET-3 release by PT-1 cells with a maximum stimulation at a CsA concentration of 500 microgram/liter (P < 0.001) and a tacrolimus concentration of 50 microgram/liter (P < 0.001). Tacrolimus 8-18 endothelin-1 Oryctolagus cuniculus 99-112 9844139-12 1998 Coincubation of PT-1 cells with CsA or tacrolimus and HGF or EGF also resulted in a marked reduction of ET-1 release. Tacrolimus 39-49 endothelin-1 Oryctolagus cuniculus 104-108 9844139-13 1998 CONCLUSIONS: The present data suggest that ET-1 and ET-3 release by cultured rabbit proximal tubule cells are regulated differently, and that the stimulatory effect of CsA and tacrolimus on ET-1 release is antagonized by HGF and EGF. Tacrolimus 176-186 endothelin-1 Oryctolagus cuniculus 43-56 9844139-13 1998 CONCLUSIONS: The present data suggest that ET-1 and ET-3 release by cultured rabbit proximal tubule cells are regulated differently, and that the stimulatory effect of CsA and tacrolimus on ET-1 release is antagonized by HGF and EGF. Tacrolimus 176-186 endothelin-1 Oryctolagus cuniculus 43-47 9844139-13 1998 CONCLUSIONS: The present data suggest that ET-1 and ET-3 release by cultured rabbit proximal tubule cells are regulated differently, and that the stimulatory effect of CsA and tacrolimus on ET-1 release is antagonized by HGF and EGF. Tacrolimus 176-186 hepatocyte growth factor Oryctolagus cuniculus 221-224 9844139-13 1998 CONCLUSIONS: The present data suggest that ET-1 and ET-3 release by cultured rabbit proximal tubule cells are regulated differently, and that the stimulatory effect of CsA and tacrolimus on ET-1 release is antagonized by HGF and EGF. Tacrolimus 176-186 pro-epidermal growth factor Oryctolagus cuniculus 229-232 9808496-1 1998 BACKGROUND: Although it is important to maintain an appropriate blood concentration of FK506 after liver transplantation, significant interindividual variability in the actual FK506 dosage has been observed, presumably due to the wide variability of cytochrome P450 3A4 activity in liver microsomes. Tacrolimus 176-181 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 250-269 9762012-0 1998 Cellular signaling mechanisms for stimulation of growth hormone secretion and growth hormone primary transcripts by immunosuppressant agents, FK506 and cyclosporin A, in cultured rat pituitary cells. Tacrolimus 142-147 gonadotropin releasing hormone receptor Rattus norvegicus 78-92 9762012-1 1998 Although an immunosuppressant, FK506, has been known to stimulate growth hormone (GH) release from rat somatotropes, the cellular signaling mechanism is unknown. Tacrolimus 31-36 gonadotropin releasing hormone receptor Rattus norvegicus 66-80 9762012-1 1998 Although an immunosuppressant, FK506, has been known to stimulate growth hormone (GH) release from rat somatotropes, the cellular signaling mechanism is unknown. Tacrolimus 31-36 gonadotropin releasing hormone receptor Rattus norvegicus 82-84 9762012-4 1998 FK506 and CsA increased GH release in a dose-dependent manner and inhibited calcineurin (CaN) activity in the cultured pituitary cells. Tacrolimus 0-5 gonadotropin releasing hormone receptor Rattus norvegicus 24-26 9762012-5 1998 The third immunosuppressant, rapamycin (RP), inhibited the FK506-induced GH release, although RP alone had no effect. Tacrolimus 59-64 gonadotropin releasing hormone receptor Rattus norvegicus 73-75 9762012-6 1998 Protein kinase A (PKA) inhibitors, H-89 and HA-1004 and EGTA blocked FK506- and CsA-induced GH release. Tacrolimus 69-74 protein kinase cAMP-activated catalytic subunit alpha Rattus norvegicus 0-16 9762012-6 1998 Protein kinase A (PKA) inhibitors, H-89 and HA-1004 and EGTA blocked FK506- and CsA-induced GH release. Tacrolimus 69-74 protein kinase cAMP-activated catalytic subunit alpha Rattus norvegicus 18-21 9762012-7 1998 TGF-beta did not alter basal GH release, but inhibited FK506-induced GH release. Tacrolimus 55-60 transforming growth factor, beta 1 Rattus norvegicus 0-8 9762012-8 1998 GH primary transcripts were increased by FK506, and the effects were blocked by H-89 and HA-1004. Tacrolimus 41-46 gonadotropin releasing hormone receptor Rattus norvegicus 0-2 9762012-9 1998 These results suggest that the immunosuppressants, FK506 and CsA, stimulate GH release by inhibiting CaN activity which results in the activation of the PKA system in the rat somatotropes. Tacrolimus 51-56 gonadotropin releasing hormone receptor Rattus norvegicus 76-78 9762012-9 1998 These results suggest that the immunosuppressants, FK506 and CsA, stimulate GH release by inhibiting CaN activity which results in the activation of the PKA system in the rat somatotropes. Tacrolimus 51-56 protein kinase cAMP-activated catalytic subunit alpha Rattus norvegicus 153-156 9762012-10 1998 TGF-beta receptors might be involved in FK506-induced GH release as a separate pathway. Tacrolimus 40-45 transforming growth factor, beta 1 Rattus norvegicus 0-8 9762012-10 1998 TGF-beta receptors might be involved in FK506-induced GH release as a separate pathway. Tacrolimus 40-45 gonadotropin releasing hormone receptor Rattus norvegicus 54-56 9762012-11 1998 FK506 also stimulates GH primary transcripts via a PKA-dependent mechanism in a manner similar to its effects on GH release. Tacrolimus 0-5 gonadotropin releasing hormone receptor Rattus norvegicus 22-24 9762012-11 1998 FK506 also stimulates GH primary transcripts via a PKA-dependent mechanism in a manner similar to its effects on GH release. Tacrolimus 0-5 protein kinase cAMP-activated catalytic subunit alpha Rattus norvegicus 51-54 9808496-5 1998 The FK506 blood clearance (CLss) was calculated from its constant infusion rate and steady-state blood concentration on day 4 after transplantation in 17 recipients. Tacrolimus 4-9 LDL receptor related protein 4 Homo sapiens 27-31 9808496-7 1998 The mean CLss of FK506 was 22.1+/-10.8 ml/min (10.1-45.2 ml/min). Tacrolimus 17-22 LDL receptor related protein 4 Homo sapiens 9-13 9808496-9 1998 A significant correlation was observed between the in vitro estimated total metabolic ability of the graft for FK506 (M-I formation rate x graft weight) and the in vivo CLss of FK506 (r=0.770, P<0.001). Tacrolimus 177-182 LDL receptor related protein 4 Homo sapiens 169-173 9808502-1 1998 BACKGROUND/AIMS: Tacrolimus is metabolized by cytochrome P450 3A4 and 2D6 and has a narrow therapeutic range. Tacrolimus 17-27 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 46-73 9800954-8 1998 In mixed lymphocyte cultures performed with peripheral blood mononuclear cells (PBMC) from normal subjects, IL-10 decreased the cell proliferation in a dose-dependent manner, and this immunosuppression was synergistic with that of cyclosporine or FK506. Tacrolimus 247-252 interleukin 10 Homo sapiens 108-113 9809157-4 1998 In the area of therapeutics, recombinant growth hormone was shown to improve statural growth of children with chronic renal failure; in addition, renal transplantation benefits from new immunosuppressants as tacrolimus and mycophenolate mofetil. Tacrolimus 208-218 growth hormone 1 Homo sapiens 41-55 9767529-1 1998 BACKGROUND: The immunosuppressive drugs cyclosporine A (CsA) and tacrolimus (FK506) are extruded from cells by the multidrug resistance P-glycoprotein (P-gp), an efflux pump for drugs and xenobiotics, which may limit their therapeutic effectiveness and/or incidence of toxic side effects. Tacrolimus 65-75 ATP binding cassette subfamily B member 1 Homo sapiens 136-150 9767529-1 1998 BACKGROUND: The immunosuppressive drugs cyclosporine A (CsA) and tacrolimus (FK506) are extruded from cells by the multidrug resistance P-glycoprotein (P-gp), an efflux pump for drugs and xenobiotics, which may limit their therapeutic effectiveness and/or incidence of toxic side effects. Tacrolimus 65-75 ATP binding cassette subfamily B member 1 Homo sapiens 152-156 9767529-1 1998 BACKGROUND: The immunosuppressive drugs cyclosporine A (CsA) and tacrolimus (FK506) are extruded from cells by the multidrug resistance P-glycoprotein (P-gp), an efflux pump for drugs and xenobiotics, which may limit their therapeutic effectiveness and/or incidence of toxic side effects. Tacrolimus 77-82 ATP binding cassette subfamily B member 1 Homo sapiens 136-150 9767529-1 1998 BACKGROUND: The immunosuppressive drugs cyclosporine A (CsA) and tacrolimus (FK506) are extruded from cells by the multidrug resistance P-glycoprotein (P-gp), an efflux pump for drugs and xenobiotics, which may limit their therapeutic effectiveness and/or incidence of toxic side effects. Tacrolimus 77-82 ATP binding cassette subfamily B member 1 Homo sapiens 152-156 9767529-2 1998 In the present study, we investigated the effect of therapeutic concentrations of CsA and FK506 on the expression of P-gp in cultured endothelial and proximal tubule cells. Tacrolimus 90-95 ATP binding cassette subfamily B member 1 Homo sapiens 117-121 9767529-8 1998 CONCLUSIONS: The data suggest that the induction of P-gp expression in HAEC and RPTC at concentrations of CsA or FK506 above 0.5 microM is part of the protective answer of cells to toxic concentrations of the drugs and could therefore interfere with the therapeutic effectiveness of CsA in vivo. Tacrolimus 113-118 ATP binding cassette subfamily B member 1 Homo sapiens 52-56 9872566-7 1998 Immunosuppressants FK506 and cyclosporin A suppressed the K252a-induced cell adhesion and abolished tyrosine phosphorylation of cellular proteins including FAK and paxillin. Tacrolimus 19-24 protein tyrosine kinase 2 Homo sapiens 156-159 9800954-10 1998 Thus, IL-10 therapy in association with cyclosporine or FK506 might be proposed after liver transplantation. Tacrolimus 56-61 interleukin 10 Homo sapiens 6-11 9727000-4 1998 FK506.FKBP12 drug-immunophilin complexes inhibited the interaction of NFAT with activated calcineurin much more effectively than they inhibited the interaction with inactive calcineurin, suggesting that part of the interaction with activated calcineurin involved the enzyme active site. Tacrolimus 0-5 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 6-12 9771754-5 1998 FK 506 immunosuppressed mice were transplanted by splenic injection with Bcl-2 hepatocytes. Tacrolimus 0-6 B cell leukemia/lymphoma 2 Mus musculus 73-78 9716519-7 1998 In addition, cFKBP/SMAP-induced smooth muscle differentiation was inhibited by FK506. Tacrolimus 79-84 FK506 binding protein 9 Gallus gallus 13-23 9733519-3 1998 Administration of the calcineurin inhibitors cyclosporin and FK506 prevented disease in mice that were genetically predisposed to develop HCM as a result of aberrant expression of tropomodulin, myosin light chain-2, or fetal beta-tropomyosin in the heart. Tacrolimus 61-66 myosin light chain, phosphorylatable, fast skeletal muscle Mus musculus 194-214 9743407-2 1998 The immunosuppressants FK506 and rapamycin both bind to FKBP12 and in turn dissociate the protein from the ryanodine receptor. Tacrolimus 23-28 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 56-62 9709036-1 1998 The immunosuppressive fungal products cyclosporin A (CsA) and FK506 bind with high affinity to intracellular receptor proteins: cyclophilin (CYP) is one of the receptors for CsA and FK506-binding protein (FKBP) is one of the receptors for FK506. Tacrolimus 62-67 peptidylprolyl isomerase G Homo sapiens 128-139 9709036-1 1998 The immunosuppressive fungal products cyclosporin A (CsA) and FK506 bind with high affinity to intracellular receptor proteins: cyclophilin (CYP) is one of the receptors for CsA and FK506-binding protein (FKBP) is one of the receptors for FK506. Tacrolimus 62-67 peptidylprolyl isomerase G Homo sapiens 141-144 9709036-1 1998 The immunosuppressive fungal products cyclosporin A (CsA) and FK506 bind with high affinity to intracellular receptor proteins: cyclophilin (CYP) is one of the receptors for CsA and FK506-binding protein (FKBP) is one of the receptors for FK506. Tacrolimus 62-67 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 174-177 9709036-1 1998 The immunosuppressive fungal products cyclosporin A (CsA) and FK506 bind with high affinity to intracellular receptor proteins: cyclophilin (CYP) is one of the receptors for CsA and FK506-binding protein (FKBP) is one of the receptors for FK506. Tacrolimus 182-187 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 53-56 9709036-1 1998 The immunosuppressive fungal products cyclosporin A (CsA) and FK506 bind with high affinity to intracellular receptor proteins: cyclophilin (CYP) is one of the receptors for CsA and FK506-binding protein (FKBP) is one of the receptors for FK506. Tacrolimus 182-187 peptidylprolyl isomerase G Homo sapiens 128-139 9709036-1 1998 The immunosuppressive fungal products cyclosporin A (CsA) and FK506 bind with high affinity to intracellular receptor proteins: cyclophilin (CYP) is one of the receptors for CsA and FK506-binding protein (FKBP) is one of the receptors for FK506. Tacrolimus 182-187 peptidylprolyl isomerase G Homo sapiens 141-144 9709036-4 1998 Here, we examine the effects of CsA and FK506 and show that the former inhibits PPIase activity in schistosome extracts, whereas the latter does not. Tacrolimus 40-45 FKBP prolyl isomerase 7 Homo sapiens 80-86 9709036-6 1998 The observation that CsA was significantly more effective than FK506 as an antischistosomal agent, both in vivo and in vitro raises the possibility that killing of schistosomes is caused by the inhibition of schistosome CYP PPIase. Tacrolimus 63-68 peptidylprolyl isomerase G Homo sapiens 220-223 9709036-6 1998 The observation that CsA was significantly more effective than FK506 as an antischistosomal agent, both in vivo and in vitro raises the possibility that killing of schistosomes is caused by the inhibition of schistosome CYP PPIase. Tacrolimus 63-68 FKBP prolyl isomerase 7 Homo sapiens 224-230 9767457-7 1998 Moreover extended studies demonstrated that in cultured rat spleen cells the expression of lymphotactin mRNA was markedly induced by phytohaemagglutinin (PHA) or phorbol myristate acetate (PMA), and such induction was inhibited by the immunosuppressive drugs FK506 and cyclosporin. Tacrolimus 259-264 X-C motif chemokine ligand 1 Rattus norvegicus 91-103 9725828-5 1998 However, Std1p functions in a calcineurin-independent ion stress response pathway, since a std1 mth1 mutant is FK506 sensitive under conditions of ion stress. Tacrolimus 111-116 Std1p Saccharomyces cerevisiae S288C 9-14 9725828-5 1998 However, Std1p functions in a calcineurin-independent ion stress response pathway, since a std1 mth1 mutant is FK506 sensitive under conditions of ion stress. Tacrolimus 111-116 Std1p Saccharomyces cerevisiae S288C 91-95 9725828-5 1998 However, Std1p functions in a calcineurin-independent ion stress response pathway, since a std1 mth1 mutant is FK506 sensitive under conditions of ion stress. Tacrolimus 111-116 Mth1p Saccharomyces cerevisiae S288C 96-100 9782077-6 1998 FKBP6 has a putative N-terminal FK-506 binding and peptidylproyl isomerase (rotamase) domain and, like known high-molecular-weight FKBPs, an imperfect C-terminal tetratricopeptide repeat domain. Tacrolimus 32-38 FKBP prolyl isomerase family member 6 (inactive) Homo sapiens 0-5 9792034-6 1998 FK506 administration attenuated hepatic-mRNA expression of interleukin-2 and both superoxide anions as well as tumour necrosis factor-alpha production by hepatic macrophages, but did not change CD14-mRNA expression in Propionibacterium acnes-treated rats. Tacrolimus 0-5 interleukin 2 Rattus norvegicus 59-72 9751843-3 1998 In this simple working hypothesis, mechanisms of clinical amelioration of atopic diseases manifested by glucocorticoids or immunosuppressants such as cyclosporin A or FK506 were easily understood to stem from the inhibition of IL-5 production and gene transcription. Tacrolimus 167-172 interleukin 5 Homo sapiens 227-231 9792034-8 1998 FK506 seems to attenuate such activation by suppressing hepatic interleukin-2 expression, without affecting CD14 expression on the cells. Tacrolimus 0-5 interleukin 2 Rattus norvegicus 64-77 9761470-14 1998 Analysis of the FK506 binding protein, and of compounds developed in SAR by NMR (Shuker SB et al., 1996, Science 274:1531-1534), indicates that CAST pocket computation may provide a priori identification of target proteins for linked-fragment design. Tacrolimus 16-21 calpastatin Homo sapiens 144-148 28976697-6 1998 FK506 administration attenuated hepatic-mRNA expression of interleukin-2 and both superoxide anions as well as tumour necrosis factor-alpha production by hepatic macrophages, but did not change CD14-mRNA expression in Propionibacterium acnes-treated rats. Tacrolimus 0-5 interleukin 2 Rattus norvegicus 59-72 28976697-8 1998 FK506 seems to attenuate such activation by suppressing hepatic interleukin-2 expression, without affecting CD14 expression on the cells. Tacrolimus 0-5 interleukin 2 Rattus norvegicus 64-77 9873523-1 1998 A series of C32-O-aralkyl ether derivatives of the FK-506 related macrolide ascomycin have been prepared based on an earlier reported C32-O-cinnamyl ether design. Tacrolimus 51-57 chemokine like factor Homo sapiens 12-15 9807817-0 1998 An Arabidopsis immunophilin, AtFKBP12, binds to AtFIP37 (FKBP interacting protein) in an interaction that is disrupted by FK506. Tacrolimus 122-127 FK506-binding protein 12 Arabidopsis thaliana 15-27 9807817-0 1998 An Arabidopsis immunophilin, AtFKBP12, binds to AtFIP37 (FKBP interacting protein) in an interaction that is disrupted by FK506. Tacrolimus 122-127 FK506-binding protein 12 Arabidopsis thaliana 29-37 9807817-0 1998 An Arabidopsis immunophilin, AtFKBP12, binds to AtFIP37 (FKBP interacting protein) in an interaction that is disrupted by FK506. Tacrolimus 122-127 FKBP12 interacting protein 37 Arabidopsis thaliana 48-55 9807817-5 1998 The interaction between AtFKBP12 and AtFIP37 in the 2-hybrid system, as assessed by histidine auxotrophy and beta-galactosidase activity, was disrupted by FK506, but not by cyclosporin A, a drug that binds to cyclophilin A. Tacrolimus 155-160 FK506-binding protein 12 Arabidopsis thaliana 24-32 9807817-5 1998 The interaction between AtFKBP12 and AtFIP37 in the 2-hybrid system, as assessed by histidine auxotrophy and beta-galactosidase activity, was disrupted by FK506, but not by cyclosporin A, a drug that binds to cyclophilin A. Tacrolimus 155-160 FKBP12 interacting protein 37 Arabidopsis thaliana 37-44 9807817-5 1998 The interaction between AtFKBP12 and AtFIP37 in the 2-hybrid system, as assessed by histidine auxotrophy and beta-galactosidase activity, was disrupted by FK506, but not by cyclosporin A, a drug that binds to cyclophilin A. Tacrolimus 155-160 beta-galactosidase Arabidopsis thaliana 109-127 9712905-11 1998 Dephosphorylation of CRHSP-24 was completely inhibited by pretreatment of acini with cyclosporin A or FK506. Tacrolimus 102-107 calcium regulated heat stable protein 1 Rattus norvegicus 21-29 9807817-7 1998 The binding was abolished by prior incubation of AtFKBP12 with FK506. Tacrolimus 63-68 FK506-binding protein 12 Arabidopsis thaliana 49-57 9807817-8 1998 These findings indicate that an Arabidopsis FKBP12 ortholog encodes a protein that binds FK506 and that the interaction between AtFKBP12 and AtFIP37 may involve the FK506 binding site of AtFKBP12. Tacrolimus 89-94 FK506-binding protein 12 Arabidopsis thaliana 44-50 9807817-8 1998 These findings indicate that an Arabidopsis FKBP12 ortholog encodes a protein that binds FK506 and that the interaction between AtFKBP12 and AtFIP37 may involve the FK506 binding site of AtFKBP12. Tacrolimus 89-94 FK506-binding protein 12 Arabidopsis thaliana 128-136 9807817-8 1998 These findings indicate that an Arabidopsis FKBP12 ortholog encodes a protein that binds FK506 and that the interaction between AtFKBP12 and AtFIP37 may involve the FK506 binding site of AtFKBP12. Tacrolimus 89-94 FKBP12 interacting protein 37 Arabidopsis thaliana 141-148 9807817-8 1998 These findings indicate that an Arabidopsis FKBP12 ortholog encodes a protein that binds FK506 and that the interaction between AtFKBP12 and AtFIP37 may involve the FK506 binding site of AtFKBP12. Tacrolimus 89-94 FK506-binding protein 12 Arabidopsis thaliana 187-195 9807817-8 1998 These findings indicate that an Arabidopsis FKBP12 ortholog encodes a protein that binds FK506 and that the interaction between AtFKBP12 and AtFIP37 may involve the FK506 binding site of AtFKBP12. Tacrolimus 165-170 FK506-binding protein 12 Arabidopsis thaliana 44-50 9807817-8 1998 These findings indicate that an Arabidopsis FKBP12 ortholog encodes a protein that binds FK506 and that the interaction between AtFKBP12 and AtFIP37 may involve the FK506 binding site of AtFKBP12. Tacrolimus 165-170 FK506-binding protein 12 Arabidopsis thaliana 128-136 9807817-8 1998 These findings indicate that an Arabidopsis FKBP12 ortholog encodes a protein that binds FK506 and that the interaction between AtFKBP12 and AtFIP37 may involve the FK506 binding site of AtFKBP12. Tacrolimus 165-170 FKBP12 interacting protein 37 Arabidopsis thaliana 141-148 9807817-8 1998 These findings indicate that an Arabidopsis FKBP12 ortholog encodes a protein that binds FK506 and that the interaction between AtFKBP12 and AtFIP37 may involve the FK506 binding site of AtFKBP12. Tacrolimus 165-170 FK506-binding protein 12 Arabidopsis thaliana 187-195 9873523-1 1998 A series of C32-O-aralkyl ether derivatives of the FK-506 related macrolide ascomycin have been prepared based on an earlier reported C32-O-cinnamyl ether design. Tacrolimus 51-57 chemokine like factor Homo sapiens 134-137 9729383-0 1998 MAP kinase phosphatase 1 is expressed and enhanced by FK506 in surviving mamillary, but not degenerating nigral neurons following axotomy. Tacrolimus 54-59 dual specificity phosphatase 1 Rattus norvegicus 0-24 9705862-5 1998 FK506 caused a moderate increase in ALP activity and a decreased expression of osteopontin mRNA. Tacrolimus 0-5 secreted phosphoprotein 1 Rattus norvegicus 79-90 9729383-8 1998 Subcutaneous injection of the immunosuppressant FK506 that protects axotomized SNC neurons against neuronal cell death, enhanced the expression of MKP-1 in CMm, but failed to do so in SNC neurons. Tacrolimus 48-53 dual specificity phosphatase 1 Rattus norvegicus 147-152 9754866-7 1998 TNF-alpha release induced by DTBHQ and CPA was inhibited by treatment with actinomycin-D, the immunosuppressant FK506 and the glucocorticoid dexamethasone (p < or = 0.01). Tacrolimus 112-117 tumor necrosis factor Rattus norvegicus 0-9 9687567-8 1998 Of the mRNAs tested, only those encoding monocyte chemotactic protein-1 (approximately 2-fold over basal) and interleukin-8 (approximately 6-fold over basal) are induced by histamine; both of these responses are suppressed by CsA and FK506. Tacrolimus 234-239 C-C motif chemokine ligand 2 Homo sapiens 41-71 9687567-8 1998 Of the mRNAs tested, only those encoding monocyte chemotactic protein-1 (approximately 2-fold over basal) and interleukin-8 (approximately 6-fold over basal) are induced by histamine; both of these responses are suppressed by CsA and FK506. Tacrolimus 234-239 C-X-C motif chemokine ligand 8 Homo sapiens 110-123 9658403-7 1998 Indeed, Northern analysis showed that Rap, KN62, and, to a lesser degree, FK506 inhibited progestin stimulation of Cyclin D1 mRNA levels, but not those of the non-steroid-regulated glyceraldehyde 3-phosphate dehydrogenase (GAPDH) gene. Tacrolimus 74-79 cyclin D1 Homo sapiens 115-124 9742506-1 1998 BACKGROUND: The 12 kD FK506 binding protein FKBP12 is a cytosolic receptor for the immunosuppressant drugs FK506 and rapamycin. Tacrolimus 22-27 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 44-50 9742506-3 1998 We investigated a role for FKBP12 in male reproductive physiology on the basis of our identification of extremely high levels of [3H]FK506 binding in male reproductive tissues. Tacrolimus 133-138 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 27-33 9742506-12 1998 FKBP12 was purified from epididymal plasma by FK506 affinity chromatography. Tacrolimus 46-51 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 0-6 9785975-8 1998 Although close monitoring of FK506 blood concentration and patient clinical signs are required, we concluded that FK506 is effective for GVHD prophylaxis after bone marrow transplantation from HLA-A, B, DR genotypically mismatched unrelated donors, and that adverse reactions due to FK506 are controllable. Tacrolimus 114-119 major histocompatibility complex, class I, A Homo sapiens 193-201 9785975-8 1998 Although close monitoring of FK506 blood concentration and patient clinical signs are required, we concluded that FK506 is effective for GVHD prophylaxis after bone marrow transplantation from HLA-A, B, DR genotypically mismatched unrelated donors, and that adverse reactions due to FK506 are controllable. Tacrolimus 114-119 major histocompatibility complex, class I, A Homo sapiens 193-201 9721433-1 1998 Based on the findings above, a number of conclusions can be made regarding the distribution, pharmacokinetics, and therapeutic range investigations with RAPA: (a) the majority of the drug is sequestered in erythrocytes, resulting in whole blood concentrations being considerably higher than plasma concentrations; (b) the drug is metabolized by the same cytochrome P450 3A enzyme involved in the metabolism of CsA and FK506. Tacrolimus 418-423 transcriptional regulating factor 1 Homo sapiens 153-157 9715421-3 1998 Using the same model, we examined the effect of FK506 on the expression of hepatocyte growth factor (HGF) and transforming growth factor-beta 1 (TGF-beta 1) and found no changes in HGF and TGF-beta 1 mRNA expression in the liver or in the HGF protein concentration in plasma. Tacrolimus 48-53 hepatocyte growth factor Homo sapiens 75-99 9715421-3 1998 Using the same model, we examined the effect of FK506 on the expression of hepatocyte growth factor (HGF) and transforming growth factor-beta 1 (TGF-beta 1) and found no changes in HGF and TGF-beta 1 mRNA expression in the liver or in the HGF protein concentration in plasma. Tacrolimus 48-53 hepatocyte growth factor Homo sapiens 101-104 9715421-3 1998 Using the same model, we examined the effect of FK506 on the expression of hepatocyte growth factor (HGF) and transforming growth factor-beta 1 (TGF-beta 1) and found no changes in HGF and TGF-beta 1 mRNA expression in the liver or in the HGF protein concentration in plasma. Tacrolimus 48-53 transforming growth factor beta 1 Homo sapiens 110-143 9715421-3 1998 Using the same model, we examined the effect of FK506 on the expression of hepatocyte growth factor (HGF) and transforming growth factor-beta 1 (TGF-beta 1) and found no changes in HGF and TGF-beta 1 mRNA expression in the liver or in the HGF protein concentration in plasma. Tacrolimus 48-53 transforming growth factor beta 1 Homo sapiens 145-155 9658403-12 1998 The nuclear 9S-[3H]Org2058-PR resulting from cells exposed to Rap, contained, in addition to the heat shock proteins of 90 kDa and 70 kDa (hsp90 and hsp70), the FK506-binding immunophilin FKBP52 but not FKBP51, although the latter was part of unliganded PR heterocomplex associated with hsp90. Tacrolimus 161-166 progesterone receptor Homo sapiens 27-29 9658403-12 1998 The nuclear 9S-[3H]Org2058-PR resulting from cells exposed to Rap, contained, in addition to the heat shock proteins of 90 kDa and 70 kDa (hsp90 and hsp70), the FK506-binding immunophilin FKBP52 but not FKBP51, although the latter was part of unliganded PR heterocomplex associated with hsp90. Tacrolimus 161-166 FKBP prolyl isomerase 4 Homo sapiens 188-194 9658403-12 1998 The nuclear 9S-[3H]Org2058-PR resulting from cells exposed to Rap, contained, in addition to the heat shock proteins of 90 kDa and 70 kDa (hsp90 and hsp70), the FK506-binding immunophilin FKBP52 but not FKBP51, although the latter was part of unliganded PR heterocomplex associated with hsp90. Tacrolimus 161-166 progesterone receptor Homo sapiens 254-256 9658403-12 1998 The nuclear 9S-[3H]Org2058-PR resulting from cells exposed to Rap, contained, in addition to the heat shock proteins of 90 kDa and 70 kDa (hsp90 and hsp70), the FK506-binding immunophilin FKBP52 but not FKBP51, although the latter was part of unliganded PR heterocomplex associated with hsp90. Tacrolimus 161-166 heat shock protein 90 alpha family class A member 1 Homo sapiens 287-292 9723345-0 1998 Clinical use of the euglycemic hyperinsulinemic clamp for diagnosis of tacrolimus-induced insulin resistance after combined pancreas-kidney transplantation. Tacrolimus 71-81 insulin Homo sapiens 36-43 9675023-1 1998 The calcium- and calmodulin-activated protein phosphatase calcineurin (CN) is the target for the immunosuppressive drugs FK506 and cyclosporin A (CsA) when bound to their intracellular receptor proteins, the immunophilins known as FK506-binding protein (FKBP) and cyclophilin A (CypA), respectively. Tacrolimus 121-126 peptidylprolyl isomerase A Homo sapiens 264-277 9675023-1 1998 The calcium- and calmodulin-activated protein phosphatase calcineurin (CN) is the target for the immunosuppressive drugs FK506 and cyclosporin A (CsA) when bound to their intracellular receptor proteins, the immunophilins known as FK506-binding protein (FKBP) and cyclophilin A (CypA), respectively. Tacrolimus 121-126 peptidylprolyl isomerase A Homo sapiens 279-283 9675023-10 1998 Inhibition of CN by the CsA x CypA complex was not time-dependent, but the data did conform to a competitive inhibition model like FK506 x FKBP. Tacrolimus 131-136 peptidylprolyl isomerase A Homo sapiens 30-34 9688583-0 1998 FK506 (tacrolimus) inhibits extravasation of lymphoid cells by abrogating VLA-4/VCAM-1 mediated transendothelial migration. Tacrolimus 0-5 vascular cell adhesion molecule 1 Homo sapiens 80-86 9688583-0 1998 FK506 (tacrolimus) inhibits extravasation of lymphoid cells by abrogating VLA-4/VCAM-1 mediated transendothelial migration. Tacrolimus 7-17 vascular cell adhesion molecule 1 Homo sapiens 80-86 9688583-4 1998 FK506 may exert its potent immunosuppressive action partly by inhibiting VLA-4/VCAM-1 mediated transendothelial migration or insinuation of lymphoid cells to tissues. Tacrolimus 0-5 vascular cell adhesion molecule 1 Homo sapiens 79-85 9749865-7 1998 Tacrolimus also significantly suppressed the production of IL-8 and GM-CSF by the TCCs. Tacrolimus 0-10 C-X-C motif chemokine ligand 8 Homo sapiens 59-63 9749865-7 1998 Tacrolimus also significantly suppressed the production of IL-8 and GM-CSF by the TCCs. Tacrolimus 0-10 colony stimulating factor 2 Homo sapiens 68-74 9605132-0 1998 Topical FK506 suppresses cytokine and costimulatory molecule expression in epidermal and local draining lymph node cells during primary skin immune responses. Tacrolimus 8-13 CD276 antigen Mus musculus 38-60 9662091-11 1998 However, in the cyclosporine- and FK506-treated groups, the PCNA index in bronchial surface epithelium was suppressed to less than 5% at 3 and 5 days. Tacrolimus 34-39 proliferating cell nuclear antigen Rattus norvegicus 60-64 9662091-12 1998 Even at 50 days after transplantation, PCNA-positive cells were rare in bronchial epithelium of FK506-treated grafts. Tacrolimus 96-101 proliferating cell nuclear antigen Rattus norvegicus 39-43 9655420-11 1998 Flap necrosis and the increase in malondialdehyde, myeloperoxidase, and neutrophil infiltration were improved by FK506 pretreatment, a neutrophil modulating agent. Tacrolimus 113-118 myeloperoxidase Rattus norvegicus 51-66 9642110-5 1998 FK506 inhibited the CD8 downregulation in both cell types. Tacrolimus 0-5 CD8a molecule Homo sapiens 20-23 9605132-6 1998 Further analysis of the LNC cytokine pattern revealed that the production of both Thl (IFN-gamma and IL-2) and Th2 (IL-4) cytokines was dramatically impaired after topical FK506 treatment. Tacrolimus 172-177 interferon gamma Mus musculus 87-96 9605132-6 1998 Further analysis of the LNC cytokine pattern revealed that the production of both Thl (IFN-gamma and IL-2) and Th2 (IL-4) cytokines was dramatically impaired after topical FK506 treatment. Tacrolimus 172-177 interleukin 2 Mus musculus 101-105 9605132-6 1998 Further analysis of the LNC cytokine pattern revealed that the production of both Thl (IFN-gamma and IL-2) and Th2 (IL-4) cytokines was dramatically impaired after topical FK506 treatment. Tacrolimus 172-177 heart and neural crest derivatives expressed 2 Mus musculus 111-114 9605132-6 1998 Further analysis of the LNC cytokine pattern revealed that the production of both Thl (IFN-gamma and IL-2) and Th2 (IL-4) cytokines was dramatically impaired after topical FK506 treatment. Tacrolimus 172-177 interleukin 4 Mus musculus 116-120 9605132-7 1998 Flow cytometric analysis showed that topical FK506 decreased the population of epidermis-infiltrating CD4+ T cells and suppressed the expression of CD54 and CD80 on I-A+ EC and LNC during hapten-induced contact hypersensitivity. Tacrolimus 45-50 intercellular adhesion molecule 1 Mus musculus 148-152 9573223-6 1998 However, the combination of FK506 plus CTLA4Ig abrogated the immune response against adenovirus proteins and dystrophin to a degree not achievable with the use of either agent alone. Tacrolimus 28-33 dystrophin, muscular dystrophy Mus musculus 109-119 9605132-7 1998 Flow cytometric analysis showed that topical FK506 decreased the population of epidermis-infiltrating CD4+ T cells and suppressed the expression of CD54 and CD80 on I-A+ EC and LNC during hapten-induced contact hypersensitivity. Tacrolimus 45-50 CD80 antigen Mus musculus 157-161 9605132-8 1998 Furthermore, topical FK506 profoundly impaired oxazolone-induced up-regulation of CD25 expression on CD4+ LNC and dramatically decreased hapten-induced expansion of I-A+/B220+ and I-A+/CD69+ LNC subsets. Tacrolimus 21-26 interleukin 2 receptor, alpha chain Mus musculus 82-86 9636495-0 1998 Tacrolimus therapy for ABO-incompatible kidney transplantation. Tacrolimus 0-10 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 23-26 9606972-2 1998 Here we report that both cobalt and the calcium chelator EGTA, inhibitors of calcium uptake, as well as cyclosporin A and FK-506, specific inhibitors of calcineurin function, abolished fibroblast growth factor (FGF)-induced expression of cyclins A and E, but not cyclin D1. Tacrolimus 122-128 cyclin A2 Homo sapiens 238-253 9654363-9 1998 The possibility that the difference in the control of GFAP phosphorylation in the two structures is related to differences in the control of GFAP dephosphorylation was investigated by incubating spinal cord slices with the calcineurin inhibitor FK506 in the presence of Ca2+. Tacrolimus 245-250 glial fibrillary acidic protein Rattus norvegicus 54-58 9654363-9 1998 The possibility that the difference in the control of GFAP phosphorylation in the two structures is related to differences in the control of GFAP dephosphorylation was investigated by incubating spinal cord slices with the calcineurin inhibitor FK506 in the presence of Ca2+. Tacrolimus 245-250 glial fibrillary acidic protein Rattus norvegicus 141-145 9606972-2 1998 Here we report that both cobalt and the calcium chelator EGTA, inhibitors of calcium uptake, as well as cyclosporin A and FK-506, specific inhibitors of calcineurin function, abolished fibroblast growth factor (FGF)-induced expression of cyclins A and E, but not cyclin D1. Tacrolimus 122-128 cyclin D1 Homo sapiens 263-272 9565564-5 1998 The transcriptional response provoked by mT was blocked by the immunosuppressive drug FK506, a potent inhibitor of TCR-mediated IL-2 gene expression. Tacrolimus 86-91 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 115-118 9565564-5 1998 The transcriptional response provoked by mT was blocked by the immunosuppressive drug FK506, a potent inhibitor of TCR-mediated IL-2 gene expression. Tacrolimus 86-91 interleukin 2 Homo sapiens 128-132 9681677-1 1998 FcepsilonRI-mediated exocytosis of preformed mediators from mast cells and basophils (e.g. histamine, serotonin, beta-hexosaminidase) is sensitive to the immunosuppressants cyclosporin A and FK506 (IC50 200 and 4 nM, respectively) but not rapamycin. Tacrolimus 191-196 O-GlcNAcase Rattus norvegicus 113-132 9565100-8 1998 Low pharmacologic concentrations of tacrolimus/cyclosporine (CsA) and mycophenolic acid (MPA) singly or in combination had no effect on the spontaneous proliferation of DBMC and had significantly less inhibitory activity on MLC responses of DBMC and its purified CD3+ or CD34+ subpopulations, compared with the responses of spleen cells. Tacrolimus 36-46 CD34 molecule Homo sapiens 271-275 9653668-1 1998 Since FK506 binding protein (FKBP12) inhibits dose-dependently the immunosuppressive activity of FK506 in vitro, plasma FKBP12 levels were measured after rat small bowel transplantation (SBTx). Tacrolimus 6-11 FKBP prolyl isomerase 1A Rattus norvegicus 29-35 9653668-5 1998 Therefore, the plasma FKBP12 level should be considered as one of the parameters related to the immunosuppressive activity of FK506 in SBTx. Tacrolimus 126-131 FKBP prolyl isomerase 1A Rattus norvegicus 22-28 9620363-3 1998 NFAT acts at the antigen receptor response element-2 (ARRE-2) sequence in the IL-2 enhancer and is the nuclear target of T cell stimulation signals and the immunosuppressant drugs cyclosporine (Sandimmune) and FK-506 (tacrolimus), which are potent inhibitors of IL-2 gene transcription. Tacrolimus 210-216 interleukin 2 Homo sapiens 78-82 9620363-3 1998 NFAT acts at the antigen receptor response element-2 (ARRE-2) sequence in the IL-2 enhancer and is the nuclear target of T cell stimulation signals and the immunosuppressant drugs cyclosporine (Sandimmune) and FK-506 (tacrolimus), which are potent inhibitors of IL-2 gene transcription. Tacrolimus 210-216 interleukin 2 Homo sapiens 262-266 9620363-3 1998 NFAT acts at the antigen receptor response element-2 (ARRE-2) sequence in the IL-2 enhancer and is the nuclear target of T cell stimulation signals and the immunosuppressant drugs cyclosporine (Sandimmune) and FK-506 (tacrolimus), which are potent inhibitors of IL-2 gene transcription. Tacrolimus 218-228 interleukin 2 Homo sapiens 78-82 9620363-3 1998 NFAT acts at the antigen receptor response element-2 (ARRE-2) sequence in the IL-2 enhancer and is the nuclear target of T cell stimulation signals and the immunosuppressant drugs cyclosporine (Sandimmune) and FK-506 (tacrolimus), which are potent inhibitors of IL-2 gene transcription. Tacrolimus 218-228 interleukin 2 Homo sapiens 262-266 9599809-0 1998 A maize FK506-sensitive immunophilin, mzFKBP-66, is a peptidylproline cis-trans-isomerase that interacts with calmodulin and a 36-kDa cytoplasmic protein. Tacrolimus 8-13 Peptidyl-prolyl cis-trans isomerase FKBP12 Zea mays 24-36 9599809-0 1998 A maize FK506-sensitive immunophilin, mzFKBP-66, is a peptidylproline cis-trans-isomerase that interacts with calmodulin and a 36-kDa cytoplasmic protein. Tacrolimus 8-13 calmodulin1 Zea mays 110-120 9545268-0 1998 Effects of FK506-binding protein 12 and FK506 on autophosphorylation of epidermal growth factor receptor. Tacrolimus 11-16 epidermal growth factor receptor Homo sapiens 72-104 9545268-6 1998 Finally, FK506 and rapamycin, which are known to block the PPIase activity of FKBP12, induced a significant stimulation of EGF receptor autophosphorylation in intact A431 cells suggesting suppression of EGF receptor autophosphorylation by intracellular FKBP12 in vivo. Tacrolimus 9-14 FKBP prolyl isomerase 7 Homo sapiens 59-65 9545268-6 1998 Finally, FK506 and rapamycin, which are known to block the PPIase activity of FKBP12, induced a significant stimulation of EGF receptor autophosphorylation in intact A431 cells suggesting suppression of EGF receptor autophosphorylation by intracellular FKBP12 in vivo. Tacrolimus 9-14 FKBP prolyl isomerase 1A Homo sapiens 78-84 9545268-6 1998 Finally, FK506 and rapamycin, which are known to block the PPIase activity of FKBP12, induced a significant stimulation of EGF receptor autophosphorylation in intact A431 cells suggesting suppression of EGF receptor autophosphorylation by intracellular FKBP12 in vivo. Tacrolimus 9-14 epidermal growth factor Homo sapiens 123-126 9545268-6 1998 Finally, FK506 and rapamycin, which are known to block the PPIase activity of FKBP12, induced a significant stimulation of EGF receptor autophosphorylation in intact A431 cells suggesting suppression of EGF receptor autophosphorylation by intracellular FKBP12 in vivo. Tacrolimus 9-14 epidermal growth factor Homo sapiens 203-206 9545268-6 1998 Finally, FK506 and rapamycin, which are known to block the PPIase activity of FKBP12, induced a significant stimulation of EGF receptor autophosphorylation in intact A431 cells suggesting suppression of EGF receptor autophosphorylation by intracellular FKBP12 in vivo. Tacrolimus 9-14 FKBP prolyl isomerase 1A Homo sapiens 253-259 9719949-1 1998 FK506 treatment markedly increased survival rates of [BALB/c-->C3H/He] bone marrow and spleen (BM/Spl) chimeras which had severe graft-versus-host disease (GVHD), marking 91% survival rates on day 60. Tacrolimus 0-5 sphingosine-1-phosphate lyase 1 Homo sapiens 101-104 9719949-5 1998 On week 12, alloreactive MLRs were still low in FK506-treated allogeneic BM/Spl and BM chimeras although those against third party alloantigen in the spleen cells from vehicle-treated allogeneic BM chimeras and syngeneic BM chimeras gradually recovered. Tacrolimus 48-53 sphingosine-1-phosphate lyase 1 Homo sapiens 76-79 9719949-8 1998 Reverse-transcription polymerase chain reaction (RT-PCR) method suggested that perforin and granzyme B gene expressions were basically unchanged or rather increased in the spleen cells from FK506-treated allogeneic BM/Spl and BM chimeras. Tacrolimus 190-195 granzyme B Homo sapiens 92-102 9719949-8 1998 Reverse-transcription polymerase chain reaction (RT-PCR) method suggested that perforin and granzyme B gene expressions were basically unchanged or rather increased in the spleen cells from FK506-treated allogeneic BM/Spl and BM chimeras. Tacrolimus 190-195 sphingosine-1-phosphate lyase 1 Homo sapiens 218-221 9563957-12 1998 The mean baseline maintenance dose of prednisone was greater by 2 mg daily in patients who received IFN-alpha with tacrolimus compared with control patients who did not receive IFN-alpha with tacrolimus (IFN-alpha 5. Tacrolimus 115-125 interferon alpha 1 Homo sapiens 100-109 9558067-9 1998 By Western blot it was determined that the levels of Fas ligand protein were increased in the CD8-depleted group compared with those in control and FK506-treated allograft recipients. Tacrolimus 148-153 Fas ligand Rattus norvegicus 53-63 9566798-4 1998 In fact, rapamycin and FK-506 block both alphabeta+, CD4+ and gammadelta+ T lymphocytes, while CsA inhibits only the alphabeta+, CD4+ T lymphocyte. Tacrolimus 23-29 CD4 antigen Mus musculus 53-56 9630010-0 1998 FK506 (Tacrolimus) decreases the cytotoxicity of cyclosporin A in rat hepatocytes in primary culture: implication of CYP3A induction. Tacrolimus 0-5 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 117-122 9630010-0 1998 FK506 (Tacrolimus) decreases the cytotoxicity of cyclosporin A in rat hepatocytes in primary culture: implication of CYP3A induction. Tacrolimus 7-17 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 117-122 9630010-1 1998 Tacrolimus (FK506) and cyclosporin A (CsA) are two potent immunosuppressants mainly metabolized by hepatic cytochrome P-450 3A (CYP3A) monooxygenase. Tacrolimus 0-10 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 107-126 9630010-1 1998 Tacrolimus (FK506) and cyclosporin A (CsA) are two potent immunosuppressants mainly metabolized by hepatic cytochrome P-450 3A (CYP3A) monooxygenase. Tacrolimus 0-10 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 128-133 9630010-1 1998 Tacrolimus (FK506) and cyclosporin A (CsA) are two potent immunosuppressants mainly metabolized by hepatic cytochrome P-450 3A (CYP3A) monooxygenase. Tacrolimus 12-17 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 107-126 9630010-1 1998 Tacrolimus (FK506) and cyclosporin A (CsA) are two potent immunosuppressants mainly metabolized by hepatic cytochrome P-450 3A (CYP3A) monooxygenase. Tacrolimus 12-17 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 128-133 9630010-7 1998 It is hypothesized that the interaction between the two drugs relies on a mechanism involving both competition of FK506 and CsA for CYP3A and of their immunophilin complexes for a common site on the calcineurin-calmodulin complex. Tacrolimus 114-119 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 132-137 9566798-6 1998 Hapten-immune lymph node cells from mice treated in vivo with CsA or FK506 failed to proliferate and to produce IL-2 when re-exposed to the specific antigen in vitro. Tacrolimus 69-74 interleukin 2 Mus musculus 112-116 9531290-8 1998 Furthermore, B cell proliferation through CD72 was blocked by the immunosuppressive agents cyclosporin A and FK506, indicating the important role for Ca2+-regulated activation events similar to BCR-stimulated cells. Tacrolimus 109-114 CD72 antigen Mus musculus 42-46 9608563-10 1998 When the dogs were immunosuppressed with FK506 combined with CsA and RS-61443, muscle fibers expressing beta-Gal were present 4 weeks after the transplantation and no antibodies reacting with donor myoblasts were detected. Tacrolimus 41-46 galactosidase beta 1 Canis lupus familiaris 104-112 9566840-15 1998 However, constitutive CYP3A4, which represents 50% of total CYP and metabolises drugs like nifedipine, warfarin, acetaminophen, cyclosporin and FK-506, is reduced during liver regeneration. Tacrolimus 144-150 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 22-25 9685211-1 1998 Although the immunosuppressive drugs FK506, rapamycin and cyclosporin A have been reported to potentiate transcriptional activation mediated by a non-saturating concentration of the glucocorticoid receptor agonist dexamethasone, the precise mechanism(s) underlying these responses remains unclear. Tacrolimus 37-42 nuclear receptor subfamily 3 group C member 1 Homo sapiens 182-205 9678635-0 1998 A calcineurin inhibitor, FK506, blocks voltage-gated calcium channel-dependent LTP in the hippocampus. Tacrolimus 25-30 calcineurin binding protein 1 Mus musculus 2-23 9510155-2 1998 Here, we utilized two pharmacophores, PD98059 and FK506, that inhibit, respectively, mitogen-activated protein (MAP) kinase kinase 1 (MEK 1) and calcineurin, to determine the relative role of the signaling pathways controlled by these enzymes in T cell activation. Tacrolimus 50-55 mitogen-activated protein kinase kinase 1 Homo sapiens 134-139 9510155-7 1998 FK506 suppressed CD3/PMA-induced production of all cytokines examined here but to a lesser extent IL-13. Tacrolimus 0-5 interleukin 13 Homo sapiens 98-103 9510155-8 1998 FK506 also reduced CD3/CD28-induced production of IL-3, IL-4, IL-10, TNF-alpha, and IL-6 but augmented that of GM-CSF, IL-5, IFN-gamma, and IL-13. Tacrolimus 0-5 CD28 molecule Homo sapiens 23-27 9510155-8 1998 FK506 also reduced CD3/CD28-induced production of IL-3, IL-4, IL-10, TNF-alpha, and IL-6 but augmented that of GM-CSF, IL-5, IFN-gamma, and IL-13. Tacrolimus 0-5 interleukin 3 Homo sapiens 50-54 9510155-8 1998 FK506 also reduced CD3/CD28-induced production of IL-3, IL-4, IL-10, TNF-alpha, and IL-6 but augmented that of GM-CSF, IL-5, IFN-gamma, and IL-13. Tacrolimus 0-5 interleukin 4 Homo sapiens 56-60 9510155-8 1998 FK506 also reduced CD3/CD28-induced production of IL-3, IL-4, IL-10, TNF-alpha, and IL-6 but augmented that of GM-CSF, IL-5, IFN-gamma, and IL-13. Tacrolimus 0-5 interleukin 10 Homo sapiens 62-67 9510155-8 1998 FK506 also reduced CD3/CD28-induced production of IL-3, IL-4, IL-10, TNF-alpha, and IL-6 but augmented that of GM-CSF, IL-5, IFN-gamma, and IL-13. Tacrolimus 0-5 tumor necrosis factor Homo sapiens 69-78 9515963-5 1998 Treatment of wild-type embryos with FK506, a specific calcineurin inhibitor, prevents nuclear localization of NF-ATc. Tacrolimus 36-41 nuclear factor of activated T cells 1 Homo sapiens 110-116 9510155-8 1998 FK506 also reduced CD3/CD28-induced production of IL-3, IL-4, IL-10, TNF-alpha, and IL-6 but augmented that of GM-CSF, IL-5, IFN-gamma, and IL-13. Tacrolimus 0-5 interleukin 6 Homo sapiens 84-88 9510155-8 1998 FK506 also reduced CD3/CD28-induced production of IL-3, IL-4, IL-10, TNF-alpha, and IL-6 but augmented that of GM-CSF, IL-5, IFN-gamma, and IL-13. Tacrolimus 0-5 colony stimulating factor 2 Homo sapiens 111-117 9510155-8 1998 FK506 also reduced CD3/CD28-induced production of IL-3, IL-4, IL-10, TNF-alpha, and IL-6 but augmented that of GM-CSF, IL-5, IFN-gamma, and IL-13. Tacrolimus 0-5 interleukin 5 Homo sapiens 119-123 9510155-8 1998 FK506 also reduced CD3/CD28-induced production of IL-3, IL-4, IL-10, TNF-alpha, and IL-6 but augmented that of GM-CSF, IL-5, IFN-gamma, and IL-13. Tacrolimus 0-5 interferon gamma Homo sapiens 125-134 9510155-8 1998 FK506 also reduced CD3/CD28-induced production of IL-3, IL-4, IL-10, TNF-alpha, and IL-6 but augmented that of GM-CSF, IL-5, IFN-gamma, and IL-13. Tacrolimus 0-5 interleukin 13 Homo sapiens 140-145 9488678-4 1998 The EGF-induced stimulation of protein synthesis and activation of eIF2B was dependent upon FK506-binding protein-rapamycin-associated protein, as shown with the immunosuppressant rapamycin, whereas NGF induction was partially dependent upon FK506-binding protein-rapamycin-associated protein. Tacrolimus 92-97 epidermal growth factor like 1 Rattus norvegicus 4-7 9488678-4 1998 The EGF-induced stimulation of protein synthesis and activation of eIF2B was dependent upon FK506-binding protein-rapamycin-associated protein, as shown with the immunosuppressant rapamycin, whereas NGF induction was partially dependent upon FK506-binding protein-rapamycin-associated protein. Tacrolimus 92-97 eukaryotic translation initiation factor 2B subunit delta Rattus norvegicus 67-72 9555045-5 1998 These results suggest that modulation of the activity of the Ca2+-dependent K+ channel by FK506 and rapamycin is directly through association of immunosuppressants with FKBP12. Tacrolimus 90-95 FKBP prolyl isomerase 1A Rattus norvegicus 169-175 9567214-11 1998 This way, the detoxicant function of P-gp against products of the ras catabolism could mediate their accumulation when the "vacuum cleaner" function is blocked by CsA or tacrolimus, contributing to the initial development of fibroblastic activation that leads to interstitial fibrosis associated with nephrotoxicity by these immunosuppressor drugs. Tacrolimus 170-180 ATP binding cassette subfamily B member 1 Homo sapiens 37-41 9570636-9 1998 However, the stratum oriens of the hippocampal CA1 sector showed a significant reduction in [3H]FK506 binding 48 h and 7 days after ischemia. Tacrolimus 96-101 carbonic anhydrase 1 Rattus norvegicus 47-50 9667420-4 1998 FK506-induced apoptosis was efficiently attenuated by co-addition of ILs including IL-1beta (2 ng/ml), IL-2 (5 ng/ml) and IL-4 (40 ng/ml) into culture. Tacrolimus 0-5 interleukin 1 beta Homo sapiens 83-91 9667420-4 1998 FK506-induced apoptosis was efficiently attenuated by co-addition of ILs including IL-1beta (2 ng/ml), IL-2 (5 ng/ml) and IL-4 (40 ng/ml) into culture. Tacrolimus 0-5 interleukin 2 Homo sapiens 103-107 9667420-4 1998 FK506-induced apoptosis was efficiently attenuated by co-addition of ILs including IL-1beta (2 ng/ml), IL-2 (5 ng/ml) and IL-4 (40 ng/ml) into culture. Tacrolimus 0-5 interleukin 4 Homo sapiens 122-126 9667420-6 1998 The data also suggested that cytokine networks including those via IL-1beta and IL-4 in addition to IL-2 prevent FK506-induced apoptosis. Tacrolimus 113-118 interleukin 1 beta Homo sapiens 67-75 9667420-6 1998 The data also suggested that cytokine networks including those via IL-1beta and IL-4 in addition to IL-2 prevent FK506-induced apoptosis. Tacrolimus 113-118 interleukin 4 Homo sapiens 80-84 9667420-6 1998 The data also suggested that cytokine networks including those via IL-1beta and IL-4 in addition to IL-2 prevent FK506-induced apoptosis. Tacrolimus 113-118 interleukin 2 Homo sapiens 100-104 9516567-9 1998 One patient treated with FK506 developed an LPD localized to the lymph nodes 8 months after the occurrence of serum protein abnormalities. Tacrolimus 25-30 acyl-CoA synthetase bubblegum family member 1 Homo sapiens 44-47 9495869-4 1998 FK-506 is metabolized by the cytochrome P-450-dependent mixed-function oxygenase system in different animal species, and we are reporting the isolation from pig liver microsomes, and the identification by electrospray ms-ms, of the FK-506 C19-C20 epoxide metabolite. Tacrolimus 0-6 cytochrome P450 family 2 subfamily D member 6 Sus scrofa 29-45 9495869-4 1998 FK-506 is metabolized by the cytochrome P-450-dependent mixed-function oxygenase system in different animal species, and we are reporting the isolation from pig liver microsomes, and the identification by electrospray ms-ms, of the FK-506 C19-C20 epoxide metabolite. Tacrolimus 232-238 cytochrome P450 family 2 subfamily D member 6 Sus scrofa 29-45 9475389-1 1998 FKBP59 is an immunophilin that binds the immunosuppressant drugs FK506 and rapamycin. Tacrolimus 65-70 FKBP prolyl isomerase 4 Rattus norvegicus 0-6 9532223-0 1998 Tacrolimus-related microangiopathy in kidney and simultaneous pancreas-kidney recipients: evidence of endothelin and cytokine involvement. Tacrolimus 0-10 TNF receptor superfamily member 8 Homo sapiens 117-125 9469459-8 1998 The immunosuppressant, FK506, while strikingly inhibiting the accumulation of IL-4 mRNA and the secretion of protein in response to IL-3/anti-IgE, had no effect on the generation of IL-13 in these cultures; the resistance was attributed to the IL-3-dependent signaling. Tacrolimus 23-28 interleukin 4 Homo sapiens 78-82 9469459-8 1998 The immunosuppressant, FK506, while strikingly inhibiting the accumulation of IL-4 mRNA and the secretion of protein in response to IL-3/anti-IgE, had no effect on the generation of IL-13 in these cultures; the resistance was attributed to the IL-3-dependent signaling. Tacrolimus 23-28 interleukin 3 Homo sapiens 132-136 9446649-1 1998 Previous studies have shown that expression of a membrane targeted chimeric protein containing the erythropoietin receptor (EpoR) cytoplasmic domain fused to the FK506-binding peptide FKBP12 allowed Ba/F3 cells to be rescued from interleukin-3 (IL-3) deprivation using a dimeric form of FK506, called FK1012. Tacrolimus 162-167 FK506 binding protein 1a Mus musculus 184-190 9446649-1 1998 Previous studies have shown that expression of a membrane targeted chimeric protein containing the erythropoietin receptor (EpoR) cytoplasmic domain fused to the FK506-binding peptide FKBP12 allowed Ba/F3 cells to be rescued from interleukin-3 (IL-3) deprivation using a dimeric form of FK506, called FK1012. Tacrolimus 287-292 FK506 binding protein 1a Mus musculus 184-190 9462649-7 1998 Complete obliteration of hemostasis and hepatic injury was also accomplished by the pretreatment with FK506 which suppressed TNF-alpha and IFN-gamma production. Tacrolimus 102-107 tumor necrosis factor Mus musculus 125-134 9462649-7 1998 Complete obliteration of hemostasis and hepatic injury was also accomplished by the pretreatment with FK506 which suppressed TNF-alpha and IFN-gamma production. Tacrolimus 102-107 interferon gamma Mus musculus 139-148 9461216-1 1998 FKBP12, a cis-trans prolyl isomerase that binds the immunosuppressants FK506 and rapamycin, is ubiquitously expressed and interacts with proteins in several intracellular signal transduction systems. Tacrolimus 71-76 FK506 binding protein 1a Mus musculus 0-6 9450972-0 1998 Genetic separation of FK506 susceptibility and drug transport in the yeast Pdr5 ATP-binding cassette multidrug resistance transporter. Tacrolimus 22-27 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 75-79 9450972-4 1998 Selected mutant Pdr5 transporters were analyzed with respect to their expression levels, subcellular localization, drug resistance profiles to cycloheximide, rhodamines, antifungal azoles, steroids, and sensitivity to the inhibitor FK506. Tacrolimus 232-237 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 16-20 9450972-8 1998 Surprisingly, a S1360F exchange in transmembrane domain 10 not only caused limited substrate specificity, but also abolished Pdr5 susceptibility to inhibition by the immunosuppressant FK506. Tacrolimus 184-189 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 125-129 9448150-12 1998 In patients converted to tacrolimus treatment, there was a -55 mg/dl (-16%) (P=0.0031) change in cholesterol, a -48 mg/dl (-25%) (P=0.0014) change in LDL cholesterol, and a -36 mg/dl (-23%) (P=0.034) change in apolipoprotein B. Tacrolimus 25-35 apolipoprotein B Homo sapiens 210-226 9502207-0 1998 The immunosuppressant FK506 increases GAP-43 mRNA levels in axotomized sensory neurons. Tacrolimus 22-27 growth associated protein 43 Rattus norvegicus 38-44 9502207-3 1998 Using the reverse-transcriptase polymerase chain reaction (RT-PCR) technique and a digoxigenin-labeled probe, we show that subcutaneous injections of FK506 (10 mg/kg/day) markedly increases the level of axotomy-induced growth-associated protein (GAP-43) mRNA in dorsal root ganglion (DRG) neurons. Tacrolimus 150-155 growth associated protein 43 Rattus norvegicus 246-252 9502207-5 1998 Increased synthesis of GAP-43 may play a role in FK506"s ability to speed nerve regeneration. Tacrolimus 49-54 growth associated protein 43 Rattus norvegicus 23-29 9448137-7 1998 In contrast to tacrolimus, indolyl-ASC binds poorly to FK506 binding protein 12 (FKBP12), the major cytosolic receptor for tacrolimus and related compounds. Tacrolimus 123-133 PYD and CARD domain containing Homo sapiens 35-38 9448150-16 1998 Tacrolimus can lower cholesterol, LDL, and apolipoprotein B. Tacrolimus 0-10 apolipoprotein B Homo sapiens 43-59 9453019-5 1998 We also measured ADO plasma level in two KTR treated with FK506, a CsA analog. Tacrolimus 58-63 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 67-70 9448137-7 1998 In contrast to tacrolimus, indolyl-ASC binds poorly to FK506 binding protein 12 (FKBP12), the major cytosolic receptor for tacrolimus and related compounds. Tacrolimus 123-133 FKBP prolyl isomerase 1A Homo sapiens 55-79 9448137-7 1998 In contrast to tacrolimus, indolyl-ASC binds poorly to FK506 binding protein 12 (FKBP12), the major cytosolic receptor for tacrolimus and related compounds. Tacrolimus 123-133 FKBP prolyl isomerase 1A Homo sapiens 81-87 9448137-8 1998 However, the stability of the interaction between the FKBP12-indolyl-ASC complex and CaN is much greater than that of the FKBP12-tacrolimus complex. Tacrolimus 129-139 FKBP prolyl isomerase 1A Homo sapiens 122-128 9448138-1 1998 BACKGROUND: Tacrolimus (FK506) has potent immunosuppressive properties reflecting its ability to block the transcription of lymphokine genes in activated T cells through formation of a complex with FK506 binding protein-12, which inhibits the phosphatase activity of calcineurin. Tacrolimus 12-22 FK506 binding protein 1a Mus musculus 198-222 9448138-1 1998 BACKGROUND: Tacrolimus (FK506) has potent immunosuppressive properties reflecting its ability to block the transcription of lymphokine genes in activated T cells through formation of a complex with FK506 binding protein-12, which inhibits the phosphatase activity of calcineurin. Tacrolimus 24-29 FK506 binding protein 1a Mus musculus 198-222 9669111-5 1998 We also examined the effect of the immunosuppressant FK506 on the production of IL-13 by antigen-stimulated PBMC from AD patients. Tacrolimus 53-58 interleukin 13 Homo sapiens 80-85 9669111-8 1998 Moreover, the enhanced production of IL-13 in AD was suppressed by FK506. Tacrolimus 67-72 interleukin 13 Homo sapiens 37-42 9486505-3 1998 Close monitoring of patients receiving FK506 and high-dose corticosteroids, for the development of TTP is warranted. Tacrolimus 39-44 ZFP36 ring finger protein Homo sapiens 99-102 9517403-7 1998 Continuous administration of an ET(A)/ET(B) antagonist, TAK-044 (3 mg/day, s.c.), which effectively blocked systemic and renal vascular responses to exogenously administered ET-1, partially attenuated the FK506-induced renal vasoconstriction. Tacrolimus 205-210 endothelin receptor type B Rattus norvegicus 38-43 9665007-0 1998 Differential effects of cyclosporin A and tacrolimus on the production of TGF-beta: implications for the development of obliterative bronchiolitis after lung transplantation. Tacrolimus 42-52 transforming growth factor beta 1 Homo sapiens 74-82 9551469-1 1998 Treatment with FK506, an inhibitor of Ca2+/calmodulin dependent phosphatase (calcineurin, CaN), within 1 hr after transient ischemia afforded protection from apoptotic death in CA1 pyramidal neurons. Tacrolimus 15-20 carbonic anhydrase 1 Homo sapiens 177-180 9697196-1 1998 Ligand-protein docking simulations are employed to analyze the binding energy landscape of the pipecolinyl fragment that serves as a recognition core of the FK506 ligand in binding with the FKBP12 protein. Tacrolimus 157-162 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 190-196 9697196-2 1998 This fragment acts as a molecular anchor that specifically binds within the protein active site in a unique binding mode, in harmony with the structure of the FK506-FKBP12 complex. Tacrolimus 159-164 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 165-171 14518324-0 1998 [Selective immunosuppression with monoclonal antibodies against ICAM-1 and LFA-1 with FK 506 after experimental small intestine transplantation in the rat]. Tacrolimus 86-92 integrin subunit alpha L Rattus norvegicus 75-80 14518324-4 1998 The results of this study indicate that in contrast to the induction of allospecific tolerance after liver transplantation, the addition of anti ICAM-1 and/or of anti LFA-1 mabs after SBT reverses the FK 506 effect and results in early graft rejection. Tacrolimus 201-207 intercellular adhesion molecule 1 Rattus norvegicus 145-151 14518324-4 1998 The results of this study indicate that in contrast to the induction of allospecific tolerance after liver transplantation, the addition of anti ICAM-1 and/or of anti LFA-1 mabs after SBT reverses the FK 506 effect and results in early graft rejection. Tacrolimus 201-207 integrin subunit alpha L Rattus norvegicus 167-172 9665008-10 1998 Numerically, EPO was higher and iron lower in the TC group than in the CsA group. Tacrolimus 50-52 erythropoietin Homo sapiens 13-16 9665007-3 1998 We investigated the effect of tacrolimus and CsA in tissue culture and found that there was indeed a negative effect on human lung small airway epithelial cell proliferation by recombinant transforming growth factor-beta (TGF-beta), which was reversed by anti-TGF-beta. Tacrolimus 30-40 transforming growth factor beta 1 Homo sapiens 189-220 9665007-3 1998 We investigated the effect of tacrolimus and CsA in tissue culture and found that there was indeed a negative effect on human lung small airway epithelial cell proliferation by recombinant transforming growth factor-beta (TGF-beta), which was reversed by anti-TGF-beta. Tacrolimus 30-40 transforming growth factor beta 1 Homo sapiens 222-230 9405642-10 1997 FK506 binding to FKBP52 did not affect the phosphorylation by CK2 and, conversely, the FK506-binding activity of FKBP52 was not affected by the phosphorylation. Tacrolimus 0-5 peptidyl-prolyl cis-trans isomerase FKBP4 Oryctolagus cuniculus 17-23 9422408-2 1997 Recently, calcineurin phosphatase inhibition by cyclosporine or tacrolimus has been postulated to lead to diastolic hypertension through the induction of transforming growth factor-beta (TGF-beta) and resultant endothelin-mediated renal arteriolar vasospasm. Tacrolimus 64-74 transforming growth factor beta 1 Homo sapiens 154-185 9422408-2 1997 Recently, calcineurin phosphatase inhibition by cyclosporine or tacrolimus has been postulated to lead to diastolic hypertension through the induction of transforming growth factor-beta (TGF-beta) and resultant endothelin-mediated renal arteriolar vasospasm. Tacrolimus 64-74 transforming growth factor beta 1 Homo sapiens 187-195 9422427-6 1997 FK506-treated kidneys had a progressive increase in the expression of TGF-beta1 and matrix proteins (biglycan, tenascin, fibronectin, and type I collagen). Tacrolimus 0-5 transforming growth factor, beta 1 Rattus norvegicus 70-79 9422427-6 1997 FK506-treated kidneys had a progressive increase in the expression of TGF-beta1 and matrix proteins (biglycan, tenascin, fibronectin, and type I collagen). Tacrolimus 0-5 biglycan Rattus norvegicus 101-109 9422427-6 1997 FK506-treated kidneys had a progressive increase in the expression of TGF-beta1 and matrix proteins (biglycan, tenascin, fibronectin, and type I collagen). Tacrolimus 0-5 tenascin C Rattus norvegicus 111-119 9422427-6 1997 FK506-treated kidneys had a progressive increase in the expression of TGF-beta1 and matrix proteins (biglycan, tenascin, fibronectin, and type I collagen). Tacrolimus 0-5 fibronectin 1 Rattus norvegicus 121-132 9422427-9 1997 The expression of plasminogen activator inhibitor-1, a protease inhibitor influenced by TGF-beta, followed TGF-beta1 and matrix proteins, suggesting that the fibrosis of chronic FK506 nephropathy likely involves the dual action of TGF-beta1 on matrix deposition and degradation. Tacrolimus 178-183 serpin family E member 1 Rattus norvegicus 18-51 9422427-10 1997 Since both peripheral and tissue renin expression were elevated with FK506, the renin-angiotensin system may play a role in the pathogenesis of this condition. Tacrolimus 69-74 renin Rattus norvegicus 33-38 9422427-10 1997 Since both peripheral and tissue renin expression were elevated with FK506, the renin-angiotensin system may play a role in the pathogenesis of this condition. Tacrolimus 69-74 renin Rattus norvegicus 80-85 9405642-10 1997 FK506 binding to FKBP52 did not affect the phosphorylation by CK2 and, conversely, the FK506-binding activity of FKBP52 was not affected by the phosphorylation. Tacrolimus 87-92 peptidyl-prolyl cis-trans isomerase FKBP4 Oryctolagus cuniculus 113-119 9440622-5 1997 When FK 506 or CsA was added, Fas-Ag expression with FK 506 at a concentration of 0.01-0.1 microg/ml was significantly lower than that with CsA (P < 0.05). Tacrolimus 53-59 Fas (TNF receptor superfamily member 6) Mus musculus 30-36 9432041-0 1997 FK506 inhibits anti-IgM antibody-induced apoptosis and 17 kD endonuclease activity in the human B-cell line, MBC-1, established from Burkitt"s lymphoma. Tacrolimus 0-5 coiled-coil domain containing 112 Homo sapiens 109-114 9432041-3 1997 FK506, an immunosuppressive agent and calcineurin inhibitor, partially rescued the anti-IgM antibody-induced cell death in these MBC-1 cells. Tacrolimus 0-5 coiled-coil domain containing 112 Homo sapiens 129-134 9466703-6 1997 The direct or indirect association of calcineurin with GFAP was suggested by observations showing that FK506, a specific inhibitor of calcineurin, increased the phosphorylation state of GFAP in immature slices and of GFAP and vimentin in astrocyte cultures. Tacrolimus 103-108 glial fibrillary acidic protein Rattus norvegicus 55-59 9466703-6 1997 The direct or indirect association of calcineurin with GFAP was suggested by observations showing that FK506, a specific inhibitor of calcineurin, increased the phosphorylation state of GFAP in immature slices and of GFAP and vimentin in astrocyte cultures. Tacrolimus 103-108 glial fibrillary acidic protein Rattus norvegicus 186-190 9466703-6 1997 The direct or indirect association of calcineurin with GFAP was suggested by observations showing that FK506, a specific inhibitor of calcineurin, increased the phosphorylation state of GFAP in immature slices and of GFAP and vimentin in astrocyte cultures. Tacrolimus 103-108 glial fibrillary acidic protein Rattus norvegicus 186-190 9466703-6 1997 The direct or indirect association of calcineurin with GFAP was suggested by observations showing that FK506, a specific inhibitor of calcineurin, increased the phosphorylation state of GFAP in immature slices and of GFAP and vimentin in astrocyte cultures. Tacrolimus 103-108 vimentin Rattus norvegicus 226-234 9403721-1 1997 P-glycoprotein (P-gp) expels hydrophobic substances from the cell, including chemotherapeutic agents and immunosuppressants such as cyclosporin A (CsA) and FK506. Tacrolimus 156-161 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 0-14 9403721-1 1997 P-glycoprotein (P-gp) expels hydrophobic substances from the cell, including chemotherapeutic agents and immunosuppressants such as cyclosporin A (CsA) and FK506. Tacrolimus 156-161 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 16-20 9440622-2 1997 Thus, in liver transplantation, Fas-Ag expression of hepatocytes and its modification by immunosuppressive agents such as FK 506 or CsA can theoretically influence allograft survival. Tacrolimus 122-128 Fas (TNF receptor superfamily member 6) Mus musculus 32-38 9440622-5 1997 When FK 506 or CsA was added, Fas-Ag expression with FK 506 at a concentration of 0.01-0.1 microg/ml was significantly lower than that with CsA (P < 0.05). Tacrolimus 5-11 Fas (TNF receptor superfamily member 6) Mus musculus 30-36 9457703-2 1997 FK506 is inactive by itself and requires binding to an FK506 binding protein-12 (FKBP-12), or immunophilin, for activation. Tacrolimus 0-5 FKBP prolyl isomerase 1A Rattus norvegicus 55-79 9407429-3 1997 Both tacrolimus and CsA interfere with the early stage of lymphocyte proliferation by blocking interleukin-2 synthesis. Tacrolimus 5-15 interleukin 2 Homo sapiens 95-108 9457703-2 1997 FK506 is inactive by itself and requires binding to an FK506 binding protein-12 (FKBP-12), or immunophilin, for activation. Tacrolimus 0-5 FKBP prolyl isomerase 1A Rattus norvegicus 81-88 9457703-3 1997 In this regard, FK506 is analogous to cyclosporin A, which must bind to its immunophilin (cyclophilin A) to display activity. Tacrolimus 16-21 peptidylprolyl isomerase A Rattus norvegicus 90-103 9457703-9 1997 The nerve regenerative property of this class of agents is separate from their immunosuppressant action because FK506-related compounds that bind to FKBP-12 but do not inhibit calcineurin are also able to increase nerve regeneration. Tacrolimus 112-117 FKBP prolyl isomerase 1A Rattus norvegicus 149-156 9371677-3 1997 METHODS: We investigated the relationship between the presence of anti-FKBP12 autoantibodies and rejection episodes in 47 patients treated with FK506 after living-related partial liver transplantation (LRLT). Tacrolimus 144-149 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 71-77 9371682-5 1997 Increased thymic apoptosis resulted in the disappearance of CD4+CD8+ thymocytes after FK506/dexamethasone injection. Tacrolimus 86-91 CD4 molecule Homo sapiens 60-63 9371682-5 1997 Increased thymic apoptosis resulted in the disappearance of CD4+CD8+ thymocytes after FK506/dexamethasone injection. Tacrolimus 86-91 CD8a molecule Homo sapiens 64-67 9353327-4 1997 p27 exhibits isomerase activity with a kcat/Km of 0.18 microM-1 s-1 for a peptide substrate; this activity is inhibited by cyclosporin A but is not affected by FK506. Tacrolimus 160-165 interferon alpha inducible protein 27 Homo sapiens 0-3 9402514-8 1997 RESULTS: Allografts from mice that received FK506 treatment daily showed significant neointimal thickening with increased expression of ICAM-1, VCAM-1, and PDGF-B mRNA, whereas there was almost no intimal thickening and ICAM-1, VCAM-1, and PDGF-B mRNA expression in the mice that received anti-ICAM-1 plus anti-LFA-1 mAbs. Tacrolimus 44-49 intercellular adhesion molecule 1 Mus musculus 136-142 9374148-7 1997 The increase in accumulation of neutrophils, and MDA and MPO levels (which were induced by ischemia) observed 1 and 24 hours after flap elevation was diminished by FK506 pretreatment. Tacrolimus 164-169 myeloperoxidase Rattus norvegicus 57-60 9394799-6 1997 Such up-regulation could be suppressed by RAPA, FK506, and cyclosporin A. RAPA and FK506 also repressed the up-regulated PA28 alpha messages in phytohemagglutinin (PHA)-stimulated T cells. Tacrolimus 48-53 proteasome activator subunit 1 Homo sapiens 121-131 9373220-6 1997 Excess levels of the FK-506 analogue ascomycin reversed the antagonistic effect of rapamycin on IL-6 mediated growth suppression, suggesting that this biological action of rapamycin is mediated by a rapamycin/immunophilin complex. Tacrolimus 21-27 interleukin 6 Homo sapiens 96-100 9363914-0 1997 FK506 enhances IL-13 production by T cells activated through CD3/CD28. Tacrolimus 0-5 interleukin 13 Homo sapiens 15-20 9363914-0 1997 FK506 enhances IL-13 production by T cells activated through CD3/CD28. Tacrolimus 0-5 CD28 molecule Homo sapiens 65-69 9402514-8 1997 RESULTS: Allografts from mice that received FK506 treatment daily showed significant neointimal thickening with increased expression of ICAM-1, VCAM-1, and PDGF-B mRNA, whereas there was almost no intimal thickening and ICAM-1, VCAM-1, and PDGF-B mRNA expression in the mice that received anti-ICAM-1 plus anti-LFA-1 mAbs. Tacrolimus 44-49 vascular cell adhesion molecule 1 Mus musculus 144-150 9402514-8 1997 RESULTS: Allografts from mice that received FK506 treatment daily showed significant neointimal thickening with increased expression of ICAM-1, VCAM-1, and PDGF-B mRNA, whereas there was almost no intimal thickening and ICAM-1, VCAM-1, and PDGF-B mRNA expression in the mice that received anti-ICAM-1 plus anti-LFA-1 mAbs. Tacrolimus 44-49 platelet derived growth factor, B polypeptide Mus musculus 156-162 9402514-8 1997 RESULTS: Allografts from mice that received FK506 treatment daily showed significant neointimal thickening with increased expression of ICAM-1, VCAM-1, and PDGF-B mRNA, whereas there was almost no intimal thickening and ICAM-1, VCAM-1, and PDGF-B mRNA expression in the mice that received anti-ICAM-1 plus anti-LFA-1 mAbs. Tacrolimus 44-49 intercellular adhesion molecule 1 Mus musculus 220-226 9402514-8 1997 RESULTS: Allografts from mice that received FK506 treatment daily showed significant neointimal thickening with increased expression of ICAM-1, VCAM-1, and PDGF-B mRNA, whereas there was almost no intimal thickening and ICAM-1, VCAM-1, and PDGF-B mRNA expression in the mice that received anti-ICAM-1 plus anti-LFA-1 mAbs. Tacrolimus 44-49 vascular cell adhesion molecule 1 Mus musculus 228-234 9402514-8 1997 RESULTS: Allografts from mice that received FK506 treatment daily showed significant neointimal thickening with increased expression of ICAM-1, VCAM-1, and PDGF-B mRNA, whereas there was almost no intimal thickening and ICAM-1, VCAM-1, and PDGF-B mRNA expression in the mice that received anti-ICAM-1 plus anti-LFA-1 mAbs. Tacrolimus 44-49 platelet derived growth factor, B polypeptide Mus musculus 240-246 9402514-8 1997 RESULTS: Allografts from mice that received FK506 treatment daily showed significant neointimal thickening with increased expression of ICAM-1, VCAM-1, and PDGF-B mRNA, whereas there was almost no intimal thickening and ICAM-1, VCAM-1, and PDGF-B mRNA expression in the mice that received anti-ICAM-1 plus anti-LFA-1 mAbs. Tacrolimus 44-49 intercellular adhesion molecule 1 Mus musculus 220-226 9402514-8 1997 RESULTS: Allografts from mice that received FK506 treatment daily showed significant neointimal thickening with increased expression of ICAM-1, VCAM-1, and PDGF-B mRNA, whereas there was almost no intimal thickening and ICAM-1, VCAM-1, and PDGF-B mRNA expression in the mice that received anti-ICAM-1 plus anti-LFA-1 mAbs. Tacrolimus 44-49 integrin beta 2 Mus musculus 311-316 9509307-8 1997 The serum antibody levels to S-Ag and proliferative response of lymphocytes to S-Ag were also significantly suppressed by the combination therapy with low doses of tacrolimus and rapamycin. Tacrolimus 164-174 S-antigen visual arrestin Rattus norvegicus 29-33 9509307-8 1997 The serum antibody levels to S-Ag and proliferative response of lymphocytes to S-Ag were also significantly suppressed by the combination therapy with low doses of tacrolimus and rapamycin. Tacrolimus 164-174 S-antigen visual arrestin Rattus norvegicus 79-83 9355847-8 1997 The associated changes in the expression of CD11c, CD29, and CD31 were also significantly altered by FK506. Tacrolimus 101-106 integrin subunit alpha X Homo sapiens 44-49 9346894-0 1997 FKBP12 binds the inositol 1,4,5-trisphosphate receptor at leucine-proline (1400-1401) and anchors calcineurin to this FK506-like domain. Tacrolimus 118-123 FKBP prolyl isomerase 1A Homo sapiens 0-6 9346894-2 1997 It is the primary receptor for the immunosuppressant actions of the drug FK506 in whose presence FKBP12 binds to and inhibits calcineurin, disrupting interleukin formation in lymphocytes. Tacrolimus 73-78 FKBP prolyl isomerase 1A Homo sapiens 97-103 9346894-4 1997 We now report that FKBP12 binds the IP3R at residues 1400-1401, a leucyl-prolyl dipeptide epitope that structurally resembles FK506. Tacrolimus 126-131 FKBP prolyl isomerase 1A Homo sapiens 19-25 9346894-4 1997 We now report that FKBP12 binds the IP3R at residues 1400-1401, a leucyl-prolyl dipeptide epitope that structurally resembles FK506. Tacrolimus 126-131 inositol 1,4,5-trisphosphate receptor type 3 Homo sapiens 36-40 9346894-6 1997 We propose that FK506 promotes an FKBP12-calcineurin interaction by mimicking structurally similar dipeptide epitopes present within proteins that use FKBP12 to anchor calcineurin to the appropriate physiologic substrates. Tacrolimus 16-21 FKBP prolyl isomerase 1A Homo sapiens 34-40 9346894-6 1997 We propose that FK506 promotes an FKBP12-calcineurin interaction by mimicking structurally similar dipeptide epitopes present within proteins that use FKBP12 to anchor calcineurin to the appropriate physiologic substrates. Tacrolimus 16-21 FKBP prolyl isomerase 1A Homo sapiens 151-157 9355847-8 1997 The associated changes in the expression of CD11c, CD29, and CD31 were also significantly altered by FK506. Tacrolimus 101-106 integrin subunit beta 1 Homo sapiens 51-55 9355847-8 1997 The associated changes in the expression of CD11c, CD29, and CD31 were also significantly altered by FK506. Tacrolimus 101-106 platelet and endothelial cell adhesion molecule 1 Homo sapiens 61-65 9344552-1 1997 The immunosuppressant drugs FK506 and cyclosporin A inhibit T-cell proliferation via a common mechanism: calcineurin inhibition following binding to their respective binding proteins, the peptidyl prolyl isomerases FKBP-12 and cyclophilin A. Tacrolimus 28-33 FKBP prolyl isomerase 1A Rattus norvegicus 215-222 9344552-1 1997 The immunosuppressant drugs FK506 and cyclosporin A inhibit T-cell proliferation via a common mechanism: calcineurin inhibition following binding to their respective binding proteins, the peptidyl prolyl isomerases FKBP-12 and cyclophilin A. Tacrolimus 28-33 peptidylprolyl isomerase A Rattus norvegicus 227-240 9336339-4 1997 The expression of interferon-gamma mRNA in reverse transcriptase-polymerase chain reaction in the ear was inhibited by FK-506 and cyclosporin A. Tacrolimus 119-125 interferon gamma Mus musculus 18-34 9349985-3 1997 The effects of cyclosporine A (CsA), tacrolimus (FK506), and dexamethasone (DEX) on cytokine-induced production of interleukin (IL)-8 in a human colonic cancer cell line (HT-29) were examined. Tacrolimus 37-47 C-X-C motif chemokine ligand 8 Homo sapiens 115-133 9349985-3 1997 The effects of cyclosporine A (CsA), tacrolimus (FK506), and dexamethasone (DEX) on cytokine-induced production of interleukin (IL)-8 in a human colonic cancer cell line (HT-29) were examined. Tacrolimus 49-54 C-X-C motif chemokine ligand 8 Homo sapiens 115-133 9336339-6 1997 Contrary to the above in vivo findings, the immunosuppressors, FK-506 and cyclosporin A, inhibited the production of interferon-gamma and interleukin-2 by cultured Th1 cells (1E10.H2 cells) and of interleukin-4 and -5 by Th2 cells (D10.G4.1 cells) in vitro. Tacrolimus 63-69 interferon gamma Mus musculus 117-133 9336339-6 1997 Contrary to the above in vivo findings, the immunosuppressors, FK-506 and cyclosporin A, inhibited the production of interferon-gamma and interleukin-2 by cultured Th1 cells (1E10.H2 cells) and of interleukin-4 and -5 by Th2 cells (D10.G4.1 cells) in vitro. Tacrolimus 63-69 interleukin 2 Mus musculus 138-151 9336339-6 1997 Contrary to the above in vivo findings, the immunosuppressors, FK-506 and cyclosporin A, inhibited the production of interferon-gamma and interleukin-2 by cultured Th1 cells (1E10.H2 cells) and of interleukin-4 and -5 by Th2 cells (D10.G4.1 cells) in vitro. Tacrolimus 63-69 negative elongation factor complex member C/D, Th1l Mus musculus 164-167 9336339-6 1997 Contrary to the above in vivo findings, the immunosuppressors, FK-506 and cyclosporin A, inhibited the production of interferon-gamma and interleukin-2 by cultured Th1 cells (1E10.H2 cells) and of interleukin-4 and -5 by Th2 cells (D10.G4.1 cells) in vitro. Tacrolimus 63-69 interleukin 4 Mus musculus 197-217 9336339-6 1997 Contrary to the above in vivo findings, the immunosuppressors, FK-506 and cyclosporin A, inhibited the production of interferon-gamma and interleukin-2 by cultured Th1 cells (1E10.H2 cells) and of interleukin-4 and -5 by Th2 cells (D10.G4.1 cells) in vitro. Tacrolimus 63-69 heart and neural crest derivatives expressed 2 Mus musculus 221-224 9336339-7 1997 These results indicated that FK-506 and cyclosporin A selectively inhibited the Th1 cell-mediated contact dermatitis and potentiated the Th2 cell-mediated IgE antibody production in vivo. Tacrolimus 29-35 negative elongation factor complex member C/D, Th1l Mus musculus 80-83 9336339-7 1997 These results indicated that FK-506 and cyclosporin A selectively inhibited the Th1 cell-mediated contact dermatitis and potentiated the Th2 cell-mediated IgE antibody production in vivo. Tacrolimus 29-35 heart and neural crest derivatives expressed 2 Mus musculus 137-140 9336339-9 1997 However, because FK-506 and cyclosporin A inhibited the production of cytokines by both Th1 and Th2 cells in vitro and these two immunosuppressors showed higher selectivity toward inhibiting Th1 cell-mediated reactions by limitations in vivo experiments. Tacrolimus 17-23 negative elongation factor complex member C/D, Th1l Mus musculus 88-91 9336339-9 1997 However, because FK-506 and cyclosporin A inhibited the production of cytokines by both Th1 and Th2 cells in vitro and these two immunosuppressors showed higher selectivity toward inhibiting Th1 cell-mediated reactions by limitations in vivo experiments. Tacrolimus 17-23 heart and neural crest derivatives expressed 2 Mus musculus 96-99 9336339-9 1997 However, because FK-506 and cyclosporin A inhibited the production of cytokines by both Th1 and Th2 cells in vitro and these two immunosuppressors showed higher selectivity toward inhibiting Th1 cell-mediated reactions by limitations in vivo experiments. Tacrolimus 17-23 negative elongation factor complex member C/D, Th1l Mus musculus 191-194 9349628-3 1997 Previous studies in pancreatic islet cell lines have shown that cyclosporin A and FK506 through inhibition of calcineurin interfere also with the function of the transcription factor cAMP response element binding protein (CREB) that is activated by cAMP and calcium signals and binds to cAMP/calcium response elements (CRE). Tacrolimus 82-87 cAMP responsive element binding protein 1 Homo sapiens 183-220 10837558-1 1997 The objective of this section is to evaluate the contributions of hepatic metabolism, intestinal metabolism and intestinal p-glycoprotein to the pharmacokinetics of orally administered cyclosporine and tacrolimus. Tacrolimus 202-212 ATP binding cassette subfamily B member 1 Homo sapiens 123-137 9349628-3 1997 Previous studies in pancreatic islet cell lines have shown that cyclosporin A and FK506 through inhibition of calcineurin interfere also with the function of the transcription factor cAMP response element binding protein (CREB) that is activated by cAMP and calcium signals and binds to cAMP/calcium response elements (CRE). Tacrolimus 82-87 cAMP responsive element binding protein 1 Homo sapiens 222-226 9349628-4 1997 By transient expression of CRE-reporter genes or GAL4-CREB fusion proteins, the present study shows that inhibition of CREB/CRE-directed transcription by cyclosporin A and FK506 occurs in a great variety of cell types including in cell lines derived from tissues in which adverse effects of the immunosuppressants develop. Tacrolimus 172-177 galectin 4 Homo sapiens 49-53 9349628-4 1997 By transient expression of CRE-reporter genes or GAL4-CREB fusion proteins, the present study shows that inhibition of CREB/CRE-directed transcription by cyclosporin A and FK506 occurs in a great variety of cell types including in cell lines derived from tissues in which adverse effects of the immunosuppressants develop. Tacrolimus 172-177 cAMP responsive element binding protein 1 Homo sapiens 54-58 9349628-4 1997 By transient expression of CRE-reporter genes or GAL4-CREB fusion proteins, the present study shows that inhibition of CREB/CRE-directed transcription by cyclosporin A and FK506 occurs in a great variety of cell types including in cell lines derived from tissues in which adverse effects of the immunosuppressants develop. Tacrolimus 172-177 cAMP responsive element binding protein 1 Homo sapiens 119-123 9349628-6 1997 When taken together with recent evidence for an essential role of CREB in T-cell activation and proliferation, the present results suggest that inhibition of CREB/CRE-directed transcription may be a molecular mechanism of the immunosuppressive effect of cyclosporin A and FK506. Tacrolimus 272-277 cAMP responsive element binding protein 1 Homo sapiens 158-162 9295299-10 1997 The immunosuppressants cyclosporin A and FK506 abolish calcineurin-mediated induction of CTF-1 activity. Tacrolimus 41-46 cardiotrophin 1 Mus musculus 89-94 10837558-4 1997 Cyclosporine and tacrolimus are also substrates for p-glycoprotein, which acts as a counter-transport pump, actively transporting cyclosporine and tacrolimus back into the intestinal lumen. Tacrolimus 17-27 ATP binding cassette subfamily B member 1 Homo sapiens 52-66 10837558-4 1997 Cyclosporine and tacrolimus are also substrates for p-glycoprotein, which acts as a counter-transport pump, actively transporting cyclosporine and tacrolimus back into the intestinal lumen. Tacrolimus 147-157 ATP binding cassette subfamily B member 1 Homo sapiens 52-66 9349628-0 1997 Inhibition of CREB- and cAMP response element-mediated gene transcription by the immunosuppressive drugs cyclosporin A and FK506 in T cells. Tacrolimus 123-128 cAMP responsive element binding protein 1 Homo sapiens 14-18 9349628-1 1997 The clinically important immunosuppressant drugs cyclosporin A and FK506 (tacrolimus) inhibit in T-cells calcineurin phosphatase activity and nuclear translocation of the cytosolic component of the transcription factor nuclear factor of activated T-cells (NF-ATc) that is involved in the induction of early genes during T-cell activation. Tacrolimus 67-72 nuclear factor of activated T cells 1 Homo sapiens 256-262 9349628-1 1997 The clinically important immunosuppressant drugs cyclosporin A and FK506 (tacrolimus) inhibit in T-cells calcineurin phosphatase activity and nuclear translocation of the cytosolic component of the transcription factor nuclear factor of activated T-cells (NF-ATc) that is involved in the induction of early genes during T-cell activation. Tacrolimus 74-84 nuclear factor of activated T cells 1 Homo sapiens 256-262 9396011-9 1997 By influencing phosphorylation of neuronal nitric oxide synthase, FKBP12 regulates nitric oxide formation, which is reduced by FK506. Tacrolimus 127-132 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 66-72 10837558-14 1997 It seems that compounds that alter (either induce or inhibit) CYP3A4 and/or p-glycoprotein will alter the oral pharmacokinetics of cyclosporine and tacrolimus. Tacrolimus 148-158 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 10837558-14 1997 It seems that compounds that alter (either induce or inhibit) CYP3A4 and/or p-glycoprotein will alter the oral pharmacokinetics of cyclosporine and tacrolimus. Tacrolimus 148-158 ATP binding cassette subfamily B member 1 Homo sapiens 76-90 10837558-2 1997 Cyclosporine and tacrolimus are metabolized primarily by cytochrome P450 3A4 (CYP3A4) in the liver and small intestine. Tacrolimus 17-27 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 57-76 10837558-2 1997 Cyclosporine and tacrolimus are metabolized primarily by cytochrome P450 3A4 (CYP3A4) in the liver and small intestine. Tacrolimus 17-27 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 78-84 9252447-6 1997 Rapamycin potently blocked both the basal and the CCK-stimulated p70s6k activity, and this inhibition was reversed by an excess of FK-506. Tacrolimus 131-137 cholecystokinin Rattus norvegicus 50-53 9268302-5 1997 In Jurkat cells, TLiSA1/PTA1 mRNA and surface protein expression is greatly stimulated by treatment of the cells with phorbol ester, but the T cell proliferative signal of phorbol ester and ionophore combined greatly reduces or abrogates this response, and this suppressive effect of the ionophore is not reversed by incorporating FK506 to inhibit calcineurin. Tacrolimus 331-336 CD226 molecule Homo sapiens 17-23 9268302-5 1997 In Jurkat cells, TLiSA1/PTA1 mRNA and surface protein expression is greatly stimulated by treatment of the cells with phorbol ester, but the T cell proliferative signal of phorbol ester and ionophore combined greatly reduces or abrogates this response, and this suppressive effect of the ionophore is not reversed by incorporating FK506 to inhibit calcineurin. Tacrolimus 331-336 CD226 molecule Homo sapiens 24-28 9271313-0 1997 Cyclosporin A and FK506 reduce interleukin-5 mRNA abundance by inhibiting gene transcription. Tacrolimus 18-23 interleukin 5 Homo sapiens 31-44 9292524-6 1997 The results showed that IL-3 was induced by calcium ionophore and that the IL-3 induced by Fc gammaRIII stimulation was blocked by EGTA or FK506, but not by staurosporine (protein kinase C [PKC] inhibitor), indicating the important role of calcium-calcineurin in this system. Tacrolimus 139-144 interleukin 3 Mus musculus 75-79 9292524-6 1997 The results showed that IL-3 was induced by calcium ionophore and that the IL-3 induced by Fc gammaRIII stimulation was blocked by EGTA or FK506, but not by staurosporine (protein kinase C [PKC] inhibitor), indicating the important role of calcium-calcineurin in this system. Tacrolimus 139-144 Fc receptor, IgG, low affinity III Mus musculus 91-103 9325011-4 1997 The dependence on Ca2+ in the induction of GM-CSF, but not of apoptosis, was further confirmed by the inhibition of TNF- or IL-1-induced cytokine production by cyclosporin A or FK506, drugs that block the Ca2+/calmodulin-dependent protein Ser/Thr phosphatase calcineurin. Tacrolimus 177-182 colony stimulating factor 2 Rattus norvegicus 43-49 9325011-4 1997 The dependence on Ca2+ in the induction of GM-CSF, but not of apoptosis, was further confirmed by the inhibition of TNF- or IL-1-induced cytokine production by cyclosporin A or FK506, drugs that block the Ca2+/calmodulin-dependent protein Ser/Thr phosphatase calcineurin. Tacrolimus 177-182 tumor necrosis factor Rattus norvegicus 116-119 9271313-3 1997 We investigated the mechanisms controlling IL-5 messenger RNA (mRNA) expression in human T-lymphocytes in the presence of CsA or FK506. Tacrolimus 129-134 interleukin 5 Homo sapiens 43-47 9271313-7 1997 CsA and FK506 strongly inhibited cellular IL-5 mRNA expression in response to phytohemagglutinin (PHA), or to phorbol myristate acetate (PMA), and/or calcium ionophore. Tacrolimus 8-13 interleukin 5 Homo sapiens 42-46 9271313-9 1997 Nuclear run-on assays done with either 7-day cultured PBMC or HSB-2 cells demonstrated striking inhibition of IL-5 gene transcription by both CsA and FK506 at levels reflecting the degree of reduction of total cellular IL-5 mRNA abundance. Tacrolimus 150-155 interleukin 5 Homo sapiens 110-114 9271313-9 1997 Nuclear run-on assays done with either 7-day cultured PBMC or HSB-2 cells demonstrated striking inhibition of IL-5 gene transcription by both CsA and FK506 at levels reflecting the degree of reduction of total cellular IL-5 mRNA abundance. Tacrolimus 150-155 interleukin 5 Homo sapiens 219-223 9271313-11 1997 Thus, the inhibitory effect of CsA and FK506 on cellular IL-5 mRNA expression can be explained by inhibition of the rate of IL-5 gene transcription. Tacrolimus 39-44 interleukin 5 Homo sapiens 57-61 9271313-11 1997 Thus, the inhibitory effect of CsA and FK506 on cellular IL-5 mRNA expression can be explained by inhibition of the rate of IL-5 gene transcription. Tacrolimus 39-44 interleukin 5 Homo sapiens 124-128 9233699-9 1997 However, local expression of IgM, C3 and T helper 1 cytokines, serum antibodies of IgG and IgM, and delayed-type hypersensitivity were suppressed in the anti-LFA-1 mAb- plus FK506-treated group. Tacrolimus 174-179 immunoglobulin heavy constant mu Mus musculus 29-32 9233699-9 1997 However, local expression of IgM, C3 and T helper 1 cytokines, serum antibodies of IgG and IgM, and delayed-type hypersensitivity were suppressed in the anti-LFA-1 mAb- plus FK506-treated group. Tacrolimus 174-179 immunoglobulin heavy constant mu Mus musculus 91-94 9233699-9 1997 However, local expression of IgM, C3 and T helper 1 cytokines, serum antibodies of IgG and IgM, and delayed-type hypersensitivity were suppressed in the anti-LFA-1 mAb- plus FK506-treated group. Tacrolimus 174-179 integrin alpha L Mus musculus 158-163 9246018-1 1997 OBJECTIVE: To quantitate the effect of ketoconazole, an azole antifungal agent and potent inhibitor of CYP3A4 and P-glycoprotein, on the bioavailability of tacrolimus, a substrate of the CYP3A system and of P-glycoprotein. Tacrolimus 156-166 ATP binding cassette subfamily B member 1 Homo sapiens 114-128 9246018-1 1997 OBJECTIVE: To quantitate the effect of ketoconazole, an azole antifungal agent and potent inhibitor of CYP3A4 and P-glycoprotein, on the bioavailability of tacrolimus, a substrate of the CYP3A system and of P-glycoprotein. Tacrolimus 156-166 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 103-108 9246018-1 1997 OBJECTIVE: To quantitate the effect of ketoconazole, an azole antifungal agent and potent inhibitor of CYP3A4 and P-glycoprotein, on the bioavailability of tacrolimus, a substrate of the CYP3A system and of P-glycoprotein. Tacrolimus 156-166 ATP binding cassette subfamily B member 1 Homo sapiens 207-221 9223178-0 1997 Mean field analysis of FKBP12 complexes with FK506 and rapamycin: implications for a role of crystallographic water molecules in molecular recognition and specificity. Tacrolimus 45-50 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 23-29 9247567-2 1997 Inhibition of T cell activation by the immunosuppressants cyclosporin A (CsA) and FK506 exerts a suppressive effect on the induction of these NF-kappa B-controlled cytokine promoters. Tacrolimus 82-87 nuclear factor kappa B subunit 1 Homo sapiens 142-152 9199340-7 1997 Similar to the upregulation of IL-2 secretion, the transcriptional upregulation of the RE/AP composite element by CD28 is FK506 insensitive. Tacrolimus 122-127 CD28 molecule Homo sapiens 114-118 9223178-1 1997 Mean field analysis of FKBP12 complexes with FK506 and rapamycin has been performed by using structures obtained from molecular docking simulations on a simple, yet robust molecular recognition energy landscape. Tacrolimus 45-50 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 23-29 9223178-4 1997 The stability gap in the FKBP12-FK506 system is determined by two critical water molecules from the effector region that participate in a network of specific hydrogen bond interactions. Tacrolimus 32-37 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 25-31 9223178-5 1997 This interaction pattern protects the integrity and precision of the composite ligand-protein effector surface in the binary FKBP12-FK506 complex and is preserved in the crystal structure of the FKBP12-FK506-calcineurin ternary complex. Tacrolimus 132-137 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 125-131 9223178-5 1997 This interaction pattern protects the integrity and precision of the composite ligand-protein effector surface in the binary FKBP12-FK506 complex and is preserved in the crystal structure of the FKBP12-FK506-calcineurin ternary complex. Tacrolimus 132-137 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 195-201 9223178-5 1997 This interaction pattern protects the integrity and precision of the composite ligand-protein effector surface in the binary FKBP12-FK506 complex and is preserved in the crystal structure of the FKBP12-FK506-calcineurin ternary complex. Tacrolimus 202-207 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 125-131 9223178-5 1997 This interaction pattern protects the integrity and precision of the composite ligand-protein effector surface in the binary FKBP12-FK506 complex and is preserved in the crystal structure of the FKBP12-FK506-calcineurin ternary complex. Tacrolimus 202-207 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 195-201 9169521-6 1997 Synaptic potentiation induced by FK-506 was significantly attenuated by co-injecting BAPTA, heparin/dantrolene (inhibitors of intracellular Ca2+ release), a CaM-binding peptide, or CaM-KII/PKC pseudosubstrate peptides. Tacrolimus 33-39 carbonic anhydrase 2 Rattus norvegicus 140-143 9195923-0 1997 Protein phosphatase 5 is a major component of glucocorticoid receptor.hsp90 complexes with properties of an FK506-binding immunophilin. Tacrolimus 108-113 nuclear receptor subfamily 3 group C member 1 Homo sapiens 46-69 9195923-0 1997 Protein phosphatase 5 is a major component of glucocorticoid receptor.hsp90 complexes with properties of an FK506-binding immunophilin. Tacrolimus 108-113 heat shock protein 90 alpha family class A member 1 Homo sapiens 70-75 9195923-2 1997 The immunophilin, which can be of the FK506- or cyclosporin A-binding class, binds to hsp90 via its tetratricopeptide repeat (TPR) domain, and different receptor heterocomplexes exist depending upon which immunophilin occupies the TPR-binding region of hsp90. Tacrolimus 38-43 heat shock protein 90 alpha family class A member 1 Homo sapiens 86-91 9195923-7 1997 Approximately one-half of the GR.hsp90 heterocomplexes in L cell cytosol contains an immunophilin with high affinity FK506 binding activity, such as FKBP51 or FKBP52, and approximately 35% contains PP5. Tacrolimus 117-122 nuclear receptor subfamily 3 group C member 1 Homo sapiens 30-32 9195923-7 1997 Approximately one-half of the GR.hsp90 heterocomplexes in L cell cytosol contains an immunophilin with high affinity FK506 binding activity, such as FKBP51 or FKBP52, and approximately 35% contains PP5. Tacrolimus 117-122 heat shock protein 90 alpha family class A member 1 Homo sapiens 33-38 9195923-11 1997 Of the 9 residues in this portion of FKBP52 involved in high affinity interactions with FK506, 3 residues are retained and 4 have homologous substitutions in PP5. Tacrolimus 88-93 FKBP prolyl isomerase 4 Homo sapiens 37-43 9195923-12 1997 Although immunoadsorbed PP5 did not bind [3H]FK506, we found that both rabbit PP5 in reticulocyte lysate and purified rat PP5 were specifically retained by an FK506 affinity matrix. Tacrolimus 159-164 protein phosphatase 5 catalytic subunit Homo sapiens 78-81 9195923-12 1997 Although immunoadsorbed PP5 did not bind [3H]FK506, we found that both rabbit PP5 in reticulocyte lysate and purified rat PP5 were specifically retained by an FK506 affinity matrix. Tacrolimus 159-164 protein phosphatase 5, catalytic subunit Rattus norvegicus 78-81 9195923-13 1997 Thus, we propose that PP5 possesses properties of an immunophilin with low affinity FK506 binding activity and that it determines a major portion of the native GR heterocomplexes in L cell cytosol. Tacrolimus 84-89 protein phosphatase 5 catalytic subunit Homo sapiens 22-25 9182928-4 1997 RESULTS: Interleukin-2 production was suppressed in patients treated with tacrolimus. Tacrolimus 74-84 interleukin 2 Homo sapiens 9-22 9190906-2 1997 Furthermore we show that IL-10 production by human T cell lines, such as IFN-gamma and IL-2, is inhibited by the immunosuppressive drugs cyclosporin A and FK506. Tacrolimus 155-160 interleukin 10 Homo sapiens 25-30 9190906-2 1997 Furthermore we show that IL-10 production by human T cell lines, such as IFN-gamma and IL-2, is inhibited by the immunosuppressive drugs cyclosporin A and FK506. Tacrolimus 155-160 interferon gamma Homo sapiens 73-82 9190906-2 1997 Furthermore we show that IL-10 production by human T cell lines, such as IFN-gamma and IL-2, is inhibited by the immunosuppressive drugs cyclosporin A and FK506. Tacrolimus 155-160 interleukin 2 Homo sapiens 87-91 9304807-2 1997 FK-506 non-competitively inhibited the aniline p-hydroxylase, p-nitroanisole O-demethylase and lidocaine N-deethylase activities of cytochrome P-450-linked monooxygenase systems, these activities being mainly catalyzed by cytochromes P-450 CYP2E1, CYP2C11 and CYP3A4, respectively, and the Ki values of the activities for FK-506 were determined to be 605, 491 and 97 microM, respectively. Tacrolimus 0-6 cytochrome P450, subfamily 2, polypeptide 11 Rattus norvegicus 248-255 9182723-6 1997 These St-induced effects were inhibited by pretreatment with FK506, indicating that CaN activity was required for the observed effects on NFATp. Tacrolimus 61-66 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 2 Mus musculus 138-143 9304807-3 1997 The inhibition of cytochrome P-450-linked monooxygenase systems by FK-506 seemed to involve the direct inhibition of cytochromes P-450 because the NADPH-cytochrome c reductase and NADPH-ferricyanide reductase activities of NADPH-cytochrome P-450 reductase were not affected by the presence of 1 mM FK-506 at all. Tacrolimus 67-73 cytochrome p450 oxidoreductase Rattus norvegicus 223-255 9145907-3 1997 Disruption of the association between immunophilin FKBP12 and Ry1R with FK 506 or rapamycin completely eliminates PCB 95-enhanced binding of [3H]ryanodine (IC50 approximately 35 microM) to Ry1R and PCB 95-induced release of Ca2+ from actively loaded SR vesicles (IC50 approximately 11 microM), demonstrating a FKBP12-dependent mechanism. Tacrolimus 72-78 FKBP prolyl isomerase 1A Homo sapiens 38-57 9154824-7 1997 FK-506, another ligand of FKBP12 affecting the phosphatase calcineurin, did not antagonize but shared the effect of rapamycin. Tacrolimus 0-6 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 26-32 9145907-4 1997 FK 506 selectively eliminates PCB 95-induced Ca2+ release from SR because Ry1R maintains responsiveness to caffeine and Ca2+. Tacrolimus 0-6 pyruvate carboxylase Homo sapiens 30-33 9145907-9 1997 The actions of PCB 95 on SR-loading capacity are additive with those of FK 506. Tacrolimus 72-78 pyruvate carboxylase Homo sapiens 15-18 9145907-3 1997 Disruption of the association between immunophilin FKBP12 and Ry1R with FK 506 or rapamycin completely eliminates PCB 95-enhanced binding of [3H]ryanodine (IC50 approximately 35 microM) to Ry1R and PCB 95-induced release of Ca2+ from actively loaded SR vesicles (IC50 approximately 11 microM), demonstrating a FKBP12-dependent mechanism. Tacrolimus 72-78 pyruvate carboxylase Homo sapiens 114-117 9145907-3 1997 Disruption of the association between immunophilin FKBP12 and Ry1R with FK 506 or rapamycin completely eliminates PCB 95-enhanced binding of [3H]ryanodine (IC50 approximately 35 microM) to Ry1R and PCB 95-induced release of Ca2+ from actively loaded SR vesicles (IC50 approximately 11 microM), demonstrating a FKBP12-dependent mechanism. Tacrolimus 72-78 FKBP prolyl isomerase 1A Homo sapiens 51-57 9111057-4 1997 One of the ligand-dependent clones, ARA9, encodes a novel 330-amino acid protein with regions of amino acid sequence similarity to the 52-kDa FK506-binding protein known to be associated with the glucocorticoid receptor. Tacrolimus 142-147 aryl hydrocarbon receptor interacting protein Homo sapiens 36-40 9165550-0 1997 Time-dependent disposition of tacrolimus and its effect on endothelin-1 in liver allograft recipients. Tacrolimus 30-40 endothelin 1 Homo sapiens 59-71 9165550-1 1997 STUDY OBJECTIVE: To characterize a time-dependent disposition of oral tacrolimus and its relationship with plasma endothelin-1 concentrations. Tacrolimus 70-80 endothelin 1 Homo sapiens 114-126 9111057-4 1997 One of the ligand-dependent clones, ARA9, encodes a novel 330-amino acid protein with regions of amino acid sequence similarity to the 52-kDa FK506-binding protein known to be associated with the glucocorticoid receptor. Tacrolimus 142-147 nuclear receptor subfamily 3 group C member 1 Homo sapiens 196-219 9103428-0 1997 IL-2-induced IL-5 synthesis, but not proliferation, of human CD4+ T cells is suppressed by FK506. Tacrolimus 91-96 interleukin 2 Homo sapiens 0-4 9103428-0 1997 IL-2-induced IL-5 synthesis, but not proliferation, of human CD4+ T cells is suppressed by FK506. Tacrolimus 91-96 interleukin 5 Homo sapiens 13-17 9103428-0 1997 IL-2-induced IL-5 synthesis, but not proliferation, of human CD4+ T cells is suppressed by FK506. Tacrolimus 91-96 CD4 molecule Homo sapiens 61-64 9103428-2 1997 Immunosuppressant FK506 suppressed IL-5 synthesis of T cells activated through TCR in a dose-dependent manner. Tacrolimus 18-23 interleukin 5 Homo sapiens 35-39 9103428-3 1997 IL-5 gene transcription and protein synthesis were also induced in the same T cell clones upon stimulation with IL-2 and were suppressed by FK506 in a dose response similar to that induced by TCR stimulation. Tacrolimus 140-145 interleukin 5 Homo sapiens 0-4 9103428-5 1997 Human IL-5 promoter/enhancer-luciferase gene construct transfected to T cell clones was transcribed upon either TCR or IL-2 stimulation and was clearly down-regulated by FK506, indicating that the approximately 500-bp human IL-5 gene segment located 5" upstream of the coding region contained FK506-sensitive enhancer elements. Tacrolimus 170-175 interleukin 5 Homo sapiens 6-10 9103428-5 1997 Human IL-5 promoter/enhancer-luciferase gene construct transfected to T cell clones was transcribed upon either TCR or IL-2 stimulation and was clearly down-regulated by FK506, indicating that the approximately 500-bp human IL-5 gene segment located 5" upstream of the coding region contained FK506-sensitive enhancer elements. Tacrolimus 170-175 interleukin 5 Homo sapiens 224-228 9103428-5 1997 Human IL-5 promoter/enhancer-luciferase gene construct transfected to T cell clones was transcribed upon either TCR or IL-2 stimulation and was clearly down-regulated by FK506, indicating that the approximately 500-bp human IL-5 gene segment located 5" upstream of the coding region contained FK506-sensitive enhancer elements. Tacrolimus 293-298 interleukin 5 Homo sapiens 6-10 9103428-5 1997 Human IL-5 promoter/enhancer-luciferase gene construct transfected to T cell clones was transcribed upon either TCR or IL-2 stimulation and was clearly down-regulated by FK506, indicating that the approximately 500-bp human IL-5 gene segment located 5" upstream of the coding region contained FK506-sensitive enhancer elements. Tacrolimus 293-298 interleukin 5 Homo sapiens 224-228 9103428-6 1997 Our present findings clearly indicate that FK506-sensitive signaling molecules are involved in T cell IL-5 production induced by both TCR and IL-2 stimulation and suggest that IL-2 receptor signal leading to IL-5 gene transcription is transduced by a unique FK506-sensitive pathway other than the Ca2+-dependent signal transduction pathway, such as the calcineurin-NF-AT system. Tacrolimus 43-48 interleukin 5 Homo sapiens 102-106 9112351-2 1997 BACKGROUND: Tacrolimus (FK506), a macrolide molecule that potently inhibits the expression of interleukin 2 by T lymphocytes, represents a potential major advance in the management of rejection following solid-organ transplantation. Tacrolimus 12-22 interleukin 2 Homo sapiens 94-107 9112351-2 1997 BACKGROUND: Tacrolimus (FK506), a macrolide molecule that potently inhibits the expression of interleukin 2 by T lymphocytes, represents a potential major advance in the management of rejection following solid-organ transplantation. Tacrolimus 24-29 interleukin 2 Homo sapiens 94-107 9103428-6 1997 Our present findings clearly indicate that FK506-sensitive signaling molecules are involved in T cell IL-5 production induced by both TCR and IL-2 stimulation and suggest that IL-2 receptor signal leading to IL-5 gene transcription is transduced by a unique FK506-sensitive pathway other than the Ca2+-dependent signal transduction pathway, such as the calcineurin-NF-AT system. Tacrolimus 43-48 interleukin 2 Homo sapiens 142-146 9103428-6 1997 Our present findings clearly indicate that FK506-sensitive signaling molecules are involved in T cell IL-5 production induced by both TCR and IL-2 stimulation and suggest that IL-2 receptor signal leading to IL-5 gene transcription is transduced by a unique FK506-sensitive pathway other than the Ca2+-dependent signal transduction pathway, such as the calcineurin-NF-AT system. Tacrolimus 43-48 interleukin 2 Homo sapiens 176-180 9103428-6 1997 Our present findings clearly indicate that FK506-sensitive signaling molecules are involved in T cell IL-5 production induced by both TCR and IL-2 stimulation and suggest that IL-2 receptor signal leading to IL-5 gene transcription is transduced by a unique FK506-sensitive pathway other than the Ca2+-dependent signal transduction pathway, such as the calcineurin-NF-AT system. Tacrolimus 43-48 interleukin 5 Homo sapiens 208-212 9103428-6 1997 Our present findings clearly indicate that FK506-sensitive signaling molecules are involved in T cell IL-5 production induced by both TCR and IL-2 stimulation and suggest that IL-2 receptor signal leading to IL-5 gene transcription is transduced by a unique FK506-sensitive pathway other than the Ca2+-dependent signal transduction pathway, such as the calcineurin-NF-AT system. Tacrolimus 258-263 interleukin 2 Homo sapiens 176-180 9103428-6 1997 Our present findings clearly indicate that FK506-sensitive signaling molecules are involved in T cell IL-5 production induced by both TCR and IL-2 stimulation and suggest that IL-2 receptor signal leading to IL-5 gene transcription is transduced by a unique FK506-sensitive pathway other than the Ca2+-dependent signal transduction pathway, such as the calcineurin-NF-AT system. Tacrolimus 258-263 interleukin 5 Homo sapiens 208-212 9147322-8 1997 Inhibition of FKBP by the immunosuppressants FK506 or rapamycin increased the duration of spontaneous or depolarization-evoked Ca2+ sparks 6- to 7-fold. Tacrolimus 45-50 carbonic anhydrase 2 Rattus norvegicus 127-130 9147322-11 1997 FK506 potentiated and prolonged electrically stimulated [Ca2+]i transients and contractions, but did not affect the amplitude and kinetics of the L-type Ca2+ channel current. Tacrolimus 0-5 carbonic anhydrase 2 Rattus norvegicus 57-60 9147322-13 1997 In planar lipid bilayers, FK506 (15 microM) prolonged approximately 7-fold the mean open lifetime of reconstituted single RyRs, induced the appearance of long-lasting subconductance states, and markedly slowed the spontaneous decay of RyR activity elicited by fast and sustained Ca2+ stimuli. Tacrolimus 26-31 ryanodine receptor 2 Rattus norvegicus 122-125 9104801-0 1997 Extremely high serum level of IgE during immunosuppressive therapy: paradoxical effect of cyclosporine A and tacrolimus. Tacrolimus 109-119 immunoglobulin heavy constant epsilon Homo sapiens 30-33 9147322-13 1997 In planar lipid bilayers, FK506 (15 microM) prolonged approximately 7-fold the mean open lifetime of reconstituted single RyRs, induced the appearance of long-lasting subconductance states, and markedly slowed the spontaneous decay of RyR activity elicited by fast and sustained Ca2+ stimuli. Tacrolimus 26-31 carbonic anhydrase 2 Rattus norvegicus 279-282 9113492-8 1997 Cyclosporin and tacrolimus cause post-transplant diabetes mellitus by a number of mechanisms, including decreased insulin secretion, increased insulin resistance or a direct toxic effect on the beta cell. Tacrolimus 16-26 insulin Homo sapiens 114-121 9104801-1 1997 A case of X-linked autoimmune enteropathy was successfully treated with cyclosporine A (CsA) or tacrolimus (FK506) and developed extremely high serum levels of IgE during the immunosuppressive therapy. Tacrolimus 108-113 immunoglobulin heavy constant epsilon Homo sapiens 160-163 9104801-2 1997 Serum IgE levels increased from 190 to 1,000-2,500 IU/ml with CsA therapy and as high as 80,000 IU/ml with subsequent FK506 therapy. Tacrolimus 118-123 immunoglobulin heavy constant epsilon Homo sapiens 6-9 9104801-4 1997 Thereafter, serum IgE levels progressively decreased in parallel with a reduced dosage of FK506. Tacrolimus 90-95 immunoglobulin heavy constant epsilon Homo sapiens 18-21 9096348-3 1997 FK1012 is used as a pharmacological mediator of dimerization to bring together FK506 binding domains, taken from the endogenous protein FKBP12. Tacrolimus 79-84 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 136-142 9143223-0 1997 Calcineurin inhibitor, FK506, prevents reduction in the binding capacity of cyclic AMP-dependent protein kinase in ischemic gerbil brain. Tacrolimus 23-28 calcineurin binding protein 1 Homo sapiens 0-21 9143223-7 1997 In the ischemia group of gerbils, FK506 prevented any significant reduction of cAMP binding in the hippocampus CA1 and cerebral cortices on the ischemic side, whereas it exerted no significant influence on the cAMP binding of the nonischemic side. Tacrolimus 34-39 carbonic anhydrase 1 Homo sapiens 111-114 9101372-19 1997 BGP levels were significantly increased in the CsA (p < 0.01) and FK506 (p < 0.001) groups on day 28. Tacrolimus 69-74 bone gamma-carboxyglutamate protein Rattus norvegicus 0-3 9103242-9 1997 Blocking the resulting damage of this inflammatory reaction with anti-LFA-1 in animals also treated with FK506, a powerful immunosuppressant for gene therapy, largely increased the long-term transgene expression compared with mice only treated with FK506. Tacrolimus 105-110 integrin alpha L Mus musculus 70-75 9103242-9 1997 Blocking the resulting damage of this inflammatory reaction with anti-LFA-1 in animals also treated with FK506, a powerful immunosuppressant for gene therapy, largely increased the long-term transgene expression compared with mice only treated with FK506. Tacrolimus 249-254 integrin alpha L Mus musculus 70-75 9127694-0 1997 Identification of a 37 kDa tacrolimus, sirolimus and cyclosporine binding immunophilin possessing glyceraldehyde 3-phosphate dehydrogenase activity isolated from the Jurkat T cell line. Tacrolimus 27-37 glyceraldehyde-3-phosphate dehydrogenase Homo sapiens 98-138 9125197-5 1997 The 51.2 kDa protein encoded by this gene shares 87% identity to murine FKBP51 and demonstrates a similar IC50 value for the FK506-mediated inhibition of calcineurin phosphatase in vitro. Tacrolimus 125-130 FK506 binding protein 5 Mus musculus 72-78 9178371-2 1997 The in vitro metabolism of tacrolimus (TAC, FK 506) was investigated in the liver microsomes prepared from normal rats as well as rats treated with dexamethasone (DEX) and rifampin (RIF). Tacrolimus 27-37 tachykinin precursor 3 Rattus norvegicus 39-42 9113092-0 1997 Decrease in kidney calbindin-D 28kDa as a possible mechanism mediating cyclosporine A- and FK-506-induced calciuria and tubular mineralization. Tacrolimus 91-97 calbindin 1 Rattus norvegicus 19-33 9127694-1 1997 OBJECTIVE: The isolation and partial characterization of a 37 kDa minor immunophilin from the Jurkat cell line which binds to cyclosporine (CsA), Tacrolimus (FK506) and Sirolimus (RAPA). Tacrolimus 158-163 transcriptional regulating factor 1 Homo sapiens 180-184 9032415-6 1997 The assay is very sensitive and selective for immunosuppressive compounds inhibiting IL-2 gene expression such as cyclosporine (CsA) and FK506, their active metabolites and derivatives, but not for others such as rapamycin. Tacrolimus 137-142 interleukin 2 Homo sapiens 85-89 9061187-0 1997 Modeling the interaction between FK506 and FKBP12: a mechanism for formation of the calcineurin inhibitory complex. Tacrolimus 33-38 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 43-49 9013543-4 1997 cADPR as well as FK506 bound to FK506-binding protein 12.6 (FKBP12.6), which we also found occurs naturally in islet microsomes. Tacrolimus 17-22 FKBP prolyl isomerase 1B Homo sapiens 32-58 9013543-4 1997 cADPR as well as FK506 bound to FK506-binding protein 12.6 (FKBP12.6), which we also found occurs naturally in islet microsomes. Tacrolimus 17-22 FKBP prolyl isomerase 1B Homo sapiens 60-68 9061187-1 1997 FK506 is a naturally occurring immunosuppressant whose mode of action involves formation of an initial complex with the cytosolic protein FKBP12. Tacrolimus 0-5 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 138-144 9061187-7 1997 Comparison of the structure of Z-Arg32-ascomycin in water with structures of FK506 bound to FKBP12 indicate that the conformation of the pipecolate region is conserved during the binding process. Tacrolimus 77-82 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 92-98 9061187-10 1997 From computer models and molecular dynamics simulations of interactions between FK506 and FKBP12 we suggest that the conformational changes observed in bound FK506 are induced by the interaction between the 80"s loop of FKBP12 and the pyranose ring of FKBP12. Tacrolimus 80-85 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 90-96 9061187-10 1997 From computer models and molecular dynamics simulations of interactions between FK506 and FKBP12 we suggest that the conformational changes observed in bound FK506 are induced by the interaction between the 80"s loop of FKBP12 and the pyranose ring of FKBP12. Tacrolimus 80-85 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 220-226 9061187-10 1997 From computer models and molecular dynamics simulations of interactions between FK506 and FKBP12 we suggest that the conformational changes observed in bound FK506 are induced by the interaction between the 80"s loop of FKBP12 and the pyranose ring of FKBP12. Tacrolimus 80-85 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 220-226 9061187-10 1997 From computer models and molecular dynamics simulations of interactions between FK506 and FKBP12 we suggest that the conformational changes observed in bound FK506 are induced by the interaction between the 80"s loop of FKBP12 and the pyranose ring of FKBP12. Tacrolimus 158-163 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 90-96 9051292-4 1997 The effect on ODC was specific for the intracellular signalling pathway leading to activation of p70S6k, as the immunosuppressant FK 506 was without effect on ODC activity. Tacrolimus 130-136 ornithine decarboxylase 1 Rattus norvegicus 14-17 9051292-4 1997 The effect on ODC was specific for the intracellular signalling pathway leading to activation of p70S6k, as the immunosuppressant FK 506 was without effect on ODC activity. Tacrolimus 130-136 ribosomal protein S6 kinase B1 Rattus norvegicus 97-103 9061187-10 1997 From computer models and molecular dynamics simulations of interactions between FK506 and FKBP12 we suggest that the conformational changes observed in bound FK506 are induced by the interaction between the 80"s loop of FKBP12 and the pyranose ring of FKBP12. Tacrolimus 158-163 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 220-226 9061187-10 1997 From computer models and molecular dynamics simulations of interactions between FK506 and FKBP12 we suggest that the conformational changes observed in bound FK506 are induced by the interaction between the 80"s loop of FKBP12 and the pyranose ring of FKBP12. Tacrolimus 158-163 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 220-226 9080295-7 1997 FK506 also suppressed anti-H alpha 125-147 and anti-rat AChR antibody production accompanied by a decrease in the antigen-specific T cell response against H alpha 125-147. Tacrolimus 0-5 cholinergic receptor nicotinic alpha 2 subunit Rattus norvegicus 56-60 9045922-4 1997 Blocking the IL-2 pathway by cyclosporin A, FK506, rapamycin, anti-IL-2 or CD25 antibodies, prevented the development of sensitivity to apoptosis. Tacrolimus 44-49 interleukin 2 Homo sapiens 13-17 9123208-0 1997 Impaired T-cell IL-10 secretion and CD4 helper function in liver transplant patients treated with tacrolimus. Tacrolimus 98-108 interleukin 10 Homo sapiens 16-21 9123208-0 1997 Impaired T-cell IL-10 secretion and CD4 helper function in liver transplant patients treated with tacrolimus. Tacrolimus 98-108 CD4 molecule Homo sapiens 36-39 9123209-0 1997 High-dose cellular IL-10 exacerbates rejection and reverses effects of cyclosporine and tacrolimus in Mouse cardiac transplantation. Tacrolimus 88-98 interleukin 10 Mus musculus 19-24 9209689-0 1997 Implication of CYP 3A in the toxicity of cyclosporin G (CsG), cyclosporin A (CsA) and FK506 on rat hepatocytes in primary culture. Tacrolimus 86-91 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 15-21 9016789-2 1997 FK506, in contrast, had no effect on DEX-induced apoptosis; moreover, an excess of FK506 reversed the potentiation of apoptosis by RAP, indicating that RAP exerts its effects through binding to FKBP. Tacrolimus 83-88 regulatory associated protein of MTOR, complex 1 Mus musculus 131-134 9016789-2 1997 FK506, in contrast, had no effect on DEX-induced apoptosis; moreover, an excess of FK506 reversed the potentiation of apoptosis by RAP, indicating that RAP exerts its effects through binding to FKBP. Tacrolimus 83-88 regulatory associated protein of MTOR, complex 1 Mus musculus 152-155 9016789-2 1997 FK506, in contrast, had no effect on DEX-induced apoptosis; moreover, an excess of FK506 reversed the potentiation of apoptosis by RAP, indicating that RAP exerts its effects through binding to FKBP. Tacrolimus 83-88 FK506 binding protein 1a Mus musculus 194-198 9016789-5 1997 The reduction of JNK activity by RAP was reversed by the addition of an excess of FK506. Tacrolimus 82-87 mitogen-activated protein kinase 8 Mus musculus 17-20 9016789-5 1997 The reduction of JNK activity by RAP was reversed by the addition of an excess of FK506. Tacrolimus 82-87 regulatory associated protein of MTOR, complex 1 Mus musculus 33-36 9209689-1 1997 FK506, cyclosporin A (CsA), and its structural analog cyclosporin G (CsG) are immunosuppressant drugs mainly metabolized by hepatic cytochrome P-450 3A (CYP 3A) oxygenase. Tacrolimus 0-5 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 132-151 9209689-1 1997 FK506, cyclosporin A (CsA), and its structural analog cyclosporin G (CsG) are immunosuppressant drugs mainly metabolized by hepatic cytochrome P-450 3A (CYP 3A) oxygenase. Tacrolimus 0-5 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 153-159 9209689-12 1997 Our results show that the toxicity of the three drugs in rat hepatocytes is dependent on CYP 3A induction: increased for FK506, decreased for CsA and CsG. Tacrolimus 121-126 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 89-95 8955134-3 1996 FKBP59 and FKBP12 belong to the large family of immunophilins that bind the macrolide immunosuppressant drugs FK506 and rapamycin. Tacrolimus 110-115 FKBP prolyl isomerase 4 Homo sapiens 0-6 9117088-7 1997 The antigen-induced interleukin (IL)-5 formation in the BALF and serum was inhibited by FK-506 by 75% in both instances. Tacrolimus 88-94 interleukin 5 Mus musculus 20-38 9117088-13 1997 These findings indicate that antigen-induced in vivo IL-5 release and eosinophil, but not T-cell, infiltration into the bronchial lumen of sensitized BP2 mice are targets for the anti-allergic activities of FK-506. Tacrolimus 207-213 interleukin 5 Mus musculus 53-57 8955134-3 1996 FKBP59 and FKBP12 belong to the large family of immunophilins that bind the macrolide immunosuppressant drugs FK506 and rapamycin. Tacrolimus 110-115 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 11-17 8955134-10 1996 The formation of the complexes between FKBP59 or FKBP12 and FAP48 is prevented by FK506 and rapamycin in a dose-dependent manner. Tacrolimus 82-87 FKBP prolyl isomerase 4 Homo sapiens 39-45 8955134-10 1996 The formation of the complexes between FKBP59 or FKBP12 and FAP48 is prevented by FK506 and rapamycin in a dose-dependent manner. Tacrolimus 82-87 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 49-55 8955134-10 1996 The formation of the complexes between FKBP59 or FKBP12 and FAP48 is prevented by FK506 and rapamycin in a dose-dependent manner. Tacrolimus 82-87 glomulin, FKBP associated protein Homo sapiens 60-65 8981925-0 1996 Effects of tacrolimus (FK506) on human insulin gene expression, insulin mRNA levels, and insulin secretion in HIT-T15 cells. Tacrolimus 23-28 insulin Homo sapiens 39-46 8981925-2 1996 Regulation of insulin gene expression in the beta cell may also involve Ca2+-signaling pathways and FK506 has been associated with insulin-requiring diabetes mellitus during clinical use. Tacrolimus 100-105 insulin Homo sapiens 14-21 8981925-3 1996 The purpose of this study was to characterize the effects of FK506 on human insulin gene transcription, insulin mRNA levels, and insulin secretion using as a model the HIT-T15 beta cell line. Tacrolimus 61-66 insulin Homo sapiens 76-83 8994885-1 1996 Recently, two structures of the Ser/Thr phosphorylase calcineurin in complex with FK506 and its cognate immunophilin, FKBP12, have been reported, both solved by small pharmaceutical companies focused on structure-based drug design. Tacrolimus 82-87 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 118-124 8981925-5 1996 Decreased insulin secretion in the presence of FK506 was also accompanied by a dose-dependent decrease in HIT cell insulin content, insulin mRNA levels, and expression of a human insulin promoter-chloramphenicol acetyl transferase (CAT) reporter gene. Tacrolimus 47-52 insulin Homo sapiens 10-17 8981925-5 1996 Decreased insulin secretion in the presence of FK506 was also accompanied by a dose-dependent decrease in HIT cell insulin content, insulin mRNA levels, and expression of a human insulin promoter-chloramphenicol acetyl transferase (CAT) reporter gene. Tacrolimus 47-52 insulin Homo sapiens 115-122 8981925-5 1996 Decreased insulin secretion in the presence of FK506 was also accompanied by a dose-dependent decrease in HIT cell insulin content, insulin mRNA levels, and expression of a human insulin promoter-chloramphenicol acetyl transferase (CAT) reporter gene. Tacrolimus 47-52 insulin Homo sapiens 115-122 8981925-6 1996 FK506 decreased HIT cell expression of the human insulin promoter-CAT reporter gene by 40% in the presence of both low (0.4 mM) at high (20 mM) glucose concentrations. Tacrolimus 0-5 insulin Homo sapiens 49-56 8981925-8 1996 These findings suggest that FK506 may have direct effects to reversibly inhibit insulin gene transcription, leading to a decline in insulin mRNA levels, insulin synthesis, and ultimately insulin secretion. Tacrolimus 28-33 insulin Homo sapiens 80-87 8981925-8 1996 These findings suggest that FK506 may have direct effects to reversibly inhibit insulin gene transcription, leading to a decline in insulin mRNA levels, insulin synthesis, and ultimately insulin secretion. Tacrolimus 28-33 insulin Homo sapiens 132-139 8981925-8 1996 These findings suggest that FK506 may have direct effects to reversibly inhibit insulin gene transcription, leading to a decline in insulin mRNA levels, insulin synthesis, and ultimately insulin secretion. Tacrolimus 28-33 insulin Homo sapiens 132-139 8981925-8 1996 These findings suggest that FK506 may have direct effects to reversibly inhibit insulin gene transcription, leading to a decline in insulin mRNA levels, insulin synthesis, and ultimately insulin secretion. Tacrolimus 28-33 insulin Homo sapiens 132-139 8971291-12 1996 Water immersion stress increased gastric mucosal IL-1 beta and IL-2 contents 6 h after stress treatment, and these increases were prevented by FK506. Tacrolimus 143-148 interleukin 1 beta Rattus norvegicus 49-58 8971291-12 1996 Water immersion stress increased gastric mucosal IL-1 beta and IL-2 contents 6 h after stress treatment, and these increases were prevented by FK506. Tacrolimus 143-148 interleukin 2 Rattus norvegicus 63-67 8939643-3 1996 The immunosuppressants cyclosporin A and FK506, when complexed with immunophilins, inactivate the protein phosphatase calcineurin, resulting in the inhibition of interleukin-2 gene activation. Tacrolimus 41-46 interleukin 2 Homo sapiens 162-175 8974006-3 1996 NFAT acts at the antigen receptor response element-2 (ARRE-2) sequence in the IL-2 enhancer and is the nuclear target of T cell stimulation signals and the immunosuppressant drugs cyclosporine and FK506, which are potent inhibitors of IL-2 gene transcription. Tacrolimus 197-202 interleukin 2 Homo sapiens 78-82 8906804-4 1996 Interference with the IL-2 pathway was achieved by 1) inhibition of cytokine synthesis using cyclosporin A or FK506, 2) neutralization of IL-2 by anti-IL-2 Ab, 3) inhibition of binding to IL-2R by CD25 mAb, and 4) blocking of IL-2R signaling by rapamycin. Tacrolimus 110-115 interleukin 2 Homo sapiens 22-26 8906804-6 1996 While the addition of rIL-2 reversed the inhibitory effect of cyclosporin A and FK506, the addition of rIL-4, rIL-7, or rIFN-gamma did not, although these cytokines induced progression into the S phase of the cell cycle. Tacrolimus 80-85 interleukin 2 Rattus norvegicus 22-27 8946659-1 1996 In a prospective study, we evaluated a novel enzyme-linked immunosorbent assay (ELISA) (Pro-Trac) for determining tacrolimus (FK 506) concentrations in whole blood. Tacrolimus 114-124 T cell receptor alpha constant Homo sapiens 92-96 8946659-1 1996 In a prospective study, we evaluated a novel enzyme-linked immunosorbent assay (ELISA) (Pro-Trac) for determining tacrolimus (FK 506) concentrations in whole blood. Tacrolimus 126-132 T cell receptor alpha constant Homo sapiens 92-96 8946659-8 1996 We conclude that if assay precision in the upper range is improved, the Pro-Trac ELISA might be a valuable alternative to the MEIA for therapeutic drug monitoring of tacrolimus. Tacrolimus 166-176 T cell receptor alpha constant Homo sapiens 76-80 8962237-0 1996 Decreased Fas antigen expression of cultured hepatocytes with FK 506. Tacrolimus 62-68 Fas cell surface death receptor Homo sapiens 10-21 8917502-7 1996 Using this system, we screened a Jurkat cDNA library fused to the transcriptional activation domain in yeast expressing the hormone binding domain of rat glucocorticoid receptor-LexA DNA binding domain fusion protein in the presence of dexamethasone-FK506 heterodimer. Tacrolimus 250-255 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 154-177 8974006-3 1996 NFAT acts at the antigen receptor response element-2 (ARRE-2) sequence in the IL-2 enhancer and is the nuclear target of T cell stimulation signals and the immunosuppressant drugs cyclosporine and FK506, which are potent inhibitors of IL-2 gene transcription. Tacrolimus 197-202 interleukin 2 Homo sapiens 235-239 8816436-2 1996 In both cases, induction of TNF-alpha gene transcription can be blocked by the immunosuppressants cyclosporin A and FK506, which suggested a role for the NFAT family of proteins in the regulation of the gene in B cells. Tacrolimus 116-121 tumor necrosis factor Homo sapiens 28-37 8986461-1 1996 FK-506 (Tacrolimus) has been shown to block T cell proliferation in vitro by inhibiting the generation of several lymphokines, especially interleukin (IL)-2, but little direct evidence is available to support the view that the immunosuppressive effects of FK-506 in vivo are mediated by a similar inhibition of lymphokine cascade. Tacrolimus 0-6 interleukin 2 Mus musculus 138-156 8986461-1 1996 FK-506 (Tacrolimus) has been shown to block T cell proliferation in vitro by inhibiting the generation of several lymphokines, especially interleukin (IL)-2, but little direct evidence is available to support the view that the immunosuppressive effects of FK-506 in vivo are mediated by a similar inhibition of lymphokine cascade. Tacrolimus 8-18 interleukin 2 Mus musculus 138-156 8986461-6 1996 In contrast to the effects of FK-506 on IL-2 and IFN-gamma productions in MLN, IL-1 and tumor necrosis factor-alpha in the intestinal wall, which were enhanced by H. nana infection, were not completely decreased as a result of 10.0 mg/kg FK-506 treatment. Tacrolimus 30-36 interleukin 2 Mus musculus 40-44 8986461-6 1996 In contrast to the effects of FK-506 on IL-2 and IFN-gamma productions in MLN, IL-1 and tumor necrosis factor-alpha in the intestinal wall, which were enhanced by H. nana infection, were not completely decreased as a result of 10.0 mg/kg FK-506 treatment. Tacrolimus 30-36 interferon gamma Mus musculus 49-58 8986461-7 1996 The reverse transcriptase-PCR revealed complete inhibition of IL-2 and IFN-gamma mRNA expression on mesenteric L3T4+ cells that were induced by H. nana infection, when mice were given 10.0 mg/kg/day FK-506 for 5 days. Tacrolimus 199-205 interleukin 2 Mus musculus 62-66 8986461-7 1996 The reverse transcriptase-PCR revealed complete inhibition of IL-2 and IFN-gamma mRNA expression on mesenteric L3T4+ cells that were induced by H. nana infection, when mice were given 10.0 mg/kg/day FK-506 for 5 days. Tacrolimus 199-205 interferon gamma Mus musculus 71-80 9010851-9 1996 The complex of CsA or FK506 with CyP or FKBP, respectively, inhibits the activation of calcineurin. Tacrolimus 22-27 peptidylprolyl isomerase G Homo sapiens 33-36 8906215-5 1996 Cyclosporin A, FK-506, and cyclophosphamide clearly inhibited the antigen-induced increase of IL-5 and eosinophils in BALF. Tacrolimus 15-21 interleukin 5 Mus musculus 94-98 8824230-8 1996 On the other hand, both PP1 and PP2B activities but not PP2A activity of cell extracts were suppressed by the addition of cyclosporin A or FK506 in the culture medium. Tacrolimus 139-144 protein phosphatase 1 catalytic subunit gamma Mus musculus 24-27 8816436-2 1996 In both cases, induction of TNF-alpha gene transcription can be blocked by the immunosuppressants cyclosporin A and FK506, which suggested a role for the NFAT family of proteins in the regulation of the gene in B cells. Tacrolimus 116-121 nuclear factor of activated T cells 2 Homo sapiens 154-158 8702959-6 1996 FKBP12 dissociates from TbetaR-I in the presence of a high concentration of FK506. Tacrolimus 76-81 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 0-6 8798476-2 1996 As the FK506 motif that competes with TbetaR-I for FKBP12 resembles an invariant Leu-Pro dipeptide in TbetaR-I, we replaced Leu193 and Pro194 with Ala, along with mutations across the Gly/Ser box. Tacrolimus 7-12 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 51-57 8774885-3 1996 To examine whether induced oligomerization can alter Raf kinase activity, sequences encoding the FK506-binding protein FKBP12 were fused to the amino terminus of c-Raf-1, introducing a binding site for FK506. Tacrolimus 97-102 FKBP prolyl isomerase 1A pseudogene 3 Homo sapiens 119-125 8774885-4 1996 Oligomerization of recombinant FKBP-Raf in situ, induced by the addition of the dimeric FK506 derivative FK1012A, activated Raf kinase activity at least half as well as epidermal growth factor (EGF). Tacrolimus 88-93 zinc fingers and homeoboxes 2 Homo sapiens 36-39 8774885-4 1996 Oligomerization of recombinant FKBP-Raf in situ, induced by the addition of the dimeric FK506 derivative FK1012A, activated Raf kinase activity at least half as well as epidermal growth factor (EGF). Tacrolimus 88-93 zinc fingers and homeoboxes 2 Homo sapiens 124-127 8702959-6 1996 FKBP12 dissociates from TbetaR-I in the presence of a high concentration of FK506. Tacrolimus 76-81 transforming growth factor beta receptor 1 Homo sapiens 24-32 8880916-4 1996 Analysis of 1H-13C HSQC data acquired for the FKBP-12/ 13C-FK506 and FKBP-12/13C-FK506/CnB/BBD complexes indicates that FKBP-12/FK506 and CnB/BBD are in fast exchange in the quaternary complex. Tacrolimus 81-86 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 69-76 8787699-7 1996 Stepwise regression analysis including tacrolimus, its metabolites, and liver function parameters suggested a model including serum activities of gamma-glutamyltransferase, alkaline phosphatase, and alanine aminotransferase as predictors for increased concentrations of demethyl tacrolimus, didemethyl tacrolimus, and the parent drug. Tacrolimus 39-49 glutamic--pyruvic transaminase Homo sapiens 199-223 8883951-8 1996 Complementary experiments with the selective calcineurin inhibitor, FK506, also showed the reduction of LTP threshold in a dose-dependent manner. Tacrolimus 68-73 calcineurin binding protein 1 Rattus norvegicus 45-66 8884530-4 1996 FK506 and CsA inhibit keratinocyte proliferation induced by EGF, TGF-alpha or IL-6. Tacrolimus 0-5 epidermal growth factor Homo sapiens 60-63 8884530-4 1996 FK506 and CsA inhibit keratinocyte proliferation induced by EGF, TGF-alpha or IL-6. Tacrolimus 0-5 transforming growth factor alpha Homo sapiens 65-74 8884530-4 1996 FK506 and CsA inhibit keratinocyte proliferation induced by EGF, TGF-alpha or IL-6. Tacrolimus 0-5 interleukin 6 Homo sapiens 78-82 8884530-6 1996 These findings might indicate that the effects of FK506 and CsA on proliferation of cultured normal human keratinocytes are probably related to direct effects on growth regulation of keratinocytes via EGF, TGF-alpha or IL-6 stimulation. Tacrolimus 50-55 epidermal growth factor Homo sapiens 201-204 8884530-6 1996 These findings might indicate that the effects of FK506 and CsA on proliferation of cultured normal human keratinocytes are probably related to direct effects on growth regulation of keratinocytes via EGF, TGF-alpha or IL-6 stimulation. Tacrolimus 50-55 transforming growth factor alpha Homo sapiens 206-215 8884530-6 1996 These findings might indicate that the effects of FK506 and CsA on proliferation of cultured normal human keratinocytes are probably related to direct effects on growth regulation of keratinocytes via EGF, TGF-alpha or IL-6 stimulation. Tacrolimus 50-55 interleukin 6 Homo sapiens 219-223 8880916-0 1996 Dynamic NMR studies of ligand-receptor interactions: design and analysis of a rapidly exchanging complex of FKBP-12/FK506 with a 24 kDa calcineurin fragment. Tacrolimus 116-121 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 108-115 8880916-2 1996 Using a combination of isotope labeling and isotope edited NMR, we have extended these techniques to characterize interactions of a much larger protein/drug complex, FKBP-12/ FK506 with its receptor protein, calcineurin. Tacrolimus 175-180 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 166-173 8880916-4 1996 Analysis of 1H-13C HSQC data acquired for the FKBP-12/ 13C-FK506 and FKBP-12/13C-FK506/CnB/BBD complexes indicates that FKBP-12/FK506 and CnB/BBD are in fast exchange in the quaternary complex. Tacrolimus 81-86 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 69-76 8880916-4 1996 Analysis of 1H-13C HSQC data acquired for the FKBP-12/ 13C-FK506 and FKBP-12/13C-FK506/CnB/BBD complexes indicates that FKBP-12/FK506 and CnB/BBD are in fast exchange in the quaternary complex. Tacrolimus 59-64 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 46-53 8854096-0 1996 Transient immunosuppression by FK506 permits a sustained high-level dystrophin expression after adenovirus-mediated dystrophin minigene transfer to skeletal muscles of adult dystrophic (mdx) mice. Tacrolimus 31-36 dystrophin, muscular dystrophy Mus musculus 68-78 8880916-4 1996 Analysis of 1H-13C HSQC data acquired for the FKBP-12/ 13C-FK506 and FKBP-12/13C-FK506/CnB/BBD complexes indicates that FKBP-12/FK506 and CnB/BBD are in fast exchange in the quaternary complex. Tacrolimus 81-86 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 69-76 8880916-4 1996 Analysis of 1H-13C HSQC data acquired for the FKBP-12/ 13C-FK506 and FKBP-12/13C-FK506/CnB/BBD complexes indicates that FKBP-12/FK506 and CnB/BBD are in fast exchange in the quaternary complex. Tacrolimus 81-86 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 69-76 8702500-7 1996 Similarly, murine mdr3 expression confers resistance to the immunosuppressants cyclosporin A (CsA) and FK506 in a CsA-FK506-sensitive vph6 mutant yeast strain. Tacrolimus 103-108 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 18-22 8702500-7 1996 Similarly, murine mdr3 expression confers resistance to the immunosuppressants cyclosporin A (CsA) and FK506 in a CsA-FK506-sensitive vph6 mutant yeast strain. Tacrolimus 118-123 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 18-22 8702500-8 1996 CsA and FK506 are known to partially overcome Pgp-mediated drug resistance, suggesting the targets of these drugs might regulate Pgp function. Tacrolimus 8-13 ATP binding cassette subfamily B member 1 Homo sapiens 46-49 8702500-8 1996 CsA and FK506 are known to partially overcome Pgp-mediated drug resistance, suggesting the targets of these drugs might regulate Pgp function. Tacrolimus 8-13 ATP binding cassette subfamily B member 1 Homo sapiens 129-132 8702500-12 1996 Our results support the model that immunosuppressants reverse multidrug resistance by competing with other Pgp substrates but reveal that inhibition of FKBP12-dependent Pgp function may also contribute to reversal of multidrug resistance by FK506 and rapamycin. Tacrolimus 241-246 ATP binding cassette subfamily B member 1 Homo sapiens 107-110 8702500-12 1996 Our results support the model that immunosuppressants reverse multidrug resistance by competing with other Pgp substrates but reveal that inhibition of FKBP12-dependent Pgp function may also contribute to reversal of multidrug resistance by FK506 and rapamycin. Tacrolimus 241-246 ATP binding cassette subfamily B member 1 Homo sapiens 169-172 8757216-0 1996 Effects of cyclosporin A and FK-506 on stem cell factor-induced histamine secretion and growth of human mast cells. Tacrolimus 29-35 KIT ligand Homo sapiens 39-55 8757216-2 1996 In this study the effects of cyclosporin A (CSA) and FK-506, two potent immunosuppressive drugs, on SCF-dependent histamine release and growth of human MCs were analyzed. Tacrolimus 53-59 KIT ligand Homo sapiens 100-103 8757216-5 1996 The effects of CSA and FK-506 on SCF-dependent release of histamine were dose-dependent (IC50: CSA, 1 to 10 ng/ml; FK-506, 0.3 to 3 ng/ml). Tacrolimus 23-29 KIT ligand Homo sapiens 33-36 8757216-5 1996 The effects of CSA and FK-506 on SCF-dependent release of histamine were dose-dependent (IC50: CSA, 1 to 10 ng/ml; FK-506, 0.3 to 3 ng/ml). Tacrolimus 23-29 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 95-98 8757216-5 1996 The effects of CSA and FK-506 on SCF-dependent release of histamine were dose-dependent (IC50: CSA, 1 to 10 ng/ml; FK-506, 0.3 to 3 ng/ml). Tacrolimus 115-121 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 15-18 8757216-5 1996 The effects of CSA and FK-506 on SCF-dependent release of histamine were dose-dependent (IC50: CSA, 1 to 10 ng/ml; FK-506, 0.3 to 3 ng/ml). Tacrolimus 115-121 KIT ligand Homo sapiens 33-36 8757216-9 1996 Together, CSA and FK-506 inhibit SCF-dependent release of histamine from human MCs and even augment SCF-dependent growth of human MCs in long-term culture. Tacrolimus 18-24 KIT ligand Homo sapiens 33-36 8757216-9 1996 Together, CSA and FK-506 inhibit SCF-dependent release of histamine from human MCs and even augment SCF-dependent growth of human MCs in long-term culture. Tacrolimus 18-24 KIT ligand Homo sapiens 100-103 8757620-2 1996 Here we show that FK506-suppressible, delayed c-Rel induction is similar in B and T cells and is regulated by mRNA production. Tacrolimus 18-23 REL proto-oncogene, NF-kB subunit Homo sapiens 46-51 8857552-2 1996 Interpatient differences in liver CYP3A4 activity, as measured by the ERMBT, seem to account, for the most part, for interindividual differences in the kinetics of cyclosporin A and FK506. Tacrolimus 182-187 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 34-40 8794888-2 1996 CsA and FK506 associate with intracellular binding proteins (i.e., CsA with cyclophilin A and FK506 with FKBP12) to form protein/drug complexes that suppress the immune system by preventing activation of T cells in response to antigen presentation. Tacrolimus 8-13 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 105-111 8794888-2 1996 CsA and FK506 associate with intracellular binding proteins (i.e., CsA with cyclophilin A and FK506 with FKBP12) to form protein/drug complexes that suppress the immune system by preventing activation of T cells in response to antigen presentation. Tacrolimus 94-99 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 105-111 8794888-4 1996 In previous studies, we identified calcineurin mutations that block binding by the cyclophilin A/CsA or FKBP12/FK506 complexes and thereby render yeast cells resistant to the antifungal effects of CsA or FK506. Tacrolimus 111-116 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 104-110 8794888-4 1996 In previous studies, we identified calcineurin mutations that block binding by the cyclophilin A/CsA or FKBP12/FK506 complexes and thereby render yeast cells resistant to the antifungal effects of CsA or FK506. Tacrolimus 204-209 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 104-110 8874888-1 1996 The effects of FK506, a Ca2+/calmodulin-dependent phosphatase 2B (calcineurin) inhibitor, on the NMDA receptor-mediated potentials and synaptic plasticity were investigated in the CA1 region of the rat hippocampus. Tacrolimus 15-20 carbonic anhydrase 2 Rattus norvegicus 24-27 8874888-1 1996 The effects of FK506, a Ca2+/calmodulin-dependent phosphatase 2B (calcineurin) inhibitor, on the NMDA receptor-mediated potentials and synaptic plasticity were investigated in the CA1 region of the rat hippocampus. Tacrolimus 15-20 carbonic anhydrase 1 Rattus norvegicus 180-183 8874888-6 1996 The differential effects of FK506 on LTP and depotentiation may attribute to the partial inhibition on the activity of NMDA receptors and the subsequent attenuation of intracellular Ca2+ increase. Tacrolimus 28-33 carbonic anhydrase 2 Rattus norvegicus 182-185 8757620-1 1996 c-Rel induction in activated lymphocytes is suppressed by the immunosuppressive drug, FK506. Tacrolimus 86-91 REL proto-oncogene, NF-kB subunit Homo sapiens 0-5 8854096-3 1996 In this study we treated adult dystrophic (mdx) mice with daily subcutaneous injections of the immunosuppressive drug FK506 (tacrolimus) over 5, 10, 30 and 60 days after AV-mediated dystrophin gene transfer and compared the transduction level with saline-injected mdx controls. Tacrolimus 118-123 dystrophin, muscular dystrophy Mus musculus 182-192 8854096-4 1996 We show that daily FK506 treatment after AV-mediated dystrophin gene transfer into adult mdx muscle results in the maintenance of the initial transgene expression for at least 2 months, even when FK506 treatment was discontinued after 1 month. Tacrolimus 19-24 dystrophin, muscular dystrophy Mus musculus 53-63 8854096-6 1996 Moreover, we find that FK506 efficiently suppresses the humoral immune response against both the vector proteins and the transgene protein product (dystrophin). Tacrolimus 23-28 dystrophin, muscular dystrophy Mus musculus 148-158 8757947-1 1996 Differential regulation of Fas (CD95) versus Fas ligand expression by cyclosporin A and FK506. Tacrolimus 88-93 Fas (TNF receptor superfamily member 6) Mus musculus 32-36 8692927-11 1996 Like the mammalian and yeast FKBP13, the recombinant VfFKBP15 protein has rotamase activity that is inhibited by both FK506 and rapamycin with a Ki value of 30 nM and 0.9 nM, respectively, illustrating that VfFKBP15 binds rapamycin in preference over FK506. Tacrolimus 118-123 FKBP prolyl isomerase 2 Homo sapiens 29-35 8663352-4 1996 In yeast, the immunosuppressive drug FK506 inhibited Pdr5p, thereby potentiating activation of the glucocorticoid receptor by dexamethasone, a ligand that is exported by Pdr5p. Tacrolimus 37-42 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 53-58 8663352-4 1996 In yeast, the immunosuppressive drug FK506 inhibited Pdr5p, thereby potentiating activation of the glucocorticoid receptor by dexamethasone, a ligand that is exported by Pdr5p. Tacrolimus 37-42 nuclear receptor subfamily 3 group C member 1 Homo sapiens 99-122 8663352-4 1996 In yeast, the immunosuppressive drug FK506 inhibited Pdr5p, thereby potentiating activation of the glucocorticoid receptor by dexamethasone, a ligand that is exported by Pdr5p. Tacrolimus 37-42 ATP-binding cassette multidrug transporter PDR5 Saccharomyces cerevisiae S288C 170-175 8752921-0 1996 Non-T cell-derived IL-4 plays an important role in IgE production induced by antigen resensitization and is resistant to FK506. Tacrolimus 121-126 interleukin 4 Mus musculus 19-23 8752921-4 1996 In contrast to its known potent immunosuppressive effects on T cells, FK506 only partially inhibited IL-4 produced by non-T cells; therefore, FK506 was found to be a useful tool for identifying a cell source of IL-4 production. Tacrolimus 70-75 interleukin 4 Mus musculus 101-105 8752921-4 1996 In contrast to its known potent immunosuppressive effects on T cells, FK506 only partially inhibited IL-4 produced by non-T cells; therefore, FK506 was found to be a useful tool for identifying a cell source of IL-4 production. Tacrolimus 142-147 interleukin 4 Mus musculus 211-215 8760046-3 1996 Of interest, FK-506 and RAP, but not CyA, are bound by the steroid receptor-associated FK-506-binding heat shock protein of 56 kDa, HSP56. Tacrolimus 13-19 FKBP prolyl isomerase 4 Homo sapiens 132-137 8757947-4 1996 We have found that CsA- and FK506-treated cells did not exhibit transcription of FasL mRNA after activation and were lacking functional FasL protein on their surface as determined by staining and the ability to induce apoptosis in Fas+ target cells. Tacrolimus 28-33 Fas ligand (TNF superfamily, member 6) Mus musculus 136-140 8757947-9 1996 We therefore conclude that CsA and FK506 block activation-induced apoptosis in T cell hybridomas predominantly by interfering with activation signals leading to FasL expression and, further, that the regulation of the expression of Fas and FasL on activated T cells is differentially controlled. Tacrolimus 35-40 Fas ligand (TNF superfamily, member 6) Mus musculus 161-165 8757947-9 1996 We therefore conclude that CsA and FK506 block activation-induced apoptosis in T cell hybridomas predominantly by interfering with activation signals leading to FasL expression and, further, that the regulation of the expression of Fas and FasL on activated T cells is differentially controlled. Tacrolimus 35-40 Fas ligand (TNF superfamily, member 6) Mus musculus 240-244 21136324-0 1996 Practical applications of time-averaged restrained molecular dynamics to ligand-receptor systems: FK506 bound to the Q50R,A95H,K98I triple mutant of FKBP-13. Tacrolimus 98-103 FKBP prolyl isomerase 2 Homo sapiens 149-156 21136324-2 1996 Practical suggestions are offered for performing TARMD calculations with ligand-receptor systems, and are illustrated for the complex of the immunosuppressant FK506 bound to Q50R,A95H,K98I triple mutant FKBP-13. Tacrolimus 159-164 FKBP prolyl isomerase 2 Homo sapiens 203-210 21136324-3 1996 The structure of (13)C-labeled FK506 bound to triple-mutant FKBP-13 was determined using a set of 87 NOE distance restraints derived from HSQC-NOESY experiments. Tacrolimus 31-36 FKBP prolyl isomerase 2 Homo sapiens 60-67 21136324-4 1996 TARMD was found to be superior to conventional simulated-annealing methods, and produced structures that were conformationally similar to FK506 bound to wild-type FKBP-12. Tacrolimus 138-143 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 163-170 8647935-0 1996 Immunosuppressant FK506 induces interleukin-6 production through the activation of transcription factor nuclear factor (NF)-kappa(B). Tacrolimus 18-23 interleukin 6 Homo sapiens 32-45 8965835-0 1996 Increased interferon-gamma mRNA expression following alloincompatible myoblast transplantation is inhibited by FK506. Tacrolimus 111-116 interferon gamma Mus musculus 10-26 8965835-5 1996 Short-term immunosuppressive treatment with FK506 inhibited the transcription of IFN-gamma mRNA compared with that in untreated mice. Tacrolimus 44-49 interferon gamma Mus musculus 81-90 8669110-7 1996 Noninhibitory doses of CsA (8 nM) or FK506 (0.2 nM) suppressed mitogen-induced IL-2 production by 60-80% when combined with a noninhibitory dose (25 nM) of Ro 31-8220, indicating the potent synergy between these agents. Tacrolimus 37-42 interleukin 2 Homo sapiens 79-83 8647935-0 1996 Immunosuppressant FK506 induces interleukin-6 production through the activation of transcription factor nuclear factor (NF)-kappa(B). Tacrolimus 18-23 nuclear factor kappa B subunit 1 Homo sapiens 104-131 8647935-2 1996 FK506 is a powerful immunosuppressive drug currently in use that inhibits the activation of several transcription factors (nuclear factor (NF)-AT and NF-kappaB) critical for T cell activation. Tacrolimus 0-5 nuclear factor kappa B subunit 1 Homo sapiens 150-159 8647935-3 1996 We show here that, contrary to the situation in T cells, FK506 activates transcription factor NF-kappaB in nonlymphoid cells such as fibroblasts and renal mesangial cells. Tacrolimus 57-62 nuclear factor kappa B subunit 1 Homo sapiens 94-103 8647935-4 1996 We further show that FK506 induces NF-kappaB-regulated IL-6 production in vitro and in vivo, in particular in kidney. Tacrolimus 21-26 nuclear factor kappa B subunit 1 Homo sapiens 35-44 8647935-4 1996 We further show that FK506 induces NF-kappaB-regulated IL-6 production in vitro and in vivo, in particular in kidney. Tacrolimus 21-26 interleukin 6 Homo sapiens 55-59 8647935-7 1996 These results thus suggest a causal relationship between FK506-induced NF-kappaB activation/IL-6 production and some of FK506-induced renal abnormalities. Tacrolimus 57-62 nuclear factor kappa B subunit 1 Homo sapiens 71-80 8647935-7 1996 These results thus suggest a causal relationship between FK506-induced NF-kappaB activation/IL-6 production and some of FK506-induced renal abnormalities. Tacrolimus 57-62 interleukin 6 Homo sapiens 92-96 8647935-7 1996 These results thus suggest a causal relationship between FK506-induced NF-kappaB activation/IL-6 production and some of FK506-induced renal abnormalities. Tacrolimus 120-125 nuclear factor kappa B subunit 1 Homo sapiens 71-80 8647935-7 1996 These results thus suggest a causal relationship between FK506-induced NF-kappaB activation/IL-6 production and some of FK506-induced renal abnormalities. Tacrolimus 120-125 interleukin 6 Homo sapiens 92-96 8642394-7 1996 When the monkeys were immunosuppressed with FK506, muscle fibers expressing beta-galactosidase (beta-gal) were present 1, 4 and 12 weeks after the transplantation. Tacrolimus 44-49 galactosidase beta 1 Homo sapiens 76-94 8642394-7 1996 When the monkeys were immunosuppressed with FK506, muscle fibers expressing beta-galactosidase (beta-gal) were present 1, 4 and 12 weeks after the transplantation. Tacrolimus 44-49 galactosidase beta 1 Homo sapiens 76-84 8843591-6 1996 The doses of tacrolimus in unmatched cases tended to be larger than those in matched cases for every locus except for DQA1. Tacrolimus 13-23 major histocompatibility complex, class II, DQ alpha 1 Homo sapiens 118-122 8843593-0 1996 Inhibition by CsA and FK506 of the in vitro proliferative response of gamma delta T cells on stimulation with anti-TCR delta monoclonal antibody. Tacrolimus 22-27 T cell receptor delta chain Mus musculus 115-124 8843593-4 1996 Using spleen cells from Tg.Tlaa-3-1 mice, we showed that cyclosporin A (CsA) and FK506 inhibited the in vitro proliferative response of gamma delta T cells on stimulation with anti-TCR delta mAb. Tacrolimus 81-86 T cell receptor delta chain Mus musculus 181-190 8843593-5 1996 The dose-dependent inhibitory effect of CsA and FK506 on proliferation of gamma delta T cells on stimulation with anti-TCR delta mAb was similar to that on proliferation of alpha beta T cells on stimulation with anti-TCF beta mAb. Tacrolimus 48-53 T cell receptor delta chain Mus musculus 119-128 8843593-7 1996 The proliferative response of gamma delta T cells among spleen cells from TCR alpha beta-depleted Tg.Tlaa-3-1 mice was also inhibited by CsA and FK506, suggesting that the inhibition directly affected gamma delta T cells without mediation by alpha beta T cells. Tacrolimus 145-150 T cell receptor alpha chain Mus musculus 74-83 8787547-11 1996 Together with earlier results showing downmodulation for IL-8 receptor type A expression in cultured KC treated with FK506, these results suggest that both the mitogenic IL-8/IL-8R system and the cell cycle inhibitor p53 represent potential targets for the antipsoriatic action of the drug, whereas protooncogenes acting downstream in mitogenic signal transduction cascades are unaffected. Tacrolimus 117-122 C-X-C motif chemokine ligand 8 Homo sapiens 57-61 8787547-11 1996 Together with earlier results showing downmodulation for IL-8 receptor type A expression in cultured KC treated with FK506, these results suggest that both the mitogenic IL-8/IL-8R system and the cell cycle inhibitor p53 represent potential targets for the antipsoriatic action of the drug, whereas protooncogenes acting downstream in mitogenic signal transduction cascades are unaffected. Tacrolimus 117-122 C-X-C motif chemokine ligand 8 Homo sapiens 170-174 8787547-0 1996 Antioncogene P53 and mitogenic cytokine interleukin-8 aberrantly expressed in psoriatic skin are inversely regulated by the antipsoriatic drug tacrolimus (FK506). Tacrolimus 143-153 tumor protein p53 Homo sapiens 13-16 8787547-11 1996 Together with earlier results showing downmodulation for IL-8 receptor type A expression in cultured KC treated with FK506, these results suggest that both the mitogenic IL-8/IL-8R system and the cell cycle inhibitor p53 represent potential targets for the antipsoriatic action of the drug, whereas protooncogenes acting downstream in mitogenic signal transduction cascades are unaffected. Tacrolimus 117-122 tumor protein p53 Homo sapiens 217-220 8787547-0 1996 Antioncogene P53 and mitogenic cytokine interleukin-8 aberrantly expressed in psoriatic skin are inversely regulated by the antipsoriatic drug tacrolimus (FK506). Tacrolimus 143-153 C-X-C motif chemokine ligand 8 Homo sapiens 40-53 8787547-0 1996 Antioncogene P53 and mitogenic cytokine interleukin-8 aberrantly expressed in psoriatic skin are inversely regulated by the antipsoriatic drug tacrolimus (FK506). Tacrolimus 155-160 tumor protein p53 Homo sapiens 13-16 8787547-0 1996 Antioncogene P53 and mitogenic cytokine interleukin-8 aberrantly expressed in psoriatic skin are inversely regulated by the antipsoriatic drug tacrolimus (FK506). Tacrolimus 155-160 C-X-C motif chemokine ligand 8 Homo sapiens 40-53 8787547-6 1996 After 1-3 hr, IL-8 mRNA levels were dose-dependently decreased in tacrolimus (FK506)-treated cells. Tacrolimus 66-76 C-X-C motif chemokine ligand 8 Homo sapiens 14-18 8787547-6 1996 After 1-3 hr, IL-8 mRNA levels were dose-dependently decreased in tacrolimus (FK506)-treated cells. Tacrolimus 78-83 C-X-C motif chemokine ligand 8 Homo sapiens 14-18 8787547-8 1996 Interestingly, p53 transcription was clearly induced by FK506 treatment. Tacrolimus 56-61 tumor protein p53 Homo sapiens 15-18 8665940-7 1996 Low concentrations (2 nM) of okadaic acid, a serine/threonine phosphatase inhibitor, prevented TNF-alpha-induced inhibition of MAPK and restored insulin"s effect on MAPK activity, while orthovanadate (a tyrosine phosphatase inhibitor), inhibitor 2 (phosphatase-1 inhibitor) and FK506 (phosphatase-2B inhibitor) were ineffective. Tacrolimus 278-283 tumor necrosis factor Rattus norvegicus 95-104 8784785-1 1996 BACKGROUND: The immunophilins are proteins that mediate actions of immunosuppressant drugs such as FK506 and cyclosporin A by binding to calcineurin, inhibiting its phosphatase activity, and increasing the phosphorylation level of transcription factors required for interleukin 2 formation. Tacrolimus 99-104 interleukin 2 Rattus norvegicus 266-279 8628289-2 1996 Previous work showed that Ca2+ tolerance can be restored to pmc1 mutants by inactivation of calcineurin, a Ca2+/calmodulin-dependent protein phosphatase sensitive to the immunosuppressive drug FK506. Tacrolimus 193-198 calcium-transporting ATPase PMC1 Saccharomyces cerevisiae S288C 60-64 8784785-13 1996 Synapsin I, a synaptic vesicle phosphoprotein, displays enhanced phosphorylation in the presence of FK506. Tacrolimus 100-105 synapsin I Rattus norvegicus 0-10 8894806-9 1996 Both CSA and M17 synergized more strongly with FK 506 than they did between themselves. Tacrolimus 47-53 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 5-8 8627611-3 1996 Analyses of several X-ray crystal structures of FKBP12-urea complexes demonstrate that the urea-containing inhibitors associate with FKBP12 in a manner that is similar to, but significantly different from, that observed for the natural product FK506. Tacrolimus 244-249 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 48-54 8627611-3 1996 Analyses of several X-ray crystal structures of FKBP12-urea complexes demonstrate that the urea-containing inhibitors associate with FKBP12 in a manner that is similar to, but significantly different from, that observed for the natural product FK506. Tacrolimus 244-249 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 133-139 21232286-5 1996 The immunosuppressant drugs FK506 and rapamycin inhibit the prolyl isomerase activity of FKBP12 and could cause cardiac dysfunction by inducing a Ca(2+) leak from the sarcoplasmic reticulum. Tacrolimus 28-33 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 89-95 8623224-0 1996 The inhibition of T-cell-receptor-induced Fas ligand upregulation by cyclosporine and FK 506. Tacrolimus 86-92 Fas ligand Homo sapiens 42-52 8812665-0 1996 Immunophilin Modulation of Calcium Channel Gating The FK506 binding protein (FKBP12) is the cytosolic receptor for the immunosuppressant drugs FK506 and rapamycin. Tacrolimus 54-59 peptidylprolyl isomerase A (cyclophilin A) L homeolog Xenopus laevis 0-12 8812665-0 1996 Immunophilin Modulation of Calcium Channel Gating The FK506 binding protein (FKBP12) is the cytosolic receptor for the immunosuppressant drugs FK506 and rapamycin. Tacrolimus 54-59 FKBP prolyl isomerase 1A L homeolog Xenopus laevis 77-83 8812665-4 1996 These effects were reversed by adding FK506 or rapamycin, both of which inhibit FKBP12 isomerase activity and dissociate the FKBP-RyR complex. Tacrolimus 38-43 FKBP prolyl isomerase 1A L homeolog Xenopus laevis 80-86 8623162-1 1996 FK506 blocks T cell activation by preventing the transcription of lymphokine genes through binding to the intracellular protein FKBP12 and formation of complex that inhibits the phosphatase activity of calcineurin. Tacrolimus 0-5 FKBP prolyl isomerase 1A Rattus norvegicus 128-134 8762016-6 1996 Tacrolimus significantly increased NAG (N-acetyl-beta-glucosaminidase) excretion and associated histological damage, finally decreasing creatinine clearance. Tacrolimus 0-10 O-GlcNAcase Rattus norvegicus 35-38 8785272-1 1996 The FK506-binding protein (FKBP12) is important in the immunosuppressant action of FK506 and rapamycin. Tacrolimus 4-9 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 27-33 8785272-5 1996 Results obtained for the FKBP12/rapamycin complex are similar to those found for the FKBP12/FK506 complex. Tacrolimus 92-97 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 85-91 8786538-1 1996 FK-506 blocks T cell activation by preventing lymphokine gene transcription through formation of a complex with FKBP12 that inhibits calcineurin phosphatase activity. Tacrolimus 0-6 interleukin 2 Homo sapiens 46-56 8786538-1 1996 FK-506 blocks T cell activation by preventing lymphokine gene transcription through formation of a complex with FKBP12 that inhibits calcineurin phosphatase activity. Tacrolimus 0-6 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 112-118 8603582-0 1996 Immunosuppressant agent FK506 stimulates growth hormone (GH) secretion and gene expression of hypothalamic GH-releasing hormone in the rat. Tacrolimus 24-29 gonadotropin releasing hormone receptor Rattus norvegicus 41-55 8603582-0 1996 Immunosuppressant agent FK506 stimulates growth hormone (GH) secretion and gene expression of hypothalamic GH-releasing hormone in the rat. Tacrolimus 24-29 gonadotropin releasing hormone receptor Rattus norvegicus 57-59 8603582-0 1996 Immunosuppressant agent FK506 stimulates growth hormone (GH) secretion and gene expression of hypothalamic GH-releasing hormone in the rat. Tacrolimus 24-29 gonadotropin releasing hormone receptor Rattus norvegicus 107-109 8603582-6 1996 The FK506 significantly stimulated GH release from the rat anterior pituitary cells in a dose-dependent manner in vitro. Tacrolimus 4-9 gonadotropin releasing hormone receptor Rattus norvegicus 35-37 8603582-7 1996 In in vivo experiments, the area under the curve of GH surges was significantly increased in FK506-treated rats, although the peak height and the trough level of GH surges were not altered. Tacrolimus 93-98 gonadotropin releasing hormone receptor Rattus norvegicus 52-54 8603582-7 1996 In in vivo experiments, the area under the curve of GH surges was significantly increased in FK506-treated rats, although the peak height and the trough level of GH surges were not altered. Tacrolimus 93-98 gonadotropin releasing hormone receptor Rattus norvegicus 162-164 8603582-8 1996 Pituitary GH messenger RNA (mRNA) levels were significantly increased by the FK506 treatment. Tacrolimus 77-82 gonadotropin releasing hormone receptor Rattus norvegicus 10-12 8603582-10 1996 These findings indicate that FK506 stimulates GH secretion and gene expression of hypothalamic GRH in the rat. Tacrolimus 29-34 gonadotropin releasing hormone receptor Rattus norvegicus 46-48 8626235-2 1996 The antascomicins bind strongly to the FK506-binding protein FKBP12 and antagonize the immunosuppressive activity of FK506 and rapamycin. Tacrolimus 39-44 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 61-67 8762016-6 1996 Tacrolimus significantly increased NAG (N-acetyl-beta-glucosaminidase) excretion and associated histological damage, finally decreasing creatinine clearance. Tacrolimus 0-10 O-GlcNAcase Rattus norvegicus 40-69 8626756-4 1996 MRP and Mdr3 expression produced pleiotropic effects on drug resistance in this mutant, as corresponding VASY2563 transformants also acquired resistance to the anti-fungal agent FK506 and to the K+/H+ ionophore valinomycin. Tacrolimus 178-183 ATP binding cassette subfamily C member 3 Homo sapiens 0-3 8599949-6 1996 The rapamycin-FK506 binding protein complex is the effector of the inhibition of 4E-BP1 phosphorylation as excess of FK506 over rapamycin reversed the rapamycin-mediated inhibition of 4E-BP1 phosphorylation. Tacrolimus 14-19 eukaryotic translation initiation factor 4E binding protein 1 Homo sapiens 81-87 8599949-6 1996 The rapamycin-FK506 binding protein complex is the effector of the inhibition of 4E-BP1 phosphorylation as excess of FK506 over rapamycin reversed the rapamycin-mediated inhibition of 4E-BP1 phosphorylation. Tacrolimus 14-19 eukaryotic translation initiation factor 4E binding protein 1 Homo sapiens 184-190 8599949-6 1996 The rapamycin-FK506 binding protein complex is the effector of the inhibition of 4E-BP1 phosphorylation as excess of FK506 over rapamycin reversed the rapamycin-mediated inhibition of 4E-BP1 phosphorylation. Tacrolimus 117-122 eukaryotic translation initiation factor 4E binding protein 1 Homo sapiens 81-87 8599949-6 1996 The rapamycin-FK506 binding protein complex is the effector of the inhibition of 4E-BP1 phosphorylation as excess of FK506 over rapamycin reversed the rapamycin-mediated inhibition of 4E-BP1 phosphorylation. Tacrolimus 117-122 eukaryotic translation initiation factor 4E binding protein 1 Homo sapiens 184-190 8680047-15 1996 Tacrolimus binds to FK binding protein-12 (FKBP-12) to create a complex that inhibits CN. Tacrolimus 0-10 FKBP prolyl isomerase 1A Homo sapiens 20-41 8617295-7 1996 B7-1 stimulation is not mediated by the enhancement of IL-2 production: B7-1 enhancement of IL-2R alpha expression was FK506 resistant, while the inclusion of FK506 abrogated IL-2 production of the Th1 cells. Tacrolimus 119-124 CD80 antigen Mus musculus 72-76 8680047-15 1996 Tacrolimus binds to FK binding protein-12 (FKBP-12) to create a complex that inhibits CN. Tacrolimus 0-10 FKBP prolyl isomerase 1A Homo sapiens 43-50 8617295-7 1996 B7-1 stimulation is not mediated by the enhancement of IL-2 production: B7-1 enhancement of IL-2R alpha expression was FK506 resistant, while the inclusion of FK506 abrogated IL-2 production of the Th1 cells. Tacrolimus 119-124 interleukin 2 receptor, alpha chain Mus musculus 92-103 8617295-7 1996 B7-1 stimulation is not mediated by the enhancement of IL-2 production: B7-1 enhancement of IL-2R alpha expression was FK506 resistant, while the inclusion of FK506 abrogated IL-2 production of the Th1 cells. Tacrolimus 119-124 interleukin 2 Mus musculus 92-96 8617295-7 1996 B7-1 stimulation is not mediated by the enhancement of IL-2 production: B7-1 enhancement of IL-2R alpha expression was FK506 resistant, while the inclusion of FK506 abrogated IL-2 production of the Th1 cells. Tacrolimus 159-164 CD80 antigen Mus musculus 0-4 8617295-7 1996 B7-1 stimulation is not mediated by the enhancement of IL-2 production: B7-1 enhancement of IL-2R alpha expression was FK506 resistant, while the inclusion of FK506 abrogated IL-2 production of the Th1 cells. Tacrolimus 159-164 negative elongation factor complex member C/D, Th1l Mus musculus 198-201 8566225-3 1996 Accumulation of Ca2+ within the cls2 cells is synergistically elevated by the addition of immunosuppressant, FK506. Tacrolimus 109-114 mannosylinositol phosphorylceramide synthase regulatory subunit Saccharomyces cerevisiae S288C 32-36 8627154-6 1996 CNA alpha -/- T cells remained sensitive to both cyclosporin A and FK506, suggesting that CNA beta or another CNA-like molecule can mediate the action of these immunosuppressive drugs. Tacrolimus 67-72 protein phosphatase 3, catalytic subunit, alpha isoform Mus musculus 0-3 8627154-7 1996 CNA alpha -/- mice provide an animal model for dissecting the physiologic functions of calcineurin as well as the effects of FK506 and CsA. Tacrolimus 125-130 protein phosphatase 3, catalytic subunit, alpha isoform Mus musculus 0-3 8611359-3 1996 Although insulin requirement with tacrolimus therapy has been occasionally reported in adolescent patients post liver transplant, only a single case in a pediatric kidney transplant recipient has been previously documented. Tacrolimus 34-44 insulin Homo sapiens 9-16 9005438-1 1996 The 12 kDa FK506-binding protein FKBP12 is a cis-trans peptidyl-prolyl isomerase that binds the macrolides FK506 and rapamycin. Tacrolimus 11-16 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 33-39 9005438-3 1996 For each mutant FKBP12, we measured the affinity for FK506 and rapamycin and the catalytic efficiency in the cis-frans peptidyl-prolyl isomerase reaction. Tacrolimus 53-58 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 16-22 9005438-4 1996 The mutation of Trp59 or Phe99 generates an FKBP12 with a significantly lower affinity for FK506 than wild-type protein. Tacrolimus 91-96 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 44-50 8566225-4 1996 Moreover, in the vma3 background, toxicity caused by the cls2 mutation is greatly enhanced by FK506. Tacrolimus 94-99 H(+)-transporting V0 sector ATPase subunit c Saccharomyces cerevisiae S288C 17-21 8566225-4 1996 Moreover, in the vma3 background, toxicity caused by the cls2 mutation is greatly enhanced by FK506. Tacrolimus 94-99 mannosylinositol phosphorylceramide synthase regulatory subunit Saccharomyces cerevisiae S288C 57-61 8566225-5 1996 Given that FK506 inhibits the calcineurin activity, Cls2p likely functions in releasing Ca2+ flux from the endoplasmic reticulum, somehow cooperating with calcineurin. Tacrolimus 11-16 mannosylinositol phosphorylceramide synthase regulatory subunit Saccharomyces cerevisiae S288C 52-57 8576111-3 1996 Inhibition of calcineurin by the immunosuppressive drugs cyclosporin A and FK506 prevents dephosphorylation of NFATp and its translocation to the nucleus. Tacrolimus 75-80 nuclear factor of activated T cells 2 Homo sapiens 111-116 8576111-9 1996 Furthermore, the binding of both forms of NFATp to calcineurin was inhibited by pretreatment of calcineurin with a complex of FK506 and its ligand FKBP12. Tacrolimus 126-131 nuclear factor of activated T cells 2 Homo sapiens 42-47 8576111-9 1996 Furthermore, the binding of both forms of NFATp to calcineurin was inhibited by pretreatment of calcineurin with a complex of FK506 and its ligand FKBP12. Tacrolimus 126-131 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 147-153 8717522-3 1996 The FKBP12.FK506 complex specifically binds to and inhibits calcineurin, a signaling protein required for transcriptional activation of the interleukin (IL)-2 gene in response to T cell antigen receptor engagement. Tacrolimus 11-16 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 4-10 8717522-3 1996 The FKBP12.FK506 complex specifically binds to and inhibits calcineurin, a signaling protein required for transcriptional activation of the interleukin (IL)-2 gene in response to T cell antigen receptor engagement. Tacrolimus 11-16 interleukin 2 Homo sapiens 140-158 8822088-5 1996 FK506 treatment suppressed the accumulation of ED-1-positive cells, CD4-positive cells, CD8-positive cells, interleukin-2 (IL-2)-receptor-positive cells, leukocyte-function-associated antigen-1 (LFA-1)-positive cells and intercellular adhesion molecule-1 (ICAM-1)-expression in nephritic glomeruli. Tacrolimus 0-5 Cd4 molecule Rattus norvegicus 68-71 8822088-5 1996 FK506 treatment suppressed the accumulation of ED-1-positive cells, CD4-positive cells, CD8-positive cells, interleukin-2 (IL-2)-receptor-positive cells, leukocyte-function-associated antigen-1 (LFA-1)-positive cells and intercellular adhesion molecule-1 (ICAM-1)-expression in nephritic glomeruli. Tacrolimus 0-5 interleukin 2 Rattus norvegicus 123-127 8822088-5 1996 FK506 treatment suppressed the accumulation of ED-1-positive cells, CD4-positive cells, CD8-positive cells, interleukin-2 (IL-2)-receptor-positive cells, leukocyte-function-associated antigen-1 (LFA-1)-positive cells and intercellular adhesion molecule-1 (ICAM-1)-expression in nephritic glomeruli. Tacrolimus 0-5 interleukin 2 Rattus norvegicus 108-121 8822088-5 1996 FK506 treatment suppressed the accumulation of ED-1-positive cells, CD4-positive cells, CD8-positive cells, interleukin-2 (IL-2)-receptor-positive cells, leukocyte-function-associated antigen-1 (LFA-1)-positive cells and intercellular adhesion molecule-1 (ICAM-1)-expression in nephritic glomeruli. Tacrolimus 0-5 integrin subunit alpha L Rattus norvegicus 154-193 8822088-5 1996 FK506 treatment suppressed the accumulation of ED-1-positive cells, CD4-positive cells, CD8-positive cells, interleukin-2 (IL-2)-receptor-positive cells, leukocyte-function-associated antigen-1 (LFA-1)-positive cells and intercellular adhesion molecule-1 (ICAM-1)-expression in nephritic glomeruli. Tacrolimus 0-5 integrin subunit alpha L Rattus norvegicus 195-200 8822088-5 1996 FK506 treatment suppressed the accumulation of ED-1-positive cells, CD4-positive cells, CD8-positive cells, interleukin-2 (IL-2)-receptor-positive cells, leukocyte-function-associated antigen-1 (LFA-1)-positive cells and intercellular adhesion molecule-1 (ICAM-1)-expression in nephritic glomeruli. Tacrolimus 0-5 intercellular adhesion molecule 1 Rattus norvegicus 221-254 8822088-5 1996 FK506 treatment suppressed the accumulation of ED-1-positive cells, CD4-positive cells, CD8-positive cells, interleukin-2 (IL-2)-receptor-positive cells, leukocyte-function-associated antigen-1 (LFA-1)-positive cells and intercellular adhesion molecule-1 (ICAM-1)-expression in nephritic glomeruli. Tacrolimus 0-5 intercellular adhesion molecule 1 Rattus norvegicus 256-262 8822088-7 1996 On the other hand, IL-2 production from the spleen cells isolated from nephritic rats treated with FK506 was lower than that in the nephritic control rats. Tacrolimus 99-104 interleukin 2 Rattus norvegicus 19-23 8822088-8 1996 These results suggest that FK506 is effective against crescentic-type anti-GBM nephritis and that the antinephritic mechanisms of FK506 is due to the inhibition of intraglomerular accumulation and activation of leukocytes through the suppression of ICAM-1 expression and IL-2 production. Tacrolimus 130-135 intercellular adhesion molecule 1 Rattus norvegicus 249-255 8822088-8 1996 These results suggest that FK506 is effective against crescentic-type anti-GBM nephritis and that the antinephritic mechanisms of FK506 is due to the inhibition of intraglomerular accumulation and activation of leukocytes through the suppression of ICAM-1 expression and IL-2 production. Tacrolimus 130-135 interleukin 2 Rattus norvegicus 271-275 8622563-7 1996 Nevertheless, if Dex and TGF-beta 1 were dosed together with one of these immunosuppressants, suppressions of histamine and ischemic edema were 53%, 45% (FK506), 45%, 49% (CsA), 44%, 48% (Rapa) and 39%, 51% (DSP), respectively. Tacrolimus 154-159 transforming growth factor, beta 1 Mus musculus 25-35 8890921-3 1996 It has been shown that glucocorticoids and immunosuppressive agents such as cyclosporin and FK-506 inhibit TNF-alpha generation by T-lymphocytes and monocytes. Tacrolimus 92-98 tumor necrosis factor Mus musculus 107-116 8597171-5 1995 Observations on the FK 506-induced release of endothelin-1 from isolated rat kidney mesangial cells suggest that this cell may be an important target associated with the nephrotoxic potential of the drug, and that this action may be mediated via the FKBP. Tacrolimus 20-26 endothelin 1 Rattus norvegicus 46-58 8805102-6 1996 Rapamycin depressed the production of IL-6 and TNF-alpha, and FK506 depressed the production of TNF-alpha. Tacrolimus 62-67 tumor necrosis factor Homo sapiens 96-105 8545894-0 1995 Inhibitory effect of plasma FKBP12 on immunosuppressive activity of FK506. Tacrolimus 68-73 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 28-34 8545894-1 1995 To evaluate the roles of extracellular FKBP12, we examined the effect of extracellular FKBP12 on the immunosuppressive activity of FK506 in vitro and clinically. Tacrolimus 131-136 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 87-93 8545894-2 1995 The ability of FK506 to suppress phytohemagglutinin-induced proliferative response of human peripheral blood mononuclear cells was inhibited in the presence of recombinant FKBP12 dose-dependently. Tacrolimus 15-20 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 172-178 8545894-3 1995 We measured plasma levels of FKBP12 using a newly developed enzyme-linked immunosorbent assay system in 34 patients receiving FK506 after liver transplantation. Tacrolimus 126-131 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 29-35 8545894-9 1995 These results suggest that the rapid increase in plasma levels of FKBP12 may contribute to the occurrence and progress of acute cellular rejection probably by inhibiting the immunosuppressive activity of FK506. Tacrolimus 204-209 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 66-72 8524402-0 1995 Crystal structures of human calcineurin and the human FKBP12-FK506-calcineurin complex. Tacrolimus 61-66 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 54-60 8524402-2 1995 CaN is the target of the immunosuppressive drugs cyclosporin A and FK506, which inhibit CaN after forming complexes with cytoplasmic binding proteins (cyclophilin and FKBP12, respectively). Tacrolimus 67-72 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 167-173 8524402-3 1995 We report here the crystal structures of full-length human CaN at 2.1 A resolution and of the complex of human CaN with FKBP12-FK506 at 3.5 A resolution. Tacrolimus 127-132 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 120-126 8524402-6 1995 In the FKBP12-FK506-CaN complex, the auto-inhibitory element is displaced from the active site. Tacrolimus 14-19 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 7-13 8524402-7 1995 The site of binding of FKBP12-FK506 appears to be shared by other non-competitive inhibitors of calcineurin, including a natural anchoring protein. Tacrolimus 30-35 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 23-29 8689938-0 1995 Metabolism of the immunosuppressant tacrolimus in the small intestine: cytochrome P450, drug interactions, and interindividual variability. Tacrolimus 36-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 8689938-5 1995 The cytochrome P450 (CYP) enzymes responsible for tacrolimus metabolism in small intestine were identified using specific CYP antibodies and inhibitors. Tacrolimus 50-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 8689938-5 1995 The cytochrome P450 (CYP) enzymes responsible for tacrolimus metabolism in small intestine were identified using specific CYP antibodies and inhibitors. Tacrolimus 50-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 8689938-5 1995 The cytochrome P450 (CYP) enzymes responsible for tacrolimus metabolism in small intestine were identified using specific CYP antibodies and inhibitors. Tacrolimus 50-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-125 8689938-9 1995 The metabolism of tacrolimus by human small intestine was inhibited by anti-CYP3A, troleandomycin, and erythromycin, indicating that, as in the liver, CYP3A enzymes are the major enzymes for tacrolimus metabolism in the human small intestine. Tacrolimus 18-28 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 76-81 8689938-9 1995 The metabolism of tacrolimus by human small intestine was inhibited by anti-CYP3A, troleandomycin, and erythromycin, indicating that, as in the liver, CYP3A enzymes are the major enzymes for tacrolimus metabolism in the human small intestine. Tacrolimus 18-28 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 151-156 8689938-9 1995 The metabolism of tacrolimus by human small intestine was inhibited by anti-CYP3A, troleandomycin, and erythromycin, indicating that, as in the liver, CYP3A enzymes are the major enzymes for tacrolimus metabolism in the human small intestine. Tacrolimus 191-201 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 76-81 8689938-9 1995 The metabolism of tacrolimus by human small intestine was inhibited by anti-CYP3A, troleandomycin, and erythromycin, indicating that, as in the liver, CYP3A enzymes are the major enzymes for tacrolimus metabolism in the human small intestine. Tacrolimus 191-201 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 151-156 8689938-13 1995 It is concluded that tacrolimus is metabolized by cytochrome CYP3A enzymes in the small intestine. Tacrolimus 21-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 61-66 8689938-14 1995 The rate of the CYP3A enzymatic activities varies about 5 times from patient to patient, and drugs that interfere with the in vitro metabolism of tacrolimus in the liver also inhibit its small intestinal metabolism. Tacrolimus 146-156 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 16-21 8530625-0 1995 Expression of intercellular adhesion molecule-1 on human thyroid cells from patients with autoimmune thyroid disease: study of thyroid xenografts in nude and severe combined immunodeficient mice and treatment with FK-506. Tacrolimus 214-220 intercellular adhesion molecule 1 Homo sapiens 14-47 8530625-6 1995 When the SCID mice engrafted with AITD tissue were treated with the anti-CD4+ T (helper) cell agent FK-506, the expression of ICAM-1 was reduced significantly compared with that in the original tissue or that in nontreated mice engrafted with the same tissue. Tacrolimus 100-106 intercellular adhesion molecule 1 Mus musculus 126-132 8521476-1 1995 The immunosuppressant drug FK506 binds to the immunophilin protein FKBP12 and inhibits its prolyl isomerase activity. Tacrolimus 27-32 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 67-73 8521476-4 1995 FKBP12 is physically associated with the RyR and IP3R Ca2+ channels in the absence of FK506, with added FK506 disrupting these complexes. Tacrolimus 104-109 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 0-6 7592869-1 1995 FK506, an immunosuppressant that prolongs allograft survival, is a co-drug with its intracellular receptor, FKBP12. Tacrolimus 0-5 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 108-114 8521476-6 1995 We now report that calcineurin is physiologically associated with the IP3R-FKBP12 and RyR-FKBP12 receptor complexes and that this interaction can be disrupted by FK506 or rapamycin. Tacrolimus 162-167 inositol 1,4,5-trisphosphate receptor type 3 Homo sapiens 70-74 7592869-2 1995 The FKBP12.FK506 complex inhibits calcineurin, a critical signaling molecule during T-cell activation. Tacrolimus 11-16 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 4-10 7592869-3 1995 FKBP12 was, until recently, the sole FKBP known to mediate calcineurin inhibition at clinically relevant FK506 concentrations. Tacrolimus 105-110 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 0-6 7592869-5 1995 Recently, a novel protein, FKBP12.6, was found to inhibit calcineurin at clinically relevant FK506 concentrations. Tacrolimus 93-98 FKBP prolyl isomerase 1B Homo sapiens 27-35 8521476-6 1995 We now report that calcineurin is physiologically associated with the IP3R-FKBP12 and RyR-FKBP12 receptor complexes and that this interaction can be disrupted by FK506 or rapamycin. Tacrolimus 162-167 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 75-81 7592869-7 1995 In transfected Jurkat cells, FKBP12.6 is equivalent to FKBP12 at mediating the inhibitory effects of FK506. Tacrolimus 101-106 FKBP prolyl isomerase 1B Homo sapiens 29-37 8521476-6 1995 We now report that calcineurin is physiologically associated with the IP3R-FKBP12 and RyR-FKBP12 receptor complexes and that this interaction can be disrupted by FK506 or rapamycin. Tacrolimus 162-167 ryanodine receptor 2 Homo sapiens 86-89 7592869-7 1995 In transfected Jurkat cells, FKBP12.6 is equivalent to FKBP12 at mediating the inhibitory effects of FK506. Tacrolimus 101-106 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 29-35 7592869-10 1995 Our results suggest that FKBP12.6 has both a unique physiological role in excitation-contraction coupling in cardiac muscle and the potential to contribute to the immunosuppressive and toxic effects of FK506 and rapamycin. Tacrolimus 202-207 FKBP prolyl isomerase 1B Homo sapiens 25-33 8521476-6 1995 We now report that calcineurin is physiologically associated with the IP3R-FKBP12 and RyR-FKBP12 receptor complexes and that this interaction can be disrupted by FK506 or rapamycin. Tacrolimus 162-167 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 90-96 8576317-0 1995 Inhibitory effect of the immunosuppressant FK506 on apoptotic cell death induced by HIV-1 gp120. Tacrolimus 43-48 Envelope surface glycoprotein gp160, precursor Human immunodeficiency virus 1 90-95 8591053-4 1995 The target of the cyclophilin A-cyclosporine A and FKBP12-FK506 complexes is calcineurin, a protein phosphatase required for signaling via the T-cell receptor. Tacrolimus 58-63 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 51-57 8750397-3 1995 Cyclosporine and tacrolimus are metabolized by the cytochrome P-450 enzyme system. Tacrolimus 17-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 8582630-2 1995 A single nuclear mutation, designated cev1 for calcineurin essential for viability, is responsible for the CsA-FK506-sensitive phenotype. Tacrolimus 111-116 Vma22p Saccharomyces cerevisiae S288C 38-42 8582630-3 1995 The peptidyl-prolyl cis-trans isomerases cyclophilin A and FKBP12, respectively, mediate CsA and FK506 toxicity in the cev1 mutant strain. Tacrolimus 97-102 Vma22p Saccharomyces cerevisiae S288C 119-123 8576317-4 1995 Treatment of gp120-primed cells with an immunosuppressant (FK506) before TCR signaling inhibited apoptotic cell death, and this blocking effect of FK506 was concentration dependent. Tacrolimus 59-64 inter-alpha-trypsin inhibitor heavy chain 4 Homo sapiens 13-18 8576317-4 1995 Treatment of gp120-primed cells with an immunosuppressant (FK506) before TCR signaling inhibited apoptotic cell death, and this blocking effect of FK506 was concentration dependent. Tacrolimus 147-152 inter-alpha-trypsin inhibitor heavy chain 4 Homo sapiens 13-18 7559654-5 1995 p70 was purified to homogeneity; analysis of four tryptic peptides revealed that p70 is identical to the recently described FPR3 gene product, a nucleolarly localized proline rotamase of the FK506- and rapamycin-binding family. Tacrolimus 191-196 peptidylprolyl isomerase FPR3 Saccharomyces cerevisiae S288C 124-128 8584032-0 1995 Evidence that the FK506-binding immunophilin heat shock protein 56 is required for trafficking of the glucocorticoid receptor from the cytoplasm to the nucleus. Tacrolimus 18-23 FKBP prolyl isomerase 4 Homo sapiens 45-66 8584032-0 1995 Evidence that the FK506-binding immunophilin heat shock protein 56 is required for trafficking of the glucocorticoid receptor from the cytoplasm to the nucleus. Tacrolimus 18-23 nuclear receptor subfamily 3 group C member 1 Homo sapiens 102-125 8584032-1 1995 The FK506-binding immunophilin hsp56 (FKBP52) is one of several chaperone proteins associated with untrasformed steroid receptors in a multiprotein heterocomplex. Tacrolimus 4-9 FKBP prolyl isomerase 4 Homo sapiens 31-36 8584032-1 1995 The FK506-binding immunophilin hsp56 (FKBP52) is one of several chaperone proteins associated with untrasformed steroid receptors in a multiprotein heterocomplex. Tacrolimus 4-9 FKBP prolyl isomerase 4 Homo sapiens 38-44 8563622-0 1995 Structure comparison of native and mutant human recombinant FKBP12 complexes with the immunosuppressant drug FK506 (tacrolimus). Tacrolimus 109-114 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 60-66 8563622-0 1995 Structure comparison of native and mutant human recombinant FKBP12 complexes with the immunosuppressant drug FK506 (tacrolimus). Tacrolimus 116-126 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 60-66 8563622-2 1995 Here, we examine the effects of "conservative" and "nonconservative" substitutions on the X-ray crystal structures of human recombinant FKBP12 mutants in complex with the immunosuppressant drug FK506 (tacrolimus). Tacrolimus 194-199 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 136-142 8563622-2 1995 Here, we examine the effects of "conservative" and "nonconservative" substitutions on the X-ray crystal structures of human recombinant FKBP12 mutants in complex with the immunosuppressant drug FK506 (tacrolimus). Tacrolimus 201-211 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 136-142 7559604-7 1995 In vitro, both wild-type and CNB1-G2A mutant proteins formed complexes with both cyclophilin A-cyclosporin A (CsA) and FKBP12-FK506 that contained calcineurin A. Interestingly, expression of the nonmyristoylated CNB1-G2A mutant protein rendered yeast cells partially resistant to the immunosuppressant CsA, but not to FK506. Tacrolimus 126-131 calcineurin regulatory subunit B Saccharomyces cerevisiae S288C 29-33 7559604-7 1995 In vitro, both wild-type and CNB1-G2A mutant proteins formed complexes with both cyclophilin A-cyclosporin A (CsA) and FKBP12-FK506 that contained calcineurin A. Interestingly, expression of the nonmyristoylated CNB1-G2A mutant protein rendered yeast cells partially resistant to the immunosuppressant CsA, but not to FK506. Tacrolimus 318-323 calcineurin regulatory subunit B Saccharomyces cerevisiae S288C 29-33 8748119-6 1995 Despite the fact these drugs did not modulate the actions of glucocorticoids on corticotrope cells, both FK506 and CsA were potent stimulators of basal beta-endorphin secretion (4-6 fold) from rat anterior pituitary cultures and AtT20 cells. Tacrolimus 105-110 pro-opiomelanocortin-alpha Mus musculus 152-166 8748119-7 1995 In addition, FK506 and CsA potentiated beta-endorphin secretion induced by corticotropin releasing factor (CRF) and phorbol ester, but had no apparent acute (60 min) effect on POMC hnRNA levels. Tacrolimus 13-18 pro-opiomelanocortin-alpha Mus musculus 39-53 8748119-7 1995 In addition, FK506 and CsA potentiated beta-endorphin secretion induced by corticotropin releasing factor (CRF) and phorbol ester, but had no apparent acute (60 min) effect on POMC hnRNA levels. Tacrolimus 13-18 corticotropin releasing hormone Mus musculus 75-105 8748119-9 1995 Taken together these data suggest that FK506 and CsA do not alter GR activation or function in corticotrope cells, however, they are potent but short lived stimulators of POMC-derived peptide secretion. Tacrolimus 39-44 pro-opiomelanocortin-alpha Mus musculus 171-175 8748119-10 1995 The observation that CsA and FK506 stimulate POMC-derived peptide secretion, and potentiate both phorbol ester and CRF induced secretion, suggests that these immunosuppressant drugs are acting upon a common point within these intracellular pathways. Tacrolimus 29-34 pro-opiomelanocortin-alpha Mus musculus 45-49 9346613-5 1995 FK506 (0.5 ng/ mL) reduced both the percentage of interleukin-2 receptor expressing T cells and the cell surface density of this receptor by 7.1% and 8.7% (P < .01), whereas it only reduced the proportion of HLA-DR expressing T cells by 6.8%. Tacrolimus 0-5 interleukin 2 Homo sapiens 50-63 7549508-6 1995 Moreover, FK-506, mizoribine and cyclophosphamide clearly inhibited the antigen-induced IL-5 production and cyclosporin A showed the tendency to inhibit IL-5 production. Tacrolimus 10-16 interleukin 5 Mus musculus 88-92 7488022-5 1995 Both CsA and FK506 cause disruption of the CnA1 delta-AID interaction, whereas their presence permits CnA1 delta to bind more strongly to CnB. Tacrolimus 13-18 calcineurin catalytic subunit A Saccharomyces cerevisiae S288C 43-47 16535142-0 1995 Microbial Demethylation of Immunosuppressant FK-506: Isolation of 31-O-FK-506-Specific Demethylase Showing Cytochrome P-450 Characteristics from Streptomyces rimosus MA187. Tacrolimus 45-51 DF17_RS20860 Streptomyces rimosus 107-123 16535142-2 1995 Treatment of the biotransforming culture with FK-506 increased demethylase activity 2.4-fold and stabilized the cytochrome P-450 protein. Tacrolimus 46-52 DF17_RS20860 Streptomyces rimosus 112-128 16535142-6 1995 The isolated demethylase is therefore a cytochrome P-450 protein that can be used as a catalyst for the synthesis of 31-O-desmethylFK-506, an important immunosuppressant and a known metabolite of FK-506 metabolism by human liver microsomes. Tacrolimus 131-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 8557519-1 1995 We investigated the transcription and expression of the interleukin-2 receptor (IL-2R, CD25) in human T-lymphocytes after different modes of T-lymphocyte stimulation in the presence of the immunosuppressants cyclosporin (CsA) and tacrolimus (FK506) as well as the structurally related macrolide rapamycin. Tacrolimus 230-240 interleukin 2 receptor subunit alpha Homo sapiens 56-78 7575502-1 1995 The molecular complex formed by the immunosuppressant FK506 and the immunophilin protein FKBP12 potently inhibits the Ca2+/calmodulin-activated protein phosphatase calcineurin. Tacrolimus 54-59 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 89-95 8575571-6 1995 Interleukin-5 production by stimulated mononuclear cells was also inhibited both by FK506 and cyclosporin A. Tacrolimus 84-89 interleukin 5 Homo sapiens 0-13 7495746-7 1995 Cross-linking of TCR-CD3 together with CD4, CD8 or LFA-1 markedly inhibited glucocorticoid-induced death and the inhibition was canceled by FK506. Tacrolimus 140-145 CD4 molecule Homo sapiens 39-42 7495746-7 1995 Cross-linking of TCR-CD3 together with CD4, CD8 or LFA-1 markedly inhibited glucocorticoid-induced death and the inhibition was canceled by FK506. Tacrolimus 140-145 CD8a molecule Homo sapiens 44-47 7495746-7 1995 Cross-linking of TCR-CD3 together with CD4, CD8 or LFA-1 markedly inhibited glucocorticoid-induced death and the inhibition was canceled by FK506. Tacrolimus 140-145 integrin subunit alpha L Homo sapiens 51-56 7495746-8 1995 Furthermore, cross-linking of TCR-CD3 together with LFA-1 potentially induces both an apoptosis-inducing signal and an FK506-sensitive anti-apoptotic signal, and that the latter signal may be related to an essential signal for positive selection. Tacrolimus 119-124 integrin subunit alpha L Homo sapiens 52-57 8557519-2 1995 We demonstrate that CsA and FK506 inhibited IL-2R (CD25) gene transcription and protein expression after stimulation by anti-CD3 or ionomycin, but not by phorbol ester or IL-2. Tacrolimus 28-33 interleukin 2 receptor subunit alpha Homo sapiens 44-49 8557519-2 1995 We demonstrate that CsA and FK506 inhibited IL-2R (CD25) gene transcription and protein expression after stimulation by anti-CD3 or ionomycin, but not by phorbol ester or IL-2. Tacrolimus 28-33 interleukin 2 receptor subunit alpha Homo sapiens 51-55 7565718-9 1995 We have previously shown that growth of fks1 null mutants is highly sensitive to the calcineurin inhibitors FK506 and cyclosporin A. Tacrolimus 108-113 1,3-beta-D-glucan synthase Saccharomyces cerevisiae S288C 40-44 7565718-10 1995 Expression of FKS2 from the heterologous ADH1 promoter results in FK506-resistant growth. Tacrolimus 66-71 1,3-beta-glucan synthase GSC2 Saccharomyces cerevisiae S288C 14-18 7565718-10 1995 Expression of FKS2 from the heterologous ADH1 promoter results in FK506-resistant growth. Tacrolimus 66-71 alcohol dehydrogenase ADH1 Saccharomyces cerevisiae S288C 41-45 7545680-8 1995 Both of these events were blocked by preincubation of the cells with FK506, a calcineurin inhibitor, consistent with the hypothesis that NFATp is a calcineurin substrate in cells. Tacrolimus 69-74 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 2 Mus musculus 137-142 7545671-4 1995 The inhibitory effect of rapamycin on serum-induced p70s6k activation and the phosphorylation of Thr229, Thr389, Ser404, and Ser411 is rescued by FK506, providing further evidence that the inhibitory effect is exerted through a complex of rapamycin-FKBP12. Tacrolimus 146-151 ribosomal protein S6 kinase, polypeptide 1 Mus musculus 52-58 7545671-4 1995 The inhibitory effect of rapamycin on serum-induced p70s6k activation and the phosphorylation of Thr229, Thr389, Ser404, and Ser411 is rescued by FK506, providing further evidence that the inhibitory effect is exerted through a complex of rapamycin-FKBP12. Tacrolimus 146-151 FK506 binding protein 1a Mus musculus 249-255 8575571-0 1995 FK506 and cyclosporin A inhibit granulocyte/macrophage colony-stimulating factor production by mononuclear cells in asthma. Tacrolimus 0-5 colony stimulating factor 2 Homo sapiens 32-80 8575571-3 1995 To assess this, we compared the inhibitory effects of FK506 and cyclosporin A on production of granulocyte/macrophage colony-stimulating factor and interleukin-5 by interleukin-2- or Dermatophagoides farinae-stimulated mononuclear cells from patients with asthma, and their contribution to proliferation and survival of eosinophils in vitro. Tacrolimus 54-59 colony stimulating factor 2 Homo sapiens 95-143 8575571-3 1995 To assess this, we compared the inhibitory effects of FK506 and cyclosporin A on production of granulocyte/macrophage colony-stimulating factor and interleukin-5 by interleukin-2- or Dermatophagoides farinae-stimulated mononuclear cells from patients with asthma, and their contribution to proliferation and survival of eosinophils in vitro. Tacrolimus 54-59 interleukin 5 Homo sapiens 148-161 8575571-4 1995 FK506 inhibited granulocyte/macrophage colony-stimulating factor production by stimulated mononuclear cells from asthma patients at lower concentrations than cyclosporin A. Tacrolimus 0-5 colony stimulating factor 2 Homo sapiens 16-64 7495746-0 1995 Cross-linking of the TCR-CD3 complex with CD4, CD8 or LFA-1 induces an anti-apoptotic signal in thymocytes: the signal is canceled by FK506. Tacrolimus 134-139 CD4 molecule Homo sapiens 42-45 7495746-0 1995 Cross-linking of the TCR-CD3 complex with CD4, CD8 or LFA-1 induces an anti-apoptotic signal in thymocytes: the signal is canceled by FK506. Tacrolimus 134-139 CD8a molecule Homo sapiens 47-50 7495746-0 1995 Cross-linking of the TCR-CD3 complex with CD4, CD8 or LFA-1 induces an anti-apoptotic signal in thymocytes: the signal is canceled by FK506. Tacrolimus 134-139 integrin subunit alpha L Homo sapiens 54-59 7495746-3 1995 As positive selection depends on the inhibition of thymocyte apoptosis at its DP stage by signaling through the TCR-CD3 complex and some of the accessory molecules, including CD4, CD8 and LFA-1, we studied the possibility that FK506 enhanced apoptosis by itself or canceled the inhibition of apoptosis. Tacrolimus 227-232 CD4 molecule Homo sapiens 175-178 7495746-3 1995 As positive selection depends on the inhibition of thymocyte apoptosis at its DP stage by signaling through the TCR-CD3 complex and some of the accessory molecules, including CD4, CD8 and LFA-1, we studied the possibility that FK506 enhanced apoptosis by itself or canceled the inhibition of apoptosis. Tacrolimus 227-232 integrin subunit alpha L Homo sapiens 188-193 7495746-5 1995 On the other hand, upon cross-linking TCR-CD3 together with CD4, CD8 or LFA-1, FK506 markedly enhanced DNA fragmentation and cytolysis. Tacrolimus 79-84 CD4 molecule Homo sapiens 60-63 7495746-5 1995 On the other hand, upon cross-linking TCR-CD3 together with CD4, CD8 or LFA-1, FK506 markedly enhanced DNA fragmentation and cytolysis. Tacrolimus 79-84 CD8a molecule Homo sapiens 65-68 7495746-5 1995 On the other hand, upon cross-linking TCR-CD3 together with CD4, CD8 or LFA-1, FK506 markedly enhanced DNA fragmentation and cytolysis. Tacrolimus 79-84 integrin subunit alpha L Homo sapiens 72-77 8557519-2 1995 We demonstrate that CsA and FK506 inhibited IL-2R (CD25) gene transcription and protein expression after stimulation by anti-CD3 or ionomycin, but not by phorbol ester or IL-2. Tacrolimus 28-33 interleukin 2 Homo sapiens 44-48 8557519-1 1995 We investigated the transcription and expression of the interleukin-2 receptor (IL-2R, CD25) in human T-lymphocytes after different modes of T-lymphocyte stimulation in the presence of the immunosuppressants cyclosporin (CsA) and tacrolimus (FK506) as well as the structurally related macrolide rapamycin. Tacrolimus 242-247 interleukin 2 receptor subunit alpha Homo sapiens 56-78 8846199-6 1995 FK506 strongly suppressed the production of IL-2 (IC50 = 0.01 microM) and GM-CSF (IC50 = 0.03 microM), but was inactive (< 30% inhibition at 1 microM) against IL-5 and IFN-gamma. Tacrolimus 0-5 interleukin 2 Homo sapiens 44-48 7650004-7 1995 Additionally, thymic NFATc3 undergoes modifications in response to agents that mimic T cell receptor signaling, including a decrease in apparent molecular mass upon elevation of intracellular calcium that is inhibited by the immunosuppressant FK506. Tacrolimus 243-248 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 3 Mus musculus 21-27 8846199-6 1995 FK506 strongly suppressed the production of IL-2 (IC50 = 0.01 microM) and GM-CSF (IC50 = 0.03 microM), but was inactive (< 30% inhibition at 1 microM) against IL-5 and IFN-gamma. Tacrolimus 0-5 colony stimulating factor 2 Homo sapiens 74-80 8846199-6 1995 FK506 strongly suppressed the production of IL-2 (IC50 = 0.01 microM) and GM-CSF (IC50 = 0.03 microM), but was inactive (< 30% inhibition at 1 microM) against IL-5 and IFN-gamma. Tacrolimus 0-5 interferon gamma Homo sapiens 171-180 7657624-0 1995 The cyclosporin A-binding immunophilin CyP-40 and the FK506-binding immunophilin hsp56 bind to a common site on hsp90 and exist in independent cytosolic heterocomplexes with the untransformed glucocorticoid receptor. Tacrolimus 54-59 glucocorticoid receptor Oryctolagus cuniculus 192-215 7643878-7 1995 When such transplantations were done in mice immunosuppressed with cyclosporine or FK-506, normal dystrophin mRNA accounted for 31% and 36% of the total dystrophin mRNA, respectively. Tacrolimus 83-89 dystrophin, muscular dystrophy Mus musculus 98-108 7643878-8 1995 In fact, one animal immunosuppressed with FK-506 expressed as much as 57% of normal dystrophin mRNA. Tacrolimus 42-48 dystrophin, muscular dystrophy Mus musculus 84-94 7643878-9 1995 These results thus show that FK-506 makes it possible to restore dystrophin expression to a level comparable to that observed in DMD carriers that are usually asymptomatic. Tacrolimus 29-35 dystrophin, muscular dystrophy Mus musculus 65-75 7642551-6 1995 Mutations at FKBP12 residues Asp-37, Arg-42, His-87, and Ile-90 decrease calcineurin affinity of the mutant FKBP12.FK506 complex by as much as 2600-fold in the case of I90K. Tacrolimus 115-120 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 13-19 7642551-2 1995 The 12- and 13-kDa FK506 binding proteins (FKBP12 and FKBP13) are cis-trans peptidyl-prolyl isomerases that bind the macrolides FK506 (Tacrolimus) and rapamycin (Sirolimus). Tacrolimus 19-24 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 43-49 7642551-6 1995 Mutations at FKBP12 residues Asp-37, Arg-42, His-87, and Ile-90 decrease calcineurin affinity of the mutant FKBP12.FK506 complex by as much as 2600-fold in the case of I90K. Tacrolimus 115-120 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 108-114 7642551-2 1995 The 12- and 13-kDa FK506 binding proteins (FKBP12 and FKBP13) are cis-trans peptidyl-prolyl isomerases that bind the macrolides FK506 (Tacrolimus) and rapamycin (Sirolimus). Tacrolimus 19-24 FKBP prolyl isomerase 2 Homo sapiens 54-60 7543369-0 1995 X-ray structure of calcineurin inhibited by the immunophilin-immunosuppressant FKBP12-FK506 complex. Tacrolimus 86-91 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 79-85 7642551-7 1995 Replacement of three FKBP13 surface residues (Gln-50, Ala-95, and Lys-98) with the corresponding homologous FKBP12 residues (Arg-42, His-87, and Ile-90) generates an FKBP13 variant that is equivalent to FKBP12 in its affinity for FK506, rapamycin, and calcineurin. Tacrolimus 230-235 FKBP prolyl isomerase 2 Homo sapiens 21-27 7642551-2 1995 The 12- and 13-kDa FK506 binding proteins (FKBP12 and FKBP13) are cis-trans peptidyl-prolyl isomerases that bind the macrolides FK506 (Tacrolimus) and rapamycin (Sirolimus). Tacrolimus 128-133 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 43-49 7642551-2 1995 The 12- and 13-kDa FK506 binding proteins (FKBP12 and FKBP13) are cis-trans peptidyl-prolyl isomerases that bind the macrolides FK506 (Tacrolimus) and rapamycin (Sirolimus). Tacrolimus 128-133 FKBP prolyl isomerase 2 Homo sapiens 54-60 7642551-2 1995 The 12- and 13-kDa FK506 binding proteins (FKBP12 and FKBP13) are cis-trans peptidyl-prolyl isomerases that bind the macrolides FK506 (Tacrolimus) and rapamycin (Sirolimus). Tacrolimus 135-145 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 43-49 7642551-2 1995 The 12- and 13-kDa FK506 binding proteins (FKBP12 and FKBP13) are cis-trans peptidyl-prolyl isomerases that bind the macrolides FK506 (Tacrolimus) and rapamycin (Sirolimus). Tacrolimus 135-145 FKBP prolyl isomerase 2 Homo sapiens 54-60 7543369-1 1995 The X-ray structure of the ternary complex of a calcineurin A fragment, calcineurin B, FKBP12, and the immunosuppressant drug FK506 (also known as tacrolimus) has been determined at 2.5 A resolution, providing a description of how FK506 functions at the atomic level. Tacrolimus 126-131 protein phosphatase 3 regulatory subunit B, alpha Homo sapiens 72-85 7543369-1 1995 The X-ray structure of the ternary complex of a calcineurin A fragment, calcineurin B, FKBP12, and the immunosuppressant drug FK506 (also known as tacrolimus) has been determined at 2.5 A resolution, providing a description of how FK506 functions at the atomic level. Tacrolimus 126-131 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 87-93 7543369-1 1995 The X-ray structure of the ternary complex of a calcineurin A fragment, calcineurin B, FKBP12, and the immunosuppressant drug FK506 (also known as tacrolimus) has been determined at 2.5 A resolution, providing a description of how FK506 functions at the atomic level. Tacrolimus 147-157 protein phosphatase 3 regulatory subunit B, alpha Homo sapiens 72-85 7543369-1 1995 The X-ray structure of the ternary complex of a calcineurin A fragment, calcineurin B, FKBP12, and the immunosuppressant drug FK506 (also known as tacrolimus) has been determined at 2.5 A resolution, providing a description of how FK506 functions at the atomic level. Tacrolimus 147-157 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 87-93 7543369-1 1995 The X-ray structure of the ternary complex of a calcineurin A fragment, calcineurin B, FKBP12, and the immunosuppressant drug FK506 (also known as tacrolimus) has been determined at 2.5 A resolution, providing a description of how FK506 functions at the atomic level. Tacrolimus 231-236 protein phosphatase 3 regulatory subunit B, alpha Homo sapiens 72-85 7642551-7 1995 Replacement of three FKBP13 surface residues (Gln-50, Ala-95, and Lys-98) with the corresponding homologous FKBP12 residues (Arg-42, His-87, and Ile-90) generates an FKBP13 variant that is equivalent to FKBP12 in its affinity for FK506, rapamycin, and calcineurin. Tacrolimus 230-235 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 108-114 7543369-2 1995 In the structure, the FKBP12-FK506 binary complex does not contact the phosphatase active site on calcineurin A that is more than 10 A removed. Tacrolimus 29-34 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 22-28 7642551-7 1995 Replacement of three FKBP13 surface residues (Gln-50, Ala-95, and Lys-98) with the corresponding homologous FKBP12 residues (Arg-42, His-87, and Ile-90) generates an FKBP13 variant that is equivalent to FKBP12 in its affinity for FK506, rapamycin, and calcineurin. Tacrolimus 230-235 FKBP prolyl isomerase 2 Homo sapiens 166-172 7543369-3 1995 Instead, FKBP12-FK506 is so positioned that it can inhibit the dephosphorylation of its macromolecular substrates by physically hindering their approach to the active site. Tacrolimus 16-21 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 9-15 7543369-4 1995 The ternary complex described here represents the three-dimensional structure of a Ser/Thr protein phosphatase and provides a structural basis for understanding calcineurin inhibition by FKBP12-FK506. Tacrolimus 194-199 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 187-193 7642551-3 1995 The FKBP12.FK506 complex is immunosuppressive, acting as an inhibitor of the protein phosphatase calcineurin. Tacrolimus 11-16 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 4-10 7544285-3 1995 FKBP59 binds immunosuppressant FK506 and has peptidylprolyl cis-trans-isomerase activity, both properties being localized in the N-terminal domain (FKBP59-I). Tacrolimus 31-36 FKBP prolyl isomerase 4 Homo sapiens 0-6 7639278-1 1995 P-glycoprotein 170 (P-gp), the multidrug transport pump, excludes drugs from the interior of cells and is inhibited by agents such as cyclosporin A (CsA), verapamil, and FK-506, which are also substrates for the P-gp pump. Tacrolimus 170-176 phosphoglycolate phosphatase Homo sapiens 0-18 7639278-1 1995 P-glycoprotein 170 (P-gp), the multidrug transport pump, excludes drugs from the interior of cells and is inhibited by agents such as cyclosporin A (CsA), verapamil, and FK-506, which are also substrates for the P-gp pump. Tacrolimus 170-176 phosphoglycolate phosphatase Homo sapiens 20-24 7639278-1 1995 P-glycoprotein 170 (P-gp), the multidrug transport pump, excludes drugs from the interior of cells and is inhibited by agents such as cyclosporin A (CsA), verapamil, and FK-506, which are also substrates for the P-gp pump. Tacrolimus 170-176 phosphoglycolate phosphatase Homo sapiens 212-216 7583777-1 1995 The effects of cyclosporin A (CsA), FK506 and rapamycin (Rapa) on the intracellular localization of a mutated rabbit progesterone receptor (PR) which lacks the main constitutive nuclear localization signal (NLS) (delta 638-642) and is cytoplasmic in the absence of progesterone (Prog), were assayed by indirect immunofluorescence in Lcl3 cells, a mouse L-cell line stably expressing this mutant. Tacrolimus 36-41 progesterone receptor Oryctolagus cuniculus 117-138 7543492-5 1995 FK506 treatment significantly augmented thymic apoptosis induced by in vivo anti-CD3 Ab administration. Tacrolimus 0-5 CD3 antigen, epsilon polypeptide Mus musculus 81-84 7543492-6 1995 Increased thymic apoptosis resulted in the disappearance of CD4+ CD8+ thymocytes after anti-CD3 Ab/FK506 treatment. Tacrolimus 99-104 CD4 antigen Mus musculus 60-63 7543492-6 1995 Increased thymic apoptosis resulted in the disappearance of CD4+ CD8+ thymocytes after anti-CD3 Ab/FK506 treatment. Tacrolimus 99-104 CD3 antigen, epsilon polypeptide Mus musculus 92-95 7542743-7 1995 As judged from peptidyl prolyl isomerase activity, FKBP51 had a slightly higher affinity for rapamycin than for FK520, an FK506 analog. Tacrolimus 122-127 FK506 binding protein 5 Mus musculus 51-57 7542815-11 1995 FK506 decreased steady-state insulin secretion during the last 60 min of the clamp, regardless of initial glucose tolerance. Tacrolimus 0-5 insulin Homo sapiens 29-36 7623828-10 1995 Furthermore, the activating function of p95vav is blocked by FK506, suggesting that its activity also depends on calcineurin. Tacrolimus 61-66 vav 1 oncogene Mus musculus 40-46 7542741-6 1995 CND1 is identical to the gene identified previously as FKS1, ETG1, and CWH53, cnd1 mutants are sensitive to FK506 and cyclosporin A and exhibit slow growth that is improved by the addition of osmotic stabilizing agents. Tacrolimus 108-113 1,3-beta-D-glucan synthase Saccharomyces cerevisiae S288C 0-4 7542741-6 1995 CND1 is identical to the gene identified previously as FKS1, ETG1, and CWH53, cnd1 mutants are sensitive to FK506 and cyclosporin A and exhibit slow growth that is improved by the addition of osmotic stabilizing agents. Tacrolimus 108-113 1,3-beta-D-glucan synthase Saccharomyces cerevisiae S288C 55-59 7542741-6 1995 CND1 is identical to the gene identified previously as FKS1, ETG1, and CWH53, cnd1 mutants are sensitive to FK506 and cyclosporin A and exhibit slow growth that is improved by the addition of osmotic stabilizing agents. Tacrolimus 108-113 1,3-beta-D-glucan synthase Saccharomyces cerevisiae S288C 61-65 7542741-6 1995 CND1 is identical to the gene identified previously as FKS1, ETG1, and CWH53, cnd1 mutants are sensitive to FK506 and cyclosporin A and exhibit slow growth that is improved by the addition of osmotic stabilizing agents. Tacrolimus 108-113 1,3-beta-D-glucan synthase Saccharomyces cerevisiae S288C 71-76 7542741-6 1995 CND1 is identical to the gene identified previously as FKS1, ETG1, and CWH53, cnd1 mutants are sensitive to FK506 and cyclosporin A and exhibit slow growth that is improved by the addition of osmotic stabilizing agents. Tacrolimus 108-113 1,3-beta-D-glucan synthase Saccharomyces cerevisiae S288C 78-82 7541044-0 1995 Interaction of FKBP12-FK506 with calcineurin A at the B subunit-binding domain. Tacrolimus 22-27 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 15-21 7552894-2 1995 Tacrolimus prevents rejection of the transplanted organ by inhibiting the expression of interleukin-2 in T cells and inhibiting T-cell growth and proliferation. Tacrolimus 0-10 interleukin 2 Homo sapiens 88-101 7552894-4 1995 Tacrolimus is extensively metabolized by cytochrome P-450 3A4 isoenzyme, resulting in several known drug interactions. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-61 15299838-2 1995 FK506, in complex with its 12 kDa cytosolic receptor (FKBP12), is a potent agonist of immunosuppression through the inhibition of the phosphatase activity of calcineurin. Tacrolimus 0-5 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 54-60 15299838-3 1995 Rapamycin (sirolimus), which is itself an immunosuppressant by a different mechanism, completes with FK506 for binding to FKBP12 and thereby acts as an antagonist of calcineurin inhibition. Tacrolimus 101-106 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 122-128 15299838-4 1995 We have solved the X-ray structure of unliganded FKBP12 and of FKBP12 in complex with FK506 and with rapamycin; these structures show localized differences in conformation and mobility in those regions of the protein that are known, by site-directed mutagenesis, to be involved in calcineurin inhibition. Tacrolimus 86-91 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 49-55 15299838-4 1995 We have solved the X-ray structure of unliganded FKBP12 and of FKBP12 in complex with FK506 and with rapamycin; these structures show localized differences in conformation and mobility in those regions of the protein that are known, by site-directed mutagenesis, to be involved in calcineurin inhibition. Tacrolimus 86-91 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 63-69 15299839-0 1995 Design, synthesis and structure of non-macrocyclic inhibitors of FKBP12, the major binding protein for the immunosuppressant FK506. Tacrolimus 125-130 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 65-71 15299839-1 1995 We have synthesized a series of non-macrocyclic ligands to FKBP12 that are comparable in binding potency and peptidyl prolyl isomerase (PPIase) inhibition to FK506 itself. Tacrolimus 158-163 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 59-65 15299839-5 1995 In the FKBP12-FK506 complex, a significant portion of the FK506 ligand, its "effector domain", projects beyond the envelope of the binding protein in a manner that is suggestive of a potential interaction with a second protein, the calcium-dependent phosphatase, calcineurin, whose inhibition by the FKBP 12-FK506 complex interrupts the T-cell activation events leading to immunosuppression. Tacrolimus 14-19 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 7-13 15299839-5 1995 In the FKBP12-FK506 complex, a significant portion of the FK506 ligand, its "effector domain", projects beyond the envelope of the binding protein in a manner that is suggestive of a potential interaction with a second protein, the calcium-dependent phosphatase, calcineurin, whose inhibition by the FKBP 12-FK506 complex interrupts the T-cell activation events leading to immunosuppression. Tacrolimus 14-19 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 300-307 15299839-5 1995 In the FKBP12-FK506 complex, a significant portion of the FK506 ligand, its "effector domain", projects beyond the envelope of the binding protein in a manner that is suggestive of a potential interaction with a second protein, the calcium-dependent phosphatase, calcineurin, whose inhibition by the FKBP 12-FK506 complex interrupts the T-cell activation events leading to immunosuppression. Tacrolimus 58-63 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 7-13 15299839-5 1995 In the FKBP12-FK506 complex, a significant portion of the FK506 ligand, its "effector domain", projects beyond the envelope of the binding protein in a manner that is suggestive of a potential interaction with a second protein, the calcium-dependent phosphatase, calcineurin, whose inhibition by the FKBP 12-FK506 complex interrupts the T-cell activation events leading to immunosuppression. Tacrolimus 58-63 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 300-307 15299839-5 1995 In the FKBP12-FK506 complex, a significant portion of the FK506 ligand, its "effector domain", projects beyond the envelope of the binding protein in a manner that is suggestive of a potential interaction with a second protein, the calcium-dependent phosphatase, calcineurin, whose inhibition by the FKBP 12-FK506 complex interrupts the T-cell activation events leading to immunosuppression. Tacrolimus 58-63 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 7-13 15299839-5 1995 In the FKBP12-FK506 complex, a significant portion of the FK506 ligand, its "effector domain", projects beyond the envelope of the binding protein in a manner that is suggestive of a potential interaction with a second protein, the calcium-dependent phosphatase, calcineurin, whose inhibition by the FKBP 12-FK506 complex interrupts the T-cell activation events leading to immunosuppression. Tacrolimus 58-63 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 300-307 7476967-0 1995 Interaction of the progesterone receptor with binding proteins for FK506 and cyclosporin A. Tacrolimus 67-72 progesterone receptor Homo sapiens 19-40 7476967-5 1995 A third 59-kilodalton (kDa) protein observed previously was confirmed to be p59 (also called hsp56 or FKBP52), which has been shown to bind the immunosuppressant drug FK506. Tacrolimus 167-172 HLA complex P5B Homo sapiens 76-79 7476967-5 1995 A third 59-kilodalton (kDa) protein observed previously was confirmed to be p59 (also called hsp56 or FKBP52), which has been shown to bind the immunosuppressant drug FK506. Tacrolimus 167-172 FKBP prolyl isomerase 4 Homo sapiens 93-98 7476967-5 1995 A third 59-kilodalton (kDa) protein observed previously was confirmed to be p59 (also called hsp56 or FKBP52), which has been shown to bind the immunosuppressant drug FK506. Tacrolimus 167-172 FKBP prolyl isomerase 4 Homo sapiens 102-108 7541044-6 1995 Furthermore, calcineurin A subunit mutants of residues Thr351, Leu354, and Lys360 showed NF kappa B transactivation activity and phosphatase activity with increased resistance to FKBP12-FK506 but displayed no or minimal increase in resistance for cyclosporin A inhibition. Tacrolimus 186-191 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 179-185 7541044-7 1995 Together, these results strongly suggest that the B subunit-binding domain is required for calcineurin activity intracellulary and interacts with the FKBP12-FK506 complex. Tacrolimus 157-162 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 150-156 7539960-10 1995 Thus, as in the case with NF-AT in T cells, these findings point to the reduction of unidentified nuclear factors for insulin mRNA transcription caused by the binding of FK506 to FKBP-12 and a subsequent inhibition of calcineurin in the beta-cells. Tacrolimus 170-175 FKBP prolyl isomerase 1A Rattus norvegicus 179-186 7541038-4 1995 Cyclosporin A and FK506 efficiently disrupt the YY1-CyPA and YY1-FKBP12 interactions. Tacrolimus 18-23 YY1 transcription factor Homo sapiens 48-51 7541038-4 1995 Cyclosporin A and FK506 efficiently disrupt the YY1-CyPA and YY1-FKBP12 interactions. Tacrolimus 18-23 peptidylprolyl isomerase A Homo sapiens 52-56 7541038-4 1995 Cyclosporin A and FK506 efficiently disrupt the YY1-CyPA and YY1-FKBP12 interactions. Tacrolimus 18-23 YY1 transcription factor Homo sapiens 61-64 7541038-4 1995 Cyclosporin A and FK506 efficiently disrupt the YY1-CyPA and YY1-FKBP12 interactions. Tacrolimus 18-23 FKBP prolyl isomerase 1A Homo sapiens 65-71 7540976-4 1995 One mutation that confers dominant FK506 resistance alters a single residue (W430C) in the calcineurin A catalytic subunit CMP2. Tacrolimus 35-40 calcineurin catalytic subunit A Saccharomyces cerevisiae S288C 123-127 7759865-7 1995 p40cdk6 also increased in amount in cells activated in the presence of cyclosporin A or FK506, drugs that inhibit production of IL-2 and cell proliferation, suggesting that initial induction occurred independently of IL-2-mediated cell cycle progression. Tacrolimus 88-93 interleukin 2 Homo sapiens 128-132 7540976-6 1995 When introduced into the CMP1 subunit, the FK506 resistance mutation (W388C) blocks binding by FKBP12-FK506, but not by cyclophilin A-CsA. Tacrolimus 43-48 calcineurin catalytic subunit A Saccharomyces cerevisiae S288C 25-29 7759865-7 1995 p40cdk6 also increased in amount in cells activated in the presence of cyclosporin A or FK506, drugs that inhibit production of IL-2 and cell proliferation, suggesting that initial induction occurred independently of IL-2-mediated cell cycle progression. Tacrolimus 88-93 interleukin 2 Homo sapiens 217-221 7540976-6 1995 When introduced into the CMP1 subunit, the FK506 resistance mutation (W388C) blocks binding by FKBP12-FK506, but not by cyclophilin A-CsA. Tacrolimus 102-107 calcineurin catalytic subunit A Saccharomyces cerevisiae S288C 25-29 7540976-8 1995 Double mutant calcineurin A subunits (Y377F, W388C CMP1 and Y419F, W430C CMP2) confer resistance to CsA, to FK506 and to CsA plus FK506. Tacrolimus 108-113 calcineurin catalytic subunit A Saccharomyces cerevisiae S288C 73-77 7540976-8 1995 Double mutant calcineurin A subunits (Y377F, W388C CMP1 and Y419F, W430C CMP2) confer resistance to CsA, to FK506 and to CsA plus FK506. Tacrolimus 130-135 calcineurin catalytic subunit A Saccharomyces cerevisiae S288C 73-77 7544633-8 1995 Activities of myeloperoxidase and concentrations of 6-keto-prostaglandin F1 alpha (6-keto-PGF1 alpha), PGF2 alpha and PGE2 in colonic tissue increased significantly following induction of experimental colitis, however, FK506 did not affect these changes. Tacrolimus 219-224 myeloperoxidase Rattus norvegicus 14-29 7577807-0 1995 Suppression of adult T cell leukemia-derived factor/human thioredoxin induction by FK506 and cyclosporin A: a new mechanism of immune modulation via redox control. Tacrolimus 83-88 thioredoxin Homo sapiens 58-69 7538911-5 1995 These mutants were still fully sensitive to FK-506, an immunosuppressant structurally related to RAP whose mode of action also involves an interaction with FKBPs. Tacrolimus 44-50 regulatory associated protein of MTOR, complex 1 Mus musculus 97-100 7544064-7 1995 With decrease in the dose of FK506, the TL was normalized, and the dose of IS could be decreased. Tacrolimus 29-34 insulin Homo sapiens 75-77 7544064-8 1995 FK506 has been reported to inhibit IS secretion. Tacrolimus 0-5 insulin Homo sapiens 35-37 7577807-9 1995 These results suggest that ADF/TRX produced and released from PBMC may be a crucial event in lymphocyte activation, and that FK506 and CsA may exert the immune suppression partly through inhibiting the induction of the endogenous reducing factor ADF/TRX. Tacrolimus 125-130 thioredoxin Homo sapiens 246-249 7577807-4 1995 ADF/TRX induction in PBMC by PHA, Con A or OKT3 mAb was almost completely suppressed by FK506. Tacrolimus 88-93 thioredoxin Homo sapiens 0-3 7577807-9 1995 These results suggest that ADF/TRX produced and released from PBMC may be a crucial event in lymphocyte activation, and that FK506 and CsA may exert the immune suppression partly through inhibiting the induction of the endogenous reducing factor ADF/TRX. Tacrolimus 125-130 thioredoxin Homo sapiens 250-253 7577807-4 1995 ADF/TRX induction in PBMC by PHA, Con A or OKT3 mAb was almost completely suppressed by FK506. Tacrolimus 88-93 thioredoxin Homo sapiens 4-7 7577807-8 1995 Furthermore, when recombinant ADF (rADF) was added to a culture of PBMC 1 h before the addition of PHA and FK506, the action of FK506 was partially reversed as determined by [3H]thymidine incorporation and viable cell counts. Tacrolimus 107-112 thioredoxin Homo sapiens 30-33 7577807-8 1995 Furthermore, when recombinant ADF (rADF) was added to a culture of PBMC 1 h before the addition of PHA and FK506, the action of FK506 was partially reversed as determined by [3H]thymidine incorporation and viable cell counts. Tacrolimus 128-133 thioredoxin Homo sapiens 30-33 7538541-13 1995 Finally, immunosuppressant cyclosporin A (100 nM) and FK-506 (10 nM) significantly inhibited mast cell adhesion to both fibronectin and laminin (p < 0.05). Tacrolimus 54-60 fibronectin 1 Homo sapiens 120-131 7538591-0 1995 Determination of the differential effects of hydrogen bonding and water release on the binding of FK506 to native and Tyr82-->Phe82 FKBP-12 proteins using free energy simulations. Tacrolimus 98-103 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 135-142 7539555-9 1995 Cellular expression of IL-1 alpha, IL-2, IL-6, gamma-INF, FGF, and TGF-beta mRNA was suppressed with FK506 to below isograft levels, and clinical rejection was not observed with the doses, routes, and schedules used. Tacrolimus 101-106 interleukin 1 alpha Rattus norvegicus 23-33 7539555-9 1995 Cellular expression of IL-1 alpha, IL-2, IL-6, gamma-INF, FGF, and TGF-beta mRNA was suppressed with FK506 to below isograft levels, and clinical rejection was not observed with the doses, routes, and schedules used. Tacrolimus 101-106 interleukin 2 Rattus norvegicus 35-39 7539555-9 1995 Cellular expression of IL-1 alpha, IL-2, IL-6, gamma-INF, FGF, and TGF-beta mRNA was suppressed with FK506 to below isograft levels, and clinical rejection was not observed with the doses, routes, and schedules used. Tacrolimus 101-106 interleukin 6 Rattus norvegicus 41-45 7539555-9 1995 Cellular expression of IL-1 alpha, IL-2, IL-6, gamma-INF, FGF, and TGF-beta mRNA was suppressed with FK506 to below isograft levels, and clinical rejection was not observed with the doses, routes, and schedules used. Tacrolimus 101-106 transforming growth factor, beta 1 Rattus norvegicus 67-75 7538591-1 1995 We use the thermodynamic integration technique to calculate the free energy associated with the Tyr82-->Phe82 mutation (Y82F) in the protein FKBP-12, both free and bound to known inhibitor FK506 (tacrolimis). Tacrolimus 192-197 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 144-151 7545487-2 1995 FK506 which has a similar immunosuppressive mechanism to that of CsA also showed the same inhibitory effects except for decreased IL-5 and IL-6 mRNA expression. Tacrolimus 0-5 interleukin 5 Homo sapiens 130-134 7545487-2 1995 FK506 which has a similar immunosuppressive mechanism to that of CsA also showed the same inhibitory effects except for decreased IL-5 and IL-6 mRNA expression. Tacrolimus 0-5 interleukin 6 Homo sapiens 139-143 7545487-3 1995 In contrast, both CsA and FK506 enhanced transforming growth factor beta (TGF beta) and IL-1 beta mRNA expression. Tacrolimus 26-31 transforming growth factor beta 1 Homo sapiens 41-72 7545487-3 1995 In contrast, both CsA and FK506 enhanced transforming growth factor beta (TGF beta) and IL-1 beta mRNA expression. Tacrolimus 26-31 transforming growth factor beta 1 Homo sapiens 74-82 7545487-3 1995 In contrast, both CsA and FK506 enhanced transforming growth factor beta (TGF beta) and IL-1 beta mRNA expression. Tacrolimus 26-31 interleukin 1 beta Homo sapiens 88-97 7613145-3 1995 The immunosuppressive agents CsA and FK506 enhanced the release of leukotriene B4 (LTB4) from human neutrophil granulocytes dependent on the priming with G-CSF and IL-3. Tacrolimus 37-42 colony stimulating factor 3 Homo sapiens 154-159 7537219-5 1995 The amino acids in human FKBP12 which are proposed to be important for FK506 interaction are conserved in the schistosome protein. Tacrolimus 71-76 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 25-31 7542077-6 1995 FK506 suppressed IL-5 production and gene expression in vitro in a dose-dependent manner. Tacrolimus 0-5 interleukin 5 Homo sapiens 17-21 7613145-3 1995 The immunosuppressive agents CsA and FK506 enhanced the release of leukotriene B4 (LTB4) from human neutrophil granulocytes dependent on the priming with G-CSF and IL-3. Tacrolimus 37-42 interleukin 3 Homo sapiens 164-168 7613145-4 1995 IL-3-primed neutrophils released up to threefold higher amounts of LTB4 after subsequent stimulation with FK506 and fMLP. Tacrolimus 106-111 interleukin 3 Homo sapiens 0-4 7537264-2 1995 We report here that the immunosuppressants FK506 and cyclosporin A cause general growth inhibition of the vma3 mutant. Tacrolimus 43-48 H(+)-transporting V0 sector ATPase subunit c Saccharomyces cerevisiae S288C 106-110 7543725-8 1995 CsA- and FK506-treated rats, but not those treated with rapamycin, demonstrated high turnover osteoporosis with raised serum 1,25(OH)2D (p < 0.05) and elevated serum osteocalcin (p < 0.05). Tacrolimus 9-14 bone gamma-carboxyglutamate protein Rattus norvegicus 169-180 7537264-5 1995 The addition of FK506 decreases the cytosolic free concentration of Ca2+ in the vma3 mutant cells. Tacrolimus 16-21 H(+)-transporting V0 sector ATPase subunit c Saccharomyces cerevisiae S288C 80-84 7536932-7 1995 Neutrophil activation by mast cell-derived FKBP12 is prevented by complexing FKBP12 with FK506 or rapamycin. Tacrolimus 89-94 FKBP prolyl isomerase 1A Homo sapiens 43-49 7538962-0 1995 FKBP39, a Drosophila member of a family of proteins that bind the immunosuppressive drug FK506. Tacrolimus 89-94 FK506-binding protein 39kD Drosophila melanogaster 0-6 7709436-4 1995 Cox"s proportional hazards univariate and multivariate analyses showed that significant first-episode risk factors were: CMV seropositive donors for negative recipients (relative risk [RR], 3.86; P = 0.02), the average daily plasma trough level of tacrolimus (RR, 2.15; P = 0.04), and total amount of steroid boluses (RR, 2.90; P = 0.02). Tacrolimus 248-258 cytochrome c oxidase subunit 8A Homo sapiens 0-3 7536932-7 1995 Neutrophil activation by mast cell-derived FKBP12 is prevented by complexing FKBP12 with FK506 or rapamycin. Tacrolimus 89-94 FKBP prolyl isomerase 1A Homo sapiens 77-83 7536932-9 1995 They further suggest a pathophysiological role for FKBP12 as a mediator in immediate or type I hypersensitivity and may have implications for novel therapeutic strategies in the treatment of allergic disorders with FK506 and rapamycin. Tacrolimus 215-220 FKBP prolyl isomerase 1A Homo sapiens 51-57 7543780-5 1995 Simultaneous addition of Rap and CsA or Rap and FK506 inhibit the IL-2-mediated proliferation of TS1 beta and TS1 alpha beta cells and therefore FK506 does not revert the inhibition mediated by Rap in TS1 alpha beta cells. Tacrolimus 48-53 interleukin 2 Homo sapiens 66-70 7535208-0 1995 Combined effects of FK506 (tacrolimus) and cyclophosphamide on atypical B220+ T cells, cytokine gene expression and disease activity in MRL/MpJ-lpr/lpr mice. Tacrolimus 20-25 Fas (TNF receptor superfamily member 6) Mus musculus 144-147 7535208-9 1995 IL-10 and interferon-gamma (IFN-gamma) mRNAs were induced by FK506, CY and by the drug combination. Tacrolimus 61-66 interleukin 10 Mus musculus 0-5 7535208-9 1995 IL-10 and interferon-gamma (IFN-gamma) mRNAs were induced by FK506, CY and by the drug combination. Tacrolimus 61-66 interferon gamma Mus musculus 10-26 7535208-9 1995 IL-10 and interferon-gamma (IFN-gamma) mRNAs were induced by FK506, CY and by the drug combination. Tacrolimus 61-66 interferon gamma Mus musculus 28-37 7547679-6 1995 The effect of CsA on Bcl-2 expression is controlled by calcineurin since we have shown that FK506 but not rapamycin had the same effect on Bcl-2 expression, whereas okadaic acid, an inhibitor of phosphatases 1, 2A and 2C, was ineffective. Tacrolimus 92-97 BCL2 apoptosis regulator Homo sapiens 21-26 7547679-6 1995 The effect of CsA on Bcl-2 expression is controlled by calcineurin since we have shown that FK506 but not rapamycin had the same effect on Bcl-2 expression, whereas okadaic acid, an inhibitor of phosphatases 1, 2A and 2C, was ineffective. Tacrolimus 92-97 BCL2 apoptosis regulator Homo sapiens 139-144 7536825-3 1995 The complex of FK506 and FKBP-12 inhibits the calcium activated phosphatase, calcineurin, increasing phosphorylated levels of calcineurin substrates with growth associated protein-43 (GAP-43), being most prominent in the brain. Tacrolimus 15-20 FKBP prolyl isomerase 1A Homo sapiens 25-32 7534792-6 1995 Expression of p68 c-rel protein, but not p50 or p65, was suppressed by the immunosuppressive drug FK506. Tacrolimus 98-103 DNA polymerase delta 3, accessory subunit Homo sapiens 14-17 7534792-6 1995 Expression of p68 c-rel protein, but not p50 or p65, was suppressed by the immunosuppressive drug FK506. Tacrolimus 98-103 REL proto-oncogene, NF-kB subunit Homo sapiens 18-23 7534792-7 1995 Because FK506 specifically inhibits the appearance of mature single-positive thymocytes, gene expression regulated by p68 c-rel may play a role in selection and maturational signals involved in the double-positive to single-positive transition. Tacrolimus 8-13 DNA polymerase delta 3, accessory subunit Homo sapiens 118-121 7534792-7 1995 Because FK506 specifically inhibits the appearance of mature single-positive thymocytes, gene expression regulated by p68 c-rel may play a role in selection and maturational signals involved in the double-positive to single-positive transition. Tacrolimus 8-13 REL proto-oncogene, NF-kB subunit Homo sapiens 122-127 7536825-3 1995 The complex of FK506 and FKBP-12 inhibits the calcium activated phosphatase, calcineurin, increasing phosphorylated levels of calcineurin substrates with growth associated protein-43 (GAP-43), being most prominent in the brain. Tacrolimus 15-20 growth associated protein 43 Homo sapiens 154-182 7536825-3 1995 The complex of FK506 and FKBP-12 inhibits the calcium activated phosphatase, calcineurin, increasing phosphorylated levels of calcineurin substrates with growth associated protein-43 (GAP-43), being most prominent in the brain. Tacrolimus 15-20 growth associated protein 43 Homo sapiens 184-190 7628296-9 1995 Therefore, compounds interacting with CYP3A proteins are expected to cause drug-drug interactions (i.e. the antimycotics ketoconazole and clotrimazole, the steroids ethinylestradiol and testosterone, the ergots, the calcium channel blocker nifedipine, and the immunosuppressants FK-506 and rapamycin). Tacrolimus 279-285 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 38-43 9383417-3 1995 Rapamycin, ascomycin and FK506 have a common domain responsible for binding to FKBP12, their cellular receptor, and different effector domains that determine the target of the complex. Tacrolimus 25-30 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 79-85 9383417-7 1995 CONCLUSIONS: The designed rapamycin-based FKBP12 ligand exhibits powerful binding properties but, unlike rapamycin, shows no activity in IL-6 dependent B-cell proliferation and, in contrast to FK506, shows no activity in the IL-2 reporter assay. Tacrolimus 193-198 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 42-48 7532912-9 1995 These data demonstrate that: 1) FK 506 can be used to treat PSC; 2) the response to FK 506 by patients with PSC is rapid; and, 3) no adverse effect on the serum BUN and creatinine levels was observed. Tacrolimus 32-38 proteasome 20S subunit alpha 2 Homo sapiens 60-66 7538492-6 1995 The presence of IFN-gamma and LPS in the culture increased NO2- production by casein-elicited macrophages and partially eliminated the inhibition exerted by CsA and FK506. Tacrolimus 165-170 interferon gamma Mus musculus 16-25 7540862-0 1995 IL-5 production by CD4+ T cells of asthmatic patients is suppressed by glucocorticoids and the immunosuppressants FK506 and cyclosporin A. Tacrolimus 114-119 interleukin 5 Homo sapiens 0-4 7540862-0 1995 IL-5 production by CD4+ T cells of asthmatic patients is suppressed by glucocorticoids and the immunosuppressants FK506 and cyclosporin A. Tacrolimus 114-119 CD4 molecule Homo sapiens 19-22 7540862-6 1995 Dexamethasone, FK506 and cyclosporin A suppressed IL-5 production in vitro in a dose-dependent manner. Tacrolimus 15-20 interleukin 5 Homo sapiens 50-54 7540862-7 1995 Clear dose-dependent suppression of IL-5 gene expression by FK506 was also observed. Tacrolimus 60-65 interleukin 5 Homo sapiens 36-40 7538492-7 1995 Both drugs acted directly on the nitric oxide synthase (NOS), since CsA and FK506 reduced by 35% and by 17%, respectively, NOS activity in the crude cytosolic fraction. Tacrolimus 76-81 nitric oxide synthase 1, neuronal Mus musculus 33-54 7532665-3 1995 In this report, we investigated the effects of dexamethasone, cyclosporin A, FK506, and pyrrolidine dithiocarbamate (PDTC) on the induction of the ICAM-1 gene by cytokines in fibroblasts. Tacrolimus 77-82 intercellular adhesion molecule 1 Homo sapiens 147-153 7878764-11 1995 Tacrolimus immunosuppression of transplant rejection prevented the appearance of hsp-reactive lymphocytes in allografts. Tacrolimus 0-10 selenoprotein K Rattus norvegicus 81-84 7532685-3 1995 The effect of FK506, an inhibitor of calcineurin activation, on positive and negative selection in CD4+CD8+ double positive (DP) thymocytes was examined in normal mice and in a TCR transgenic mouse model. Tacrolimus 14-19 CD4 antigen Mus musculus 99-102 7532685-4 1995 In vivo FK506 treatment blocked the generation of mature TCRhighCD4+CD8- and TCRhighCD4-CD8+ thymocytes, and the induction of CD69 expression on DP thymocytes. Tacrolimus 8-13 CD69 antigen Mus musculus 126-130 7532685-5 1995 In addition, the shutdown of recombination activating gene 1 (RAG-1) transcription and the downregulation of CD4 and CD8 expression were inhibited by FK506 treatment suggesting that the activation of calcineurin is required for the first step (or the very early intracellular signaling events) of TCR-mediated positive selection of DP thymocytes. Tacrolimus 150-155 recombination activating 1 Mus musculus 29-60 7532685-5 1995 In addition, the shutdown of recombination activating gene 1 (RAG-1) transcription and the downregulation of CD4 and CD8 expression were inhibited by FK506 treatment suggesting that the activation of calcineurin is required for the first step (or the very early intracellular signaling events) of TCR-mediated positive selection of DP thymocytes. Tacrolimus 150-155 recombination activating 1 Mus musculus 62-67 7532685-5 1995 In addition, the shutdown of recombination activating gene 1 (RAG-1) transcription and the downregulation of CD4 and CD8 expression were inhibited by FK506 treatment suggesting that the activation of calcineurin is required for the first step (or the very early intracellular signaling events) of TCR-mediated positive selection of DP thymocytes. Tacrolimus 150-155 CD4 antigen Mus musculus 109-112 7538218-1 1995 We investigated the inhibitory action of FK506 (0.0005-5 micrograms/ml) on the metabolism of arachidonate 5-lipoxygenase in rat basophilic leukemia-1 cells. Tacrolimus 41-46 arachidonate 5-lipoxygenase Rattus norvegicus 93-120 7538218-8 1995 These results indicate that FK506 inhibits the production of peptide LTs, LTB4 and 5-HETE by inhibiting 5-lipoxygenase activity in intact cells. Tacrolimus 28-33 arachidonate 5-lipoxygenase Rattus norvegicus 104-118 7551978-1 1995 This study evaluates the ability of the immunosuppressive drugs dexamethasone, cyclosporine, FK506 and rapamycin, alone and in combination to suppress interleukin-1 beta (IL-1 beta) secretion in vitro by THP-1 cells when stimulated by lipopolysaccharide. Tacrolimus 93-98 interleukin 1 beta Homo sapiens 151-169 7551978-1 1995 This study evaluates the ability of the immunosuppressive drugs dexamethasone, cyclosporine, FK506 and rapamycin, alone and in combination to suppress interleukin-1 beta (IL-1 beta) secretion in vitro by THP-1 cells when stimulated by lipopolysaccharide. Tacrolimus 93-98 interleukin 1 beta Homo sapiens 171-180 7551978-4 1995 Cyclosporine, FK506 and rapamycin only partially suppress secretion of IL-1 beta at concentrations within their therapeutic ranges and increasing concentrations of the drugs do not result in further suppression of secretion. Tacrolimus 14-19 interleukin 1 beta Homo sapiens 71-80 7551978-7 1995 These data suggest that cyclosporine, FK506 and rapamycin all share a common effect on the production of IL-1 beta, different from that of dexamethasone. Tacrolimus 38-43 interleukin 1 beta Homo sapiens 105-114 7532676-0 1995 FK506 inhibits antigen receptor-mediated induction of c-rel in B and T lymphoid cells. Tacrolimus 0-5 REL proto-oncogene, NF-kB subunit Homo sapiens 54-59 7532676-4 1995 Furthermore, c-rel induction is blocked by the immunosuppressive drug FK506 that is known to inhibit B and T cell activation. Tacrolimus 70-75 REL proto-oncogene, NF-kB subunit Homo sapiens 13-18 7530743-1 1995 FK506 and cyclosporin A (CsA) are immunosuppressive agents that inhibit IL-2 production by activated T cells, but only CsA inhibits IgE activation-induced cytokine transcripts in mouse IL-3-dependent, bone marrow-derived mast cells (BMMC). Tacrolimus 0-5 interleukin 2 Mus musculus 72-76 7530743-1 1995 FK506 and cyclosporin A (CsA) are immunosuppressive agents that inhibit IL-2 production by activated T cells, but only CsA inhibits IgE activation-induced cytokine transcripts in mouse IL-3-dependent, bone marrow-derived mast cells (BMMC). Tacrolimus 0-5 interleukin 3 Mus musculus 185-189 7532879-5 1995 In contrast, only the highest concentrations of FK506 and CsA significantly altered PDGF- or bFGF-induced VSMC DNA synthesis. Tacrolimus 48-53 fibroblast growth factor 2 Rattus norvegicus 93-97 7530743-2 1995 We previously associated the resistance of BMMC to FK506 with a deficiency in the expression of FK506 binding protein (FKBP) 12, a molecule that forms a complex with FK506 capable of inhibiting calcineurin phosphatase activity in vitro. Tacrolimus 51-56 FK506 binding protein 1a Mus musculus 96-117 7532879-7 1995 The extent of the antagonism of RPM"s inhibition of bFGF-induced VSMC DNA synthesis by FK506 was inversely proportional to RPM concentration and directly proportional to FK506 concentration. Tacrolimus 87-92 fibroblast growth factor 2 Rattus norvegicus 52-56 7530743-2 1995 We previously associated the resistance of BMMC to FK506 with a deficiency in the expression of FK506 binding protein (FKBP) 12, a molecule that forms a complex with FK506 capable of inhibiting calcineurin phosphatase activity in vitro. Tacrolimus 51-56 FK506 binding protein 1a Mus musculus 119-123 7532879-7 1995 The extent of the antagonism of RPM"s inhibition of bFGF-induced VSMC DNA synthesis by FK506 was inversely proportional to RPM concentration and directly proportional to FK506 concentration. Tacrolimus 170-175 fibroblast growth factor 2 Rattus norvegicus 52-56 7530743-2 1995 We previously associated the resistance of BMMC to FK506 with a deficiency in the expression of FK506 binding protein (FKBP) 12, a molecule that forms a complex with FK506 capable of inhibiting calcineurin phosphatase activity in vitro. Tacrolimus 96-101 FK506 binding protein 1a Mus musculus 119-123 7530743-3 1995 In this report, we establish that FKBP12 mediates FK506 inhibition of both calcineurin phosphatase activity and IgE activation-induced cytokine transcripts in a Kirsten murine sarcoma virus-immortalized mast cell line that is FKBP12 deficient. Tacrolimus 50-55 FK506 binding protein 1a Mus musculus 34-40 7530743-3 1995 In this report, we establish that FKBP12 mediates FK506 inhibition of both calcineurin phosphatase activity and IgE activation-induced cytokine transcripts in a Kirsten murine sarcoma virus-immortalized mast cell line that is FKBP12 deficient. Tacrolimus 50-55 FK506 binding protein 1a Mus musculus 226-232 7530743-4 1995 Overexpression of FKBP12 by transfection enhanced the ability of FK506 to inhibit calcineurin phosphatase activity (IC50 = 2 nM), compared with cells transfected with the expression vector alone (IC50 > 30 nM). Tacrolimus 65-70 FK506 binding protein 1a Mus musculus 18-24 7531689-2 1995 FKBP-12 (FKBP), the soluble receptor for the immunosuppresant drug FK-506, is tightly bound to the calcium release channel (CRC)/ryanodine receptor (RyR) of skeletal muscle terminal cisternae (TC) of sarcoplasmic reticulum with a stoichiometry of 4 mol of FKBP per tetrameric RyR complex. Tacrolimus 67-73 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 0-7 7530743-5 1995 The IC50 value for FK506 inhibition of IgE activation-induced transcripts for TNF-alpha decreased from 40 nM in vector control cells to 10 nM in FKBP12 transfectants. Tacrolimus 19-24 tumor necrosis factor Mus musculus 78-87 7531689-2 1995 FKBP-12 (FKBP), the soluble receptor for the immunosuppresant drug FK-506, is tightly bound to the calcium release channel (CRC)/ryanodine receptor (RyR) of skeletal muscle terminal cisternae (TC) of sarcoplasmic reticulum with a stoichiometry of 4 mol of FKBP per tetrameric RyR complex. Tacrolimus 67-73 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 0-4 7530743-5 1995 The IC50 value for FK506 inhibition of IgE activation-induced transcripts for TNF-alpha decreased from 40 nM in vector control cells to 10 nM in FKBP12 transfectants. Tacrolimus 19-24 FK506 binding protein 1a Mus musculus 145-151 7531689-2 1995 FKBP-12 (FKBP), the soluble receptor for the immunosuppresant drug FK-506, is tightly bound to the calcium release channel (CRC)/ryanodine receptor (RyR) of skeletal muscle terminal cisternae (TC) of sarcoplasmic reticulum with a stoichiometry of 4 mol of FKBP per tetrameric RyR complex. Tacrolimus 67-73 ryanodine receptor 1 Homo sapiens 149-152 7531689-2 1995 FKBP-12 (FKBP), the soluble receptor for the immunosuppresant drug FK-506, is tightly bound to the calcium release channel (CRC)/ryanodine receptor (RyR) of skeletal muscle terminal cisternae (TC) of sarcoplasmic reticulum with a stoichiometry of 4 mol of FKBP per tetrameric RyR complex. Tacrolimus 67-73 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 9-13 7530743-7 1995 In contrast, activation-elicited release of the secretory granule mediator beta-hexosaminidase was only partially inhibited by FK506 at 1000 nM, regardless of the levels of FKBP12 expressed by the cells. Tacrolimus 127-132 O-GlcNAcase Mus musculus 75-94 7530743-8 1995 Thus, FKBP12 is the dominant cytosolic protein that mediates FK506 inhibition of TNF-alpha and IL-6 transcripts. Tacrolimus 61-66 FK506 binding protein 1a Mus musculus 6-12 7530743-8 1995 Thus, FKBP12 is the dominant cytosolic protein that mediates FK506 inhibition of TNF-alpha and IL-6 transcripts. Tacrolimus 61-66 tumor necrosis factor Mus musculus 81-90 7530743-8 1995 Thus, FKBP12 is the dominant cytosolic protein that mediates FK506 inhibition of TNF-alpha and IL-6 transcripts. Tacrolimus 61-66 interleukin 6 Mus musculus 95-99 7533090-7 1995 Increased expression of FKBP12 resulted in increased sensitivity to FK506 and rapamycin, as measured by inhibition of calcineurin activity and p70 S6 kinase activity, respectively. Tacrolimus 68-73 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 24-30 7531627-0 1995 IL-8/IL-8 receptor expression in psoriasis and the response to systemic tacrolimus (FK506) therapy. Tacrolimus 72-82 C-X-C motif chemokine ligand 8 Homo sapiens 0-4 7531627-0 1995 IL-8/IL-8 receptor expression in psoriasis and the response to systemic tacrolimus (FK506) therapy. Tacrolimus 72-82 C-X-C motif chemokine ligand 8 Homo sapiens 5-9 7531627-0 1995 IL-8/IL-8 receptor expression in psoriasis and the response to systemic tacrolimus (FK506) therapy. Tacrolimus 84-89 C-X-C motif chemokine ligand 8 Homo sapiens 0-4 7531627-0 1995 IL-8/IL-8 receptor expression in psoriasis and the response to systemic tacrolimus (FK506) therapy. Tacrolimus 84-89 C-X-C motif chemokine ligand 8 Homo sapiens 5-9 7531627-5 1995 IL-8 mRNA was not detected in the skin of any patient after the start of systemic tacrolimus therapy; IL-1 beta, IL-6 and IFN-gamma transcripts were also reduced. Tacrolimus 82-92 C-X-C motif chemokine ligand 8 Homo sapiens 0-4 7531627-8 1995 Estimation of circulating IL-8 levels by enzyme immunoassay showed that all patients with detectable IL-8 before treatment had decreased levels in response to treatment with tacrolimus; reductions in PASI scores were accompanied by decreases in IL-8 levels, that varied both in rate and extent. Tacrolimus 174-184 C-X-C motif chemokine ligand 8 Homo sapiens 26-30 7531627-8 1995 Estimation of circulating IL-8 levels by enzyme immunoassay showed that all patients with detectable IL-8 before treatment had decreased levels in response to treatment with tacrolimus; reductions in PASI scores were accompanied by decreases in IL-8 levels, that varied both in rate and extent. Tacrolimus 174-184 C-X-C motif chemokine ligand 8 Homo sapiens 101-105 7531627-8 1995 Estimation of circulating IL-8 levels by enzyme immunoassay showed that all patients with detectable IL-8 before treatment had decreased levels in response to treatment with tacrolimus; reductions in PASI scores were accompanied by decreases in IL-8 levels, that varied both in rate and extent. Tacrolimus 174-184 C-X-C motif chemokine ligand 8 Homo sapiens 101-105 7531627-11 1995 They further suggest that interference with IL-8 production and/or that of other key chemokines may be an important mechanism underlying the therapeutic efficacy of tacrolimus, and other agents such as cyclosporin A, with similar molecular actions. Tacrolimus 165-175 C-X-C motif chemokine ligand 8 Homo sapiens 44-48 7533090-9 1995 Two distinct point mutations in FKBP12, one altering a hydrophobic residue within the drug-binding pocket and the other changing a charged surface residue of FKBP12, abrogated its ability to mediate sensitivity to FK506 and rapamycin. Tacrolimus 214-219 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 32-38 7533090-9 1995 Two distinct point mutations in FKBP12, one altering a hydrophobic residue within the drug-binding pocket and the other changing a charged surface residue of FKBP12, abrogated its ability to mediate sensitivity to FK506 and rapamycin. Tacrolimus 214-219 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 158-164 7533090-10 1995 These results establish that FKBP12 can mediate sensitivity to both FK506 and rapamycin in mammalian cells. Tacrolimus 68-73 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 29-35 7532989-2 1995 Based on functional and sequence homology studies, it was recently discovered that hsp56 also belongs to the FKBP class of immunophilin proteins, which are thought to mediate the actions of the immunosuppressive drugs FK506 and rapamycin. Tacrolimus 218-223 FKBP prolyl isomerase 4 Homo sapiens 83-88 7875200-7 1995 The immunosuppressant FK506 as well as dominant negative alleles of Ras and Raf inhibited HamT-induced IL-2 transcription. Tacrolimus 22-27 interleukin 2 Mus musculus 103-107 7532989-6 1995 It was also found that the untransformed but not the transformed GR was retained following affinity chromatography with FK506-affigel resin, reinforcing the possibility that hsp56 within the untransformed GR complex could be a target for the actions of FK506. Tacrolimus 253-258 FKBP prolyl isomerase 4 Homo sapiens 174-179 7532989-8 1995 This inhibition of GR transformation by FK506 was shown to correlate with an inhibition of Dex-induced GR/hsp90 dissociation, with 10 microM FK506 preventing 48% of the GR/hsp90 complexes from dissociating. Tacrolimus 40-45 heat shock protein 86, pseudogene 1 Mus musculus 106-111 7532989-8 1995 This inhibition of GR transformation by FK506 was shown to correlate with an inhibition of Dex-induced GR/hsp90 dissociation, with 10 microM FK506 preventing 48% of the GR/hsp90 complexes from dissociating. Tacrolimus 141-146 heat shock protein 86, pseudogene 1 Mus musculus 106-111 7532989-8 1995 This inhibition of GR transformation by FK506 was shown to correlate with an inhibition of Dex-induced GR/hsp90 dissociation, with 10 microM FK506 preventing 48% of the GR/hsp90 complexes from dissociating. Tacrolimus 141-146 heat shock protein 86, pseudogene 1 Mus musculus 172-177 7532989-11 1995 Although we speculate that these actions of FK506 on the GR complex are mediated by the associated hsp56 component, other possible mechanisms are also discussed. Tacrolimus 44-49 FKBP prolyl isomerase 4 Homo sapiens 99-104 8744655-2 1995 The present study examines the in vitro interactions of classical immunosuppressive agents--FK 506 and Cyclosporine A (CsA) with 2-CdA at the level of T and B cells proliferation, expression receptor for interleukin 2 (R-IL-2) and Ig synthesis. Tacrolimus 92-98 interleukin 2 Homo sapiens 204-217 7533409-0 1995 Inhibition of insulin production by FK 506 is caused at the transcriptional level in pancreatic beta cell when FK BP-12 content is relatively high. Tacrolimus 36-42 FKBP prolyl isomerase 1A Homo sapiens 111-119 7530217-0 1995 Rapamycin, FK506 and cyclosporin A inhibit human prolactin gene expression. Tacrolimus 11-16 prolactin Homo sapiens 49-58 7530217-1 1995 In this work we demonstrate that transcription of the human prolactin gene is inhibited by the immunosuppressants FK506 (IC50 = 25 nM), cyclosporin A (IC50 = 190 nM) and rapamycin (IC50 = 25 nM). Tacrolimus 114-119 prolactin Homo sapiens 60-69 7530217-2 1995 Whereas the effect of FK506 and cyclosporin A is specific for prolactin gene transcription, rapamycin has a more general effect on transcription and/or translation in pituitary cells. Tacrolimus 22-27 prolactin Homo sapiens 62-71 7531976-1 1995 The ability of the immunosuppressive agent FK506 to affect growth of the epidermal growth factor-receptor (EGF-R) overexpressing cell line, A431, was compared with that of the structurally unrelated immunosuppressive compound, cyclosporin A (CyA). Tacrolimus 43-48 epidermal growth factor receptor Homo sapiens 73-105 7531976-1 1995 The ability of the immunosuppressive agent FK506 to affect growth of the epidermal growth factor-receptor (EGF-R) overexpressing cell line, A431, was compared with that of the structurally unrelated immunosuppressive compound, cyclosporin A (CyA). Tacrolimus 43-48 epidermal growth factor receptor Homo sapiens 107-112 7529414-4 1995 They interrupt the cytoplasmic portion of T-cell signaling by forming a complex with a binding protein--FKBP12 in the case of FK506 and rapamycin and cyclophilin A (CyPA) in the case of cyclosporin A (CsA). Tacrolimus 126-131 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 104-110 7529414-7 1995 The existence of strong FK506-FKBP12 binding suggests that FK506 is mimicking some natural ligand for FKBP12. Tacrolimus 24-29 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 30-36 7529414-7 1995 The existence of strong FK506-FKBP12 binding suggests that FK506 is mimicking some natural ligand for FKBP12. Tacrolimus 24-29 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 102-108 7487142-0 1995 FK506: therapeutic effects on lupus dermatoses in autoimmune-prone MRL/Mp-lpr/lpr mice. Tacrolimus 0-5 Fas (TNF receptor superfamily member 6) Mus musculus 67-81 22827272-6 1995 IL-8 production by TCC from the ocular fluid was further up-regulated upon stimulatation with PHA, but was suppressed by FK506 and hydrocortisone, though not by diclofenac sodium. Tacrolimus 121-126 C-X-C motif chemokine ligand 8 Homo sapiens 0-4 7529995-3 1995 The existence of a slow cis-trans interconversion of an imidic bond in the inhibitor molecule during the course of the formation of the CsA-CyP18cy complex (where CyP18cy is human 18 kDa cytosolic CyP) prompted us to investigate the reaction of the peptidomacrolides FK506, ascomycin and rapamycin with two specific binding-proteins in more detail. Tacrolimus 267-272 peptidylprolyl isomerase G Homo sapiens 140-143 7529995-5 1995 For FK506, the kinetics of inhibition of human 12 kDa cytosolic FKBP (FKBP12cy) were clearly dependent on time. Tacrolimus 4-9 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 70-76 7529995-9 1995 On the other hand, the kinetics and amplitudes of the inhibition of FKBP12cy varied significantly if rapamycin was used as an inhibitor instead of FK 506. Tacrolimus 147-153 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 68-74 7544748-4 1995 Using a cytotoxicity assay and a radioimmunoassay, we observed significant increases in levels of tumor necrosis factor-alpha (TNF-alpha) in the colitis mucosa and detected interleukin-1 alpha in the mucosa of 3 of 5 DSS rats and an increase in TNF-alpha had a tendency to be inhibited by treatment with FK506. Tacrolimus 304-309 tumor necrosis factor Rattus norvegicus 98-125 7544748-4 1995 Using a cytotoxicity assay and a radioimmunoassay, we observed significant increases in levels of tumor necrosis factor-alpha (TNF-alpha) in the colitis mucosa and detected interleukin-1 alpha in the mucosa of 3 of 5 DSS rats and an increase in TNF-alpha had a tendency to be inhibited by treatment with FK506. Tacrolimus 304-309 tumor necrosis factor Rattus norvegicus 127-136 7529023-8 1994 The interaction between FKBP-12 (FK506 binding protein) and the ryanodine-binding Ca2+ channel may be an essential link in the chain of events by which FK506 alters Ca(2+)-dependent cellular processes. Tacrolimus 33-38 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 24-31 7530227-0 1994 The yeast FKS1 gene encodes a novel membrane protein, mutations in which confer FK506 and cyclosporin A hypersensitivity and calcineurin-dependent growth. Tacrolimus 80-85 1,3-beta-D-glucan synthase Saccharomyces cerevisiae S288C 10-14 7530227-4 1994 We and others have defined the Saccharomyces cerevisiae FKS1 gene by recessive mutations resulting in 100-1000-fold hypersensitivity to FK506 and CsA (as compared to wild type), but which do not affect sensitivity to a variety of other antifungal drugs. Tacrolimus 136-141 1,3-beta-D-glucan synthase Saccharomyces cerevisiae S288C 56-60 7530227-12 1994 These data suggest that FKS1 provides a unique cellular function which, when absent, increases FK506 and CsA sensitivity by making the CNs (or a CN-dependent function) essential. Tacrolimus 95-100 1,3-beta-D-glucan synthase Saccharomyces cerevisiae S288C 24-28 7806527-3 1994 This up-regulation of Cdk4 mRNA and protein was resistant to the immunosuppressant drugs cyclosporin A (CsA) and FK506. Tacrolimus 113-118 cyclin dependent kinase 4 Homo sapiens 22-26 7527548-1 1994 A soluble 12-kDa FK506 binding protein (FKBP12), the cellular receptor of the immunosuppressive drug FK506, is tightly associated with the Ca2+ release channel of rabbit skeletal muscle sarcoplasmic reticulum [Jayaraman, T., Brillantes, A. M., Timerman, A. P., Fleischer, S., Erdjument-Bromage, H., Tempst, P. & Marks, A. Tacrolimus 17-22 peptidyl-prolyl cis-trans isomerase FKBP1A Oryctolagus cuniculus 40-46 7529175-1 1994 The peptidyl-prolyl isomerases FKBP12 and cyclophilin A (immunophilins) form complexes with the immunosuppressants FK506 and cyclosporin A that inhibit the phosphatase calcineurin. Tacrolimus 115-120 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 31-37 7529175-2 1994 With the yeast two hybrid system, we detect complexes between FKBP12 and the calcineurin A catalytic subunit in both the presence and absence of FK506. Tacrolimus 145-150 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 62-68 7529175-3 1994 Mutations in FKBP12 surface residues or the absence of the calcineurin B regulatory subunit perturb the FK506-dependent, but not the ligand-independent, FKBP12-calcineurin complex. Tacrolimus 104-109 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 13-19 7529175-3 1994 Mutations in FKBP12 surface residues or the absence of the calcineurin B regulatory subunit perturb the FK506-dependent, but not the ligand-independent, FKBP12-calcineurin complex. Tacrolimus 104-109 protein phosphatase 3 regulatory subunit B, alpha Homo sapiens 59-72 7529739-1 1994 FKBP12 is an 11.8-kDa protein that binds the potent immunosuppressants FK506 and rapamycin. Tacrolimus 71-76 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 0-6 7529739-2 1994 When bound to FK506, FKBP12 forms an inhibitory complex with calcineurin and interferes with signal transduction in activated T lymphocytes. Tacrolimus 14-19 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 21-27 7528676-5 1994 However, the effects of the drug on proliferative responses elicited by anti-Ig or anti-CD40 differ significantly: firstly, the response to anti-CD40 + IL-4 becomes completely FK506-resistant within 24 h, whilst that to anti-Ig remains sensitive significantly longer. Tacrolimus 176-181 CD40 antigen Mus musculus 88-92 7528676-5 1994 However, the effects of the drug on proliferative responses elicited by anti-Ig or anti-CD40 differ significantly: firstly, the response to anti-CD40 + IL-4 becomes completely FK506-resistant within 24 h, whilst that to anti-Ig remains sensitive significantly longer. Tacrolimus 176-181 CD40 antigen Mus musculus 145-149 7528676-1 1994 Ligation of CD40 activates B cells via a Ca(++)-dependent, FK506-sensitive pathway. Tacrolimus 59-64 CD40 antigen Mus musculus 12-16 7528676-5 1994 However, the effects of the drug on proliferative responses elicited by anti-Ig or anti-CD40 differ significantly: firstly, the response to anti-CD40 + IL-4 becomes completely FK506-resistant within 24 h, whilst that to anti-Ig remains sensitive significantly longer. Tacrolimus 176-181 interleukin 4 Mus musculus 152-156 7528676-6 1994 Secondly, stimulating B cells concurrently via CD40, surface Ig (sIg) and IL-4 receptors invokes an FK506-resistant activation pathway. Tacrolimus 100-105 CD40 antigen Mus musculus 47-51 7528676-6 1994 Secondly, stimulating B cells concurrently via CD40, surface Ig (sIg) and IL-4 receptors invokes an FK506-resistant activation pathway. Tacrolimus 100-105 interleukin 4 Mus musculus 74-78 7528676-7 1994 We previously reported that ligation of either sIg or CD40 receptors, in conjunction with IL-4, induces the transcription factor, nuclear factor of activated T cells (NF-AT) in B cells, via a CsA/FK506-sensitive pathway. Tacrolimus 196-201 CD40 antigen Mus musculus 54-58 7528676-7 1994 We previously reported that ligation of either sIg or CD40 receptors, in conjunction with IL-4, induces the transcription factor, nuclear factor of activated T cells (NF-AT) in B cells, via a CsA/FK506-sensitive pathway. Tacrolimus 196-201 interleukin 4 Mus musculus 90-94 7528676-8 1994 However, NF-AT induction elicited by anti-Ig/anti-CD40/IL-4 is still FK506 sensitive, implying that the drug resistance of the response to these three stimuli involves additional components than NF-AT. Tacrolimus 69-74 CD40 antigen Mus musculus 50-54 7526495-1 1994 The microbial products FK506 and CsA are potent immunosuppressive agents that prevent early transcriptional events in TcR-mediated activation. Tacrolimus 23-28 T cell receptor alpha variable 6-3 Mus musculus 118-121 7708060-2 1994 These untransformed nuclear receptors exist in a heterocomplex containing three heat shock proteins, hsp90, hsp70, and hsp56, the latter being an immunophilin of the FK506 binding type whose cellular function is unknown. Tacrolimus 166-171 heat shock protein 90 alpha family class A member 1 Rattus norvegicus 101-106 7524662-0 1994 Solution structure of FK506 bound to the R42K, H87V double mutant of FKBP-12. Tacrolimus 22-27 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 69-76 7524662-1 1994 The binding of the FK506/FKBP-12 complex to calcineurin (CN), its putative target for immunosuppression, involves recognition of solvent-exposed regions of the ligand as well as FKBP-12 residues near the active site. Tacrolimus 19-24 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 25-32 7524662-1 1994 The binding of the FK506/FKBP-12 complex to calcineurin (CN), its putative target for immunosuppression, involves recognition of solvent-exposed regions of the ligand as well as FKBP-12 residues near the active site. Tacrolimus 19-24 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 178-185 7524662-6 1994 This work reports the solution structure of 13C-labeled FK506 bound to R42K, H87V FKBP-12. Tacrolimus 56-61 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 82-89 7524662-10 1994 Comparison with the NMR structure of FK506 bound to wild-type FKBP-12 reveals that the R42K, H87V mutation causes the ligand backbone near C16 to move by 2.5 to 4.5 A, reorients 15-MeO by 90 degrees, and shifts 13-MeO by approximately 1.5 A. Tacrolimus 37-42 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 62-69 7536706-1 1994 The immunosuppressant FK506 inhibits N-alpha-benzyloxylcarbonyl-L-lysine thiobenzyl ester (BLT) esterase release from cytotoxic T lymphocytes (CTL). Tacrolimus 22-27 granzyme A Mus musculus 37-104 7525596-0 1994 A novel FK506- and rapamycin-binding protein (FPR3 gene product) in the yeast Saccharomyces cerevisiae is a proline rotamase localized to the nucleolus. Tacrolimus 8-13 peptidylprolyl isomerase FPR3 Saccharomyces cerevisiae S288C 46-50 7531458-0 1994 FK506 can inhibit apoptotic cell death induced by the HIV-1 envelope glycoprotein gp120. Tacrolimus 0-5 Envelope surface glycoprotein gp160, precursor Human immunodeficiency virus 1 82-87 7530693-2 1994 The effect of the immunosuppressant FK506 on the expression of TCR alpha beta in rat CD4- CD8- thymocytes was also examined. Tacrolimus 36-41 Cd4 molecule Rattus norvegicus 85-88 7530693-6 1994 In contrast, continuous treatment with FK506 for 7 days markedly decreased not only the percentages of CD4+ CD8- TCR alpha beta high and CD4- CD8+ TCR alpha beta high thymocytes, but also that of CD4- CD8- TCR alpha beta high thymocytes. Tacrolimus 39-44 Cd4 molecule Rattus norvegicus 103-106 7530693-6 1994 In contrast, continuous treatment with FK506 for 7 days markedly decreased not only the percentages of CD4+ CD8- TCR alpha beta high and CD4- CD8+ TCR alpha beta high thymocytes, but also that of CD4- CD8- TCR alpha beta high thymocytes. Tacrolimus 39-44 Cd4 molecule Rattus norvegicus 137-140 7530693-6 1994 In contrast, continuous treatment with FK506 for 7 days markedly decreased not only the percentages of CD4+ CD8- TCR alpha beta high and CD4- CD8+ TCR alpha beta high thymocytes, but also that of CD4- CD8- TCR alpha beta high thymocytes. Tacrolimus 39-44 Cd4 molecule Rattus norvegicus 137-140 7525815-4 1994 On the other hand, an immunosuppressant, FK506, and a glucocorticoid inhibit the gene transcription as well as the production of IL-8. Tacrolimus 41-46 C-X-C motif chemokine ligand 8 Homo sapiens 129-133 7530727-0 1994 A soluble binding assay for measuring 3H-FK506 binding to the hsp56 immunophilin. Tacrolimus 41-46 FKBP prolyl isomerase 4 Homo sapiens 62-67 7530727-1 1994 Heat shock protein 56 (hsp56) was previously identified as an immunophilin based on its ability to specifically bind to FK506-Affi-Gel 10. Tacrolimus 120-125 FKBP prolyl isomerase 4 Homo sapiens 0-21 7530727-1 1994 Heat shock protein 56 (hsp56) was previously identified as an immunophilin based on its ability to specifically bind to FK506-Affi-Gel 10. Tacrolimus 120-125 FKBP prolyl isomerase 4 Homo sapiens 23-28 7530727-6 1994 These results demonstrate that hsp56 binds FK506 and rapamycin with similar affinities, and suggest that hsp56 may play a role in mediating the cellular function of both of these drugs. Tacrolimus 43-48 FKBP prolyl isomerase 4 Homo sapiens 31-36 7530727-6 1994 These results demonstrate that hsp56 binds FK506 and rapamycin with similar affinities, and suggest that hsp56 may play a role in mediating the cellular function of both of these drugs. Tacrolimus 43-48 FKBP prolyl isomerase 4 Homo sapiens 105-110 7522130-2 1994 Although no change in cytoplasmic calcium level ([Ca2+]i) was detectable during antigen-specific signal transduction of 171-CD4+ cells, IL2 induction was inhibited by FK506 and CsA. Tacrolimus 167-172 interleukin 2 Homo sapiens 136-139 7522636-7 1994 Cyclosporin A and FK506 act on two distinct levels of the IL-2 control mechanism. Tacrolimus 18-23 interleukin 2 Homo sapiens 58-62 7522130-5 1994 We suggest that IL2 secretion induced by antigen presentation to TCR/CD4/p56lck requires an FK506 and cyclosporin A-sensitive step which may be independent of calcium signaling. Tacrolimus 92-97 interleukin 2 Homo sapiens 16-19 7522130-5 1994 We suggest that IL2 secretion induced by antigen presentation to TCR/CD4/p56lck requires an FK506 and cyclosporin A-sensitive step which may be independent of calcium signaling. Tacrolimus 92-97 CD4 molecule Homo sapiens 69-72 7522130-5 1994 We suggest that IL2 secretion induced by antigen presentation to TCR/CD4/p56lck requires an FK506 and cyclosporin A-sensitive step which may be independent of calcium signaling. Tacrolimus 92-97 LCK proto-oncogene, Src family tyrosine kinase Homo sapiens 73-79 7523855-0 1994 Two FK506 resistance-conferring genes in Saccharomyces cerevisiae, TAT1 and TAT2, encode amino acid permeases mediating tyrosine and tryptophan uptake. Tacrolimus 4-9 amino acid transporter TAT1 Saccharomyces cerevisiae S288C 67-71 7523855-0 1994 Two FK506 resistance-conferring genes in Saccharomyces cerevisiae, TAT1 and TAT2, encode amino acid permeases mediating tyrosine and tryptophan uptake. Tacrolimus 4-9 aromatic amino acid transmembrane transporter TAT2 Saccharomyces cerevisiae S288C 76-80 7523855-2 1994 We reported previously that an FK506-sensitive target in S. cerevisiae is required for amino acid import and that overexpression of two new genes, TAT1 and TAT2 (formerly called TAP1 and TAP2), confers resistance to the drug. Tacrolimus 31-36 amino acid transporter TAT1 Saccharomyces cerevisiae S288C 147-151 7528255-7 1994 After the induction of FK506, the number of CD8 positive cells decreased and cardiac rejection was successfully resolved. Tacrolimus 23-28 CD8a molecule Homo sapiens 44-47 7523855-2 1994 We reported previously that an FK506-sensitive target in S. cerevisiae is required for amino acid import and that overexpression of two new genes, TAT1 and TAT2 (formerly called TAP1 and TAP2), confers resistance to the drug. Tacrolimus 31-36 aromatic amino acid transmembrane transporter TAT2 Saccharomyces cerevisiae S288C 156-160 7523855-2 1994 We reported previously that an FK506-sensitive target in S. cerevisiae is required for amino acid import and that overexpression of two new genes, TAT1 and TAT2 (formerly called TAP1 and TAP2), confers resistance to the drug. Tacrolimus 31-36 amino acid transporter TAT1 Saccharomyces cerevisiae S288C 178-182 7523855-2 1994 We reported previously that an FK506-sensitive target in S. cerevisiae is required for amino acid import and that overexpression of two new genes, TAT1 and TAT2 (formerly called TAP1 and TAP2), confers resistance to the drug. Tacrolimus 31-36 aromatic amino acid transmembrane transporter TAT2 Saccharomyces cerevisiae S288C 187-191 7522447-2 1994 Contrarily to the inhibition observed with the immunosuppressant complex FKBP-12-FK506, no significant inhibition was observed with FKBP-59/HBI or FKBP-59/HBI-I in the presence of FK506, even though FKBP-59/HBI-1 is nearly 55% homologous to the immunophilin FKBP-12. Tacrolimus 81-86 peptidyl-prolyl cis-trans isomerase FKBP1A Oryctolagus cuniculus 73-80 7522303-1 1994 The immunosuppressive action of the drug FK506 involves inhibition of calcineurin in T-lymphocytes by a complex of FK506 and an FK506 binding protein, FKBP12, a member of the immunophilin protein family. Tacrolimus 41-46 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 151-157 7522303-1 1994 The immunosuppressive action of the drug FK506 involves inhibition of calcineurin in T-lymphocytes by a complex of FK506 and an FK506 binding protein, FKBP12, a member of the immunophilin protein family. Tacrolimus 115-120 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 151-157 7521210-0 1994 Potentiation of progesterone receptor-mediated transcription by the immunosuppressant FK506. Tacrolimus 86-91 progesterone receptor Homo sapiens 16-37 7521210-7 1994 To gain insight into the mechanism of FK506"s regulation of PR action, we questioned whether calcineurin is involved, because it has been shown that FK506 is a specific inhibitor of calcineurin, a Ca(2+)- and calmodulin-regulated phosphatase, through the formation of an FKBP12-FK506-calcineurin-calmodulin complex. Tacrolimus 149-154 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 271-277 7521210-7 1994 To gain insight into the mechanism of FK506"s regulation of PR action, we questioned whether calcineurin is involved, because it has been shown that FK506 is a specific inhibitor of calcineurin, a Ca(2+)- and calmodulin-regulated phosphatase, through the formation of an FKBP12-FK506-calcineurin-calmodulin complex. Tacrolimus 149-154 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 271-277 7521210-10 1994 Furthermore, immunoblot analysis showed that both FK506 and calmidazolium potentiated the effect of progesterone in decreasing the mobility of hPR-B upon sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Tacrolimus 50-55 RB transcriptional corepressor 1 Homo sapiens 143-148 7521210-11 1994 This suggests that FK506 and calmidazolium may cooperate with progesterone in increasing the level of hPR-B phosphorylation. Tacrolimus 19-24 RB transcriptional corepressor 1 Homo sapiens 102-107 7520353-0 1994 Inhibition of anthralin-caused skin tumor promotion and interleukin-1 alpha production by potent immunosuppressant FK506. Tacrolimus 115-120 interleukin 1 alpha Mus musculus 56-75 7974924-2 1994 Several new drugs have been found to be effective immunosuppressive agents: FK 506, a macrolid antibiotic inhibiting lymphokine gene transcription, lymphokine production and secretion, rapamycin, which blocks effects of lymphokine-induced signal transduction, and RS 61443, brequinar and mizoribine, which inhibit DNA/RNA synthesis and lymphocyte proliferation. Tacrolimus 76-82 interleukin 2 Homo sapiens 117-127 7520912-0 1994 The native v-Raf.hsp90.p50 heterocomplex contains a novel immunophilin of the FK506 binding class. Tacrolimus 78-83 heat shock protein 90 alpha family class A member 1 Rattus norvegicus 17-22 7520912-10 1994 We have immunoadsorbed v-Raf from stably transfected rat 3Y1 fibroblasts and show that the immunoadsorbed v-Raf.hsp90.p50 heterocomplex binds the immunosuppressant drug [3H]FK506. Tacrolimus 173-178 heat shock protein 90 alpha family class A member 1 Rattus norvegicus 112-117 7528280-0 1994 FK506, an immunosuppressant, partially inhibits interleukin 6 production by adherent rheumatoid synovial cells. Tacrolimus 0-5 interleukin 6 Homo sapiens 48-61 7528280-1 1994 OBJECTIVE: To investigate the effect of a new immunosuppressant, FK506, on interleukin 6 (IL-6) production by freshly prepared rheumatoid synovial cells. Tacrolimus 65-70 interleukin 6 Homo sapiens 75-88 7528280-1 1994 OBJECTIVE: To investigate the effect of a new immunosuppressant, FK506, on interleukin 6 (IL-6) production by freshly prepared rheumatoid synovial cells. Tacrolimus 65-70 interleukin 6 Homo sapiens 90-94 7528280-7 1994 This spontaneous production of IL-6 was significantly inhibited by FK506 at the concentration of 10(-8) to 10(-6) M in a dose dependent manner. Tacrolimus 67-72 interleukin 6 Homo sapiens 31-35 7528280-8 1994 In the preincubation study, FK506 required more than 12 h to inhibit IL-6 production by synovial cells. Tacrolimus 28-33 interleukin 6 Homo sapiens 69-73 7528280-9 1994 CONCLUSION: These results suggest that FK506 may be beneficial for patients with RA via inhibiting IL-6 production in inflammatory joints. Tacrolimus 39-44 interleukin 6 Homo sapiens 99-103 7520433-5 1994 Preincubation with the calcineurin inhibitors FK-506 or cyclosporin A strongly enhanced expression of NGFI-A and blocked transcription of NGFI-B, but it had no significant effect on Ca(2+)-stimulated transcription of c-fos. Tacrolimus 46-52 early growth response 1 Rattus norvegicus 102-108 7520433-5 1994 Preincubation with the calcineurin inhibitors FK-506 or cyclosporin A strongly enhanced expression of NGFI-A and blocked transcription of NGFI-B, but it had no significant effect on Ca(2+)-stimulated transcription of c-fos. Tacrolimus 46-52 nuclear receptor subfamily 4, group A, member 1 Rattus norvegicus 138-144 7520433-5 1994 Preincubation with the calcineurin inhibitors FK-506 or cyclosporin A strongly enhanced expression of NGFI-A and blocked transcription of NGFI-B, but it had no significant effect on Ca(2+)-stimulated transcription of c-fos. Tacrolimus 46-52 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 217-222 7520438-3 1994 The FKBP12.FK-506 complex inhibits calcineurin, a calcium-dependent phosphatase that is a component of the signal transduction pathway leading to early lymphokine gene transcription. Tacrolimus 11-17 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 4-10 7520438-8 1994 When complexed with FK-506, FKBP12.6 binds to and inhibits calcineurin, making it only the second FKBP discovered thus far to do so. Tacrolimus 20-26 FKBP prolyl isomerase 1B Homo sapiens 28-36 7520438-9 1994 The ability to inhibit calcineurin establishes the potential relevance of FKBP12.6 to the immunosuppressive or toxic side effects of FK-506. Tacrolimus 133-139 FKBP prolyl isomerase 1B Homo sapiens 74-82 7520353-5 1994 Both production and release of Il-1 alpha were markedly inhibited by FK506 (0.1 or 1 microM). Tacrolimus 69-74 interleukin 1 alpha Mus musculus 31-41 7520353-7 1994 It may be possible that the inhibition of IL-1 alpha production by FK506 is related to its anti-tumor-promoting action. Tacrolimus 67-72 interleukin 1 alpha Mus musculus 42-52 7798592-2 1994 Both methods were used to measure inhibition of proliferative responses of phytohaemagglutinin (PHA1)-stimulated human peripheral blood lymphocytes by tacrolimus (FK 506(1)), cyclosporine A (CsA1), rapamycin (RA1), dexamethasone (DEX1), prednisolone (PR1), and methylprednisolone (MP1). Tacrolimus 151-161 sodium channel epithelial 1 subunit gamma Homo sapiens 96-100 7519613-0 1994 Cloning and expression of cyclosporin A- and FK506-sensitive nuclear factor of activated T-cells: NF45 and NF90. Tacrolimus 45-50 interleukin enhancer binding factor 2 Homo sapiens 98-102 7519613-0 1994 Cloning and expression of cyclosporin A- and FK506-sensitive nuclear factor of activated T-cells: NF45 and NF90. Tacrolimus 45-50 interleukin enhancer binding factor 3 Homo sapiens 107-111 7519613-11 1994 Histidine-tagged NF45 and NF90 proteins, affinity-purified on nickel chelate columns, encode a NF-AT DNA-binding activity that is enhanced following T-cell stimulation, and this enhancement is blocked when T-cells are stimulated in the presence of cyclosporin A or FK506. Tacrolimus 265-270 interleukin enhancer binding factor 2 Homo sapiens 17-21 7519613-11 1994 Histidine-tagged NF45 and NF90 proteins, affinity-purified on nickel chelate columns, encode a NF-AT DNA-binding activity that is enhanced following T-cell stimulation, and this enhancement is blocked when T-cells are stimulated in the presence of cyclosporin A or FK506. Tacrolimus 265-270 interleukin enhancer binding factor 3 Homo sapiens 26-30 7518783-10 1994 Increased hepatic collagen and higher messenger RNA levels of transforming growth factor beta 1 and collagens I, III, and IV were found in the FK506-treated group. Tacrolimus 143-148 transforming growth factor, beta 1 Rattus norvegicus 62-95 7518783-11 1994 Rat fibroblasts treated with FK506 expressed higher levels of collagens I and III, fibronectin, macrophage-colony stimulating factor, tissue inhibitor of metalloprotease, and transforming growth factor beta 1 messenger RNAs. Tacrolimus 29-34 fibronectin 1 Rattus norvegicus 83-94 7798592-2 1994 Both methods were used to measure inhibition of proliferative responses of phytohaemagglutinin (PHA1)-stimulated human peripheral blood lymphocytes by tacrolimus (FK 506(1)), cyclosporine A (CsA1), rapamycin (RA1), dexamethasone (DEX1), prednisolone (PR1), and methylprednisolone (MP1). Tacrolimus 163-169 sodium channel epithelial 1 subunit gamma Homo sapiens 96-100 8046352-1 1994 The tumor necrosis factor alpha (TNF-alpha) gene is rapidly transcribed in activated T cells via a calcium-dependent pathway that does not require de novo protein synthesis, but is completely blocked by the immunosuppressive drugs cyclosporin A (CsA) and FK506. Tacrolimus 255-260 tumor necrosis factor Homo sapiens 4-31 8046352-1 1994 The tumor necrosis factor alpha (TNF-alpha) gene is rapidly transcribed in activated T cells via a calcium-dependent pathway that does not require de novo protein synthesis, but is completely blocked by the immunosuppressive drugs cyclosporin A (CsA) and FK506. Tacrolimus 255-260 tumor necrosis factor Homo sapiens 33-42 8046352-3 1994 The ability of analogues of CsA and FK506 to block calcineurin phosphatase activity correlates completely with their ability to inhibit induction of TNF-alpha mRNA, induction of a TNF-alpha promoter reporter plasmid in transiently transfected T cells, and induction of the kappa 3 binding factor in an electrophoretic mobility shift assay. Tacrolimus 36-41 tumor necrosis factor Homo sapiens 149-158 8046352-3 1994 The ability of analogues of CsA and FK506 to block calcineurin phosphatase activity correlates completely with their ability to inhibit induction of TNF-alpha mRNA, induction of a TNF-alpha promoter reporter plasmid in transiently transfected T cells, and induction of the kappa 3 binding factor in an electrophoretic mobility shift assay. Tacrolimus 36-41 tumor necrosis factor Homo sapiens 180-189 8046352-6 1994 Using the panel of CsA and FK506 analogues, we show that calcineurin participates in the induction of TNF-alpha transcription in activated B cells. Tacrolimus 27-32 tumor necrosis factor Homo sapiens 102-111 7518925-2 1994 Antibody directed at surface immunoglobulin (anti-Ig) on B cells has previously been shown to induce a rapid burst of TNF-alpha gene transcription, which can be blocked by the immunosuppressants cyclosporin A (CsA) and FK506. Tacrolimus 219-224 tumor necrosis factor Homo sapiens 118-127 7518461-1 1994 The immunosuppressant FK-506 (tacrolimus) forms a complex with a ubiquitous intracellular receptor, FK-506 binding protein (FKBP12), and this complex inhibits the heterodimeric Ca2+/calmodulin-dependent phosphatase, calcineurin, an essential component of the T-cell receptor signal transduction pathway. Tacrolimus 22-28 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 124-130 7518461-1 1994 The immunosuppressant FK-506 (tacrolimus) forms a complex with a ubiquitous intracellular receptor, FK-506 binding protein (FKBP12), and this complex inhibits the heterodimeric Ca2+/calmodulin-dependent phosphatase, calcineurin, an essential component of the T-cell receptor signal transduction pathway. Tacrolimus 30-40 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 124-130 7518461-6 1994 To assess the role of the autoinhibitory domain in regulating the interaction of CaN with the FK-506.FKBP12 complex, we reconstituted wild type and mutant phosphatase heterodimers using in vitro transcribed and translated subunits. Tacrolimus 94-100 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 101-107 7518461-7 1994 Association of the reconstituted calcineurin heterodimers with FKBP12 was dependent on FK-506. Tacrolimus 87-93 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 63-69 7518461-8 1994 In the case of the wild type heterodimer, association with the FK-506.FKBP12 complex was also dependent upon Ca2+; however, mutant catalytic subunits, in which the autoinhibitory domains were deleted, associated with the drug-binding protein complex in the presence of 10 mM EGTA. Tacrolimus 63-69 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 70-76 7518461-9 1994 These results indicate that the conserved autoinhibitory domain regulates both Ca(2+)-dependent phosphatase activity and association with the FK-506.FKBP12 complex. Tacrolimus 142-148 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 149-155 7518925-3 1994 Here, TNF-alpha gene transcription is shown also to be highly and rapidly induced in human B cells after stimulation via the CD40 and interleukin 4 pathways, which similarly is inhibited by CsA and a panel of CsA or FK506 analogues that block calcineurin phosphatase activity. Tacrolimus 216-221 tumor necrosis factor Homo sapiens 6-15 7518925-3 1994 Here, TNF-alpha gene transcription is shown also to be highly and rapidly induced in human B cells after stimulation via the CD40 and interleukin 4 pathways, which similarly is inhibited by CsA and a panel of CsA or FK506 analogues that block calcineurin phosphatase activity. Tacrolimus 216-221 CD40 molecule Homo sapiens 125-129 7518925-3 1994 Here, TNF-alpha gene transcription is shown also to be highly and rapidly induced in human B cells after stimulation via the CD40 and interleukin 4 pathways, which similarly is inhibited by CsA and a panel of CsA or FK506 analogues that block calcineurin phosphatase activity. Tacrolimus 216-221 interleukin 4 Homo sapiens 134-147 7518356-1 1994 The immunosuppressants rapamycin and FK506 bind to the same intracellular protein, the immunophilin FKBP12. Tacrolimus 37-42 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 100-106 8068174-1 1994 The transcription factor NF-ATp is a target in activated T cells for the calcium-regulated phosphatase calcineurin, and is therefore a secondary target for the immunosuppressive drugs cyclosporin A and FK506. Tacrolimus 202-207 nuclear factor of activated T cells 2 Homo sapiens 25-31 7813402-6 1994 For each isoform of the enzyme the peptidyl-prolyl cis/trans isomerase activity of the separated proteins was inhibited by cyclosporin A but was resistant toward FK 506. Tacrolimus 162-168 FKBP prolyl isomerase like Homo sapiens 35-70 7517981-5 1994 In regard to FK-506, we found that 1) FK-506 completely blocked the production of IL-2; 2) exogeneous IL-2 consistently restored the FK-506-induced inhibition; 3) FK-506 affected the phorbol myristate acetate-induced IL-2 responsiveness very little, if any; and 4) the significant suppression was observed only when FK-506 was added within 24 h after the initiation of culture. Tacrolimus 38-44 interleukin 2 Homo sapiens 82-86 7517981-5 1994 In regard to FK-506, we found that 1) FK-506 completely blocked the production of IL-2; 2) exogeneous IL-2 consistently restored the FK-506-induced inhibition; 3) FK-506 affected the phorbol myristate acetate-induced IL-2 responsiveness very little, if any; and 4) the significant suppression was observed only when FK-506 was added within 24 h after the initiation of culture. Tacrolimus 38-44 interleukin 2 Homo sapiens 82-86 7517981-5 1994 In regard to FK-506, we found that 1) FK-506 completely blocked the production of IL-2; 2) exogeneous IL-2 consistently restored the FK-506-induced inhibition; 3) FK-506 affected the phorbol myristate acetate-induced IL-2 responsiveness very little, if any; and 4) the significant suppression was observed only when FK-506 was added within 24 h after the initiation of culture. Tacrolimus 38-44 interleukin 2 Homo sapiens 82-86 7517981-5 1994 In regard to FK-506, we found that 1) FK-506 completely blocked the production of IL-2; 2) exogeneous IL-2 consistently restored the FK-506-induced inhibition; 3) FK-506 affected the phorbol myristate acetate-induced IL-2 responsiveness very little, if any; and 4) the significant suppression was observed only when FK-506 was added within 24 h after the initiation of culture. Tacrolimus 38-44 interleukin 2 Homo sapiens 82-86 7517981-7 1994 Conversely, FK-506 acts as a strong inhibitor of IL-2 production without a prominent effect on IL-2 responsiveness. Tacrolimus 12-18 interleukin 2 Homo sapiens 49-53 8008069-1 1994 The structurally related natural products rapamycin and FK506 bind to the same intracellular receptor, FKBP12, yet the resulting complexes interfere with distinct signalling pathways. Tacrolimus 56-61 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 103-109 7518678-1 1994 Tacrolimus(FK506) is a strong immuno-suppressant and shows its activity through inhibiting IL-2 mRNA transcription by forming pentameric complex with intracellular receptor(FK506 binding protein 12 kDa or FKBP12), Ca2+, calmodulin, and calcineurin. Tacrolimus 0-10 interleukin 2 Homo sapiens 91-95 7518678-1 1994 Tacrolimus(FK506) is a strong immuno-suppressant and shows its activity through inhibiting IL-2 mRNA transcription by forming pentameric complex with intracellular receptor(FK506 binding protein 12 kDa or FKBP12), Ca2+, calmodulin, and calcineurin. Tacrolimus 0-10 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 205-211 7518678-1 1994 Tacrolimus(FK506) is a strong immuno-suppressant and shows its activity through inhibiting IL-2 mRNA transcription by forming pentameric complex with intracellular receptor(FK506 binding protein 12 kDa or FKBP12), Ca2+, calmodulin, and calcineurin. Tacrolimus 11-16 interleukin 2 Homo sapiens 91-95 7518678-1 1994 Tacrolimus(FK506) is a strong immuno-suppressant and shows its activity through inhibiting IL-2 mRNA transcription by forming pentameric complex with intracellular receptor(FK506 binding protein 12 kDa or FKBP12), Ca2+, calmodulin, and calcineurin. Tacrolimus 11-16 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 205-211 7519528-11 1994 RESULTS: After 4-6 weeks of FK-506 therapy, human IgG, all thyroid antibodies and IFN-gamma were suppressed, while the levels remained elevated in the control group. Tacrolimus 28-34 immunoglobulin heavy variable V1-62 Mus musculus 50-53 7519528-11 1994 RESULTS: After 4-6 weeks of FK-506 therapy, human IgG, all thyroid antibodies and IFN-gamma were suppressed, while the levels remained elevated in the control group. Tacrolimus 28-34 interferon gamma Homo sapiens 82-91 7923750-4 1994 Cyclosporine and related agents such as FK-506 and rapamycin selectively inhibit adaptive immune responses by blocking T cell-dependent biosynthesis of lymphokines, particularly interleukin 2 at the level of messenger ribonucleic acid (mRNA) transcription. Tacrolimus 40-46 interleukin 2 Homo sapiens 178-191 7517078-0 1994 The direct effect of FK506 and rapamycin on interleukin 1(beta) and immunoglobulin production in vitro. Tacrolimus 21-26 interleukin 1 beta Homo sapiens 44-63 7514503-1 1994 FK506-binding protein (FKBP12) was originally identified as the cytosolic receptor for the immunosuppressant drugs FK506 and rapamycin. Tacrolimus 0-5 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 23-29 7524055-6 1994 Correlation of the 6 beta-testosterone hydroxylase activity with the FK 506 metabolite (M1) initial formation rate is consistent with the belief that CYP 3A isozymes are involved in FK 506 metabolism in male rats. Tacrolimus 69-75 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 150-156 7524055-6 1994 Correlation of the 6 beta-testosterone hydroxylase activity with the FK 506 metabolite (M1) initial formation rate is consistent with the belief that CYP 3A isozymes are involved in FK 506 metabolism in male rats. Tacrolimus 182-188 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 150-156 7584489-7 1994 These results suggest that combined treatment with FK506 and GC acts complexly to decrease rat CD4+8+ thymocytes and prevents thymocyte differentiation and maturation. Tacrolimus 51-56 Cd4 molecule Rattus norvegicus 95-98 7514503-5 1994 FK506 or rapamycin, inhibitors of FKBP12 isomerase activity, reverse these stabilizing effects. Tacrolimus 0-5 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 34-40 7514602-3 1994 Chemical cross-linking studies with disuccinimidyl suberate in the presence of either cyclophilin A, B, or C in complex with cyclosporin A or FK506 binding protein-FK506 result on the other hand in the apparently exclusive and strictly immunosuppressant-dependent formation of covalent immunophilin-calcineurin B subunit products. Tacrolimus 142-147 peptidyl-prolyl cis-trans isomerase A Bos taurus 86-99 7514602-3 1994 Chemical cross-linking studies with disuccinimidyl suberate in the presence of either cyclophilin A, B, or C in complex with cyclosporin A or FK506 binding protein-FK506 result on the other hand in the apparently exclusive and strictly immunosuppressant-dependent formation of covalent immunophilin-calcineurin B subunit products. Tacrolimus 164-169 peptidyl-prolyl cis-trans isomerase A Bos taurus 86-99 7514602-3 1994 Chemical cross-linking studies with disuccinimidyl suberate in the presence of either cyclophilin A, B, or C in complex with cyclosporin A or FK506 binding protein-FK506 result on the other hand in the apparently exclusive and strictly immunosuppressant-dependent formation of covalent immunophilin-calcineurin B subunit products. Tacrolimus 164-169 protein phosphatase 3 regulatory subunit B, alpha Bos taurus 299-312 7512379-1 1994 Backbone dynamics of the ligand- (FK506-) bound protein FKBP-12 (107 amino acids) have been examined using 15N relaxation data derived from inverse-detected two-dimensional 1H-15N NMR spectra. Tacrolimus 34-39 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 56-63 7521274-5 1994 Both CsA and FK506 (at 1 mumol/L) significantly inhibited nitrite production induced by recombinant murine interleukin-1 beta (rIL-1 beta). Tacrolimus 13-18 interleukin 1 beta Rattus norvegicus 127-137 7518966-2 1994 Nonspecific inhibitors of cytochrome P-450, such as ketoconazole, itraconazole, fluconazole and SKF 525 A, and most of the cytochrome P-450 IIIA specific substrates used in this study significantly inhibited FK 506 metabolism. Tacrolimus 208-214 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 26-42 7518966-2 1994 Nonspecific inhibitors of cytochrome P-450, such as ketoconazole, itraconazole, fluconazole and SKF 525 A, and most of the cytochrome P-450 IIIA specific substrates used in this study significantly inhibited FK 506 metabolism. Tacrolimus 208-214 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 123-139 7518966-4 1994 Cytochrome P-450 II substrates had minimal but significant effect on FK 506 metabolism. Tacrolimus 69-75 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 0-16 7518966-5 1994 This data supports our earlier observations that FK 506 metabolism is mediated predominantly by the steroid inducible cytochrome P-450 IIIA enzyme subfamily. Tacrolimus 49-55 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 118-134 7518966-6 1994 The results of this study indicate that in transplant patients there is a potential for an interaction of FK 506 with other drugs that are metabolized by the cytochrome P-450 IIIA subfamily or those that alter the activity of cytochrome P-450 IIIA subfamily. Tacrolimus 106-112 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 158-174 7518966-6 1994 The results of this study indicate that in transplant patients there is a potential for an interaction of FK 506 with other drugs that are metabolized by the cytochrome P-450 IIIA subfamily or those that alter the activity of cytochrome P-450 IIIA subfamily. Tacrolimus 106-112 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 226-242 7510691-0 1994 The interleukin-8 AP-1 and kappa B-like sites are genetic end targets of FK506-sensitive pathway accompanied by calcium mobilization. Tacrolimus 73-78 C-X-C motif chemokine ligand 8 Homo sapiens 4-17 7510691-2 1994 We observed that FK506 suppressed the transcription of a chemotactic cytokine, interleukin-8 (IL-8) in a human T cell line, Jurkat cells, activated by phorbol 12-myristate 13-acetate (PMA) and calcium (Ca2+) ionophore (ionomycin). Tacrolimus 17-22 C-X-C motif chemokine ligand 8 Homo sapiens 79-92 7510691-2 1994 We observed that FK506 suppressed the transcription of a chemotactic cytokine, interleukin-8 (IL-8) in a human T cell line, Jurkat cells, activated by phorbol 12-myristate 13-acetate (PMA) and calcium (Ca2+) ionophore (ionomycin). Tacrolimus 17-22 C-X-C motif chemokine ligand 8 Homo sapiens 94-98 7510691-6 1994 In contrast, FK506 or EGTA (Ca2+ chelator) similarly affected the formation of kappa B-like site binding complexes, which were not recognized by any antibodies against the human Rel family proteins (c-Rel, p65, p50, and p49). Tacrolimus 13-18 REL proto-oncogene, NF-kB subunit Homo sapiens 199-204 7510691-6 1994 In contrast, FK506 or EGTA (Ca2+ chelator) similarly affected the formation of kappa B-like site binding complexes, which were not recognized by any antibodies against the human Rel family proteins (c-Rel, p65, p50, and p49). Tacrolimus 13-18 RELA proto-oncogene, NF-kB subunit Homo sapiens 206-209 7510691-6 1994 In contrast, FK506 or EGTA (Ca2+ chelator) similarly affected the formation of kappa B-like site binding complexes, which were not recognized by any antibodies against the human Rel family proteins (c-Rel, p65, p50, and p49). Tacrolimus 13-18 nuclear factor kappa B subunit 1 Homo sapiens 211-214 7510691-6 1994 In contrast, FK506 or EGTA (Ca2+ chelator) similarly affected the formation of kappa B-like site binding complexes, which were not recognized by any antibodies against the human Rel family proteins (c-Rel, p65, p50, and p49). Tacrolimus 13-18 DNA primase subunit 1 Homo sapiens 220-223 7510691-7 1994 Furthermore, we confirmed the previous report that FK506 suppressed the PMA/ionomycin-induced activation through authentic kappa B site of immunoglobulin (Ig) gene, to which NF-kappa B binding was also decreased by FK506, indicating that both IL-8 kappa B-like site and Ig kappa B site are FK506-sensitive in spite of the difference of binding factors. Tacrolimus 51-56 nuclear factor kappa B subunit 1 Homo sapiens 174-184 7510691-7 1994 Furthermore, we confirmed the previous report that FK506 suppressed the PMA/ionomycin-induced activation through authentic kappa B site of immunoglobulin (Ig) gene, to which NF-kappa B binding was also decreased by FK506, indicating that both IL-8 kappa B-like site and Ig kappa B site are FK506-sensitive in spite of the difference of binding factors. Tacrolimus 51-56 C-X-C motif chemokine ligand 8 Homo sapiens 243-247 7510691-7 1994 Furthermore, we confirmed the previous report that FK506 suppressed the PMA/ionomycin-induced activation through authentic kappa B site of immunoglobulin (Ig) gene, to which NF-kappa B binding was also decreased by FK506, indicating that both IL-8 kappa B-like site and Ig kappa B site are FK506-sensitive in spite of the difference of binding factors. Tacrolimus 215-220 nuclear factor kappa B subunit 1 Homo sapiens 174-184 7510691-7 1994 Furthermore, we confirmed the previous report that FK506 suppressed the PMA/ionomycin-induced activation through authentic kappa B site of immunoglobulin (Ig) gene, to which NF-kappa B binding was also decreased by FK506, indicating that both IL-8 kappa B-like site and Ig kappa B site are FK506-sensitive in spite of the difference of binding factors. Tacrolimus 215-220 C-X-C motif chemokine ligand 8 Homo sapiens 243-247 7510691-7 1994 Furthermore, we confirmed the previous report that FK506 suppressed the PMA/ionomycin-induced activation through authentic kappa B site of immunoglobulin (Ig) gene, to which NF-kappa B binding was also decreased by FK506, indicating that both IL-8 kappa B-like site and Ig kappa B site are FK506-sensitive in spite of the difference of binding factors. Tacrolimus 215-220 nuclear factor kappa B subunit 1 Homo sapiens 174-184 7510691-7 1994 Furthermore, we confirmed the previous report that FK506 suppressed the PMA/ionomycin-induced activation through authentic kappa B site of immunoglobulin (Ig) gene, to which NF-kappa B binding was also decreased by FK506, indicating that both IL-8 kappa B-like site and Ig kappa B site are FK506-sensitive in spite of the difference of binding factors. Tacrolimus 215-220 C-X-C motif chemokine ligand 8 Homo sapiens 243-247 7510691-8 1994 Our results indicate that not only the reported IL-2 NF-AT and NFIL-2A sites and Ig kappa B site, but also the IL-8 AP-1 and kappa B-like sites are terminals of FK506-sensitive pathway involving Ca2+ mobilization. Tacrolimus 161-166 C-X-C motif chemokine ligand 8 Homo sapiens 111-115 7521274-5 1994 Both CsA and FK506 (at 1 mumol/L) significantly inhibited nitrite production induced by recombinant murine interleukin-1 beta (rIL-1 beta). Tacrolimus 13-18 interleukin 1 beta Mus musculus 107-125 7514263-2 1994 The macrolides FK506 and FK520 stimulate the Pgp-ATPase activity with affinities in the 100 nM range, nearly 10 times higher than that of verapamil, a well known Pgp substrate. Tacrolimus 15-20 ATP binding cassette subfamily B member 1 Homo sapiens 45-48 7514263-2 1994 The macrolides FK506 and FK520 stimulate the Pgp-ATPase activity with affinities in the 100 nM range, nearly 10 times higher than that of verapamil, a well known Pgp substrate. Tacrolimus 15-20 dynein axonemal heavy chain 8 Homo sapiens 49-55 7514263-2 1994 The macrolides FK506 and FK520 stimulate the Pgp-ATPase activity with affinities in the 100 nM range, nearly 10 times higher than that of verapamil, a well known Pgp substrate. Tacrolimus 15-20 ATP binding cassette subfamily B member 1 Homo sapiens 162-165 7511258-0 1994 Anti-CD2 monoclonal antibodies synergize with FK506 but not with cyclosporine or rapamycin to induce tolerance. Tacrolimus 46-51 CD2 molecule Homo sapiens 5-8 7511258-15 1994 This suggests that FK506 acts at a different locus in allograft immunity compared with the other immunosuppressants and this may be related to the alternative CD2 T cell activation pathway. Tacrolimus 19-24 CD2 molecule Homo sapiens 159-162 8137548-10 1994 Treatment with low-dose cyclosporin A (CsA) or FK506 in combination with BRC has proved more effective than either drug alone in suppression of T cell proliferation and CD25 antigen expression. Tacrolimus 47-52 interleukin 2 receptor subunit alpha Homo sapiens 169-173 7507662-0 1994 Catalytic and ligand binding properties of the FK506 binding protein FKBP12: effects of the single amino acid substitution of Tyr82 to Leu. Tacrolimus 47-52 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 69-75 7510408-2 1994 Changing tyrosine-82 to phenylalanine in FKBP-12 abolishes protein-ligand hydrogen bond interactions in the FKBP-12 complexes with tacrolimus or rapamycin and leads to a large apparent enthalpic stabilization of binding in both H2O and D2O. Tacrolimus 131-141 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 41-48 7510408-2 1994 Changing tyrosine-82 to phenylalanine in FKBP-12 abolishes protein-ligand hydrogen bond interactions in the FKBP-12 complexes with tacrolimus or rapamycin and leads to a large apparent enthalpic stabilization of binding in both H2O and D2O. Tacrolimus 131-141 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 108-115 8112299-3 1994 Calcineurin, a Ca2+/calmodulin-dependent protein phosphatase, is the FK-506- and CsA-sensitive enzyme required for TcR mediated activation of the IL-2 promoter. Tacrolimus 69-75 T cell receptor beta variable 20/OR9-2 (non-functional) Homo sapiens 115-118 8112299-3 1994 Calcineurin, a Ca2+/calmodulin-dependent protein phosphatase, is the FK-506- and CsA-sensitive enzyme required for TcR mediated activation of the IL-2 promoter. Tacrolimus 69-75 interleukin 2 Homo sapiens 146-150 7511990-2 1994 The immunosuppressant drug FK-506 effectively dissociates FKBP-12 from the calcium release channel of terminal cisternae (TC) vesicles. Tacrolimus 27-33 peptidyl-prolyl cis-trans isomerase FKBP1A Oryctolagus cuniculus 58-65 7507493-6 1994 Mutations in calcineurin A or B subunits or the inhibitory compounds FK506 and cyclosporin A restore growth of pmc1 mutants in high Ca2+ media. Tacrolimus 69-74 calcium-transporting ATPase PMC1 Saccharomyces cerevisiae S288C 111-115 7507662-1 1994 The binding of FK506 and rapamycin to their cytosolic receptor FKBP12 is an intermediate step in the paths leading to their potent immunosuppressive properties. Tacrolimus 15-20 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 63-69 7510997-3 1994 A recombinant p59/HBI-glutathione-S-transferase fusion protein, purified by Sephadex LH-20 filtration of tritiated drug-p59/HBI complexes, binds FK506 and RAP with apparent Kd values of 75 +/- 40 and 40 +/- 15 nM, respectively. Tacrolimus 145-150 FKBP prolyl isomerase 4 Homo sapiens 124-127 7512020-3 1994 CYP3A enzymes are thought to be responsible for metabolizing FK 506 in male rats. Tacrolimus 61-67 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 0-5 7512393-0 1994 Regulation of interleukin-5 production by peripheral blood mononuclear cells from atopic patients with FK506, cyclosporin A and glucocorticoid. Tacrolimus 103-108 interleukin 5 Homo sapiens 14-27 7512393-3 1994 IL-5 induction in vitro was completely inhibited by immunosuppressant FK506, cyclosporin A and dexamethasone. Tacrolimus 70-75 interleukin 5 Homo sapiens 0-4 7512393-4 1994 FK506 applied in vivo effectively suppressed clinical symptoms of atopic dermatitis and IL-5 production of PBMC. Tacrolimus 0-5 interleukin 5 Homo sapiens 88-92 7510997-0 1994 Effects of immunosuppressants FK506 and rapamycin on the heterooligomeric form of the progesterone receptor. Tacrolimus 30-35 progesterone receptor Homo sapiens 86-107 7887301-4 1994 The activation of calcineurin and the nuclear import of NF-ATc can both be blocked by cyclosporin A or FK506 in complex with their respective immunophilins. Tacrolimus 103-108 nuclear factor of activated T cells 1 Homo sapiens 56-62 7516291-0 1994 Evidence that FK506 alleviates ischemia/reperfusion injury to the rat liver: in vivo demonstration for suppression of TNF-a production in response to endotoxemia. Tacrolimus 14-19 tumor necrosis factor Rattus norvegicus 118-123 7507994-1 1994 The structurally related immunosuppressive macrolides FK-506 and rapamycin (RAP) exert distinct biological effects: inhibition of interleukin-2 production and inhibition of interleukin-2-induced proliferation, respectively, through binding to intracellular receptors, termed FKBPs. Tacrolimus 54-60 interleukin 2 Homo sapiens 130-143 7507994-1 1994 The structurally related immunosuppressive macrolides FK-506 and rapamycin (RAP) exert distinct biological effects: inhibition of interleukin-2 production and inhibition of interleukin-2-induced proliferation, respectively, through binding to intracellular receptors, termed FKBPs. Tacrolimus 54-60 interleukin 2 Homo sapiens 173-186 7510997-9 1994 Purification of cytosol PR on immobilized FK506 yields a 9S form still containing hsp90, hsp70 and p59/HBI associated to PR units. Tacrolimus 42-47 progesterone receptor Homo sapiens 24-26 7510997-9 1994 Purification of cytosol PR on immobilized FK506 yields a 9S form still containing hsp90, hsp70 and p59/HBI associated to PR units. Tacrolimus 42-47 heat shock protein 90 alpha family class A member 1 Homo sapiens 82-87 7510997-3 1994 A recombinant p59/HBI-glutathione-S-transferase fusion protein, purified by Sephadex LH-20 filtration of tritiated drug-p59/HBI complexes, binds FK506 and RAP with apparent Kd values of 75 +/- 40 and 40 +/- 15 nM, respectively. Tacrolimus 145-150 HLA complex P5B Homo sapiens 14-17 7510997-3 1994 A recombinant p59/HBI-glutathione-S-transferase fusion protein, purified by Sephadex LH-20 filtration of tritiated drug-p59/HBI complexes, binds FK506 and RAP with apparent Kd values of 75 +/- 40 and 40 +/- 15 nM, respectively. Tacrolimus 145-150 peptidyl-prolyl cis-trans isomerase FKBP4 Oryctolagus cuniculus 18-21 7526029-0 1994 Inhibitory effect of FK506 on intercellular adhesion molecule-1 (ICAM-1) expression on cultured thyroid cells. Tacrolimus 21-26 intercellular adhesion molecule 1 Homo sapiens 30-63 7510997-3 1994 A recombinant p59/HBI-glutathione-S-transferase fusion protein, purified by Sephadex LH-20 filtration of tritiated drug-p59/HBI complexes, binds FK506 and RAP with apparent Kd values of 75 +/- 40 and 40 +/- 15 nM, respectively. Tacrolimus 145-150 HLA complex P5B Homo sapiens 120-123 7510997-6 1994 In contrast 30 to 50% of the original 9S-PR species containing hsps and p59/HBI, was eluted in the absence of tungstate ions but after exposure of cytosol to 5 microM FK506 or RAP. Tacrolimus 167-172 progesterone receptor Homo sapiens 41-43 7510997-9 1994 Purification of cytosol PR on immobilized FK506 yields a 9S form still containing hsp90, hsp70 and p59/HBI associated to PR units. Tacrolimus 42-47 heat shock protein family A (Hsp70) member 4 Homo sapiens 89-94 7510997-9 1994 Purification of cytosol PR on immobilized FK506 yields a 9S form still containing hsp90, hsp70 and p59/HBI associated to PR units. Tacrolimus 42-47 HLA complex P5B Homo sapiens 99-102 7510997-9 1994 Purification of cytosol PR on immobilized FK506 yields a 9S form still containing hsp90, hsp70 and p59/HBI associated to PR units. Tacrolimus 42-47 FKBP prolyl isomerase 4 Homo sapiens 103-106 7510997-9 1994 Purification of cytosol PR on immobilized FK506 yields a 9S form still containing hsp90, hsp70 and p59/HBI associated to PR units. Tacrolimus 42-47 progesterone receptor Homo sapiens 121-123 7538609-3 1994 P-glycoprotein also transports MDR modulators such as cyclosporin A, FK506, and calcium channel blockers. Tacrolimus 69-74 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 7526029-0 1994 Inhibitory effect of FK506 on intercellular adhesion molecule-1 (ICAM-1) expression on cultured thyroid cells. Tacrolimus 21-26 intercellular adhesion molecule 1 Homo sapiens 65-71 7526029-1 1994 The effect of FK506, an immunosuppressive agent, on phytohemagglutinin (PHA) or interferon gamma (IFN gamma)-induced intercellular adhesion molecule-1 (ICAM-1) expression on cultured human thyroid cells from patients with Graves" disease was investigated. Tacrolimus 14-19 intercellular adhesion molecule 1 Homo sapiens 117-150 7526029-1 1994 The effect of FK506, an immunosuppressive agent, on phytohemagglutinin (PHA) or interferon gamma (IFN gamma)-induced intercellular adhesion molecule-1 (ICAM-1) expression on cultured human thyroid cells from patients with Graves" disease was investigated. Tacrolimus 14-19 intercellular adhesion molecule 1 Homo sapiens 152-158 7526029-2 1994 Primary cultured thyroid cells were incubated for three days with IFN gamma (10 to 800 U/ml) or PHA (1 to 50 micrograms/ml) in the presence of FK506. Tacrolimus 143-148 interferon gamma Homo sapiens 66-75 7526029-5 1994 FK506 inhibited the PHA-induced ICAM-1 expression in thyroid cells, but not the induction by IFN gamma. Tacrolimus 0-5 intercellular adhesion molecule 1 Homo sapiens 32-38 7526029-8 1994 This data indicates that the inhibitory effect of FK506 on ICAM-1 expression in primary cultured thyroid cells may be due to actions on infiltrating lymphocytes in the thyroid gland. Tacrolimus 50-55 intercellular adhesion molecule 1 Homo sapiens 59-65 7526029-9 1994 Further studies are necessary to elucidate whether the inhibition of ICAM-1 by FK506 results in the suppression of autoimmune reactions in the thyroid gland. Tacrolimus 79-84 intercellular adhesion molecule 1 Homo sapiens 69-75 7520521-6 1994 FK506, not cyclosporin A, was demonstrated for the first time in vivo to enhance GR and a hypothesis is proposed that FK506 might enhance GR and AP-1 signalings in a system reciprocally controlled by NO and H2O2. Tacrolimus 118-123 nuclear receptor subfamily 3, group C, member 1 Mus musculus 138-140 7520521-1 1994 An immunosuppressant FK506 binds with a component (hsp 56) of glucocorticoid receptor (GR) complex. Tacrolimus 21-26 nuclear receptor subfamily 3, group C, member 1 Mus musculus 62-85 8186461-2 1994 The expression of GM-CSF and IL-2 is inhibited by immunosuppressive drugs such as cyclosporin A (CsA) and FK506. Tacrolimus 106-111 colony stimulating factor 2 Homo sapiens 18-24 7520521-1 1994 An immunosuppressant FK506 binds with a component (hsp 56) of glucocorticoid receptor (GR) complex. Tacrolimus 21-26 nuclear receptor subfamily 3, group C, member 1 Mus musculus 87-89 7520521-6 1994 FK506, not cyclosporin A, was demonstrated for the first time in vivo to enhance GR and a hypothesis is proposed that FK506 might enhance GR and AP-1 signalings in a system reciprocally controlled by NO and H2O2. Tacrolimus 0-5 nuclear receptor subfamily 3, group C, member 1 Mus musculus 81-83 18475583-1 1994 The effect of FK506 and cyclosporin A (CsA) on the production of interleukin 6 (IL-6) in adherent monocytes was studied at a single-cell level by the avidinbiotin- peroxidase complex methods. Tacrolimus 14-19 interleukin 6 Homo sapiens 65-78 18475583-1 1994 The effect of FK506 and cyclosporin A (CsA) on the production of interleukin 6 (IL-6) in adherent monocytes was studied at a single-cell level by the avidinbiotin- peroxidase complex methods. Tacrolimus 14-19 interleukin 6 Homo sapiens 80-84 18475583-3 1994 Both FK506 and CsA enhanced the percentage of IL-6- producing monocytes stimulated with 100 pg/ml-1 mug/ml of LPS up to values near those obtained with 10 mug/ml of LPS. Tacrolimus 5-10 interleukin 6 Homo sapiens 46-50 18475583-8 1994 Moreover, pretreatment of monocytes with FK506 and CsA had a significant enhancing effect on LPS-induced IL-6 production, while treatment with FK506 or CsA after LPS stimulation had no effects on IL-6 production, suggesting that the enhancing effect of each drug is exerted before LPS stimulation or at an early stage of the post-receptor pathway after LPS stimulation. Tacrolimus 41-46 interleukin 6 Homo sapiens 105-109 18475583-9 1994 These experiments demonstrate that FK506 and CsA can selectively enhance IL-6 production in monocytes under certain conditions in vitro and, possibly, also in vivo. Tacrolimus 35-40 interleukin 6 Homo sapiens 73-77 8186461-2 1994 The expression of GM-CSF and IL-2 is inhibited by immunosuppressive drugs such as cyclosporin A (CsA) and FK506. Tacrolimus 106-111 interleukin 2 Homo sapiens 29-33 8302298-11 1994 The immunosuppressive drugs Cyclosporin A and FK506 completely blocked CD28 and CD3 mediated IL-2 production in these transfectants whereas rapamycin had only a partial inhibitory effect. Tacrolimus 46-51 CD28 molecule Homo sapiens 71-75 7539646-7 1994 In the histological examination, the congestion observed in the periportal region of the control group was mild, while there was less induction of ICAM-1 in the endothelial cells of the portal veins and hepatic veins in the FK506 group. Tacrolimus 224-229 intercellular adhesion molecule 1 Rattus norvegicus 147-153 8302298-11 1994 The immunosuppressive drugs Cyclosporin A and FK506 completely blocked CD28 and CD3 mediated IL-2 production in these transfectants whereas rapamycin had only a partial inhibitory effect. Tacrolimus 46-51 interleukin 2 Homo sapiens 93-97 7529381-0 1994 In-vitro effects of cyclosporin A, FK506, 6-mercaptopurine, and prednisolone on lymphokine-activated killer cells. Tacrolimus 35-40 interleukin 2 Homo sapiens 80-90 7517020-0 1994 FK 506 for vascular permeability factor production in minimal change nephrotic syndrome. Tacrolimus 0-6 vascular endothelial growth factor A Homo sapiens 11-39 11271304-5 1994 Thus, although FK 506 and cyclosporin inhibited calcium-dependent signalling to the IL-2 promoter via inhibition of the protein phosphatase calcineurin, it is possible that IL-2 induction via TCR/CD4 requires an FK 506 (and cyclosporin) sensitive step which is independent of cytoplasmic calcium changes. Tacrolimus 15-21 interleukin 2 Homo sapiens 84-88 7521743-3 1994 Continuous daily treatment of syngeneically reconstituted B10 mice with FK506 delayed the development of thymocytes from the CD4+CD8+ to CD4+CD8- stage, while no effect was observed at the earlier CD4-CD8- to CD4+CD8+ stage. Tacrolimus 72-77 granzyme C Mus musculus 58-61 7521743-3 1994 Continuous daily treatment of syngeneically reconstituted B10 mice with FK506 delayed the development of thymocytes from the CD4+CD8+ to CD4+CD8- stage, while no effect was observed at the earlier CD4-CD8- to CD4+CD8+ stage. Tacrolimus 72-77 CD4 antigen Mus musculus 125-128 7521743-3 1994 Continuous daily treatment of syngeneically reconstituted B10 mice with FK506 delayed the development of thymocytes from the CD4+CD8+ to CD4+CD8- stage, while no effect was observed at the earlier CD4-CD8- to CD4+CD8+ stage. Tacrolimus 72-77 CD4 antigen Mus musculus 137-140 7521743-3 1994 Continuous daily treatment of syngeneically reconstituted B10 mice with FK506 delayed the development of thymocytes from the CD4+CD8+ to CD4+CD8- stage, while no effect was observed at the earlier CD4-CD8- to CD4+CD8+ stage. Tacrolimus 72-77 CD4 antigen Mus musculus 137-140 7521743-3 1994 Continuous daily treatment of syngeneically reconstituted B10 mice with FK506 delayed the development of thymocytes from the CD4+CD8+ to CD4+CD8- stage, while no effect was observed at the earlier CD4-CD8- to CD4+CD8+ stage. Tacrolimus 72-77 CD4 antigen Mus musculus 137-140 7521743-7 1994 Levels of expression of V beta 5+ and V beta 11+ T cells in FK506-treated B10.BR-->B10.BR recipients were similar to those observed in unmanipulated, syngeneically reconstituted B10.BR-->B10.BR controls. Tacrolimus 60-65 granzyme C Mus musculus 74-77 7521743-7 1994 Levels of expression of V beta 5+ and V beta 11+ T cells in FK506-treated B10.BR-->B10.BR recipients were similar to those observed in unmanipulated, syngeneically reconstituted B10.BR-->B10.BR controls. Tacrolimus 60-65 granzyme C Mus musculus 86-89 7521743-7 1994 Levels of expression of V beta 5+ and V beta 11+ T cells in FK506-treated B10.BR-->B10.BR recipients were similar to those observed in unmanipulated, syngeneically reconstituted B10.BR-->B10.BR controls. Tacrolimus 60-65 granzyme C Mus musculus 86-89 7521743-7 1994 Levels of expression of V beta 5+ and V beta 11+ T cells in FK506-treated B10.BR-->B10.BR recipients were similar to those observed in unmanipulated, syngeneically reconstituted B10.BR-->B10.BR controls. Tacrolimus 60-65 granzyme C Mus musculus 86-89 7532330-3 1994 However, defects in the maturation of CD4+8+ cells to CD4+8- cells and of TCR alpha beta low MHC class I+ cells to TCR alpha beta high MHC class I+ cells were observed after FK506 administration. Tacrolimus 174-179 Cd4 molecule Rattus norvegicus 38-41 7532330-6 1994 These results suggest that the maturation of CD4-8- TCR alpha beta- MHC class I- cells to CD4+8+ TCR alpha beta low MHC class I+ cells may be FK506 resistant, whereas FK506 may interfere with the maturation of CD4+8+ TCR alpha beta low MHC class I+ thymocytes not only to CD4+8- TCR alpha beta high MHC class I+ cells but also to CD4-8+ TCR alpha beta high MHC class I+ cells. Tacrolimus 142-147 Cd4 molecule Rattus norvegicus 45-48 7532330-6 1994 These results suggest that the maturation of CD4-8- TCR alpha beta- MHC class I- cells to CD4+8+ TCR alpha beta low MHC class I+ cells may be FK506 resistant, whereas FK506 may interfere with the maturation of CD4+8+ TCR alpha beta low MHC class I+ thymocytes not only to CD4+8- TCR alpha beta high MHC class I+ cells but also to CD4-8+ TCR alpha beta high MHC class I+ cells. Tacrolimus 142-147 Cd4 molecule Rattus norvegicus 90-93 7532330-6 1994 These results suggest that the maturation of CD4-8- TCR alpha beta- MHC class I- cells to CD4+8+ TCR alpha beta low MHC class I+ cells may be FK506 resistant, whereas FK506 may interfere with the maturation of CD4+8+ TCR alpha beta low MHC class I+ thymocytes not only to CD4+8- TCR alpha beta high MHC class I+ cells but also to CD4-8+ TCR alpha beta high MHC class I+ cells. Tacrolimus 142-147 Cd4 molecule Rattus norvegicus 90-93 7532330-6 1994 These results suggest that the maturation of CD4-8- TCR alpha beta- MHC class I- cells to CD4+8+ TCR alpha beta low MHC class I+ cells may be FK506 resistant, whereas FK506 may interfere with the maturation of CD4+8+ TCR alpha beta low MHC class I+ thymocytes not only to CD4+8- TCR alpha beta high MHC class I+ cells but also to CD4-8+ TCR alpha beta high MHC class I+ cells. Tacrolimus 142-147 Cd4 molecule Rattus norvegicus 90-93 11271304-0 1994 FK 506 and cyclosporin each block antigen-induced T cell receptor signalling that is dependent on CD4 co-receptor and operates in the absence of detectable cytoplasmic calcium fluxes. Tacrolimus 0-6 CD4 molecule Homo sapiens 98-101 11271304-3 1994 Using 171, 171-CD4 (wild-type) and 171-CD4 (mutant), we found that IL-2 secretion was inhibited by FK 506 and cyclosporin but not by rapamycin. Tacrolimus 99-105 CD4 molecule Homo sapiens 15-18 11271304-3 1994 Using 171, 171-CD4 (wild-type) and 171-CD4 (mutant), we found that IL-2 secretion was inhibited by FK 506 and cyclosporin but not by rapamycin. Tacrolimus 99-105 CD4 molecule Homo sapiens 39-42 11271304-3 1994 Using 171, 171-CD4 (wild-type) and 171-CD4 (mutant), we found that IL-2 secretion was inhibited by FK 506 and cyclosporin but not by rapamycin. Tacrolimus 99-105 interleukin 2 Homo sapiens 67-71 11271305-0 1994 Effect of immunosuppressive agents FK 506 and cyclosporin and steroids on the expression of IL-6 and its receptor by stimulated lymphocytes and monocytes. Tacrolimus 35-41 interleukin 6 Homo sapiens 92-96 11271305-5 1994 Dexamethazone, cyclosporin (CyA), and FK 506 at immunosuppressive concentrations induced a dose-dependent inhibition of IL-6 secretion from adherent monocytes (MO) stimulated with phytohemagglutinin (PHA). Tacrolimus 38-44 interleukin 6 Homo sapiens 120-124 11271305-9 1994 Treatment of PHA-stimulated adherent MO with different concentrations of CyA and FK 506 induced a restoration of IL-6R expression. Tacrolimus 81-87 interleukin 6 receptor Homo sapiens 113-118 11271305-10 1994 FK 506 was 100 time more effective in restoring IL-6R than CyA. Tacrolimus 0-6 interleukin 6 receptor Homo sapiens 48-53 11271305-12 1994 FK 506, CyA, and steroids may exert their immunosuppressive effect by inhibiting IL-6 secretion and partially restoring MO IL-6R, which may be important in protecting the cell target against IL-6 autocrine stimulation. Tacrolimus 0-6 interleukin 6 Homo sapiens 81-85 11271305-12 1994 FK 506, CyA, and steroids may exert their immunosuppressive effect by inhibiting IL-6 secretion and partially restoring MO IL-6R, which may be important in protecting the cell target against IL-6 autocrine stimulation. Tacrolimus 0-6 interleukin 6 receptor Homo sapiens 123-128 11271305-12 1994 FK 506, CyA, and steroids may exert their immunosuppressive effect by inhibiting IL-6 secretion and partially restoring MO IL-6R, which may be important in protecting the cell target against IL-6 autocrine stimulation. Tacrolimus 0-6 interleukin 6 Homo sapiens 123-127 7532330-6 1994 These results suggest that the maturation of CD4-8- TCR alpha beta- MHC class I- cells to CD4+8+ TCR alpha beta low MHC class I+ cells may be FK506 resistant, whereas FK506 may interfere with the maturation of CD4+8+ TCR alpha beta low MHC class I+ thymocytes not only to CD4+8- TCR alpha beta high MHC class I+ cells but also to CD4-8+ TCR alpha beta high MHC class I+ cells. Tacrolimus 142-147 Cd48 molecule Rattus norvegicus 45-50 7522118-5 1993 Following incubation of this FK506-affinity purified 9S-PR with BF4, a specific anti-chick hsp90 monoclonal antibody, a shift of the [3H]Org 2058-PR complexes from 9S to 11S has been observed, indicating the presence of hsp90, hsp70 also is included in the 9S-PR complexes as demonstrated by Western blotting and density gradient experiments. Tacrolimus 29-34 heat shock protein family A (Hsp70) member 2 Gallus gallus 227-232 7532330-6 1994 These results suggest that the maturation of CD4-8- TCR alpha beta- MHC class I- cells to CD4+8+ TCR alpha beta low MHC class I+ cells may be FK506 resistant, whereas FK506 may interfere with the maturation of CD4+8+ TCR alpha beta low MHC class I+ thymocytes not only to CD4+8- TCR alpha beta high MHC class I+ cells but also to CD4-8+ TCR alpha beta high MHC class I+ cells. Tacrolimus 167-172 Cd4 molecule Rattus norvegicus 45-48 7532330-6 1994 These results suggest that the maturation of CD4-8- TCR alpha beta- MHC class I- cells to CD4+8+ TCR alpha beta low MHC class I+ cells may be FK506 resistant, whereas FK506 may interfere with the maturation of CD4+8+ TCR alpha beta low MHC class I+ thymocytes not only to CD4+8- TCR alpha beta high MHC class I+ cells but also to CD4-8+ TCR alpha beta high MHC class I+ cells. Tacrolimus 167-172 Cd4 molecule Rattus norvegicus 90-93 7532330-6 1994 These results suggest that the maturation of CD4-8- TCR alpha beta- MHC class I- cells to CD4+8+ TCR alpha beta low MHC class I+ cells may be FK506 resistant, whereas FK506 may interfere with the maturation of CD4+8+ TCR alpha beta low MHC class I+ thymocytes not only to CD4+8- TCR alpha beta high MHC class I+ cells but also to CD4-8+ TCR alpha beta high MHC class I+ cells. Tacrolimus 167-172 Cd4 molecule Rattus norvegicus 90-93 7532330-6 1994 These results suggest that the maturation of CD4-8- TCR alpha beta- MHC class I- cells to CD4+8+ TCR alpha beta low MHC class I+ cells may be FK506 resistant, whereas FK506 may interfere with the maturation of CD4+8+ TCR alpha beta low MHC class I+ thymocytes not only to CD4+8- TCR alpha beta high MHC class I+ cells but also to CD4-8+ TCR alpha beta high MHC class I+ cells. Tacrolimus 167-172 Cd4 molecule Rattus norvegicus 90-93 7532330-6 1994 These results suggest that the maturation of CD4-8- TCR alpha beta- MHC class I- cells to CD4+8+ TCR alpha beta low MHC class I+ cells may be FK506 resistant, whereas FK506 may interfere with the maturation of CD4+8+ TCR alpha beta low MHC class I+ thymocytes not only to CD4+8- TCR alpha beta high MHC class I+ cells but also to CD4-8+ TCR alpha beta high MHC class I+ cells. Tacrolimus 167-172 Cd48 molecule Rattus norvegicus 45-50 7509103-1 1993 We investigated the effects of FK506, a novel immunosuppressive agent, on the phytohemagglutinin (PHA) or interferon-gamma (IFN-gamma)-induced expression of HLA-DR antigen, accessory cell function and proliferation of primary cultured human thyroid cells. Tacrolimus 31-36 interferon gamma Homo sapiens 106-133 7505578-6 1993 This protochordate protein is substantially similar to 12-13 kDa FKBPs, most remarkably to one of the receptors that had been proposed to mediate the immunosuppressive actions of FK506, the human FKBP-13 (62% amino acid identity and 74% similarity). Tacrolimus 179-184 FKBP prolyl isomerase 2 Homo sapiens 196-203 7522118-0 1993 Immunological identification of a 50 kDa Mr FK506-binding immunophilin as a component of the non-DNA binding, hsp90 and hsp70 containing, heterooligomeric form of the chick oviduct progesterone receptor. Tacrolimus 44-49 heat shock protein family A (Hsp70) member 2 Gallus gallus 120-125 7522118-2 1993 P59/HBI belongs to the FKBP family since it binds the immunosuppressants FK506 and Rapamycin. Tacrolimus 73-78 peptidyl-prolyl cis-trans isomerase FKBP4 Oryctolagus cuniculus 0-3 7522118-2 1993 P59/HBI belongs to the FKBP family since it binds the immunosuppressants FK506 and Rapamycin. Tacrolimus 73-78 peptidyl-prolyl cis-trans isomerase FKBP4 Oryctolagus cuniculus 4-7 8251509-0 1993 Mechanism for the rotamase activity of FK506 binding protein from molecular dynamics simulations. Tacrolimus 39-44 FKBP prolyl isomerase 10 Homo sapiens 18-26 7510323-1 1993 The immunosuppressants FK506 and cyclosporin A (CsA) bound to their receptors, FKBP12 or cyclophilin, inhibit the Ca2+/calmodulin-dependent protein phosphatase, calcineurin, preventing T cell activation or, in yeast, recovery from alpha-mating factor arrest. Tacrolimus 23-28 peptidylprolyl isomerase FPR1 Saccharomyces cerevisiae S288C 79-85 7510323-7 1993 Overexpression of CNA1 or CNA2, in conjunction with CNB1, results in a significant decrease in hypersensitivity to FK506 and CsA. Tacrolimus 115-120 calcineurin catalytic subunit A Saccharomyces cerevisiae S288C 18-22 7510323-7 1993 Overexpression of CNA1 or CNA2, in conjunction with CNB1, results in a significant decrease in hypersensitivity to FK506 and CsA. Tacrolimus 115-120 calcineurin catalytic subunit A Saccharomyces cerevisiae S288C 26-30 7510323-7 1993 Overexpression of CNA1 or CNA2, in conjunction with CNB1, results in a significant decrease in hypersensitivity to FK506 and CsA. Tacrolimus 115-120 calcineurin regulatory subunit B Saccharomyces cerevisiae S288C 52-56 7506454-3 1993 In the present study, we have compared the effects of CsA and FK506 on glucose-stimulated insulin release from the isolated dog pancreatic islets, which have been maintained in culture for 3 days after isolation. Tacrolimus 62-67 insulin Homo sapiens 90-97 7510077-1 1993 The major immunophilins that bind cyclosporine (cyclophilin) and FK-506/rapamycin (FK-BP 12) have been well characterized. Tacrolimus 65-71 FKBP prolyl isomerase 1A Homo sapiens 83-91 7504525-0 1993 An active FK506-binding domain of 17,000 daltons is isolated following limited proteolysis of chicken thymus hsp56. Tacrolimus 10-15 FKBP prolyl isomerase 4 Homo sapiens 109-114 7504525-5 1993 We now report that hsp56 is also found to be a major immunophilin in chicken thymus, by virtue of binding to FK506-Affi-Gel-10 as well as positive cross-reactivity with a polyclonal antiserum directed against human hsp56. Tacrolimus 109-114 FKBP prolyl isomerase 4 Homo sapiens 19-24 7504525-7 1993 Peptide mapping provided additional proof that p17 is a fragment which comprises the entire FK506 binding domain I of chicken hsp56, terminating with an Arg-Lys which might represent a processing site. Tacrolimus 92-97 FKBP prolyl isomerase 4 Homo sapiens 126-131 7504525-11 1993 This work demonstrates the excision of a domain from an hsp56 protein that is active in binding FK506 and functionally distinct from FKBP-12, a protein of similar size and structure. Tacrolimus 96-101 FKBP prolyl isomerase 4 Homo sapiens 56-61 8397339-2 1993 NFAT, a DNA-binding protein required for interleukin-2 gene transcription, is a potential target for calcineurin, cyclosporin A and FK506. Tacrolimus 132-137 nuclear factor of activated T cells 2 Homo sapiens 0-4 7693698-4 1993 Both p50 and p54 bind an FK506 affinity resin at 0.5 M KCl, but only p50 binds efficiently in low salt conditions. Tacrolimus 25-30 nuclear factor kappa B subunit 1 Homo sapiens 5-8 7693698-4 1993 Both p50 and p54 bind an FK506 affinity resin at 0.5 M KCl, but only p50 binds efficiently in low salt conditions. Tacrolimus 25-30 FKBP prolyl isomerase 5 Homo sapiens 13-16 7693698-6 1993 The poor retention of p54 on FK506 resin at low ionic strength compared with the high retention of p50 suggests that these proteins may largely exist in separate complexes and may interact with other proteins, such as progesterone receptor, in distinctive manners. Tacrolimus 29-34 FKBP prolyl isomerase 5 Homo sapiens 22-25 8235597-2 1993 The DNA-binding specificity of NFAT is conferred by NFATp, a phosphoprotein that is a target for the immunosuppressive compounds cyclosporin A and FK506. Tacrolimus 147-152 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 2 Mus musculus 31-35 8235597-2 1993 The DNA-binding specificity of NFAT is conferred by NFATp, a phosphoprotein that is a target for the immunosuppressive compounds cyclosporin A and FK506. Tacrolimus 147-152 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 2 Mus musculus 52-57 7691614-6 1993 Induction of apoptosis by anti-CD2 mAb was prevented by cyclosporine A and FK 506. Tacrolimus 75-81 CD2 molecule Homo sapiens 31-34 7693684-3 1993 CsA and FK506 inhibited depolarization-induced glucagon gene transcription, FK506 being more potent than CsA. Tacrolimus 76-81 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 0-3 7693684-4 1993 CsA/FK506 responsiveness was mediated by the glucagon CRE and also by well characterized CREs of the choriogonadotropin and somatostatin genes. Tacrolimus 4-9 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 0-3 7693684-5 1993 Rapamycin antagonized the inhibitory effect of FK506 but not CsA, suggesting that FK506 and CsA may act through complex formation with distinct intracellular immunophilins. Tacrolimus 47-52 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 92-95 7693684-7 1993 These results demonstrate an inhibition by CsA and FK506 of CRE-mediated, calcium-induced transcription and suggest that membrane depolarization relies on calcineurin phosphatase activity for activation of CREB/CRE-mediated gene transcription. Tacrolimus 51-56 cAMP responsive element binding protein 1 Homo sapiens 206-210 7693684-8 1993 The interference with CRE-mediated gene transcription represents a novel mechanism of CsA/FK506 action, which may underlie pharmacological effects and toxic manifestations of these potent immunosuppressive drugs. Tacrolimus 90-95 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 86-89 7507316-4 1993 In contrast, rapamycin, a structural analogue of FK 506, interferes with the immune response at a different level, by blocking the response induced by cytokines such as IL-2. Tacrolimus 49-55 interleukin 2 Homo sapiens 169-173 7507316-6 1993 For instance, CsA and FK 506 inhibit the transcription of IL-3, IL-4, IFN gamma, TNF alpha or GM-CSF by activated T cells, and rapamycin has been shown to block the response to various growth factors such as IL-3, IL-4 or IL-6. Tacrolimus 22-28 interleukin 3 Homo sapiens 58-62 7507316-6 1993 For instance, CsA and FK 506 inhibit the transcription of IL-3, IL-4, IFN gamma, TNF alpha or GM-CSF by activated T cells, and rapamycin has been shown to block the response to various growth factors such as IL-3, IL-4 or IL-6. Tacrolimus 22-28 interleukin 4 Homo sapiens 64-68 7507316-6 1993 For instance, CsA and FK 506 inhibit the transcription of IL-3, IL-4, IFN gamma, TNF alpha or GM-CSF by activated T cells, and rapamycin has been shown to block the response to various growth factors such as IL-3, IL-4 or IL-6. Tacrolimus 22-28 interferon gamma Homo sapiens 70-79 7507316-6 1993 For instance, CsA and FK 506 inhibit the transcription of IL-3, IL-4, IFN gamma, TNF alpha or GM-CSF by activated T cells, and rapamycin has been shown to block the response to various growth factors such as IL-3, IL-4 or IL-6. Tacrolimus 22-28 tumor necrosis factor Homo sapiens 81-90 7507316-6 1993 For instance, CsA and FK 506 inhibit the transcription of IL-3, IL-4, IFN gamma, TNF alpha or GM-CSF by activated T cells, and rapamycin has been shown to block the response to various growth factors such as IL-3, IL-4 or IL-6. Tacrolimus 22-28 colony stimulating factor 2 Homo sapiens 94-100 7507316-6 1993 For instance, CsA and FK 506 inhibit the transcription of IL-3, IL-4, IFN gamma, TNF alpha or GM-CSF by activated T cells, and rapamycin has been shown to block the response to various growth factors such as IL-3, IL-4 or IL-6. Tacrolimus 22-28 interleukin 3 Homo sapiens 208-212 7507316-6 1993 For instance, CsA and FK 506 inhibit the transcription of IL-3, IL-4, IFN gamma, TNF alpha or GM-CSF by activated T cells, and rapamycin has been shown to block the response to various growth factors such as IL-3, IL-4 or IL-6. Tacrolimus 22-28 interleukin 4 Homo sapiens 214-218 7507316-6 1993 For instance, CsA and FK 506 inhibit the transcription of IL-3, IL-4, IFN gamma, TNF alpha or GM-CSF by activated T cells, and rapamycin has been shown to block the response to various growth factors such as IL-3, IL-4 or IL-6. Tacrolimus 22-28 interleukin 6 Homo sapiens 222-226 7693482-3 1993 Furthermore, other immunosuppressive agents FK506 and dexamethasone inhibit E-selectin expression, indicating that the enhancement observed in the presence of Cys A is not a consequence of general immunosuppression. Tacrolimus 44-49 selectin E Homo sapiens 76-86 7507077-1 1993 We have isolated a unique gene from a mouse JB6 epidermal cell cDNA expression library, termed FKBPRP, that codes for a protein having domains that share between 37 and 44% amino acid sequence identity and 60% similarity with members of the family of FK506-binding proteins. Tacrolimus 251-256 FK506 binding protein 10 Mus musculus 95-101 7509460-8 1993 These results support the notion that the interaction of drug-immunophilin complexes with calcineurin may be the molecular basis of cyclosporin A/FK506-induced inhibition of CREB/CRE-mediated gene transcription. Tacrolimus 146-151 cAMP responsive element binding protein 1 Rattus norvegicus 174-178 7509460-9 1993 The ability to interfere with CREB/CRE-mediated gene transcription represents a novel mechanism of cyclosporin A/FK506 action which may underlie pharmacological effects and toxic manifestations of these potent immunuosuppressive drugs. Tacrolimus 113-118 cAMP responsive element binding protein 1 Rattus norvegicus 30-34 7692637-12 1993 In conclusion, the need for insulin with FK-506 compares favorably to that of previous immunosuppressive regimens, and FK-506 may have a reversible diabetogenic effect that is not dose dependent. Tacrolimus 41-47 insulin Homo sapiens 28-35 7692640-2 1993 FK-506 is known to suppress the transcription of several genes encoding cytokines (e.g., IL-2, IFN-gamma) thought to play an important role in the allograft response. Tacrolimus 0-6 interleukin 2 Mus musculus 89-93 7692640-2 1993 FK-506 is known to suppress the transcription of several genes encoding cytokines (e.g., IL-2, IFN-gamma) thought to play an important role in the allograft response. Tacrolimus 0-6 interferon gamma Mus musculus 95-104 7692640-5 1993 Supernatants from Th2 cells treated with FK-506 showed marked suppression of IL-4 but only moderate suppression of IL-10 levels. Tacrolimus 41-47 interleukin 4 Mus musculus 77-81 7692640-5 1993 Supernatants from Th2 cells treated with FK-506 showed marked suppression of IL-4 but only moderate suppression of IL-10 levels. Tacrolimus 41-47 interleukin 10 Mus musculus 115-120 7692640-8 1993 Similar to results at the protein level, FK-506 suppressed steady state levels of IL-4 mRNA markedly but had a lesser effect on steady state levels of IL-10 mRNA. Tacrolimus 41-47 interleukin 4 Mus musculus 82-86 7692640-8 1993 Similar to results at the protein level, FK-506 suppressed steady state levels of IL-4 mRNA markedly but had a lesser effect on steady state levels of IL-10 mRNA. Tacrolimus 41-47 interleukin 10 Mus musculus 151-156 7692640-9 1993 Furthermore, FK-506 completely abrogated Con A-induced upregulation of IL-4 mRNA, but only slightly suppressed Con A-induced upregulation of IL-10 mRNA. Tacrolimus 13-19 interleukin 4 Mus musculus 71-75 7692640-9 1993 Furthermore, FK-506 completely abrogated Con A-induced upregulation of IL-4 mRNA, but only slightly suppressed Con A-induced upregulation of IL-10 mRNA. Tacrolimus 13-19 interleukin 10 Mus musculus 141-146 7692640-12 1993 Therefore, the ability of FK-506 to differentially suppress the mRNA levels of immunostimulatory cytokines such as IL-2, IL-4, and IFN-gamma more than the mRNA levels of the immunosuppressive cytokine IL-10 suggests that the effective immunosuppression seen in vivo with FK-506 may be a combination of these 2 effects. Tacrolimus 26-32 interleukin 2 Mus musculus 115-119 7692640-12 1993 Therefore, the ability of FK-506 to differentially suppress the mRNA levels of immunostimulatory cytokines such as IL-2, IL-4, and IFN-gamma more than the mRNA levels of the immunosuppressive cytokine IL-10 suggests that the effective immunosuppression seen in vivo with FK-506 may be a combination of these 2 effects. Tacrolimus 26-32 interleukin 4 Mus musculus 121-125 7692640-12 1993 Therefore, the ability of FK-506 to differentially suppress the mRNA levels of immunostimulatory cytokines such as IL-2, IL-4, and IFN-gamma more than the mRNA levels of the immunosuppressive cytokine IL-10 suggests that the effective immunosuppression seen in vivo with FK-506 may be a combination of these 2 effects. Tacrolimus 26-32 interferon gamma Mus musculus 131-140 7692640-12 1993 Therefore, the ability of FK-506 to differentially suppress the mRNA levels of immunostimulatory cytokines such as IL-2, IL-4, and IFN-gamma more than the mRNA levels of the immunosuppressive cytokine IL-10 suggests that the effective immunosuppression seen in vivo with FK-506 may be a combination of these 2 effects. Tacrolimus 26-32 interleukin 10 Mus musculus 201-206 8397339-2 1993 NFAT, a DNA-binding protein required for interleukin-2 gene transcription, is a potential target for calcineurin, cyclosporin A and FK506. Tacrolimus 132-137 interleukin 2 Homo sapiens 41-54 7689858-2 1993 Recently, we showed that p59 purified from human lymphocytes is an immunophilin (FKBP59) which binds both FK506 and rapamycin. Tacrolimus 106-111 HLA complex P5B Homo sapiens 25-28 7690248-1 1993 Backbone dynamics of the major tacrolimus (FK506) binding protein (FKBP-12, 107 amino acids) have been studied using 15N relaxation data derived from proton-detected two-dimensional 1H-15N NMR spectroscopy. Tacrolimus 31-41 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 67-74 7690248-1 1993 Backbone dynamics of the major tacrolimus (FK506) binding protein (FKBP-12, 107 amino acids) have been studied using 15N relaxation data derived from proton-detected two-dimensional 1H-15N NMR spectroscopy. Tacrolimus 43-48 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 67-74 7690315-8 1993 FK506 was found to inhibit the synthesis of IL3 and GM-CSF, whereas rapamycin failed to suppress the cytokine production of eosinophils. Tacrolimus 0-5 interleukin 3 Homo sapiens 44-47 7690315-8 1993 FK506 was found to inhibit the synthesis of IL3 and GM-CSF, whereas rapamycin failed to suppress the cytokine production of eosinophils. Tacrolimus 0-5 colony stimulating factor 2 Homo sapiens 52-58 8373400-3 1993 It binds the immunosuppressants FK506 and rapamycin and possesses three FKBP-12 (FK506 binding protein of M(r) 12,000)--like domains (I to III), plus a tail containing a putative calmodulin binding site (domain IV). Tacrolimus 32-37 calmodulin 1 Homo sapiens 179-189 7689858-2 1993 Recently, we showed that p59 purified from human lymphocytes is an immunophilin (FKBP59) which binds both FK506 and rapamycin. Tacrolimus 106-111 FKBP prolyl isomerase 4 Homo sapiens 81-87 7689858-4 1993 Here we provide evidence that rabbit uterine p59 also binds FK506 and rapamycin and that p59 or its homologue is associated with nontransformed progesterone receptors of rabbit uterus and chicken oviduct. Tacrolimus 60-65 peptidyl-prolyl cis-trans isomerase FKBP4 Oryctolagus cuniculus 45-48 7689858-8 1993 The association of p63 to the p59 complex was inhibited by FK506 and rapamycin, suggesting that p63 could be a potential target for the immunosuppressive actions of these two drugs. Tacrolimus 59-64 peptidyl-prolyl cis-trans isomerase FKBP4 Oryctolagus cuniculus 30-33 7687744-3 1993 We found that overexpression of cyclophilin A or B or FKBP12 increased T-cell sensitivity to CsA or FK506, respectively, demonstrating that they are able to mediate the inhibitory effects of their respective immunosuppressants in vivo. Tacrolimus 100-105 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 54-60 7687745-9 1993 FK506 induces GCN4 expression, which is normally induced by amino acid starvation. Tacrolimus 0-5 amino acid starvation-responsive transcription factor GCN4 Saccharomyces cerevisiae S288C 14-18 8377379-0 1993 CsA, FK506, corticosteroids and rapamycin inhibit TNF alpha production by cultured PTEC. Tacrolimus 5-10 tumor necrosis factor Homo sapiens 50-59 8377379-6 1993 FK506, corticosteroids and rapamycin also inhibited TNF alpha production in a dose dependent fashion, although not as effectively as CsA. Tacrolimus 0-5 tumor necrosis factor Homo sapiens 52-61 7687745-7 1993 Amino acid-auxotrophic yeast strains (trp1 his4 leu2) are FK506 sensitive, whereas prototrophic strains (TRP1 his4 leu2, trp1 HIS4 leu2, and trp1 his4 LEU2) are FK506 resistant. Tacrolimus 58-63 phosphoribosylanthranilate isomerase TRP1 Saccharomyces cerevisiae S288C 38-42 7685932-7 1993 FK506 and CsA-treated animals had better survival and diminished neutrophil liver infiltration, as well as MPO and MDA levels. Tacrolimus 0-5 myeloperoxidase Rattus norvegicus 107-110 8325502-8 1993 The single immunoblot-positive allele was found to contain a mutation altering a specific residue (Tyr89) which is conserved among the known FKBPs, and which, based on the solution and x-ray structures of human FKBP12, has been proposed to be part of a hydrophobic drug-binding pocket for FK506 and Rm. Tacrolimus 289-294 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 211-217 7687429-9 1993 Both immunosuppressive agents enhanced the calcium-dependent release of adrenocorticotropic hormone into the medium, once more, FK506 was 10-fold more potent than cyclosporin A. Tacrolimus 128-133 pro-opiomelanocortin-alpha Mus musculus 72-99 8335913-2 1993 NF-AT is thought to consist of two components: a ubiquitous, inducible nuclear component that we have identified as Fos and Jun proteins, and a preexisting, T cell-specific component (NF-ATp) which is the target for the immunosuppressive agents cyclosporin A (CsA) and FK506. Tacrolimus 269-274 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 2 Mus musculus 0-5 7689811-0 1993 FK-506 inhibits the IL-2-independent induction of the activation antigen CD 69. Tacrolimus 0-6 interleukin 2 Homo sapiens 20-24 7689811-0 1993 FK-506 inhibits the IL-2-independent induction of the activation antigen CD 69. Tacrolimus 0-6 CD69 molecule Homo sapiens 73-78 7684925-4 1993 Two largely nonpolar, structurally related macrolide ligands, tacrolimus (also known as FK506) and rapamycin, each bind with high affinity to a common site on a small FK506 binding protein (FKBP-12) and inhibit its peptidylprolyl cis-trans-isomerase activity. Tacrolimus 62-72 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 190-197 7684925-4 1993 Two largely nonpolar, structurally related macrolide ligands, tacrolimus (also known as FK506) and rapamycin, each bind with high affinity to a common site on a small FK506 binding protein (FKBP-12) and inhibit its peptidylprolyl cis-trans-isomerase activity. Tacrolimus 88-93 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 190-197 7685382-6 1993 FK-506-dependent MDA formation, studied only in the rat model, paralleled CyA-induced MDA formation but showed greater inhibition with CAT and less inhibition with SOD or GLUT. Tacrolimus 0-6 catalase Rattus norvegicus 135-138 7685949-0 1993 Effect of FK 506 and cyclosporine on the expression of IL-6 and its receptor on stimulated monocytes. Tacrolimus 10-16 interleukin 6 Homo sapiens 55-59 7683633-5 1993 These results suggest that the inhibitory effects of FK506 and CsA are independent of IL-2, and are associated with the reduction of c-myc and c-fos gene expression. Tacrolimus 53-58 MYC proto-oncogene, bHLH transcription factor Homo sapiens 133-138 7684380-8 1993 In addition, FK-506 complexes with FKBP12 proteins from several species all inhibit mammalian calcineurin. Tacrolimus 13-19 FKBP prolyl isomerase 1A Homo sapiens 35-41 7683633-3 1993 FK506 and CsA also reduced expression of the proto-oncogenes, c-myc and c-fos, but not c-jun and interleukin-2-receptor-alpha (IL-2R alpha) gene in H109 cells. Tacrolimus 0-5 MYC proto-oncogene, bHLH transcription factor Homo sapiens 62-67 7683633-3 1993 FK506 and CsA also reduced expression of the proto-oncogenes, c-myc and c-fos, but not c-jun and interleukin-2-receptor-alpha (IL-2R alpha) gene in H109 cells. Tacrolimus 0-5 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 72-77 7683633-5 1993 These results suggest that the inhibitory effects of FK506 and CsA are independent of IL-2, and are associated with the reduction of c-myc and c-fos gene expression. Tacrolimus 53-58 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 143-148 7680241-4 1993 Liquid-suspension-limiting dilution assay with IL-3 showed that FK506 directly stimulated the growth of blood progenitors in a dose-dependent manner with single-hit kinetics. Tacrolimus 64-69 interleukin 3 Homo sapiens 47-51 8475567-6 1993 Cyclosporine and FK506 had similar effects on the frequency of IL-5 gene expression in rejecting and nonrejecting allografts. Tacrolimus 17-22 interleukin 5 Homo sapiens 63-67 7682438-0 1993 FK506 binding to the 56-kilodalton immunophilin (Hsp56) in the glucocorticoid receptor heterocomplex has no effect on receptor folding or function. Tacrolimus 0-5 nuclear receptor subfamily 3, group C, member 1 Mus musculus 63-86 7682438-1 1993 It has recently been reported that the hsp56 component of glucocorticoid receptor heterocomplexes is an immunophilin of the FK506 binding class [Yem, A. W., Tomasselli, A. G., Heinrikson, R. L., Zurcher-Neely, H., Ruff, V. A., Johnson, R. A., & Deibel, M. R. (1992) J. Biol. Tacrolimus 124-129 nuclear receptor subfamily 3, group C, member 1 Mus musculus 58-81 7683935-0 1993 Reversion of the P-glycoprotein-mediated multidrug resistance of cancer cells by FK-506 derivatives. Tacrolimus 81-87 phosphoglycolate phosphatase Mus musculus 17-31 7683935-1 1993 FK-506 is a resistance-modulating agent (RMA) for tumor cells whose multidrug resistance (MDR) involves a P-glycoprotein (Pgp)-mediated anti-cancer drug efflux. Tacrolimus 0-6 phosphoglycolate phosphatase Mus musculus 106-120 7683935-1 1993 FK-506 is a resistance-modulating agent (RMA) for tumor cells whose multidrug resistance (MDR) involves a P-glycoprotein (Pgp)-mediated anti-cancer drug efflux. Tacrolimus 0-6 phosphoglycolate phosphatase Mus musculus 122-125 7683267-8 1993 Immunocytochemistry demonstrated that FK506 reduced the expression of interferon-gamma induced major histocompatibility complex (MHC) class I on the proximal tubular cells, but had no effect on the expression of the MHC class II antigens or the intercellular adhesion molecule (ICAM-1) on the cultured cells. Tacrolimus 38-43 interferon gamma Homo sapiens 70-86 7683267-8 1993 Immunocytochemistry demonstrated that FK506 reduced the expression of interferon-gamma induced major histocompatibility complex (MHC) class I on the proximal tubular cells, but had no effect on the expression of the MHC class II antigens or the intercellular adhesion molecule (ICAM-1) on the cultured cells. Tacrolimus 38-43 intercellular adhesion molecule 1 Homo sapiens 278-284 7681074-7 1993 The FK506-binding immunophilin FKBP12, as well as calcineurin, are shown to be present in these cells by immunoblotting analysis. Tacrolimus 4-9 FK506 binding protein 1a Mus musculus 18-37 7681058-0 1993 Potentiation of glucocorticoid receptor-mediated gene expression by the immunophilin ligands FK506 and rapamycin. Tacrolimus 93-98 nuclear receptor subfamily 3 group C member 1 Homo sapiens 16-39 7681058-2 1993 The ability of hsp56 to bind the immunosuppressive macrolide FK506 has led to the speculation that the steroid receptor and immunophilin signal transduction pathways are functionally interrelated. Tacrolimus 61-66 FKBP prolyl isomerase 4 Homo sapiens 15-20 7681058-3 1993 We have tested this idea by assessing the effects of FK506 on glucocorticoid receptor (GR)-mediated expression of the murine mammary tumor virus-chloramphenicol acetyltransferase (MMTV-CAT) reporter plasmid. Tacrolimus 53-58 nuclear receptor subfamily 3, group C, member 1 Mus musculus 62-85 7681058-3 1993 We have tested this idea by assessing the effects of FK506 on glucocorticoid receptor (GR)-mediated expression of the murine mammary tumor virus-chloramphenicol acetyltransferase (MMTV-CAT) reporter plasmid. Tacrolimus 53-58 nuclear receptor subfamily 3, group C, member 1 Mus musculus 87-89 7681058-7 1993 Finally, we provide evidence that FK506 potentiation of GR-mediated gene expression is the result of increased translocation to the nucleus of the GR. Tacrolimus 34-39 nuclear receptor subfamily 3 group C member 1 Homo sapiens 56-58 7681058-7 1993 Finally, we provide evidence that FK506 potentiation of GR-mediated gene expression is the result of increased translocation to the nucleus of the GR. Tacrolimus 34-39 nuclear receptor subfamily 3 group C member 1 Homo sapiens 147-149 7681059-0 1993 Human P-glycoprotein transports cyclosporin A and FK506. Tacrolimus 50-55 ATP binding cassette subfamily B member 1 Homo sapiens 6-20 7681059-4 1993 We were interested to determine whether cyclosporin A and FK506 are substrates for P-glycoprotein to transport, and we studied their transcellular transport. Tacrolimus 58-63 ATP binding cassette subfamily B member 1 Homo sapiens 83-97 7681059-10 1993 These results indicate that P-glycoprotein transports the immunosuppressive agents cyclosporin A and FK506. Tacrolimus 101-106 ATP binding cassette subfamily B member 1 Homo sapiens 28-42 7680241-5 1993 Liquid-suspension preincubation of blood cells with FK506 before culture in methylcellulose induced a significant increase in the amount of IL-3-supported growth of CFU-GM and BFU-E, whereas initial preincubation with IL-3 and subsequent culture with FK506 plus IL-3 exerted its stimulatory effect only on BFU-E. Tacrolimus 52-57 interleukin 3 Homo sapiens 140-144 7680241-5 1993 Liquid-suspension preincubation of blood cells with FK506 before culture in methylcellulose induced a significant increase in the amount of IL-3-supported growth of CFU-GM and BFU-E, whereas initial preincubation with IL-3 and subsequent culture with FK506 plus IL-3 exerted its stimulatory effect only on BFU-E. Tacrolimus 52-57 interleukin 3 Homo sapiens 218-222 7680241-5 1993 Liquid-suspension preincubation of blood cells with FK506 before culture in methylcellulose induced a significant increase in the amount of IL-3-supported growth of CFU-GM and BFU-E, whereas initial preincubation with IL-3 and subsequent culture with FK506 plus IL-3 exerted its stimulatory effect only on BFU-E. Tacrolimus 52-57 interleukin 3 Homo sapiens 218-222 7680241-5 1993 Liquid-suspension preincubation of blood cells with FK506 before culture in methylcellulose induced a significant increase in the amount of IL-3-supported growth of CFU-GM and BFU-E, whereas initial preincubation with IL-3 and subsequent culture with FK506 plus IL-3 exerted its stimulatory effect only on BFU-E. Tacrolimus 251-256 interleukin 3 Homo sapiens 140-144 7680613-2 1993 SEB-activated lamina propria T cells produced interleukin-2 and interferon-gamma and T cell activation was accompanied by tissue damage, which was inhibited by FK506. Tacrolimus 160-165 interferon gamma Homo sapiens 64-80 7680293-6 1993 FK 506 administration reduced markedly the incidence of infiltrating TCR alpha/beta +, CD5+, CD4+, CD8+, and LFA-1 beta + cells and the expression both of MHC class II antigens and ICAM-1 on MNC, endothelial cells and thyrocytes. Tacrolimus 0-6 similar to T cell receptor V-alpha J-alpha Rattus norvegicus 69-78 7680293-6 1993 FK 506 administration reduced markedly the incidence of infiltrating TCR alpha/beta +, CD5+, CD4+, CD8+, and LFA-1 beta + cells and the expression both of MHC class II antigens and ICAM-1 on MNC, endothelial cells and thyrocytes. Tacrolimus 0-6 Cd5 molecule Rattus norvegicus 87-90 7680293-6 1993 FK 506 administration reduced markedly the incidence of infiltrating TCR alpha/beta +, CD5+, CD4+, CD8+, and LFA-1 beta + cells and the expression both of MHC class II antigens and ICAM-1 on MNC, endothelial cells and thyrocytes. Tacrolimus 0-6 Cd4 molecule Rattus norvegicus 93-96 7680293-6 1993 FK 506 administration reduced markedly the incidence of infiltrating TCR alpha/beta +, CD5+, CD4+, CD8+, and LFA-1 beta + cells and the expression both of MHC class II antigens and ICAM-1 on MNC, endothelial cells and thyrocytes. Tacrolimus 0-6 intercellular adhesion molecule 1 Rattus norvegicus 181-187 7680293-10 1993 The results are consistent with a therapeutic effect of FK 506 mediated via interference with CD4+ T lymphocyte function and adhesion molecule-dependent cytotoxic effector mechanisms. Tacrolimus 56-62 Cd4 molecule Rattus norvegicus 94-97 7687472-4 1993 As previously found in other systems, induction of this lymphokine by ionomycin + PMA was not inhibited by RAP, although it was highly sensitive to inhibition by FK-506, an immunosuppressive macrolide structurally related to RAP. Tacrolimus 162-168 peroneal muscular atrophy Mus musculus 82-85 7687472-4 1993 As previously found in other systems, induction of this lymphokine by ionomycin + PMA was not inhibited by RAP, although it was highly sensitive to inhibition by FK-506, an immunosuppressive macrolide structurally related to RAP. Tacrolimus 162-168 regulatory associated protein of MTOR, complex 1 Mus musculus 225-228 7687472-5 1993 In contrast, the induction of the same lymphokine by IL-1 alpha + PMA was highly sensitive to RAP but resistant to FK-506. Tacrolimus 115-121 interleukin 1 alpha Mus musculus 53-63 7687472-5 1993 In contrast, the induction of the same lymphokine by IL-1 alpha + PMA was highly sensitive to RAP but resistant to FK-506. Tacrolimus 115-121 peroneal muscular atrophy Mus musculus 66-69 7687472-9 1993 While the inhibitory action of RAP could not be removed by extensive washing of the cells, it was readily reversed by a hundred-fold excess of FK-506 added to the cultures simultaneously with RAP or up to 16-18 h later. Tacrolimus 143-149 regulatory associated protein of MTOR, complex 1 Mus musculus 31-34 7687472-9 1993 While the inhibitory action of RAP could not be removed by extensive washing of the cells, it was readily reversed by a hundred-fold excess of FK-506 added to the cultures simultaneously with RAP or up to 16-18 h later. Tacrolimus 143-149 regulatory associated protein of MTOR, complex 1 Mus musculus 192-195 7687472-11 1993 The inhibition of lymphokine gene expression by RAP takes place at a late stage of the inductive response, and through a mechanism that involves interaction with the same cellular binding proteins as FK-506. Tacrolimus 200-206 regulatory associated protein of MTOR, complex 1 Mus musculus 48-51 7686193-8 1993 Based upon these data, it is probable that FK 506 attenuates hepatic ischemia/reperfusion injury, at least in part, by reducing TNF and IL-6 levels. Tacrolimus 43-49 tumor necrosis factor-like Rattus norvegicus 128-131 7683619-11 1993 Furthermore, the PCNA-labeling index 36 hrs after the partial hepatectomy was enhanced by the administration with FK-506 in the FHF mice. Tacrolimus 114-120 proliferating cell nuclear antigen Mus musculus 17-21 7686193-0 1993 FK 506 pre-treatment is associated with reduced levels of tumor necrosis factor and interleukin 6 following hepatic ischemia/reperfusion. Tacrolimus 0-6 tumor necrosis factor-like Rattus norvegicus 58-79 7683641-0 1993 FKBP12-FK506 complex inhibits phosphatase activity of two mammalian isoforms of calcineurin irrespective of their substrates or activation mechanisms. Tacrolimus 7-12 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 0-6 7683641-1 1993 The interaction of calcineurin (Ca2+/calmodulin-dependent protein phosphatase) with the potent immunosuppressive agent FK506 and its 12 kDa isoform binding protein (FKBP12) was investigated. Tacrolimus 119-124 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 165-171 7683641-2 1993 The FKBP12-FK506 complex inhibited the Ca2+/calmodulin-stimulated phosphatase activity of each of two calcineurin isoforms, which contain either the catalytic subunit A alpha or A beta (calcineurin A alpha or A beta) of bovine calcineurin. Tacrolimus 11-16 peptidyl-prolyl cis-trans isomerase FKBP1A Bos taurus 4-10 7683641-3 1993 Calcineurin phosphatase activity was inhibited by the FKBP12-FK506 complex irrespective of the substrate or the enzyme activation mechanism. Tacrolimus 61-66 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 54-60 7683641-4 1993 FK506 and FKBP-12 inhibited calcineurin in a concentration-dependent manner, and complete inhibition of the phosphatase activity appeared to require a molar excess of FKBP12-FK506 complex. Tacrolimus 174-179 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 10-17 7683641-4 1993 FK506 and FKBP-12 inhibited calcineurin in a concentration-dependent manner, and complete inhibition of the phosphatase activity appeared to require a molar excess of FKBP12-FK506 complex. Tacrolimus 174-179 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 167-173 7683641-6 1993 Direct binding studies with [3H]dihydro-FK506 suggest that the ratio of FKBP12-FK506 complex to calcineurin in vivo when IL2 production is inhibited is well correlated with the ratio when calcineurin phosphatase activity is inhibited in vitro. Tacrolimus 40-45 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 72-78 7683641-6 1993 Direct binding studies with [3H]dihydro-FK506 suggest that the ratio of FKBP12-FK506 complex to calcineurin in vivo when IL2 production is inhibited is well correlated with the ratio when calcineurin phosphatase activity is inhibited in vitro. Tacrolimus 40-45 interleukin 2 Homo sapiens 121-124 7683641-7 1993 These results suggest that calcineurin is a relevant cellular target of FK506 when bound to FKBP-12. Tacrolimus 72-77 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 92-99 7686193-0 1993 FK 506 pre-treatment is associated with reduced levels of tumor necrosis factor and interleukin 6 following hepatic ischemia/reperfusion. Tacrolimus 0-6 interleukin 6 Rattus norvegicus 84-97 7686193-8 1993 Based upon these data, it is probable that FK 506 attenuates hepatic ischemia/reperfusion injury, at least in part, by reducing TNF and IL-6 levels. Tacrolimus 43-49 interleukin 6 Rattus norvegicus 136-140 7682200-0 1993 Regulation of liver regeneration by interleukin-2 and its inhibitors: cyclosporine A and FK 506. Tacrolimus 89-95 interleukin 2 Mus musculus 36-49 7679116-6 1993 Moreover, dephosphorylation of NF-ATp in cell extracts is inhibited by prior treatment of T cells with the immunosuppressive drugs cyclosporin A or FK506, which inhibit the phosphatase activity of calcineurin when complexed with their specific binding proteins, cyclophilin and FK506-binding protein. Tacrolimus 148-153 nuclear factor of activated T cells 1 Bos taurus 31-37 7679116-7 1993 This work identifies NF-ATp as a DNA-binding phosphoprotein and a target for the drug/immunophilin/calcineurin complexes thought to mediate the inhibition of interleukin-2 gene induction by cyclosporin A and FK506. Tacrolimus 208-213 nuclear factor of activated T cells 1 Bos taurus 21-27 7679116-7 1993 This work identifies NF-ATp as a DNA-binding phosphoprotein and a target for the drug/immunophilin/calcineurin complexes thought to mediate the inhibition of interleukin-2 gene induction by cyclosporin A and FK506. Tacrolimus 208-213 interleukin 2 Bos taurus 158-171 7682200-6 1993 On the other hand, treatment with CsA and FK506, which inhibit IL-2 production, increased the mitotic indices of the regenerating livers. Tacrolimus 42-47 interleukin 2 Mus musculus 63-67 7682200-8 1993 Based on these results, CsA and FK506 would appear to stimulate liver cell proliferation by suppressing IL-2 production and inhibiting of NK cell activity. Tacrolimus 32-37 interleukin 2 Mus musculus 104-108 7679839-0 1993 Long-term insulin requirement after liver transplantation with FK 506 in American veterans. Tacrolimus 63-69 insulin Homo sapiens 10-17 7678400-4 1993 It was found that C-18 hydroxyl analogues of ascomycin, an analogue of FK-506 also called FR900520, bound tightly to immunophilin FKBP-12, but do not show any immunosuppressive activity in vitro or in vivo despite good bioavailability. Tacrolimus 71-77 FKBP prolyl isomerase 1A Homo sapiens 117-137 7678431-0 1993 Atomic structures of the human immunophilin FKBP-12 complexes with FK506 and rapamycin. Tacrolimus 67-72 FKBP prolyl isomerase 1A Homo sapiens 31-51 8419347-9 1993 The peptidyl prolyl isomerase hsp59 of the FK506 binding class is known to bind to hsp90. Tacrolimus 43-48 heat shock protein 90 alpha family class A member 1 Rattus norvegicus 83-88 7678431-1 1993 High resolution structures for the complexes formed by the immunosuppressive agents FK506 and rapamycin with the human immunophilin FKBP-12 have been determined by X-ray diffraction. Tacrolimus 84-89 FKBP prolyl isomerase 1A Homo sapiens 119-139 22827197-1 1993 In vitro studies of FK506 demonstrated that the agent inhibited the T-cell receptormediated signal transduction that results in the transcription of interleukin 2. Tacrolimus 20-25 interleukin 2 Rattus norvegicus 149-162 7678228-4 1993 In the present report we describe the effects of three immunosuppressive drugs, cyclosporin A (CsA), FK 506, and mycalamide A on mB7-mediated T cell activation. Tacrolimus 101-107 CD52 antigen Mus musculus 129-132 7678228-5 1993 The immunophilin ligands CsA and FK 506 block activation of murine CD4+ T cells by the combination of anti-CD3 mAb and CHO-mB7 cells but do not affect activation by CHO-mB7 cells and PMA. Tacrolimus 33-39 CD52 antigen Mus musculus 123-126 7678231-0 1993 Inhibition of activation-induced changes in the structure of the T cell interleukin-7 receptor by cyclosporin A and FK506. Tacrolimus 116-121 interleukin 7 receptor Homo sapiens 72-94 7678231-7 1993 In this study, the effect of the potent immunosuppressive agents cyclosporin A and FK506 on the activation-induced responsiveness to IL-7-driven proliferation and the concomitant changes in receptor structure have been investigated. Tacrolimus 83-88 interleukin 7 Homo sapiens 133-137 7678231-14 1993 As reported for IL-2R and IL-4R, our data also show that the expression of another T cell growth factor receptor is sensitive to the effects of cyclosporin A and FK506. Tacrolimus 162-167 interleukin 2 receptor subunit alpha Homo sapiens 16-21 7678231-14 1993 As reported for IL-2R and IL-4R, our data also show that the expression of another T cell growth factor receptor is sensitive to the effects of cyclosporin A and FK506. Tacrolimus 162-167 interleukin 4 receptor Homo sapiens 26-31 7509973-2 1993 This study was undertaken to examine the effect of FK 506 on stimulation of endothelin-1 (ET-1) mRNA and secretion of ET-1 in human endothelial cells (EC) compared with the effects of CyA. Tacrolimus 51-57 endothelin 1 Homo sapiens 76-88 7509973-2 1993 This study was undertaken to examine the effect of FK 506 on stimulation of endothelin-1 (ET-1) mRNA and secretion of ET-1 in human endothelial cells (EC) compared with the effects of CyA. Tacrolimus 51-57 endothelin 1 Homo sapiens 90-94 7509973-2 1993 This study was undertaken to examine the effect of FK 506 on stimulation of endothelin-1 (ET-1) mRNA and secretion of ET-1 in human endothelial cells (EC) compared with the effects of CyA. Tacrolimus 51-57 endothelin 1 Homo sapiens 118-122 7505632-0 1993 ICAM-1 and E-selectin expression in lesional biopsies of psoriasis patients responding to systemic FK 506 therapy. Tacrolimus 99-105 intercellular adhesion molecule 1 Homo sapiens 0-6 7505632-0 1993 ICAM-1 and E-selectin expression in lesional biopsies of psoriasis patients responding to systemic FK 506 therapy. Tacrolimus 99-105 selectin E Homo sapiens 11-21 7505632-5 1993 In contrast, the number of epidermal CD1+ (Langerhans) cells increased in response to FK 506 therapy. Tacrolimus 86-92 CD1c molecule Homo sapiens 37-40 7505632-8 1993 In response to FK 506 treatment, both ICAM-1 and E-selectin expression on blood vessels was reduced consistently but nevertheless persisted, even in individuals exhibiting total clearance of psoriatic lesions. Tacrolimus 15-21 intercellular adhesion molecule 1 Homo sapiens 38-44 7505632-8 1993 In response to FK 506 treatment, both ICAM-1 and E-selectin expression on blood vessels was reduced consistently but nevertheless persisted, even in individuals exhibiting total clearance of psoriatic lesions. Tacrolimus 15-21 selectin E Homo sapiens 49-59 1282000-3 1992 Using this and immunohistochemical staining, we found that the immunosuppressants cyclosporin A and FK506 decreased CD4 expression in cultured murine microglia without causing any significant decrease in cell viability. Tacrolimus 100-105 CD4 antigen Mus musculus 116-119 7678356-0 1993 Inhibition of glucose-stimulated insulin release from beta TC3 cells and rodent islets by an analog of FK506. Tacrolimus 103-108 insulin Homo sapiens 33-40 7678356-4 1993 We have studied the effects of an analog of a newer agent (FK506) termed L-683,590 on insulin secretion by an islet tumor line, beta TC3, and rat islets, and compared the effects of this drug to those of cyclosporine, since both cause similar immunosuppression. Tacrolimus 59-64 insulin Homo sapiens 86-93 7692621-8 1993 The analysis of lymphocyte subsets proved the decrease of W3/25: OX8 ratio in both FK506- and cyclosporin-treated groups. Tacrolimus 83-88 Cd4 molecule Rattus norvegicus 58-63 7507676-0 1993 Living related liver transplantation across ABO blood groups with FK506 and OKT3. Tacrolimus 66-71 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 44-47 7507676-14 1993 The present results suggest that graft livers from living related donors across ABO blood groups can function well with FK506, low-dose steroids, and prophylactic OKT3 without causing lethal complications. Tacrolimus 120-125 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 80-83 1281550-0 1992 Transcription of the tumor necrosis factor alpha gene is rapidly induced by anti-immunoglobulin and blocked by cyclosporin A and FK506 in human B cells. Tacrolimus 129-134 tumor necrosis factor Homo sapiens 21-48 1281550-5 1992 Moreover, induction of TNF-alpha gene transcription by anti-immunoglobulin was blocked by the immunosuppressants cyclosporin A and FK506. Tacrolimus 131-136 tumor necrosis factor Homo sapiens 23-32 1281562-6 1992 The damage induced by FK506 and CsA was characterized by a direct cytotoxic effect on tubular cells, as expressed by release of 3H thymidine from prelabeled cells, N-acetyl-beta-D-glucosaminidase release, and cell detachment. Tacrolimus 22-27 O-GlcNAcase Rattus norvegicus 164-195 1281562-12 1992 The concentrations of FK506 or CsA that induced ET-1 secretion were not cytolytic for tubular cells in vitro. Tacrolimus 22-27 endothelin 1 Rattus norvegicus 48-52 1281562-13 1992 FK506- or CsA-treated rats showed an increase in serum level of ET-1 in comparison with the control. Tacrolimus 0-5 endothelin 1 Rattus norvegicus 64-68 1279700-1 1992 Using an FK506 affinity column to identify mammalian immunosuppressant-binding proteins, we identified an immunophilin with an apparent M(r) approximately 55,000, which we have named FKBP52. Tacrolimus 9-14 FKBP prolyl isomerase 4 Homo sapiens 183-189 1279700-4 1992 Recombinant hFKBP52 has peptidyl-prolyl cis-trans isomerase activity that is inhibited by FK506 and rapamycin and an FKBP12-like consensus sequence that probably defines the immunosuppressant-binding site. Tacrolimus 90-95 FKBP prolyl isomerase 4 Homo sapiens 12-19 1383226-2 1992 FK-506 binds to a growing family of receptors termed FK-506-binding proteins (FKBPs), the most abundant being a 12-kDa cytosolic receptor, FKBP12. Tacrolimus 0-6 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 139-145 1383222-2 1992 Heat shock protein 56 (hsp56) has been shown to be involved in two cellular pathways, as an immunophilin for FK506 and as a component of steroid receptor complexes. Tacrolimus 109-114 FKBP prolyl isomerase 4 Homo sapiens 0-21 1383226-7 1992 The 110-kDa activity observed in brain extracts appears to be the FKBP12.FK-506.calcineurin (CaN) complex previously reported (Liu, J., Farmer, J., Lane, W., Friedman, J., Weissman, I., and Schreiber, S. (1991) Cell 66, 807-815) while the 110 kDa activity observed in JURKAT cells is a novel FK-506-binding protein. Tacrolimus 73-79 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 66-72 1383222-2 1992 Heat shock protein 56 (hsp56) has been shown to be involved in two cellular pathways, as an immunophilin for FK506 and as a component of steroid receptor complexes. Tacrolimus 109-114 FKBP prolyl isomerase 4 Homo sapiens 23-28 1383226-8 1992 Our characterization of the FKBP12.FK-506.CaN complex reveals a dependence upon calmodulin (CaM) for formation of the complex and demonstrates that the peptidyl-prolyl cis-trans isomerase (PPIase) activity of FKBP12 is not required for binding of FKBP12.FK-506 to CaN or for inhibition of CaN phosphatase activity. Tacrolimus 35-41 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 28-34 1383226-8 1992 Our characterization of the FKBP12.FK-506.CaN complex reveals a dependence upon calmodulin (CaM) for formation of the complex and demonstrates that the peptidyl-prolyl cis-trans isomerase (PPIase) activity of FKBP12 is not required for binding of FKBP12.FK-506 to CaN or for inhibition of CaN phosphatase activity. Tacrolimus 35-41 FKBP prolyl isomerase 5 Homo sapiens 189-195 1383226-8 1992 Our characterization of the FKBP12.FK-506.CaN complex reveals a dependence upon calmodulin (CaM) for formation of the complex and demonstrates that the peptidyl-prolyl cis-trans isomerase (PPIase) activity of FKBP12 is not required for binding of FKBP12.FK-506 to CaN or for inhibition of CaN phosphatase activity. Tacrolimus 35-41 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 209-215 1383226-8 1992 Our characterization of the FKBP12.FK-506.CaN complex reveals a dependence upon calmodulin (CaM) for formation of the complex and demonstrates that the peptidyl-prolyl cis-trans isomerase (PPIase) activity of FKBP12 is not required for binding of FKBP12.FK-506 to CaN or for inhibition of CaN phosphatase activity. Tacrolimus 35-41 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 209-215 1383226-8 1992 Our characterization of the FKBP12.FK-506.CaN complex reveals a dependence upon calmodulin (CaM) for formation of the complex and demonstrates that the peptidyl-prolyl cis-trans isomerase (PPIase) activity of FKBP12 is not required for binding of FKBP12.FK-506 to CaN or for inhibition of CaN phosphatase activity. Tacrolimus 254-260 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 28-34 1383226-8 1992 Our characterization of the FKBP12.FK-506.CaN complex reveals a dependence upon calmodulin (CaM) for formation of the complex and demonstrates that the peptidyl-prolyl cis-trans isomerase (PPIase) activity of FKBP12 is not required for binding of FKBP12.FK-506 to CaN or for inhibition of CaN phosphatase activity. Tacrolimus 254-260 FKBP prolyl isomerase 5 Homo sapiens 189-195 1281674-0 1992 FK506 and rapamycin selectively enhance degradation of IL-2 and GM-CSF mRNA. Tacrolimus 0-5 interleukin 2 Homo sapiens 55-59 1384129-4 1992 A similar reduction in motility on vitronectin occurred when cells were treated with the immunosuppressant FK506, which also inhibits calcineurin when bound to its binding protein, FKBP. Tacrolimus 107-112 vitronectin Homo sapiens 35-46 1382988-3 1992 We have previously shown that T cell receptor/CD3-mediated induction of apoptosis in a murine T cell hybridoma is inhibited by the immunosuppressive drugs cyclosporin A (CsA) and FK506. Tacrolimus 179-184 CD3 antigen, epsilon polypeptide Mus musculus 46-49 1281674-0 1992 FK506 and rapamycin selectively enhance degradation of IL-2 and GM-CSF mRNA. Tacrolimus 0-5 colony stimulating factor 2 Homo sapiens 64-70 1281674-2 1992 Using Northern analyses and promoter-reporter constructs we analyzed the transcriptional and posttranscriptional effects of FK506 and rapamycin on IL-2, GM-CSF, and IL-2R alpha gene expression. Tacrolimus 124-129 interleukin 2 Homo sapiens 147-151 1281674-2 1992 Using Northern analyses and promoter-reporter constructs we analyzed the transcriptional and posttranscriptional effects of FK506 and rapamycin on IL-2, GM-CSF, and IL-2R alpha gene expression. Tacrolimus 124-129 colony stimulating factor 2 Homo sapiens 153-159 1281674-2 1992 Using Northern analyses and promoter-reporter constructs we analyzed the transcriptional and posttranscriptional effects of FK506 and rapamycin on IL-2, GM-CSF, and IL-2R alpha gene expression. Tacrolimus 124-129 interleukin 2 receptor subunit alpha Homo sapiens 165-176 1281674-3 1992 FK506 completely inhibited activation of the IL-2 promoter, but only partially blocked GM-CSF promoter activity. Tacrolimus 0-5 interleukin 2 Homo sapiens 45-49 1355105-6 1992 Growth of the cells required activation because cyclosporin A and FK506 blocked stimulator cell-induced IL-2 production and proliferation as well as the spontaneous growth of the lines. Tacrolimus 66-71 interleukin 2 Homo sapiens 104-108 1381629-8 1992 This is supported by the ability of FK506 and rapamycin to directly compete the binding of the photoaffinity analogue 125I-iodoaryl azidoprazosin to the P-glycoprotein. Tacrolimus 36-41 ATP binding cassette subfamily B member 1 Homo sapiens 153-167 1385058-0 1992 Cytochrome P-450 3A enzymes are responsible for biotransformation of FK506 and rapamycin in man and rat. Tacrolimus 69-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 1385058-1 1992 The hepatic cytochrome P-450 responsible for metabolism of the structurally related macrolides FK506 and rapamycin in humans was identified using in vitro studies. Tacrolimus 95-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 1385058-7 1992 It is concluded that in human and rat liver FK506 and rapamycin are metabolized primarily by cytochrome P-450 3A4. Tacrolimus 44-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 1382293-1 1992 The inhibitory effects of cyclosporin A (CsA) and FK506 on Fc epsilon receptor type I-initiated increases in cytokine mRNA and the expression of their intracellular binding proteins were studied in interleukin 3 (IL-3)-dependent, mouse bone marrow-derived mast cells (BMMCs). Tacrolimus 50-55 interleukin 3 Mus musculus 198-217 1382293-9 1992 The resistance of BMMCs to inhibition of Fc epsilon receptor type I-mediated increases in cytokine mRNA by FK506 is most likely due to their deficiency of FKBP12 and the related inability to inhibit the activity of calcineurin. Tacrolimus 107-112 FK506 binding protein 1a Mus musculus 155-161 1380976-4 1992 FK-506 is known to interact with FK-binding protein-12 (FKBP-12), an abundant cytosolic protein with cis-trans peptidyl-prolyl isomerase activity (PPIase) activity. Tacrolimus 0-6 FKBP prolyl isomerase family member 6 Rattus norvegicus 147-153 1380976-6 1992 Recently, it was found that the complex of FKBP-12 with FK-506, but not with RAP, inhibits the phosphatase activity of calcineurin. Tacrolimus 56-62 FKBP prolyl isomerase 1A Rattus norvegicus 43-50 1380976-4 1992 FK-506 is known to interact with FK-binding protein-12 (FKBP-12), an abundant cytosolic protein with cis-trans peptidyl-prolyl isomerase activity (PPIase) activity. Tacrolimus 0-6 FKBP prolyl isomerase 1A Rattus norvegicus 33-54 1380976-4 1992 FK-506 is known to interact with FK-binding protein-12 (FKBP-12), an abundant cytosolic protein with cis-trans peptidyl-prolyl isomerase activity (PPIase) activity. Tacrolimus 0-6 FKBP prolyl isomerase 1A Rattus norvegicus 56-63 1379588-0 1992 Charged surface residues of FKBP12 participate in formation of the FKBP12-FK506-calcineurin complex. Tacrolimus 74-79 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 28-34 1384815-1 1992 The immunosuppressive drugs FK506 and cyclosporin A have an identical spectrum of activities with respect to IgE receptor (Fc epsilon RI)-mediated exocytosis from mast cells and T cell receptor-mediated transcription of IL-2. Tacrolimus 28-33 Fc receptor, IgE, high affinity I, alpha polypeptide Mus musculus 123-136 1384815-1 1992 The immunosuppressive drugs FK506 and cyclosporin A have an identical spectrum of activities with respect to IgE receptor (Fc epsilon RI)-mediated exocytosis from mast cells and T cell receptor-mediated transcription of IL-2. Tacrolimus 28-33 interleukin 2 Mus musculus 220-224 1379588-4 1992 We have identified, however, three FKBP12 surface residues (Asp-37, Arg-42, and His-87) proximal to a solvent-exposed segment of bound FK506 that may be direct contacts in the calcineurin complex. Tacrolimus 135-140 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 35-41 1379588-5 1992 Site-directed mutagenesis of two of these residues decreases the affinity of FKBP12-FK506 for calcineurin (Ki) from 6 nM for wild-type FKBP12 to 3.7 microM for a R42K/H87V double mutant, without affecting the peptidylprolyl isomerase activity or FK506 affinity of the mutant protein. Tacrolimus 84-89 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 77-83 1379588-5 1992 Site-directed mutagenesis of two of these residues decreases the affinity of FKBP12-FK506 for calcineurin (Ki) from 6 nM for wild-type FKBP12 to 3.7 microM for a R42K/H87V double mutant, without affecting the peptidylprolyl isomerase activity or FK506 affinity of the mutant protein. Tacrolimus 84-89 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 135-141 1379588-5 1992 Site-directed mutagenesis of two of these residues decreases the affinity of FKBP12-FK506 for calcineurin (Ki) from 6 nM for wild-type FKBP12 to 3.7 microM for a R42K/H87V double mutant, without affecting the peptidylprolyl isomerase activity or FK506 affinity of the mutant protein. Tacrolimus 246-251 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 77-83 1379588-6 1992 These FKBP12 mutations along with several substitutions on FK506 known to affect calcineurin binding form a roughly 100-A2 region of the FKBP12-FK506 complex surface that is likely to be within the calcineurin binding site. Tacrolimus 59-64 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 137-143 1379588-0 1992 Charged surface residues of FKBP12 participate in formation of the FKBP12-FK506-calcineurin complex. Tacrolimus 74-79 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 67-73 1379588-1 1992 The mechanism of FK506 immunosuppression has been proposed to proceed by formation of a tight-binding complex with the intracellular 12-kDa FK506-binding protein (FKBP12). Tacrolimus 17-22 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 163-169 1279908-3 1992 Yeast FKBP-13 is homologous to human FKBP-13 (52% amino acid identity) and to FKBP-12, the major cytosolic receptor for FK506. Tacrolimus 120-125 FKBP prolyl isomerase 2 Homo sapiens 6-13 1379588-6 1992 These FKBP12 mutations along with several substitutions on FK506 known to affect calcineurin binding form a roughly 100-A2 region of the FKBP12-FK506 complex surface that is likely to be within the calcineurin binding site. Tacrolimus 144-149 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 6-12 1379588-6 1992 These FKBP12 mutations along with several substitutions on FK506 known to affect calcineurin binding form a roughly 100-A2 region of the FKBP12-FK506 complex surface that is likely to be within the calcineurin binding site. Tacrolimus 144-149 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 137-143 1383612-0 1992 FK-506 inhibits proliferation and IL-4 messenger RNA production by a T-helper 2 cell line. Tacrolimus 0-6 interleukin 4 Homo sapiens 34-38 1383612-7 1992 FK-506 markedly inhibits Con A-induced proliferation and IL-4 mRNA production by the T-helper 2 cell line Ly1+2-/9. Tacrolimus 0-6 interleukin 4 Homo sapiens 57-61 1383612-8 1992 The ability of FK-506 to block the proliferation and IL-4 production by this helper cell subset suggests that this effect may contribute to its observed marked immunosuppressive properties in vitro and in vivo. Tacrolimus 15-21 interleukin 4 Homo sapiens 53-57 1380130-1 1992 The immunophilins cyclophilin and FK506 binding protein (FKBP) are small, predominantly soluble proteins that bind the immunosuppressant drugs cyclosporin A and FK506, respectively, with high affinity, and which seem to mediate their pharmacological actions. Tacrolimus 34-39 FK506-binding protein 12kD Drosophila melanogaster 57-61 1380130-2 1992 The Ca(2+)-dependent protein phosphatase, calcineurin, binds the cyclophilin-cyclosporin A and FKBP-FK506 complexes, indicating that calcineurin might mediate the actions of these drugs. Tacrolimus 100-105 FK506-binding protein 12kD Drosophila melanogaster 95-99 1279908-3 1992 Yeast FKBP-13 is homologous to human FKBP-13 (52% amino acid identity) and to FKBP-12, the major cytosolic receptor for FK506. Tacrolimus 120-125 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 78-85 1380951-3 1992 While 1 microgram/ml CSA and 0.1 microgram/ml FK 506 completely suppressed PHA-stimulated accumulation of mRNA for IL2, IL4, IL9, GM-CSF, TNF-alpha and IFN-gamma in PBMC, flu, keto and TGF-beta failed to inhibit any (except TNF-alpha blocked by TGF-beta). Tacrolimus 46-52 interleukin 2 Homo sapiens 115-118 1380951-3 1992 While 1 microgram/ml CSA and 0.1 microgram/ml FK 506 completely suppressed PHA-stimulated accumulation of mRNA for IL2, IL4, IL9, GM-CSF, TNF-alpha and IFN-gamma in PBMC, flu, keto and TGF-beta failed to inhibit any (except TNF-alpha blocked by TGF-beta). Tacrolimus 46-52 interleukin 4 Homo sapiens 120-123 1380951-3 1992 While 1 microgram/ml CSA and 0.1 microgram/ml FK 506 completely suppressed PHA-stimulated accumulation of mRNA for IL2, IL4, IL9, GM-CSF, TNF-alpha and IFN-gamma in PBMC, flu, keto and TGF-beta failed to inhibit any (except TNF-alpha blocked by TGF-beta). Tacrolimus 46-52 interleukin 9 Homo sapiens 125-128 1380951-3 1992 While 1 microgram/ml CSA and 0.1 microgram/ml FK 506 completely suppressed PHA-stimulated accumulation of mRNA for IL2, IL4, IL9, GM-CSF, TNF-alpha and IFN-gamma in PBMC, flu, keto and TGF-beta failed to inhibit any (except TNF-alpha blocked by TGF-beta). Tacrolimus 46-52 colony stimulating factor 2 Homo sapiens 130-136 1380951-3 1992 While 1 microgram/ml CSA and 0.1 microgram/ml FK 506 completely suppressed PHA-stimulated accumulation of mRNA for IL2, IL4, IL9, GM-CSF, TNF-alpha and IFN-gamma in PBMC, flu, keto and TGF-beta failed to inhibit any (except TNF-alpha blocked by TGF-beta). Tacrolimus 46-52 tumor necrosis factor Homo sapiens 138-147 1380951-3 1992 While 1 microgram/ml CSA and 0.1 microgram/ml FK 506 completely suppressed PHA-stimulated accumulation of mRNA for IL2, IL4, IL9, GM-CSF, TNF-alpha and IFN-gamma in PBMC, flu, keto and TGF-beta failed to inhibit any (except TNF-alpha blocked by TGF-beta). Tacrolimus 46-52 interferon gamma Homo sapiens 152-161 1380951-3 1992 While 1 microgram/ml CSA and 0.1 microgram/ml FK 506 completely suppressed PHA-stimulated accumulation of mRNA for IL2, IL4, IL9, GM-CSF, TNF-alpha and IFN-gamma in PBMC, flu, keto and TGF-beta failed to inhibit any (except TNF-alpha blocked by TGF-beta). Tacrolimus 46-52 transforming growth factor beta 1 Homo sapiens 185-193 1380951-3 1992 While 1 microgram/ml CSA and 0.1 microgram/ml FK 506 completely suppressed PHA-stimulated accumulation of mRNA for IL2, IL4, IL9, GM-CSF, TNF-alpha and IFN-gamma in PBMC, flu, keto and TGF-beta failed to inhibit any (except TNF-alpha blocked by TGF-beta). Tacrolimus 46-52 tumor necrosis factor Homo sapiens 224-233 1380951-3 1992 While 1 microgram/ml CSA and 0.1 microgram/ml FK 506 completely suppressed PHA-stimulated accumulation of mRNA for IL2, IL4, IL9, GM-CSF, TNF-alpha and IFN-gamma in PBMC, flu, keto and TGF-beta failed to inhibit any (except TNF-alpha blocked by TGF-beta). Tacrolimus 46-52 transforming growth factor beta 1 Homo sapiens 245-253 1375189-7 1992 As shown by both Western blots and FK506 binding activity, FKBP-12 was eluted only in the flow-through and wash fractions. Tacrolimus 35-40 FKBP prolyl isomerase 1A Homo sapiens 59-66 1375932-1 1992 Two FK506 binding proteins of molecular mass 12 kDa (FKBP12) and 13 kDa (FKBP13) have been identified as common cellular receptors of the immunosuppressants FK506 and rapamycin. Tacrolimus 4-9 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 53-59 1375932-1 1992 Two FK506 binding proteins of molecular mass 12 kDa (FKBP12) and 13 kDa (FKBP13) have been identified as common cellular receptors of the immunosuppressants FK506 and rapamycin. Tacrolimus 4-9 FKBP prolyl isomerase 2 Homo sapiens 73-79 1375932-1 1992 Two FK506 binding proteins of molecular mass 12 kDa (FKBP12) and 13 kDa (FKBP13) have been identified as common cellular receptors of the immunosuppressants FK506 and rapamycin. Tacrolimus 157-162 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 53-59 1375932-1 1992 Two FK506 binding proteins of molecular mass 12 kDa (FKBP12) and 13 kDa (FKBP13) have been identified as common cellular receptors of the immunosuppressants FK506 and rapamycin. Tacrolimus 157-162 FKBP prolyl isomerase 2 Homo sapiens 73-79 1375932-6 1992 The PPIase activity of FKBP25 was far more sensitive to inhibition by rapamycin (IC50 = 50 nM) than FK506 (IC50 = 400 nM). Tacrolimus 100-105 FKBP prolyl isomerase 3 Homo sapiens 23-29 1375932-7 1992 PPIase activity of 100 nM FKBP25 was almost completely inhibited by 150 nM rapamycin while only 90% inhibition was achieved by 4 microM FK506. Tacrolimus 136-141 FKBP prolyl isomerase 5 Homo sapiens 0-6 1375932-7 1992 PPIase activity of 100 nM FKBP25 was almost completely inhibited by 150 nM rapamycin while only 90% inhibition was achieved by 4 microM FK506. Tacrolimus 136-141 FKBP prolyl isomerase 3 Homo sapiens 26-32 1375932-8 1992 These data demonstrate that FKBP25 has a higher affinity for rapamycin than for FK506 and suggest that this cellular receptor may be an important target molecule for immunosuppression by rapamycin. Tacrolimus 80-85 FKBP prolyl isomerase 3 Homo sapiens 28-34 1377361-0 1992 FK-506- and CsA-sensitive activation of the interleukin-2 promoter by calcineurin. Tacrolimus 0-6 interleukin 2 Homo sapiens 44-57 1377361-2 1992 The immunosuppressive drugs, cyclosporin A (CsA) and FK-506, prevent T-cell proliferation by inhibiting a Ca(2+)-dependent event required for induction of interleukin-2 transcription. Tacrolimus 53-59 interleukin 2 Homo sapiens 155-168 1377361-6 1992 Here we report that transfection of a calcineurin catalytic subunit increases the 50% inhibitory concentration (IC50) of the immunosuppressants FK-506 and CsA, and that a mutant subunit acts in synergy with phorbol ester alone to activate the interleukin-2 promoter in a drug-sensitive manner. Tacrolimus 144-150 interleukin 2 Homo sapiens 243-256 1377606-3 1992 The structurally related drug FK506 had no effect on pp70S6K activation but at high concentrations reversed the rapamycin-induced block, confirming the requirement for the rapamycin and FK506 receptor, FKBP. Tacrolimus 30-35 FK506 binding protein 1a Mus musculus 202-206 1376361-3 1992 In this report, we examined the ability of rapamycin (RAP) to inhibit cytokine production and cytokine-induced proliferation by a factor-dependent murine mast cell line and compared its activity to that of the structurally related macrolide FK506. Tacrolimus 241-246 regulatory associated protein of MTOR, complex 1 Mus musculus 43-52 1376361-3 1992 In this report, we examined the ability of rapamycin (RAP) to inhibit cytokine production and cytokine-induced proliferation by a factor-dependent murine mast cell line and compared its activity to that of the structurally related macrolide FK506. Tacrolimus 241-246 regulatory associated protein of MTOR, complex 1 Mus musculus 54-57 1376361-8 1992 Because RAP and FK506 are structurally related and yet have divergent biological effects, we examined the ability of RAP to antagonize inhibitory effects of FK506 on mast cell cytokine production and the ability of FK506 to antagonize inhibitory effects of RAP on cytokine-induced mast cell proliferation. Tacrolimus 157-162 regulatory associated protein of MTOR, complex 1 Mus musculus 117-120 1376361-8 1992 Because RAP and FK506 are structurally related and yet have divergent biological effects, we examined the ability of RAP to antagonize inhibitory effects of FK506 on mast cell cytokine production and the ability of FK506 to antagonize inhibitory effects of RAP on cytokine-induced mast cell proliferation. Tacrolimus 157-162 regulatory associated protein of MTOR, complex 1 Mus musculus 117-120 1376361-8 1992 Because RAP and FK506 are structurally related and yet have divergent biological effects, we examined the ability of RAP to antagonize inhibitory effects of FK506 on mast cell cytokine production and the ability of FK506 to antagonize inhibitory effects of RAP on cytokine-induced mast cell proliferation. Tacrolimus 157-162 regulatory associated protein of MTOR, complex 1 Mus musculus 117-120 1376361-8 1992 Because RAP and FK506 are structurally related and yet have divergent biological effects, we examined the ability of RAP to antagonize inhibitory effects of FK506 on mast cell cytokine production and the ability of FK506 to antagonize inhibitory effects of RAP on cytokine-induced mast cell proliferation. Tacrolimus 157-162 regulatory associated protein of MTOR, complex 1 Mus musculus 117-120 1376361-9 1992 The addition of RAP in molar excess reversed inhibition of mast cell cytokine production mediated by FK506, but not that of CSA. Tacrolimus 101-106 regulatory associated protein of MTOR, complex 1 Mus musculus 16-19 1375751-3 1992 Two largely nonpolar, immunosuppressive agents, FK506 and rapamycin, each bind with high affinity to a common hydrophobic pocket on a small peptidylproline cis-trans isomerase known as FK506 binding protein (FKBP-12) and inhibit its activity. Tacrolimus 48-53 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 208-215 1375751-4 1992 In an effort to elucidate the structural features of these ligands responsible for the observed energetics, we have undertaken an investigation of the thermodynamics of binding of FK506 and rapamycin to FKBP-12. Tacrolimus 180-185 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 203-210 1375751-6 1992 By analyzing the distribution of changes in solvent-accessible surface area upon binding of FK506 to FKBP-12 from crystallographic data, it is found that 99% of the net surface buried upon binding involves nonpolar groups. Tacrolimus 92-97 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 101-108 1376003-3 1992 A 59-kilodalton member of the FK506- and rapamycin-binding class was found to associate in the absence of these drugs with two heat shock proteins (hsp90 and hsp70) and the glucocorticoid receptor (GR). Tacrolimus 30-35 heat shock protein 90 alpha family class A member 1 Homo sapiens 148-153 1604471-14 1992 The hypersecretory reaction of the beta cell may be of help in further investigations of mechanisms of CsA- and FK506-induced inhibition of insulin release. Tacrolimus 112-117 insulin Canis lupus familiaris 140-147 1376003-3 1992 A 59-kilodalton member of the FK506- and rapamycin-binding class was found to associate in the absence of these drugs with two heat shock proteins (hsp90 and hsp70) and the glucocorticoid receptor (GR). Tacrolimus 30-35 heat shock protein family A (Hsp70) member 4 Homo sapiens 158-163 1376003-3 1992 A 59-kilodalton member of the FK506- and rapamycin-binding class was found to associate in the absence of these drugs with two heat shock proteins (hsp90 and hsp70) and the glucocorticoid receptor (GR). Tacrolimus 30-35 nuclear receptor subfamily 3 group C member 1 Homo sapiens 173-196 1376003-3 1992 A 59-kilodalton member of the FK506- and rapamycin-binding class was found to associate in the absence of these drugs with two heat shock proteins (hsp90 and hsp70) and the glucocorticoid receptor (GR). Tacrolimus 30-35 nuclear receptor subfamily 3 group C member 1 Homo sapiens 198-200 1373887-5 1992 Both CsA and FK 506 specifically inhibit cellular calcineurin at drug concentrations that inhibit interleukin 2 production in activated T cells. Tacrolimus 13-19 interleukin 2 Homo sapiens 98-111 1375171-0 1992 Solution structure of FK506 bound to FKBP-12. Tacrolimus 22-27 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 37-44 1375171-1 1992 The complex of the immunosuppressant FK506 bound to FKBP-12 has been studied in solution using 1H and inverse-detected 13C NMR methods. Tacrolimus 37-42 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 52-59 1374404-4 1992 In humans, FK506 binds to the 12-kDa FK506-binding protein (FKBP12) and blocks calcium-dependent T cell activation. Tacrolimus 11-16 FKBP prolyl isomerase 1A Homo sapiens 30-58 1374404-4 1992 In humans, FK506 binds to the 12-kDa FK506-binding protein (FKBP12) and blocks calcium-dependent T cell activation. Tacrolimus 11-16 FKBP prolyl isomerase 1A Homo sapiens 60-66 1374947-0 1992 FK506 as an agonist to induce inhibition of interleukin 2 production. Tacrolimus 0-5 interleukin 2 Homo sapiens 44-57 1375473-8 1992 However, nonimmunosuppressive analogs of CsA and FK-506 are also inhibitory, indicating that inhibition of PPIase activity is not directly implicated in immunosuppression. Tacrolimus 49-55 FKBP prolyl isomerase 5 Homo sapiens 107-113 1374948-8 1992 FK506 pretreatment significantly reduced the serum levels of BUN (P less than 0.02), creatinine (P less than 0.02), and TNF (P less than 0.05) as compared with that seen in controls. Tacrolimus 0-5 tumor necrosis factor Rattus norvegicus 120-123 1374948-11 1992 Based upon these data, it can be concluded that (1) FK506 pretreatment ameliorates the functional renal injury associated with I/R, (2) renal ischemia induces the release of TNF, and (3) FK506 pretreatment results in a significant inhibition of TNF production. Tacrolimus 52-57 tumor necrosis factor Rattus norvegicus 174-177 1374948-11 1992 Based upon these data, it can be concluded that (1) FK506 pretreatment ameliorates the functional renal injury associated with I/R, (2) renal ischemia induces the release of TNF, and (3) FK506 pretreatment results in a significant inhibition of TNF production. Tacrolimus 52-57 tumor necrosis factor Rattus norvegicus 245-248 1371698-4 1992 FKBP25 displays the rotamase activity characteristic of FKBPs; the activity is inhibited by the immunosuppressants rapamycin (Ki = 0.9 nM) and FK506 (Ki = 160 nM), but not cyclosporin A. Tacrolimus 143-148 FKBP prolyl isomerase 3 Bos taurus 0-6 1378764-0 1992 Cyclosporin A and FK506 prevent the derepression of the IL-2 gene in mitogen-induced primary T lymphocytes. Tacrolimus 18-23 interleukin 2 Homo sapiens 56-60 1378764-3 1992 Cyclosporin A (CsA) and FK506 interfere with normal derepression of the IL-2 gene. Tacrolimus 24-29 interleukin 2 Homo sapiens 72-76 1371623-0 1992 Blockade of interleukin-2 production from cloned T cells by cyclosporine and FK 506 assessed by proliferation assays in vitro. Tacrolimus 77-83 interleukin 2 Homo sapiens 12-25 1371624-0 1992 Enhanced liver regeneration by FK 506 can be blocked by interleukin-1 alpha and interleukin-2. Tacrolimus 31-37 interleukin 1 alpha Homo sapiens 56-75 1371624-0 1992 Enhanced liver regeneration by FK 506 can be blocked by interleukin-1 alpha and interleukin-2. Tacrolimus 31-37 interleukin 2 Homo sapiens 80-93 1380242-3 1992 Cyclosporin A (CsA) and FK506 inhibit the production of IL-2 in T lymphocytes at the level of gene transcription. Tacrolimus 24-29 interleukin 2 Homo sapiens 56-60 1380242-6 1992 However, CsA and FK506 inhibit the appearance of DNA binding activity of factors that bind to the NF-AT and AP-1 sites in the IL-2 enhancer. Tacrolimus 17-22 interleukin 2 Homo sapiens 126-130 1380242-7 1992 Since the induction of NF-AT and AP-1 is induced by the same stimuli that stimulate IL-2 production, these results indicate that the immunosuppressant action of CsA and FK506 is exerted at the level of these trans-activating factors. Tacrolimus 169-174 interleukin 2 Homo sapiens 84-88 1371107-0 1992 The Hsp56 component of steroid receptor complexes binds to immobilized FK506 and shows homology to FKBP-12 and FKBP-13. Tacrolimus 71-76 FKBP prolyl isomerase 4 Homo sapiens 4-9 1371107-5 1992 It would appear, therefore, that this 60-kDa protein, or as we refer to it provisionally, "Hsp56," could have the capacity to bind FK506 directly. Tacrolimus 131-136 FKBP prolyl isomerase 4 Homo sapiens 91-96 1371117-1 1992 FK506-binding protein (FKBP) catalyzes the cis-trans isomerization of the peptidyl-prolyl amide bond (the PPIase reaction) and is the major intracellular receptor for the immunosuppressive drugs FK506 and rapamycin. Tacrolimus 0-5 FKBP prolyl isomerase 10 Homo sapiens 106-112 1371158-7 1992 Similarly, FK506 completely inhibited cytokine production by MC/9, partially inhibited cytokine production by MCIII and had differential effects on IL-3/granulocyte-macrophage colony-stimulating factor and IL-2/IL-4 production by MCVI. Tacrolimus 11-16 interleukin 3 Mus musculus 148-152 1371158-7 1992 Similarly, FK506 completely inhibited cytokine production by MC/9, partially inhibited cytokine production by MCIII and had differential effects on IL-3/granulocyte-macrophage colony-stimulating factor and IL-2/IL-4 production by MCVI. Tacrolimus 11-16 interleukin 2 Mus musculus 206-210 1371158-7 1992 Similarly, FK506 completely inhibited cytokine production by MC/9, partially inhibited cytokine production by MCIII and had differential effects on IL-3/granulocyte-macrophage colony-stimulating factor and IL-2/IL-4 production by MCVI. Tacrolimus 11-16 interleukin 4 Mus musculus 211-215 1371491-5 1992 FK506 or CsA inhibited SEA-induced tumour necrosis factor-alpha (TNF-alpha) production both in monocytes (P less than 0.01) and in lymphocytes (P less than 0.001), at a drug concentration of 1-25 ng/ml for FK506 and 100-500 ng/ml for CsA. Tacrolimus 0-5 tumor necrosis factor Homo sapiens 65-74 1384862-4 1992 However, the response of the PHA-pulsed T cells to IL-6 was still inhibited by FK-506 or Cs A, but the inhibitory effect gradually decreased as the time in which the PHA-pulsed T cells interacted with IL-6 was prolonged. Tacrolimus 79-85 interleukin 6 Homo sapiens 51-55 1371491-5 1992 FK506 or CsA inhibited SEA-induced tumour necrosis factor-alpha (TNF-alpha) production both in monocytes (P less than 0.01) and in lymphocytes (P less than 0.001), at a drug concentration of 1-25 ng/ml for FK506 and 100-500 ng/ml for CsA. Tacrolimus 206-211 tumor necrosis factor Homo sapiens 65-74 1384862-7 1992 It is likely that the two drugs inhibit the expression of lymphokine receptors, by interfering Ca(2+)-related signals and that IL-6 induces T cell proliferation in a different way than IL-2 and IL-4, which are FK-506- and Cs A-sensitive. Tacrolimus 210-216 interleukin 4 Homo sapiens 194-198 1371491-8 1992 When the cells were stimulated by phorbol ester (phorbol 12-myristate 13-acetate, PMA) plus calcium ionophore (ionomycin), FK506 and CsA inhibited, in a dose-dependent manner, the production of IL-2, IL-4, IL-5, IFN-gamma and TNF-alpha. Tacrolimus 123-128 interleukin 5 Homo sapiens 206-210 1371491-8 1992 When the cells were stimulated by phorbol ester (phorbol 12-myristate 13-acetate, PMA) plus calcium ionophore (ionomycin), FK506 and CsA inhibited, in a dose-dependent manner, the production of IL-2, IL-4, IL-5, IFN-gamma and TNF-alpha. Tacrolimus 123-128 interferon gamma Homo sapiens 212-221 1371491-8 1992 When the cells were stimulated by phorbol ester (phorbol 12-myristate 13-acetate, PMA) plus calcium ionophore (ionomycin), FK506 and CsA inhibited, in a dose-dependent manner, the production of IL-2, IL-4, IL-5, IFN-gamma and TNF-alpha. Tacrolimus 123-128 interleukin 2 Homo sapiens 194-198 1371491-8 1992 When the cells were stimulated by phorbol ester (phorbol 12-myristate 13-acetate, PMA) plus calcium ionophore (ionomycin), FK506 and CsA inhibited, in a dose-dependent manner, the production of IL-2, IL-4, IL-5, IFN-gamma and TNF-alpha. Tacrolimus 123-128 tumor necrosis factor Homo sapiens 226-235 1371491-10 1992 Further stimulation by addition of anti-CD28 mAb to the cultures resulted in an augmented IL-2 and IFN-gamma production which was resistant to both FK506 and CsA. Tacrolimus 148-153 interleukin 2 Homo sapiens 90-94 1371491-10 1992 Further stimulation by addition of anti-CD28 mAb to the cultures resulted in an augmented IL-2 and IFN-gamma production which was resistant to both FK506 and CsA. Tacrolimus 148-153 interferon gamma Homo sapiens 99-108 1371491-12 1992 Depending on the mode of cell activation the two drugs inhibited not only cytokine production in lymphocytes but also antigen-induced monokine (TNF-alpha) production in macrophages, although the optimal immunomodulatory effect of FK506 was achieved at a concentration approximately 50-fold lower than that of CsA. Tacrolimus 230-235 tumor necrosis factor Homo sapiens 144-153 1384862-0 1992 The effects of FK-506 and cyclosporin A on the proliferation of PHA-stimulated T cells in response to IL-2, IL-4 or IL-6. Tacrolimus 15-21 interleukin 2 Homo sapiens 102-106 1384862-0 1992 The effects of FK-506 and cyclosporin A on the proliferation of PHA-stimulated T cells in response to IL-2, IL-4 or IL-6. Tacrolimus 15-21 interleukin 6 Homo sapiens 116-120 1371491-8 1992 When the cells were stimulated by phorbol ester (phorbol 12-myristate 13-acetate, PMA) plus calcium ionophore (ionomycin), FK506 and CsA inhibited, in a dose-dependent manner, the production of IL-2, IL-4, IL-5, IFN-gamma and TNF-alpha. Tacrolimus 123-128 interleukin 4 Homo sapiens 200-204 1380720-7 1992 FK 506 pretreatment reduced the serum levels of BUN (p less than .02), creatinine (p less than .02) and TNF (p less than .05) as compared to that seen in controls. Tacrolimus 0-6 tumor necrosis factor-like Rattus norvegicus 104-107 1371575-0 1992 Effect of FK506 on insulin secretion in normal dogs. Tacrolimus 10-15 insulin Canis lupus familiaris 19-26 1371575-1 1992 In this report, we describe the effect of FK506 on glucose-mediated insulin release in normal dogs. Tacrolimus 42-47 insulin Canis lupus familiaris 68-75 1371575-9 1992 Insulin secretion after the recovery period remained unchanged in group 1 dogs, but continued to be significantly reduced in group 2 dogs that had received FK506 for 4 weeks. Tacrolimus 156-161 insulin Canis lupus familiaris 0-7 1372739-6 1992 The release of amylase in response to CCK was reduced by FK506 in a dose-related manner (p less than 0.01) and by CS at 25 mg/kg (p less than 0.01). Tacrolimus 57-62 cholecystokinin Rattus norvegicus 38-41 1282197-4 1992 This in vitro synergism of gamma-IFN and FK506 may have clinical application in that low doses of gamma-IFN and FK506 combinations may be effective to correct polyclonal B cell activation of patients with SLE. Tacrolimus 41-46 interferon gamma Mus musculus 98-107 1282197-4 1992 This in vitro synergism of gamma-IFN and FK506 may have clinical application in that low doses of gamma-IFN and FK506 combinations may be effective to correct polyclonal B cell activation of patients with SLE. Tacrolimus 112-117 interferon gamma Mus musculus 27-36 1380720-8 1992 Based upon these data, it appears that: (a) renal ischemia induces the release of TNF; (b) FK 506 pretreatment inhibits TNF production; and (c) FK 506 reduces renal injury association with I/R. Tacrolimus 91-97 tumor necrosis factor-like Rattus norvegicus 120-123 1380720-8 1992 Based upon these data, it appears that: (a) renal ischemia induces the release of TNF; (b) FK 506 pretreatment inhibits TNF production; and (c) FK 506 reduces renal injury association with I/R. Tacrolimus 144-150 tumor necrosis factor-like Rattus norvegicus 82-85 14621859-3 1992 Treatment with low dose FK 506 in combination with splenectomy (Spx) synergistically prolonged the heart allograft survival in this sensitized rat model. Tacrolimus 24-30 spexin hormone Rattus norvegicus 64-67 14621743-4 1992 Exposure of tubular cells to high concentrations of FK506 or CyA (10, 50 and 100 microM) induced a time- and dose-dependent cell injury in vitro characterized by a direct cytotoxic effect on tubular cells as expressed by release of 3H-thymidine from prelabelled cells, N-acetyl-beta-D-glucosaminidase (NAG) release and cell detachment. Tacrolimus 52-57 O-GlcNAcase Homo sapiens 269-300 14621743-4 1992 Exposure of tubular cells to high concentrations of FK506 or CyA (10, 50 and 100 microM) induced a time- and dose-dependent cell injury in vitro characterized by a direct cytotoxic effect on tubular cells as expressed by release of 3H-thymidine from prelabelled cells, N-acetyl-beta-D-glucosaminidase (NAG) release and cell detachment. Tacrolimus 52-57 O-GlcNAcase Homo sapiens 302-305 1721455-0 1991 FK 506 modulates accessory cell adhesion molecule expression and inhibits CD4 lymphocyte adhesion to retinal pigment epithelial cells in vitro: implications for therapy of uveoretinitis. Tacrolimus 0-6 CD4 molecule Homo sapiens 74-77 14621744-7 1992 The concentrations of FK506 or CyA which induced ET-1 secretion by tubular cells and kidney cells were not cytolytic as assessed by N-acetyl-beta-D-glucosaminidase (NAG) release and lactic dehydrogenase (LDH) release. Tacrolimus 22-27 O-GlcNAcase Homo sapiens 165-168 1721644-5 1991 FK-506 also inhibited the de novo synthesis of 5-lipoxygenase (sulfidopeptide leukotriene C4) and cyclo-oxygenase (prostaglandin D2) metabolites of arachidonic acid from mast cells challenged with anti-IgE. Tacrolimus 0-6 arachidonate 5-lipoxygenase Homo sapiens 47-61 1721462-0 1991 The ability of myelin basic protein-sensitised leukocytes to adoptively transfer experimental allergic encephalomyelitis following coculture with FK 506, cyclosporine, or prednisolone. Tacrolimus 146-152 myelin basic protein Homo sapiens 15-35 1721276-0 1991 Effects of FK 506 on human hepatic microsomal cytochrome P-450-dependent drug metabolism in vitro. Tacrolimus 11-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 1960417-2 1991 Here we show that the generation of interferon-gamma producing cells (IFN-gamma pc) in rat spleen cell cultures stimulated with concanavalin A (ConA) is dose-dependently inhibited by a wide variety of immunosuppressants such as cyclosporin A, FK506, hydrocortisone, dexamethasone, azathioprine and ART-18, a monoclonal antibody (mAb) with established immunosuppressive activity in organ transplantation and autoimmunity. Tacrolimus 243-248 interferon gamma Rattus norvegicus 36-52 1721286-0 1991 Inhibition of insulin release by FK 506 and its prevention by rioprostil, a stable prostaglandin E1 analogue. Tacrolimus 33-39 insulin Homo sapiens 14-21 1721310-0 1991 Activation of transcription factor NF kappa B in Jurkat cells is inhibited selectively by FK 506 in a signal-dependent manner. Tacrolimus 90-96 nuclear factor kappa B subunit 1 Homo sapiens 35-45 1721312-0 1991 FK 506, rapamycin, and cyclosporine: effects on IL-4 and IL-10 mRNA levels in a T-helper 2 cell line. Tacrolimus 0-6 interleukin 4 Homo sapiens 48-52 1721312-0 1991 FK 506, rapamycin, and cyclosporine: effects on IL-4 and IL-10 mRNA levels in a T-helper 2 cell line. Tacrolimus 0-6 interleukin 10 Homo sapiens 57-62 1721313-0 1991 Kinetics of early T-cell repopulation in the mouse following syngeneic bone marrow transplantation: FK 506 causes a maturational defect of CD4+ CD8- T cells. Tacrolimus 100-106 CD4 antigen Mus musculus 139-142 1721337-0 1991 FK 506: reversal of humorally mediated rejection following ABO-incompatible liver transplantation. Tacrolimus 0-6 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 59-62 1721394-0 1991 Synexin: a target protein for toxic effects of cyclosporine and FK 506 in endocrine cells. Tacrolimus 64-70 annexin A7 Homo sapiens 0-7 1960417-2 1991 Here we show that the generation of interferon-gamma producing cells (IFN-gamma pc) in rat spleen cell cultures stimulated with concanavalin A (ConA) is dose-dependently inhibited by a wide variety of immunosuppressants such as cyclosporin A, FK506, hydrocortisone, dexamethasone, azathioprine and ART-18, a monoclonal antibody (mAb) with established immunosuppressive activity in organ transplantation and autoimmunity. Tacrolimus 243-248 interferon gamma Rattus norvegicus 70-79 1722778-9 1991 On the other hand, the number of CD4+ CD8+ thymocytes did not decrease significantly in the 0.3 mg/kg/day-treated group, whereas it did decrease significantly in the 1 mg/kg/day-treated group, decreased further with the increase in dosage of FK-506, and decreased markedly in the 30 mg/kg/day group, The number of CD4-CD8-thymocytes did not show any change, even in the high-dosage groups. Tacrolimus 242-248 CD4 antigen Mus musculus 33-36 1721767-4 1991 Although full recovery was not observed in FK506, this finding indicated that FK506 as well as CsA inhibit IL2 secretion from HTL403. Tacrolimus 78-83 interleukin 2 Homo sapiens 107-110 1716149-1 1991 FKBP-12, the major T-cell binding protein for the immunosuppressive agents FK506 and rapamycin, catalyzes the interconversion of the cis and trans rotamers of the peptidyl-prolyl amide bond of peptide and protein substrates. Tacrolimus 75-80 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 0-7 1726895-0 1991 FK506 lacks the ability to inhibit expression of interleukin-2 receptor beta-chain on human lymphocytes. Tacrolimus 0-5 interleukin 2 receptor subunit beta Homo sapiens 49-76 1726895-3 1991 FK506 is a new immunosuppressant known to inhibit synthesis of interleukin 2 and expression of the membrane bound alpha chain. Tacrolimus 0-5 interleukin 2 Homo sapiens 63-76 1911152-9 1991 Unfortunately, one of the most effective ingredients of such combination therapies in animal models (anti-CD4) appears to have its tolerogenic potential abrogated by cyclosporin A and FK-506. Tacrolimus 184-190 CD4 molecule Homo sapiens 106-109 1716797-13 1991 Higher doses of FK506 produced a significant delay in glucose disappearance in groups 3 and 4, and a significant inhibition of glucose-mediated C-peptide response in group 4. Tacrolimus 16-21 insulin Homo sapiens 144-153 1715094-4 1991 An FKBP-rapamycin complex is concluded to be the toxic agent because (i) strains that lack FKBP proline rotamase, encoded by FPR1, were viable and fully resistant to rapamycin and (ii) FK506 antagonized rapamycin toxicity in vivo. Tacrolimus 185-190 peptidylprolyl isomerase FPR1 Saccharomyces cerevisiae S288C 125-129 1719972-1 1991 Recently, the amino acid sequence of a 12 Kd endogenous protein inhibitor of protein kinase C (PKC-I 2) has been shown to be identical to that of the 12 KDa receptor for the immunosuppressive drug, FK-506. Tacrolimus 198-204 proline rich transmembrane protein 2 Homo sapiens 77-93 1719972-1 1991 Recently, the amino acid sequence of a 12 Kd endogenous protein inhibitor of protein kinase C (PKC-I 2) has been shown to be identical to that of the 12 KDa receptor for the immunosuppressive drug, FK-506. Tacrolimus 198-204 FKBP prolyl isomerase 1A Homo sapiens 95-102 1930186-1 1991 FKBP-12 is the major T cell binding protein for the immunosuppressive drugs FK506 and rapamycin. Tacrolimus 76-81 FKBP prolyl isomerase 1A Homo sapiens 0-7 1720417-0 1991 The immunosuppressive agent FK506 inhibits in vitro expression of membrane-bound and soluble interleukin-2 receptors on resting but not on activated human lymphocytes. Tacrolimus 28-33 ATP binding cassette subfamily A member 1 Homo sapiens 66-80 1720417-0 1991 The immunosuppressive agent FK506 inhibits in vitro expression of membrane-bound and soluble interleukin-2 receptors on resting but not on activated human lymphocytes. Tacrolimus 28-33 interleukin 2 Homo sapiens 93-106 1720417-4 1991 The membrane-bound IL-2 receptor expression was inhibited by FK506 in resting lymphocytes at a concentration of 1 pmol/l. Tacrolimus 61-66 ATP binding cassette subfamily A member 1 Homo sapiens 4-18 1720417-4 1991 The membrane-bound IL-2 receptor expression was inhibited by FK506 in resting lymphocytes at a concentration of 1 pmol/l. Tacrolimus 61-66 interleukin 2 receptor subunit beta Homo sapiens 19-32 1709302-4 1991 The nature of this complex has implications for the mechanism of rotamase catalysis and for the biological actions of FK506 and rapamycin. Tacrolimus 118-123 FKBP prolyl isomerase 10 Homo sapiens 65-73 1712901-0 1991 The immunosuppressant FK-506 specifically inhibits mitogen-induced activation of the interleukin-2 promoter and the isolated enhancer elements NFIL-2A and NF-AT1. Tacrolimus 22-28 interleukin 2 Homo sapiens 85-98 1712901-0 1991 The immunosuppressant FK-506 specifically inhibits mitogen-induced activation of the interleukin-2 promoter and the isolated enhancer elements NFIL-2A and NF-AT1. Tacrolimus 22-28 nuclear factor of activated T cells 2 Homo sapiens 155-161 1712901-1 1991 The macrolide FK-506, like the cyclic undecapeptide cyclosporin A (CsA), is a potent immunosuppressant that interferes with the transcriptional activation of several early-phase genes in T lymphocytes, including that for interleukin-2 (IL-2). Tacrolimus 14-20 interleukin 2 Homo sapiens 221-234 1712901-1 1991 The macrolide FK-506, like the cyclic undecapeptide cyclosporin A (CsA), is a potent immunosuppressant that interferes with the transcriptional activation of several early-phase genes in T lymphocytes, including that for interleukin-2 (IL-2). Tacrolimus 14-20 interleukin 2 Homo sapiens 236-240 1712901-2 1991 We compared the effects of FK-506 and CsA on transcription from the 5" upstream activating sequences (UAS) of the human IL-2 gene and several cellular and viral UAS to define cis-acting sites which may be responsive to FK-506. Tacrolimus 27-33 interleukin 2 Homo sapiens 120-124 1712901-7 1991 The induced transcription driven by the IL-2 promoter elements NF-AT1 and NFIL-2A could be blocked completely by FK-506 or CsA. Tacrolimus 113-119 interleukin 2 Homo sapiens 40-44 1712901-7 1991 The induced transcription driven by the IL-2 promoter elements NF-AT1 and NFIL-2A could be blocked completely by FK-506 or CsA. Tacrolimus 113-119 nuclear factor of activated T cells 2 Homo sapiens 63-69 1710844-0 1991 Evidence that FK506 and rapamycin block T cell activation at different sites relative to early reversible phosphorylation involving the protein phosphatases PP1 and PP2A. Tacrolimus 14-19 inorganic pyrophosphatase 1 Homo sapiens 157-160 1710844-0 1991 Evidence that FK506 and rapamycin block T cell activation at different sites relative to early reversible phosphorylation involving the protein phosphatases PP1 and PP2A. Tacrolimus 14-19 protein phosphatase 2 phosphatase activator Homo sapiens 165-169 1713687-6 1991 Therefore, the three-dimensional structures of FKBP-12 and the FKBP-12/FK506 complex are likely to be excellent models of the corresponding FKBP-13 structure. Tacrolimus 71-76 FKBP prolyl isomerase 1A Homo sapiens 47-54 1713687-6 1991 Therefore, the three-dimensional structures of FKBP-12 and the FKBP-12/FK506 complex are likely to be excellent models of the corresponding FKBP-13 structure. Tacrolimus 71-76 FKBP prolyl isomerase 1A Homo sapiens 63-70 1713687-6 1991 Therefore, the three-dimensional structures of FKBP-12 and the FKBP-12/FK506 complex are likely to be excellent models of the corresponding FKBP-13 structure. Tacrolimus 71-76 FKBP prolyl isomerase 2 Homo sapiens 140-147 1712484-1 1991 Investigations of the actions and interactions of the immunophilin ligands FK506, cyclosporin A (CsA), rapamycin, and 506BD suggest that complexes of FK506 with an FK506-binding protein or of CsA with a cyclophilin (CsA-binding protein) inhibit the T-cell receptor-mediated signal transduction that results in the transcription of interleukin 2. Tacrolimus 150-155 interleukin 2 Rattus norvegicus 331-344 1712484-1 1991 Investigations of the actions and interactions of the immunophilin ligands FK506, cyclosporin A (CsA), rapamycin, and 506BD suggest that complexes of FK506 with an FK506-binding protein or of CsA with a cyclophilin (CsA-binding protein) inhibit the T-cell receptor-mediated signal transduction that results in the transcription of interleukin 2. Tacrolimus 150-155 interleukin 2 Rattus norvegicus 331-344 1715317-0 1991 Comparison of the effects of FK-506, cyclosporin A and rapamycin on IL-2 production. Tacrolimus 29-35 interleukin 2 Homo sapiens 68-72 1717376-1 1991 Cyclosporin (CsA) and FK-506 are structurally distinct fungal metabolites, which exert powerful inhibitory effects on CD4+ T (helper) cell activation and on the secretion of interleukin-2 (IL-2) and other cytokines, including various cell growth factors and interferon-gamma. Tacrolimus 22-28 interleukin 2 Homo sapiens 174-187 1717376-1 1991 Cyclosporin (CsA) and FK-506 are structurally distinct fungal metabolites, which exert powerful inhibitory effects on CD4+ T (helper) cell activation and on the secretion of interleukin-2 (IL-2) and other cytokines, including various cell growth factors and interferon-gamma. Tacrolimus 22-28 interleukin 2 Homo sapiens 189-193 1717376-1 1991 Cyclosporin (CsA) and FK-506 are structurally distinct fungal metabolites, which exert powerful inhibitory effects on CD4+ T (helper) cell activation and on the secretion of interleukin-2 (IL-2) and other cytokines, including various cell growth factors and interferon-gamma. Tacrolimus 22-28 interferon gamma Homo sapiens 258-274 1717376-8 1991 Compared with CsA, the inhibitory action of FK-506 appears more difficult to reverse, e.g., in response to pre-formed IL-2. Tacrolimus 44-50 interleukin 2 Homo sapiens 118-122 1713361-4 1991 FK506 and CsA showed pharmacologic antagonism in inhibiting in vitro proliferation upon phytohemagglutinin, anti-CD3 antibody, and mixed lymphocyte reaction (MLR) stimulation, and interleukin 2 generation by activated normal human peripheral blood lymphocytes. Tacrolimus 0-5 interleukin 2 Homo sapiens 180-193 1707760-3 1991 Using measurements of the proliferative response, IL-2 production, and changes in intracellular Ca2+ ([Ca2+]i), we demonstrate that FK-506 exerts its inhibitory effect on early events of T-cell activation in a manner indistinguishable from that of CsA. Tacrolimus 132-138 interleukin 2 Homo sapiens 50-54 1715185-0 1991 Cyclosporin A and FK-506 both affect DNA binding of regulatory nuclear proteins to the human interleukin-2 promoter. Tacrolimus 18-24 interleukin 2 Homo sapiens 93-106 1706398-0 1991 FK-506, a potent novel inhibitor of the release of proinflammatory mediators from human Fc epsilon RI+ cells. Tacrolimus 0-6 Fc epsilon receptor Ia Homo sapiens 88-101 1706398-9 1991 IL-3 (3 and 10 ng/ml), but not IL-1 beta (10 and 100 ng/ml), reversed the inhibitory effect of both FK-506 and CsA on basophils challenged with anti-IgE or A23187. Tacrolimus 100-106 interleukin 3 Homo sapiens 0-4 1715185-1 1991 The structurally unrelated immunosuppressive drugs cyclosporin A (Sandimmun) and FK-506 both interfere with the process of T-cell proliferation by blocking the transcription of the T-cell growth factor interleukin-2 (IL-2). Tacrolimus 81-87 interleukin 2 Homo sapiens 202-215 1715185-1 1991 The structurally unrelated immunosuppressive drugs cyclosporin A (Sandimmun) and FK-506 both interfere with the process of T-cell proliferation by blocking the transcription of the T-cell growth factor interleukin-2 (IL-2). Tacrolimus 81-87 interleukin 2 Homo sapiens 217-221 1715185-3 1991 We present evidence that the binding by regulatory nuclear proteins to the kappa B element of the IL-2 promoter is affected negatively by cyclosporin A and FK-506 at concentrations paralleling their immunosuppressive activity in vivo. Tacrolimus 156-162 interleukin 2 Homo sapiens 98-102 1715185-5 1991 FK-506 is 10 to 100 times more potent than cyclosporin A in its ability to inhibit sequence-specific DNA binding and IL-2 production. Tacrolimus 0-6 interleukin 2 Homo sapiens 117-121 1705513-1 1991 Structurally unrelated, FK-506 and cyclosporin (CsA) bind to and inhibit the action of distinct cytoplasmic receptors, FK-506-binding protein (FKBP) and cyclophilin (CyP), respectively. Tacrolimus 24-30 peptidylprolyl isomerase G Homo sapiens 153-164 1705713-8 1991 Overexpression or disruption of FPR1 confers resistance to growth inhibition by FK 506, suggesting that FKBP is a target for FK 506 in yeast. Tacrolimus 80-86 peptidylprolyl isomerase FPR1 Saccharomyces cerevisiae S288C 32-36 1705513-4 1991 We have shown that FK-506, like CsA, is able to inhibit T cell activation mediated not only by the T cell receptor-CD3 complex, but also via another surface molecule, CD2. Tacrolimus 19-25 CD2 molecule Homo sapiens 167-170 1703047-0 1991 Effects of an immunosuppressant, FK506, on interleukin 1 alpha production by human macrophages and a macrophage-like cell line, U937. Tacrolimus 33-38 interleukin 1 alpha Homo sapiens 43-62 1703047-3 1991 FK506 partially suppressed IL-1 alpha release, from macrophage-like U937 cells stimulated with phorbol myristate acetate and from human monocytes and alveolar macrophages activated with lipopolysaccharide, in a dose-dependent manner. Tacrolimus 0-5 interleukin 1 alpha Homo sapiens 27-37 1703047-4 1991 Moreover, it was indicated that FK506 suppressed not only IL-1 release but also IL-1 synthesis itself, by measurement of cell-associated IL-1 alpha of U937 cells. Tacrolimus 32-37 interleukin 1 alpha Homo sapiens 58-62 1703047-4 1991 Moreover, it was indicated that FK506 suppressed not only IL-1 release but also IL-1 synthesis itself, by measurement of cell-associated IL-1 alpha of U937 cells. Tacrolimus 32-37 interleukin 1 alpha Homo sapiens 80-84 1703047-4 1991 Moreover, it was indicated that FK506 suppressed not only IL-1 release but also IL-1 synthesis itself, by measurement of cell-associated IL-1 alpha of U937 cells. Tacrolimus 32-37 interleukin 1 alpha Homo sapiens 137-147 1703047-5 1991 The optimal concentrations of FK506 for suppressing IL-1 alpha did not affect cell viability or proliferation, and were 10- to 100-fold lower than those of cyclosporin A. Tacrolimus 30-35 interleukin 1 alpha Homo sapiens 52-62 1703047-6 1991 It is concluded that FK506 affects macrophage physiology, suppressing IL-1 alpha production significantly. Tacrolimus 21-26 interleukin 1 alpha Homo sapiens 70-80 1705513-1 1991 Structurally unrelated, FK-506 and cyclosporin (CsA) bind to and inhibit the action of distinct cytoplasmic receptors, FK-506-binding protein (FKBP) and cyclophilin (CyP), respectively. Tacrolimus 24-30 peptidylprolyl isomerase G Homo sapiens 166-169 1707162-0 1991 The immunosuppressives FK 506 and cyclosporin A inhibit the generation of protein factors binding to the two purine boxes of the interleukin 2 enhancer. Tacrolimus 23-29 interleukin 2 Homo sapiens 129-142 1707162-1 1991 Like Cyclosporin A (CsA), the macrolide FK 506 is a potent immunosuppressive that inhibits early steps of T cell activation, including the synthesis of Interleukin 2 (II-2) and numerous other lymphokines. Tacrolimus 40-46 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 20-23 1707162-1 1991 Like Cyclosporin A (CsA), the macrolide FK 506 is a potent immunosuppressive that inhibits early steps of T cell activation, including the synthesis of Interleukin 2 (II-2) and numerous other lymphokines. Tacrolimus 40-46 interleukin 2 Homo sapiens 152-165 1707162-1 1991 Like Cyclosporin A (CsA), the macrolide FK 506 is a potent immunosuppressive that inhibits early steps of T cell activation, including the synthesis of Interleukin 2 (II-2) and numerous other lymphokines. Tacrolimus 40-46 interleukin 2 Homo sapiens 167-171 1707162-3 1991 At concentrations that block T cell activation, FK 506 and CsA inhibit the proto-enhancer activity of Purine boxes of the II-2 promoter and the generation of lymphocyte-specific factors binding to the Purine boxes. Tacrolimus 48-54 interleukin 2 Homo sapiens 122-126 1721613-11 1991 However, RAP (110 nM) reversed the apoptosis-inhibitory effect of FK-506, even if added 1-2 h after the latter to the cultures. Tacrolimus 66-72 regulatory associated protein of MTOR, complex 1 Mus musculus 9-12 1721613-12 1991 Consistent with this antagonism, RAP also reversed the binding of a radiolabeled derivative of FK-506 in DO-11.10 cells. Tacrolimus 95-101 regulatory associated protein of MTOR, complex 1 Mus musculus 33-36 1726093-0 1991 Unexpected up-regulation of gene expression by cyclosporin A and FK-506 in a T-cell lymphoma: both immunosuppressants augment Ly-6E antigen induction by interferon-gamma in the presence of ionomycin. Tacrolimus 65-71 lymphocyte antigen 6 complex, locus E Mus musculus 126-131 1726093-0 1991 Unexpected up-regulation of gene expression by cyclosporin A and FK-506 in a T-cell lymphoma: both immunosuppressants augment Ly-6E antigen induction by interferon-gamma in the presence of ionomycin. Tacrolimus 65-71 interferon gamma Mus musculus 153-169 1726093-6 1991 Cyclosporin A or FK-506 also markedly affected Ly-6E induction when the cultures were co-treated with the calcium ionophore, ionomycin. Tacrolimus 17-23 lymphocyte antigen 6 complex, locus E Mus musculus 47-52 1726093-11 1991 Therefore, through mechanisms apparently related to those involved in their immunosuppressive action, both CsA and FK-506 convert the negative effect of ionomycin on IFN-gamma-mediated Ly-6E induction into an overall positive effect. Tacrolimus 115-121 interferon gamma Mus musculus 166-175 1702372-1 1990 The effect of administration of FK506 at 1 mg/kg body weight for 14 days on rat lymphoid tissues, especially the thymus, and recovery after discontinuation of treatment, were investigated by the immunoperoxidase technique and flow cytofluorometry using monoclonal antibodies OX6, OX7, OX8, OX18 and W3/25, reactive with rat lymphocytes. Tacrolimus 32-37 Cd4 molecule Rattus norvegicus 299-304 1726093-11 1991 Therefore, through mechanisms apparently related to those involved in their immunosuppressive action, both CsA and FK-506 convert the negative effect of ionomycin on IFN-gamma-mediated Ly-6E induction into an overall positive effect. Tacrolimus 115-121 lymphocyte antigen 6 complex, locus E Mus musculus 185-190 1702372-6 1990 Flow cytometric analysis of the thymus showed that the percentages of cells labelled positively with OX7, OX8 and W3/25 were increased with FK506 treatment, and recovered to the normal level soon after withdrawal. Tacrolimus 140-145 Cd4 molecule Rattus norvegicus 114-119 1702372-7 1990 Furthermore, the peak of fluorescence intensity of OX7+, OX8+ and W3/25+ cells showed a temporary shift to the right during FK506 treatment; however, the peak of fluorescence intensity of OX18+ cells showed a temporary shift to the left. Tacrolimus 124-129 Cd4 molecule Rattus norvegicus 66-71 1702375-6 1990 Either CyA or FK506 significantly reduced the number of cells expressing OX-39 (interleukin-2 receptors) and OX-40. Tacrolimus 14-19 interleukin 2 Mus musculus 80-93 1702375-6 1990 Either CyA or FK506 significantly reduced the number of cells expressing OX-39 (interleukin-2 receptors) and OX-40. Tacrolimus 14-19 tumor necrosis factor receptor superfamily, member 4 Mus musculus 109-114 1702375-8 1990 In contrast, the absolute spleen cell numbers of OX-19+, W3/25+ and OX-8+ cells were significantly reduced in transfused animals given 14 days of FK506 treatment, while the corresponding blood cells were unaffected. Tacrolimus 146-151 AF4/FMR2 family, member 2 Mus musculus 49-54 1701825-5 1990 Therefore, CSA and FK506, while chemically different, seem to act upon a similar pathway that leads to IL-2 gene expression, whereas glucocorticoids do not affect this pathway. Tacrolimus 19-24 interleukin 2 Homo sapiens 103-107 1702408-3 1990 It was found that spleens from S-antigen-immunized and FK506-treated rats contained a population of Ts cells inhibiting the proliferative responses of S-antigen-sensitized lymphocytes to S-antigen, yet these cells did not affect the proliferative responses of interphotoreceptor retinoid-binding protein (IRBP)-sensitized lymphocytes to IRBP. Tacrolimus 55-60 S-antigen visual arrestin Rattus norvegicus 151-160 1702408-3 1990 It was found that spleens from S-antigen-immunized and FK506-treated rats contained a population of Ts cells inhibiting the proliferative responses of S-antigen-sensitized lymphocytes to S-antigen, yet these cells did not affect the proliferative responses of interphotoreceptor retinoid-binding protein (IRBP)-sensitized lymphocytes to IRBP. Tacrolimus 55-60 S-antigen visual arrestin Rattus norvegicus 151-160 1702408-3 1990 It was found that spleens from S-antigen-immunized and FK506-treated rats contained a population of Ts cells inhibiting the proliferative responses of S-antigen-sensitized lymphocytes to S-antigen, yet these cells did not affect the proliferative responses of interphotoreceptor retinoid-binding protein (IRBP)-sensitized lymphocytes to IRBP. Tacrolimus 55-60 retinol binding protein 3 Rattus norvegicus 260-303 1702408-3 1990 It was found that spleens from S-antigen-immunized and FK506-treated rats contained a population of Ts cells inhibiting the proliferative responses of S-antigen-sensitized lymphocytes to S-antigen, yet these cells did not affect the proliferative responses of interphotoreceptor retinoid-binding protein (IRBP)-sensitized lymphocytes to IRBP. Tacrolimus 55-60 retinol binding protein 3 Rattus norvegicus 305-309 1702408-3 1990 It was found that spleens from S-antigen-immunized and FK506-treated rats contained a population of Ts cells inhibiting the proliferative responses of S-antigen-sensitized lymphocytes to S-antigen, yet these cells did not affect the proliferative responses of interphotoreceptor retinoid-binding protein (IRBP)-sensitized lymphocytes to IRBP. Tacrolimus 55-60 retinol binding protein 3 Rattus norvegicus 337-341 1702408-5 1990 Furthermore, transfer of Ts cells from S-antigen-immunized and FK506-treated rats to naive syngenic rats induced partial inhibition of EAU induction or delay of EAU onset after immunizing the recipient rats with S-antigen. Tacrolimus 63-68 S-antigen visual arrestin Rattus norvegicus 212-221 1702408-7 1990 These data thus indicate that FK506 treatment after S-antigen immunization induces an activation of Ts cells specific to S-antigen and that the Ts cells might contribute, at least in part, to the uniquely prolonged and intensive immunosuppression by FK506. Tacrolimus 30-35 S-antigen visual arrestin Rattus norvegicus 52-61 1702408-7 1990 These data thus indicate that FK506 treatment after S-antigen immunization induces an activation of Ts cells specific to S-antigen and that the Ts cells might contribute, at least in part, to the uniquely prolonged and intensive immunosuppression by FK506. Tacrolimus 30-35 S-antigen visual arrestin Rattus norvegicus 121-130 2123553-2 1990 On the other hand, interleukin 2 (IL-2)-induced signals are blocked by rapamycin but not by FK506. Tacrolimus 92-97 interleukin 2 Homo sapiens 34-38 1701148-2 1990 First, rats actively immunized with S-antigen were treated with FK 506 only after the onset of EAU. Tacrolimus 64-70 S-antigen visual arrestin Rattus norvegicus 36-45 1701570-1 1990 The fungal metabolite FK506 was discovered because it shared an important property, the ability to inhibit production of IL-2, with another well-known immunosuppressive fungal metabolite, CsA. Tacrolimus 22-27 interleukin 2 Homo sapiens 121-125 2123553-5 1990 However, an excess of rapamycin is needed to revert FK506-mediated inhibition of IL-2 production, apoptosis, and transcriptional activation of NF-AT, a T-cell-specific transcription factor necessary for IL-2 gene activation. Tacrolimus 52-57 interleukin 2 Homo sapiens 81-85 2123553-5 1990 However, an excess of rapamycin is needed to revert FK506-mediated inhibition of IL-2 production, apoptosis, and transcriptional activation of NF-AT, a T-cell-specific transcription factor necessary for IL-2 gene activation. Tacrolimus 52-57 interleukin 2 Homo sapiens 203-207 2123553-6 1990 Similarly, an excess of FK506 is needed to revert rapamycin-mediated inhibition of IL-2-induced proliferation. Tacrolimus 24-29 interleukin 2 Homo sapiens 83-87 1701148-9 1990 The antibody levels to S-antigen, the antigen-specific proliferative responses of lymphocytes, and even the proliferative responses to Con A were markedly suppressed in the rats in which FK 506 was given only during the efferent limb of the immune response. Tacrolimus 187-193 S-antigen visual arrestin Rattus norvegicus 23-32 1696410-8 1990 Exogenous IL-2 or L3T4+ T cells could overcome the immunosuppressive effect of FK506 on the CTL induction of Ly2+ T cells in a secondary MLC. Tacrolimus 79-84 interleukin 2 Mus musculus 10-14 1702750-5 1990 FK506 caused a dose-dependent inhibition of cell cycle-related induction of locomotor capacity both of anti-CD3-cultured T cells and IL-4-cultured B cells, with an ID50 of less than 1 ng per ml. Tacrolimus 0-5 interleukin 4 Homo sapiens 133-137 2282365-10 1990 The FK-506 binding protein also has a PPIase activity, and this activity is inhibited by FK-506. Tacrolimus 4-10 FKBP prolyl isomerase like Homo sapiens 38-44 1688572-3 1990 FK-506 or CsA also inhibited proliferation, IL-2 production, and IL-2R expression in splenic T cells activated with ionomycin + PMA. Tacrolimus 0-6 interleukin 2 Mus musculus 44-48 1690915-9 1990 In contrast to corresponding PHA-stimulated cells, FK-506-treated alloactivated lymphocytes exhibited reductions in IL-2R, TR and HLA-DR antigen expression, which in the cases of IL-2R and TR were further reduced by combination of FK-506 with low-concentration CsA. Tacrolimus 51-57 interleukin 2 receptor subunit alpha Homo sapiens 116-121 1690915-9 1990 In contrast to corresponding PHA-stimulated cells, FK-506-treated alloactivated lymphocytes exhibited reductions in IL-2R, TR and HLA-DR antigen expression, which in the cases of IL-2R and TR were further reduced by combination of FK-506 with low-concentration CsA. Tacrolimus 51-57 transferrin receptor Homo sapiens 123-125 1690915-9 1990 In contrast to corresponding PHA-stimulated cells, FK-506-treated alloactivated lymphocytes exhibited reductions in IL-2R, TR and HLA-DR antigen expression, which in the cases of IL-2R and TR were further reduced by combination of FK-506 with low-concentration CsA. Tacrolimus 51-57 interleukin 2 receptor subunit alpha Homo sapiens 179-184 1690915-9 1990 In contrast to corresponding PHA-stimulated cells, FK-506-treated alloactivated lymphocytes exhibited reductions in IL-2R, TR and HLA-DR antigen expression, which in the cases of IL-2R and TR were further reduced by combination of FK-506 with low-concentration CsA. Tacrolimus 51-57 transferrin receptor Homo sapiens 189-191 1717008-7 1990 FK506 treatment resulted not only in the highest inhibition of expression of IL-2 receptors on T cells, but also in the prevention of the expression of MHC class II antigens on ocular resident cells. Tacrolimus 0-5 interleukin 2 Rattus norvegicus 77-81 1695378-2 1990 After the recent discovery that the cyclosporin A-binding protein cyclophilin is identical to peptidylprolyl cis-trans isomerase, a cellular binding protein for FK506 was found to be distinct from cyclophilin but to have the same enzymatic activity. Tacrolimus 161-166 FKBP prolyl isomerase 5 Homo sapiens 94-128 1689353-2 1990 FK-506 acts similarly to cyclosporin A (CsA) and prevents IL-2 production and IL-2R expression. Tacrolimus 0-6 interleukin 2 Mus musculus 58-62 1689353-2 1990 FK-506 acts similarly to cyclosporin A (CsA) and prevents IL-2 production and IL-2R expression. Tacrolimus 0-6 interleukin 2 receptor, alpha chain Mus musculus 78-83 1689353-6 1990 However, in the same system, RAP acted as a potent antagonist of FK-506 suppression. Tacrolimus 65-71 regulatory associated protein of MTOR, complex 1 Mus musculus 29-32 1689353-7 1990 RAP also blocked FK-506- but not CsA-mediated inhibition of IL-2 mRNA induction. Tacrolimus 17-23 regulatory associated protein of MTOR, complex 1 Mus musculus 0-3 1689353-8 1990 By using model systems sensitive to inhibition by RAP but not FK-506 we further demonstrated that FK-506 reciprocally behaves as an antagonist of RAP. Tacrolimus 98-104 regulatory associated protein of MTOR, complex 1 Mus musculus 50-53 1689353-8 1990 By using model systems sensitive to inhibition by RAP but not FK-506 we further demonstrated that FK-506 reciprocally behaves as an antagonist of RAP. Tacrolimus 98-104 regulatory associated protein of MTOR, complex 1 Mus musculus 146-149 1689353-9 1990 In one such model, the stimulation of splenic T cells with IL-2 + PMA, FK-506, but not CsA, reversed the suppressive effect of RAP on proliferation. Tacrolimus 71-77 interleukin 2 Mus musculus 59-63 1689353-9 1990 In one such model, the stimulation of splenic T cells with IL-2 + PMA, FK-506, but not CsA, reversed the suppressive effect of RAP on proliferation. Tacrolimus 71-77 regulatory associated protein of MTOR, complex 1 Mus musculus 127-130 1689353-10 1990 FK-506 also antagonized RAP-mediated inhibition with respect to the induction of Ly-6E Ag expression by IFN in YAC cells. Tacrolimus 0-6 regulatory associated protein of MTOR, complex 1 Mus musculus 24-27 1689353-10 1990 FK-506 also antagonized RAP-mediated inhibition with respect to the induction of Ly-6E Ag expression by IFN in YAC cells. Tacrolimus 0-6 lymphocyte antigen 6 complex, locus E Mus musculus 81-86 1690097-0 1990 Effects of a novel immunosuppressive agent, FK506, on human B cell activation. Tacrolimus 44-49 B cell linker Homo sapiens 60-77 1689694-7 1990 FACS analyses of the spleen cells revealed that FK506 reduced the percentage of double negative T cells (Thy-1.2+, Lyt-2-, L3T4-). Tacrolimus 48-53 acyl-CoA synthetase long-chain family member 1 Mus musculus 0-4 1689694-7 1990 FACS analyses of the spleen cells revealed that FK506 reduced the percentage of double negative T cells (Thy-1.2+, Lyt-2-, L3T4-). Tacrolimus 48-53 CD8 antigen, alpha chain Mus musculus 115-120 1689226-8 1990 FK-506 also suppressed increases in the small proportion of blood-borne OX-40+ (activated CD4+) cells and OX-39+ (interleukin-2 receptor+) cells in the 7 day period following immunization; thereafter values for activation marker expression between treatment and control groups were similar. Tacrolimus 0-6 interleukin 2 Mus musculus 114-127 1688572-3 1990 FK-506 or CsA also inhibited proliferation, IL-2 production, and IL-2R expression in splenic T cells activated with ionomycin + PMA. Tacrolimus 0-6 interleukin 2 receptor, alpha chain Mus musculus 65-70 1688572-8 1990 The proliferative response induced in D10.G4 cells by IL-1 + ionomycin but not that induced by IL-1 + PMA was sensitive to inhibition by FK-506 and CsA. Tacrolimus 137-143 interleukin 1 complex Mus musculus 54-58 1688572-10 1990 Finally, T cell proliferation driven by IL-2 or IL-4 was found to be relatively resistant to FK-506 or CsA but sensitive to RAP. Tacrolimus 93-99 interleukin 2 Mus musculus 40-44 1688572-10 1990 Finally, T cell proliferation driven by IL-2 or IL-4 was found to be relatively resistant to FK-506 or CsA but sensitive to RAP. Tacrolimus 93-99 interleukin 4 Mus musculus 48-52 33805042-6 2021 Interestingly, drugs widely used to treat skin inflammation, the calcineurin inhibitors tacrolimus and cyclosporine and the glucocorticoid dexamethasone, significantly decreased TRPA1 expression. Tacrolimus 88-98 transient receptor potential cation channel, subfamily A, member 1 Mus musculus 178-183 33768546-13 2021 The haematocrit (HCT), postoperative time (POD) and CYP3A5*3 genotypes had a significant influence on TAC clearance when combined with WZC. Tacrolimus 102-105 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 52-58 33768546-15 2021 For patients carrying the CYP3A5*3/*3 allele and with 30% HCT, the required TAC dose to achieve target trough concentrations of 10-15 ng/ml was 4 mg twice daily (q12h). Tacrolimus 76-79 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 26-32 33768546-16 2021 For patients with the CYP3A5*3/*3 allele, the required dose was 3 mg TAC q12h when combined with WZC, and for patients with the CYP3A5*1/*1 or *1/*3 allele, the required dose was 4 mg of TAC q12h when co-administered with WZC. Tacrolimus 69-72 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 22-28 33768546-16 2021 For patients with the CYP3A5*3/*3 allele, the required dose was 3 mg TAC q12h when combined with WZC, and for patients with the CYP3A5*1/*1 or *1/*3 allele, the required dose was 4 mg of TAC q12h when co-administered with WZC. Tacrolimus 187-190 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 128-134 33768546-17 2021 WHAT IS NEW AND CONCLUSION: Wuzhi Capsule co-administration and CYP3A5 variants affect the PK of TAC Dosing guidelines are made based on the PPK model to allow individualized administration of TAC, especially when co-administered with WZC. Tacrolimus 97-100 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 64-70 33768546-17 2021 WHAT IS NEW AND CONCLUSION: Wuzhi Capsule co-administration and CYP3A5 variants affect the PK of TAC Dosing guidelines are made based on the PPK model to allow individualized administration of TAC, especially when co-administered with WZC. Tacrolimus 193-196 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 64-70 33808965-6 2021 Improved systemic and ovarian immune functions, endometrial progesterone receptor and coreceptor expressions and uterine vascular adaptation to pregnancy were among features of enhanced progesterone-receptor sensitivity in the low-dose tacrolimus-treated mouse model of the disease. Tacrolimus 236-246 progesterone receptor Mus musculus 60-81 33777956-0 2021 Treatment With Calcineurin Inhibitor FK506 Attenuates Noise-Induced Hearing Loss. Tacrolimus 37-42 calcineurin binding protein 1 Mus musculus 15-36 33777956-5 2021 In agreement with our previous finding that autophagy marker microtubule-associated protein light chain 3B (LC3B) does not change in OHCs under conditions of moderate-noise-induced permanent threshold shifts, treatment with FK506 increases LC3B immunolabeling in OHCs after exposure to moderate noise. Tacrolimus 224-229 microtubule-associated protein 1 light chain 3 beta Mus musculus 108-112 33777956-5 2021 In agreement with our previous finding that autophagy marker microtubule-associated protein light chain 3B (LC3B) does not change in OHCs under conditions of moderate-noise-induced permanent threshold shifts, treatment with FK506 increases LC3B immunolabeling in OHCs after exposure to moderate noise. Tacrolimus 224-229 microtubule-associated protein 1 light chain 3 beta Mus musculus 240-244 33777956-6 2021 Additionally, prevention of NIHL by treatment with FK506 was partially abolished by pretreatment with LC3B small interfering RNA. Tacrolimus 51-56 microtubule-associated protein 1 light chain 3 beta Mus musculus 102-106 7512015-9 1994 Water-immersion stress significantly increased myeloperoxidase activity in gastric mucosa, and FK506 reduced the increase in myeloperoxidase activity induced by stress. Tacrolimus 95-100 myeloperoxidase Rattus norvegicus 125-140 27011912-1 2016 Tacrolimus (Prograf( ), Astellas Pharma Europe Ltd, Staines, United Kingdom; referred to as tacrolimus-BID) is an immunosuppressive agent to prevent and treat allograft rejection in kidney transplant recipients in combination with mycophenolate mofetil, corticosteroids, with or without basiliximab induction. Tacrolimus 0-10 BH3 interacting domain death agonist Homo sapiens 103-106 27011912-1 2016 Tacrolimus (Prograf( ), Astellas Pharma Europe Ltd, Staines, United Kingdom; referred to as tacrolimus-BID) is an immunosuppressive agent to prevent and treat allograft rejection in kidney transplant recipients in combination with mycophenolate mofetil, corticosteroids, with or without basiliximab induction. Tacrolimus 12-19 BH3 interacting domain death agonist Homo sapiens 103-106 27011912-1 2016 Tacrolimus (Prograf( ), Astellas Pharma Europe Ltd, Staines, United Kingdom; referred to as tacrolimus-BID) is an immunosuppressive agent to prevent and treat allograft rejection in kidney transplant recipients in combination with mycophenolate mofetil, corticosteroids, with or without basiliximab induction. Tacrolimus 92-102 BH3 interacting domain death agonist Homo sapiens 103-106 27011912-4 2016 Extended release tacrolimus has similar absorption, distribution, metabolism and excretion to tacrolimus-BID. Tacrolimus 94-104 BH3 interacting domain death agonist Homo sapiens 105-108 27011912-5 2016 Phase I pharmacokinetic trials comparing extended release tacrolimus and tacrolimus-BID have demonstrated a decreased maximum concentration (Cmax) and delayed time to maximum concentration (tmax) with the extended release formulation; however, AUC0-24 was comparable between formulations. Tacrolimus 73-83 BH3 interacting domain death agonist Homo sapiens 84-87 27011912-6 2016 Overall extended release tacrolimus has a very similar safety and efficacy profile to tacrolimus-BID. Tacrolimus 86-96 BH3 interacting domain death agonist Homo sapiens 97-100 17986100-2 2007 We report a 50-year-old Japanese man with anti-MuSK antibody-positive MG, who showed no or poor response to various therapies, including plasmapheresis, corticosteroid, and tacrolimus. Tacrolimus 173-183 muscle associated receptor tyrosine kinase Homo sapiens 47-51 30011421-3 2018 METHODS: In this retrospective case series, we identified consecutive CTD-ILD patients treated with tacrolimus plus intravenous (i.v.) Tacrolimus 100-110 CTD Homo sapiens 70-73 30011421-10 2018 CONCLUSION: In our cohort of CTD-ILD, two courses of pulse dose methylprednisolone therapy followed by prednisone and oral tacrolimus appeared to be well tolerated, and to have multidimensional efficacy. Tacrolimus 123-133 CTD Homo sapiens 29-32 19855314-0 2009 Time of drug administration, CYP3A5 and ABCB1 genotypes, and analytical method influence tacrolimus pharmacokinetics: a population pharmacokinetic study. Tacrolimus 89-99 ATP binding cassette subfamily B member 1 Homo sapiens 40-45 19709321-8 2009 Increased tacrolimus half-life and increased dose interval of sirolimus and tacrolimus were due to CYP3A4/5 and/or P-glycoprotein inhibition by protease inhibitors. Tacrolimus 10-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-105 19709321-8 2009 Increased tacrolimus half-life and increased dose interval of sirolimus and tacrolimus were due to CYP3A4/5 and/or P-glycoprotein inhibition by protease inhibitors. Tacrolimus 10-20 ATP binding cassette subfamily B member 1 Homo sapiens 115-129 19709321-8 2009 Increased tacrolimus half-life and increased dose interval of sirolimus and tacrolimus were due to CYP3A4/5 and/or P-glycoprotein inhibition by protease inhibitors. Tacrolimus 76-86 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-105 19709321-8 2009 Increased tacrolimus half-life and increased dose interval of sirolimus and tacrolimus were due to CYP3A4/5 and/or P-glycoprotein inhibition by protease inhibitors. Tacrolimus 76-86 ATP binding cassette subfamily B member 1 Homo sapiens 115-129 34785307-2 2022 FK-506 caused a significant attenuation in the proliferation of allogeneic CD4+ T cells and IFN-gamma secreting effector functions. Tacrolimus 0-6 Cd4 molecule Rattus norvegicus 75-78 34785307-2 2022 FK-506 caused a significant attenuation in the proliferation of allogeneic CD4+ T cells and IFN-gamma secreting effector functions. Tacrolimus 0-6 interferon gamma Rattus norvegicus 92-101 34793770-0 2022 The mTOR inhibitor everolimus attenuates tacrolimus-induced renal interstitial fibrosis in rats. Tacrolimus 41-51 mechanistic target of rapamycin kinase Rattus norvegicus 4-8 34922024-9 2022 In terms of the predictors of GI symptoms, it was determined that mycophenolate mofetil (MMF) was effective in the development of reflux and diarrhoea, cyclosporine in the development of diarrhoea and constipation, and tacrolimus in the development of indigestion, which are (p < 0.05). Tacrolimus 219-229 G protein subunit alpha i1 Homo sapiens 30-32 34688813-1 2022 AIM: The purpose of this study was to investigate the effect of CYP3A7, CYP3A4, and CYP3A5 genetic polymorphisms in liver transplant recipients and donors on tacrolimus concentrations in the early stages after liver transplantation. Tacrolimus 158-168 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 64-70 34688813-1 2022 AIM: The purpose of this study was to investigate the effect of CYP3A7, CYP3A4, and CYP3A5 genetic polymorphisms in liver transplant recipients and donors on tacrolimus concentrations in the early stages after liver transplantation. Tacrolimus 158-168 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 72-78 34688813-1 2022 AIM: The purpose of this study was to investigate the effect of CYP3A7, CYP3A4, and CYP3A5 genetic polymorphisms in liver transplant recipients and donors on tacrolimus concentrations in the early stages after liver transplantation. Tacrolimus 158-168 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 84-90 34688813-4 2022 RESULTS: Recipient CYP3A polymorphisms were associated with tacrolimus concentrations. Tacrolimus 60-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-24 34688813-7 2022 Regardless of the genotype of the matched donor, CYP3A7 rs10211, CYP3A4*1G (rs2242480), and CYP3A5*3 (rs776746) polymorphisms of recipients could affect tacrolimus concentrations. Tacrolimus 153-163 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 49-55 34688813-7 2022 Regardless of the genotype of the matched donor, CYP3A7 rs10211, CYP3A4*1G (rs2242480), and CYP3A5*3 (rs776746) polymorphisms of recipients could affect tacrolimus concentrations. Tacrolimus 153-163 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-71 34688813-7 2022 Regardless of the genotype of the matched donor, CYP3A7 rs10211, CYP3A4*1G (rs2242480), and CYP3A5*3 (rs776746) polymorphisms of recipients could affect tacrolimus concentrations. Tacrolimus 153-163 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 92-98 34688813-8 2022 For the CYP3A4 rs4646437 polymorphisms, when the donor carried CYP3A4 rs4646437 CC, the recipient CYP3A4 rs4646437 polymorphism was associated with the C0/D of tacrolimus, and when the donor carried CYP3A4 rs4646437 CT/TT genotype, the recipient CYP3A4 rs4646437 polymorphism also affected on tacrolimus C0/D, although the effect was not significant. Tacrolimus 160-170 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 98-104 34688813-8 2022 For the CYP3A4 rs4646437 polymorphisms, when the donor carried CYP3A4 rs4646437 CC, the recipient CYP3A4 rs4646437 polymorphism was associated with the C0/D of tacrolimus, and when the donor carried CYP3A4 rs4646437 CT/TT genotype, the recipient CYP3A4 rs4646437 polymorphism also affected on tacrolimus C0/D, although the effect was not significant. Tacrolimus 293-303 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 246-252 34688813-9 2022 CONCLUSION: The large inter-individual variation in tacrolimus concentrations in the early stages after liver transplantation is influenced by genetic polymorphisms of CYP3A7, CYP3A4, and CYP3A5. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 168-174 34688813-9 2022 CONCLUSION: The large inter-individual variation in tacrolimus concentrations in the early stages after liver transplantation is influenced by genetic polymorphisms of CYP3A7, CYP3A4, and CYP3A5. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 176-182 34688813-9 2022 CONCLUSION: The large inter-individual variation in tacrolimus concentrations in the early stages after liver transplantation is influenced by genetic polymorphisms of CYP3A7, CYP3A4, and CYP3A5. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 188-194 34688813-11 2022 Therefore, the detection of CYP3A polymorphisms in recipients could help to predict the tacrolimus starting dose in the early stages after liver transplantation. Tacrolimus 88-98 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 28-33 34781138-0 2022 Trans-eQTLs of the CYP3A4 and CYP3A5 associated with tacrolimus trough blood concentration in Chinese renal transplant patients. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-25 34781138-0 2022 Trans-eQTLs of the CYP3A4 and CYP3A5 associated with tacrolimus trough blood concentration in Chinese renal transplant patients. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 30-36 34781138-1 2022 This study aimed to systematically investigate trans-eQTLs of CYP3A4 and CYP3A5 affecting tacrolimus trough blood concentrations in Chinese renal transplant patients. Tacrolimus 90-100 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 34781138-1 2022 This study aimed to systematically investigate trans-eQTLs of CYP3A4 and CYP3A5 affecting tacrolimus trough blood concentrations in Chinese renal transplant patients. Tacrolimus 90-100 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 73-79 34780122-10 2022 Two and three sampling points limited sampling strategies: C0 , C2 , and C10 provide the most reliable and effective LSS for estimation of tacrolimus AUC0-24 in routine clinic use. Tacrolimus 139-149 homeobox C10 Homo sapiens 73-76 34688813-0 2022 CYP3A7, CYP3A4, and CYP3A5 genetic polymorphisms in recipients rather than donors influence tacrolimus concentrations in the early stages after liver transplantation. Tacrolimus 92-102 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 0-6 34875338-13 2022 The calcineurin inhibitor FK506 exhibited a Drp1-independent function that mitigated mitochondrial dysfunction. Tacrolimus 26-31 dynamin 1 like Homo sapiens 44-48 34875338-14 2022 Finally, we found that FK506 pretreatment ameliorated the neurite growth in neurons treated with TNF and the learning ability of mice after surgery. Tacrolimus 23-28 tumor necrosis factor Mus musculus 97-100 34889045-0 2022 Tacrolimus alleviates LPS-induced AKI by inhibiting TLR4/MyD88/NF-kappaB signalling in mice. Tacrolimus 0-10 toll-like receptor 4 Mus musculus 52-56 34889045-0 2022 Tacrolimus alleviates LPS-induced AKI by inhibiting TLR4/MyD88/NF-kappaB signalling in mice. Tacrolimus 0-10 myeloid differentiation primary response gene 88 Mus musculus 57-62 34889045-0 2022 Tacrolimus alleviates LPS-induced AKI by inhibiting TLR4/MyD88/NF-kappaB signalling in mice. Tacrolimus 0-10 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 63-72 34889045-5 2022 Moreover, the Toll-like receptor 4 (TLR4)/myeloid differential protein-88 (MyD88)/nuclear factor-kappa (NF-kappaB) signalling pathway was also dramatically inhibited by medium- and high-dose TAC administration at 12, 24 and 48 h of LPS treatment mice. Tacrolimus 191-194 toll-like receptor 4 Mus musculus 14-34 34889045-5 2022 Moreover, the Toll-like receptor 4 (TLR4)/myeloid differential protein-88 (MyD88)/nuclear factor-kappa (NF-kappaB) signalling pathway was also dramatically inhibited by medium- and high-dose TAC administration at 12, 24 and 48 h of LPS treatment mice. Tacrolimus 191-194 toll-like receptor 4 Mus musculus 36-40 34889045-5 2022 Moreover, the Toll-like receptor 4 (TLR4)/myeloid differential protein-88 (MyD88)/nuclear factor-kappa (NF-kappaB) signalling pathway was also dramatically inhibited by medium- and high-dose TAC administration at 12, 24 and 48 h of LPS treatment mice. Tacrolimus 191-194 myeloid differentiation primary response gene 88 Mus musculus 75-80 34889045-5 2022 Moreover, the Toll-like receptor 4 (TLR4)/myeloid differential protein-88 (MyD88)/nuclear factor-kappa (NF-kappaB) signalling pathway was also dramatically inhibited by medium- and high-dose TAC administration at 12, 24 and 48 h of LPS treatment mice. Tacrolimus 191-194 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 104-113 34889045-7 2022 Our findings indicate that TAC has protective effects against LPS-induced AKI by inhibiting TLR4/MyD88/NF-kappaB signalling pathway and podocyte dysfunction, providing another potential therapeutic effects for the LPS-induced SA-AKI. Tacrolimus 27-30 toll-like receptor 4 Mus musculus 92-96 34889045-7 2022 Our findings indicate that TAC has protective effects against LPS-induced AKI by inhibiting TLR4/MyD88/NF-kappaB signalling pathway and podocyte dysfunction, providing another potential therapeutic effects for the LPS-induced SA-AKI. Tacrolimus 27-30 myeloid differentiation primary response gene 88 Mus musculus 97-102 34889045-7 2022 Our findings indicate that TAC has protective effects against LPS-induced AKI by inhibiting TLR4/MyD88/NF-kappaB signalling pathway and podocyte dysfunction, providing another potential therapeutic effects for the LPS-induced SA-AKI. Tacrolimus 27-30 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 103-112 34793770-2 2022 The mammalian target of rapamycin (mTOR) inhibitor, everolimus, is an immunosuppressant used together with tacrolimus. Tacrolimus 107-117 mechanistic target of rapamycin kinase Homo sapiens 4-33 34793770-2 2022 The mammalian target of rapamycin (mTOR) inhibitor, everolimus, is an immunosuppressant used together with tacrolimus. Tacrolimus 107-117 mechanistic target of rapamycin kinase Homo sapiens 35-39 34793770-7 2022 KEY FINDINGS: Tacrolimus administration increased predominant profibrotic cytokine transforming growth factor-beta (TGF-beta) and fibroblast activation marker alpha-smooth muscle actin (alpha-SMA) expression and promoted the infiltration of macrophages in the kidney cortex, resulting in renal interstitial fibrosis in rats. Tacrolimus 14-24 transforming growth factor alpha Rattus norvegicus 116-124 34793770-7 2022 KEY FINDINGS: Tacrolimus administration increased predominant profibrotic cytokine transforming growth factor-beta (TGF-beta) and fibroblast activation marker alpha-smooth muscle actin (alpha-SMA) expression and promoted the infiltration of macrophages in the kidney cortex, resulting in renal interstitial fibrosis in rats. Tacrolimus 14-24 actin gamma 2, smooth muscle Rattus norvegicus 159-184 34793770-8 2022 Tacrolimus increased serum creatinine, blood urea nitrogen, kidney injury molecule-1 (KIM-1), and kidney injuries, such as tubular dilation, vacuolization, and glomerular atrophy. Tacrolimus 0-10 hepatitis A virus cellular receptor 1 Rattus norvegicus 60-84 34793770-8 2022 Tacrolimus increased serum creatinine, blood urea nitrogen, kidney injury molecule-1 (KIM-1), and kidney injuries, such as tubular dilation, vacuolization, and glomerular atrophy. Tacrolimus 0-10 hepatitis A virus cellular receptor 1 Rattus norvegicus 86-91 34800514-0 2022 Impact of the ACE2 activator xanthenone on tacrolimus nephrotoxicity: Modulation of uric acid/ERK/p38 MAPK and Nrf2/SOD3/GCLC signaling pathways. Tacrolimus 43-53 angiotensin converting enzyme 2 Homo sapiens 14-18 34800514-0 2022 Impact of the ACE2 activator xanthenone on tacrolimus nephrotoxicity: Modulation of uric acid/ERK/p38 MAPK and Nrf2/SOD3/GCLC signaling pathways. Tacrolimus 43-53 mitogen-activated protein kinase 1 Homo sapiens 94-97 34800514-0 2022 Impact of the ACE2 activator xanthenone on tacrolimus nephrotoxicity: Modulation of uric acid/ERK/p38 MAPK and Nrf2/SOD3/GCLC signaling pathways. Tacrolimus 43-53 NFE2 like bZIP transcription factor 2 Homo sapiens 111-115 34800514-0 2022 Impact of the ACE2 activator xanthenone on tacrolimus nephrotoxicity: Modulation of uric acid/ERK/p38 MAPK and Nrf2/SOD3/GCLC signaling pathways. Tacrolimus 43-53 superoxide dismutase 3 Homo sapiens 116-120 34800514-0 2022 Impact of the ACE2 activator xanthenone on tacrolimus nephrotoxicity: Modulation of uric acid/ERK/p38 MAPK and Nrf2/SOD3/GCLC signaling pathways. Tacrolimus 43-53 glutamate-cysteine ligase catalytic subunit Homo sapiens 121-125 34800514-8 2022 Relative protein expressions of p-ERK/ERK and p-p38 MAPK/p38 MAPK inflammatory signals were downregulated upon xanthenone administration with tacrolimus. Tacrolimus 142-152 mitogen-activated protein kinase 1 Homo sapiens 34-37 34800514-8 2022 Relative protein expressions of p-ERK/ERK and p-p38 MAPK/p38 MAPK inflammatory signals were downregulated upon xanthenone administration with tacrolimus. Tacrolimus 142-152 mitogen-activated protein kinase 1 Homo sapiens 38-41 34800514-9 2022 In addition, xanthenone reinforced antioxidant defense against tacrolimus by enhancing protein expression of the transcription factor Nrf2 with subsequently increased mRNA expressions of the antioxidants SOD3 and GCLC. Tacrolimus 63-73 NFE2 like bZIP transcription factor 2 Homo sapiens 134-138 34809463-0 2021 Leveraging Fungal and Human Calcineurin-Inhibitor Structures, Biophysical Data, and Dynamics To Design Selective and Nonimmunosuppressive FK506 Analogs. Tacrolimus 138-143 calcineurin binding protein 1 Homo sapiens 28-49 34583027-1 2022 BACKGROUND: CYP3A5 and CYP3A4 are the predominant enzymes responsible for tacrolimus metabolism, however only a proportion of the population expresses CYP3A5 secondary to genetic variation. Tacrolimus 74-84 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 34583027-1 2022 BACKGROUND: CYP3A5 and CYP3A4 are the predominant enzymes responsible for tacrolimus metabolism, however only a proportion of the population expresses CYP3A5 secondary to genetic variation. Tacrolimus 74-84 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 23-29 34583027-2 2022 CYP3A5 is expressed in both the intestine and the liver and has been shown to impact both oral tacrolimus bioavailability and metabolism. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 34583027-4 2022 OBJECTIVE: The objective of this study was to evaluate the impact of CYP3A5 genotype as well as other pharmacogenes on IV tacrolimus exposure to determine whether the current genotype-guided dosing recommendations are appropriate for this formulation. Tacrolimus 122-132 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 69-75 34583027-5 2022 Additionally, this study aimed to investigate dose conversion requirements among CYP3A5 genotypes when converting from IV to PO tacrolimus. Tacrolimus 128-138 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 81-87 34583027-12 2022 RESULTS: CYP3A5 and CYP3A4 genotypes were significantly associated with the IV C/D with CYP3A5 expressers having 20% lower and CYP3A4 rapid metabolizers having 20% lower tacrolimus exposure. Tacrolimus 170-180 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 9-15 34583027-12 2022 RESULTS: CYP3A5 and CYP3A4 genotypes were significantly associated with the IV C/D with CYP3A5 expressers having 20% lower and CYP3A4 rapid metabolizers having 20% lower tacrolimus exposure. Tacrolimus 170-180 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-26 34583027-12 2022 RESULTS: CYP3A5 and CYP3A4 genotypes were significantly associated with the IV C/D with CYP3A5 expressers having 20% lower and CYP3A4 rapid metabolizers having 20% lower tacrolimus exposure. Tacrolimus 170-180 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 127-133 34809463-3 2021 Harnessing fungal calcineurin-inhibitor crystal structures, we recently developed a less immunosuppressive FK506 analog, APX879, with broad-spectrum antifungal activity and demonstrable efficacy in a murine model of invasive fungal infection. Tacrolimus 107-112 calcineurin binding protein 1 Mus musculus 18-39 34809463-5 2021 To this end, we characterized high-resolution structures of the Mucor circinelloides FKBP12 bound to FK506 and of the Aspergillus fumigatus, M. circinelloides, and human FKBP12 proteins bound to the FK506 analog APX879, which exhibits enhanced selectivity for fungal pathogens. Tacrolimus 101-106 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 85-91 34809463-5 2021 To this end, we characterized high-resolution structures of the Mucor circinelloides FKBP12 bound to FK506 and of the Aspergillus fumigatus, M. circinelloides, and human FKBP12 proteins bound to the FK506 analog APX879, which exhibits enhanced selectivity for fungal pathogens. Tacrolimus 199-204 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 170-176 34809463-12 2021 By combining structural, genetic, biophysical, and in silico methodologies, we pinpoint regions of the FK506 scaffold and a less immunosuppressive analog, APX879, centered around the C15 to C18 and C36 to C37 positions that could be altered with selective extensions and/or deletions to enhance fungal selectivity. Tacrolimus 103-108 Bardet-Biedl syndrome 9 Homo sapiens 190-193 34912030-2 2021 While expression of ARNT can be pharmacologically induced by sub-immunosuppressive doses of FK506 or by GPI1046, its anti-fibrotic activity is only realized when ARNT-ARNT homodimers form, as opposed to formation of ARNT-AHR or ARNT-HIF1alpha heterodimers. Tacrolimus 92-97 aryl hydrocarbon receptor nuclear translocator Mus musculus 20-24 34912030-2 2021 While expression of ARNT can be pharmacologically induced by sub-immunosuppressive doses of FK506 or by GPI1046, its anti-fibrotic activity is only realized when ARNT-ARNT homodimers form, as opposed to formation of ARNT-AHR or ARNT-HIF1alpha heterodimers. Tacrolimus 92-97 aryl hydrocarbon receptor nuclear translocator Mus musculus 167-171 34912030-6 2021 In murine models of kidney fibrosis, and also of liver fibrosis, combinations of FK506 or GPI1046 (to induce ARNT expression) with LB100 (to enhance ARNT homodimerization) elicit additive anti-fibrotic activities. Tacrolimus 81-86 aryl hydrocarbon receptor nuclear translocator Mus musculus 109-113 34912030-6 2021 In murine models of kidney fibrosis, and also of liver fibrosis, combinations of FK506 or GPI1046 (to induce ARNT expression) with LB100 (to enhance ARNT homodimerization) elicit additive anti-fibrotic activities. Tacrolimus 81-86 aryl hydrocarbon receptor nuclear translocator Mus musculus 149-153 34911620-4 2022 The aim of this study was to determine retinal GCL thickness by means of OCT, analyzing the proportion of affected segments in individuals exposed to tacrolimus compared with a control group. Tacrolimus 150-160 germ cell-less 1, spermatogenesis associated Homo sapiens 47-50 34905302-2 2022 The main objective of this trial was to investigate whether tacrolimus IPV decreases after switching patients from IR-tacrolimus to either ER-tacrolimus or LCP-tacrolimus. Tacrolimus 60-70 kelch domain containing 2 Homo sapiens 156-159 34905302-2 2022 The main objective of this trial was to investigate whether tacrolimus IPV decreases after switching patients from IR-tacrolimus to either ER-tacrolimus or LCP-tacrolimus. Tacrolimus 160-170 kelch domain containing 2 Homo sapiens 156-159 34905302-8 2022 The IPV of LCP-tacrolimus (20.1%) was not significantly different from the IPV of ER-tacrolimus (16.5%) (%-difference +3.6%, 95%-CI -0.1%-7.3%, p=0.06). Tacrolimus 15-25 kelch domain containing 2 Homo sapiens 11-14 34905302-9 2022 In conclusion, the IPV did not decrease after switching from immediate-release tacrolimus to either ER-tacrolimus or LCP-tacrolimus. Tacrolimus 121-131 kelch domain containing 2 Homo sapiens 117-120 34977362-7 2021 AT-induced Bdnf mRNA expression was completely blocked by d-(-)-2-Amino-5-phosphonopentanoic acid but partially blocked by nicardipine, U0126, and FK506. Tacrolimus 147-152 brain-derived neurotrophic factor Rattus norvegicus 11-15 34911620-10 2022 Linear regression analysis showed the presence of GCL segments with decreased thickness to be associated with the duration of exposure to tacrolimus (P = .036) and the time in dialysis before kidney transplant (P = .030). Tacrolimus 138-148 germ cell-less 1, spermatogenesis associated Homo sapiens 50-53 34911620-11 2022 CONCLUSION: Although this is a preliminary study, OCT scanning could serve to detect the neurotoxic effect of tacrolimus on the retinal GCL and central nervous system in renal transplant recipients. Tacrolimus 110-120 germ cell-less 1, spermatogenesis associated Homo sapiens 136-139 34956169-0 2021 Lactobacillus acidophilus Supplementation Exerts a Synergistic Effect on Tacrolimus Efficacy by Modulating Th17/Treg Balance in Lupus-Prone Mice via the SIGNR3 Pathway. Tacrolimus 73-83 CD209d antigen Mus musculus 153-159 34938174-6 2021 We found that CYP3A5*3/*3 carriers required lower doses of TAC. Tacrolimus 59-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 34950647-9 2021 Compared with the free drug at the same concentration, FK506 liposomes effectively inhibited vascular endothelial growth factor-induced green fluorescent protein-transduced human umbilical vein endothelial cell migration and tube formation in vitro. Tacrolimus 55-60 vascular endothelial growth factor A Homo sapiens 93-127 34938174-7 2021 In TAC treated patients, most CYP3A5*3/*3 carriers presented higher C0/D and a high proportion of patients with C0 levels outside the therapeutic range relative to other genotypes. Tacrolimus 3-6 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 30-36 34871120-12 2022 CONCLUSIONS: Reduced early tacrolimus exposure, facilitated by IL-2RA induction, was associated with reduced risk for HCC recurrence among patients outside Milan criteria. Tacrolimus 27-37 interleukin 2 receptor subunit alpha Homo sapiens 63-69 34797182-0 2021 Stratification of atopic dermatitis patients by patterns of response to proactive therapy with topical tacrolimus: low serum IgE levels and inadequately controlled disease activity at the start of treatment predict its failure. Tacrolimus 103-113 immunoglobulin heavy constant epsilon Homo sapiens 125-128 34859357-0 2022 The impact of cytochrome P450 3A5 genotype on early tacrolimus metabolism and clinical outcomes in lung transplant recipients. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-33 34859357-3 2022 Aim We aim to investigate the influence of cytochrome P450 3A5 (CYP3A5) genotypes on early post-LTx Tac metabolism and whether it is affected by concomitant use of azole antifungals. Tacrolimus 100-103 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 43-62 34859357-3 2022 Aim We aim to investigate the influence of cytochrome P450 3A5 (CYP3A5) genotypes on early post-LTx Tac metabolism and whether it is affected by concomitant use of azole antifungals. Tacrolimus 100-103 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 64-70 34859357-14 2022 Conclusion CYP3A5 genotype could affect Tac metabolism early after LTx. Tacrolimus 40-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 11-17 34797182-3 2021 In this study, we stratified AD patients by patterns of response to remission maintenance therapy (proactive therapy) with topical tacrolimus, a typical TCI. Tacrolimus 131-141 transcobalamin 1 Homo sapiens 153-156 34342024-1 2021 WHAT IS KNOWN AND OBJECTIVE: Tacrolimus (FK506), an effective and potent calcineurin inhibitor, is the cornerstone of immunosuppression after kidney transplantation. Tacrolimus 29-39 calcineurin binding protein 1 Homo sapiens 73-94 34120305-8 2021 Furthermore, the frequency of granzyme B+CD19+B cells correlated with the level of alanine aminotransferase instead of tacrolimus. Tacrolimus 119-129 CD19 molecule Homo sapiens 41-45 34402788-1 2021 OBJECTIVE: The calcineurin inhibitor tacrolimus has been widely used to prevent allograft rejection after transplantation. Tacrolimus 37-47 calcineurin binding protein 1 Homo sapiens 15-36 34432902-10 2021 FK506 dampened HS-induced loss of MTP and elevation of caspase-3 activity significantly (p<0.05). Tacrolimus 0-5 caspase 3 Rattus norvegicus 55-64 34432902-12 2021 The maintenance of the MTP and protection against caspase-3 mediated endothelial cell barrier disruption are possible mechanisms by which FK506 attenuates HS-induced hyperpermeability. Tacrolimus 138-143 caspase 3 Rattus norvegicus 50-59 34735949-0 2021 Computational validation of ABCB1 gene polymorphism and its effect on tacrolimus dose concentration/levels in renal transplant individuals of South India. Tacrolimus 70-80 ATP binding cassette subfamily B member 1 Homo sapiens 28-33 34673300-0 2021 Combination of matrine and tacrolimus alleviates acute rejection in murine heart transplantation by inhibiting DCs maturation through ROS/ERK/NF-kappaB pathway. Tacrolimus 27-37 mitogen-activated protein kinase 1 Mus musculus 138-141 34673300-0 2021 Combination of matrine and tacrolimus alleviates acute rejection in murine heart transplantation by inhibiting DCs maturation through ROS/ERK/NF-kappaB pathway. Tacrolimus 27-37 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 142-151 34342024-1 2021 WHAT IS KNOWN AND OBJECTIVE: Tacrolimus (FK506), an effective and potent calcineurin inhibitor, is the cornerstone of immunosuppression after kidney transplantation. Tacrolimus 41-46 calcineurin binding protein 1 Homo sapiens 73-94 34342024-11 2021 The CYP3A5 genotype showed clearly associated with tacrolimus C0 /D, whereas no significant difference was observed in patients with CYP3A4*1B, CYP3A4*22, ABCB1, ABCC2, POR*28 or PXR alleles. Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 34339091-6 2021 RESULTS: Despite persistent low tacrolimus trough levels in GSD1b patients, none of these children developed TCMR within 1 month after LDLT (GSD1b: 0/11 (0%) vs. BA: 86/273 (31.5%), p = .038). Tacrolimus 32-42 solute carrier family 37 member 4 Homo sapiens 60-65 34668283-5 2021 With increasing concentrations of tacrolimus, there was a trend to reduction in the release of IL-2 (p = 0.0137), and IFN-gamma (p = 0.0147) in response to peptide stimulation. Tacrolimus 34-44 interleukin 2 Homo sapiens 95-99 34847842-11 2021 The officially accepted mTOR inhibitors that have undergone clinical testing are sirolimus, everolimus, temsirolimus, and tacrolimus. Tacrolimus 122-132 mechanistic target of rapamycin kinase Homo sapiens 24-28 34534579-4 2021 The yeast Fpr1 (FK506-sensitive proline rotamase) is a homologue of the mammalian prolyl isomerase FKBP12 (FK506-binding protein of 12 kDa). Tacrolimus 16-21 peptidylprolyl isomerase FPR1 Saccharomyces cerevisiae S288C 10-14 34534579-4 2021 The yeast Fpr1 (FK506-sensitive proline rotamase) is a homologue of the mammalian prolyl isomerase FKBP12 (FK506-binding protein of 12 kDa). Tacrolimus 16-21 FKBP prolyl isomerase 1A Homo sapiens 99-105 34534579-4 2021 The yeast Fpr1 (FK506-sensitive proline rotamase) is a homologue of the mammalian prolyl isomerase FKBP12 (FK506-binding protein of 12 kDa). Tacrolimus 16-21 FKBP prolyl isomerase 1A Homo sapiens 107-138 34695450-9 2021 Finally, an inhibitor of calcineurin,FK506, further increased Dicer1 protein compared to Abetao treatment alone. Tacrolimus 37-42 dicer 1, ribonuclease III Homo sapiens 62-68 34854174-3 2022 Tacrolimus can cause islet cell damage and decrease in insulin secretion which can lead to post-transplant diabetes mellitus and rarely diabetic ketoacidosis. Tacrolimus 0-10 insulin Homo sapiens 55-62 34016023-8 2021 Finally, the risk of alopecia (p = 0.008), infection (p = 0.045), leukocytosis (p = 0.002), and elevated ALT/AST (p = 0.011) in TAC group was significantly lower than CTX group, whereas TAC was associated with an increased risk of tremor than CTX (p = 0.010). Tacrolimus 128-131 solute carrier family 17 member 5 Homo sapiens 109-112 34668283-5 2021 With increasing concentrations of tacrolimus, there was a trend to reduction in the release of IL-2 (p = 0.0137), and IFN-gamma (p = 0.0147) in response to peptide stimulation. Tacrolimus 34-44 interferon gamma Homo sapiens 118-127 34775880-3 2021 Calcineurin inhibitor tacrolimus (FK506) attenuated the MDI-GSH conjugate-mediated induction of CCL2, CCL3, CCL5, and CXCL8/IL8 but not others. Tacrolimus 22-32 chemokine (C-C motif) ligand 2 Mus musculus 96-100 34775880-3 2021 Calcineurin inhibitor tacrolimus (FK506) attenuated the MDI-GSH conjugate-mediated induction of CCL2, CCL3, CCL5, and CXCL8/IL8 but not others. Tacrolimus 22-32 chemokine (C-C motif) ligand 3 Mus musculus 102-106 34775880-3 2021 Calcineurin inhibitor tacrolimus (FK506) attenuated the MDI-GSH conjugate-mediated induction of CCL2, CCL3, CCL5, and CXCL8/IL8 but not others. Tacrolimus 22-32 chemokine (C-C motif) ligand 5 Mus musculus 108-112 34775880-3 2021 Calcineurin inhibitor tacrolimus (FK506) attenuated the MDI-GSH conjugate-mediated induction of CCL2, CCL3, CCL5, and CXCL8/IL8 but not others. Tacrolimus 22-32 chemokine (C-X-C motif) ligand 15 Mus musculus 124-127 34775880-3 2021 Calcineurin inhibitor tacrolimus (FK506) attenuated the MDI-GSH conjugate-mediated induction of CCL2, CCL3, CCL5, and CXCL8/IL8 but not others. Tacrolimus 34-39 chemokine (C-C motif) ligand 2 Mus musculus 96-100 34775880-3 2021 Calcineurin inhibitor tacrolimus (FK506) attenuated the MDI-GSH conjugate-mediated induction of CCL2, CCL3, CCL5, and CXCL8/IL8 but not others. Tacrolimus 34-39 chemokine (C-C motif) ligand 3 Mus musculus 102-106 34775880-3 2021 Calcineurin inhibitor tacrolimus (FK506) attenuated the MDI-GSH conjugate-mediated induction of CCL2, CCL3, CCL5, and CXCL8/IL8 but not others. Tacrolimus 34-39 chemokine (C-C motif) ligand 5 Mus musculus 108-112 34775880-3 2021 Calcineurin inhibitor tacrolimus (FK506) attenuated the MDI-GSH conjugate-mediated induction of CCL2, CCL3, CCL5, and CXCL8/IL8 but not others. Tacrolimus 34-39 chemokine (C-X-C motif) ligand 15 Mus musculus 124-127 34775880-4 2021 Transfection of either miR-inhibitor-206-3p or miR-inhibitor-381-3p in macrophages induced chemokine CCL2, CCL3, CCL5, and CXCL8 transcription, whereas FK506 attenuated the miR-inhibitor-206-3p or miR-inhibitor-381-3p-mediated effects. Tacrolimus 152-157 microRNA 615 Mus musculus 23-26 34775880-4 2021 Transfection of either miR-inhibitor-206-3p or miR-inhibitor-381-3p in macrophages induced chemokine CCL2, CCL3, CCL5, and CXCL8 transcription, whereas FK506 attenuated the miR-inhibitor-206-3p or miR-inhibitor-381-3p-mediated effects. Tacrolimus 152-157 microRNA 615 Mus musculus 47-50 34775880-4 2021 Transfection of either miR-inhibitor-206-3p or miR-inhibitor-381-3p in macrophages induced chemokine CCL2, CCL3, CCL5, and CXCL8 transcription, whereas FK506 attenuated the miR-inhibitor-206-3p or miR-inhibitor-381-3p-mediated effects. Tacrolimus 152-157 microRNA 615 Mus musculus 173-176 34775880-4 2021 Transfection of either miR-inhibitor-206-3p or miR-inhibitor-381-3p in macrophages induced chemokine CCL2, CCL3, CCL5, and CXCL8 transcription, whereas FK506 attenuated the miR-inhibitor-206-3p or miR-inhibitor-381-3p-mediated effects. Tacrolimus 152-157 microRNA 615 Mus musculus 197-200 34916931-8 2021 We found that CYP3A5*3 polymorphism was the single most strongly associated factor determining the tacrolimus C0/D in blood at all three time points (p < 0.001). Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 34339091-10 2021 CONCLUSIONS: We found that GSD1b recipients were more likely to develop postoperative bloodstream infection than recipients with BA but did not experience TCMR despite low tacrolimus levels in the early post-LDLT period. Tacrolimus 172-182 solute carrier family 37 member 4 Homo sapiens 27-32 34838267-2 2021 This prospective cohort study was to explore the effectiveness and safety of tacrolimus for treatment of IgA (Immunoglobulin A) nephropathy patients. Tacrolimus 77-87 CD79a molecule Homo sapiens 105-108 34838267-2 2021 This prospective cohort study was to explore the effectiveness and safety of tacrolimus for treatment of IgA (Immunoglobulin A) nephropathy patients. Tacrolimus 77-87 CD79a molecule Homo sapiens 110-126 34824543-5 2021 Results: The CYP3A5 genotype was clearly associated with dose-adjusted trough blood tacrolimus concentrations (C0/D), whereas no significant difference was observed in patients with the CYP3A4*1B, CYP3A4*22, ABCB1, ABCC2, POR*28 or PXR alleles. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 34824543-9 2021 Conclusion: Clinical factors, medication, and CYP-enzyme polymorphisms accounted for tacrolimus concentration variability in kidney transplantation recipients. Tacrolimus 85-95 peptidylprolyl isomerase G Homo sapiens 46-49 34607938-9 2021 In vitro treatment of healthy CD4+ and CD8+ T cells with tacrolimus abrogated the proliferation and cytokine (INF-gamma, IL-2, and TNF-alpha) secretion associated with the type 1 inflammatory phenotype observed in pre- and post-PTx rejectors. Tacrolimus 57-67 CD4 molecule Homo sapiens 30-33 34834375-7 2021 In multivariate analysis, the log Cmin/dose of tacrolimus was significantly and positively associated with the hematocrit, ALAT, and CRP concentrations. Tacrolimus 47-57 C-reactive protein Homo sapiens 133-136 34834375-8 2021 CRP concentrations were higher (p = 0.003) for patients with tacrolimus overexposure (i.e., tacrolimus Cmin > 15 microg/L) (median CRP (10th-90th percentiles): 27 mg/L (3-149 mg/L), n = 91) than they were for patients with a tacrolimus Cmin <= 15 microg/L (13 mg/mL (3-95 mg/L), n = 1482)). Tacrolimus 61-71 C-reactive protein Homo sapiens 0-3 34834375-8 2021 CRP concentrations were higher (p = 0.003) for patients with tacrolimus overexposure (i.e., tacrolimus Cmin > 15 microg/L) (median CRP (10th-90th percentiles): 27 mg/L (3-149 mg/L), n = 91) than they were for patients with a tacrolimus Cmin <= 15 microg/L (13 mg/mL (3-95 mg/L), n = 1482)). Tacrolimus 92-102 C-reactive protein Homo sapiens 0-3 34834375-9 2021 CRP in the fourth quartile (49 to 334 mg/L) was associated with a 2.6-fold increased risk of tacrolimus Cmin overexposure. Tacrolimus 93-103 C-reactive protein Homo sapiens 0-3 34607938-9 2021 In vitro treatment of healthy CD4+ and CD8+ T cells with tacrolimus abrogated the proliferation and cytokine (INF-gamma, IL-2, and TNF-alpha) secretion associated with the type 1 inflammatory phenotype observed in pre- and post-PTx rejectors. Tacrolimus 57-67 CD8a molecule Homo sapiens 39-42 34607938-9 2021 In vitro treatment of healthy CD4+ and CD8+ T cells with tacrolimus abrogated the proliferation and cytokine (INF-gamma, IL-2, and TNF-alpha) secretion associated with the type 1 inflammatory phenotype observed in pre- and post-PTx rejectors. Tacrolimus 57-67 interleukin 2 Homo sapiens 121-125 34607938-9 2021 In vitro treatment of healthy CD4+ and CD8+ T cells with tacrolimus abrogated the proliferation and cytokine (INF-gamma, IL-2, and TNF-alpha) secretion associated with the type 1 inflammatory phenotype observed in pre- and post-PTx rejectors. Tacrolimus 57-67 tumor necrosis factor Homo sapiens 131-140 34830130-10 2021 Differential effects of FK506-binding immunophilins, FKBP4 and FKBP5, contribute to the efficiency of glucocorticoids under stress resilience. Tacrolimus 24-29 FKBP prolyl isomerase 4 Homo sapiens 53-58 34830130-10 2021 Differential effects of FK506-binding immunophilins, FKBP4 and FKBP5, contribute to the efficiency of glucocorticoids under stress resilience. Tacrolimus 24-29 FKBP prolyl isomerase 5 Homo sapiens 63-68 34841735-5 2021 The increased percentage of senescent CD8+ CD57+ T lineages and less responsive T cell receptor (TCR) pathway in the FK506 group indicate better graft acceptance. Tacrolimus 117-122 CD8a molecule Homo sapiens 38-41 34648292-3 2021 However, CYP3A5 preferentially metabolizes several clinically prescribed drugs, such as tacrolimus. Tacrolimus 88-98 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 9-15 34648292-4 2021 Genetic polymorphism in CYP3A5 makes race-based dosing adjustment of tacrolimus necessary to minimize acute rejection after organ transplantation. Tacrolimus 69-79 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-30 34732168-11 2021 A more significant reduction of MuSK-ab titers was observed in patients receiving TAC or RTX plus Pred than those receiving AZA plus Pred. Tacrolimus 82-85 muscle associated receptor tyrosine kinase Homo sapiens 32-36 34133842-0 2021 Effects of Postoperative Day and NR1I2 on Tacrolimus Clearance in Chinese Liver Transplant Recipients-A Population Model Approach. Tacrolimus 42-52 nuclear receptor subfamily 1 group I member 2 Homo sapiens 33-38 34133842-10 2021 To our knowledge, this is the first report indicating NR1I2 rs2276707 genotypes is another biomarker impacting tacrolimus clearance in liver transplant recipients. Tacrolimus 111-121 nuclear receptor subfamily 1 group I member 2 Homo sapiens 54-59 34133842-11 2021 The NR1I2 gene polymorphism may affect the in vivo behavior of tacrolimus by regulating gene expression. Tacrolimus 63-73 nuclear receptor subfamily 1 group I member 2 Homo sapiens 4-9 34841735-5 2021 The increased percentage of senescent CD8+ CD57+ T lineages and less responsive T cell receptor (TCR) pathway in the FK506 group indicate better graft acceptance. Tacrolimus 117-122 beta-1,3-glucuronyltransferase 1 Homo sapiens 43-47 34841735-6 2021 Meanwhile, percentages of regulatory T cells (Tregs) and expression of CTLA-4 were both up to two-fold higher in the RAPA group, suggesting the inconsistent reactivation potential of the FK506 and RAPA groups when an anti-tumour or anti-infection immune response is concerned. Tacrolimus 187-192 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 71-77 34435330-0 2021 Tacrolimus Decreases Cognitive Function by Impairing Hippocampal Synaptic Balance: a Possible Role of Klotho. Tacrolimus 0-10 klotho Mus musculus 102-108 34662453-0 2021 Tacrolimus ameliorates podocyte injury by restoring FK506 binding protein 12 (FKBP12) at actin cytoskeleton. Tacrolimus 0-10 FKBP prolyl isomerase 1A Homo sapiens 52-76 34662453-0 2021 Tacrolimus ameliorates podocyte injury by restoring FK506 binding protein 12 (FKBP12) at actin cytoskeleton. Tacrolimus 0-10 FKBP prolyl isomerase 1A Homo sapiens 78-84 34662453-1 2021 FKBP12 was identified as a binding protein of tacrolimus (Tac). Tacrolimus 46-56 FKBP prolyl isomerase 1A Homo sapiens 0-6 34662453-1 2021 FKBP12 was identified as a binding protein of tacrolimus (Tac). Tacrolimus 58-61 FKBP prolyl isomerase 1A Homo sapiens 0-6 34662453-2 2021 Tac binds to FKBP12 and exhibits immunosuppressive effects in T cells. Tacrolimus 0-3 FKBP prolyl isomerase 1A Homo sapiens 13-19 34662453-13 2021 Tac treatment to the normal cells increased the expression of FKBP12 at F-actin in processes and enhanced process formation. Tacrolimus 0-3 FKBP prolyl isomerase 1A Homo sapiens 62-68 34662453-14 2021 Tac enhanced the interaction of FKBP12 with synaptopodin. Tacrolimus 0-3 FKBP prolyl isomerase 1A Homo sapiens 32-38 34662453-15 2021 These observations suggested that FKBP12 at actin cytoskeleton participates in the maintenance of processes, and Tac treatment ameliorates podocyte injury by restoring FKBP12 at actin cytoskeleton. Tacrolimus 113-116 FKBP prolyl isomerase 1A Homo sapiens 168-174 34435330-6 2021 These results indicate that tacrolimus impairs cognitive function via synaptic imbalance, and that these processes are associated with Klotho downregulation at synapses through tacrolimus-induced oxidative stress in the hippocampus. Tacrolimus 177-187 klotho Mus musculus 135-141 34435330-4 2021 Furthermore, tacrolimus caused synaptic imbalance, as demonstrated by decreased excitatory synapses and increased inhibitory synapses, and downregulated Klotho in a dose-dependent manner; the downregulation of Klotho was localized to excitatory hippocampal synapses. Tacrolimus 13-23 klotho Mus musculus 153-159 34435330-4 2021 Furthermore, tacrolimus caused synaptic imbalance, as demonstrated by decreased excitatory synapses and increased inhibitory synapses, and downregulated Klotho in a dose-dependent manner; the downregulation of Klotho was localized to excitatory hippocampal synapses. Tacrolimus 13-23 klotho Mus musculus 210-216 34435330-5 2021 Moreover, tacrolimus increased oxidative stress and was associated with activation of the PI3K/AKT pathway in the hippocampus. Tacrolimus 10-20 thymoma viral proto-oncogene 1 Mus musculus 95-98 34435330-6 2021 These results indicate that tacrolimus impairs cognitive function via synaptic imbalance, and that these processes are associated with Klotho downregulation at synapses through tacrolimus-induced oxidative stress in the hippocampus. Tacrolimus 28-38 klotho Mus musculus 135-141 34725418-0 2021 CYP3A-status is associated with blood concentration and dose-requirement of tacrolimus in heart transplant recipients. Tacrolimus 76-86 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-5 34725418-4 2021 In low CYP3A4 expressers carrying CYP3A5*3/*3, the dose-corrected tacrolimus level was significantly higher than in normal CYP3A4 expressers or in those with CYP3A5*1. Tacrolimus 66-76 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 7-13 34725418-4 2021 In low CYP3A4 expressers carrying CYP3A5*3/*3, the dose-corrected tacrolimus level was significantly higher than in normal CYP3A4 expressers or in those with CYP3A5*1. Tacrolimus 66-76 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 34725418-5 2021 Modification of the initial tacrolimus dose was required for all patients: dose reduction by 20% for low CYP3A4 expressers, a 40% increase for normal expressers and a 2.4-fold increase for CYP3A5*1 carriers. Tacrolimus 28-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-111 34725418-5 2021 Modification of the initial tacrolimus dose was required for all patients: dose reduction by 20% for low CYP3A4 expressers, a 40% increase for normal expressers and a 2.4-fold increase for CYP3A5*1 carriers. Tacrolimus 28-38 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 189-195 34725418-1 2021 High inter-individual variability in tacrolimus clearance is attributed to genetic polymorphisms of CYP3A enzymes. Tacrolimus 37-47 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 100-105 34725418-2 2021 However, due to CYP3A phenoconversion induced by non-genetic factors, continuous changes in tacrolimus-metabolizing capacity entail frequent dose-refinement for optimal immunosuppression. Tacrolimus 92-102 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 16-21 34683143-1 2021 The pharmacokinetic variability of tacrolimus can be partly explained by CYP3A5 activity. Tacrolimus 35-45 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 73-79 34725418-7 2021 The fluctuation of CYP3A4 expression and tacrolimus blood concentration (C0/D) was found to be associated with tapering and cessation of corticosteroid in CYP3A5 non-expressers, but not in those carrying CYP3A5*1. Tacrolimus 41-51 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 155-161 34725418-8 2021 Although monitoring of tacrolimus blood concentration cannot be omitted, assaying recipients" CYP3A-status can guide optimization of the initial tacrolimus dose, and can facilitate personalized tacrolimus therapy during steroid withdrawal in the late post-operative period. Tacrolimus 145-155 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 94-99 34269803-8 2021 This synonymous variant in P-glycoprotein may influence the risk of CMV reactivation by altering the efflux of cyclosporine and tacrolimus from donor lymphocytes. Tacrolimus 128-138 ATP binding cassette subfamily B member 1 Homo sapiens 27-41 34788922-0 2021 (The effect of CYP3A5 gene polymorphism on tacrolimus concentration and adverse events in patients undergoing allogeneic hematopoietic stem cell transplantation). Tacrolimus 43-53 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 34788922-8 2021 Conclusion: CYP3A5 genotype-directed administration may help achieve the target blood concentration of tacrolimus after HSCT more quickly, reduce the incidence of severe aGVHD, and improve the efficacy of transplantation. Tacrolimus 103-113 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 34708436-11 2022 Furthermore, a larger proportion of patients in the CYP3A5 expression groups required Tac dose adjustment to achieve a therapeutic effect and were given Tac with WZC. Tacrolimus 86-89 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 52-58 34708436-11 2022 Furthermore, a larger proportion of patients in the CYP3A5 expression groups required Tac dose adjustment to achieve a therapeutic effect and were given Tac with WZC. Tacrolimus 153-156 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 52-58 34500058-7 2021 Tacrolimus was loaded into MSNs and the particles were characterized for particle size (TEM and DLS), zeta potential (DLS), solubility studies, FTIR, TGA, XRD, BET and cytotoxicity studies. Tacrolimus 0-10 delta/notch-like EGF repeat containing Mus musculus 160-163 34690808-9 2021 Diminished expression of the FK506 binding protein, FKBP12.6, may also contribute. Tacrolimus 29-34 FKBP prolyl isomerase 1B Homo sapiens 52-60 34683143-13 2021 Our study raised some issues about specific therapeutic tacrolimus C0 targets for CYP3A5*1/- patients and suggests to set up randomized control studies in this specific population. Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 82-88 34352707-5 2021 Tacrolimus trough levels increased only 0.3 ng/mL upon conversion in the CYP3A5*3/*3 group: 5.8 (3.4-7.2) vs 6.1 (3.8-7.9); p = 0.06. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 73-79 34709766-0 2021 The CYP3A5 genotypes of both liver transplant recipients and donors influence the time-dependent recovery of tacrolimus clearance during the early stage following transplantation. Tacrolimus 109-119 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 34575860-4 2021 We hypothesized that CsA and Tac induce functional defects and activate an inflammatory cascade via NF-kappaB signaling in ECFCs. Tacrolimus 29-32 nuclear factor kappa B subunit 1 Homo sapiens 100-109 34228988-0 2021 Combination of mesenchymal stem cells and FK506 prolongs heart allograft survival by inhibiting TBK1/IRF3-regulated-IFN-gamma production. Tacrolimus 42-47 TANK-binding kinase 1 Mus musculus 96-100 34228988-0 2021 Combination of mesenchymal stem cells and FK506 prolongs heart allograft survival by inhibiting TBK1/IRF3-regulated-IFN-gamma production. Tacrolimus 42-47 interferon regulatory factor 3 Mus musculus 101-105 34228988-0 2021 Combination of mesenchymal stem cells and FK506 prolongs heart allograft survival by inhibiting TBK1/IRF3-regulated-IFN-gamma production. Tacrolimus 42-47 interferon gamma Mus musculus 116-125 34228988-6 2021 Thereafter, in vitro and in vivo experiments were conducted to identify the mechanisms regarding MSC and FK506 combination (MF group) use in regulating IFN-gamma signaling. Tacrolimus 105-110 interferon gamma Mus musculus 152-161 34228988-11 2021 Combination use of MSC and FK506 can prolong graft survival, possibly by down-regulating TBK1/IRF3 phosphorylation, thus reducing IFN-gamma production to prevent infiltration of inflammatory cells in the graft and extend graft survival. Tacrolimus 27-32 TANK-binding kinase 1 Mus musculus 89-93 34228988-11 2021 Combination use of MSC and FK506 can prolong graft survival, possibly by down-regulating TBK1/IRF3 phosphorylation, thus reducing IFN-gamma production to prevent infiltration of inflammatory cells in the graft and extend graft survival. Tacrolimus 27-32 interferon regulatory factor 3 Mus musculus 94-98 34228988-11 2021 Combination use of MSC and FK506 can prolong graft survival, possibly by down-regulating TBK1/IRF3 phosphorylation, thus reducing IFN-gamma production to prevent infiltration of inflammatory cells in the graft and extend graft survival. Tacrolimus 27-32 interferon gamma Mus musculus 130-139 34481645-1 2021 Calcineurin-inhibitor induced pain syndrome (CIPS) also called the "symmetrical bone syndrome" is a condition describing reversible lower extremity pain in patients after organ transplantation who are receiving calcineurin inhibitors, especially tacrolimus. Tacrolimus 246-256 calcineurin binding protein 1 Homo sapiens 0-21 34620272-3 2021 Cytochrome-P450 (CYP) isoenzymes CYP3A4 and CYP3A5, as well P-glycoprotein (P-gp) are involved in TAC bioavailability. Tacrolimus 98-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 34620272-3 2021 Cytochrome-P450 (CYP) isoenzymes CYP3A4 and CYP3A5, as well P-glycoprotein (P-gp) are involved in TAC bioavailability. Tacrolimus 98-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 34620272-3 2021 Cytochrome-P450 (CYP) isoenzymes CYP3A4 and CYP3A5, as well P-glycoprotein (P-gp) are involved in TAC bioavailability. Tacrolimus 98-101 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 33-39 34620272-3 2021 Cytochrome-P450 (CYP) isoenzymes CYP3A4 and CYP3A5, as well P-glycoprotein (P-gp) are involved in TAC bioavailability. Tacrolimus 98-101 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 44-50 34620272-3 2021 Cytochrome-P450 (CYP) isoenzymes CYP3A4 and CYP3A5, as well P-glycoprotein (P-gp) are involved in TAC bioavailability. Tacrolimus 98-101 ATP binding cassette subfamily B member 1 Homo sapiens 60-74 34575860-11 2021 Furthermore, CsA and Tac led to NF-kappaB p65 subunit phosphorylation and nuclear translocation. Tacrolimus 21-24 RELA proto-oncogene, NF-kB subunit Homo sapiens 32-45 34575860-12 2021 Pharmacological inhibition of NF-kappaB by parthenolide diminished CsA- and Tac-mediated proinflammatory effects. Tacrolimus 76-79 nuclear factor kappa B subunit 1 Homo sapiens 30-39 34278483-13 2021 Furthermore, the western blotting results suggested that tacrolimus had no significant effects on the expression levels of total PI3K, Akt and mTOR proteins (P>0.05), but 25 and 50 ng/ml tacrolimus could significantly inhibit the expression levels of p-Akt and p-mTOR (P<0.01). Tacrolimus 187-197 thymoma viral proto-oncogene 1 Mus musculus 253-256 34511980-6 2021 Our final model identified creatinine clearance rate, hematocrit, Wuzhi capsule dose, CYP3A5*3 genetic polymorphisms, and tacrolimus daily dose as significant covariates for tacrolimus clearance, with the value of 14.4 L h-1, and the between-subject variability (BSV) was 25.4%. Tacrolimus 174-184 procollagen-lysine,2-oxoglutarate 5-dioxygenase 1 Homo sapiens 219-224 34136903-0 2021 A 24-Hour Extended Calibration Strategy for Quantitating Tacrolimus Concentrations by Liquid Chromatography-Tandem Mass Spectrometry. Tacrolimus 57-67 immunoglobulin kappa variable 2-23 (pseudogene) Homo sapiens 0-4 34136903-5 2021 A 24-h extended calibration of LC-MS/MS tacrolimus was designed and validated to reduce calibrator usage, improve turnaround time, and provide a more efficient workflow for urgent requests. Tacrolimus 40-50 immunoglobulin kappa variable 2-23 (pseudogene) Homo sapiens 0-4 34217824-2 2021 A FK506/HP-beta-CD inclusion compound was prepared by grinding to increase drug solubility. Tacrolimus 2-7 adrenocortical dysplasia Mus musculus 11-18 34217824-3 2021 To address the side- effects in non-target organs and systemic toxicity of FK506, pH-responsive Eudragit S100 (ES100) and hyaluronic acid (HA) with high affinity to CD44 receptor were adsorbed onto the surface of chitosan (CS) nanoparticles loaded with FK506/HP-beta-CD through electrostatic interactions to obtain FK506@ES100/HA/CS/HP-beta-CD nanoparticles (FK506@EHCh NPs). Tacrolimus 315-320 CD44 antigen Mus musculus 165-169 34217824-3 2021 To address the side- effects in non-target organs and systemic toxicity of FK506, pH-responsive Eudragit S100 (ES100) and hyaluronic acid (HA) with high affinity to CD44 receptor were adsorbed onto the surface of chitosan (CS) nanoparticles loaded with FK506/HP-beta-CD through electrostatic interactions to obtain FK506@ES100/HA/CS/HP-beta-CD nanoparticles (FK506@EHCh NPs). Tacrolimus 315-320 adrenocortical dysplasia Mus musculus 262-269 34217824-3 2021 To address the side- effects in non-target organs and systemic toxicity of FK506, pH-responsive Eudragit S100 (ES100) and hyaluronic acid (HA) with high affinity to CD44 receptor were adsorbed onto the surface of chitosan (CS) nanoparticles loaded with FK506/HP-beta-CD through electrostatic interactions to obtain FK506@ES100/HA/CS/HP-beta-CD nanoparticles (FK506@EHCh NPs). Tacrolimus 359-364 adrenocortical dysplasia Mus musculus 336-343 34217824-5 2021 FK506@EHCh NPs significantly suppressed secretion of TNF-alpha, IL-1beta and IL-6 by LPS-activated Raw 264.7 macrophages. Tacrolimus 0-5 tumor necrosis factor Mus musculus 53-62 34217824-5 2021 FK506@EHCh NPs significantly suppressed secretion of TNF-alpha, IL-1beta and IL-6 by LPS-activated Raw 264.7 macrophages. Tacrolimus 0-5 interleukin 1 alpha Mus musculus 64-72 34217824-5 2021 FK506@EHCh NPs significantly suppressed secretion of TNF-alpha, IL-1beta and IL-6 by LPS-activated Raw 264.7 macrophages. Tacrolimus 0-5 interleukin 6 Mus musculus 77-81 34278483-0 2021 Tacrolimus inhibits insulin release and promotes apoptosis of Min6 cells through the inhibition of the PI3K/Akt/mTOR pathway. Tacrolimus 0-10 thymoma viral proto-oncogene 1 Mus musculus 108-111 34278483-0 2021 Tacrolimus inhibits insulin release and promotes apoptosis of Min6 cells through the inhibition of the PI3K/Akt/mTOR pathway. Tacrolimus 0-10 mechanistic target of rapamycin kinase Mus musculus 112-116 34278483-1 2021 As a calcineurin inhibitor, tacrolimus is commonly used as a first-line immunosuppressant in organ transplant recipients. Tacrolimus 28-38 calcineurin binding protein 1 Mus musculus 5-26 34278483-5 2021 The aim of the present study was to investigate whether the PI3K/Akt/mTOR signaling pathway served an important role in the pathogenesis of PTDM induced by tacrolimus. Tacrolimus 156-166 thymoma viral proto-oncogene 1 Mus musculus 65-68 34278483-5 2021 The aim of the present study was to investigate whether the PI3K/Akt/mTOR signaling pathway served an important role in the pathogenesis of PTDM induced by tacrolimus. Tacrolimus 156-166 mechanistic target of rapamycin kinase Mus musculus 69-73 34278483-13 2021 Furthermore, the western blotting results suggested that tacrolimus had no significant effects on the expression levels of total PI3K, Akt and mTOR proteins (P>0.05), but 25 and 50 ng/ml tacrolimus could significantly inhibit the expression levels of p-Akt and p-mTOR (P<0.01). Tacrolimus 187-197 mechanistic target of rapamycin kinase Mus musculus 263-267 34278483-7 2021 The effects of tacrolimus on the insulin secretion and the activity of caspase-3 of Min6 cells stimulated by glucose exposure were measured by ELISA. Tacrolimus 15-25 caspase 3 Mus musculus 71-80 34278483-14 2021 In conclusion, tacrolimus decreased the activity and insulin secretion of pancreatic beta cells and induced the apoptosis of islet beta cells by inhibiting the mRNA expression levels of PI3K, Akt and mTOR and reducing the phosphorylation of Akt and mTOR proteins in the PI3K/Akt/mTOR signaling pathway, which may ultimately lead to the occurrence of diabetes mellitus, and may be considered as one of the specific mechanisms of PTDM caused by tacrolimus. Tacrolimus 15-25 thymoma viral proto-oncogene 1 Mus musculus 192-195 34278483-9 2021 The effects of tacrolimus on the mRNA expression levels of PI3K, Akt and mTOR were detected by reverse transcription-quantitative PCR (RT-qPCR), whereas the protein expression levels of PI3K, Akt, mTOR, phosphorylated (p)-AKT and p-mTOR in Min6 cells were assessed using western blotting. Tacrolimus 15-25 thymoma viral proto-oncogene 1 Mus musculus 65-68 34278483-14 2021 In conclusion, tacrolimus decreased the activity and insulin secretion of pancreatic beta cells and induced the apoptosis of islet beta cells by inhibiting the mRNA expression levels of PI3K, Akt and mTOR and reducing the phosphorylation of Akt and mTOR proteins in the PI3K/Akt/mTOR signaling pathway, which may ultimately lead to the occurrence of diabetes mellitus, and may be considered as one of the specific mechanisms of PTDM caused by tacrolimus. Tacrolimus 15-25 mechanistic target of rapamycin kinase Mus musculus 200-204 34278483-9 2021 The effects of tacrolimus on the mRNA expression levels of PI3K, Akt and mTOR were detected by reverse transcription-quantitative PCR (RT-qPCR), whereas the protein expression levels of PI3K, Akt, mTOR, phosphorylated (p)-AKT and p-mTOR in Min6 cells were assessed using western blotting. Tacrolimus 15-25 mechanistic target of rapamycin kinase Mus musculus 73-77 34278483-9 2021 The effects of tacrolimus on the mRNA expression levels of PI3K, Akt and mTOR were detected by reverse transcription-quantitative PCR (RT-qPCR), whereas the protein expression levels of PI3K, Akt, mTOR, phosphorylated (p)-AKT and p-mTOR in Min6 cells were assessed using western blotting. Tacrolimus 15-25 thymoma viral proto-oncogene 1 Mus musculus 222-225 34278483-11 2021 Moreover, 50 ng/ml tacrolimus markedly increased the activity of caspase-3 by 175.1% (P<0.05), it also decreased the SOD activity (P<0.01) and increased MDA levels (P<0.05). Tacrolimus 19-29 caspase 3 Mus musculus 65-74 34278483-12 2021 The RT-qPCR results demonstrated that the mRNA expression levels of PI3K, Akt and mTOR were downregulated by 25 and 50 ng/ml tacrolimus (P<0.01). Tacrolimus 125-135 thymoma viral proto-oncogene 1 Mus musculus 74-77 34278483-12 2021 The RT-qPCR results demonstrated that the mRNA expression levels of PI3K, Akt and mTOR were downregulated by 25 and 50 ng/ml tacrolimus (P<0.01). Tacrolimus 125-135 mechanistic target of rapamycin kinase Mus musculus 82-86 34278483-14 2021 In conclusion, tacrolimus decreased the activity and insulin secretion of pancreatic beta cells and induced the apoptosis of islet beta cells by inhibiting the mRNA expression levels of PI3K, Akt and mTOR and reducing the phosphorylation of Akt and mTOR proteins in the PI3K/Akt/mTOR signaling pathway, which may ultimately lead to the occurrence of diabetes mellitus, and may be considered as one of the specific mechanisms of PTDM caused by tacrolimus. Tacrolimus 15-25 thymoma viral proto-oncogene 1 Mus musculus 241-244 34278483-14 2021 In conclusion, tacrolimus decreased the activity and insulin secretion of pancreatic beta cells and induced the apoptosis of islet beta cells by inhibiting the mRNA expression levels of PI3K, Akt and mTOR and reducing the phosphorylation of Akt and mTOR proteins in the PI3K/Akt/mTOR signaling pathway, which may ultimately lead to the occurrence of diabetes mellitus, and may be considered as one of the specific mechanisms of PTDM caused by tacrolimus. Tacrolimus 15-25 mechanistic target of rapamycin kinase Mus musculus 249-253 34278483-14 2021 In conclusion, tacrolimus decreased the activity and insulin secretion of pancreatic beta cells and induced the apoptosis of islet beta cells by inhibiting the mRNA expression levels of PI3K, Akt and mTOR and reducing the phosphorylation of Akt and mTOR proteins in the PI3K/Akt/mTOR signaling pathway, which may ultimately lead to the occurrence of diabetes mellitus, and may be considered as one of the specific mechanisms of PTDM caused by tacrolimus. Tacrolimus 15-25 thymoma viral proto-oncogene 1 Mus musculus 275-278 34278483-14 2021 In conclusion, tacrolimus decreased the activity and insulin secretion of pancreatic beta cells and induced the apoptosis of islet beta cells by inhibiting the mRNA expression levels of PI3K, Akt and mTOR and reducing the phosphorylation of Akt and mTOR proteins in the PI3K/Akt/mTOR signaling pathway, which may ultimately lead to the occurrence of diabetes mellitus, and may be considered as one of the specific mechanisms of PTDM caused by tacrolimus. Tacrolimus 15-25 mechanistic target of rapamycin kinase Mus musculus 279-283 34278483-14 2021 In conclusion, tacrolimus decreased the activity and insulin secretion of pancreatic beta cells and induced the apoptosis of islet beta cells by inhibiting the mRNA expression levels of PI3K, Akt and mTOR and reducing the phosphorylation of Akt and mTOR proteins in the PI3K/Akt/mTOR signaling pathway, which may ultimately lead to the occurrence of diabetes mellitus, and may be considered as one of the specific mechanisms of PTDM caused by tacrolimus. Tacrolimus 443-453 thymoma viral proto-oncogene 1 Mus musculus 192-195 34278483-14 2021 In conclusion, tacrolimus decreased the activity and insulin secretion of pancreatic beta cells and induced the apoptosis of islet beta cells by inhibiting the mRNA expression levels of PI3K, Akt and mTOR and reducing the phosphorylation of Akt and mTOR proteins in the PI3K/Akt/mTOR signaling pathway, which may ultimately lead to the occurrence of diabetes mellitus, and may be considered as one of the specific mechanisms of PTDM caused by tacrolimus. Tacrolimus 443-453 mechanistic target of rapamycin kinase Mus musculus 200-204 34278483-14 2021 In conclusion, tacrolimus decreased the activity and insulin secretion of pancreatic beta cells and induced the apoptosis of islet beta cells by inhibiting the mRNA expression levels of PI3K, Akt and mTOR and reducing the phosphorylation of Akt and mTOR proteins in the PI3K/Akt/mTOR signaling pathway, which may ultimately lead to the occurrence of diabetes mellitus, and may be considered as one of the specific mechanisms of PTDM caused by tacrolimus. Tacrolimus 443-453 thymoma viral proto-oncogene 1 Mus musculus 241-244 34278483-14 2021 In conclusion, tacrolimus decreased the activity and insulin secretion of pancreatic beta cells and induced the apoptosis of islet beta cells by inhibiting the mRNA expression levels of PI3K, Akt and mTOR and reducing the phosphorylation of Akt and mTOR proteins in the PI3K/Akt/mTOR signaling pathway, which may ultimately lead to the occurrence of diabetes mellitus, and may be considered as one of the specific mechanisms of PTDM caused by tacrolimus. Tacrolimus 443-453 mechanistic target of rapamycin kinase Mus musculus 249-253 34278483-14 2021 In conclusion, tacrolimus decreased the activity and insulin secretion of pancreatic beta cells and induced the apoptosis of islet beta cells by inhibiting the mRNA expression levels of PI3K, Akt and mTOR and reducing the phosphorylation of Akt and mTOR proteins in the PI3K/Akt/mTOR signaling pathway, which may ultimately lead to the occurrence of diabetes mellitus, and may be considered as one of the specific mechanisms of PTDM caused by tacrolimus. Tacrolimus 443-453 thymoma viral proto-oncogene 1 Mus musculus 275-278 34278483-14 2021 In conclusion, tacrolimus decreased the activity and insulin secretion of pancreatic beta cells and induced the apoptosis of islet beta cells by inhibiting the mRNA expression levels of PI3K, Akt and mTOR and reducing the phosphorylation of Akt and mTOR proteins in the PI3K/Akt/mTOR signaling pathway, which may ultimately lead to the occurrence of diabetes mellitus, and may be considered as one of the specific mechanisms of PTDM caused by tacrolimus. Tacrolimus 443-453 mechanistic target of rapamycin kinase Mus musculus 279-283 34342774-3 2021 FKBP2 (FKBP13) and FKBP1 (FKBP12), known as immunophilins, are binding proteins for rapamycin and FK506, which are immunosuppressive drugs. Tacrolimus 98-103 FKBP prolyl isomerase 2 Homo sapiens 0-5 34931247-1 2021 OBJECTIVES: We retrospectively compared the therapeutic effects of combination therapy with prednisolone (PSL) and oral tacrolimus (TAC) or azathioprine (AZA) on progressive interstitial pneumonia with systemic sclerosis (SSc-PIP). Tacrolimus 120-130 prolactin induced protein Homo sapiens 226-229 34931247-7 2021 CONCLUSION: The inhibitory effect of PSL and TAC combination therapy on the progression of fibrosis in SSc-PIP may be superior to that of PSL and AZA in the long period. Tacrolimus 45-48 prolactin induced protein Homo sapiens 107-110 34483924-5 2021 We present the case of a 16-year-old girl with kidney and liver transplant in whom low concentrations of tacrolimus in the context of low hematocrit led to significant increase in the dosage of tacrolimus and participate, along with a genetic polymorphism of ABCB1, in nephrotoxicity. Tacrolimus 105-115 ATP binding cassette subfamily B member 1 Homo sapiens 259-264 34144106-4 2021 Among many immunosuppressants, tacrolimus (FK506) is one of the most potent interleukin-2 (IL-2)-mediated T-cell proliferation blockers. Tacrolimus 31-41 interleukin 2 Mus musculus 76-89 34144106-4 2021 Among many immunosuppressants, tacrolimus (FK506) is one of the most potent interleukin-2 (IL-2)-mediated T-cell proliferation blockers. Tacrolimus 31-41 interleukin 2 Mus musculus 91-95 34144106-4 2021 Among many immunosuppressants, tacrolimus (FK506) is one of the most potent interleukin-2 (IL-2)-mediated T-cell proliferation blockers. Tacrolimus 43-48 interleukin 2 Mus musculus 76-89 34144106-4 2021 Among many immunosuppressants, tacrolimus (FK506) is one of the most potent interleukin-2 (IL-2)-mediated T-cell proliferation blockers. Tacrolimus 43-48 interleukin 2 Mus musculus 91-95 34144106-8 2021 Interestingly, FK506-M at very low-doses (equivalent to 150 to 2400 ng FK506 per recipient) was found to inhibit the infiltration of immune cells into grafts and reduce serum IL-1beta levels, thereby improving graft survival times dose-dependently. Tacrolimus 15-20 interleukin 1 alpha Mus musculus 175-183 34144106-8 2021 Interestingly, FK506-M at very low-doses (equivalent to 150 to 2400 ng FK506 per recipient) was found to inhibit the infiltration of immune cells into grafts and reduce serum IL-1beta levels, thereby improving graft survival times dose-dependently. Tacrolimus 71-76 interleukin 1 alpha Mus musculus 175-183 34362271-3 2022 OBJECTIVE: To detect genotypes of cytochrome P450 3A5 (CYP3A5) and ABCB1 in kidney transplant patients and establish initial daily tacrolimus dosing formula based on genotypes of CYP3A5 and ABCB1 and patients" clinical parameters. Tacrolimus 131-141 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-53 34362271-3 2022 OBJECTIVE: To detect genotypes of cytochrome P450 3A5 (CYP3A5) and ABCB1 in kidney transplant patients and establish initial daily tacrolimus dosing formula based on genotypes of CYP3A5 and ABCB1 and patients" clinical parameters. Tacrolimus 131-141 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 179-185 34342774-3 2021 FKBP2 (FKBP13) and FKBP1 (FKBP12), known as immunophilins, are binding proteins for rapamycin and FK506, which are immunosuppressive drugs. Tacrolimus 98-103 FKBP prolyl isomerase 1A Homo sapiens 19-24 34342774-5 2021 Within the 15 mammalian FKBPs known, FKBP1 is merely the only one proven to form complexes with rapamycin and FK506 in the cytosol and facilitate their T cells immunosuppressive effects, FKBP2 is a luminal protein of the endoplasmic reticulum (ER) and is reported to take part in protein folding in the ER. Tacrolimus 110-115 FKBP prolyl isomerase 1A Homo sapiens 37-42 34804426-3 2021 The cytotoxic effect of TAC and protective effect of EAF co-treatment were studied in MDCK cell lines by measuring ROS, LPO, and NO levels; collagen accumulation, effect on mitochondrial membrane integrity and cell cycle analysis were studied. Tacrolimus 24-27 lactoperoxidase Canis lupus familiaris 120-123 34488389-11 2021 However, the expression of Bcl-2 increased when HC-030031, pifithrin-alpha, or FK506 was used alone, and HC-030031 combined with pifithrin-alpha or FK506 further improved the expression of Bcl-2. Tacrolimus 79-84 BCL2 apoptosis regulator Homo sapiens 27-32 34488389-11 2021 However, the expression of Bcl-2 increased when HC-030031, pifithrin-alpha, or FK506 was used alone, and HC-030031 combined with pifithrin-alpha or FK506 further improved the expression of Bcl-2. Tacrolimus 148-153 BCL2 apoptosis regulator Homo sapiens 27-32 34488389-11 2021 However, the expression of Bcl-2 increased when HC-030031, pifithrin-alpha, or FK506 was used alone, and HC-030031 combined with pifithrin-alpha or FK506 further improved the expression of Bcl-2. Tacrolimus 148-153 BCL2 apoptosis regulator Homo sapiens 189-194 34804426-6 2021 Further, ROS (P<0.05), LPO and NO (P<0.001), were significantly reduced with EAF co-treatment compared with TAC individually treated cells. Tacrolimus 108-111 lactoperoxidase Canis lupus familiaris 23-26 34804426-7 2021 TAC induced mitochondrial membrane integrity loss was found to be significantly reduced in co-treated cells, as measured by rhodamine123 (P<0.05) and translocation of cytochrome c (P<0.001) from nucleus to cytoplasm, and caspase 3 release (P<0.001). Tacrolimus 0-3 cytochrome c, somatic Canis lupus familiaris 167-179 34804426-7 2021 TAC induced mitochondrial membrane integrity loss was found to be significantly reduced in co-treated cells, as measured by rhodamine123 (P<0.05) and translocation of cytochrome c (P<0.001) from nucleus to cytoplasm, and caspase 3 release (P<0.001). Tacrolimus 0-3 caspase 3 Canis lupus familiaris 221-230 34439942-8 2021 We observed decreased levels of prolactin (11.9 ng/mL) and cortisol (87.4 mug/mL) in patients under tacrolimus-based therapy. Tacrolimus 100-110 prolactin Homo sapiens 32-41 34129685-1 2021 This study investigated the effect of CYP3A5 phenotype on time in therapeutic range (TTR) of tacrolimus post-transplant in pediatric patients. Tacrolimus 93-103 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 38-44 34367377-14 2021 Serum protein expressions of IL-17 (p = 0.0017), IL-18 (p = 0.0036), IFN-gamma (p = 0.0102), and MMP-9 (p = 0.0194) were significantly decreased in the allograft+tacrolimus+azithromycin group compared to the allograft group. Tacrolimus 162-172 interleukin 17A Mus musculus 29-34 34367377-14 2021 Serum protein expressions of IL-17 (p = 0.0017), IL-18 (p = 0.0036), IFN-gamma (p = 0.0102), and MMP-9 (p = 0.0194) were significantly decreased in the allograft+tacrolimus+azithromycin group compared to the allograft group. Tacrolimus 162-172 interleukin 18 Mus musculus 49-54 34367377-14 2021 Serum protein expressions of IL-17 (p = 0.0017), IL-18 (p = 0.0036), IFN-gamma (p = 0.0102), and MMP-9 (p = 0.0194) were significantly decreased in the allograft+tacrolimus+azithromycin group compared to the allograft group. Tacrolimus 162-172 interferon gamma Mus musculus 69-78 34367377-14 2021 Serum protein expressions of IL-17 (p = 0.0017), IL-18 (p = 0.0036), IFN-gamma (p = 0.0102), and MMP-9 (p = 0.0194) were significantly decreased in the allograft+tacrolimus+azithromycin group compared to the allograft group. Tacrolimus 162-172 matrix metallopeptidase 9 Mus musculus 97-102 34289186-4 2022 The goal of this study was to investigate the extent to which FK506 loaded biomimetic sponges support functional muscle regeneration and fracture healing in a composite trauma model involving VML injury to the tibialis anterior muscle and osteotomy (OST) to the tibia. Tacrolimus 62-67 MCF.2 cell line derived transforming sequence like Homo sapiens 250-253 34289186-5 2022 In this model, implantation of the FK-506 loaded biomimetic sponges limited the extent of inflammation while increasing the total number of myofibers, mean myofiber cross-sectional area, myosin-to-collagen ratio, and peak isometric torque compared to untreated VML+OST muscles on day 28. Tacrolimus 35-41 myosin heavy chain 14 Homo sapiens 187-193 34189904-6 2021 Western blot and immunofluorescence revealed that incorporation of CL in a lipid bilayer ameliorated the docking of CEF-FK506-nilotinib-GSH-CL-liposomes at alpha-synuclein (alpha-syn), indicating a better targeting capability of the liposomes to degenerated neurons. Tacrolimus 120-125 synuclein alpha Homo sapiens 156-171 34377239-14 2021 CONCLUSION: The combination of ultra-low dose rituximab and low dose tacrolimus is more effective in inducing proteinuria response, improving eGFR and serum albumin in non-responsive iMN patients than standard tacrolimus monotherapy. Tacrolimus 69-79 albumin Homo sapiens 151-164 34335631-7 2021 In that group, absolute CD4+CD25+CD127-Foxp3+ Tregs were negatively associated with steroid dose and tacrolimus trough levels (r=-0.377, p = 0.021, n=37; r=-0.43, p=0.033, n=25, respectively), opposite to IL-10+ Bregs, whose frequency apparently was not negatively affected by potent immunosuppression early posttransplant. Tacrolimus 101-111 CD4 molecule Homo sapiens 24-27 34335631-7 2021 In that group, absolute CD4+CD25+CD127-Foxp3+ Tregs were negatively associated with steroid dose and tacrolimus trough levels (r=-0.377, p = 0.021, n=37; r=-0.43, p=0.033, n=25, respectively), opposite to IL-10+ Bregs, whose frequency apparently was not negatively affected by potent immunosuppression early posttransplant. Tacrolimus 101-111 interleukin 2 receptor subunit alpha Homo sapiens 28-32 34335631-7 2021 In that group, absolute CD4+CD25+CD127-Foxp3+ Tregs were negatively associated with steroid dose and tacrolimus trough levels (r=-0.377, p = 0.021, n=37; r=-0.43, p=0.033, n=25, respectively), opposite to IL-10+ Bregs, whose frequency apparently was not negatively affected by potent immunosuppression early posttransplant. Tacrolimus 101-111 interleukin 7 receptor Homo sapiens 33-38 34335631-7 2021 In that group, absolute CD4+CD25+CD127-Foxp3+ Tregs were negatively associated with steroid dose and tacrolimus trough levels (r=-0.377, p = 0.021, n=37; r=-0.43, p=0.033, n=25, respectively), opposite to IL-10+ Bregs, whose frequency apparently was not negatively affected by potent immunosuppression early posttransplant. Tacrolimus 101-111 forkhead box P3 Homo sapiens 39-44 34290503-6 2021 TAC@MD-alpha-TOC also showed significantly enhanced anti-arthritic activity compared with free TAC, as reflected by improved clinical scores and decreased IL-6 and TNF-alpha levels in serum and synovial fluids. Tacrolimus 0-3 interleukin-6 Chlorocebus sabaeus 155-159 34290503-8 2021 Chondrogenic differentiation abilities of TAC@MD-alpha-TOC were proved by increased serum and synovial fluid levels of SOX9 mRNA and protein expression. Tacrolimus 42-45 transcription factor SOX-9 Chlorocebus sabaeus 119-123 34403416-5 2021 IgAN recipients, who were maintained with a regimen containing tacrolimus, experienced recurrence less frequently, compared to those maintained with cyclosporine (p = 0.01). Tacrolimus 63-73 IGAN1 Homo sapiens 0-4 34189904-6 2021 Western blot and immunofluorescence revealed that incorporation of CL in a lipid bilayer ameliorated the docking of CEF-FK506-nilotinib-GSH-CL-liposomes at alpha-synuclein (alpha-syn), indicating a better targeting capability of the liposomes to degenerated neurons. Tacrolimus 120-125 synuclein alpha Homo sapiens 173-182 34306041-9 2021 CYP3A4*22 was shown to significantly influence the pharmacokinetics of several drugs, with currently being most thoroughly investigated tacrolimus, cyclosporine, and statins. Tacrolimus 136-146 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 34403416-9 2021 Tacrolimus, as part of the KTx maintenance therapy, was associated with lower rates of IgAN recurrence in the graft, compared to the rate cyclosporine. Tacrolimus 0-10 IGAN1 Homo sapiens 87-91 34285488-2 2021 Wuzhi capsule (WZC) could improve tacrolimus blood concentration by inhibiting the metabolism of cytochrome P450 3A (CYP3A) and P-glycoprotein (P-gp). Tacrolimus 34-44 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 97-115 34285488-2 2021 Wuzhi capsule (WZC) could improve tacrolimus blood concentration by inhibiting the metabolism of cytochrome P450 3A (CYP3A) and P-glycoprotein (P-gp). Tacrolimus 34-44 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 117-122 34285488-2 2021 Wuzhi capsule (WZC) could improve tacrolimus blood concentration by inhibiting the metabolism of cytochrome P450 3A (CYP3A) and P-glycoprotein (P-gp). Tacrolimus 34-44 ATP binding cassette subfamily B member 1 Homo sapiens 128-142 34285488-2 2021 Wuzhi capsule (WZC) could improve tacrolimus blood concentration by inhibiting the metabolism of cytochrome P450 3A (CYP3A) and P-glycoprotein (P-gp). Tacrolimus 34-44 ATP binding cassette subfamily B member 1 Homo sapiens 144-148 34165588-0 2021 FK506 ameliorates osteoporosis caused by osteoblast apoptosis via suppressing the activated CaN/NFAT pathway during oxidative stress. Tacrolimus 0-5 nuclear factor of activated T-cells 5 Rattus norvegicus 96-100 34236044-7 2021 OX40 immunoPET signals also reflected the subject"s immunosuppression level with tacrolimus in this study. Tacrolimus 81-91 tumor necrosis factor receptor superfamily, member 4 Mus musculus 0-4 34290611-0 2021 CYP3A5 Genotype-Dependent Drug-Drug Interaction Between Tacrolimus and Nifedipine in Chinese Renal Transplant Patients. Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 34290611-2 2021 Nifedipine, frequently prescribed for hypertension, is a competitive CYP3A5 inhibitor which can inhibit tacrolimus metabolism. Tacrolimus 104-114 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 69-75 34290611-3 2021 The objective of this study was to investigate whether CYP3A5 genotype could influence tacrolimus-nifedipine DDI in Chinese renal transplant patients. Tacrolimus 87-97 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 55-61 34290611-9 2021 Results: In this study, a significant DDI between tacrolimus and nifedipine with respect to the CYP3A5*3 polymorphism was confirmed. Tacrolimus 50-60 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 96-102 34290611-12 2021 The co-administrated nifedipine and CYP3A5*3/*3 homozygotes significantly increased tacrolimus concentrations in multivariate line regression analysis. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 34290611-13 2021 Discussion: A CYP3A5 genotype-dependent DDI was found between tacrolimus and nifedipine. Tacrolimus 62-72 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 34165588-9 2021 FK-506 inhibited the oxidative stress and apoptosis by suppressing the activated CaN/NFAT pathway. Tacrolimus 0-6 nuclear factor of activated T-cells 5 Rattus norvegicus 85-89 34165588-12 2021 Briefly, low dose of FK-506 inhibited the oxidative stress by suppressing the activated CaN/NFAT pathway, while high dose of that further inhibited the oxidative stress by suppressing the CaN/NFAT pathway. Tacrolimus 21-27 nuclear factor of activated T-cells 5 Rattus norvegicus 92-96 34165588-12 2021 Briefly, low dose of FK-506 inhibited the oxidative stress by suppressing the activated CaN/NFAT pathway, while high dose of that further inhibited the oxidative stress by suppressing the CaN/NFAT pathway. Tacrolimus 21-27 nuclear factor of activated T-cells 5 Rattus norvegicus 192-196 34165588-13 2021 CONCLUSION: FK-506 ameliorates osteoporosis resulted from osteoblastic apoptosis which caused by suppressing the activated CaN/NFAT pathway during oxidative stress. Tacrolimus 12-18 nuclear factor of activated T-cells 5 Rattus norvegicus 127-131 34241984-2 2022 Persons carrying at least one copy of the wild-type allele, defined as CYP3A5 expressers, exhibit higher clearance and lower trough concentrations of tacrolimus than homozygous nonexpressers, and this difference may affect alloimmunization and allograft function. Tacrolimus 150-160 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 71-77 34241984-5 2022 During the first 3 years after transplant, CYP3A5 expressers tended to have lower tacrolimus trough levels than non-expressers, even though their tacrolimus dosage was as much as 80% higher. Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 43-49 34241984-10 2022 CONCLUSIONS: Early detection of CYP3A5 expressers, enabling genotype-based dose adjustment of tacrolimus immediately after renal transplant, may be a useful strategy for reducing the risk of de novo DSA production and antibody-mediated rejection. Tacrolimus 94-104 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 32-38 34168412-0 2021 Early serum albumin changes in patients with ulcerative colitis treated with tacrolimus will predict clinical outcome. Tacrolimus 77-87 albumin Homo sapiens 12-19 34619854-11 2021 Tacrolimus inhibited the proliferation of CD8+T cells when IL-2 concentration was 20.0,200.0 and 2 000.0 U/ml (P<0.05). Tacrolimus 0-10 CD8a molecule Homo sapiens 42-45 34619854-12 2021 The expression of perforin in CD8+T cells of SAA patients treated with tacrolimus was significantly lower than that in blank control group and IL-2 group ((2.25+-0.76)%, (6.70+-0.82)% vs (9.10+-1.90)%,all P<0.05). Tacrolimus 71-81 CD8a molecule Homo sapiens 30-33 34619854-13 2021 The level of IFN-gamma in CD8+T cells group after applying tacrolimus was significantly lower than that in the blank control group (P<0.05). Tacrolimus 59-69 interferon gamma Homo sapiens 13-22 34619854-13 2021 The level of IFN-gamma in CD8+T cells group after applying tacrolimus was significantly lower than that in the blank control group (P<0.05). Tacrolimus 59-69 CD8a molecule Homo sapiens 26-29 34619854-15 2021 The expression of perforin in CD8+T cells in tacrolimus group was significantly lower than that in SAA group ((18. Tacrolimus 45-55 CD8a molecule Homo sapiens 30-33 34130909-2 2022 Each of the compounds are substrates of CYP3A4/5, the cytochrome P450 enzyme family for which tacrolimus is also a substrate. Tacrolimus 94-104 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-48 34168412-2 2021 AIM: To examine serum albumin (Alb) level as a prognostic factor for the therapeutic effect of tacrolimus in clinical practice. Tacrolimus 95-105 albumin Homo sapiens 22-29 34168412-2 2021 AIM: To examine serum albumin (Alb) level as a prognostic factor for the therapeutic effect of tacrolimus in clinical practice. Tacrolimus 95-105 albumin Homo sapiens 31-34 34168412-8 2021 In the Kaplan-Meier analysis, a week 2/week 0 Alb ratio <= 1 had a significantly higher failure rate than that of > 1; Cox proportional hazard regression analysis also showed that a week 2/week 0 Alb ratio <= 1 was an independent prognostic factor for failure within 3 mo after the start of tacrolimus treatment. Tacrolimus 291-301 albumin Homo sapiens 46-49 34168412-8 2021 In the Kaplan-Meier analysis, a week 2/week 0 Alb ratio <= 1 had a significantly higher failure rate than that of > 1; Cox proportional hazard regression analysis also showed that a week 2/week 0 Alb ratio <= 1 was an independent prognostic factor for failure within 3 mo after the start of tacrolimus treatment. Tacrolimus 291-301 albumin Homo sapiens 196-199 34168412-9 2021 CONCLUSION: A week 2/week 0 Alb ratio <= 1 predicts failure within 3 mo of tacrolimus administration for UC. Tacrolimus 75-85 albumin Homo sapiens 28-31 34150686-1 2021 Background: The calcineurin inhibitor (CNI) tacrolimus (TAC) is a cornerstone agent in immunosuppressive therapy in pediatric liver transplantation (LTX). Tacrolimus 44-54 calcineurin binding protein 1 Homo sapiens 16-37 34108576-7 2021 A multivariable Cox regression model confirmed that high tacrolimus IPV was an independent risk factor for graft failure in the high risk group (HR, 2.90; 95% CI, 1.42-5.95, P = 0.004). Tacrolimus 57-67 cytochrome c oxidase subunit 8A Homo sapiens 16-19 34090756-9 2022 Immunohistochemical caspase-3 evaluation of the axon area revealed a significant difference between group 2 (PRFM alone; 8.67 +- 0.029) and group 3 (PRFM plus topical tacrolimus; 4.42 +- 0.028) (P = 0.001). Tacrolimus 167-177 caspase 3 Rattus norvegicus 20-29 34085919-1 2021 We report a case of neurotoxicity as a side effect of a calcineurin inhibitor (tacrolimus), which is used as an immunosuppressive drug after liver transplant. Tacrolimus 79-89 calcineurin binding protein 1 Homo sapiens 56-77 34150686-1 2021 Background: The calcineurin inhibitor (CNI) tacrolimus (TAC) is a cornerstone agent in immunosuppressive therapy in pediatric liver transplantation (LTX). Tacrolimus 56-59 calcineurin binding protein 1 Homo sapiens 16-37 34095318-0 2021 Tacrolimus Protects Podocytes from Apoptosis via Downregulation of TRPC6 in Diabetic Nephropathy. Tacrolimus 0-10 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 67-72 34077419-4 2021 Under nutrient-replete conditions, SRMS phosphorylates the PHLPP scaffold FK506-binding protein 51 (FKBP51), disrupts the FKBP51-PHLPP complex, and promotes FKBP51 degradation through the ubiquitin-proteasome pathway. Tacrolimus 74-79 src-related kinase lacking C-terminal regulatory tyrosine and N-terminal myristylation sites Homo sapiens 35-39 34077419-4 2021 Under nutrient-replete conditions, SRMS phosphorylates the PHLPP scaffold FK506-binding protein 51 (FKBP51), disrupts the FKBP51-PHLPP complex, and promotes FKBP51 degradation through the ubiquitin-proteasome pathway. Tacrolimus 74-79 PH domain and leucine rich repeat protein phosphatase 1 Homo sapiens 59-64 33729741-10 2021 Patients lacking detectable SARS-CoV-2-specific CD4 response by month 6 were more likely to be under tacrolimus (100.0% versus 66.7%; P-value = 0.087) and to have received tocilizumab for the previous COVID-19 episode (40.0% versus 0.0%; P-value = 0.087). Tacrolimus 101-111 CD4 molecule Homo sapiens 48-51 34072036-4 2021 Glucocorticoid signaling is mediated via glucocorticoid receptors, 11beta-hydroxysteroid dehydrogenases, and the FK506-binding immunophilins, Fkbp4 and Fkbp5. Tacrolimus 113-118 FK506 binding protein 4 Mus musculus 142-147 34072036-4 2021 Glucocorticoid signaling is mediated via glucocorticoid receptors, 11beta-hydroxysteroid dehydrogenases, and the FK506-binding immunophilins, Fkbp4 and Fkbp5. Tacrolimus 113-118 FK506 binding protein 5 Mus musculus 152-157 33149056-1 2021 BACKGROUND: The calcineurin inhibitor tacrolimus is a narrow therapeutic index medication, which requires therapeutic drug monitoring to optimize dose on the basis of systemic exposure. Tacrolimus 38-48 calcineurin binding protein 1 Homo sapiens 16-37 34095318-10 2021 Decreased expression of nephrin and increased expression of TRPC6, cleaved-caspase-3, and bax/bcl-2 ratios were found in podocytes, along with higher apoptotic percentage, while tacrolimus intervention counteracted the effect of HG on podocytes. Tacrolimus 178-188 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 60-65 34095318-11 2021 Our results suggest that tacrolimus protects podocytes during the progression of type 2 diabetic nephropathy, possibly ameliorating podocyte apoptosis by downregulating the expression of TRPC6. Tacrolimus 25-35 transient receptor potential cation channel, subfamily C, member 6 Rattus norvegicus 187-192 34570631-4 2021 In contrast, treatment with tacrolimus or mycophenolate led to reduced expression of functional molecule GITR in CB-Tregs, impaired their viability, proliferation and mitochondrial metabolism. Tacrolimus 28-38 TNF receptor superfamily member 18 Homo sapiens 105-109 34104149-0 2021 Influence of ABCB1 gene polymorphism on concentration to dose ratio and adverse effects of tacrolimus in Pakistani liver transplant recipients. Tacrolimus 91-101 ATP binding cassette subfamily B member 1 Homo sapiens 13-18 34104149-1 2021 Objective: To evaluate the possible association of ABCB1 single nucleotide polymorphism (SNPs) of the ABCB1 gene with tacrolimus dosages, concentration-to-dose ratios (CDR) and adverse effects in Pakistani liver transplant recipients. Tacrolimus 118-128 ATP binding cassette subfamily B member 1 Homo sapiens 51-56 34104149-1 2021 Objective: To evaluate the possible association of ABCB1 single nucleotide polymorphism (SNPs) of the ABCB1 gene with tacrolimus dosages, concentration-to-dose ratios (CDR) and adverse effects in Pakistani liver transplant recipients. Tacrolimus 118-128 ATP binding cassette subfamily B member 1 Homo sapiens 102-107 34104149-8 2021 Results: ABCB1 rs1045642 CC genotype showed lower tacrolimus CDR as compared to CT and TT genotype in the first week of the post-transplantation period (p=0.02). Tacrolimus 50-60 ATP binding cassette subfamily B member 1 Homo sapiens 9-14 34104149-10 2021 Conclusion: Identification of ABCB1 rs1045642 polymorphism may shorten the time to achieve optimum levels of tacrolimus during dose titration. Tacrolimus 109-119 ATP binding cassette subfamily B member 1 Homo sapiens 30-35 34104149-11 2021 ABCB1 polymorphism rs1045642, rs2032582 and rs1128503 may predict adverse effects in liver transplant recipients receiving tacrolimus. Tacrolimus 123-133 ATP binding cassette subfamily B member 1 Homo sapiens 0-5 35238110-10 2022 Different studies have consistently shown that CYP3A5 expressers require a higher tacrolimus dose and take longer time to achieve target blood tacrolimus levels than nonexpressers. Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 47-53 34525930-1 2021 BACKGROUND: Although the pharmacokinetic variability of Tacrolimus (Tac) metabolism is primarily influenced by CYP3A5 genotypes, the potential effect according to CYP3A5 polymorphisms in Tac extended-release (Tac-ER) and immediate-release (Tac-IR) and between these formulations" conversion needs further investigation. Tacrolimus 56-66 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 111-117 34735755-4 2021 The topical formulation of tacrolimus is a well-known FDAapproved anti-T cell agent that was recently identified as a potent activator of ALK1, which is involved in several processes and functions including angiogenesis. Tacrolimus 27-37 secretory leukocyte peptidase inhibitor Homo sapiens 138-142 35060126-0 2022 Reg3g ameliorates Tacrolimus-induced pancreatic beta-cell dysfunction by restoring mitochondrial function in murine models. Tacrolimus 18-28 regenerating islet-derived 3 gamma Mus musculus 0-5 35060126-3 2022 Here, we aim to investigate the potential role of Reg3g in reversing Tac-induced beta-cell dysfunction and NODM in mice. Tacrolimus 69-72 regenerating islet-derived 3 gamma Mus musculus 50-55 35060126-4 2022 EXPERIMENTAL APPROACH: Circulating REG3A (the human homolog of mouse Reg3g) of patients treated with Tac after heart transplantation (HT) was detected. Tacrolimus 101-104 regenerating family member 3 alpha Homo sapiens 35-40 35060126-4 2022 EXPERIMENTAL APPROACH: Circulating REG3A (the human homolog of mouse Reg3g) of patients treated with Tac after heart transplantation (HT) was detected. Tacrolimus 101-104 regenerating islet-derived 3 gamma Mus musculus 69-74 35060126-9 2022 Moreover, Reg3g restored GSIS suppressed by Tac in beta-cells through improving mitochondrial functions, including increased MMP, mitochondria calcium uptake, ATP production, OCR, and contributing to an intact mitochondrial morphology. Tacrolimus 44-47 regenerating family member 3 gamma Homo sapiens 10-15 35060126-10 2022 Mechanically, Reg3g increased accumulation of pSTAT3(Ser727) in mitochondria by activating ERK1/2-STAT3 signaling pathway, leading to restoration of Tac-induced mitochondrial impairment. Tacrolimus 149-152 regenerating family member 3 gamma Homo sapiens 14-19 35060126-10 2022 Mechanically, Reg3g increased accumulation of pSTAT3(Ser727) in mitochondria by activating ERK1/2-STAT3 signaling pathway, leading to restoration of Tac-induced mitochondrial impairment. Tacrolimus 149-152 mitogen-activated protein kinase 3 Mus musculus 91-97 35060126-10 2022 Mechanically, Reg3g increased accumulation of pSTAT3(Ser727) in mitochondria by activating ERK1/2-STAT3 signaling pathway, leading to restoration of Tac-induced mitochondrial impairment. Tacrolimus 149-152 signal transducer and activator of transcription 3 Mus musculus 98-103 35060126-11 2022 Finally, Reg3g overexpression also effectively mitigated Tac-induced NODM in mice. Tacrolimus 57-60 regenerating islet-derived 3 gamma Mus musculus 9-14 35060126-12 2022 CONCLUSION AND IMPLICATIONS: Reg3g can significantly ameliorate Tac-induced beta-cell dysfunction by restoring mitochondrial function in a pSTAT3(Ser727)-dependent manner. Tacrolimus 64-67 regenerating islet-derived 3 gamma Mus musculus 29-34 35060126-13 2022 Our observations identify a novel Reg3g-mediated mechanism that is involved in Tac-induced NODM, and establish the novel role of Reg3g in reversing Tac-induced beta-cell dysfunction. Tacrolimus 79-82 regenerating islet-derived 3 gamma Mus musculus 34-39 35060126-13 2022 Our observations identify a novel Reg3g-mediated mechanism that is involved in Tac-induced NODM, and establish the novel role of Reg3g in reversing Tac-induced beta-cell dysfunction. Tacrolimus 79-82 regenerating islet-derived 3 gamma Mus musculus 129-134 35060126-13 2022 Our observations identify a novel Reg3g-mediated mechanism that is involved in Tac-induced NODM, and establish the novel role of Reg3g in reversing Tac-induced beta-cell dysfunction. Tacrolimus 148-151 regenerating islet-derived 3 gamma Mus musculus 129-134 34145119-11 2021 Aspartate aminotransferase was identified to be the main covariate that influences tacrolimus CL/F. Tacrolimus 83-93 crooked neck pre-mRNA splicing factor 1 Homo sapiens 94-98 34535212-0 2021 Effect of tacrolimus on the expression of Park7 in glomerular podocytes injured by puromycin aminonucleoside. Tacrolimus 10-20 Parkinson disease (autosomal recessive, early onset) 7 Mus musculus 42-47 34535212-14 2021 Compared with the PAN group, the FK506 group had a significant reduction in the mRNA expression level of Park7 at all time points (P<0.01). Tacrolimus 33-38 Parkinson disease (autosomal recessive, early onset) 7 Mus musculus 105-110 34535212-16 2021 Compared with the PAN group, the FK506 group had a significant reduction in the protein expression level of Park7 at all time points (P<0.01). Tacrolimus 33-38 Parkinson disease (autosomal recessive, early onset) 7 Mus musculus 108-113 34535212-18 2021 Compared with the PAN group, the FK506 group had a significant improvement in the distribution of Park7 protein. Tacrolimus 33-38 Parkinson disease (autosomal recessive, early onset) 7 Mus musculus 98-103 34535212-20 2021 FK506 can downregulate the mRNA and protein expression of Park7 in the model of MPC-5 injury, maintain cellular homeostasis, reduce proteinuria, and delay glomerulosclerosis. Tacrolimus 0-5 Parkinson disease (autosomal recessive, early onset) 7 Mus musculus 58-63 35389944-0 2022 Composite CYP3A phenotypes influence tacrolimus dose-adjusted concentration in lung transplant recipients. Tacrolimus 37-47 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 10-15 35389944-1 2022 OBJECTIVES: Interpatient variability in tacrolimus pharmacokinetics is attributed to metabolism by cytochrome P-450 3A4/5 isoenzymes (encoded by CYP3A4 and CYP3A5). Tacrolimus 40-50 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-121 35389944-1 2022 OBJECTIVES: Interpatient variability in tacrolimus pharmacokinetics is attributed to metabolism by cytochrome P-450 3A4/5 isoenzymes (encoded by CYP3A4 and CYP3A5). Tacrolimus 40-50 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 145-151 35389944-1 2022 OBJECTIVES: Interpatient variability in tacrolimus pharmacokinetics is attributed to metabolism by cytochrome P-450 3A4/5 isoenzymes (encoded by CYP3A4 and CYP3A5). Tacrolimus 40-50 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 156-162 35389944-2 2022 Guidelines for adjusting tacrolimus based on CYP3A5 test results are published; however, CYP3A4 variants also contribute to the variability in tacrolimus pharmacokinetics. Tacrolimus 25-35 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 35389944-2 2022 Guidelines for adjusting tacrolimus based on CYP3A5 test results are published; however, CYP3A4 variants also contribute to the variability in tacrolimus pharmacokinetics. Tacrolimus 143-153 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 35389944-2 2022 Guidelines for adjusting tacrolimus based on CYP3A5 test results are published; however, CYP3A4 variants also contribute to the variability in tacrolimus pharmacokinetics. Tacrolimus 143-153 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 89-95 35389944-4 2022 The objective of this study is to investigate the impact of both increased and decreased function CYP3A variants on weight and dose-adjusted tacrolimus concentration (C0/D). Tacrolimus 141-151 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 98-103 35389944-13 2022 CONCLUSION: These data indicate that a composite CYP3A phenotype incorporating both increase and decrease variant information from CYP3A4 in addition to CYP3A5 may significantly influence tacrolimus C0/D during the early postoperative period. Tacrolimus 188-198 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 49-54 35389944-13 2022 CONCLUSION: These data indicate that a composite CYP3A phenotype incorporating both increase and decrease variant information from CYP3A4 in addition to CYP3A5 may significantly influence tacrolimus C0/D during the early postoperative period. Tacrolimus 188-198 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 131-137 35389944-13 2022 CONCLUSION: These data indicate that a composite CYP3A phenotype incorporating both increase and decrease variant information from CYP3A4 in addition to CYP3A5 may significantly influence tacrolimus C0/D during the early postoperative period. Tacrolimus 188-198 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 153-159 35294797-3 2022 In experimental Sprague Dawley rat model of TAC-induced DM and nephrotoxicity, dual inhibition of DPP4 and SGLT2 significantly decreased blood glucose level, HbA1C and increased plasma insulin levels and pancreatic islet size compared with each drug. Tacrolimus 44-47 dipeptidylpeptidase 4 Rattus norvegicus 98-102 35294797-3 2022 In experimental Sprague Dawley rat model of TAC-induced DM and nephrotoxicity, dual inhibition of DPP4 and SGLT2 significantly decreased blood glucose level, HbA1C and increased plasma insulin levels and pancreatic islet size compared with each drug. Tacrolimus 44-47 solute carrier family 5 member 2 Rattus norvegicus 107-112 35294797-3 2022 In experimental Sprague Dawley rat model of TAC-induced DM and nephrotoxicity, dual inhibition of DPP4 and SGLT2 significantly decreased blood glucose level, HbA1C and increased plasma insulin levels and pancreatic islet size compared with each drug. Tacrolimus 44-47 insulin Homo sapiens 185-192 35294797-6 2022 In in vitro study, TAC decreased the cell viability of human kidney-2(HK-2) cells and insulin-secreting beta-cell-derived line(INS-1) cells. Tacrolimus 19-22 insulin Homo sapiens 86-93 35294797-7 2022 SGLT2i protected TAC-induced cell death in HK-2 cells, but not in INS-1 cells. Tacrolimus 17-20 solute carrier family 5 member 2 Rattus norvegicus 0-5 35212883-1 2022 PURPOSE: Recent studies suggest that FK506 binding protein 51 (FKBP51), a negative regulator of glucocorticoid response, encoded by FKBP5, may influence insulin action. Tacrolimus 37-42 FKBP prolyl isomerase 5 Homo sapiens 132-137 35212883-1 2022 PURPOSE: Recent studies suggest that FK506 binding protein 51 (FKBP51), a negative regulator of glucocorticoid response, encoded by FKBP5, may influence insulin action. Tacrolimus 37-42 insulin Homo sapiens 153-160 35396676-0 2022 Interactions with the CYP3A inhibitor voriconazole differ between extended-LCP- and immediate-release tacrolimus formulations. Tacrolimus 102-112 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 22-27 35322385-0 2022 Correction to: The impact of IL-10 and CYP3A5 gene polymorphisms on dose-adjusted trough blood tacrolimus concentrations in early post-renal transplant recipients. Tacrolimus 95-105 interleukin 10 Homo sapiens 29-34 35322385-0 2022 Correction to: The impact of IL-10 and CYP3A5 gene polymorphisms on dose-adjusted trough blood tacrolimus concentrations in early post-renal transplant recipients. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 39-45 35238110-10 2022 Different studies have consistently shown that CYP3A5 expressers require a higher tacrolimus dose and take longer time to achieve target blood tacrolimus levels than nonexpressers. Tacrolimus 143-153 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 47-53 35599203-4 2022 Tacrolimus is metabolized by the cytochrome P450 3A4 enzyme system. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 33-52 35426036-1 2022 The FK506-binding protein 51 (FKBP51) binds progesterone receptor (PR), glucocorticoid receptor (GR), and androgen receptor (AR) to coregulate their transcriptional activity. Tacrolimus 4-9 progesterone receptor Homo sapiens 44-65 35426036-1 2022 The FK506-binding protein 51 (FKBP51) binds progesterone receptor (PR), glucocorticoid receptor (GR), and androgen receptor (AR) to coregulate their transcriptional activity. Tacrolimus 4-9 progesterone receptor Homo sapiens 67-69 35426036-1 2022 The FK506-binding protein 51 (FKBP51) binds progesterone receptor (PR), glucocorticoid receptor (GR), and androgen receptor (AR) to coregulate their transcriptional activity. Tacrolimus 4-9 nuclear receptor subfamily 3 group C member 1 Homo sapiens 72-95 35426036-1 2022 The FK506-binding protein 51 (FKBP51) binds progesterone receptor (PR), glucocorticoid receptor (GR), and androgen receptor (AR) to coregulate their transcriptional activity. Tacrolimus 4-9 nuclear receptor subfamily 3 group C member 1 Homo sapiens 97-99 35426036-1 2022 The FK506-binding protein 51 (FKBP51) binds progesterone receptor (PR), glucocorticoid receptor (GR), and androgen receptor (AR) to coregulate their transcriptional activity. Tacrolimus 4-9 androgen receptor Homo sapiens 106-123 35426036-1 2022 The FK506-binding protein 51 (FKBP51) binds progesterone receptor (PR), glucocorticoid receptor (GR), and androgen receptor (AR) to coregulate their transcriptional activity. Tacrolimus 4-9 androgen receptor Homo sapiens 125-127 35604094-3 2022 FK506 (tacrolimus) is a naturally produced macrocyclic compound that inhibits calcineurin by binding to the immunophilin FKBP12. Tacrolimus 0-5 FKBP prolyl isomerase 1A Homo sapiens 108-127 35604094-3 2022 FK506 (tacrolimus) is a naturally produced macrocyclic compound that inhibits calcineurin by binding to the immunophilin FKBP12. Tacrolimus 7-17 FKBP prolyl isomerase 1A Homo sapiens 108-127 35604094-6 2022 We now report high-resolution protein crystal structures of fungal FKBP12 and a human truncated calcineurin-FKBP12 bound to a natural FK506 analog, FK520 (ascomycin). Tacrolimus 134-139 FKBP prolyl isomerase 1A Homo sapiens 67-73 35604094-6 2022 We now report high-resolution protein crystal structures of fungal FKBP12 and a human truncated calcineurin-FKBP12 bound to a natural FK506 analog, FK520 (ascomycin). Tacrolimus 134-139 FKBP prolyl isomerase 1A Homo sapiens 108-114 35604094-10 2022 Furthermore, molecular dynamic simulations performed with JH-FK-05 binding to fungal and human FKBP12 identified additional residues outside the C-22 and C-21 positions that could be modified to generate novel FK506 analogs with improved antifungal activity. Tacrolimus 210-215 FKBP prolyl isomerase 1A Homo sapiens 95-101 35604094-10 2022 Furthermore, molecular dynamic simulations performed with JH-FK-05 binding to fungal and human FKBP12 identified additional residues outside the C-22 and C-21 positions that could be modified to generate novel FK506 analogs with improved antifungal activity. Tacrolimus 210-215 TBL1X/Y related 1 Homo sapiens 154-158 35136987-10 2022 These results indicated that intermittently blocking TCR signal by tacrolimus can promote anti-tumor immunity and inhibit the tumor growth in melanoma-bearing mice, inhibiting the transcription of several exhaustion-related genes, such as Nr4a1 and Ctla4. Tacrolimus 67-77 nuclear receptor subfamily 4, group A, member 1 Mus musculus 239-244 35136987-10 2022 These results indicated that intermittently blocking TCR signal by tacrolimus can promote anti-tumor immunity and inhibit the tumor growth in melanoma-bearing mice, inhibiting the transcription of several exhaustion-related genes, such as Nr4a1 and Ctla4. Tacrolimus 67-77 cytotoxic T-lymphocyte-associated protein 4 Mus musculus 249-254 35629245-6 2022 SNPs in CYP3A5*3 were significantly associated with greater dose-adjusted cyclosporine and tacrolimus concentrations. Tacrolimus 91-101 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 8-14 35629245-7 2022 SNPs in POR*28 were observed with significantly lower dose-adjusted concentrations, particularly with cyclosporine and tacrolimus. Tacrolimus 119-129 cytochrome p450 oxidoreductase Homo sapiens 8-11 35580897-5 2022 In a treatment approach, we partially reversed vascular pathology by BMPR2-targeted treatment with FK506 in vitro and in vivo. Tacrolimus 99-104 bone morphogenetic protein receptor, type II (serine/threonine kinase) Mus musculus 69-74 35578566-1 2022 BACKGROUND Tacrolimus is a calcineurin inhibitor (CNI) commonly used as an immunosuppressant to prevent the rejection of organ transplants. Tacrolimus 11-21 calcineurin binding protein 1 Homo sapiens 27-48 35578867-0 2022 Impact of POR*28 Variant on Tacrolimus Pharmacokinetics in Kidney Transplant Patients with Different CYP3A5 Genotypes. Tacrolimus 28-38 cytochrome p450 oxidoreductase Homo sapiens 10-13 35578867-3 2022 Although the relationship between CYP3A5 polymorphisms and TAC pharmacokinetics (PK) is well established, the effects of other genetic variants on TAC metabolism, such as POR*28, still remain uncertain. Tacrolimus 147-150 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 35549669-9 2022 Tacrolimus pretreatment prevented increases in the interleukin (IL)-6-to-IL-10 ratio, vascular cell adhesion molecule-1, circulating levels of IL-1beta, IL-6-to-IL-10 ratio and glycocalyx-derived molecules. Tacrolimus 0-10 interleukin 6 Sus scrofa 51-69 35549669-9 2022 Tacrolimus pretreatment prevented increases in the interleukin (IL)-6-to-IL-10 ratio, vascular cell adhesion molecule-1, circulating levels of IL-1beta, IL-6-to-IL-10 ratio and glycocalyx-derived molecules. Tacrolimus 0-10 IL10 Sus scrofa 73-78 35549669-9 2022 Tacrolimus pretreatment prevented increases in the interleukin (IL)-6-to-IL-10 ratio, vascular cell adhesion molecule-1, circulating levels of IL-1beta, IL-6-to-IL-10 ratio and glycocalyx-derived molecules. Tacrolimus 0-10 vascular cell adhesion molecule 1 Sus scrofa 86-119 35549669-9 2022 Tacrolimus pretreatment prevented increases in the interleukin (IL)-6-to-IL-10 ratio, vascular cell adhesion molecule-1, circulating levels of IL-1beta, IL-6-to-IL-10 ratio and glycocalyx-derived molecules. Tacrolimus 0-10 interleukin 1 alpha Sus scrofa 143-151 35549669-9 2022 Tacrolimus pretreatment prevented increases in the interleukin (IL)-6-to-IL-10 ratio, vascular cell adhesion molecule-1, circulating levels of IL-1beta, IL-6-to-IL-10 ratio and glycocalyx-derived molecules. Tacrolimus 0-10 interleukin 6 Sus scrofa 153-157 35549669-9 2022 Tacrolimus pretreatment prevented increases in the interleukin (IL)-6-to-IL-10 ratio, vascular cell adhesion molecule-1, circulating levels of IL-1beta, IL-6-to-IL-10 ratio and glycocalyx-derived molecules. Tacrolimus 0-10 IL10 Sus scrofa 161-166 35549669-10 2022 Tacrolimus partially decreased apoptosis (Bax-to-Bcl2 ratio (p=0.07) and the number of apoptotic cells in the lungs (p<0.05)) but failed to improve LIS. Tacrolimus 0-10 apoptosis regulator BAX Sus scrofa 42-45 35549669-10 2022 Tacrolimus partially decreased apoptosis (Bax-to-Bcl2 ratio (p=0.07) and the number of apoptotic cells in the lungs (p<0.05)) but failed to improve LIS. Tacrolimus 0-10 apoptosis regulator Bcl-2 Sus scrofa 49-53 35570009-0 2022 Renal Cyp3a5-Expressing Genotype Decreases Tacrolimus-to-Dose Ratio in Small Cohort of Renal Transplant Recipients-Preliminary Report. Tacrolimus 43-53 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 6-12 35536394-15 2022 CONCLUSIONS: The CYP3A5 and ABCB1 genotypes impact TAC exposure in PBMCs, which may further alter the effects of TAC. Tacrolimus 51-54 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 17-23 35536394-15 2022 CONCLUSIONS: The CYP3A5 and ABCB1 genotypes impact TAC exposure in PBMCs, which may further alter the effects of TAC. Tacrolimus 51-54 ATP binding cassette subfamily B member 1 Homo sapiens 28-33 35536394-15 2022 CONCLUSIONS: The CYP3A5 and ABCB1 genotypes impact TAC exposure in PBMCs, which may further alter the effects of TAC. Tacrolimus 113-116 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 17-23 35536394-15 2022 CONCLUSIONS: The CYP3A5 and ABCB1 genotypes impact TAC exposure in PBMCs, which may further alter the effects of TAC. Tacrolimus 113-116 ATP binding cassette subfamily B member 1 Homo sapiens 28-33 35537812-0 2022 Tacrolimus-binding protein FKBP8 directs myosin light chain kinase-dependent barrier regulation and is a potential therapeutic target in Crohn"s disease. Tacrolimus 0-10 FKBP prolyl isomerase 8 Homo sapiens 27-32 35570009-1 2022 BACKGROUND: Previous reports have established that patient CYP3A5 allelic variability may be the most important genetic contributor to interindividual variation in tacrolimus exposure in renal transplant recipients. Tacrolimus 164-174 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 59-65 35537812-0 2022 Tacrolimus-binding protein FKBP8 directs myosin light chain kinase-dependent barrier regulation and is a potential therapeutic target in Crohn"s disease. Tacrolimus 0-10 myosin light chain kinase Homo sapiens 41-66 35537812-8 2022 RESULTS: MLCK1 interacted specifically with the tacrolimus-binding FKBP8 PPI domain. Tacrolimus 48-58 myosin light chain kinase Homo sapiens 9-14 35570009-3 2022 The aim of this study was to investigate the role of the renal CYP3A5 genotype in tacrolimus concentration-to-dose ratio within 3 years posttransplant. Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 63-69 35537812-8 2022 RESULTS: MLCK1 interacted specifically with the tacrolimus-binding FKBP8 PPI domain. Tacrolimus 48-58 FKBP prolyl isomerase 8 Homo sapiens 67-72 35570009-4 2022 METHODS: A retrospective cohort study of 90 renal transplant recipients and their donors evaluated the effect of the CYP3A5 single-nucleotide polymorphism (rs776746) on tacrolimus exposure. Tacrolimus 169-179 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 117-123 35537812-10 2022 MLCK1-FKBP8 binding was blocked by tacrolimus, which reversed TNF-induced MLCK1-FKBP8 interactions, MLCK1 recruitment and barrier loss in vitro and in vivo. Tacrolimus 35-45 myosin light chain kinase Homo sapiens 0-5 35570009-6 2022 RESULTS: A significant effect of CYP3A5 expression on tacrolimus exposure was observed in both donors (mean +- SD: 23.8 +- 7.9 vs 32.6 +- 7.4 ng/mL/mg, respectively; P < .001) and recipients (mean +- SD: 27.1 +- 8.0 vs 32.2 +- 7.9 ng/mL/mg, respectively; P = .034) and was lower when CYP3A5 enzyme occurred. Tacrolimus 54-64 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 35537812-10 2022 MLCK1-FKBP8 binding was blocked by tacrolimus, which reversed TNF-induced MLCK1-FKBP8 interactions, MLCK1 recruitment and barrier loss in vitro and in vivo. Tacrolimus 35-45 FKBP prolyl isomerase 8 Homo sapiens 6-11 35537812-10 2022 MLCK1-FKBP8 binding was blocked by tacrolimus, which reversed TNF-induced MLCK1-FKBP8 interactions, MLCK1 recruitment and barrier loss in vitro and in vivo. Tacrolimus 35-45 myosin light chain kinase Homo sapiens 74-79 35570009-6 2022 RESULTS: A significant effect of CYP3A5 expression on tacrolimus exposure was observed in both donors (mean +- SD: 23.8 +- 7.9 vs 32.6 +- 7.4 ng/mL/mg, respectively; P < .001) and recipients (mean +- SD: 27.1 +- 8.0 vs 32.2 +- 7.9 ng/mL/mg, respectively; P = .034) and was lower when CYP3A5 enzyme occurred. Tacrolimus 54-64 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 284-290 35537812-10 2022 MLCK1-FKBP8 binding was blocked by tacrolimus, which reversed TNF-induced MLCK1-FKBP8 interactions, MLCK1 recruitment and barrier loss in vitro and in vivo. Tacrolimus 35-45 FKBP prolyl isomerase 8 Homo sapiens 80-85 35570009-7 2022 Thus, new groups were formed: the group in which at least 1 of the pairs, donor or recipient, had a CYP3A5 expressing allele (n = 23) had lower exposure to tacrolimus compared with nonexpressors (n = 67; mean +- SD: 26.2 +- 7.6 vs 33.2 +- 7.4 ng/mL/mg, respectively; P < .001). Tacrolimus 156-166 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 100-106 35537812-10 2022 MLCK1-FKBP8 binding was blocked by tacrolimus, which reversed TNF-induced MLCK1-FKBP8 interactions, MLCK1 recruitment and barrier loss in vitro and in vivo. Tacrolimus 35-45 myosin light chain kinase Homo sapiens 100-105 35537812-12 2022 CONCLUSION: Binding to FKBP8, which can be blocked by tacrolimus, is required for MLCK1 recruitment to intercellular junctions and downstream events leading to immune-mediated barrier loss. Tacrolimus 54-64 FKBP prolyl isomerase 8 Homo sapiens 23-28 35537812-12 2022 CONCLUSION: Binding to FKBP8, which can be blocked by tacrolimus, is required for MLCK1 recruitment to intercellular junctions and downstream events leading to immune-mediated barrier loss. Tacrolimus 54-64 myosin light chain kinase Homo sapiens 82-87 35570009-9 2022 Nonexpressors receiving kidneys with the CYP3A5*1 allele may benefit from higher tacrolimus doses to hasten achievement of target drug concentrations. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 41-47 35508605-0 2022 The importance of CYP2C19 genotype in tacrolimus dose optimization when concomitant with voriconazole in heart transplant recipients. Tacrolimus 38-48 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 18-25 35508891-9 2022 Although urine beta2-microglobulin significantly decreased following 2 months of treatment with all medications, efficacy was greater with tacrolimus than with cyclophosphamide and MMF (P < 0.001). Tacrolimus 139-149 beta-2-microglobulin Homo sapiens 15-34 35508605-8 2022 Besides, the fold changes of tacrolimus dose were associated with CYP2C19 genotype, which was found to be significantly lower in CYP2C19 extensive metabolizers than that in CYP2C19 intermediate metabolizers or poor metabolizers (4.06+-1.85 vs 5.49+-2.47, p=0.0031). Tacrolimus 29-39 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 66-73 35508605-8 2022 Besides, the fold changes of tacrolimus dose were associated with CYP2C19 genotype, which was found to be significantly lower in CYP2C19 extensive metabolizers than that in CYP2C19 intermediate metabolizers or poor metabolizers (4.06+-1.85 vs 5.49+-2.47, p=0.0031). Tacrolimus 29-39 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 129-136 35508605-8 2022 Besides, the fold changes of tacrolimus dose were associated with CYP2C19 genotype, which was found to be significantly lower in CYP2C19 extensive metabolizers than that in CYP2C19 intermediate metabolizers or poor metabolizers (4.06+-1.85 vs 5.49+-2.47, p=0.0031). Tacrolimus 29-39 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 173-180 35508605-10 2022 Moreover, CYP2C19 genotype and hematocrit acted as independent predicting factors for tacrolimus dose modification after voriconazole co-therapy. Tacrolimus 86-96 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 10-17 35508605-12 2022 CYP2C19 genotype and hematocrit should be considered in tailoring tacrolimus dose. Tacrolimus 66-76 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 0-7 35466485-14 2022 CYP3A5 expressors with high IPV of Tac showed worse outcomes, while the CYP3A5 polymorphism had no impact on IPV of Tac. Tacrolimus 35-38 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 35487881-1 2022 AIMS: To investigate the effects of ABCB1 DNA methylation in donors on individual differences in tacrolimus blood concentrations following liver transplantation. Tacrolimus 97-107 ATP binding cassette subfamily B member 1 Homo sapiens 36-41 35487881-7 2022 RESULTS: Genome-wide methylation sequencing and pyrosequencing analyses showed that the methylation levels of three ABCB1 CpG sites (cg12501229, cg00634941, and cg05496710) were significantly different between groups with different tacrolimus concentration/dose (C0 /D) ratios. Tacrolimus 232-242 ATP binding cassette subfamily B member 1 Homo sapiens 116-121 35487881-8 2022 ABCB1 mRNA expression in donor livers was found to be positively correlated with tacrolimus C0 /D ratio (r = 0.458, P < 0.05). Tacrolimus 81-91 ATP binding cassette subfamily B member 1 Homo sapiens 0-5 35563498-0 2022 Physosmotic Induction of Chondrogenic Maturation Is TGF-beta Dependent and Enhanced by Calcineurin Inhibitor FK506. Tacrolimus 109-114 transforming growth factor alpha Homo sapiens 52-60 35563498-12 2022 While hyperosmolarity alone facilitates TGF-beta superfamily signaling, FK506 further enhances signaling by releasing the FKBP12 break from the type I receptor to improve collagenous marker expression. Tacrolimus 72-77 transforming growth factor alpha Homo sapiens 40-48 35563498-12 2022 While hyperosmolarity alone facilitates TGF-beta superfamily signaling, FK506 further enhances signaling by releasing the FKBP12 break from the type I receptor to improve collagenous marker expression. Tacrolimus 72-77 FKBP prolyl isomerase 1A pseudogene 2 Homo sapiens 122-128 35090967-2 2022 FKBP8 is a member of the FK506-binding proteins family (FKBP) usually found in mitochondria and the endoplasmic reticulum. Tacrolimus 25-30 FKBP prolyl isomerase 8 Homo sapiens 0-5 35487881-9 2022 After treatment with 5-Aza-2-Dc, the methylation levels of the ABCB1 CpG sites in HepG2 cells significantly decreased, and this was confirmed by pyrosequencing; there was also a significant increase in ABCB1 transcription, which induced a decrease in intracellular tacrolimus concentrations. Tacrolimus 265-275 ATP binding cassette subfamily B member 1 Homo sapiens 63-68 35487881-9 2022 After treatment with 5-Aza-2-Dc, the methylation levels of the ABCB1 CpG sites in HepG2 cells significantly decreased, and this was confirmed by pyrosequencing; there was also a significant increase in ABCB1 transcription, which induced a decrease in intracellular tacrolimus concentrations. Tacrolimus 265-275 ATP binding cassette subfamily B member 1 Homo sapiens 202-207 35487881-10 2022 CONCLUSIONS: ABCB1 CpG site methylation affects tacrolimus metabolism in humans by regulating ABCB1 expression. Tacrolimus 48-58 ATP binding cassette subfamily B member 1 Homo sapiens 13-18 35487881-10 2022 CONCLUSIONS: ABCB1 CpG site methylation affects tacrolimus metabolism in humans by regulating ABCB1 expression. Tacrolimus 48-58 ATP binding cassette subfamily B member 1 Homo sapiens 94-99 35487881-11 2022 Therefore, ABCB1 DNA methylation in donor livers might be an important epigenetic factor that affects tacrolimus blood concentrations following liver transplantation. Tacrolimus 102-112 ATP binding cassette subfamily B member 1 Homo sapiens 11-16 35439353-9 2022 Compared to healthy subjects, the associated geometric mean ratios GMR (90% CI) for apixaban Cmax , AUC0-tlast and AUC0-inf were 197%(153, 295), 244%(184, 323) and 224%(170, 295) for transplant recipients on tacrolimus. Tacrolimus 208-218 colony stimulating factor 2 receptor subunit alpha Homo sapiens 67-70 35220592-10 2022 We also revealed the influence of genetic polymorphism of CYP3A5, POD, and a combination of COR on tacrolimus PK. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 58-64 35428708-1 2022 OBJECTIVE: The calcineurin inhibitor tacrolimus has been approved in Japan for the treatment of interstitial pneumonia in patients with polymyositis (PM) and dermatomyositis (DM). Tacrolimus 37-47 calcineurin binding protein 1 Homo sapiens 15-36 35562875-0 2022 Examination of the Impact of CYP3A4/5 on Drug-Drug Interaction between Schizandrol A/Schizandrol B and Tacrolimus (FK-506): A Physiologically Based Pharmacokinetic Modeling Approach. Tacrolimus 103-113 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-37 35562875-0 2022 Examination of the Impact of CYP3A4/5 on Drug-Drug Interaction between Schizandrol A/Schizandrol B and Tacrolimus (FK-506): A Physiologically Based Pharmacokinetic Modeling Approach. Tacrolimus 115-121 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-37 35562875-2 2022 Due to their inhibitory effects on cytochrome P450 (CYP) 3A enzymes, SZA/SZB may display drug-drug interaction (DDI) with tacrolimus. Tacrolimus 122-132 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-59 35562875-6 2022 Moreover, our PBPK model predicted that multiple doses of SZB would increase tacrolimus exposure by 26% and 57% in CYP3A5 expressers and non-expressers, respectively. Tacrolimus 77-87 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 115-121 35456020-6 2022 Recombinant human FKBP11 was able to refold IgG antibody in vitro and inhibited by FK506, strongly supporting a function as antibody peptidyl-prolyl cis-trans isomerase. Tacrolimus 83-88 FKBP prolyl isomerase 11 Homo sapiens 18-24 35066448-0 2022 Exogenous pancreatic kininogenase protects against tacrolimus-induced renal injury by inhibiting PI3K/AKT signaling: The role of bradykinin receptors. Tacrolimus 51-61 thymoma viral proto-oncogene 1 Mus musculus 102-105 35066448-9 2022 TAC-induced oxidative stress was closely associated with endoplasmic reticulum stress and mitochondrial dysfunction, resulting in excessive programmed cell death (apoptosis and autophagy) that was significantly abrogated by concurrent PK interference with PI3K/AKT signaling. Tacrolimus 0-3 thymoma viral proto-oncogene 1 Mus musculus 261-264 35103348-10 2022 The novel CYP3A5*3*6*7 metabolic composite was the significant covariate accounting for 15%-19% of tacrolimus variability in dose (p = 0.002); AUC0-12 h * (p < 0.001), and Cl (p < 0.001). Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 35428510-2 2022 The enzyme encoded by the CYP3A5 gene can regulate the metabolism of tacrolimus, and the polymorphism of the CYP3A5 gene regulates the enzyme function. Tacrolimus 69-79 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 26-32 35428510-2 2022 The enzyme encoded by the CYP3A5 gene can regulate the metabolism of tacrolimus, and the polymorphism of the CYP3A5 gene regulates the enzyme function. Tacrolimus 69-79 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 109-115 35244772-1 2022 FK506-binding protein 51 (encoded by Fkpb51, also known as Fkbp5) has been associated with stress-related mental illness. Tacrolimus 0-5 FK506 binding protein 5 Mus musculus 59-64 35395919-2 2022 In the present study, we established a population pharmacokinetic model to explore the effect of combined use of Wuzhi capsules/echinocandins and the patients" biochemical parameters such as hematocrit on blood concentrations and target doses of tacrolimus in renal transplant patients with different CYP3A5 genotypes. Tacrolimus 246-256 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 301-307 35395919-7 2022 CYP3A5 genotypes and co-administration of Wuzhi capsules, as well as time after renal transplantation and hematocrit, were important factors affecting the clearance of tacrolimus. Tacrolimus 168-178 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 35395919-9 2022 The Monte Carlo simulation indicated the following recommended doses of tacrolimus alone: 0.14 mg kg-1 d-1 for genotype CYP3A5*1*1, 0.12 mg kg-1 d-1 for CYP3A5*1*3, and 0.10 mg kg-1 d-1 for CYP3A5*3*3. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 120-126 35395919-9 2022 The Monte Carlo simulation indicated the following recommended doses of tacrolimus alone: 0.14 mg kg-1 d-1 for genotype CYP3A5*1*1, 0.12 mg kg-1 d-1 for CYP3A5*1*3, and 0.10 mg kg-1 d-1 for CYP3A5*3*3. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 153-159 35395919-9 2022 The Monte Carlo simulation indicated the following recommended doses of tacrolimus alone: 0.14 mg kg-1 d-1 for genotype CYP3A5*1*1, 0.12 mg kg-1 d-1 for CYP3A5*1*3, and 0.10 mg kg-1 d-1 for CYP3A5*3*3. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 190-196 35395919-10 2022 For patients receiving the combination with Wuzhi capsules, the recommended doses of tacrolimus were 0.10 mg kg-1 d-1 for CYP3A5*1*1, 0.08 mg kg-1 d-1 for CYP3A5*1*3, and 0.06 mg kg-1 d-1 for CYP3A5*3*3 genotypes. Tacrolimus 85-95 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 122-128 35395919-10 2022 For patients receiving the combination with Wuzhi capsules, the recommended doses of tacrolimus were 0.10 mg kg-1 d-1 for CYP3A5*1*1, 0.08 mg kg-1 d-1 for CYP3A5*1*3, and 0.06 mg kg-1 d-1 for CYP3A5*3*3 genotypes. Tacrolimus 85-95 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 155-161 35395919-10 2022 For patients receiving the combination with Wuzhi capsules, the recommended doses of tacrolimus were 0.10 mg kg-1 d-1 for CYP3A5*1*1, 0.08 mg kg-1 d-1 for CYP3A5*1*3, and 0.06 mg kg-1 d-1 for CYP3A5*3*3 genotypes. Tacrolimus 85-95 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 192-198 35395919-12 2022 For early kidney transplant recipients receiving tacrolimus treatment, not only body weight, but also CYP3A5 genotypes and drugs used in combination should be considered when determining the target dose of tacrolimus. Tacrolimus 206-216 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 102-108 35202484-2 2022 Genetic variation within the CYP3A5 gene locus impacts the metabolism of several clinically important drugs, including the immunosuppressants tacrolimus, sirolimus, cyclosporine, and the benzodiazepine midazolam. Tacrolimus 142-152 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 29-35 35202484-3 2022 Variable CYP3A5 activity is of clinical importance regarding tacrolimus metabolism. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 9-15 35129440-7 2022 As previous studies reported that tacrolimus treatment lead to a decrease of the measured plasma activity of lipoprotein lipase (LPL), we hypothesized that tacrolimus or related metabolites could also interfere by direct inhibition of LPL involved in TG analytical method used. Tacrolimus 34-44 lipoprotein lipase Homo sapiens 109-127 35129440-7 2022 As previous studies reported that tacrolimus treatment lead to a decrease of the measured plasma activity of lipoprotein lipase (LPL), we hypothesized that tacrolimus or related metabolites could also interfere by direct inhibition of LPL involved in TG analytical method used. Tacrolimus 34-44 lipoprotein lipase Homo sapiens 129-132 35129440-7 2022 As previous studies reported that tacrolimus treatment lead to a decrease of the measured plasma activity of lipoprotein lipase (LPL), we hypothesized that tacrolimus or related metabolites could also interfere by direct inhibition of LPL involved in TG analytical method used. Tacrolimus 34-44 lipoprotein lipase Homo sapiens 235-238 35129440-7 2022 As previous studies reported that tacrolimus treatment lead to a decrease of the measured plasma activity of lipoprotein lipase (LPL), we hypothesized that tacrolimus or related metabolites could also interfere by direct inhibition of LPL involved in TG analytical method used. Tacrolimus 156-166 lipoprotein lipase Homo sapiens 109-127 35129440-7 2022 As previous studies reported that tacrolimus treatment lead to a decrease of the measured plasma activity of lipoprotein lipase (LPL), we hypothesized that tacrolimus or related metabolites could also interfere by direct inhibition of LPL involved in TG analytical method used. Tacrolimus 156-166 lipoprotein lipase Homo sapiens 129-132 35129440-7 2022 As previous studies reported that tacrolimus treatment lead to a decrease of the measured plasma activity of lipoprotein lipase (LPL), we hypothesized that tacrolimus or related metabolites could also interfere by direct inhibition of LPL involved in TG analytical method used. Tacrolimus 156-166 lipoprotein lipase Homo sapiens 235-238 35091292-3 2022 The co-chaperone FK506 binding protein 51 (FKBP51) is a negative regulator of the GC receptor (GR), is highly stress responsive, and its polymorphisms have been repeatedly associated with stress-related disorders and dysfunctions in humans and rodents. Tacrolimus 17-22 FK506 binding protein 5 Mus musculus 43-49 35358111-5 2022 CD3+ T lymphocytes and CD14+ monocytes were isolated from PBMCs, and their intracellular tacrolimus concentrations were measured. Tacrolimus 89-99 CD14 molecule Homo sapiens 23-27 35180322-4 2022 We elucidate the influence of CYP3A5 and MDR1 genetic polymorphisms on the WZC dose by maintaining Ctrough of TAC in Chinese healthy volunteers. Tacrolimus 110-113 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 30-36 35180322-6 2022 The plasma TAC level in volunteers with high CYP3A5 expression was greatly lower than that in those with mutant CYP3A5. Tacrolimus 11-14 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 35180322-6 2022 The plasma TAC level in volunteers with high CYP3A5 expression was greatly lower than that in those with mutant CYP3A5. Tacrolimus 11-14 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 112-118 35143096-8 2022 CONCLUSIONS: Tacrolimus is the standard immunosuppression after LT and can be used in combination with other drugs such as corticosteroids and MMF, and in association with anti-IL2 receptor antibody (IL2Ra) induction. Tacrolimus 13-23 interleukin 2 receptor subunit beta Homo sapiens 177-189 35143096-8 2022 CONCLUSIONS: Tacrolimus is the standard immunosuppression after LT and can be used in combination with other drugs such as corticosteroids and MMF, and in association with anti-IL2 receptor antibody (IL2Ra) induction. Tacrolimus 13-23 interleukin 2 receptor subunit alpha Homo sapiens 200-205 35143096-10 2022 Low dose or delayed introduction of tacrolimus in association with corticosteroids and MMF and/or anti-IL2Ra induction can be used to reduce acute kidney injury. Tacrolimus 36-46 interleukin 2 receptor subunit alpha Homo sapiens 103-108 35132839-4 2022 METHODS: Trough(C0) and 1.5-hour blood levels (C1.5) of tacrolimus were measured in 95 kidney transplantation recipients. Tacrolimus 56-66 placenta associated 8 Homo sapiens 47-51 35132839-15 2022 Single nucleotide variants s in ABCB1 gene might influence the flat pattern peaks of tacrolimus absorption. Tacrolimus 85-95 ATP binding cassette subfamily B member 1 Homo sapiens 32-37 35103348-11 2022 CONCLUSIONS: Tacrolimus pharmacokinetics and adverse effects were different among stable kidney transplant recipient groups based upon race and sex with interpatient variability associated with the CYP3A5*3*6*7 metabolic composite. Tacrolimus 13-23 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 198-204 35213993-0 2022 Association of P450 Oxidoreductase Gene Polymorphism with Tacrolimus Pharmacokinetics in Renal Transplant Recipients: A Systematic Review and Meta-Analysis. Tacrolimus 58-68 cytochrome p450 oxidoreductase Homo sapiens 15-34 35126949-8 2022 The CNI dosage of tacrolimus group and cyclosporine A group was compared before and after MMF. Tacrolimus 18-28 5'-nucleotidase, cytosolic IA Homo sapiens 4-7 35126949-10 2022 Experimental results showed that there were 16 cases (14.55%) of CNI-related renal damage in lung transplant recipients and different immunosuppressants, including 10 cases (11.36%) in males, 6 cases (27.27%) in females, 11 cases (12.09%) in tacrolimus group, and 5 cases (26.32%) in cyclosporine A group. Tacrolimus 242-252 5'-nucleotidase, cytosolic IA Homo sapiens 65-68 35213993-1 2022 There are conflicting results regarding the effect of the P450 oxidoreductase (POR) *28 genotype on the tacrolimus (TAC) pharmacokinetics (PKs) during the early post-transplantation period in adult renal transplant recipients. Tacrolimus 104-114 cytochrome p450 oxidoreductase Homo sapiens 58-77 35213993-1 2022 There are conflicting results regarding the effect of the P450 oxidoreductase (POR) *28 genotype on the tacrolimus (TAC) pharmacokinetics (PKs) during the early post-transplantation period in adult renal transplant recipients. Tacrolimus 104-114 cytochrome p450 oxidoreductase Homo sapiens 79-82 35213993-1 2022 There are conflicting results regarding the effect of the P450 oxidoreductase (POR) *28 genotype on the tacrolimus (TAC) pharmacokinetics (PKs) during the early post-transplantation period in adult renal transplant recipients. Tacrolimus 116-119 cytochrome p450 oxidoreductase Homo sapiens 58-77 35213993-1 2022 There are conflicting results regarding the effect of the P450 oxidoreductase (POR) *28 genotype on the tacrolimus (TAC) pharmacokinetics (PKs) during the early post-transplantation period in adult renal transplant recipients. Tacrolimus 116-119 cytochrome p450 oxidoreductase Homo sapiens 79-82 35213993-2 2022 Thus, we characterized the impact of POR*28 on TAC PKs. Tacrolimus 47-50 cytochrome p450 oxidoreductase Homo sapiens 37-40 35213993-3 2022 We conducted a systematic review on the association between POR*28 and PKs of TAC in adult renal transplant recipients. Tacrolimus 78-81 cytochrome p450 oxidoreductase Homo sapiens 60-63 34651654-4 2022 Treatment with prednisolone (PSL), mycophenolate mofetil (MMF), and tacrolimus (TAC) resulted in improved SLE activity and glucose intolerance with the reduction of anti-insulin receptor autoantibodies. Tacrolimus 68-78 insulin receptor Homo sapiens 170-186 35126303-6 2021 The proportion of CD19+CD24hiCD38hi/CD19+ and CD19+CD5+CD1dhi/CD19+ lymphocyte subsets increased significantly after TAC therapy (p = 0.010 and p < 0.001). Tacrolimus 117-120 CD19 molecule Homo sapiens 18-22 35126303-6 2021 The proportion of CD19+CD24hiCD38hi/CD19+ and CD19+CD5+CD1dhi/CD19+ lymphocyte subsets increased significantly after TAC therapy (p = 0.010 and p < 0.001). Tacrolimus 117-120 CD24 molecule Homo sapiens 23-27 35126303-6 2021 The proportion of CD19+CD24hiCD38hi/CD19+ and CD19+CD5+CD1dhi/CD19+ lymphocyte subsets increased significantly after TAC therapy (p = 0.010 and p < 0.001). Tacrolimus 117-120 CD19 molecule Homo sapiens 36-40 35126303-6 2021 The proportion of CD19+CD24hiCD38hi/CD19+ and CD19+CD5+CD1dhi/CD19+ lymphocyte subsets increased significantly after TAC therapy (p = 0.010 and p < 0.001). Tacrolimus 117-120 CD19 molecule Homo sapiens 46-50 35126303-6 2021 The proportion of CD19+CD24hiCD38hi/CD19+ and CD19+CD5+CD1dhi/CD19+ lymphocyte subsets increased significantly after TAC therapy (p = 0.010 and p < 0.001). Tacrolimus 117-120 CD5 molecule Homo sapiens 51-54 35126303-6 2021 The proportion of CD19+CD24hiCD38hi/CD19+ and CD19+CD5+CD1dhi/CD19+ lymphocyte subsets increased significantly after TAC therapy (p = 0.010 and p < 0.001). Tacrolimus 117-120 CD19 molecule Homo sapiens 62-66 35126303-7 2021 The proportion of CD19+BAFFR+, CD19+IFN-gamma+, and CD19+IL-10+ subsets decreased significantly after TAC therapy (p = 0.015, 0.018, and 0.042, respectively). Tacrolimus 102-105 CD19 molecule Homo sapiens 18-22 35126303-7 2021 The proportion of CD19+BAFFR+, CD19+IFN-gamma+, and CD19+IL-10+ subsets decreased significantly after TAC therapy (p = 0.015, 0.018, and 0.042, respectively). Tacrolimus 102-105 TNF receptor superfamily member 13C Homo sapiens 23-28 35126303-7 2021 The proportion of CD19+BAFFR+, CD19+IFN-gamma+, and CD19+IL-10+ subsets decreased significantly after TAC therapy (p = 0.015, 0.018, and 0.042, respectively). Tacrolimus 102-105 CD19 molecule Homo sapiens 31-35 35126303-7 2021 The proportion of CD19+BAFFR+, CD19+IFN-gamma+, and CD19+IL-10+ subsets decreased significantly after TAC therapy (p = 0.015, 0.018, and 0.042, respectively). Tacrolimus 102-105 interferon gamma Homo sapiens 36-45 35126303-7 2021 The proportion of CD19+BAFFR+, CD19+IFN-gamma+, and CD19+IL-10+ subsets decreased significantly after TAC therapy (p = 0.015, 0.018, and 0.042, respectively). Tacrolimus 102-105 CD19 molecule Homo sapiens 52-56 35126303-7 2021 The proportion of CD19+BAFFR+, CD19+IFN-gamma+, and CD19+IL-10+ subsets decreased significantly after TAC therapy (p = 0.015, 0.018, and 0.042, respectively). Tacrolimus 102-105 interleukin 10 Homo sapiens 57-62 35126303-9 2021 Conclusion: Possibly through increasing regulatory B and suppressing BAFFR+ B and interferon (IFN)-gamma+ B subsets, TAC could decrease relapse. Tacrolimus 117-120 interferon gamma Homo sapiens 82-104 34516654-10 2022 Administration of prednisolone and tacrolimus quickly alleviated the symptoms and the CK level returned to normal. Tacrolimus 35-45 cytidine/uridine monophosphate kinase 1 Homo sapiens 86-88 34993577-0 2022 Tacrolimus induces remission in refractory and relapsing lupus nephritis by decreasing P-glycoprotein expression and function on peripheral blood lymphocytes. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 87-101 34993577-2 2022 Tacrolimus is a P-gp inhibitor and hence, may overcome this resistance. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 16-20 34993577-5 2022 Expression and function of P-gp on PBL was measured by flow cytometry (as relative fluorescence index, RFI and Rhodamine dye efflux assay) before and 3 months after tacrolimus therapy. Tacrolimus 165-175 ATP binding cassette subfamily B member 1 Homo sapiens 27-31 34993577-10 2022 Tacrolimus achieves renal response in refractory/relapsing proliferative LN patients which may be partly related to overcoming P-glycoprotein mediated treatment unresponsiveness. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 127-141 34651654-4 2022 Treatment with prednisolone (PSL), mycophenolate mofetil (MMF), and tacrolimus (TAC) resulted in improved SLE activity and glucose intolerance with the reduction of anti-insulin receptor autoantibodies. Tacrolimus 80-83 insulin receptor Homo sapiens 170-186 35413717-10 2022 Tacrolimus treatment ameliorated mesangial alterations by suppressing the expressions of Th17-related cytokines such as IL-17 and IL-6. Tacrolimus 0-10 interleukin 17A Rattus norvegicus 120-125 35413717-10 2022 Tacrolimus treatment ameliorated mesangial alterations by suppressing the expressions of Th17-related cytokines such as IL-17 and IL-6. Tacrolimus 0-10 interleukin 6 Rattus norvegicus 130-134 2472451-1 1989 FK506, a neutral macrolide with immunosuppressive properties, was shown to selectively and rapidly inhibit the accumulation of IL-2 mRNA, as well as the mRNAs of other early (E) phase T cell activation genes such as IL-3, IL-4, GM-CSF, TNF alpha, IFN-gamma, and c-myc in activated human peripheral blood T cells. Tacrolimus 0-5 interleukin 2 Homo sapiens 127-131 2483716-1 1989 Nanomolar concentrations of the novel immunosuppressive drug FK-506 inhibit the proliferation of human T lymphocytes in vitro induced by mitogenic lectins or by monoclonal antibodies directed against the CD3 or CD2 surface antigens. Tacrolimus 61-67 CD2 molecule Homo sapiens 211-214 35110472-0 2022 Pharmacokinetics of tacrolimus following an overdose in a patient with extreme obesity and genotype CYP3A5*3/*3: a case report. Tacrolimus 20-30 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 100-106 2472451-1 1989 FK506, a neutral macrolide with immunosuppressive properties, was shown to selectively and rapidly inhibit the accumulation of IL-2 mRNA, as well as the mRNAs of other early (E) phase T cell activation genes such as IL-3, IL-4, GM-CSF, TNF alpha, IFN-gamma, and c-myc in activated human peripheral blood T cells. Tacrolimus 0-5 interleukin 3 Homo sapiens 216-220 2472451-1 1989 FK506, a neutral macrolide with immunosuppressive properties, was shown to selectively and rapidly inhibit the accumulation of IL-2 mRNA, as well as the mRNAs of other early (E) phase T cell activation genes such as IL-3, IL-4, GM-CSF, TNF alpha, IFN-gamma, and c-myc in activated human peripheral blood T cells. Tacrolimus 0-5 interleukin 4 Homo sapiens 222-226 2472451-1 1989 FK506, a neutral macrolide with immunosuppressive properties, was shown to selectively and rapidly inhibit the accumulation of IL-2 mRNA, as well as the mRNAs of other early (E) phase T cell activation genes such as IL-3, IL-4, GM-CSF, TNF alpha, IFN-gamma, and c-myc in activated human peripheral blood T cells. Tacrolimus 0-5 colony stimulating factor 2 Homo sapiens 228-234 2472451-1 1989 FK506, a neutral macrolide with immunosuppressive properties, was shown to selectively and rapidly inhibit the accumulation of IL-2 mRNA, as well as the mRNAs of other early (E) phase T cell activation genes such as IL-3, IL-4, GM-CSF, TNF alpha, IFN-gamma, and c-myc in activated human peripheral blood T cells. Tacrolimus 0-5 tumor necrosis factor Homo sapiens 236-245 2472451-1 1989 FK506, a neutral macrolide with immunosuppressive properties, was shown to selectively and rapidly inhibit the accumulation of IL-2 mRNA, as well as the mRNAs of other early (E) phase T cell activation genes such as IL-3, IL-4, GM-CSF, TNF alpha, IFN-gamma, and c-myc in activated human peripheral blood T cells. Tacrolimus 0-5 interferon gamma Homo sapiens 247-256 2472451-1 1989 FK506, a neutral macrolide with immunosuppressive properties, was shown to selectively and rapidly inhibit the accumulation of IL-2 mRNA, as well as the mRNAs of other early (E) phase T cell activation genes such as IL-3, IL-4, GM-CSF, TNF alpha, IFN-gamma, and c-myc in activated human peripheral blood T cells. Tacrolimus 0-5 MYC proto-oncogene, bHLH transcription factor Homo sapiens 262-267 2472451-2 1989 The activity of FK506, when compared to Cyclosporin A, another immunosuppressant, was 10 to 100x more potent in its ability to inhibit IL-2 mRNA synthesis. Tacrolimus 16-21 interleukin 2 Homo sapiens 135-139 2472451-3 1989 FK506 inhibited IL-2 mRNA accumulation in Con A, Con A plus PMA, Ionomycin plus PMA, anti-CD3, and anti-CD3 plus PMA activated T cells. Tacrolimus 0-5 interleukin 2 Homo sapiens 16-20 2472451-6 1989 The effect of FK506 on inducible genes in non-T and non-lymphoid human cells was studied in LPS-induced monocytes and PMA or IL-1 activated synovial fibroblasts. Tacrolimus 14-19 interleukin 1 alpha Homo sapiens 125-129 2472451-8 1989 Nuclear run-off transcription studies indicate that FK506 inhibits transcription of the IL-2 gene. Tacrolimus 52-57 interleukin 2 Homo sapiens 88-92 2471685-0 1989 The immunosuppressant FK-506, like cyclosporins and didemnin B, inhibits calmodulin-dependent phosphorylation of the elongation factor 2 in vitro and biological effects of the phorbol ester TPA on mouse skin in vivo. Tacrolimus 22-28 eukaryotic translation elongation factor 2 Mus musculus 117-136 2471685-1 1989 Similar to previous observations with cyclosporins and didemnin B, the novel immunosuppressant FK-506 inhibits the Ca2+/calmodulin-dependent phosphorylation of the eukaryotic elongation factor 2 of protein synthesis in vitro and biological effects of the phorbol ester TPA on mouse skin in vivo. Tacrolimus 95-101 eukaryotic translation elongation factor 2 Mus musculus 175-194 2465593-4 1989 FK506 inhibited in a dose-dependent manner both interleukin 2 and gamma-interferon secretion of PBMC stimulated with PHA. Tacrolimus 0-5 interleukin 2 Homo sapiens 48-61 2465593-5 1989 Complete inhibition was obtained at the concentration of 0.25 nM of FK506 for IL-2 and 1 nM of FK506 for gamma-IFN production. Tacrolimus 68-73 interleukin 2 Homo sapiens 78-82 2442255-8 1987 On the other hand, it was found that FK506 inhibited both IL-2 secretion and IL-2 receptor expression of BC.21 after stimulation with the specific antigen. Tacrolimus 37-42 interleukin 2 Mus musculus 58-62 3291267-3 1988 The introduction of new drugs such as FK 506, some of which are clearly synergistic with CsA, could ameliorate past problems with drug toxicity. Tacrolimus 38-44 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 89-92 2463974-6 1988 Both humoral and cellular immune response to MBP were completely suppressed in rats treated with FK506. Tacrolimus 97-102 myelin basic protein Rattus norvegicus 45-48 2458329-11 1988 Therefore, as with cyclosporine, as demonstrated in our previous study, FK506 has the capacity to induce immunological unresponsiveness specific to the S-antigen. Tacrolimus 72-77 S-antigen visual arrestin Rattus norvegicus 152-161 2442255-8 1987 On the other hand, it was found that FK506 inhibited both IL-2 secretion and IL-2 receptor expression of BC.21 after stimulation with the specific antigen. Tacrolimus 37-42 interleukin 2 Mus musculus 77-81 33838305-1 2021 Tacrolimus (FK506), a calcineurin inhibitor, is an effective immunosuppressive agent mainly used to lower the risk of organ rejection after allogeneic organ transplant. Tacrolimus 0-10 calcineurin binding protein 1 Rattus norvegicus 22-43 33866281-0 2021 CYP3A5*3 polymorphism and age affect tacrolimus blood trough concentration in myasthenia gravis patients. Tacrolimus 37-47 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 33866281-8 2021 Thus, CYP3A5*3 polymorphism and age should be considered in optimizing the initial dose of tacrolimus for MG treatment. Tacrolimus 91-101 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 6-12 33838305-1 2021 Tacrolimus (FK506), a calcineurin inhibitor, is an effective immunosuppressive agent mainly used to lower the risk of organ rejection after allogeneic organ transplant. Tacrolimus 12-17 calcineurin binding protein 1 Rattus norvegicus 22-43 33830489-3 2021 We report three cases of cobicistat-tacrolimus co-administration, two of which also include the co-administration of mTOR inhibitors, in HIV-positive patients undergoing SOT (2 kidney and 1 liver recipient). Tacrolimus 36-46 mechanistic target of rapamycin kinase Homo sapiens 117-121 33386894-0 2021 The influence of recipient SLCO1B1 rs2291075 polymorphism on tacrolimus dose-corrected trough concentration in the early period after liver transplantation. Tacrolimus 61-71 solute carrier organic anion transporter family member 1B1 Homo sapiens 27-34 33386894-1 2021 PURPOSE: To explore the relationship between rs2291075 polymorphism in SLCO1B1 gene, which encodes an influx transmembrane protein transporter, and tacrolimus dose-corrected trough concentration (C/D, ng ml-1 mg-1 kg-1) in the early period after liver transplantation. Tacrolimus 148-158 solute carrier organic anion transporter family member 1B1 Homo sapiens 71-78 33386894-5 2021 RESULTS: The combined analysis of donor and recipient CYP3A5 rs776746 could distinguish the metabolic phenotype of tacrolimus into three groups: fast elimination (FE), intermediate elimination (IE), and slow elimination (SE), which was entitled the FIS classification system. Tacrolimus 115-125 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 54-60 33386894-5 2021 RESULTS: The combined analysis of donor and recipient CYP3A5 rs776746 could distinguish the metabolic phenotype of tacrolimus into three groups: fast elimination (FE), intermediate elimination (IE), and slow elimination (SE), which was entitled the FIS classification system. Tacrolimus 115-125 long intergenic non-protein coding RNA 1554 Homo sapiens 249-252 33386894-6 2021 Tacrolimus C/D ratios of recipient SLCO1B1 rs2291075 CT and TT carriers were very close and were significantly lower than those of recipient SLCO1B1 rs2291075 CC genotype carriers in convalescence phase (p = 0.0195) and in stationary phase (p = 0.0152). Tacrolimus 0-10 solute carrier organic anion transporter family member 1B1 Homo sapiens 35-42 33512727-0 2021 Tacrolimus (FK506) treatment protects allergen, IL-5, and IL-13-induced mucosal eosinophilia. Tacrolimus 0-10 interleukin 5 Homo sapiens 48-52 33512727-0 2021 Tacrolimus (FK506) treatment protects allergen, IL-5, and IL-13-induced mucosal eosinophilia. Tacrolimus 0-10 interleukin 13 Homo sapiens 58-63 33386894-8 2021 A model consisting of tacrolimus daily dose, total bilirubin, FIS classification, and recipient SLCO1B1 rs2291075 could predict tacrolimus C/D ratios in the convalescence phase by multivariate analysis. Tacrolimus 128-138 long intergenic non-protein coding RNA 1554 Homo sapiens 62-65 33386894-8 2021 A model consisting of tacrolimus daily dose, total bilirubin, FIS classification, and recipient SLCO1B1 rs2291075 could predict tacrolimus C/D ratios in the convalescence phase by multivariate analysis. Tacrolimus 128-138 solute carrier organic anion transporter family member 1B1 Homo sapiens 96-103 33386894-10 2021 Recipient SLCO1B1 rs2291075 genotype had significant effect on tacrolimus C/D ratios in convalescence phase (p = 0.0300) and stationary phase (p = 0.0400) in subgroup, which excluded the interference come from donor and recipient CYP3A5 rs776746. Tacrolimus 63-73 solute carrier organic anion transporter family member 1B1 Homo sapiens 10-17 33386894-11 2021 CONCLUSION: SLCO1B1 rs2291075 could be a novel genetic locus associated with tacrolimus metabolism. Tacrolimus 77-87 solute carrier organic anion transporter family member 1B1 Homo sapiens 12-19 33386894-12 2021 The combined analysis of donor and recipient CYP3A5 rs776746, recipient SLCO1B1 rs2291075 genotypes, could be helpful to guide the personalized administration of tacrolimus in early period after liver transplantation. Tacrolimus 162-172 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 33386894-12 2021 The combined analysis of donor and recipient CYP3A5 rs776746, recipient SLCO1B1 rs2291075 genotypes, could be helpful to guide the personalized administration of tacrolimus in early period after liver transplantation. Tacrolimus 162-172 solute carrier organic anion transporter family member 1B1 Homo sapiens 72-79 33398393-0 2021 CYP3A5 and PPARA genetic variants are associated with low trough concentration to dose ratio of tacrolimus in kidney transplant recipients. Tacrolimus 96-106 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 33398393-0 2021 CYP3A5 and PPARA genetic variants are associated with low trough concentration to dose ratio of tacrolimus in kidney transplant recipients. Tacrolimus 96-106 peroxisome proliferator activated receptor alpha Homo sapiens 11-16 33398393-2 2021 This study is aimed at assessing the impact of allelic variants of CYP3A5 and PPARA genes on the pharmacokinetics (PK) of TAC in Brazilian kidney transplant recipients in the first-year post-transplant. Tacrolimus 122-125 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-73 33398393-2 2021 This study is aimed at assessing the impact of allelic variants of CYP3A5 and PPARA genes on the pharmacokinetics (PK) of TAC in Brazilian kidney transplant recipients in the first-year post-transplant. Tacrolimus 122-125 peroxisome proliferator activated receptor alpha Homo sapiens 78-83 33512727-6 2021 Mechanistically, we show that tacrolimus in vitro treatment inhibited bone marrow derived eosinophil proliferation and viability by promoting eosinophils apoptosis that may be associated with down regulation of RCAN1. Tacrolimus 30-40 regulator of calcineurin 1 Homo sapiens 211-216 33512727-7 2021 Taken together, we provide in vivo and in vitro evidence that tacrolimus ameliorates eosinophil levels and associated pathogenesis in allergen-, IL-5- and IL-13-induced EoE, EG, and asthma pathogenesis. Tacrolimus 62-72 interleukin 5 Mus musculus 145-149 33512727-7 2021 Taken together, we provide in vivo and in vitro evidence that tacrolimus ameliorates eosinophil levels and associated pathogenesis in allergen-, IL-5- and IL-13-induced EoE, EG, and asthma pathogenesis. Tacrolimus 62-72 interleukin 13 Mus musculus 155-160 33512727-0 2021 Tacrolimus (FK506) treatment protects allergen, IL-5, and IL-13-induced mucosal eosinophilia. Tacrolimus 12-17 interleukin 5 Homo sapiens 48-52 33512727-0 2021 Tacrolimus (FK506) treatment protects allergen, IL-5, and IL-13-induced mucosal eosinophilia. Tacrolimus 12-17 interleukin 13 Homo sapiens 58-63 33512727-2 2021 Herein, we present evidence that pre-and post- intraperitoneal administration of tacrolimus (FK506) is very effective in reducing CCR3/SigleF+ eosinophils in Aspergillus-challenged asthma and EoE, CD2-IL-5 induced global eosinophilia and DOX regulated IL-13 induced asthma. Tacrolimus 81-91 C-C motif chemokine receptor 3 Homo sapiens 130-134 33512727-2 2021 Herein, we present evidence that pre-and post- intraperitoneal administration of tacrolimus (FK506) is very effective in reducing CCR3/SigleF+ eosinophils in Aspergillus-challenged asthma and EoE, CD2-IL-5 induced global eosinophilia and DOX regulated IL-13 induced asthma. Tacrolimus 81-91 CD2 molecule Homo sapiens 197-200 33512727-2 2021 Herein, we present evidence that pre-and post- intraperitoneal administration of tacrolimus (FK506) is very effective in reducing CCR3/SigleF+ eosinophils in Aspergillus-challenged asthma and EoE, CD2-IL-5 induced global eosinophilia and DOX regulated IL-13 induced asthma. Tacrolimus 81-91 interleukin 5 Homo sapiens 201-205 33512727-2 2021 Herein, we present evidence that pre-and post- intraperitoneal administration of tacrolimus (FK506) is very effective in reducing CCR3/SigleF+ eosinophils in Aspergillus-challenged asthma and EoE, CD2-IL-5 induced global eosinophilia and DOX regulated IL-13 induced asthma. Tacrolimus 81-91 interleukin 13 Homo sapiens 252-257 33512727-2 2021 Herein, we present evidence that pre-and post- intraperitoneal administration of tacrolimus (FK506) is very effective in reducing CCR3/SigleF+ eosinophils in Aspergillus-challenged asthma and EoE, CD2-IL-5 induced global eosinophilia and DOX regulated IL-13 induced asthma. Tacrolimus 93-98 C-C motif chemokine receptor 3 Homo sapiens 130-134 33512727-2 2021 Herein, we present evidence that pre-and post- intraperitoneal administration of tacrolimus (FK506) is very effective in reducing CCR3/SigleF+ eosinophils in Aspergillus-challenged asthma and EoE, CD2-IL-5 induced global eosinophilia and DOX regulated IL-13 induced asthma. Tacrolimus 93-98 CD2 molecule Homo sapiens 197-200 33512727-2 2021 Herein, we present evidence that pre-and post- intraperitoneal administration of tacrolimus (FK506) is very effective in reducing CCR3/SigleF+ eosinophils in Aspergillus-challenged asthma and EoE, CD2-IL-5 induced global eosinophilia and DOX regulated IL-13 induced asthma. Tacrolimus 93-98 interleukin 5 Homo sapiens 201-205 33512727-2 2021 Herein, we present evidence that pre-and post- intraperitoneal administration of tacrolimus (FK506) is very effective in reducing CCR3/SigleF+ eosinophils in Aspergillus-challenged asthma and EoE, CD2-IL-5 induced global eosinophilia and DOX regulated IL-13 induced asthma. Tacrolimus 93-98 interleukin 13 Homo sapiens 252-257 33512727-4 2021 Additionally, we also performed ELISA and Western blot analyses to show that tacrolimus treatment also reduces the levels of eosinophil-specific cytokines IL-4, IL-5, IL-13, TGF-beta, eosinophil-specific chemokines Eotaxin-1, Eotaxin-2, and progenitors target RCAN1 mRNA levels. Tacrolimus 77-87 interleukin 4 Homo sapiens 155-159 33512727-4 2021 Additionally, we also performed ELISA and Western blot analyses to show that tacrolimus treatment also reduces the levels of eosinophil-specific cytokines IL-4, IL-5, IL-13, TGF-beta, eosinophil-specific chemokines Eotaxin-1, Eotaxin-2, and progenitors target RCAN1 mRNA levels. Tacrolimus 77-87 interleukin 5 Homo sapiens 161-165 33512727-4 2021 Additionally, we also performed ELISA and Western blot analyses to show that tacrolimus treatment also reduces the levels of eosinophil-specific cytokines IL-4, IL-5, IL-13, TGF-beta, eosinophil-specific chemokines Eotaxin-1, Eotaxin-2, and progenitors target RCAN1 mRNA levels. Tacrolimus 77-87 interleukin 13 Homo sapiens 167-172 33512727-4 2021 Additionally, we also performed ELISA and Western blot analyses to show that tacrolimus treatment also reduces the levels of eosinophil-specific cytokines IL-4, IL-5, IL-13, TGF-beta, eosinophil-specific chemokines Eotaxin-1, Eotaxin-2, and progenitors target RCAN1 mRNA levels. Tacrolimus 77-87 transforming growth factor alpha Homo sapiens 174-182 34046945-7 2021 After mTOR initiation, 50% of patients were reduced or weaned off tacrolimus and 13.7% off prednisone. Tacrolimus 66-76 mechanistic target of rapamycin kinase Homo sapiens 6-10 33512727-4 2021 Additionally, we also performed ELISA and Western blot analyses to show that tacrolimus treatment also reduces the levels of eosinophil-specific cytokines IL-4, IL-5, IL-13, TGF-beta, eosinophil-specific chemokines Eotaxin-1, Eotaxin-2, and progenitors target RCAN1 mRNA levels. Tacrolimus 77-87 C-C motif chemokine ligand 11 Homo sapiens 215-224 33512727-4 2021 Additionally, we also performed ELISA and Western blot analyses to show that tacrolimus treatment also reduces the levels of eosinophil-specific cytokines IL-4, IL-5, IL-13, TGF-beta, eosinophil-specific chemokines Eotaxin-1, Eotaxin-2, and progenitors target RCAN1 mRNA levels. Tacrolimus 77-87 C-C motif chemokine ligand 24 Homo sapiens 226-235 33512727-4 2021 Additionally, we also performed ELISA and Western blot analyses to show that tacrolimus treatment also reduces the levels of eosinophil-specific cytokines IL-4, IL-5, IL-13, TGF-beta, eosinophil-specific chemokines Eotaxin-1, Eotaxin-2, and progenitors target RCAN1 mRNA levels. Tacrolimus 77-87 regulator of calcineurin 1 Homo sapiens 260-265 33649515-0 2021 Functional CYP3A variants affecting tacrolimus trough blood concentrations in Chinese renal transplant recipients. Tacrolimus 36-46 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 11-16 33649515-2 2021 We analyzed the association between 58 single nucleotide polymorphisms (SNPs) across the CYP3A gene cluster and the log-transformed tacrolimus concentration/dose ratio (log (C0/D)) in 819 renal transplant recipients (Discovery cohort). Tacrolimus 132-142 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 89-94 33649515-6 2021 Nine SNPs were significantly associated with tacrolimus log (C0/D) after adjustment for CYP3A5*3 and clinical factors. Tacrolimus 45-55 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 88-94 33649515-10 2021 CYP3A7*2, rs4646450, and rs3823812 are proposed as functional SNPs affecting tacrolimus trough blood concentrations in Chinese renal transplant recipients. Tacrolimus 77-87 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 0-6 34058078-0 2021 Association of CYP3A5 polymorphisms and parathyroid hormone with blood level of tacrolimus in patients with end-stage renal disease. Tacrolimus 80-90 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 34058078-0 2021 Association of CYP3A5 polymorphisms and parathyroid hormone with blood level of tacrolimus in patients with end-stage renal disease. Tacrolimus 80-90 parathyroid hormone Homo sapiens 40-59 34058078-1 2021 Because tacrolimus is predominantly metabolized by CYP3A, the blood concentration/dose (C/D) ratio is affected by CYP3A5 polymorphism. Tacrolimus 8-18 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 51-56 34058078-1 2021 Because tacrolimus is predominantly metabolized by CYP3A, the blood concentration/dose (C/D) ratio is affected by CYP3A5 polymorphism. Tacrolimus 8-18 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 114-120 34058078-4 2021 In this study, we examined the influence of CYP3A5 polymorphism on and association of serum intact-PTH (iPTH) level with blood tacrolimus concentration in patients with end-stage renal disease just before kidney transplantation. Tacrolimus 127-137 parathyroid hormone Homo sapiens 99-102 34058078-7 2021 The blood tacrolimus C/D (per body weight) ratio was significantly lower in CYP3A5 expressors than that in CYP3A5 nonexpressors. Tacrolimus 10-20 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 76-82 34058078-9 2021 A multiple logistic regression analysis by stepwise selection identified CYP3A5 polymorphism and serum iPTH level as significant factors associated with tacrolimus C/D. Tacrolimus 153-163 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 73-79 34057812-5 2022 Treatment of prostate cancer cells with FK506, which binds to the FK1 domain of FKBPs, or with MJC13, an inhibitor of FKBP52-AR signaling, also inhibited AR dimer formation. Tacrolimus 40-45 androgen receptor Homo sapiens 154-156 34042216-6 2021 The cyclosporine actions appear to be a class rather than a drug effect because similar exacerbation of LPS nephrotoxicity was observed in rats treated with tacrolimus, another calcineurin inhibitor (CNI). Tacrolimus 157-167 calcineurin binding protein 1 Rattus norvegicus 177-198 34042216-7 2021 Moreover, the deteriorated renal outcomes in LPS/tacrolimus-treated rats were reduced after castration or androgen receptor blockade by flutamide. Tacrolimus 49-59 androgen receptor Rattus norvegicus 106-123 34048854-4 2021 OBJECTIVE: We sought to evaluate whether ocular lesions that FADS mice spontaneously develop are similar to those of AKC patients and to estimate the efficacy of topical treatments with tacrolimus and betamethasone for FADS mice using a novel biomarker for allergic conjunctival disease, tear periostin. Tacrolimus 186-196 periostin, osteoblast specific factor Mus musculus 293-302 33977768-3 2021 Time dependent study was conducted using primary rat hepatocytes treated with TAC (36 microM) for 6, 12, 24 and 48 h. Western blot analysis was performed using cell lysate in order to analyze the regulation of HH pathway proteins including HHIP, SMO, PTCH, IHH, SHH, and GLI transcription factors. Tacrolimus 78-81 Hedgehog-interacting protein Rattus norvegicus 240-244 34006401-8 2021 RESULTS: The wounds treated with 0.1% tacrolimus showed weak labeling and a lower percentage of labeling for smooth muscle actin, a higher proportion of mucin absence, weak staining, fine and organized fibers for Gomori"s Trichrome, strong staining and organized fibers for Verhoeff when compared to controls. Tacrolimus 38-48 actin Oryctolagus cuniculus 123-128 33977768-3 2021 Time dependent study was conducted using primary rat hepatocytes treated with TAC (36 microM) for 6, 12, 24 and 48 h. Western blot analysis was performed using cell lysate in order to analyze the regulation of HH pathway proteins including HHIP, SMO, PTCH, IHH, SHH, and GLI transcription factors. Tacrolimus 78-81 smoothened, frizzled class receptor Rattus norvegicus 246-249 34006401-9 2021 The wounds treated with 0.03% tacrolimus showed weak labeling for smooth muscle actin, a higher proportion of mucin absence, strong staining for Verhoeff when compared to the controls. Tacrolimus 30-40 actin Oryctolagus cuniculus 80-85 33977768-3 2021 Time dependent study was conducted using primary rat hepatocytes treated with TAC (36 microM) for 6, 12, 24 and 48 h. Western blot analysis was performed using cell lysate in order to analyze the regulation of HH pathway proteins including HHIP, SMO, PTCH, IHH, SHH, and GLI transcription factors. Tacrolimus 78-81 patched 1 Rattus norvegicus 251-255 33952632-2 2021 In this study, we tested the hypothesis that tacrolimus, a calcineurin inhibitor, prevents age-associated microstructural atrophy, which we measured using higher-order diffusion MRI, in the middle-aged beagle brain (n = 30, male and female). Tacrolimus 45-55 calcineurin binding protein 1 Homo sapiens 59-80 33977768-3 2021 Time dependent study was conducted using primary rat hepatocytes treated with TAC (36 microM) for 6, 12, 24 and 48 h. Western blot analysis was performed using cell lysate in order to analyze the regulation of HH pathway proteins including HHIP, SMO, PTCH, IHH, SHH, and GLI transcription factors. Tacrolimus 78-81 Indian hedgehog signaling molecule Rattus norvegicus 257-260 33977768-3 2021 Time dependent study was conducted using primary rat hepatocytes treated with TAC (36 microM) for 6, 12, 24 and 48 h. Western blot analysis was performed using cell lysate in order to analyze the regulation of HH pathway proteins including HHIP, SMO, PTCH, IHH, SHH, and GLI transcription factors. Tacrolimus 78-81 sonic hedgehog signaling molecule Rattus norvegicus 262-265 33977768-3 2021 Time dependent study was conducted using primary rat hepatocytes treated with TAC (36 microM) for 6, 12, 24 and 48 h. Western blot analysis was performed using cell lysate in order to analyze the regulation of HH pathway proteins including HHIP, SMO, PTCH, IHH, SHH, and GLI transcription factors. Tacrolimus 78-81 GLI family zinc finger 1 Rattus norvegicus 271-274 33963666-7 2021 The calcineurin inhibitor (CNI) tacrolimus is the mainstay of immunosuppression but can result in AKI. Tacrolimus 32-42 calcineurin binding protein 1 Homo sapiens 4-25 33952632-8 2021 In this study, we show that administration of the calcineurin inhibitor, tacrolimus, over one year prevents age and AD-associated microstructural changes in the hippocampus, parahippocampal cortex, and prefrontal cortex of the middle-aged beagle brain, with no noticeable adverse effects. Tacrolimus 73-83 calcineurin binding protein 1 Homo sapiens 50-71 33962777-1 2021 Tacrolimus, an immunosuppressant prescribed to reduce the risk of organ rejection, is metabolized by cytochrome P450 and is a substrate for P-glycoprotein. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 140-154 33938785-11 2021 FK506 therapy restored cardiac BMP signaling, reduced RV fibrosis in a BMP-dependent manner independent from its immunosuppressive effect, preserved RV capillarization and improved RV function and strain over the time-course of disease. Tacrolimus 0-5 bone morphogenetic protein receptor, type II (serine/threonine kinase) Mus musculus 31-34 33938785-11 2021 FK506 therapy restored cardiac BMP signaling, reduced RV fibrosis in a BMP-dependent manner independent from its immunosuppressive effect, preserved RV capillarization and improved RV function and strain over the time-course of disease. Tacrolimus 0-5 bone morphogenetic protein receptor, type II (serine/threonine kinase) Mus musculus 71-74 33938785-13 2021 Mechanistically, FK506 required ALK1 in hCFs as BMPR2 co-receptor to reduce TGFbeta1-induced proliferation and collagen production. Tacrolimus 17-22 secretory leukocyte peptidase inhibitor Homo sapiens 32-36 33938785-0 2021 Improving Right Ventricular Function by Increasing BMP Signaling with FK506. Tacrolimus 70-75 bone morphogenetic protein receptor, type II (serine/threonine kinase) Mus musculus 51-54 33938785-2 2021 In pre-clinical models, pharmacological activation of bone morphogenetic protein (BMP) signaling with FK506 (Tacrolimus) improved RV function by decreasing RV afterload. Tacrolimus 102-107 bone morphogenetic protein receptor, type II (serine/threonine kinase) Mus musculus 82-85 33938785-2 2021 In pre-clinical models, pharmacological activation of bone morphogenetic protein (BMP) signaling with FK506 (Tacrolimus) improved RV function by decreasing RV afterload. Tacrolimus 109-119 bone morphogenetic protein receptor, type II (serine/threonine kinase) Mus musculus 82-85 33938785-13 2021 Mechanistically, FK506 required ALK1 in hCFs as BMPR2 co-receptor to reduce TGFbeta1-induced proliferation and collagen production. Tacrolimus 17-22 bone morphogenetic protein receptor, type II (serine/threonine kinase) Mus musculus 48-53 33711846-9 2021 While ACE2 level increased in the groups receiving a combination of tacrolimus and losartan or captopril, the level of increase was insignificant, compared to the group treated with tacrolimus alone. Tacrolimus 68-78 angiotensin I converting enzyme 2 Rattus norvegicus 6-10 33938785-13 2021 Mechanistically, FK506 required ALK1 in hCFs as BMPR2 co-receptor to reduce TGFbeta1-induced proliferation and collagen production. Tacrolimus 17-22 transforming growth factor beta 1 Homo sapiens 76-84 33938785-14 2021 Our study demonstrates that increasing cardiac BMP signaling with FK506 improves RV structure and function independent from its previously described beneficial effects on pulmonary vascular remodeling. Tacrolimus 66-71 bone morphogenetic protein receptor, type II (serine/threonine kinase) Mus musculus 47-50 33938785-5 2021 We hypothesized that increasing cardiac BMP signaling with FK506 improves RV structure and function in a model of fixed RV afterload after pulmonary artery banding (PAB). Tacrolimus 59-64 bone morphogenetic protein receptor, type II (serine/threonine kinase) Mus musculus 40-43 33711846-12 2021 CONCLUSION: According to this study, tacrolimus increased the BUN and Cr levels while decreasing the ACE2 levels. Tacrolimus 37-47 angiotensin I converting enzyme 2 Rattus norvegicus 101-105 33716110-4 2021 In the first part of this study, we demonstrated in vitro that tacrolimus and cyclosporine were able to affect viability, inhibit leucocyte proliferation and suppress il2 expression in vitro. Tacrolimus 63-73 interleukin 2 Oncorhynchus mykiss 167-170 33383132-10 2021 CONCLUSIONS: NFATC2 rs150348438, rs6013219, rs1052653, and NFATC1 rs754093, ranking high in scoring, significantly affected the post-transplant eGFR and the incidence of pneumonia, acute rejection, and nephrotoxicity in renal transplant patients taking tacrolimus. Tacrolimus 253-263 nuclear factor of activated T cells 2 Homo sapiens 13-19 33716110-5 2021 In in vivo experiments, both doses of tacrolimus (0.5 and 1.5 mg/kg) and the lower dose of cyclosporine (20 mg/kg) significantly inhibited the expression of il2 in head kidney, three days post-injection. Tacrolimus 38-48 interleukin 2 Oncorhynchus mykiss 157-160 33716110-10 2021 il2 expression in head kidney was also suppressed in grafted animals treated with tacrolimus compared to non-treated group. Tacrolimus 82-92 interleukin 2 Oncorhynchus mykiss 0-3 33925140-9 2021 Indeed, blockade of the CXCL10 receptor CXCR3 at the time of tacrolimus withdrawal prevented CD8+ T cell infiltration and the regression of SCC. Tacrolimus 61-71 chemokine (C-X-C motif) ligand 10 Mus musculus 24-30 33516819-6 2021 In addition, tacrolimus treatment combined with IR injury increased histological injury (tubular vacuolation, glomerulosclerosis and interstitial fibrosis), as well as alpha-smooth muscle actin (alpha-SMA), transforming growth factor-beta (TGF-beta), and kidney injury molecule-1 (KIM-1) expression in the renal cortex. Tacrolimus 13-23 actin gamma 2, smooth muscle Rattus norvegicus 168-193 33516819-6 2021 In addition, tacrolimus treatment combined with IR injury increased histological injury (tubular vacuolation, glomerulosclerosis and interstitial fibrosis), as well as alpha-smooth muscle actin (alpha-SMA), transforming growth factor-beta (TGF-beta), and kidney injury molecule-1 (KIM-1) expression in the renal cortex. Tacrolimus 13-23 transforming growth factor alpha Rattus norvegicus 240-248 33516819-6 2021 In addition, tacrolimus treatment combined with IR injury increased histological injury (tubular vacuolation, glomerulosclerosis and interstitial fibrosis), as well as alpha-smooth muscle actin (alpha-SMA), transforming growth factor-beta (TGF-beta), and kidney injury molecule-1 (KIM-1) expression in the renal cortex. Tacrolimus 13-23 hepatitis A virus cellular receptor 1 Rattus norvegicus 255-279 33516819-6 2021 In addition, tacrolimus treatment combined with IR injury increased histological injury (tubular vacuolation, glomerulosclerosis and interstitial fibrosis), as well as alpha-smooth muscle actin (alpha-SMA), transforming growth factor-beta (TGF-beta), and kidney injury molecule-1 (KIM-1) expression in the renal cortex. Tacrolimus 13-23 hepatitis A virus cellular receptor 1 Rattus norvegicus 281-286 33185367-7 2021 Tacrolimus/m-TOR inhibitors were reduced by 50% and all antimetabolites were discontinued. Tacrolimus 0-10 RAR related orphan receptor C Homo sapiens 13-16 32761929-8 2021 Further, in a gene expression analysis of infected Kupffer cells, the TREM-1 pathway was the one with the most significant downregulation after tacrolimus treatment. Tacrolimus 144-154 triggering receptor expressed on myeloid cells 1 Mus musculus 70-76 32761929-11 2021 Our results indicate that tacrolimus treatment has a significant impact directly on Kupffer cells and on TREM-1, thereby compromising their capacity to fend off infections. Tacrolimus 26-36 triggering receptor expressed on myeloid cells 1 Mus musculus 105-111 33996955-0 2021 Case Report: Clinical Remission in a Cat With Severe Bilateral Eosinophilic Keratitis Receiving Combined Immunosuppressive Therapy (Triamcinolone Acetonide and Tacrolimus). Tacrolimus 160-170 catalase Homo sapiens 37-40 33925140-9 2021 Indeed, blockade of the CXCL10 receptor CXCR3 at the time of tacrolimus withdrawal prevented CD8+ T cell infiltration and the regression of SCC. Tacrolimus 61-71 chemokine (C-X-C motif) receptor 3 Mus musculus 40-45 33887727-0 2021 Outcomes of Interleukin-2 Receptor Antagonist Induction Therapy in Standard-Risk Renal Transplant Recipients Maintained on Tacrolimus: A Systematic Review and Meta-Analysis. Tacrolimus 123-133 interleukin 2 receptor subunit alpha Homo sapiens 12-45 33896035-9 2021 CONCLUSION: Our data support the quick conversion to tacrolimus in the condition of persistent low CSA levels with acceptable efficacy and safety. Tacrolimus 53-63 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 99-102 33896348-7 2021 CONCLUSIONS: The inhibitory effect of PSL and TAC combination therapy on the progression of fibrosis in SSc-PIP may be superior to that of PSL and AZA in the long period. Tacrolimus 46-49 prolactin induced protein Homo sapiens 108-111 33887167-4 2021 Tac group showed significantly lower expression of HO-1. Tacrolimus 0-3 heme oxygenase 1 Rattus norvegicus 51-55 33967795-16 2021 Conclusions: These results support that CYP3A5 and UGT1A9 genotyping in pediatric recipients might be useful and advisable to guide TAC and MPA dosing and monitoring in children that undergo kidney transplantation. Tacrolimus 132-135 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 40-46 33887167-7 2021 Tac group showed the highest expression of caspase-3. Tacrolimus 0-3 caspase 3 Rattus norvegicus 43-52 33967795-16 2021 Conclusions: These results support that CYP3A5 and UGT1A9 genotyping in pediatric recipients might be useful and advisable to guide TAC and MPA dosing and monitoring in children that undergo kidney transplantation. Tacrolimus 132-135 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 51-57 33882512-5 2021 For in vitro assessment, human renal epithelium HK-2 cells were cultured under serum starvation conditions or with tacrolimus.The administration of SLPI (250 mug/kg, i.p) reduced elevated plasma creatinine and blood urea nitrogen levels, tissue myeloperoxidase content, and acute tubular necrosis induced by kidney damage. Tacrolimus 115-125 secretory leukocyte peptidase inhibitor Homo sapiens 148-152 33883562-9 2021 Consistent with these data, expression levels of genes encoding lysyl hydroxylase 2 (LH2) and its molecular chaperone FK506-binding protein 65 were both significantly increased in neoplastic samples. Tacrolimus 118-123 procollagen-lysine,2-oxoglutarate 5-dioxygenase 2 Homo sapiens 64-83 33883562-9 2021 Consistent with these data, expression levels of genes encoding lysyl hydroxylase 2 (LH2) and its molecular chaperone FK506-binding protein 65 were both significantly increased in neoplastic samples. Tacrolimus 118-123 procollagen-lysine,2-oxoglutarate 5-dioxygenase 2 Homo sapiens 85-88 33865397-8 2021 Moreover, HGC-TAC administration regulated renal injury via the TGF-beta1/MAPK/NF-kappaB signaling pathway. Tacrolimus 14-17 transforming growth factor, beta 1 Mus musculus 64-73 33847343-8 2021 The combination of vedolizumab with tacrolimus further reduced the number of infiltrating CD3+ T cells and CD68+ monocytes/macrophages and was superior in ameliorating intestinal inflammation when compared to vedolizumab monotreatment. Tacrolimus 36-46 CD68 molecule Homo sapiens 107-111 33894161-4 2021 We performed a high-throughput phenotypic screen and identified a series of FK506 analogs that act as potent BMP potentiators by sequestering FKBP12 from BMP type I receptors. Tacrolimus 76-81 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 142-148 33873145-4 2021 We observed that in a 4-Nitroquinoline N-oxide (4NQO)-induced oral carcinogenesis model, tacrolimus treatment was associated with a significantly lower ratio of cancer formation (52.94% vs. 90%) and a lower proportion of Ki67 and proliferation cell nuclear antigen (PCNA) -positive cells in lesion areas (P < 0.001). Tacrolimus 89-99 proliferating cell nuclear antigen Homo sapiens 230-271 33873145-9 2021 Tacrolimus induced G1/S phase arrest and significantly downregulated the expression of cyclinD1, cyclinE1, and c-Myc. Tacrolimus 0-10 cyclin D1 Homo sapiens 87-95 33873145-9 2021 Tacrolimus induced G1/S phase arrest and significantly downregulated the expression of cyclinD1, cyclinE1, and c-Myc. Tacrolimus 0-10 cyclin E1 Homo sapiens 97-105 33873145-9 2021 Tacrolimus induced G1/S phase arrest and significantly downregulated the expression of cyclinD1, cyclinE1, and c-Myc. Tacrolimus 0-10 MYC proto-oncogene, bHLH transcription factor Homo sapiens 111-116 33873145-10 2021 These results suggest that tacrolimus induces G1/S phase arrest via inhibition of cyclinD1, cyclinE1, and c-Myc expression and retards oral cell carcinogenesis in vitro and in vivo. Tacrolimus 27-37 cyclin D1 Homo sapiens 82-90 33873145-10 2021 These results suggest that tacrolimus induces G1/S phase arrest via inhibition of cyclinD1, cyclinE1, and c-Myc expression and retards oral cell carcinogenesis in vitro and in vivo. Tacrolimus 27-37 cyclin E1 Homo sapiens 92-100 33873145-10 2021 These results suggest that tacrolimus induces G1/S phase arrest via inhibition of cyclinD1, cyclinE1, and c-Myc expression and retards oral cell carcinogenesis in vitro and in vivo. Tacrolimus 27-37 MYC proto-oncogene, bHLH transcription factor Homo sapiens 106-111 33571621-3 2021 A thin-film hydration method was used to encapsulate TAC within the chitosan-based amphiphile: N-palmitoyl-N-monomethyl-N,N-dimethyl-N,N,N-trimethyl-6-O-glycolchitosan (Molecular Envelope Technology - MET) in an aqueous formulation. Tacrolimus 53-56 hepatocyte growth factor receptor Oryctolagus cuniculus 201-204 33853841-2 2021 The vast majority of patients are treated with the calcineurin inhibitor tacrolimus as the primary agent in combination with mycophenolate, with or without corticosteroids. Tacrolimus 73-83 calcineurin binding protein 1 Homo sapiens 51-72 33894161-0 2021 Phenotypic screen identifies calcineurin-sparing FK506 analogs as BMP potentiators for treatment of acute kidney injury. Tacrolimus 49-54 bone morphogenetic protein 1 Homo sapiens 66-69 33894161-4 2021 We performed a high-throughput phenotypic screen and identified a series of FK506 analogs that act as potent BMP potentiators by sequestering FKBP12 from BMP type I receptors. Tacrolimus 76-81 bone morphogenetic protein 1 Homo sapiens 109-112 33936035-7 2021 Through gene set enrichment analysis, we found that this subpopulation is similar to virus-specific CD8+ T cells and T cells that mediate acute rejection in patients using tacrolimus and belatacept, a selective costimulation blocker. Tacrolimus 172-182 CD8a molecule Homo sapiens 100-103 33166580-7 2021 Anti-PLA2R titers showed a significant decrease in both groups but the proportion of anti-PLA2R-positive patients who achieved immunological response (depletion of anti-PLA2R antibodies) was significantly higher at three and six months in the corticosteroid-cyclophosphamide group (77% and 92%, respectively), as compared to the tacrolimus-rituximab group (45% and 70%, respectively). Tacrolimus 329-339 phospholipase A2 receptor 1 Homo sapiens 90-95 33920149-0 2021 Relationship between CYP3A5 Polymorphism and Tacrolimus Blood Concentration Changes in Allogeneic Hematopoietic Stem Cell Transplant Recipients during Continuous Infusion. Tacrolimus 45-55 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 21-27 33920149-1 2021 A polymorphism in the gene encoding the metabolic enzyme cytochrome P450 family 3 subfamily A member 5 (CYP3A5) is a particularly influential factor in the use of tacrolimus in Japanese patients. Tacrolimus 163-173 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 57-102 33920149-1 2021 A polymorphism in the gene encoding the metabolic enzyme cytochrome P450 family 3 subfamily A member 5 (CYP3A5) is a particularly influential factor in the use of tacrolimus in Japanese patients. Tacrolimus 163-173 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 104-110 33920149-2 2021 Those who are homozygotic for the *3 mutation lack CYP3A5 activity, which results in substantial individual differences in tacrolimus metabolism. Tacrolimus 123-133 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 51-57 33920149-3 2021 The aim of this study was to analyze the relationship between individual differences in tacrolimus blood concentration changes and CYP3A5 polymorphisms in allogeneic hematopoietic stem cell transplantation recipients during the period of increasing blood concentration of the drug following treatment onset. Tacrolimus 88-98 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 131-137 33920149-7 2021 This study reveals the effects of CYP3A5 polymorphism on continuous changes in tacrolimus blood concentration. Tacrolimus 79-89 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 32986876-0 2021 Clinical significance of personalized tacrolimus dosing by adjusting to donor CYP3A-status in liver transplant recipients. Tacrolimus 38-48 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 78-83 32986876-1 2021 AIMS: Donor"s CYP3A-status (CYP3A5 genotype and CYP3A4 expression) can provide prognostic information regarding tacrolimus-metabolizing capacity of the liver graft and initial tacrolimus dosing for therapeutic blood concentrations in liver transplants. Tacrolimus 112-122 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-19 32986876-1 2021 AIMS: Donor"s CYP3A-status (CYP3A5 genotype and CYP3A4 expression) can provide prognostic information regarding tacrolimus-metabolizing capacity of the liver graft and initial tacrolimus dosing for therapeutic blood concentrations in liver transplants. Tacrolimus 112-122 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 28-34 32986876-1 2021 AIMS: Donor"s CYP3A-status (CYP3A5 genotype and CYP3A4 expression) can provide prognostic information regarding tacrolimus-metabolizing capacity of the liver graft and initial tacrolimus dosing for therapeutic blood concentrations in liver transplants. Tacrolimus 112-122 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-54 32986876-1 2021 AIMS: Donor"s CYP3A-status (CYP3A5 genotype and CYP3A4 expression) can provide prognostic information regarding tacrolimus-metabolizing capacity of the liver graft and initial tacrolimus dosing for therapeutic blood concentrations in liver transplants. Tacrolimus 176-186 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-19 32986876-1 2021 AIMS: Donor"s CYP3A-status (CYP3A5 genotype and CYP3A4 expression) can provide prognostic information regarding tacrolimus-metabolizing capacity of the liver graft and initial tacrolimus dosing for therapeutic blood concentrations in liver transplants. Tacrolimus 176-186 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 28-34 32986876-2 2021 The present work prospectively investigated whether CYP3A-status guided tacrolimus therapy has any potential clinical benefit for recipients in the early post-operative period. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 52-57 32986876-3 2021 METHODS: The contribution of preliminary assaying of donor CYP3A-status to the optimization of initial tacrolimus therapy and to the reduction of adverse events (acute rejection, infection, nephrotoxicity) was investigated in 112 liver transplant recipients (CYPtest group) comparing to 101 control patients on tacrolimus concentration guided therapy. Tacrolimus 103-113 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 59-64 33597150-8 2021 PD1 upregulation is accentuated in the presence of rapamycin but prevented by tacrolimus. Tacrolimus 78-88 programmed cell death 1 Homo sapiens 0-3 33166580-7 2021 Anti-PLA2R titers showed a significant decrease in both groups but the proportion of anti-PLA2R-positive patients who achieved immunological response (depletion of anti-PLA2R antibodies) was significantly higher at three and six months in the corticosteroid-cyclophosphamide group (77% and 92%, respectively), as compared to the tacrolimus-rituximab group (45% and 70%, respectively). Tacrolimus 329-339 phospholipase A2 receptor 1 Homo sapiens 90-95 33724664-4 2021 Utilizing a combination of in vitro techniques and a mouse model of CNI nephrotoxicity, we found that the CNIs, cyclosporine A (CsA) and tacrolimus (TAC), share a similar mechanism of tubular epithelial kidney cell injury, including mitochondrial dysfunction and release of High Mobility Group Box I (HMGB1). Tacrolimus 137-147 high mobility group box 1 Mus musculus 301-306 33027230-12 2021 Obesity, high mycophenolate mofetil daily dose at M3, and CYP3A5 expression were independently associated with higher tacrolimus exposure. Tacrolimus 118-128 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 58-64 33756310-14 2021 One study found that for each 5 ng/mL per year of tacrolimus exposure, defined by consecutive AUC, eGFR declined by 1.3 mL/min/1.73m2 (p < 0.001). Tacrolimus 50-60 epidermal growth factor receptor Homo sapiens 99-103 33754647-0 2021 Effects of Tacrolimus on c-Fos in Hippocampus and Memory Performances in Streptozotocin Model of Alzheimer"s Disease of Rats. Tacrolimus 11-21 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 25-30 33754647-4 2021 Our hypothesis is that a calcineurin inhibitor, tacrolimus, could prevent the accumulation and the decrease of the neuronal cells. Tacrolimus 48-58 calcineurin binding protein 1 Rattus norvegicus 25-46 33724664-4 2021 Utilizing a combination of in vitro techniques and a mouse model of CNI nephrotoxicity, we found that the CNIs, cyclosporine A (CsA) and tacrolimus (TAC), share a similar mechanism of tubular epithelial kidney cell injury, including mitochondrial dysfunction and release of High Mobility Group Box I (HMGB1). Tacrolimus 149-152 high mobility group box 1 Mus musculus 301-306 33751414-0 2021 Clinical Impact of the Adaptation of Initial Tacrolimus Dosing to the CYP3A5 Genotype After Kidney Transplantation: Systematic Review and Meta-Analysis of Randomized Controlled Trials. Tacrolimus 45-55 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 70-76 33746516-0 2021 Evaluating the Impact of CYP3A5 Genotype on Post-Transplant Healthcare Resource Utilization in Pediatric Renal and Heart Transplant Recipients Receiving Tacrolimus. Tacrolimus 153-163 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 25-31 33746516-1 2021 Purpose: CYP3A5 genotype is a significant contributor to inter-individual tacrolimus exposure and may impact the time required to achieve therapeutic concentrations and number of tacrolimus dose adjustments in transplant patients. Tacrolimus 74-84 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 9-15 33746516-1 2021 Purpose: CYP3A5 genotype is a significant contributor to inter-individual tacrolimus exposure and may impact the time required to achieve therapeutic concentrations and number of tacrolimus dose adjustments in transplant patients. Tacrolimus 179-189 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 9-15 33746516-11 2021 CYP3A5 genotype was not associated with any outcomes in renal transplant, however, a CYP3A5 expresser phenotype was a predictor of more dose changes, more tacrolimus concentrations, longer length of stay, and higher total charges in heart transplant recipients. Tacrolimus 155-165 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 85-91 33746516-14 2021 Conclusion: CYP3A5 genotype may predict healthcare resource utilization in the first year post-transplant, although this may be mitigated by differences in tacrolimus management. Tacrolimus 156-166 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 33732042-11 2021 The results of the GSE84908 data analysis showed that after tacrolimus treatment, the expression of DAAM1 was significantly increased (p = 0.015). Tacrolimus 60-70 dishevelled associated activator of morphogenesis 1 Homo sapiens 100-105 33692448-12 2021 Gal-9 can be regarded as a new biomarker for evaluating RA activity and therapeutic effect, including TAC. Tacrolimus 102-105 galectin 9 Homo sapiens 0-5 33248001-0 2021 Temporary decrease in tacrolimus clearance in cytochrome P450 3A5 non-expressors early after living donor kidney transplantation: Effect of interleukin 6-induced suppression of the cytochrome P450 3A gene. Tacrolimus 22-32 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-65 33248001-4 2021 The blood tacrolimus concentration per dose ratio (C/D) temporarily increased post-kidney transplantation on days 3 to 4 only in CYP3A5 non-expressors. Tacrolimus 10-20 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 129-135 33248001-8 2021 We hypothesise that CYP3A5 non-expressors might exhibit a temporary decrease in the oral clearance of tacrolimus via an increase in serum IL-6 concentrations early after kidney transplantation. Tacrolimus 102-112 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 33248001-8 2021 We hypothesise that CYP3A5 non-expressors might exhibit a temporary decrease in the oral clearance of tacrolimus via an increase in serum IL-6 concentrations early after kidney transplantation. Tacrolimus 102-112 interleukin 6 Homo sapiens 138-142 32749698-0 2021 Roles of BATF/JUN/IRF4 complex in tacrolimus mediated immunosuppression on Tfh cells in acute rejection after liver transplantation. Tacrolimus 34-44 basic leucine zipper ATF-like transcription factor Rattus norvegicus 9-13 32749698-0 2021 Roles of BATF/JUN/IRF4 complex in tacrolimus mediated immunosuppression on Tfh cells in acute rejection after liver transplantation. Tacrolimus 34-44 interferon regulatory factor 4 Rattus norvegicus 18-22 32749698-4 2021 This study aimed to explore the role of the BATF/JUN/IRF4 complex in rejection after LTx by treatment with Tac. Tacrolimus 107-110 basic leucine zipper ATF-like transcription factor Rattus norvegicus 44-48 32749698-4 2021 This study aimed to explore the role of the BATF/JUN/IRF4 complex in rejection after LTx by treatment with Tac. Tacrolimus 107-110 interferon regulatory factor 4 Rattus norvegicus 53-57 32749698-8 2021 The results demonstrated that Tac prolonged the allografts survival and attenuated inflammation injury, and decreased the percentage frequencies of T follicular helper (Tfh) cells in peripheral blood mononuclear cells and inhibited B-cell lymphoma 6 (Bcl-6) and IL-6 expression in Tfh cells. Tacrolimus 30-33 BCL6, transcription repressor Rattus norvegicus 251-256 32749698-8 2021 The results demonstrated that Tac prolonged the allografts survival and attenuated inflammation injury, and decreased the percentage frequencies of T follicular helper (Tfh) cells in peripheral blood mononuclear cells and inhibited B-cell lymphoma 6 (Bcl-6) and IL-6 expression in Tfh cells. Tacrolimus 30-33 interleukin 6 Rattus norvegicus 262-266 32749698-10 2021 In conclusion, the attenuation of rejection injury may be dependent on the NFAT-BATF/JUN/IRF4-IL-21 axis, and the BATF/JUN/IRF4 complex participates in the inhibition of IL-21-producing Tfh cells after treatment with Tac. Tacrolimus 217-220 basic leucine zipper ATF-like transcription factor Rattus norvegicus 114-118 32749698-10 2021 In conclusion, the attenuation of rejection injury may be dependent on the NFAT-BATF/JUN/IRF4-IL-21 axis, and the BATF/JUN/IRF4 complex participates in the inhibition of IL-21-producing Tfh cells after treatment with Tac. Tacrolimus 217-220 interferon regulatory factor 4 Rattus norvegicus 123-127 32749698-10 2021 In conclusion, the attenuation of rejection injury may be dependent on the NFAT-BATF/JUN/IRF4-IL-21 axis, and the BATF/JUN/IRF4 complex participates in the inhibition of IL-21-producing Tfh cells after treatment with Tac. Tacrolimus 217-220 interleukin 21 Rattus norvegicus 170-175 33732042-12 2021 Conclusion: Tacrolimus may inhibit the human immune response by affecting the expression of DAAM1 in liver transplant patients. Tacrolimus 12-22 dishevelled associated activator of morphogenesis 1 Homo sapiens 92-97 32541562-5 2021 As we enter the fourth decade of tacrolimus use, newer studies utilizing of novel combinations (as with the mammalian target of rapamycin (mTOR) inhibitor, everolimus, and T-cell co-stimulation blockade with belatacept) offer potential for enhanced benefits. Tacrolimus 33-43 mechanistic target of rapamycin kinase Homo sapiens 108-137 33673653-0 2021 Investigation of the Impact of CYP3A5 Polymorphism on Drug-Drug Interaction between Tacrolimus and Schisantherin A/Schisandrin A Based on Physiologically-Based Pharmacokinetic Modeling. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 31-37 33673653-8 2021 An optimized physiologically-based pharmacokinetic (PBPK) model integrated with CYP3A5 polymorphism was successfully established, providing more insights regarding the long-term DDI between tacrolimus and Wuzhi capsules in patients with different CYP3A5 genotypes. Tacrolimus 190-200 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 80-86 33673653-8 2021 An optimized physiologically-based pharmacokinetic (PBPK) model integrated with CYP3A5 polymorphism was successfully established, providing more insights regarding the long-term DDI between tacrolimus and Wuzhi capsules in patients with different CYP3A5 genotypes. Tacrolimus 190-200 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 247-253 33692688-0 2021 Commentary: The Clinical Impact of the C0/D Ratio and the CYP3A5 Genotype on Outcome in Tacrolimus Treated Kidney Transplant Recipients. Tacrolimus 88-98 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 58-64 33654003-0 2021 Unraveling the Genomic Architecture of the CYP3A Locus and ADME Genes for Personalized Tacrolimus Dosing. Tacrolimus 87-97 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 43-48 33638630-0 2021 CD28 confers CD4+ T cells with resistance to cyclosporin A and tacrolimus but to different degrees. Tacrolimus 63-73 CD4 molecule Homo sapiens 13-16 33638630-9 2021 RESULTS: Anti-CD28 mAb conferred CD4+ T cells with resistance to both CSA and TAC, and CD28"s effect on the latter was approximately twice that on the former. Tacrolimus 78-81 CD4 molecule Homo sapiens 33-36 33638630-11 2021 The addition of LPS to culture containing DCs seemed to make CD4+ T cells slightly resistant to TAC but not to CSA. Tacrolimus 96-99 CD4 molecule Homo sapiens 61-64 33637323-7 2021 We also found the higher frequency of Eomes+dCD4+T cells from miscarriage in response to cyclosporine, tacrolimus, Trophoblasts, and HTR8/SVneo cell line, might provide new strategy for therapy to promote maternal-fetal tolerance and prevent pregnancy loss. Tacrolimus 103-113 eomesodermin Homo sapiens 38-43 33539328-8 2021 The patient had several FSGS relapses that were treated by different combinations of plasmapheresis, pulse steroid, mycophenolic acid, tacrolimus, prednisolone, IVIG, and IV rituximab. Tacrolimus 135-145 actinin alpha 4 Homo sapiens 24-28 33654003-6 2021 RESULTS: GWAS verified that CYP3A5*3 is the only common variant associated with TAC PK variability in Koreans. Tacrolimus 80-83 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 28-34 33654003-7 2021 We detected several CYP3A5 and CYP3A4 rare variants which could potentially affect TAC metabolism. Tacrolimus 83-86 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 33654003-7 2021 We detected several CYP3A5 and CYP3A4 rare variants which could potentially affect TAC metabolism. Tacrolimus 83-86 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 33654003-10 2021 Gene-based association tests in the 61 absorption, distribution, metabolism, and excretion (ADME) genes revealed that CYP1A1 are associated with additional TAC PK variability: CYP1A1 rare variant carriers among CYP3A5 poor metabolizers showed lower TAC trough levels than the noncarrier controls. Tacrolimus 156-159 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 118-124 33654003-10 2021 Gene-based association tests in the 61 absorption, distribution, metabolism, and excretion (ADME) genes revealed that CYP1A1 are associated with additional TAC PK variability: CYP1A1 rare variant carriers among CYP3A5 poor metabolizers showed lower TAC trough levels than the noncarrier controls. Tacrolimus 156-159 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 176-182 33654003-10 2021 Gene-based association tests in the 61 absorption, distribution, metabolism, and excretion (ADME) genes revealed that CYP1A1 are associated with additional TAC PK variability: CYP1A1 rare variant carriers among CYP3A5 poor metabolizers showed lower TAC trough levels than the noncarrier controls. Tacrolimus 156-159 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 211-217 32843687-0 2021 Influence CYP3A polymorphisms on tacrolimus pharmacokinetics in kidney transplant recipients. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 10-15 33604334-10 2020 In cultured podocytes, FK506 activated OSR1/SPAK, increased lamellipodia, accelerated cell migration, and promoted traction force. Tacrolimus 23-28 serine/threonine kinase 39 Mus musculus 44-48 33604334-11 2020 These actions of FK506 were reduced by depletion of WNK1. Tacrolimus 17-22 WNK lysine deficient protein kinase 1 Mus musculus 52-56 33093072-3 2021 From December 2016 to October 2019, five vitiligo patients from Peking Union Medical College Hospital were treated with simvastatin and tacrolimus. Tacrolimus 136-146 VAMAS6 Homo sapiens 41-49 32908236-0 2021 Correction: Influence of CYP3A polymorphisms on tacrolimus pharmacokinetics in kidney transplant recipients. Tacrolimus 48-58 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-30 32843687-3 2021 This study aimed to determine, in kidney transplant patients, the influence of different genotypic clusters involving these SNPs CYP3A4*1B, CYP3A4*22, and CYP3A5*3 on Tacrolimus bioavailability during the first (PTP1) and the second (PTP2) posttransplant phase (PT). Tacrolimus 167-177 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 129-135 32843687-3 2021 This study aimed to determine, in kidney transplant patients, the influence of different genotypic clusters involving these SNPs CYP3A4*1B, CYP3A4*22, and CYP3A5*3 on Tacrolimus bioavailability during the first (PTP1) and the second (PTP2) posttransplant phase (PT). Tacrolimus 167-177 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 140-146 32843687-3 2021 This study aimed to determine, in kidney transplant patients, the influence of different genotypic clusters involving these SNPs CYP3A4*1B, CYP3A4*22, and CYP3A5*3 on Tacrolimus bioavailability during the first (PTP1) and the second (PTP2) posttransplant phase (PT). Tacrolimus 167-177 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 155-161 33348024-0 2021 Multiple microRNAs regulate tacrolimus metabolism through CYP3A5. Tacrolimus 28-38 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 58-64 33348024-1 2021 The CYP3A5 gene polymorphism accounts for the majority of inter-individual variability in tacrolimus pharmacokinetics. Tacrolimus 90-100 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 33572801-7 2021 The 2.17 A structure of this ALK2-FKBP12.6 complex bound to the inhibitor dorsomorphin showed FKBP12.6 binding to the GS domain of ALK2 in a manner equivalent to the FKBP12 complex, with ALK2 residues Phe198 and Leu199 extending into the FK506-binding pocket of FKBP12.6. Tacrolimus 238-243 activin A receptor type 1 Homo sapiens 29-33 33348024-2 2021 We found that the basal expression of CYP3A5 in donor grafts also played a significant role in tacrolimus metabolism under the same genetic conditions after pediatric liver transplantation. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 38-44 33348024-6 2021 We found that these miRNAs inhibited tacrolimus metabolism that was dependent on CYP3A5. Tacrolimus 37-47 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 81-87 33348024-8 2021 Luciferase reporter assays and functional studies showed that miR-26b-5p inhibited tacrolimus metabolism by directly regulating CYP3A5, while miR-29a-5p, miR-99a-5p, and miR-532-5p targeted HNF4alpha, NR1I3, and NR1I2, respectively, in turn regulating the downstream expression of CYP3A5; the corresponding target gene siRNAs markedly abolished the effects caused by miRNA inhibitors. Tacrolimus 83-93 microRNA 26b Homo sapiens 62-69 33348024-8 2021 Luciferase reporter assays and functional studies showed that miR-26b-5p inhibited tacrolimus metabolism by directly regulating CYP3A5, while miR-29a-5p, miR-99a-5p, and miR-532-5p targeted HNF4alpha, NR1I3, and NR1I2, respectively, in turn regulating the downstream expression of CYP3A5; the corresponding target gene siRNAs markedly abolished the effects caused by miRNA inhibitors. Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 128-134 33348024-9 2021 Also, the expression of miR-29a-3p, miR-99a-5p, miR-532-5p, and miR-26b-5p in donor grafts were negatively correlated with tacrolimus C/D following pediatric liver transplantation. Tacrolimus 123-133 microRNA 532 Homo sapiens 48-55 33348024-9 2021 Also, the expression of miR-29a-3p, miR-99a-5p, miR-532-5p, and miR-26b-5p in donor grafts were negatively correlated with tacrolimus C/D following pediatric liver transplantation. Tacrolimus 123-133 microRNA 26b Homo sapiens 64-71 33515077-0 2021 Correction to: CYP3A5 and CYP3A7 genetic polymorphisms affect tacrolimus concentration in pediatric patients with nephrotic syndrome. Tacrolimus 62-72 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 33515077-0 2021 Correction to: CYP3A5 and CYP3A7 genetic polymorphisms affect tacrolimus concentration in pediatric patients with nephrotic syndrome. Tacrolimus 62-72 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 26-32 33572801-7 2021 The 2.17 A structure of this ALK2-FKBP12.6 complex bound to the inhibitor dorsomorphin showed FKBP12.6 binding to the GS domain of ALK2 in a manner equivalent to the FKBP12 complex, with ALK2 residues Phe198 and Leu199 extending into the FK506-binding pocket of FKBP12.6. Tacrolimus 238-243 FKBP prolyl isomerase 1B Homo sapiens 34-42 33572801-7 2021 The 2.17 A structure of this ALK2-FKBP12.6 complex bound to the inhibitor dorsomorphin showed FKBP12.6 binding to the GS domain of ALK2 in a manner equivalent to the FKBP12 complex, with ALK2 residues Phe198 and Leu199 extending into the FK506-binding pocket of FKBP12.6. Tacrolimus 238-243 FKBP prolyl isomerase 1B Homo sapiens 94-102 33572801-7 2021 The 2.17 A structure of this ALK2-FKBP12.6 complex bound to the inhibitor dorsomorphin showed FKBP12.6 binding to the GS domain of ALK2 in a manner equivalent to the FKBP12 complex, with ALK2 residues Phe198 and Leu199 extending into the FK506-binding pocket of FKBP12.6. Tacrolimus 238-243 activin A receptor type 1 Homo sapiens 131-135 33572801-7 2021 The 2.17 A structure of this ALK2-FKBP12.6 complex bound to the inhibitor dorsomorphin showed FKBP12.6 binding to the GS domain of ALK2 in a manner equivalent to the FKBP12 complex, with ALK2 residues Phe198 and Leu199 extending into the FK506-binding pocket of FKBP12.6. Tacrolimus 238-243 FKBP prolyl isomerase 1A Homo sapiens 34-40 33572801-7 2021 The 2.17 A structure of this ALK2-FKBP12.6 complex bound to the inhibitor dorsomorphin showed FKBP12.6 binding to the GS domain of ALK2 in a manner equivalent to the FKBP12 complex, with ALK2 residues Phe198 and Leu199 extending into the FK506-binding pocket of FKBP12.6. Tacrolimus 238-243 activin A receptor type 1 Homo sapiens 131-135 33572801-7 2021 The 2.17 A structure of this ALK2-FKBP12.6 complex bound to the inhibitor dorsomorphin showed FKBP12.6 binding to the GS domain of ALK2 in a manner equivalent to the FKBP12 complex, with ALK2 residues Phe198 and Leu199 extending into the FK506-binding pocket of FKBP12.6. Tacrolimus 238-243 FKBP prolyl isomerase 1B Homo sapiens 94-102 33534527-0 2021 COMMENTARY: Unraveling the Genomic Architecture of the CYP3A Locus and ADME Genes for Personalized Tacrolimus Dosing. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 55-60 33536919-0 2020 Astragaloside IV Alleviates Tacrolimus-Induced Chronic Nephrotoxicity via p62-Keap1-Nrf2 Pathway. Tacrolimus 28-38 nucleoporin 62 Homo sapiens 74-77 33536919-0 2020 Astragaloside IV Alleviates Tacrolimus-Induced Chronic Nephrotoxicity via p62-Keap1-Nrf2 Pathway. Tacrolimus 28-38 kelch like ECH associated protein 1 Homo sapiens 78-83 33536919-0 2020 Astragaloside IV Alleviates Tacrolimus-Induced Chronic Nephrotoxicity via p62-Keap1-Nrf2 Pathway. Tacrolimus 28-38 NFE2 like bZIP transcription factor 2 Homo sapiens 84-88 33431785-0 2021 Suppression of Allograft Fibrosis by Regulation of Mammalian Target of Rapamycin-Related Protein Expression in Kidney-Transplanted Recipients Treated with Everolimus and Reduced Tacrolimus. Tacrolimus 178-188 mechanistic target of rapamycin kinase Homo sapiens 51-80 32555441-0 2021 L-Carnitine protects against tacrolimus-induced renal injury by attenuating programmed cell death via PI3K/AKT/PTEN signaling. Tacrolimus 29-39 AKT serine/threonine kinase 1 Rattus norvegicus 107-110 33432012-1 2021 Tacrolimus (TAC) pharmacokinetics is influenced by the donor CYP3A5 genotype and the age of pediatric liver recipients. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-67 33432012-1 2021 Tacrolimus (TAC) pharmacokinetics is influenced by the donor CYP3A5 genotype and the age of pediatric liver recipients. Tacrolimus 12-15 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-67 33432012-6 2021 TAC concentration/weight-adjusted dose ratio was significantly lower in recipients from CYP3A5*1/*3 donors or with extra-large (GRWR > 5%) or large (GRWR 3-5%) grafts. Tacrolimus 0-3 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 88-94 33432012-9 2021 In conclusion, TAC starting dose could be guided according to the donor CYP3A5 genotype and GRWR, allowing for a quicker achievement of target concentrations and eventually reducing the risk of rejection. Tacrolimus 15-18 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 72-78 33422095-5 2021 In this study, we identified a significant increase in the expression of miRNA-690 (miR-690) in the medial prefrontal cortex (mPFC) of FK506-binding protein 51 knock-out (Fkbp5 KO) mice. Tacrolimus 135-140 FK506 binding protein 5 Mus musculus 171-176 32956624-5 2021 We developed a vector-free clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9-based good manufacturing practice (GMP)-compliant protocol that efficiently targets and knocks out the gene for the adaptor protein FK506-binding protein 12 (FKBP12), required for the immunosuppressive function of tacrolimus. Tacrolimus 313-323 FKBP prolyl isomerase 1A Homo sapiens 231-255 32555441-0 2021 L-Carnitine protects against tacrolimus-induced renal injury by attenuating programmed cell death via PI3K/AKT/PTEN signaling. Tacrolimus 29-39 phosphatase and tensin homolog Rattus norvegicus 111-115 32555441-10 2021 Human kidney proximal tubular epithelial cells (HK-2 cells) were treated with TAC (50 mug/mL) in vitro, which induced production of intracellular reactive oxygen species and expression of an array of genes controlling programmed cell death (pyroptosis, apoptosis, and autophagy) through interfering with PI3K/AKT/PTEN signaling. Tacrolimus 78-81 AKT serine/threonine kinase 1 Homo sapiens 309-312 32555441-10 2021 Human kidney proximal tubular epithelial cells (HK-2 cells) were treated with TAC (50 mug/mL) in vitro, which induced production of intracellular reactive oxygen species and expression of an array of genes controlling programmed cell death (pyroptosis, apoptosis, and autophagy) through interfering with PI3K/AKT/PTEN signaling. Tacrolimus 78-81 phosphatase and tensin homolog Homo sapiens 313-317 32238515-11 2021 Urine protein:creatinine ratio, serum anti-dsDNA antibody levels, complement C3 levels, and steroid-sparing effect were all significantly improved from 4 weeks after tacrolimus treatment initiation (p<0.001), and were sustained over 5 years. Tacrolimus 166-176 complement C3 Homo sapiens 66-79 33790106-2 2021 However, the optimal dose of TAC in combination therapy with anti-TNFalpha antibodies (TAC + anti-TNFalpha therapy) remains unclear. Tacrolimus 29-32 tumor necrosis factor Mus musculus 98-106 33790106-8 2021 The nuclear expression of NFATc1 was inversely proportional to the administered doses of TAC. Tacrolimus 89-92 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 26-32 33790106-10 2021 The serum concentration of anti-TNFalpha antibodies in the high-dose TAC + anti-TNFalpha therapy was significantly higher than those in the other groups. Tacrolimus 69-72 tumor necrosis factor Mus musculus 32-40 33790106-11 2021 Low-dose TAC exerted its immunosuppressive effect on T-cells, and additionally, high-dose TAC maintained the serum anti-TNFalpha antibody concentration. Tacrolimus 90-93 tumor necrosis factor Mus musculus 120-128 33790106-12 2021 When administered in combination with anti-TNFalpha antibodies, the dose of TAC should be adjusted according to the disease severity. Tacrolimus 76-79 tumor necrosis factor Mus musculus 43-51 33618532-10 2021 RESULTS: In our study, serum MCP-1 levels were significantly higher in cyclosporine and tacrolimus groups than in sirolimus (p<0.05). Tacrolimus 88-98 C-C motif chemokine ligand 2 Homo sapiens 29-34 33631989-7 2021 FK506 clearly decreased the numbers of Th17 cells and FoxP3+IL-17+ cells. Tacrolimus 0-5 forkhead box P3 Homo sapiens 54-59 33631989-7 2021 FK506 clearly decreased the numbers of Th17 cells and FoxP3+IL-17+ cells. Tacrolimus 0-5 interleukin 17A Homo sapiens 60-65 33631989-8 2021 The proliferation capacity of cells was also inhibited by FK506, which had a greater effect on FoxP3- cells than FoxP3+ cells. Tacrolimus 58-63 forkhead box P3 Homo sapiens 95-100 33631989-8 2021 The proliferation capacity of cells was also inhibited by FK506, which had a greater effect on FoxP3- cells than FoxP3+ cells. Tacrolimus 58-63 forkhead box P3 Homo sapiens 113-118 33631989-10 2021 Our study provides that FK506 reduced the number of FoxP3low CD45RA- T cells (FrIII) by inhibiting its proliferation. Tacrolimus 24-29 forkhead box P3 Homo sapiens 52-57 33631989-10 2021 Our study provides that FK506 reduced the number of FoxP3low CD45RA- T cells (FrIII) by inhibiting its proliferation. Tacrolimus 24-29 protein tyrosine phosphatase receptor type C Homo sapiens 61-67 33220920-7 2021 Particularly, tacrolimus, an inhibitor of phosphatase calcineurin, strongly suppressed the CYP11B2 expression even at 10 nM. Tacrolimus 14-24 cytochrome P450 family 11 subfamily B member 2 Homo sapiens 91-98 33790106-0 2021 The Optimal Dose of Tacrolimus in Combination Therapy with an Anti-TNFalpha Antibody in a Mouse Colitis Model. Tacrolimus 20-30 tumor necrosis factor Mus musculus 67-75 32803289-7 2021 Subpopulation carrying CYP3A4*1G, CYP3A5*1, ABCB1-3435TT, or SLCO1B3-699AA was presented with enhanced increment in tacrolimus C0/D by 38.8-102.9%. Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 23-29 32803289-7 2021 Subpopulation carrying CYP3A4*1G, CYP3A5*1, ABCB1-3435TT, or SLCO1B3-699AA was presented with enhanced increment in tacrolimus C0/D by 38.8-102.9%. Tacrolimus 116-126 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 32803289-7 2021 Subpopulation carrying CYP3A4*1G, CYP3A5*1, ABCB1-3435TT, or SLCO1B3-699AA was presented with enhanced increment in tacrolimus C0/D by 38.8-102.9%. Tacrolimus 116-126 ATP binding cassette subfamily B member 1 Homo sapiens 44-49 32803289-7 2021 Subpopulation carrying CYP3A4*1G, CYP3A5*1, ABCB1-3435TT, or SLCO1B3-699AA was presented with enhanced increment in tacrolimus C0/D by 38.8-102.9%. Tacrolimus 116-126 solute carrier organic anion transporter family member 1B3 Homo sapiens 61-68 32803289-8 2021 CONCLUSION: Moderate effect of diltiazem on tacrolimus sparing, which might relate to the polymorphisms of CYP3A4, CYP3A5, ABCB1, and SLCO1B3, was documented. Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 107-113 32803289-8 2021 CONCLUSION: Moderate effect of diltiazem on tacrolimus sparing, which might relate to the polymorphisms of CYP3A4, CYP3A5, ABCB1, and SLCO1B3, was documented. Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 115-121 32803289-8 2021 CONCLUSION: Moderate effect of diltiazem on tacrolimus sparing, which might relate to the polymorphisms of CYP3A4, CYP3A5, ABCB1, and SLCO1B3, was documented. Tacrolimus 44-54 ATP binding cassette subfamily B member 1 Homo sapiens 123-128 32803289-8 2021 CONCLUSION: Moderate effect of diltiazem on tacrolimus sparing, which might relate to the polymorphisms of CYP3A4, CYP3A5, ABCB1, and SLCO1B3, was documented. Tacrolimus 44-54 solute carrier organic anion transporter family member 1B3 Homo sapiens 134-141 32768638-3 2020 However, WZ possesses a biphasic effect on regulating CYP3A (the major metabolizing enzyme of tacrolimus), which could induce the mRNA and protein expression after long-term treatment while transiently inhibit the activity of CYP3A. Tacrolimus 94-104 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 54-59 33368914-0 2021 Age and sex determine conversion from immediate-release to extended-release tacrolimus in a multi-center cohort of Canadian pediatric renal transplant recipients. Tacrolimus 76-86 renin binding protein Homo sapiens 0-3 33135259-1 2021 Post-Transplant Diabetes Mellitus (PTDM) shows a relationship with risk factors including obesity and tacrolimus-based immunosuppression, which decreases pancreatic insulin secretion. Tacrolimus 102-112 solute carrier family 35 member G1 Homo sapiens 0-4 33019979-11 2020 In addition, podocyte apoptosis induced by HDAC4 overexpression was effectively rescued by FK506, a pharmacological inhibitor of CaN, which was accompanied by decreased Bax and increased Bcl-2 expression. Tacrolimus 91-96 histone deacetylase 4 Homo sapiens 43-48 33019979-11 2020 In addition, podocyte apoptosis induced by HDAC4 overexpression was effectively rescued by FK506, a pharmacological inhibitor of CaN, which was accompanied by decreased Bax and increased Bcl-2 expression. Tacrolimus 91-96 BCL2 associated X, apoptosis regulator Homo sapiens 169-172 33019979-11 2020 In addition, podocyte apoptosis induced by HDAC4 overexpression was effectively rescued by FK506, a pharmacological inhibitor of CaN, which was accompanied by decreased Bax and increased Bcl-2 expression. Tacrolimus 91-96 BCL2 apoptosis regulator Homo sapiens 187-192 33363168-0 2020 FK506 Induces the TGF-beta1/Smad 3 Pathway Independently of Calcineurin Inhibition to Prevent Intervertebral Disk Degeneration. Tacrolimus 0-5 transforming growth factor, beta 1 Rattus norvegicus 18-27 33363168-0 2020 FK506 Induces the TGF-beta1/Smad 3 Pathway Independently of Calcineurin Inhibition to Prevent Intervertebral Disk Degeneration. Tacrolimus 0-5 SMAD family member 3 Rattus norvegicus 28-34 33363168-10 2020 FK506 could attenuate NP degeneration induced by IL-1beta. Tacrolimus 0-5 interleukin 1 alpha Rattus norvegicus 49-57 33363168-11 2020 Furthermore, FK506 exerted its function via TGFbeta/Smad3 activation instead of through calcineurin inhibition. Tacrolimus 13-18 transforming growth factor alpha Rattus norvegicus 44-51 33363168-11 2020 Furthermore, FK506 exerted its function via TGFbeta/Smad3 activation instead of through calcineurin inhibition. Tacrolimus 13-18 SMAD family member 3 Rattus norvegicus 52-57 33363168-12 2020 Inhibition of the TGF-beta pathway prevented the protective effect of FK506 on IVD degeneration. Tacrolimus 70-75 transforming growth factor alpha Rattus norvegicus 18-26 33363168-13 2020 In an in vivo study, FK506 injection reversed the development of rat caudal IVD degeneration influenced by Smad3. Tacrolimus 21-26 SMAD family member 3 Rattus norvegicus 107-112 33363168-14 2020 Conclusion: Our current study demonstrates the positive effect of FK506 on delaying the degeneration of IVD via the TGFbeta/Smad3 pathway. Tacrolimus 66-71 SMAD family member 3 Rattus norvegicus 124-129 32768638-3 2020 However, WZ possesses a biphasic effect on regulating CYP3A (the major metabolizing enzyme of tacrolimus), which could induce the mRNA and protein expression after long-term treatment while transiently inhibit the activity of CYP3A. Tacrolimus 94-104 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 226-231 32828804-0 2020 Proof that the high molecular weight immunophilin FKBP52 mediates the in vivo neuroregenerative effect of the macrolide FK506. Tacrolimus 120-125 FK506 binding protein 4 Mus musculus 37-56 33273604-4 2020 Administration of FK506, a calcineurin inhibitor, in the heterozygous fetuses improved bone mineral content (BMC) of the neonates, although it did not save the neonates from the lethal effects of the mutation, whereas rapamycin, an mTOR inhibitor, reduced BMC, suggesting that mTOR signaling is involved in the bone mineralization of heterozygous mutants. Tacrolimus 18-23 calcineurin binding protein 1 Mus musculus 27-48 33273604-4 2020 Administration of FK506, a calcineurin inhibitor, in the heterozygous fetuses improved bone mineral content (BMC) of the neonates, although it did not save the neonates from the lethal effects of the mutation, whereas rapamycin, an mTOR inhibitor, reduced BMC, suggesting that mTOR signaling is involved in the bone mineralization of heterozygous mutants. Tacrolimus 18-23 mechanistic target of rapamycin kinase Mus musculus 232-236 33273604-4 2020 Administration of FK506, a calcineurin inhibitor, in the heterozygous fetuses improved bone mineral content (BMC) of the neonates, although it did not save the neonates from the lethal effects of the mutation, whereas rapamycin, an mTOR inhibitor, reduced BMC, suggesting that mTOR signaling is involved in the bone mineralization of heterozygous mutants. Tacrolimus 18-23 mechanistic target of rapamycin kinase Mus musculus 277-281 32956634-2 2020 Among the seven mTOR inhibitors evaluated, tacrolimus (TAC) showed significant dose- and time-dependent killing of cultured protoscoleces and cysts in vitro. Tacrolimus 43-53 mechanistic target of rapamycin kinase Homo sapiens 16-20 32956634-2 2020 Among the seven mTOR inhibitors evaluated, tacrolimus (TAC) showed significant dose- and time-dependent killing of cultured protoscoleces and cysts in vitro. Tacrolimus 55-58 mechanistic target of rapamycin kinase Homo sapiens 16-20 32956634-6 2020 We further observed that the ATP levels and glucose content of cysts reduced upon TAC treatment, indicating that inhibiting mTORC1 activity possibly affects glucose metabolism in the cysts of mice. Tacrolimus 82-85 CREB regulated transcription coactivator 1 Mus musculus 124-130 33278239-0 2021 High Intra-Patient Variability in Tacrolimus Exposure Calculated over a Long Period Is Associated with De Novo Donor-Specific Antibody Development and/or Late Rejection in Thai Kidney Transplant Patients Receiving Concomitant CYP3A4/5 Inhibitors. Tacrolimus 34-44 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 226-234 32828804-6 2020 The experimental evidence demonstrates that the neurotrophic actions of FK506 are the consequence of its binding to FKBP52, whereas FK506 interaction with the close-related partner immunophilin FKBP51 antagonises the function of FKBP52. Tacrolimus 72-77 FK506 binding protein 4 Mus musculus 116-122 32828804-6 2020 The experimental evidence demonstrates that the neurotrophic actions of FK506 are the consequence of its binding to FKBP52, whereas FK506 interaction with the close-related partner immunophilin FKBP51 antagonises the function of FKBP52. Tacrolimus 132-137 FK506 binding protein 4 Mus musculus 229-235 33202516-12 2020 Conclusion: In tacrolimus-based immunosuppressive drugs regimen, impaired insulin secretory function for reduced insulin sensitivity contributed to the development of PTDM than insulin resistance during 1 year after transplantation. Tacrolimus 15-25 insulin Homo sapiens 74-81 33202516-12 2020 Conclusion: In tacrolimus-based immunosuppressive drugs regimen, impaired insulin secretory function for reduced insulin sensitivity contributed to the development of PTDM than insulin resistance during 1 year after transplantation. Tacrolimus 15-25 insulin Homo sapiens 113-120 33125703-0 2020 A twist in the ABC: Regulation of ABC transporter trafficking and transport by FK506-binding proteins. Tacrolimus 79-84 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 34-49 32712713-11 2020 CONCLUSIONS: Flucloxacillin decreases tacrolimus trough levels, possibly through a CYP3A4 and/or P-gp-inducing effect. Tacrolimus 38-48 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-89 32712713-11 2020 CONCLUSIONS: Flucloxacillin decreases tacrolimus trough levels, possibly through a CYP3A4 and/or P-gp-inducing effect. Tacrolimus 38-48 ATP binding cassette subfamily B member 1 Homo sapiens 97-101 33125703-2 2020 Yet, recent accumulating evidence implicates FK506-binding proteins (FKBPs), a type of peptidylprolyl cis-trans isomerase (PPIase) proteins, in ABC transporter regulation. Tacrolimus 45-50 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 144-159 33322998-0 2020 Effects of tacrolimus on autophagy protein LC3 in puromycin-damaged mouse podocytes. Tacrolimus 11-21 microtubule-associated protein 1 light chain 3 alpha Mus musculus 43-46 33039969-3 2020 The aim of the study was to examine the correlation between tacrolimus (TAC) dose and genetic variation for interleukin-10 (IL-10) and its effect on the therapeutic outcome. Tacrolimus 60-70 interleukin 10 Homo sapiens 108-122 33039969-3 2020 The aim of the study was to examine the correlation between tacrolimus (TAC) dose and genetic variation for interleukin-10 (IL-10) and its effect on the therapeutic outcome. Tacrolimus 60-70 interleukin 10 Homo sapiens 124-129 33039969-3 2020 The aim of the study was to examine the correlation between tacrolimus (TAC) dose and genetic variation for interleukin-10 (IL-10) and its effect on the therapeutic outcome. Tacrolimus 72-75 interleukin 10 Homo sapiens 108-122 33039969-3 2020 The aim of the study was to examine the correlation between tacrolimus (TAC) dose and genetic variation for interleukin-10 (IL-10) and its effect on the therapeutic outcome. Tacrolimus 72-75 interleukin 10 Homo sapiens 124-129 33039969-8 2020 The analysis of the correlation between TAC dose and IL-10 genetic variation for the -1082A > G polymorphism revealed that patients with the AA genotype required lower immunosuppressive drug doses (AA: 3.54 +- 2.38 mg/day vs AG: 6.18 +- 5.10 mg/day, GG: 4.44 +- 3.01 mg/day). Tacrolimus 40-43 interleukin 10 Homo sapiens 53-58 33039969-10 2020 The results of the study indicated that the IL-10 -1082A > G polymorphism may in fact influence the TAC dose. Tacrolimus 100-103 interleukin 10 Homo sapiens 44-49 32180132-7 2020 IFN-gamma potently enhanced TNFA, IL12, HLADRA1 and LRRK2 expression, which was suppressed by FK506, a calcineurin-specific inhibitor, but further enhanced by LRRK2-specific kinase inhibitor (GSK2578215A). Tacrolimus 94-99 interferon gamma Homo sapiens 0-9 33322998-6 2020 The low level of LC3 mRNA observed in untreated podocytes was decreased by puromycin treatment; however, levels of LC3 mRNA were higher in the FK506 group versus PAN group. Tacrolimus 143-148 microtubule-associated protein 1 light chain 3 alpha Mus musculus 17-20 32180132-7 2020 IFN-gamma potently enhanced TNFA, IL12, HLADRA1 and LRRK2 expression, which was suppressed by FK506, a calcineurin-specific inhibitor, but further enhanced by LRRK2-specific kinase inhibitor (GSK2578215A). Tacrolimus 94-99 tumor necrosis factor Homo sapiens 28-32 32180132-7 2020 IFN-gamma potently enhanced TNFA, IL12, HLADRA1 and LRRK2 expression, which was suppressed by FK506, a calcineurin-specific inhibitor, but further enhanced by LRRK2-specific kinase inhibitor (GSK2578215A). Tacrolimus 94-99 major histocompatibility complex, class II, DR alpha Homo sapiens 40-47 32180132-7 2020 IFN-gamma potently enhanced TNFA, IL12, HLADRA1 and LRRK2 expression, which was suppressed by FK506, a calcineurin-specific inhibitor, but further enhanced by LRRK2-specific kinase inhibitor (GSK2578215A). Tacrolimus 94-99 leucine rich repeat kinase 2 Homo sapiens 52-57 32180132-9 2020 CD14+ monocytes from G2019S and R1441C LRRK2 mutated PD cases and carriers show no changes in IFN-gamma responses for TNFA or IL12, reduced response for HLADRA1, and enhanced responses for LRRK2 in FK506-sensitive manner. Tacrolimus 198-203 CD14 molecule Homo sapiens 0-4 33322998-6 2020 The low level of LC3 mRNA observed in untreated podocytes was decreased by puromycin treatment; however, levels of LC3 mRNA were higher in the FK506 group versus PAN group. Tacrolimus 143-148 microtubule-associated protein 1 light chain 3 alpha Mus musculus 115-118 33322998-9 2020 Cytoplasmic LC3-related fluorescence intensity was stronger in control and FK506 podocytes versus the PAN group. Tacrolimus 75-80 microtubule-associated protein 1 light chain 3 alpha Mus musculus 12-15 33322998-10 2020 CONCLUSIONS: Tacrolimus inhibited puromycin-induced mouse podocyte damage by regulating LC3 expression and enhancing autophagy. Tacrolimus 13-23 microtubule-associated protein 1 light chain 3 alpha Mus musculus 88-91 32393842-5 2020 Among patients receiving tacrolimus/sirolimus, nivolumab-exposed patients had a higher incidence of severe aGVHD and a more effector T-cell profile compared with anti-PD-1-naive patients. Tacrolimus 25-35 programmed cell death 1 Homo sapiens 167-171 32783100-8 2020 Studies related variability in tacrolimus whole blood clearance among transplant recipients to either cytochrome P450 (CYP) 3A5 genotype (41%), days post-transplant (30%), or hematocrit (29%). Tacrolimus 31-41 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 102-127 32600641-11 2020 CONCLUSIONS: Our findings suggest that EVR-based regimens with tacrolimus and corticosteroid therapy for de novo KT reduce the recurrence of IgAN compared with MMF-based regimens with tacrolimus and corticosteroid therapy. Tacrolimus 63-73 IGAN1 Homo sapiens 141-145 33312896-2 2020 In order to expand successful renal transplant care to children and adolescents at the lowest possible cost, our pediatric renal transplant clinic uses a post-transplant tacrolimus-sparing strategy via inhibition of CYP3A4. Tacrolimus 170-180 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 216-222 33207690-1 2020 The protective effects of alpha-1 antitrypsin (AAT) in tacrolimus (TAC)-induced renal injury was evaluated in a rat model. Tacrolimus 55-65 serpin family A member 1 Rattus norvegicus 47-50 33207690-1 2020 The protective effects of alpha-1 antitrypsin (AAT) in tacrolimus (TAC)-induced renal injury was evaluated in a rat model. Tacrolimus 67-70 serpin family A member 1 Rattus norvegicus 47-50 33207690-3 2020 The TAC with AAT group was cotreated with daily subcutaneous injections of TAC and intraperitoneal injections of AAT (80 mg/kg) for four weeks. Tacrolimus 4-7 serpin family A member 1 Rattus norvegicus 13-16 33207690-3 2020 The TAC with AAT group was cotreated with daily subcutaneous injections of TAC and intraperitoneal injections of AAT (80 mg/kg) for four weeks. Tacrolimus 4-7 serpin family A member 1 Rattus norvegicus 113-116 33207690-4 2020 The effects of AAT on TAC-induced renal injury were evaluated using serum biochemistry, histopathology, and Western blotting. Tacrolimus 22-25 serpin family A member 1 Rattus norvegicus 15-18 33207690-7 2020 Immunohistochemical staining for inflammation (osteopontin and ED-1 staining) revealed improved interstitial inflammation in the TAC with AAT group compared to that in the TAC group. Tacrolimus 129-132 serpin family A member 1 Rattus norvegicus 138-141 33207690-8 2020 The TAC treatment increased renal apoptosis compared to the control treatment, based on the results of increased immunohistochemical staining of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL), increased caspase-3 activity, and lower Bcl-2 to Bad expression ratio. Tacrolimus 4-7 caspase 3 Rattus norvegicus 234-243 33207690-8 2020 The TAC treatment increased renal apoptosis compared to the control treatment, based on the results of increased immunohistochemical staining of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL), increased caspase-3 activity, and lower Bcl-2 to Bad expression ratio. Tacrolimus 4-7 BCL2, apoptosis regulator Rattus norvegicus 264-269 33207690-10 2020 AAT protects against TAC-induced renal injury via antifibrotic, anti-inflammatory, and antiapoptotic effects. Tacrolimus 21-24 serpin family A member 1 Rattus norvegicus 0-3 32453653-0 2020 Effects of CYP3A5, ABCB1 and POR*28 polymorphisms on pharmacokinetics of tacrolimus in the early period after renal transplantation. Tacrolimus 73-83 cytochrome p450 oxidoreductase Homo sapiens 29-32 32453653-2 2020 We aimed to establish a population pharmacokinetic (PK) model of tacrolimus and identify clinical covariates, especially the genetic polymorphisms of CYP3A5, ABCB1 and POR*28 that affected the PK to prevent fluctuation in the trough concentration of tacrolimus during the early period after renal transplantation. Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 150-156 32453653-2 2020 We aimed to establish a population pharmacokinetic (PK) model of tacrolimus and identify clinical covariates, especially the genetic polymorphisms of CYP3A5, ABCB1 and POR*28 that affected the PK to prevent fluctuation in the trough concentration of tacrolimus during the early period after renal transplantation. Tacrolimus 65-75 ATP binding cassette subfamily B member 1 Homo sapiens 158-163 32453653-2 2020 We aimed to establish a population pharmacokinetic (PK) model of tacrolimus and identify clinical covariates, especially the genetic polymorphisms of CYP3A5, ABCB1 and POR*28 that affected the PK to prevent fluctuation in the trough concentration of tacrolimus during the early period after renal transplantation. Tacrolimus 65-75 cytochrome p450 oxidoreductase Homo sapiens 168-171 32453653-2 2020 We aimed to establish a population pharmacokinetic (PK) model of tacrolimus and identify clinical covariates, especially the genetic polymorphisms of CYP3A5, ABCB1 and POR*28 that affected the PK to prevent fluctuation in the trough concentration of tacrolimus during the early period after renal transplantation. Tacrolimus 250-260 cytochrome p450 oxidoreductase Homo sapiens 168-171 32453653-14 2020 CYP3A5 genotype, post-operative day and hematocrit were confirmed as critical PK factors of tacrolimus. Tacrolimus 92-102 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 33188441-5 2020 A tacrolimus trough > 2.5 ng/mL was recently shown to be associated with a lower risk of PEP in liver transplant patients undergoing ERCP. Tacrolimus 2-12 prolyl endopeptidase Homo sapiens 89-92 33188441-9 2020 Tacrolimus is a promising potential agent to prevent PEP but needs further clinical study. Tacrolimus 0-10 prolyl endopeptidase Homo sapiens 53-56 33206751-8 2020 Moreover, the drug combination of mycophenolate mofetil and tacrolimus greatly reduced the expression levels of serum TGF-beta1 and cystatin C. Tacrolimus 60-70 transforming growth factor beta 1 Homo sapiens 118-127 33206751-8 2020 Moreover, the drug combination of mycophenolate mofetil and tacrolimus greatly reduced the expression levels of serum TGF-beta1 and cystatin C. Tacrolimus 60-70 cystatin C Homo sapiens 132-142 33000210-0 2020 Effects of tacrolimus on the TGF-beta1/SMAD signaling pathway in paraquat-exposed rat alveolar type II epithelial cells. Tacrolimus 11-21 transforming growth factor, beta 1 Rattus norvegicus 29-38 32738395-6 2020 Knock down of CYP2E1 mRNA using specific shRNA, FK506, a Calcineurin inhibitor, and Mdivi-1, a DRP1 inhibitor, ameliorated alcohol-induced mitochondrial retrograde signaling, and hepatic steatosis. Tacrolimus 48-53 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 14-20 32894758-0 2020 Differential effects of voclosporin and tacrolimus on insulin secretion from human islets. Tacrolimus 40-50 insulin Homo sapiens 54-61 32894758-7 2020 TAC, but not VCS, caused a significant impairment of 15 mM glucose-stimulated and 30 mM KCl-stimulated insulin secretion. Tacrolimus 0-3 insulin Homo sapiens 103-110 32894758-11 2020 Clinically relevant doses of TAC, but not VCS, directly inhibit insulin secretion from human islets, likely via transcriptional control of exocytosis machinery. Tacrolimus 29-32 insulin Homo sapiens 64-71 33275448-5 2020 Expression of miR-155-5p and miR-223-3p correlated with tacrolimus dose (p < 0.05), miR-223-3p with serum creatinine (p < 0.05), and miR-223-3p and miR-1228-3p with blood leukocytes (p < 0.05). Tacrolimus 56-66 microRNA 155 Homo sapiens 14-21 33000210-8 2020 The expression levels of TGF-beta1, SMAD3 and CTGF, as well as their concentrations in the culture supernatant, were significantly downregulated in the tacrolimus group compared with the paraquat group. Tacrolimus 152-162 transforming growth factor, beta 1 Rattus norvegicus 25-34 33000210-8 2020 The expression levels of TGF-beta1, SMAD3 and CTGF, as well as their concentrations in the culture supernatant, were significantly downregulated in the tacrolimus group compared with the paraquat group. Tacrolimus 152-162 SMAD family member 3 Rattus norvegicus 36-41 33000210-8 2020 The expression levels of TGF-beta1, SMAD3 and CTGF, as well as their concentrations in the culture supernatant, were significantly downregulated in the tacrolimus group compared with the paraquat group. Tacrolimus 152-162 cellular communication network factor 2 Rattus norvegicus 46-50 33149706-14 2020 FK506 use has significantly (P<0.01) increased biomarkers levels of cystatin C (325% and 477%), urea (177% and 245%), MDA (1253%), except calcineurin B1 that has decreased (97%). Tacrolimus 0-5 cystatin C Rattus norvegicus 68-78 33000210-9 2020 However, both the concentration and expression levels of SMAD7 were significantly upregulated in the tacrolimus group compared with the paraquat group. Tacrolimus 101-111 SMAD family member 7 Rattus norvegicus 57-62 33000210-10 2020 In conclusion, tacrolimus can reduce the levels of TGF-beta1, SMAD3 and CTGF, increase the level of SMAD7 in TGF-beta1 signaling pathway and protect the development of pulmonary fibrosis in paraquat exposed alveolar epithelial cells. Tacrolimus 15-25 transforming growth factor, beta 1 Rattus norvegicus 51-60 33000210-10 2020 In conclusion, tacrolimus can reduce the levels of TGF-beta1, SMAD3 and CTGF, increase the level of SMAD7 in TGF-beta1 signaling pathway and protect the development of pulmonary fibrosis in paraquat exposed alveolar epithelial cells. Tacrolimus 15-25 SMAD family member 3 Rattus norvegicus 62-67 33000210-10 2020 In conclusion, tacrolimus can reduce the levels of TGF-beta1, SMAD3 and CTGF, increase the level of SMAD7 in TGF-beta1 signaling pathway and protect the development of pulmonary fibrosis in paraquat exposed alveolar epithelial cells. Tacrolimus 15-25 cellular communication network factor 2 Rattus norvegicus 72-76 33000210-10 2020 In conclusion, tacrolimus can reduce the levels of TGF-beta1, SMAD3 and CTGF, increase the level of SMAD7 in TGF-beta1 signaling pathway and protect the development of pulmonary fibrosis in paraquat exposed alveolar epithelial cells. Tacrolimus 15-25 SMAD family member 7 Rattus norvegicus 100-105 33000210-10 2020 In conclusion, tacrolimus can reduce the levels of TGF-beta1, SMAD3 and CTGF, increase the level of SMAD7 in TGF-beta1 signaling pathway and protect the development of pulmonary fibrosis in paraquat exposed alveolar epithelial cells. Tacrolimus 15-25 transforming growth factor, beta 1 Rattus norvegicus 109-118 33350247-1 2020 The purpose of this study was to investigate the inhibitory effects of the main active components of Salviae Miltiorrhizae Radix et Rhizoma on the metabolism of tacrolimus mediated by CYP3 A4/5 enzyme, so as to predict the potential drug-drug interaction(DDI) in clinical use. Tacrolimus 161-171 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 184-193 33350247-5 2020 The results showed that dihydrotanshinone I had a strong inhibitory effect on the metabolism of tacrolimus in both HLM and rCYP3 A4/5 enzyme systems, and the inhibitory potential IC_(50) in HLM was 6.0 mumol L~(-1), while the other four active components of Salviae Miltiorrhizae Radix et Rhizoma exhibited relatively weak inhibition on CYP3 A4/5 activity with inhibition rate less than 30% at 10 mumol L~(-1). Tacrolimus 96-106 oxysterol binding protein 2 Homo sapiens 115-118 33350247-5 2020 The results showed that dihydrotanshinone I had a strong inhibitory effect on the metabolism of tacrolimus in both HLM and rCYP3 A4/5 enzyme systems, and the inhibitory potential IC_(50) in HLM was 6.0 mumol L~(-1), while the other four active components of Salviae Miltiorrhizae Radix et Rhizoma exhibited relatively weak inhibition on CYP3 A4/5 activity with inhibition rate less than 30% at 10 mumol L~(-1). Tacrolimus 96-106 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 124-133 32622856-2 2020 The original member of this family, FKBP12, is a well-known binding partner for the immunosuppressive drugs tacrolimus (FK506) and sirolimus (rapamycin). Tacrolimus 108-118 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 36-42 32622856-2 2020 The original member of this family, FKBP12, is a well-known binding partner for the immunosuppressive drugs tacrolimus (FK506) and sirolimus (rapamycin). Tacrolimus 120-125 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 36-42 32687845-6 2020 Then the role of calcineurin (CnA)-mediated Drp1 signaling pathway on KGN cells was confirmed by treating with Mdivi-1 or FK506T. Tacrolimus 122-127 utrophin Homo sapiens 44-48 33127654-5 2020 RESULTS: In two embodiments of our strategy, we fuse the chronically endocytosing domain of human folate receptor alpha to either a murine scFv that binds fluorescein or human FK506 binding protein that binds FK506, thereby creating a fusion receptor composed of largely human components. Tacrolimus 209-214 folate receptor alpha Homo sapiens 98-119 33067715-1 2020 AIM: The high doses of oral tacrolimus (TAC) (1,2) necessary to prevent acute rejection (AR) after vascularized composite allotransplantation (VCA) are associated with systemic adverse effects. Tacrolimus 28-38 ferredoxin reductase Rattus norvegicus 89-91 33067715-1 2020 AIM: The high doses of oral tacrolimus (TAC) (1,2) necessary to prevent acute rejection (AR) after vascularized composite allotransplantation (VCA) are associated with systemic adverse effects. Tacrolimus 40-43 ferredoxin reductase Rattus norvegicus 89-91 33067715-3 2020 However, the short-term use of topical TAC (Protopic ), as an off-label adjunct to oral TAC, to treat AR episodes pro re nata (PRN), has yielded inconsistent results. Tacrolimus 39-42 ferredoxin reductase Rattus norvegicus 102-104 32985896-9 2020 Results: Examination of the 15 SNPs and several clinical factors identified the CYP3A5 genotype (p = 5.6 x 10-11) and hemoglobin (p = 8.4 x 10-10) as the most significant determinants of tacrolimus C0/D. Tacrolimus 187-197 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 80-86 32985896-11 2020 Conclusion: A new classification and regression tree model was developed for establishing the starting dose of tacrolimus based on the CYP3A5 genotype and hemoglobin values. Tacrolimus 111-121 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 135-141 33195437-5 2020 Two small molecules FK506 and rapamycin bind to FKBP12 immunophilin and the resulting complexes (FK506-FKBP12 and rapamycin-FKBP12) target calcineurin and TOR, respectively in both humans and fungi. Tacrolimus 20-25 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 48-54 33195437-5 2020 Two small molecules FK506 and rapamycin bind to FKBP12 immunophilin and the resulting complexes (FK506-FKBP12 and rapamycin-FKBP12) target calcineurin and TOR, respectively in both humans and fungi. Tacrolimus 20-25 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 103-109 33195437-5 2020 Two small molecules FK506 and rapamycin bind to FKBP12 immunophilin and the resulting complexes (FK506-FKBP12 and rapamycin-FKBP12) target calcineurin and TOR, respectively in both humans and fungi. Tacrolimus 20-25 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 103-109 33195437-5 2020 Two small molecules FK506 and rapamycin bind to FKBP12 immunophilin and the resulting complexes (FK506-FKBP12 and rapamycin-FKBP12) target calcineurin and TOR, respectively in both humans and fungi. Tacrolimus 20-25 RAR related orphan receptor C Homo sapiens 155-158 33195437-5 2020 Two small molecules FK506 and rapamycin bind to FKBP12 immunophilin and the resulting complexes (FK506-FKBP12 and rapamycin-FKBP12) target calcineurin and TOR, respectively in both humans and fungi. Tacrolimus 97-102 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 48-54 33195437-5 2020 Two small molecules FK506 and rapamycin bind to FKBP12 immunophilin and the resulting complexes (FK506-FKBP12 and rapamycin-FKBP12) target calcineurin and TOR, respectively in both humans and fungi. Tacrolimus 97-102 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 103-109 33195437-5 2020 Two small molecules FK506 and rapamycin bind to FKBP12 immunophilin and the resulting complexes (FK506-FKBP12 and rapamycin-FKBP12) target calcineurin and TOR, respectively in both humans and fungi. Tacrolimus 97-102 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 103-109 33195437-5 2020 Two small molecules FK506 and rapamycin bind to FKBP12 immunophilin and the resulting complexes (FK506-FKBP12 and rapamycin-FKBP12) target calcineurin and TOR, respectively in both humans and fungi. Tacrolimus 97-102 RAR related orphan receptor C Homo sapiens 155-158 33195437-8 2020 The current review highlights FK506/rapamycin-FKBP12 interactions with calcineurin/TOR kinase in human and fungi, and development of non-immunosuppressive analogs of FK506, rapamycin, and novel small molecules in inhibition of fungal calcineurin and TOR kinase. Tacrolimus 30-35 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 46-52 33195437-8 2020 The current review highlights FK506/rapamycin-FKBP12 interactions with calcineurin/TOR kinase in human and fungi, and development of non-immunosuppressive analogs of FK506, rapamycin, and novel small molecules in inhibition of fungal calcineurin and TOR kinase. Tacrolimus 30-35 RAR related orphan receptor C Homo sapiens 83-86 33195437-8 2020 The current review highlights FK506/rapamycin-FKBP12 interactions with calcineurin/TOR kinase in human and fungi, and development of non-immunosuppressive analogs of FK506, rapamycin, and novel small molecules in inhibition of fungal calcineurin and TOR kinase. Tacrolimus 30-35 RAR related orphan receptor C Homo sapiens 250-253 32749395-4 2020 The LN distribution study showed that both DiR and FK506 were delivered into the LNs effectively via GM-mediated transport after 24 h and were present in the LNs for at least 48 h. The FK506-loaded GM (GM-FK506) significantly prolonged allograft survival compared with the PBS group (mean survival time, 17.8 +- 1.9 versus 7.3 +- 1.0 days; P < 0.01), and marked decreased the acute rejection grade. Tacrolimus 185-190 arginine vasopressin receptor 2 Homo sapiens 43-46 32749395-5 2020 Furthermore, T cell infiltration, and secretion of IL-2 and IFN-gamma were dramatically reduced in the GM-FK506 group. Tacrolimus 106-111 interleukin 2 Homo sapiens 51-55 32749395-5 2020 Furthermore, T cell infiltration, and secretion of IL-2 and IFN-gamma were dramatically reduced in the GM-FK506 group. Tacrolimus 106-111 interferon gamma Homo sapiens 60-69 32728937-8 2020 Meanwhile, tacrolimus-treated ITP mice displayed a significant decrease in the mRNA expression of T-bet and plasma level of IFN-gamma and IL-12p70 compared with ITP mice but without differences when compared with normal mice. Tacrolimus 11-21 T-box 21 Mus musculus 98-103 32728937-8 2020 Meanwhile, tacrolimus-treated ITP mice displayed a significant decrease in the mRNA expression of T-bet and plasma level of IFN-gamma and IL-12p70 compared with ITP mice but without differences when compared with normal mice. Tacrolimus 11-21 interferon gamma Mus musculus 124-133 32728937-9 2020 Furthermore, the expression of GATA3, Foxp3, and plasma level of IL-4 and TGF-beta were upregulated in tacrolimus-treated ITP mice without significant differences to normal mice (except TGF-beta). Tacrolimus 103-113 GATA binding protein 3 Mus musculus 31-36 32728937-9 2020 Furthermore, the expression of GATA3, Foxp3, and plasma level of IL-4 and TGF-beta were upregulated in tacrolimus-treated ITP mice without significant differences to normal mice (except TGF-beta). Tacrolimus 103-113 forkhead box P3 Mus musculus 38-43 32728937-9 2020 Furthermore, the expression of GATA3, Foxp3, and plasma level of IL-4 and TGF-beta were upregulated in tacrolimus-treated ITP mice without significant differences to normal mice (except TGF-beta). Tacrolimus 103-113 interleukin 4 Mus musculus 65-69 32728937-9 2020 Furthermore, the expression of GATA3, Foxp3, and plasma level of IL-4 and TGF-beta were upregulated in tacrolimus-treated ITP mice without significant differences to normal mice (except TGF-beta). Tacrolimus 103-113 transforming growth factor alpha Mus musculus 74-82 32728937-9 2020 Furthermore, the expression of GATA3, Foxp3, and plasma level of IL-4 and TGF-beta were upregulated in tacrolimus-treated ITP mice without significant differences to normal mice (except TGF-beta). Tacrolimus 103-113 transforming growth factor alpha Mus musculus 186-194 32888708-0 2020 Effect of ABCB1 3435C>T Genetic Polymorphism on Pharmacokinetic Variables of Tacrolimus in Adult Renal Transplant Recipients: a Systematic Review and Meta-analysis. Tacrolimus 77-87 ATP binding cassette subfamily B member 1 Homo sapiens 10-15 32888708-1 2020 PURPOSE: Tacrolimus is the substrate of multidrug-resistance 1 (ABCB1). Tacrolimus 9-19 ATP binding cassette subfamily B member 1 Homo sapiens 40-62 32888708-1 2020 PURPOSE: Tacrolimus is the substrate of multidrug-resistance 1 (ABCB1). Tacrolimus 9-19 ATP binding cassette subfamily B member 1 Homo sapiens 64-69 32888708-2 2020 However, the effect of ABCB1 C3435T polymorphism on pharmacokinetic variables of tacrolimus is controversial in different studies. Tacrolimus 81-91 ATP binding cassette subfamily B member 1 Homo sapiens 23-28 32888708-3 2020 This meta-analysis was conducted to explore the relationship between ABCB1 3435C>T genetic polymorphism and pharmacokinetic variables of tacrolimus. Tacrolimus 137-147 ATP binding cassette subfamily B member 1 Homo sapiens 69-74 32888708-6 2020 The study explored the relationship between ABCB1 3435C>T genetic polymorphism and pharmacokinetic variables of tacrolimus stratified according to time of posttransplantation, ethnicity, methods of concentration measurement, and the initial doses of tacrolimus. Tacrolimus 112-122 ATP binding cassette subfamily B member 1 Homo sapiens 44-49 32888708-9 2020 The subgroup analysis then revealed that the tacrolimus concentration/weight-adjusted daily dose ratio of ABCB1 3435T carriers was significantly higher than that of the ABCB1 3435CC group at 1 and 6 months. Tacrolimus 45-55 ATP binding cassette subfamily B member 1 Homo sapiens 106-111 32888708-10 2020 Meanwhile, ABCB1 3435CT and TT both had a higher tacrolimus concentration/weight-adjusted daily dose ratio compared with ABCB1 3435CC. Tacrolimus 49-59 ATP binding cassette subfamily B member 1 Homo sapiens 11-16 32888708-11 2020 IMPLICATIONS: Our meta-analysis identified that the ABCB1 3435C>T genetic polymorphism affected the pharmacokinetic variables of tacrolimus in adult renal transplant recipients. Tacrolimus 129-139 ATP binding cassette subfamily B member 1 Homo sapiens 52-57 32945359-10 2020 TAC inhibited HMC proliferation by affecting the Smad2 signaling pathway. Tacrolimus 0-3 SMAD family member 2 Homo sapiens 49-54 32945964-4 2020 Here, we characterized the function of two FK506-binding proteins, namely, FKBP15-1 and FKBP15-2, in Arabidopsis. Tacrolimus 43-48 FK506-binding protein 15 kD-1 Arabidopsis thaliana 75-83 32247597-1 2020 BACKGROUND: The aim of the study was to assess bioavailability aspects of tacrolimus formulations during conversion from twice-daily (TAC BID) to once-daily (TAC OD) formulation in 89 stable kidney transplant recipients. Tacrolimus 74-84 BH3 interacting domain death agonist Homo sapiens 138-141 32247597-9 2020 CONCLUSIONS: Conversion from TAC BID to TAC OD is associated with a significant increase in tacrolimus dose during the first 3 months. Tacrolimus 92-102 BH3 interacting domain death agonist Homo sapiens 33-36 32307148-1 2020 INTRODUCTION: After kidney transplantation (KTx) in patients with diagnosed cancers, calcineurin inhibitor tacrolimus (TAC) is replaced by sirolimus or everolimus (EV). Tacrolimus 107-117 calcineurin binding protein 1 Homo sapiens 85-106 32307148-1 2020 INTRODUCTION: After kidney transplantation (KTx) in patients with diagnosed cancers, calcineurin inhibitor tacrolimus (TAC) is replaced by sirolimus or everolimus (EV). Tacrolimus 119-122 calcineurin binding protein 1 Homo sapiens 85-106 33061827-9 2020 Tacrolimus therapy-induced methylation and overexpression of NFAT5 could significantly reduce the expression of G0S2 in AChR MG patients. Tacrolimus 0-10 G0/G1 switch 2 Homo sapiens 112-116 33061827-12 2020 Therefore, G0S2 could be an immune regulatory factor in both AChR MG occurrence and treatment with tacrolimus. Tacrolimus 99-109 G0/G1 switch 2 Homo sapiens 11-15 32971783-2 2020 The objective of this study was to estimate the frequency of CYP3A5 genetic polymorphisms and their relationship with tacrolimus requirements in the pediatric population. Tacrolimus 118-128 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-67 32992801-0 2020 Conjunctival Injection Reduction in Patients with Atopic Keratoconjunctivitis Due to Synergic Effect of Bovine Enteric-Coated Lactoferrin in 0.1% Tacrolimus Ophthalmic Suspension. Tacrolimus 146-156 lactotransferrin Bos taurus 126-137 32945964-4 2020 Here, we characterized the function of two FK506-binding proteins, namely, FKBP15-1 and FKBP15-2, in Arabidopsis. Tacrolimus 43-48 FK506- and rapamycin-binding protein 15 kD-2 Arabidopsis thaliana 88-96 32911703-1 2020 Cytochrome P450 (CYP) 3A5 polymorphism influences tacrolimus metabolism, but its effect on the drug pharmacokinetics in liver transplant recipients switched to once-daily extended-release formulation remains unknown. Tacrolimus 50-60 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-25 33193695-9 2020 In addition, for the first time, our results suggested several antiviral drugs, such as FK506, with molecular binding energies of -11.06 and -10.1 kcal/mol with ACE2 and the spike protein, respectively, could be potentially used to prevent SARS-CoV-2 and remains to further validation. Tacrolimus 88-93 angiotensin converting enzyme 2 Homo sapiens 161-165 33193695-9 2020 In addition, for the first time, our results suggested several antiviral drugs, such as FK506, with molecular binding energies of -11.06 and -10.1 kcal/mol with ACE2 and the spike protein, respectively, could be potentially used to prevent SARS-CoV-2 and remains to further validation. Tacrolimus 88-93 surface glycoprotein Severe acute respiratory syndrome coronavirus 2 174-179 32907953-6 2020 We also demonstrate that, in contrast to the case with Saccharomyces cerevisiae, NCS1 expression in C. neoformans is regulated by the calcineurin pathway via the transcription factor Crz1, as NCS1 expression is reduced by FK506 treatment and CRZ1 deletion. Tacrolimus 222-227 neuronal calcium sensor 1 Mus musculus 81-85 32907953-6 2020 We also demonstrate that, in contrast to the case with Saccharomyces cerevisiae, NCS1 expression in C. neoformans is regulated by the calcineurin pathway via the transcription factor Crz1, as NCS1 expression is reduced by FK506 treatment and CRZ1 deletion. Tacrolimus 222-227 neuronal calcium sensor 1 Mus musculus 192-196 32911703-0 2020 Effect of CYP3A5 on the Once-Daily Tacrolimus Conversion in Stable Liver Transplant Patients. Tacrolimus 35-45 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 32911703-2 2020 The aim of this study is to analyze the effect of CYP3A5 polymorphism on liver function after once-daily tacrolimus conversion in liver transplant patients. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 50-56 32911703-12 2020 CYP3A5 genotyping in liver transplant recipients leads to prediction of pharmacokinetics after switching from a twice-daily regimen to a once-daily dosage form, which makes it possible to establish an appropriate dose of tacrolimus. Tacrolimus 221-231 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 32243709-6 2020 The butyrate-GRP43-GLP-1 pathway in the intestine crypts may involve in the pathogenesis of normalization of hyperglycemia caused by the tacrolimus. Tacrolimus 137-147 glucagon Mus musculus 19-24 32243709-7 2020 Therefore, tacrolimus affects glucose metabolism through the butyrate-associated GLP-1 pathway in the gut, and oral supplementation with butyrate provides new insights for the prevention and treatment of tacrolimus-induced hyperglycemia in transplant recipients. Tacrolimus 11-21 glucagon Mus musculus 81-86 32983131-2 2020 Methods: To understand the biology of Tfr cells in kidney transplant patients treated with tacrolimus and mycophenolate mofetil (MMF) combination immunosuppression, we measured circulating (c)Tfh and cTfr cells in peripheral blood by flow cytometry in n = 211 kidney transplant recipients. Tacrolimus 91-101 transferrin receptor Homo sapiens 38-41 33145270-0 2020 Differential expression of genes related to calcineurin and mTOR signaling and regulatory miRNAs in peripheral blood from kidney recipients under tacrolimus-based therapy. Tacrolimus 146-156 mechanistic target of rapamycin kinase Homo sapiens 60-64 33145270-10 2020 Conclusions: The expression of PPP3CA, MTOR and miR-99a in the peripheral blood of renal recipients is influenced by tacrolimus-based therapy and by PPP3CA and MTOR variants. Tacrolimus 117-127 protein phosphatase 3 catalytic subunit alpha Homo sapiens 31-37 33145270-10 2020 Conclusions: The expression of PPP3CA, MTOR and miR-99a in the peripheral blood of renal recipients is influenced by tacrolimus-based therapy and by PPP3CA and MTOR variants. Tacrolimus 117-127 mechanistic target of rapamycin kinase Homo sapiens 39-43 33145270-10 2020 Conclusions: The expression of PPP3CA, MTOR and miR-99a in the peripheral blood of renal recipients is influenced by tacrolimus-based therapy and by PPP3CA and MTOR variants. Tacrolimus 117-127 microRNA 99a Homo sapiens 48-55 33145270-10 2020 Conclusions: The expression of PPP3CA, MTOR and miR-99a in the peripheral blood of renal recipients is influenced by tacrolimus-based therapy and by PPP3CA and MTOR variants. Tacrolimus 117-127 mechanistic target of rapamycin kinase Homo sapiens 160-164 32779844-0 2020 Tacrolimus ameliorates tubulointerstitial inflammation in diabetic nephropathy via inhibiting the NFATc1/TRPC6 pathway. Tacrolimus 0-10 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 98-104 32779844-0 2020 Tacrolimus ameliorates tubulointerstitial inflammation in diabetic nephropathy via inhibiting the NFATc1/TRPC6 pathway. Tacrolimus 0-10 transient receptor potential cation channel, subfamily C, member 6 Mus musculus 105-110 32779844-7 2020 The expression of NFATc1 and TRPC6 also increased in the kidneys of db/db mice and HK-2 cells with high glucose (HG), while TAC inhibited these effects. Tacrolimus 124-127 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 18-24 32779844-9 2020 Furthermore, HG-induced TRPC6 expression was inhibited by NFATc1 siRNA, while NFATc1 nuclear translocation was inhibited by TAC, but was restored by TRPC6 plasmid in HK-2 cells under HG conditions. Tacrolimus 124-127 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 78-84 32779844-10 2020 These findings suggest that TAC ameliorates tubulointerstitial inflammation in DN through NFATc1/TRPC6 feedback loop. Tacrolimus 28-31 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 90-96 32779844-10 2020 These findings suggest that TAC ameliorates tubulointerstitial inflammation in DN through NFATc1/TRPC6 feedback loop. Tacrolimus 28-31 transient receptor potential cation channel, subfamily C, member 6 Mus musculus 97-102 32552577-1 2020 INTRODUCTION: Although the association between CYP3A5 gene polymorphism and tacrolimus dosing requirements was well established, the impact on how CYP3A5 genotype affects the acute rejection and long-term renal function in patients who received kidney transplants and were treated with tacrolimus remained controversial. Tacrolimus 286-296 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 147-153 32552577-7 2020 Mean daily tacrolimus dose in the CYP3A5 expressers and nonexpressers was 0.08 (0.03) and 0.05 (0.02) mg/kg, respectively (P < .01). Tacrolimus 11-21 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 32933627-14 2020 CYP3A5 genotyping has a certain guiding significance for determining the dosage of tacrolimus. Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 32621968-7 2020 Hematocrit, postoperative days, tacrolimus daily dose, voriconazole concomitant therapy, and CYP3A5*3 genotype were identified as significant covariates for tacrolimus clearance. Tacrolimus 157-167 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 93-99 32563057-6 2020 Systemic MDSC plus FK-506 therapy was found to have a significant anti-psoriatic effect in the murine model, to reduce levels of pro-inflammatory cytokines Th1 cytokines (TNF-alpha and IFN-gamma) and Th17 cytokines (IL-17A and IL-23) in serum and skin. Tacrolimus 19-25 tumor necrosis factor Mus musculus 171-180 32563057-6 2020 Systemic MDSC plus FK-506 therapy was found to have a significant anti-psoriatic effect in the murine model, to reduce levels of pro-inflammatory cytokines Th1 cytokines (TNF-alpha and IFN-gamma) and Th17 cytokines (IL-17A and IL-23) in serum and skin. Tacrolimus 19-25 interferon gamma Mus musculus 185-194 32563057-6 2020 Systemic MDSC plus FK-506 therapy was found to have a significant anti-psoriatic effect in the murine model, to reduce levels of pro-inflammatory cytokines Th1 cytokines (TNF-alpha and IFN-gamma) and Th17 cytokines (IL-17A and IL-23) in serum and skin. Tacrolimus 19-25 interleukin 17A Mus musculus 216-222 32563057-6 2020 Systemic MDSC plus FK-506 therapy was found to have a significant anti-psoriatic effect in the murine model, to reduce levels of pro-inflammatory cytokines Th1 cytokines (TNF-alpha and IFN-gamma) and Th17 cytokines (IL-17A and IL-23) in serum and skin. Tacrolimus 19-25 interleukin 23, alpha subunit p19 Mus musculus 227-232 32878009-1 2020 FK506 Induces Ligand-Independent Activation of the Bone Morphogenetic Protein Pathway and Osteogenesis. Tacrolimus 0-5 bone morphogenetic protein 1 Homo sapiens 51-77 32848803-0 2020 CYP3A5 Gene-Guided Tacrolimus Treatment of Living-Donor Egyptian Kidney Transplanted Patients. Tacrolimus 19-29 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 32825653-7 2020 All SSD1 disruptants displayed susceptibility to the calcineurin inhibitor FK506, similar to fks1 . Tacrolimus 75-80 mRNA-binding translational repressor SSD1 Saccharomyces cerevisiae S288C 4-8 32849848-0 2020 Beyond Single Nucleotide Polymorphisms: CYP3A5*3*6*7 Composite and ABCB1 Haplotype Associations to Tacrolimus Pharmacokinetics in Black and White Renal Transplant Recipients. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 40-46 32848803-8 2020 Conclusion: This is the first study in Egypt contributing to the individualization of tacrolimus dosing in Egyptian patients, informed by the CYP3A5 genotype. Tacrolimus 86-96 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 142-148 32849848-0 2020 Beyond Single Nucleotide Polymorphisms: CYP3A5*3*6*7 Composite and ABCB1 Haplotype Associations to Tacrolimus Pharmacokinetics in Black and White Renal Transplant Recipients. Tacrolimus 99-109 ATP binding cassette subfamily B member 1 Homo sapiens 67-72 32848803-2 2020 Part of interindividual and interethnic differences in the response of patients to tacrolimus is attributed to polymorphisms at CYP3A5 metabolic enzyme. Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 128-134 32849848-1 2020 Interpatient variability in tacrolimus pharmacokinetics is attributed to metabolism by cytochrome P-450 3A5 (CYP3A5) isoenzymes and membrane transport by P-glycoprotein. Tacrolimus 28-38 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 87-107 32849848-1 2020 Interpatient variability in tacrolimus pharmacokinetics is attributed to metabolism by cytochrome P-450 3A5 (CYP3A5) isoenzymes and membrane transport by P-glycoprotein. Tacrolimus 28-38 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 109-115 32848803-3 2020 CYP3A5 gene expression status is associated with tacrolimus dose requirement in renal transplant recipients. Tacrolimus 49-59 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 32849848-1 2020 Interpatient variability in tacrolimus pharmacokinetics is attributed to metabolism by cytochrome P-450 3A5 (CYP3A5) isoenzymes and membrane transport by P-glycoprotein. Tacrolimus 28-38 ATP binding cassette subfamily B member 1 Homo sapiens 154-168 32848803-5 2020 Secondly, we evaluated the influence of the CYP3A5 gene variant on tacrolimus doses required for these patients as well on dose-adjusted tacrolimus trough-concentrations. Tacrolimus 67-77 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 44-50 32849848-3 2020 Tacrolimus pharmacokinetics was investigated in 65 stable Black and Caucasian post-renal transplant patients by assessing the effects of multiple alleles in both CYP3A5 and ABCB1. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 162-168 32849848-3 2020 Tacrolimus pharmacokinetics was investigated in 65 stable Black and Caucasian post-renal transplant patients by assessing the effects of multiple alleles in both CYP3A5 and ABCB1. Tacrolimus 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 173-178 32848803-7 2020 Additionally, our results showed that, mean tacrolimus daily requirements for heterozygous patients (CYP3A5*1/*3) were significantly higher compared to homozygous patients (CYP3A5*3/*3) during the first year after kidney transplantation. Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 101-107 32849848-6 2020 Finally, a combined analysis using both CYP3A5 and ABCB1 polymorphisms was developed to assess their inter-related influence on tacrolimus pharmacokinetics. Tacrolimus 128-138 ATP binding cassette subfamily B member 1 Homo sapiens 51-56 32821416-14 2020 Finally, we offer a simplified approach that can aid in distinguishing between a primary psychiatric diagnosis versus tacrolimus-associated psychosis. Tacrolimus 118-128 activation induced cytidine deaminase Homo sapiens 49-52 32849848-13 2020 The ABCB1 haplotype analysis detected significant associations of the wildtype 1236T-2677T-3435T haplotype to tacrolimus dose (P = 0.03), CL (P = 0.023), CL/LBW (P = 0.022), and AUC* (P = 0.078). Tacrolimus 110-120 ATP binding cassette subfamily B member 1 Homo sapiens 4-9 32849848-14 2020 Finally, analysis combining CYP3A5 and ABCB1 genotypes indicated that the presence of the ABCB1 3435 T allele significantly reduced tacrolimus clearance for all three CPY3A5 metabolic composite groups. Tacrolimus 132-142 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 28-34 32849848-14 2020 Finally, analysis combining CYP3A5 and ABCB1 genotypes indicated that the presence of the ABCB1 3435 T allele significantly reduced tacrolimus clearance for all three CPY3A5 metabolic composite groups. Tacrolimus 132-142 ATP binding cassette subfamily B member 1 Homo sapiens 39-44 32849848-14 2020 Finally, analysis combining CYP3A5 and ABCB1 genotypes indicated that the presence of the ABCB1 3435 T allele significantly reduced tacrolimus clearance for all three CPY3A5 metabolic composite groups. Tacrolimus 132-142 ATP binding cassette subfamily B member 1 Homo sapiens 90-95 32849848-15 2020 Genotypic associations of tacrolimus pharmacokinetics can be improved by using the novel composite CYP3A5*3*4*5 and ABCB1 haplotypes. Tacrolimus 26-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 99-105 32849848-15 2020 Genotypic associations of tacrolimus pharmacokinetics can be improved by using the novel composite CYP3A5*3*4*5 and ABCB1 haplotypes. Tacrolimus 26-36 ATP binding cassette subfamily B member 1 Homo sapiens 116-121 32849848-16 2020 Consideration of multiple alleles using CYP3A5 metabolic composites and drug transporter ABCB1 haplotypes provides a more comprehensive appraisal of genetic factors contributing to interpatient variability in tacrolimus pharmacokinetics among Whites and Blacks. Tacrolimus 209-219 ATP binding cassette subfamily B member 1 Homo sapiens 89-94 32817992-4 2020 Results Tacrolimus significantly increased the expression of LIF, IL-10, and IL-17 and decreased the expression of IL-4, IFN-gamma, and the IFN-gamma/IL-10 ratio in RIF patients. Tacrolimus 8-18 LIF interleukin 6 family cytokine Homo sapiens 61-64 32817992-0 2020 Tacrolimus Improves the Implantation Rate in Patients with Elevated Th1/2 Helper Cell Ratio and Repeated Implantation Failure (RIF). Tacrolimus 0-10 negative elongation factor complex member C/D Homo sapiens 68-73 32170643-10 2020 CONCLUSIONS: For patients with the CYP3A5*3*3 allele, the required tacrolimus dose for 75% of subjects to achieve target trough concentrations of 4.8-15 ng/mL was 2 mg every 12 h (q12h). Tacrolimus 67-77 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 35-41 32170643-11 2020 For patients with the CYP3A5*1*1 allele, the required dose was 2 mg tacrolimus q12h with a Wuzhi capsule, and for patients with the CYP3A5*1*3 allele, the required dose was 3 mg of tacrolimus q12h or 4 mg q24h co-administered with a Wuzhi capsule. Tacrolimus 68-78 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 22-28 32817992-4 2020 Results Tacrolimus significantly increased the expression of LIF, IL-10, and IL-17 and decreased the expression of IL-4, IFN-gamma, and the IFN-gamma/IL-10 ratio in RIF patients. Tacrolimus 8-18 interleukin 10 Homo sapiens 66-71 32817992-4 2020 Results Tacrolimus significantly increased the expression of LIF, IL-10, and IL-17 and decreased the expression of IL-4, IFN-gamma, and the IFN-gamma/IL-10 ratio in RIF patients. Tacrolimus 8-18 interleukin 17A Homo sapiens 77-82 32817992-4 2020 Results Tacrolimus significantly increased the expression of LIF, IL-10, and IL-17 and decreased the expression of IL-4, IFN-gamma, and the IFN-gamma/IL-10 ratio in RIF patients. Tacrolimus 8-18 interleukin 4 Homo sapiens 115-119 32817992-4 2020 Results Tacrolimus significantly increased the expression of LIF, IL-10, and IL-17 and decreased the expression of IL-4, IFN-gamma, and the IFN-gamma/IL-10 ratio in RIF patients. Tacrolimus 8-18 interferon gamma Homo sapiens 121-130 32817992-4 2020 Results Tacrolimus significantly increased the expression of LIF, IL-10, and IL-17 and decreased the expression of IL-4, IFN-gamma, and the IFN-gamma/IL-10 ratio in RIF patients. Tacrolimus 8-18 interferon gamma Homo sapiens 140-149 32817992-4 2020 Results Tacrolimus significantly increased the expression of LIF, IL-10, and IL-17 and decreased the expression of IL-4, IFN-gamma, and the IFN-gamma/IL-10 ratio in RIF patients. Tacrolimus 8-18 interleukin 10 Homo sapiens 150-155 32301802-7 2020 These data support that the antimelanoma effect of FK506 and FK520 is partially mediated by inhibiting the oncogenic factor NFAT3, suggesting that therapeutics based on NFAT3 inhibition may be effective in clinical melanoma treatment. Tacrolimus 51-56 nuclear factor of activated T cells 4 Homo sapiens 124-129 32301802-6 2020 Mechanistic studies revealed that FK506 or FK520 blocked the nuclear translocation and reduced the transcriptional activity of NFAT3. Tacrolimus 34-39 nuclear factor of activated T cells 4 Homo sapiens 127-132 32301802-7 2020 These data support that the antimelanoma effect of FK506 and FK520 is partially mediated by inhibiting the oncogenic factor NFAT3, suggesting that therapeutics based on NFAT3 inhibition may be effective in clinical melanoma treatment. Tacrolimus 51-56 nuclear factor of activated T cells 4 Homo sapiens 169-174 32848756-0 2020 The Clinical Impact of the C0/D Ratio and the CYP3A5 Genotype on Outcome in Tacrolimus Treated Kidney Transplant Recipients. Tacrolimus 76-86 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 32732848-9 2020 CONCLUSIONS: This single-center retrospective cohort analysis suggests that in hypersensitized kidney transplant recipients receiving tacrolimus-based immunosuppressive therapy similar clinical outcomes may be obtained using mTOR inhibitors compared to mycophenolate. Tacrolimus 134-144 mechanistic target of rapamycin kinase Homo sapiens 225-229 31902946-7 2020 Tacrolimus C0/D was higher in CYP3A5 nonexpressers than in CYP3A5 expressers (p = 0.003). Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 30-36 31902946-7 2020 Tacrolimus C0/D was higher in CYP3A5 nonexpressers than in CYP3A5 expressers (p = 0.003). Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 59-65 31902946-8 2020 ACTN4 rs62121818, MYH9 rs2239781, CYP3A5*3, and age explained 20.5% interindividual variability of tacrolimus concentration in the total cohort. Tacrolimus 99-109 actinin alpha 4 Homo sapiens 0-5 31902946-8 2020 ACTN4 rs62121818, MYH9 rs2239781, CYP3A5*3, and age explained 20.5% interindividual variability of tacrolimus concentration in the total cohort. Tacrolimus 99-109 myosin heavy chain 9 Homo sapiens 18-22 31902946-9 2020 In CYP3A5 nonexpressers, ACTN4 rs62121818 and MYH9 rs2239781 together explained 14.6% variation of tacrolimus C0/D. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 3-9 31902946-9 2020 In CYP3A5 nonexpressers, ACTN4 rs62121818 and MYH9 rs2239781 together explained 14.6% variation of tacrolimus C0/D. Tacrolimus 99-109 actinin alpha 4 Homo sapiens 25-30 31902946-9 2020 In CYP3A5 nonexpressers, ACTN4 rs62121818 and MYH9 rs2239781 together explained 14.6% variation of tacrolimus C0/D. Tacrolimus 99-109 myosin heavy chain 9 Homo sapiens 46-50 31902946-10 2020 MYH9 rs2239781, LAMB2 rs62119873 and age together explained 22.3% variability of tacrolimus level in CYP3A5 expressers. Tacrolimus 81-91 myosin heavy chain 9 Homo sapiens 0-4 31902946-10 2020 MYH9 rs2239781, LAMB2 rs62119873 and age together explained 22.3% variability of tacrolimus level in CYP3A5 expressers. Tacrolimus 81-91 laminin subunit beta 2 Homo sapiens 16-21 31902946-10 2020 MYH9 rs2239781, LAMB2 rs62119873 and age together explained 22.3% variability of tacrolimus level in CYP3A5 expressers. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 101-107 31902946-11 2020 CYP3A5*3 was still an important factor affecting tacrolimus concentration in patients with NS. Tacrolimus 49-59 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 31902946-12 2020 Podocyte-associated gene polymorphisms, especially ACTN4 rs62121818 and MYH9 rs2239781, were the other most important biomarkers for tacrolimus whole blood levels. Tacrolimus 133-143 actinin alpha 4 Homo sapiens 51-56 31902946-12 2020 Podocyte-associated gene polymorphisms, especially ACTN4 rs62121818 and MYH9 rs2239781, were the other most important biomarkers for tacrolimus whole blood levels. Tacrolimus 133-143 myosin heavy chain 9 Homo sapiens 72-76 31902946-13 2020 Genotyping of CYP3A5, ACTN4, and MYH9 polymorphisms may be helpful for better guiding tacrolimus dosing in pediatric patients with refractory NS. Tacrolimus 86-96 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 31902946-13 2020 Genotyping of CYP3A5, ACTN4, and MYH9 polymorphisms may be helpful for better guiding tacrolimus dosing in pediatric patients with refractory NS. Tacrolimus 86-96 myosin heavy chain 9 Homo sapiens 33-37 31902947-0 2020 CYP3A5 gene polymorphisms and their impact on dosage and trough concentration of tacrolimus among kidney transplant patients: a systematic review and meta-analysis. Tacrolimus 81-91 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 31902947-2 2020 Cytochrome P450 3A5 (CYP3A5) protein is involved in tacrolimus metabolism. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-19 31902947-2 2020 Cytochrome P450 3A5 (CYP3A5) protein is involved in tacrolimus metabolism. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 21-27 31902947-3 2020 Single nucleotide polymorphism in the CYP3A5 gene (6986A>G) results in alteration in metabolic activity of CYP3A5 protein which eventually affects the tacrolimus concentration. Tacrolimus 151-161 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 38-44 31902947-3 2020 Single nucleotide polymorphism in the CYP3A5 gene (6986A>G) results in alteration in metabolic activity of CYP3A5 protein which eventually affects the tacrolimus concentration. Tacrolimus 151-161 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 107-113 31902947-4 2020 Patients with CYP3A5 expresser genotypes (A/A *1/*1 and A/G *1/*3) metabolize tacrolimus more rapidly than CYP3A5 nonexpressers (G/G *3/*3). Tacrolimus 78-88 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 31902947-6 2020 Our results showed that the tacrolimus Co/D ratio is significantly lower in CYP3A5 expresser group as compared with nonexpresser in Asian as well as in European populations at any post-transplant period (p < 0.00001). Tacrolimus 28-38 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 76-82 32736525-0 2020 FK506 induces lung lymphatic endothelial cell senescence and downregulates LYVE-1 expression, with associated decreased hyaluronan uptake. Tacrolimus 0-5 lymphatic vessel endothelial hyaluronan receptor 1 Homo sapiens 75-81 32736525-8 2020 Matrigel tubulation assay were used to investigate the effects of FK506 on TNF-alpha-induced lymphangiogenesis. Tacrolimus 66-71 tumor necrosis factor Homo sapiens 75-84 32848756-1 2020 Tacrolimus is metabolized by CYP3A4 and CYP3A5 enzymes. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-35 32736525-10 2020 Flow cytometry was used to examine the effects of FK506 on LYVE-1 in precision-cut-lung-slices ex vivo and on hyaluronan uptake in vitro. Tacrolimus 50-55 lymphatic vessel endothelial hyaluronan receptor 1 Homo sapiens 59-65 32736525-11 2020 RESULTS: In vitro, FK506 downregulated telomerase reverse transcriptase expression, resulting in decreased telomerase activity and subsequent induction of p21 expression and cell senescence. Tacrolimus 19-24 H3 histone pseudogene 16 Homo sapiens 155-158 32736525-12 2020 Treatment with FK506 decreased LYVE-1 mRNA and protein levels and resulted in decreased LEC HA uptake. Tacrolimus 15-20 lymphatic vessel endothelial hyaluronan receptor 1 Homo sapiens 31-37 32848756-1 2020 Tacrolimus is metabolized by CYP3A4 and CYP3A5 enzymes. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 40-46 32736525-13 2020 Similar result showing reduction of LYVE-1 expression when treated with FK506 was observed ex vivo. Tacrolimus 72-77 lymphatic vessel endothelial hyaluronan receptor 1 Homo sapiens 36-42 32848756-6 2020 At first sight, the expression of CYP3A5 and a low C0/D ratio seem to be overlapping factors, both pointing towards patients in whom a higher tacrolimus dose is needed to reach the tacrolimus target concentration. Tacrolimus 142-152 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 32736525-15 2020 We showed that this NFAT binding site regulates LYVE-1 transcription, and mutation of this binding site blunted FK506-dependent downregulation of LYVE-1 promoter-dependent transcription. Tacrolimus 112-117 lymphatic vessel endothelial hyaluronan receptor 1 Homo sapiens 48-54 32736525-15 2020 We showed that this NFAT binding site regulates LYVE-1 transcription, and mutation of this binding site blunted FK506-dependent downregulation of LYVE-1 promoter-dependent transcription. Tacrolimus 112-117 lymphatic vessel endothelial hyaluronan receptor 1 Homo sapiens 146-152 32848756-6 2020 At first sight, the expression of CYP3A5 and a low C0/D ratio seem to be overlapping factors, both pointing towards patients in whom a higher tacrolimus dose is needed to reach the tacrolimus target concentration. Tacrolimus 181-191 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 32736525-16 2020 Finally, FK506-treated lymphatic endothelial cells show a blunted response to TNF-alpha-mediated lymphangiogenesis. Tacrolimus 9-14 tumor necrosis factor Homo sapiens 78-87 32848756-11 2020 Steroids induce the metabolism of tacrolimus via pregnane X receptor mediated increased CYP3A4 expression, resulting in lower tacrolimus C0/D ratio in high risk patients. Tacrolimus 34-44 nuclear receptor subfamily 1 group I member 2 Homo sapiens 49-68 32848756-11 2020 Steroids induce the metabolism of tacrolimus via pregnane X receptor mediated increased CYP3A4 expression, resulting in lower tacrolimus C0/D ratio in high risk patients. Tacrolimus 34-44 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-94 32848756-11 2020 Steroids induce the metabolism of tacrolimus via pregnane X receptor mediated increased CYP3A4 expression, resulting in lower tacrolimus C0/D ratio in high risk patients. Tacrolimus 126-136 nuclear receptor subfamily 1 group I member 2 Homo sapiens 49-68 32848756-11 2020 Steroids induce the metabolism of tacrolimus via pregnane X receptor mediated increased CYP3A4 expression, resulting in lower tacrolimus C0/D ratio in high risk patients. Tacrolimus 126-136 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-94 32690569-4 2020 Tacrolimus is metabolised by the cytochrome P (CYP) 450 3A enzyme system, and utilisation of CYP 3A inducers to accelerate its clearance may be used as a successful therapy to treat tacrolimus toxicity. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-99 32719413-2 2020 The aim of our study was to investigate the short- and long-term efficacy of anti-TNF agents [adalimumab (ADA) and infliximab (IFX)] and TAC in anti-TNF agent- and TAC-naive steroid-refractory UC patients. Tacrolimus 137-140 tumor necrosis factor Homo sapiens 149-152 32719413-2 2020 The aim of our study was to investigate the short- and long-term efficacy of anti-TNF agents [adalimumab (ADA) and infliximab (IFX)] and TAC in anti-TNF agent- and TAC-naive steroid-refractory UC patients. Tacrolimus 164-167 tumor necrosis factor Homo sapiens 82-85 32719413-5 2020 Logistic regression analysis showed the male sex and higher C-reactive protein to be independent factors for response to anti-TNF agents and TAC, respectively. Tacrolimus 141-144 C-reactive protein Homo sapiens 60-78 32690569-4 2020 Tacrolimus is metabolised by the cytochrome P (CYP) 450 3A enzyme system, and utilisation of CYP 3A inducers to accelerate its clearance may be used as a successful therapy to treat tacrolimus toxicity. Tacrolimus 182-192 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-99 32674627-0 2021 Association of the IL-6 Rs1800796 SNP with Concentration/dose Ratios of Tacrolimus and Donor Liver Function after Transplantation. Tacrolimus 72-82 interleukin 6 Homo sapiens 19-23 32674627-2 2021 Here, we aimed to assess the potential influence of the IL-6 single nucleotide polymorphism (SNP) rs1800796 on the concentration/dose (C/D) ratios of tacrolimus and donor liver function in Chinese liver transplant patients. Tacrolimus 150-160 interleukin 6 Homo sapiens 56-60 32674627-5 2021 The IL-6 rs1800796 polymorphism in recipients was found to be correlated with the C/D ratios of tacrolimus at months 2 to month 6 after transplantation. Tacrolimus 96-106 interleukin 6 Homo sapiens 4-8 32674627-7 2021 In conclusion, the IL-6 rs1800796 polymorphism was associated with the C/D ratios of tacrolimus and post-transplant donor liver function. Tacrolimus 85-95 interleukin 6 Homo sapiens 19-23 32664531-1 2020 Tacrolimus is a first-line calcineurin inhibitor (CNI) and an integral part of the immunosuppressive strategy in solid organ transplantation. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 27-48 32661607-8 2020 Pre-exposure of RBCs to FKBP12 followed by exposure to tacrolimus significantly decreased tacrolimus distribution in RBCs in a concentration-dependent manner. Tacrolimus 90-100 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 24-30 32661607-9 2020 In addition, preincubation of tacrolimus with FKBP12 significantly reduced the rate of tacrolimus distribution in RBCs. Tacrolimus 30-40 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 46-52 32661607-9 2020 In addition, preincubation of tacrolimus with FKBP12 significantly reduced the rate of tacrolimus distribution in RBCs. Tacrolimus 87-97 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 46-52 32422433-4 2020 Application of drugs that target either calcineurin (cyclosporine A) or FKBP12 (tacrolimus known as FK506 and sirolimus known as rapamycin) caused a decrease in TMEM16A activity. Tacrolimus 80-90 FKBP prolyl isomerase 1A Homo sapiens 72-78 32664235-0 2020 Tacrolimus Prevents TWEAK-Induced PLA2R Expression in Cultured Human Podocytes. Tacrolimus 0-10 TNF superfamily member 12 Homo sapiens 20-25 32664235-0 2020 Tacrolimus Prevents TWEAK-Induced PLA2R Expression in Cultured Human Podocytes. Tacrolimus 0-10 phospholipase A2 receptor 1 Homo sapiens 34-39 32664235-9 2020 Interestingly, IRF4 encodes the FK506-binding protein 52 (FKBP52), a protein associated with tacrolimus. Tacrolimus 93-103 interferon regulatory factor 4 Homo sapiens 15-19 32664235-9 2020 Interestingly, IRF4 encodes the FK506-binding protein 52 (FKBP52), a protein associated with tacrolimus. Tacrolimus 93-103 FKBP prolyl isomerase 4 Homo sapiens 32-56 32664235-9 2020 Interestingly, IRF4 encodes the FK506-binding protein 52 (FKBP52), a protein associated with tacrolimus. Tacrolimus 93-103 FKBP prolyl isomerase 4 Homo sapiens 58-64 32664235-10 2020 Tacrolimus prevented the increased expression of PLA2R, NFKB1 and IRF4 induced by TWEAK in cultured podocytes. Tacrolimus 0-10 phospholipase A2 receptor 1 Homo sapiens 49-54 32664235-10 2020 Tacrolimus prevented the increased expression of PLA2R, NFKB1 and IRF4 induced by TWEAK in cultured podocytes. Tacrolimus 0-10 nuclear factor kappa B subunit 1 Homo sapiens 56-61 32664235-10 2020 Tacrolimus prevented the increased expression of PLA2R, NFKB1 and IRF4 induced by TWEAK in cultured podocytes. Tacrolimus 0-10 interferon regulatory factor 4 Homo sapiens 66-70 32664235-10 2020 Tacrolimus prevented the increased expression of PLA2R, NFKB1 and IRF4 induced by TWEAK in cultured podocytes. Tacrolimus 0-10 TNF superfamily member 12 Homo sapiens 82-87 32664235-11 2020 In conclusion, TWEAK upregulates the expression of PLA2R and of other genes linked to membranous nephropathy in podocytes, and this is prevented by tacrolimus. Tacrolimus 148-158 TNF superfamily member 12 Homo sapiens 15-20 32664235-11 2020 In conclusion, TWEAK upregulates the expression of PLA2R and of other genes linked to membranous nephropathy in podocytes, and this is prevented by tacrolimus. Tacrolimus 148-158 phospholipase A2 receptor 1 Homo sapiens 51-56 32664235-12 2020 An impact of tacrolimus on the expression of PLA2R and other genes in podocytes may underlie its efficacy in treating the disease as well as the frequent recurrence of nephrotic syndrome upon tacrolimus withdrawal. Tacrolimus 13-23 phospholipase A2 receptor 1 Homo sapiens 45-50 32664235-12 2020 An impact of tacrolimus on the expression of PLA2R and other genes in podocytes may underlie its efficacy in treating the disease as well as the frequent recurrence of nephrotic syndrome upon tacrolimus withdrawal. Tacrolimus 192-202 phospholipase A2 receptor 1 Homo sapiens 45-50 32298692-8 2020 FK506, which also forms a complex with FKBP12 but does not target mTOR, reduced platelet procoagulant responses to a similar extent as rapamycin. Tacrolimus 0-5 FKBP prolyl isomerase 1A Homo sapiens 39-45 31888346-0 2020 Impact of Donor and Recipient CYP3A5*3 Genotype on Tacrolimus Population Pharmacokinetics in Chinese Adult Liver Transplant Recipients. Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 30-36 32422433-4 2020 Application of drugs that target either calcineurin (cyclosporine A) or FKBP12 (tacrolimus known as FK506 and sirolimus known as rapamycin) caused a decrease in TMEM16A activity. Tacrolimus 80-90 anoctamin 1 Homo sapiens 161-168 32422433-4 2020 Application of drugs that target either calcineurin (cyclosporine A) or FKBP12 (tacrolimus known as FK506 and sirolimus known as rapamycin) caused a decrease in TMEM16A activity. Tacrolimus 100-105 FKBP prolyl isomerase 1A Homo sapiens 72-78 32422433-4 2020 Application of drugs that target either calcineurin (cyclosporine A) or FKBP12 (tacrolimus known as FK506 and sirolimus known as rapamycin) caused a decrease in TMEM16A activity. Tacrolimus 100-105 anoctamin 1 Homo sapiens 161-168 32422433-5 2020 In addition, FK506 and BAPTA-AM prevented co-immunoprecipitation between FKBP12 and TMEM16A. Tacrolimus 13-18 FKBP prolyl isomerase 1A Homo sapiens 73-79 32422433-5 2020 In addition, FK506 and BAPTA-AM prevented co-immunoprecipitation between FKBP12 and TMEM16A. Tacrolimus 13-18 anoctamin 1 Homo sapiens 84-91 32422433-7 2020 Rapamycin decreased TMEM16A activity in cells pre-treated with cyclosporine A or FK506. Tacrolimus 81-86 anoctamin 1 Homo sapiens 20-27 32570960-0 2020 CYP3A5 Genotype as a Potential Pharmacodynamic Biomarker for Tacrolimus Therapy in Ulcerative Colitis in Japanese Patients. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 32580389-3 2020 This complex glucocorticoid regulation is mediated through the glucocorticoid receptor, also known as nuclear receptor subfamily 3 group C member 1 (NR3C1/GR) and related genes, like 11beta-hydroxysteroid dehydrogenases (HSD11Bs) and the FK506-binding immunophilins, FKBP5 and FKBP4. Tacrolimus 238-243 nuclear receptor subfamily 3 group C member 1 Homo sapiens 63-86 32580389-3 2020 This complex glucocorticoid regulation is mediated through the glucocorticoid receptor, also known as nuclear receptor subfamily 3 group C member 1 (NR3C1/GR) and related genes, like 11beta-hydroxysteroid dehydrogenases (HSD11Bs) and the FK506-binding immunophilins, FKBP5 and FKBP4. Tacrolimus 238-243 nuclear receptor subfamily 3 group C member 1 Homo sapiens 102-147 32580389-3 2020 This complex glucocorticoid regulation is mediated through the glucocorticoid receptor, also known as nuclear receptor subfamily 3 group C member 1 (NR3C1/GR) and related genes, like 11beta-hydroxysteroid dehydrogenases (HSD11Bs) and the FK506-binding immunophilins, FKBP5 and FKBP4. Tacrolimus 238-243 nuclear receptor subfamily 3 group C member 1 Homo sapiens 149-154 32580389-3 2020 This complex glucocorticoid regulation is mediated through the glucocorticoid receptor, also known as nuclear receptor subfamily 3 group C member 1 (NR3C1/GR) and related genes, like 11beta-hydroxysteroid dehydrogenases (HSD11Bs) and the FK506-binding immunophilins, FKBP5 and FKBP4. Tacrolimus 238-243 FKBP prolyl isomerase 5 Homo sapiens 267-272 32580389-3 2020 This complex glucocorticoid regulation is mediated through the glucocorticoid receptor, also known as nuclear receptor subfamily 3 group C member 1 (NR3C1/GR) and related genes, like 11beta-hydroxysteroid dehydrogenases (HSD11Bs) and the FK506-binding immunophilins, FKBP5 and FKBP4. Tacrolimus 238-243 FKBP prolyl isomerase 4 Homo sapiens 277-282 32603360-1 2020 Fpr1 (FK506-sensitive proline rotamase 1), a protein of the FKBP12 (FK506-binding protein 12 kDa) family in Saccharomyces cerevisiae, is a primary target for the immunosuppressive agents FK506 and rapamycin. Tacrolimus 6-11 peptidylprolyl isomerase FPR1 Saccharomyces cerevisiae S288C 0-4 32603360-2 2020 Fpr1 inhibits calcineurin and TORC1 (target of rapamycin complex 1) when bound to FK506 and rapamycin, respectively. Tacrolimus 82-87 peptidylprolyl isomerase FPR1 Saccharomyces cerevisiae S288C 0-4 32570960-8 2020 The relationship between CYP3A5 genotype and blood concentration, dose, and concentration/dose (C/D) ratio of tacrolimus in 15 subjects was studied. Tacrolimus 110-120 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 25-31 32570960-9 2020 The tacrolimus dose to maintain equivalent blood concentrations was lower in CYP3A5*3/*3 than in CYP3A5*1 carriers, and the C/D ratio was significantly higher in the latter. Tacrolimus 4-14 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 77-83 32570960-9 2020 The tacrolimus dose to maintain equivalent blood concentrations was lower in CYP3A5*3/*3 than in CYP3A5*1 carriers, and the C/D ratio was significantly higher in the latter. Tacrolimus 4-14 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 97-103 32570960-10 2020 Thus, CYP3A5 polymorphism information played a role in determining the initial dose of tacrolimus. Tacrolimus 87-97 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 6-12 32570960-11 2020 Furthermore, since the effect of tacrolimus appears earlier in CYP3A5*3/*3 than in CYP3A5*1/*1 and *1/*3, it seems necessary to change the evaluation time of therapeutic effect by CYP3A5 genotype. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 63-69 32570960-11 2020 Furthermore, since the effect of tacrolimus appears earlier in CYP3A5*3/*3 than in CYP3A5*1/*1 and *1/*3, it seems necessary to change the evaluation time of therapeutic effect by CYP3A5 genotype. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 83-89 32570960-11 2020 Furthermore, since the effect of tacrolimus appears earlier in CYP3A5*3/*3 than in CYP3A5*1/*1 and *1/*3, it seems necessary to change the evaluation time of therapeutic effect by CYP3A5 genotype. Tacrolimus 33-43 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 83-89 32570960-12 2020 Additionally, the relationship between CYP3A5 genotype and C/D ratio of tacrolimus in colonic mucosa was investigated in 10 subjects. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 39-45 32570960-13 2020 Tacrolimus concentration in the mucosa was two-fold higher in CYP3A5*3/*3 than in CYP3A5*1 carriers, although no significant difference in tacrolimus-blood levels was observed. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 62-68 32570960-13 2020 Tacrolimus concentration in the mucosa was two-fold higher in CYP3A5*3/*3 than in CYP3A5*1 carriers, although no significant difference in tacrolimus-blood levels was observed. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 82-88 32570960-14 2020 Therefore, the local concentration of tacrolimus affected by CYP3A5 polymorphism might be related to its therapeutic effect. Tacrolimus 38-48 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-67 32078103-1 2020 BACKGROUND AND OBJECTIVES: P-glycoprotein (P-gp) has been shown previously to contribute to the intestinal absorption of verapamil, diltiazem, tacrolimus, colchicine and indinavir in situ; however, its contribution in vivo is unknown. Tacrolimus 143-153 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 27-41 32578817-0 2020 Tacrolimus therapeutic efficacy in post-liver transplant patients with Cytochrome P450 3A5 (CYP3A5) genetic polymorphisms. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 71-90 32578817-0 2020 Tacrolimus therapeutic efficacy in post-liver transplant patients with Cytochrome P450 3A5 (CYP3A5) genetic polymorphisms. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 92-98 32578817-1 2020 Genetic polymorphisms of CYP3A5 have been pointed out as factors that influenciates tacrolimus immunosuppressive efficacy in post liver transplant patients. Tacrolimus 84-94 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 25-31 32578817-2 2020 This study aims to review the literature on the influence of cytochrome P450 3A5 (CYP3A5) genetic polymorphisms of tacrolimus in post-liver transplant patients. Tacrolimus 115-125 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-80 32578817-2 2020 This study aims to review the literature on the influence of cytochrome P450 3A5 (CYP3A5) genetic polymorphisms of tacrolimus in post-liver transplant patients. Tacrolimus 115-125 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 82-88 32578817-6 2020 Initially, the pharmacological aspects of tacrolimus were discussed, including details on its pharmacodynamics, pharmacokinetics and toxicity After that, we analyzed the studies that correlates CYP3A5 genetic polymorphisms and tacrolimus efficacy, including the ethnical specifications and the general limittions of the studies. Tacrolimus 42-52 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 194-200 32578817-7 2020 The CYP3A5 polymorphisms have pointed to alterations in the metabolism of tacrolimus according to the ethnic and populational genotype, specially the *1 and *3*3 alleles, reflecting in the need for dose adjustment and also in post liver transplant rejection. Tacrolimus 74-84 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 32060883-3 2020 Here, we aim to study the psoriasis treatment with TAC and siRNA for one of most cytokines expressed in psoriasis, the TNF-alpha. Tacrolimus 51-54 tumor necrosis factor Mus musculus 119-128 32078103-1 2020 BACKGROUND AND OBJECTIVES: P-glycoprotein (P-gp) has been shown previously to contribute to the intestinal absorption of verapamil, diltiazem, tacrolimus, colchicine and indinavir in situ; however, its contribution in vivo is unknown. Tacrolimus 143-153 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 43-47 32192767-0 2020 FKBP12 dimerization mutations effect FK506 binding and differentially alter calcineurin inhibition in the human pathogen Aspergillus fumigatus. Tacrolimus 37-42 FKBP prolyl isomerase 1A Homo sapiens 0-6 32305124-7 2020 Baseline GLP-1 levels in cyclosporine-treated renal transplant patients were higher than in both tacrolimus-treated renal transplant patients (p = 0,016) and control groups (p < 0,001). Tacrolimus 97-107 glucagon Homo sapiens 9-14 32305124-8 2020 GLP-1 levels at the 30th minute were higher in tacrolimus-treated renal transplant patients when compared to the cyclosporine-treated renal transplant patients (p = 0,024). Tacrolimus 47-57 glucagon Homo sapiens 0-5 32305124-9 2020 GLP-1 levels at the 120th minute were higher in tacrolimus-treated renal transplant patients than the control group (p = 0,024). Tacrolimus 48-58 glucagon Homo sapiens 0-5 32305124-10 2020 The areas under the curve of GLP-1 was higher in tacrolimus-treated renal transplant patients when compared to the control group (p = 0,018). Tacrolimus 49-59 glucagon Homo sapiens 29-34 32305124-12 2020 CONCLUSION: These findings showed a temporally affected incretin hormones in renal transplant patients, a preserved GLP-1 response to an oral glucose load in renal transplant patients on cyclosporine and increased GLP -1 response to an oral glucose load in those on tacrolimus. Tacrolimus 266-276 glucagon like peptide 1 receptor Homo sapiens 214-220 32450827-3 2020 RESULTS: Patients with the A allele of CYP3A5 treated with tacrolimus had a higher risk of acute rejection than those without the A allele, while patients carrying the homozygous GG variant for SXR A7635GG did not show any episode of acute rejection. Tacrolimus 59-69 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 39-45 32192767-1 2020 The 12-kDa FK506-binding protein (FKBP12) is the target of the commonly used immunosuppressive drug FK506. Tacrolimus 11-16 FKBP prolyl isomerase 1A Homo sapiens 34-40 32192767-2 2020 The FKBP12-FK506 complex binds to calcineurin and inhibits its activity, leading to immunosuppression and preventing organ transplant rejection. Tacrolimus 11-16 FKBP prolyl isomerase 1A Homo sapiens 4-10 32192767-8 2020 Molecular dynamics and pulling simulations for each dimeric FKBP12 protein revealed a two-fold increase in dimer strength and significantly higher number of contacts for the F37M, F37L, and W60V mutations, further confirming their varying degree of impact on FK506 binding and calcineurin inhibition in vivo. Tacrolimus 259-264 FKBP prolyl isomerase 1A Homo sapiens 60-66 32369692-8 2020 RESULTS: The enhanced pro-apoptotic and anti-proliferative properties of Tacrolimus versus mTOR inhibitors were associated with increased protein kinase RNA-like endoplasmic reticulum kinase (PERK)-related ER stress, Ser15P-p53/p53 ratio and p21 protein expression that may counterbalance the risk of proliferative upregulation caused by enhanced Thr172P-Cdk4/Cdk4 activation in liver cancer cells. Tacrolimus 73-83 eukaryotic translation initiation factor 2 alpha kinase 3 Homo sapiens 138-190 32499781-7 2020 Cyclosporine A, tacrolimus and steroids dose-dependently inhibited IFN-gamma secretion, and reactivity was further reduced when calcineurin inhibitors were combined with steroids. Tacrolimus 16-26 interferon gamma Homo sapiens 67-76 32369692-8 2020 RESULTS: The enhanced pro-apoptotic and anti-proliferative properties of Tacrolimus versus mTOR inhibitors were associated with increased protein kinase RNA-like endoplasmic reticulum kinase (PERK)-related ER stress, Ser15P-p53/p53 ratio and p21 protein expression that may counterbalance the risk of proliferative upregulation caused by enhanced Thr172P-Cdk4/Cdk4 activation in liver cancer cells. Tacrolimus 73-83 eukaryotic translation initiation factor 2 alpha kinase 3 Homo sapiens 192-196 32369692-8 2020 RESULTS: The enhanced pro-apoptotic and anti-proliferative properties of Tacrolimus versus mTOR inhibitors were associated with increased protein kinase RNA-like endoplasmic reticulum kinase (PERK)-related ER stress, Ser15P-p53/p53 ratio and p21 protein expression that may counterbalance the risk of proliferative upregulation caused by enhanced Thr172P-Cdk4/Cdk4 activation in liver cancer cells. Tacrolimus 73-83 tumor protein p53 Homo sapiens 224-227 32369692-8 2020 RESULTS: The enhanced pro-apoptotic and anti-proliferative properties of Tacrolimus versus mTOR inhibitors were associated with increased protein kinase RNA-like endoplasmic reticulum kinase (PERK)-related ER stress, Ser15P-p53/p53 ratio and p21 protein expression that may counterbalance the risk of proliferative upregulation caused by enhanced Thr172P-Cdk4/Cdk4 activation in liver cancer cells. Tacrolimus 73-83 tumor protein p53 Homo sapiens 228-231 32369692-8 2020 RESULTS: The enhanced pro-apoptotic and anti-proliferative properties of Tacrolimus versus mTOR inhibitors were associated with increased protein kinase RNA-like endoplasmic reticulum kinase (PERK)-related ER stress, Ser15P-p53/p53 ratio and p21 protein expression that may counterbalance the risk of proliferative upregulation caused by enhanced Thr172P-Cdk4/Cdk4 activation in liver cancer cells. Tacrolimus 73-83 H3 histone pseudogene 16 Homo sapiens 242-245 32369692-8 2020 RESULTS: The enhanced pro-apoptotic and anti-proliferative properties of Tacrolimus versus mTOR inhibitors were associated with increased protein kinase RNA-like endoplasmic reticulum kinase (PERK)-related ER stress, Ser15P-p53/p53 ratio and p21 protein expression that may counterbalance the risk of proliferative upregulation caused by enhanced Thr172P-Cdk4/Cdk4 activation in liver cancer cells. Tacrolimus 73-83 cyclin dependent kinase 4 Homo sapiens 355-359 32369692-8 2020 RESULTS: The enhanced pro-apoptotic and anti-proliferative properties of Tacrolimus versus mTOR inhibitors were associated with increased protein kinase RNA-like endoplasmic reticulum kinase (PERK)-related ER stress, Ser15P-p53/p53 ratio and p21 protein expression that may counterbalance the risk of proliferative upregulation caused by enhanced Thr172P-Cdk4/Cdk4 activation in liver cancer cells. Tacrolimus 73-83 cyclin dependent kinase 4 Homo sapiens 360-364 32269108-4 2020 Using an animal model of CIPS, we found that systemic administration of FK506 in male and female mice significantly increased the amount of alpha2delta-1-GluN1 complexes in the spinal cord and the level of alpha2delta-1-bound GluN1 proteins in spinal synaptosomes. Tacrolimus 72-77 glutamate receptor, ionotropic, NMDA1 (zeta 1) Mus musculus 154-159 32269108-4 2020 Using an animal model of CIPS, we found that systemic administration of FK506 in male and female mice significantly increased the amount of alpha2delta-1-GluN1 complexes in the spinal cord and the level of alpha2delta-1-bound GluN1 proteins in spinal synaptosomes. Tacrolimus 72-77 glutamate receptor, ionotropic, NMDA1 (zeta 1) Mus musculus 226-231 32369692-10 2020 Tacrolimus and mTOR inhibitors increased the protein expression of FKBP12 and FKBP51 that appeared to play pro-survival role. Tacrolimus 0-10 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 67-73 32269108-5 2020 Treatment with FK506 significantly increased the frequency of miniature excitatory postsynaptic currents (EPSCs) and the amplitudes of monosynaptic EPSCs evoked from the dorsal root and puff NMDAR currents in spinal dorsal horn neurons. Tacrolimus 15-20 glutamate receptor, ionotropic, NMDA1 (zeta 1) Mus musculus 191-196 32369692-12 2020 Tacrolimus and mTOR inhibitors decreased miR-92a-1-5p, miR-197-3p, miR-483-3p and miR-720, and increased miR-22-3p, miR-376a-3p, miR-663b, miR-886-5p, miR-1300 and miR-1303 expressions in HepG2 cells. Tacrolimus 0-10 microRNA 92a-1 Homo sapiens 41-50 32269108-6 2020 Inhibiting alpha2delta-1 with gabapentin or disrupting the alpha2delta-1-NMDAR interaction with alpha2delta-1Tat peptide completely reversed the effects of FK506. Tacrolimus 156-161 glutamate receptor, ionotropic, NMDA1 (zeta 1) Mus musculus 73-78 32269108-9 2020 In addition, genetically deleting GluN1 in primary sensory neurons or alpha2delta-1 similarly attenuated FK506-induced thermal and mechanical hypersensitivity. Tacrolimus 105-110 glutamate receptor, ionotropic, NMDA1 (zeta 1) Mus musculus 34-39 32369692-12 2020 Tacrolimus and mTOR inhibitors decreased miR-92a-1-5p, miR-197-3p, miR-483-3p and miR-720, and increased miR-22-3p, miR-376a-3p, miR-663b, miR-886-5p, miR-1300 and miR-1303 expressions in HepG2 cells. Tacrolimus 0-10 microRNA 197 Homo sapiens 55-62 32369692-12 2020 Tacrolimus and mTOR inhibitors decreased miR-92a-1-5p, miR-197-3p, miR-483-3p and miR-720, and increased miR-22-3p, miR-376a-3p, miR-663b, miR-886-5p, miR-1300 and miR-1303 expressions in HepG2 cells. Tacrolimus 0-10 microRNA 223 Homo sapiens 105-114 32369692-12 2020 Tacrolimus and mTOR inhibitors decreased miR-92a-1-5p, miR-197-3p, miR-483-3p and miR-720, and increased miR-22-3p, miR-376a-3p, miR-663b, miR-886-5p, miR-1300 and miR-1303 expressions in HepG2 cells. Tacrolimus 0-10 microRNA 663b Homo sapiens 129-137 32369692-12 2020 Tacrolimus and mTOR inhibitors decreased miR-92a-1-5p, miR-197-3p, miR-483-3p and miR-720, and increased miR-22-3p, miR-376a-3p, miR-663b, miR-886-5p, miR-1300 and miR-1303 expressions in HepG2 cells. Tacrolimus 0-10 vault RNA 2-1 Homo sapiens 139-146 32369692-12 2020 Tacrolimus and mTOR inhibitors decreased miR-92a-1-5p, miR-197-3p, miR-483-3p and miR-720, and increased miR-22-3p, miR-376a-3p, miR-663b, miR-886-5p, miR-1300 and miR-1303 expressions in HepG2 cells. Tacrolimus 0-10 microRNA 1303 Homo sapiens 164-172 32369692-13 2020 CONCLUSION: The more potent pro-apoptotic and anti-proliferative properties of Tacrolimus versus mTOR inhibitors were associated with an increased activation of PERK and p53 signaling, and p21 protein expression. Tacrolimus 79-89 eukaryotic translation initiation factor 2 alpha kinase 3 Homo sapiens 161-165 32369692-13 2020 CONCLUSION: The more potent pro-apoptotic and anti-proliferative properties of Tacrolimus versus mTOR inhibitors were associated with an increased activation of PERK and p53 signaling, and p21 protein expression. Tacrolimus 79-89 tumor protein p53 Homo sapiens 170-173 32369692-13 2020 CONCLUSION: The more potent pro-apoptotic and anti-proliferative properties of Tacrolimus versus mTOR inhibitors were associated with an increased activation of PERK and p53 signaling, and p21 protein expression. Tacrolimus 79-89 H3 histone pseudogene 16 Homo sapiens 189-192 32369692-14 2020 FKBP12 and FKBP51 appeared to be the most relevant partners of Tacrolimus and mTOR inhibitors exerting a pro-survival effect in HepG2 cells. Tacrolimus 63-73 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 0-6 31622734-3 2020 Tacrolimus and rectal indomethacin have each been reported to reduce risk of PEP. Tacrolimus 0-10 progestagen associated endometrial protein Homo sapiens 77-80 31654367-4 2020 METHODS: This was a multi-center, single-arm, prospective trial with a planned interim analysis after 16 patients, in which the tacrolimus starting dose was based on bodyweight, cytochrome P450 3A5 genotype, and donor status (living vs. deceased donor). Tacrolimus 128-138 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 178-197 31654367-9 2020 Cytochrome P450 3A5 genotype, hematocrit, and creatinine influenced the tacrolimus clearance. Tacrolimus 72-82 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-19 31622734-4 2020 We investigated the incidence of PEP in patients who have undergone ERCP after liver transplantation and the effectiveness of tacrolimus and/or indomethacin in reducing risk of PEP. Tacrolimus 126-136 progestagen associated endometrial protein Homo sapiens 177-180 31654367-12 2020 CONCLUSIONS: The weight-normalized starting dose of tacrolimus should be higher in patients with a lower bodyweight and in those who are cytochrome P450 3A5 expressers. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 137-156 31622734-10 2020 We calculated adjusted odds ratios (ORs) for the association between tacrolimus and indomethacin use and risk of PEP using mixed-effects multivariable logistic regression. Tacrolimus 69-79 progestagen associated endometrial protein Homo sapiens 113-116 31622734-13 2020 A trough level of tacrolimus above 2.5 ng/mL was associated with 79% lower odds of PEP (OR, 0.21; 95% CI, 0.06-0.72; P=.01). Tacrolimus 18-28 progestagen associated endometrial protein Homo sapiens 83-86 31622734-16 2020 In patients with trough levels of tacrolimus above 2.5 ng/mL, addition of indomethacin reduced the odds of PEP by 93% compared to patients who were unexposed to indomethacin. Tacrolimus 34-44 progestagen associated endometrial protein Homo sapiens 107-110 31622734-18 2020 CONCLUSIONS: In a retrospective study of patients who underwent ERCP for biliary complications after liver transplantation, we found trough levels of tacrolimus above 2.5 ng/mL to significantly reduce risk for PEP. Tacrolimus 150-160 progestagen associated endometrial protein Homo sapiens 210-213 31955224-6 2020 Based on the prediction, dual-luciferase reporter assay and miRNA transfection were used to discover the mechanism of how SNP rs15524 controls tacrolimus serum concentration through influencing CYP3A5 expression. Tacrolimus 143-153 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 194-200 31955224-7 2020 RESULTS: In this article, we found multiple SNPs on CYP3A4, CYP3A5, FKBP1A, NFATC2 genes were predicted closely related to tacrolimus serum concentration, therapeutic effect which reflected by QMG score changes or even reasonable drug dose. Tacrolimus 123-133 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 52-58 31955224-7 2020 RESULTS: In this article, we found multiple SNPs on CYP3A4, CYP3A5, FKBP1A, NFATC2 genes were predicted closely related to tacrolimus serum concentration, therapeutic effect which reflected by QMG score changes or even reasonable drug dose. Tacrolimus 123-133 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 60-66 31955224-7 2020 RESULTS: In this article, we found multiple SNPs on CYP3A4, CYP3A5, FKBP1A, NFATC2 genes were predicted closely related to tacrolimus serum concentration, therapeutic effect which reflected by QMG score changes or even reasonable drug dose. Tacrolimus 123-133 FKBP prolyl isomerase 1A Homo sapiens 68-74 31955224-7 2020 RESULTS: In this article, we found multiple SNPs on CYP3A4, CYP3A5, FKBP1A, NFATC2 genes were predicted closely related to tacrolimus serum concentration, therapeutic effect which reflected by QMG score changes or even reasonable drug dose. Tacrolimus 123-133 nuclear factor of activated T cells 2 Homo sapiens 76-82 32467610-11 2020 Finally, we found that FK506 reversed hypoxia-induced activation of the calcineurin/NFAT signaling pathway in NP cells and an ex vivo model. Tacrolimus 23-28 nuclear factor of activated T-cells 1 Rattus norvegicus 84-88 31530218-0 2020 Optimization of initial dosing scheme of tacrolimus in pediatric refractory nephrotic syndrome patients based on CYP3A5 genotype and coadministration with wuzhi-capsule. Tacrolimus 41-51 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 113-119 32151541-0 2020 Tacrolimus, a calcineurin inhibitor, promotes capsaicin-induced colonic pain in mice. Tacrolimus 0-10 calcineurin binding protein 1 Mus musculus 14-35 32151541-2 2020 Since the clinical use of calcineurin-inhibiting immunosuppressants is commonly associated with chronic diarrhea, we examined if tacrolimus, a calcineurin inhibitor, promotes TRPV1-dependent colonic hypersensitivity in mice. Tacrolimus 129-139 calcineurin binding protein 1 Mus musculus 143-164 32151541-2 2020 Since the clinical use of calcineurin-inhibiting immunosuppressants is commonly associated with chronic diarrhea, we examined if tacrolimus, a calcineurin inhibitor, promotes TRPV1-dependent colonic hypersensitivity in mice. Tacrolimus 129-139 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 175-180 32151541-6 2020 Thus, tacrolimus may aggravate TRPV1-related colonic pain accompanying irritable bowel syndrome. Tacrolimus 6-16 transient receptor potential cation channel, subfamily V, member 1 Mus musculus 31-36 31530218-10 2020 The present study recommended the tacrolimus initial dosing scheme in pediatric refractory nephrotic syndrome patients based on CYP3A5 genotype and coadministration with wuzhi-capsule. Tacrolimus 34-44 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 128-134 32213713-6 2020 We further demonstrated that immunotherapy with recombinant IL-22 treatment ameliorated the ST258 pulmonary infection in both FK506 treated WT mice and Rag2-/-Il2rg-/- mice via hepatic IL-22ra1 signaling. Tacrolimus 126-131 interleukin 22 Mus musculus 60-65 32425928-8 2020 Interestingly, when we compared the ability of Mo-MDSC to suppress T cell proliferation, we observed that tacrolimus, but not rapamycin-treated KTR, was able to inhibit CD4+ T cell proliferation in vitro. Tacrolimus 106-116 CD4 molecule Homo sapiens 169-172 32317681-5 2020 The ex vivo study showed that TAC-loaded NPs caused a significant suppression of the proliferation of CD4+ and CD8+ cells, which was comparable to the control formulation (Prograf). Tacrolimus 30-33 CD4 antigen Mus musculus 102-105 32340188-0 2020 Donor CYP3A5 Gene Polymorphism Alone Cannot Predict Tacrolimus Intrarenal Concentration in Renal Transplant Recipients. Tacrolimus 52-62 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 6-12 32340188-1 2020 CYP3A5 gene polymorphism in recipients plays an important role in tacrolimus blood pharmacokinetics after renal transplantation. Tacrolimus 66-76 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 32340188-3 2020 The aim of our study was to investigate how the tacrolimus intrarenal concentration (Ctissue) could be predicted based on donor CYP3A5 gene polymorphism in renal transplant recipients. Tacrolimus 48-58 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 128-134 32368128-0 2020 Effects of CYP3A5 Polymorphisms on Efficacy and Safety of Tacrolimus Therapy in Patients with Idiopathic Membranous Nephropathy. Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 11-17 32368128-4 2020 The aim of this study was to analyze the effects of CYP3A5 gene polymorphisms on the efficacy and safety of TAC in IMN patients. Tacrolimus 108-111 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 52-58 32368128-10 2020 The significant association between the CYP3A5 phenotype and TAC metabolism was observed. Tacrolimus 61-64 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 40-46 32368128-15 2020 Conclusion: Our results demonstrated that CYP3A5 polymorphisms had important guiding roles in the treatment of IMN with tacrolimus. Tacrolimus 120-130 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 42-48 32368128-16 2020 CYP3A5 expressers required higher daily doses of TAC to achieve the target drug concentration, but with fewer side effects. Tacrolimus 49-52 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 32363004-0 2020 Correction: Tacrolimus dose requirement based on the CYP3A5 genotype in renal transplant patients. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 53-59 31721244-0 2020 A novel random forest integrative approach based on endogenous CYP3A4 phenotype for predicting tacrolimus concentrations and dosages in Chinese renal transplant patients. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 63-69 32290462-11 2020 Finally, CD56bright and CD94/NK group 2 member A receptor positive (NKG2A+) Natural Killer (NK) cell subsets increased in mTORi- compared to tacrolimus-treated patients (both p < 0.001). Tacrolimus 141-151 killer cell lectin like receptor D1 Homo sapiens 24-28 32290462-11 2020 Finally, CD56bright and CD94/NK group 2 member A receptor positive (NKG2A+) Natural Killer (NK) cell subsets increased in mTORi- compared to tacrolimus-treated patients (both p < 0.001). Tacrolimus 141-151 killer cell lectin like receptor C1 Homo sapiens 68-73 32260456-3 2020 rs776746 (CYP3A5) and rs1137115 (CYP2A6) are single nucleotide polymorphisms (SNPs) that can affect exposure to tacrolimus. Tacrolimus 112-122 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 32260456-3 2020 rs776746 (CYP3A5) and rs1137115 (CYP2A6) are single nucleotide polymorphisms (SNPs) that can affect exposure to tacrolimus. Tacrolimus 112-122 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 33-39 31838663-0 2020 Tacrolimus Inhibits TNF-alpha/IL-17A-Produced pro-Inflammatory Effect on Human Keratinocytes by Regulating IkappaBzeta. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 20-29 31838663-0 2020 Tacrolimus Inhibits TNF-alpha/IL-17A-Produced pro-Inflammatory Effect on Human Keratinocytes by Regulating IkappaBzeta. Tacrolimus 0-10 interleukin 17A Homo sapiens 30-36 31838663-0 2020 Tacrolimus Inhibits TNF-alpha/IL-17A-Produced pro-Inflammatory Effect on Human Keratinocytes by Regulating IkappaBzeta. Tacrolimus 0-10 NFKB inhibitor zeta Homo sapiens 107-118 31838663-3 2020 This study aimed to investigate the potential regulatory effect of tacrolimus on TNF-alpha/ IL-17A-costimulated human keratinocytes in the mimic psoriatic microenvironment. Tacrolimus 67-77 tumor necrosis factor Homo sapiens 81-90 31838663-3 2020 This study aimed to investigate the potential regulatory effect of tacrolimus on TNF-alpha/ IL-17A-costimulated human keratinocytes in the mimic psoriatic microenvironment. Tacrolimus 67-77 interleukin 17A Homo sapiens 92-98 31838663-8 2020 Tacrolimus significantly inhibited TNF-alpha/IL-17A-induced IL-36gamma, CCL-20, IL-1beta, and S100-A9 expression at gene level and IL-36gamma and CCL-20 expression at protein level. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 35-44 31838663-8 2020 Tacrolimus significantly inhibited TNF-alpha/IL-17A-induced IL-36gamma, CCL-20, IL-1beta, and S100-A9 expression at gene level and IL-36gamma and CCL-20 expression at protein level. Tacrolimus 0-10 interleukin 17A Homo sapiens 45-51 31838663-8 2020 Tacrolimus significantly inhibited TNF-alpha/IL-17A-induced IL-36gamma, CCL-20, IL-1beta, and S100-A9 expression at gene level and IL-36gamma and CCL-20 expression at protein level. Tacrolimus 0-10 C-C motif chemokine ligand 20 Homo sapiens 72-78 31838663-8 2020 Tacrolimus significantly inhibited TNF-alpha/IL-17A-induced IL-36gamma, CCL-20, IL-1beta, and S100-A9 expression at gene level and IL-36gamma and CCL-20 expression at protein level. Tacrolimus 0-10 interleukin 1 beta Homo sapiens 80-88 31838663-8 2020 Tacrolimus significantly inhibited TNF-alpha/IL-17A-induced IL-36gamma, CCL-20, IL-1beta, and S100-A9 expression at gene level and IL-36gamma and CCL-20 expression at protein level. Tacrolimus 0-10 C-C motif chemokine ligand 20 Homo sapiens 146-152 31838663-9 2020 We further discovered TNF-alpha/IL-17A induced significant IkappaBzeta mRNA and protein expression in NHKs, which could be inhibited by tacrolimus. Tacrolimus 136-146 tumor necrosis factor Homo sapiens 22-31 31838663-9 2020 We further discovered TNF-alpha/IL-17A induced significant IkappaBzeta mRNA and protein expression in NHKs, which could be inhibited by tacrolimus. Tacrolimus 136-146 interleukin 17A Homo sapiens 32-38 31838663-9 2020 We further discovered TNF-alpha/IL-17A induced significant IkappaBzeta mRNA and protein expression in NHKs, which could be inhibited by tacrolimus. Tacrolimus 136-146 NFKB inhibitor zeta Homo sapiens 59-70 31838663-10 2020 Tacrolimus can inhibit pro-inflammatory synergistic action of TNF-alpha/IL-17A on human keratinocytes by regulating IkappaBzeta expression. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 62-71 31838663-10 2020 Tacrolimus can inhibit pro-inflammatory synergistic action of TNF-alpha/IL-17A on human keratinocytes by regulating IkappaBzeta expression. Tacrolimus 0-10 interleukin 17A Homo sapiens 72-78 31838663-10 2020 Tacrolimus can inhibit pro-inflammatory synergistic action of TNF-alpha/IL-17A on human keratinocytes by regulating IkappaBzeta expression. Tacrolimus 0-10 NFKB inhibitor zeta Homo sapiens 116-127 31755126-0 2020 Initial dose optimization of tacrolimus for children with systemic lupus erythematosus based on the CYP3A5 polymorphism and coadministration with Wuzhi capsule. Tacrolimus 29-39 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 100-106 31755126-4 2020 RESULTS AND DISCUSSION: The results showed that weight, the CYP3A5 genotype and combined treatment with Wuzhi capsule can affect tacrolimus clearance in children with systemic lupus erythematosus. Tacrolimus 129-139 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 60-66 31755126-8 2020 WHAT IS NEW AND CONCLUSION: This study is the first study to recommend an optimal initial regimen of tacrolimus for children with systemic lupus erythematosus based on the CYP3A5 polymorphism and coadministration of Wuzhi capsule. Tacrolimus 101-111 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 172-178 31756280-6 2020 Gene screening for CYP3A5 6986 A>G and ABCB1 3435 C>T in organ transplant recipients may help in preventing DDI and facilitating tacrolimus dose adjustment. Tacrolimus 129-139 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 19-25 31756280-6 2020 Gene screening for CYP3A5 6986 A>G and ABCB1 3435 C>T in organ transplant recipients may help in preventing DDI and facilitating tacrolimus dose adjustment. Tacrolimus 129-139 ATP binding cassette subfamily B member 1 Homo sapiens 39-44 31811646-7 2020 The inhibition of calcineurin with FK506 significantly inhibited the nuclear levels of NFAT2 and NFAT4 and the inhibition of P38 MAPK with SB203580 inhibited the nuclear and cytoplasmic accumulation of NFAT1. Tacrolimus 35-40 nuclear factor of activated T-cells 3 Rattus norvegicus 97-102 31820394-1 2020 The calcineurin inhibitor tacrolimus is an effective immunosuppressant and is extensively used in solid organ transplantation. Tacrolimus 26-36 calcineurin binding protein 1 Homo sapiens 4-25 32170593-1 2020 BACKGROUND: The aim of this meta-analysis is to explore the effect of IL-2RA vs rATG on the rate of acute rejection, post-transplant infections, and graft as well as patient"s survival in standard- and high-risk renal transplant patients receiving tacrolimus-based maintenance immunotherapy. Tacrolimus 248-258 interleukin 2 receptor subunit alpha Homo sapiens 70-76 31721244-2 2020 The present study aimed to evaluate the potential of an integrative approach to predict individual tacrolimus concentrations and dosages based on endogenous CYP3A4 phenotype, CYP3A5 genotype and clinical variables. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 157-163 31721244-4 2020 RESULTS AND DISCUSSION: The results suggested that endogenous CYP3A4 phenotype was the most important determinant of tacrolimus concentrations and dose requirements. Tacrolimus 117-127 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 31721244-7 2020 WHAT IS NEW AND CONCLUSION: In summary, endogenous CYP3A4 phenotype is a critical biomarker for the determination of tacrolimus disposition. Tacrolimus 117-127 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 51-57 31721244-8 2020 This predictive RF approach based on CYP3A4 biomarker with the combination of CYP3A5*3 genotype and other clinical variables can be used for predicting tacrolimus concentrations and dosages, which may serve as a useful tool in individualized tacrolimus dosing. Tacrolimus 152-162 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-43 31721244-8 2020 This predictive RF approach based on CYP3A4 biomarker with the combination of CYP3A5*3 genotype and other clinical variables can be used for predicting tacrolimus concentrations and dosages, which may serve as a useful tool in individualized tacrolimus dosing. Tacrolimus 152-162 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 78-84 31721244-8 2020 This predictive RF approach based on CYP3A4 biomarker with the combination of CYP3A5*3 genotype and other clinical variables can be used for predicting tacrolimus concentrations and dosages, which may serve as a useful tool in individualized tacrolimus dosing. Tacrolimus 242-252 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-43 31721244-8 2020 This predictive RF approach based on CYP3A4 biomarker with the combination of CYP3A5*3 genotype and other clinical variables can be used for predicting tacrolimus concentrations and dosages, which may serve as a useful tool in individualized tacrolimus dosing. Tacrolimus 242-252 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 78-84 31379240-9 2020 The SNPs in 3 genes (CYP1A2, DRD2, and PON1) were associated with risk of tacrolimus-induced nephrotoxicity. Tacrolimus 74-84 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 21-27 32308133-5 2020 CYP3A5 genotype significantly impacts oral tacrolimus concentrations and response after solid organ transplantation. Tacrolimus 43-53 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 31908055-6 2020 hILC showed lower TMG binding comparing to Lin+ cells, reduced expression of CD25 (basiliximab target), and diminished calcineurin activity with undetectable calcineurin and FKBP12 (tacrolimus target). Tacrolimus 182-192 C-C motif chemokine ligand 27 Homo sapiens 0-4 31049814-1 2020 BACKGROUND: Tacrolimus, a calcineurin inhibitor, is recommended by the recent guidelines from the Kidney Disease Improving Global Outcomes Group as the first-line treatment for steroid-resistant nephrotic syndrome (SRNS), but its clinical application in China is still limited. Tacrolimus 12-22 calcineurin binding protein 1 Homo sapiens 26-47 31379240-9 2020 The SNPs in 3 genes (CYP1A2, DRD2, and PON1) were associated with risk of tacrolimus-induced nephrotoxicity. Tacrolimus 74-84 paraoxonase 1 Homo sapiens 39-43 31654553-5 2020 In addition, TAC caused hepatic insulin resistance and triglyceride accumulation through IRS2/AKT and SREBP1 signaling, respectively. Tacrolimus 13-16 insulin receptor substrate 2 Mus musculus 89-93 32225074-0 2020 Influence of POR*28 Polymorphisms on CYP3A5*3-Associated Variations in Tacrolimus Blood Levels at an Early Stage after Liver Transplantation. Tacrolimus 71-81 cytochrome p450 oxidoreductase Homo sapiens 13-16 32225074-0 2020 Influence of POR*28 Polymorphisms on CYP3A5*3-Associated Variations in Tacrolimus Blood Levels at an Early Stage after Liver Transplantation. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 32225074-1 2020 It is well known that the CYP3A5*3 polymorphism is an important marker that correlates with the tacrolimus dose requirement after organ transplantation. Tacrolimus 96-106 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 26-32 32225074-2 2020 Recently, it has been revealed that the POR*28 polymorphism affects the pharmacokinetics of tacrolimus in renal transplant patients. Tacrolimus 92-102 cytochrome p450 oxidoreductase Homo sapiens 40-43 32225074-3 2020 In this study, we examined whether POR*28 as well as CYP3A5*3 polymorphism in Japanese recipients and donors would be another biomarker for the variation of tacrolimus blood levels in the recipients during the first month after living-donor liver transplantation. Tacrolimus 157-167 cytochrome p450 oxidoreductase Homo sapiens 35-38 32225074-3 2020 In this study, we examined whether POR*28 as well as CYP3A5*3 polymorphism in Japanese recipients and donors would be another biomarker for the variation of tacrolimus blood levels in the recipients during the first month after living-donor liver transplantation. Tacrolimus 157-167 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 53-59 32225074-8 2020 In conclusion, to our knowledge, this is the first report suggesting that the POR*28 polymorphism is another biomarker for the tacrolimus oral dosage after liver transplantation in patients carrying CYP3A5*1 rather than CYP3A5*3/*3. Tacrolimus 127-137 cytochrome p450 oxidoreductase Homo sapiens 78-81 32225074-8 2020 In conclusion, to our knowledge, this is the first report suggesting that the POR*28 polymorphism is another biomarker for the tacrolimus oral dosage after liver transplantation in patients carrying CYP3A5*1 rather than CYP3A5*3/*3. Tacrolimus 127-137 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 199-205 32210081-4 2020 We show that treatment of cultured hippocampal mouse and fetal human astrocytes with a CaN inhibitor FK506 resulted in a dynamic modulation of GLAST protein expression, being downregulated after 24-48 h, but upregulated after 7 days of continuous FK506 (200 nM) treatment. Tacrolimus 101-106 solute carrier family 1 member 3 Homo sapiens 143-148 32210081-4 2020 We show that treatment of cultured hippocampal mouse and fetal human astrocytes with a CaN inhibitor FK506 resulted in a dynamic modulation of GLAST protein expression, being downregulated after 24-48 h, but upregulated after 7 days of continuous FK506 (200 nM) treatment. Tacrolimus 247-252 solute carrier family 1 member 3 Homo sapiens 143-148 32235696-5 2020 In Nc/Nga mice, FX or TAC applied to the skin inhibited eosinophil infiltration with decreased expression of Il-33. Tacrolimus 22-25 interleukin 33 Mus musculus 109-114 32296037-4 2020 In this study, we observed that tacrolimus could induce triglyceride accumulation in hepatocytes by stimulating sterol response element-binding proteins (SREBPs) and miR-33a. Tacrolimus 32-42 microRNA 33a Homo sapiens 166-173 32296037-6 2020 Tacrolimus could downregulate circFASN and result in elevated miR-33a in vivo and in vitro. Tacrolimus 0-10 microRNA 33a Homo sapiens 62-69 32296037-7 2020 Overexpression of circFASN or silencing of miR-33a decreased the promoting effects of tacrolimus on triglyceride accumulation. Tacrolimus 86-96 microRNA 33a Homo sapiens 43-50 32296037-9 2020 Our results showed that the circFASN/miR-33a regulatory system plays a distinct role in tacrolimus-induced disruption of lipid homeostasis. Tacrolimus 88-98 microRNA 33a Homo sapiens 37-44 32296037-10 2020 MiR-33a is likely a risk factor for tacrolimus-related dyslipidemia, providing a potential therapeutic target to combat tacrolimus-induced dyslipidemia after liver transplantation. Tacrolimus 36-46 microRNA 33a Homo sapiens 0-7 32296037-10 2020 MiR-33a is likely a risk factor for tacrolimus-related dyslipidemia, providing a potential therapeutic target to combat tacrolimus-induced dyslipidemia after liver transplantation. Tacrolimus 120-130 microRNA 33a Homo sapiens 0-7 32308538-10 2020 This can be explained by the increase in TAC concentration caused by CYP3A4 inhibition due to LMV and by the decrease in TAC concentration ascribed to the decrease in VRCZ concentration by CYP2C19 induction due to LMV. Tacrolimus 41-44 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-75 32308538-10 2020 This can be explained by the increase in TAC concentration caused by CYP3A4 inhibition due to LMV and by the decrease in TAC concentration ascribed to the decrease in VRCZ concentration by CYP2C19 induction due to LMV. Tacrolimus 121-124 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 189-196 31654553-5 2020 In addition, TAC caused hepatic insulin resistance and triglyceride accumulation through IRS2/AKT and SREBP1 signaling, respectively. Tacrolimus 13-16 thymoma viral proto-oncogene 1 Mus musculus 94-97 31654553-5 2020 In addition, TAC caused hepatic insulin resistance and triglyceride accumulation through IRS2/AKT and SREBP1 signaling, respectively. Tacrolimus 13-16 sterol regulatory element binding transcription factor 1 Mus musculus 102-108 31654553-6 2020 Furthermore, we found a pivotal role of CRTC2 in TAC-induced metabolic disorders. Tacrolimus 49-52 CREB regulated transcription coactivator 2 Mus musculus 40-45 31654553-10 2020 Taken together, in addition to its impact on pancreatic cells, TAC induces "hepatogenous diabetes" via CRTC2 signaling. Tacrolimus 63-66 CREB regulated transcription coactivator 2 Mus musculus 103-108 31705533-13 2020 CONCLUSION AND IMPLICATIONS: The hypertensive effects of the immunosuppressant drugs FK506 and rapamycin, while mediated by endothelial cells, do not appear to be exerted at documented cellular targets of the drugs on Ca2+ release and altered FKBP binding to IP3 and RyR. Tacrolimus 85-90 ryanodine receptor 2 Homo sapiens 267-270 31678537-8 2020 Rising serum tacrolimus or sirolimus levels, new acute kidney injury, and increasing platelet transfusion requirements were significant early predictors of onset in the week preceding prior VOD/SOS diagnosis. Tacrolimus 13-23 xylosyltransferase 2 Homo sapiens 194-197 32058973-8 2020 In parallel, chemical inhibition of the Cam/Calcineurin pathway by Cyclosporin A or FK506 also reduces CDTa phenotypes, potentially opening new avenues for treating CDIs. Tacrolimus 84-89 Calcineurin A1 Drosophila melanogaster 44-55 30890073-9 2020 Next, using the calcineurin inhibitor FK506, we implicated this phosphatase in activation of the nuclear factor of activated T-cells (NFAT; a transcription factor activated through calcineurin-mediated dephosphorylation) and propose that this pathway is involved in transcriptional upregulation of the IL-4 synthesis in NMDA-treated neurons. Tacrolimus 38-43 nuclear factor of activated T-cells 5 Rattus norvegicus 134-138 30890073-9 2020 Next, using the calcineurin inhibitor FK506, we implicated this phosphatase in activation of the nuclear factor of activated T-cells (NFAT; a transcription factor activated through calcineurin-mediated dephosphorylation) and propose that this pathway is involved in transcriptional upregulation of the IL-4 synthesis in NMDA-treated neurons. Tacrolimus 38-43 interleukin 4 Rattus norvegicus 302-306 31733802-2 2020 Although dasatinib is the first-line treatment for CML, it has inhibitory activity against CYP3A4; this might increase the blood concentration of tacrolimus (administered to KT patients for immune suppression). Tacrolimus 146-156 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 91-97 31931373-0 2020 Effects of donor-recipient combinational CYP3A5 genotypes on tacrolimus dosing in Chinese DDLT adult recipients. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 41-47 31931373-3 2020 This retrospective study was conducted to investigate the combined effects of donor-recipient CYP3A5 genotype on tacrolimus pharmacokinetics in Chinese LT adult patients. Tacrolimus 113-123 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 94-100 31931373-12 2020 CONCLUSIONS: To predict the initial dose of tacrolimus in LT patients, both donor and recipient CYP3A5 genotypes must be taken into account; during the maintenance phase of targeted blood concentration, the donor"s CYP3A5 genotype may be of prime importance, especially at three months after transplantation. Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 215-221 32006551-5 2020 Transcription assays of calcineurin (CaN)- and mTOR (mammalian target of rapamycin)-downstream target genes confirm that BbFKBP12 is the target of both FK506 and rapamycin, associated with CaN- and mTOR-signal pathways in B. bassiana. Tacrolimus 152-157 mechanistic target of rapamycin kinase Homo sapiens 47-51 32006551-5 2020 Transcription assays of calcineurin (CaN)- and mTOR (mammalian target of rapamycin)-downstream target genes confirm that BbFKBP12 is the target of both FK506 and rapamycin, associated with CaN- and mTOR-signal pathways in B. bassiana. Tacrolimus 152-157 mechanistic target of rapamycin kinase Homo sapiens 53-82 32006551-5 2020 Transcription assays of calcineurin (CaN)- and mTOR (mammalian target of rapamycin)-downstream target genes confirm that BbFKBP12 is the target of both FK506 and rapamycin, associated with CaN- and mTOR-signal pathways in B. bassiana. Tacrolimus 152-157 mechanistic target of rapamycin kinase Homo sapiens 198-202 32067305-6 2020 Two patients with severe food allergy responded favorably to conversion from tacrolimus-based immunosuppression to MMF and corticosteroids with reduction in clinical symptoms, total IgE, specific IgE, IL-1ra, IL-4, RANTES, PDGF, MIP-1a, and TNFalpha. Tacrolimus 77-87 interleukin 1 receptor antagonist Homo sapiens 201-207 32067305-6 2020 Two patients with severe food allergy responded favorably to conversion from tacrolimus-based immunosuppression to MMF and corticosteroids with reduction in clinical symptoms, total IgE, specific IgE, IL-1ra, IL-4, RANTES, PDGF, MIP-1a, and TNFalpha. Tacrolimus 77-87 interleukin 4 Homo sapiens 209-213 32067305-6 2020 Two patients with severe food allergy responded favorably to conversion from tacrolimus-based immunosuppression to MMF and corticosteroids with reduction in clinical symptoms, total IgE, specific IgE, IL-1ra, IL-4, RANTES, PDGF, MIP-1a, and TNFalpha. Tacrolimus 77-87 C-C motif chemokine ligand 5 Homo sapiens 215-221 32067305-6 2020 Two patients with severe food allergy responded favorably to conversion from tacrolimus-based immunosuppression to MMF and corticosteroids with reduction in clinical symptoms, total IgE, specific IgE, IL-1ra, IL-4, RANTES, PDGF, MIP-1a, and TNFalpha. Tacrolimus 77-87 C-C motif chemokine ligand 3 Homo sapiens 229-235 32067305-6 2020 Two patients with severe food allergy responded favorably to conversion from tacrolimus-based immunosuppression to MMF and corticosteroids with reduction in clinical symptoms, total IgE, specific IgE, IL-1ra, IL-4, RANTES, PDGF, MIP-1a, and TNFalpha. Tacrolimus 77-87 tumor necrosis factor Homo sapiens 241-249 31807785-1 2020 FKBP53 is one of the seven multi-domain FK506-binding proteins present in Arabidopsis thaliana, and it is known to get targeted to the nucleus. Tacrolimus 40-45 FK506 BINDING PROTEIN 53 Arabidopsis thaliana 0-6 32048900-1 2022 Background: The pathogenesis of vitiligo is complex and multifactorial, accumulating evidence of increased oxidative stress and reduction in catalase levels in vitiligo patients has been shown, hence, pseudocatalase/superoxide dismutase (PSD) gel has been used as treatment option for vitiligo.Aim: To assesses the synergic effect of PSD when combines with Tacrolimus 0.1% ointment versus Tacrolimus 0.1% alone.Method: A randomized controlled trial that included 49 children with vitiligo with limited area (10% or less). Tacrolimus 357-367 catalase Homo sapiens 141-149 32048900-1 2022 Background: The pathogenesis of vitiligo is complex and multifactorial, accumulating evidence of increased oxidative stress and reduction in catalase levels in vitiligo patients has been shown, hence, pseudocatalase/superoxide dismutase (PSD) gel has been used as treatment option for vitiligo.Aim: To assesses the synergic effect of PSD when combines with Tacrolimus 0.1% ointment versus Tacrolimus 0.1% alone.Method: A randomized controlled trial that included 49 children with vitiligo with limited area (10% or less). Tacrolimus 389-399 catalase Homo sapiens 141-149 32034192-0 2020 Author Correction: Whole exome sequencing for the identification of CYP3A7 variants associated with tacrolimus concentrations in kidney transplant patients. Tacrolimus 100-110 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 68-74 31386765-2 2020 Tacrolimus (or FK506), a calcineurin inhibitor (CNI) introduced in field of transplantation in the 1990s, is the cornerstone of most immunosuppressive regimens in solid organ transplantation. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 25-46 31441337-6 2020 Data Synthesis: Studies demonstrate pharmacokinetic differences between cyclosporine and tacrolimus, particularly with regard to inhibition of 2 hepatic transporters: P-glycoprotein and organic anion transporting polypeptide (OATP). Tacrolimus 89-99 ATP binding cassette subfamily B member 1 Homo sapiens 167-181 31441337-6 2020 Data Synthesis: Studies demonstrate pharmacokinetic differences between cyclosporine and tacrolimus, particularly with regard to inhibition of 2 hepatic transporters: P-glycoprotein and organic anion transporting polypeptide (OATP). Tacrolimus 89-99 solute carrier organic anion transporter family member 1A2 Homo sapiens 186-224 31441337-6 2020 Data Synthesis: Studies demonstrate pharmacokinetic differences between cyclosporine and tacrolimus, particularly with regard to inhibition of 2 hepatic transporters: P-glycoprotein and organic anion transporting polypeptide (OATP). Tacrolimus 89-99 solute carrier organic anion transporter family member 1A2 Homo sapiens 226-230 31386765-2 2020 Tacrolimus (or FK506), a calcineurin inhibitor (CNI) introduced in field of transplantation in the 1990s, is the cornerstone of most immunosuppressive regimens in solid organ transplantation. Tacrolimus 15-20 calcineurin binding protein 1 Homo sapiens 25-46 31732500-0 2020 Tacrolimus-Induced BMP/SMAD Signaling Associates with Metabolic Stress-Activated FOXO1 to Trigger beta-Cell Failure. Tacrolimus 0-10 bone morphogenetic protein 1 Homo sapiens 19-22 31725919-9 2020 Body weight, age, CYP3A5 genotype, cystatin-C and daily dose of tacrolimus may have significant effects on the pharmacokinetics of tacrolimus in children with glomerular disease. Tacrolimus 131-141 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 18-24 31725919-9 2020 Body weight, age, CYP3A5 genotype, cystatin-C and daily dose of tacrolimus may have significant effects on the pharmacokinetics of tacrolimus in children with glomerular disease. Tacrolimus 131-141 cystatin C Homo sapiens 35-45 31732500-0 2020 Tacrolimus-Induced BMP/SMAD Signaling Associates with Metabolic Stress-Activated FOXO1 to Trigger beta-Cell Failure. Tacrolimus 0-10 forkhead box O1 Homo sapiens 81-86 31732500-3 2020 Here we show that tacrolimus induces loss of human beta-cell maturity and beta-cell failure through activation of the BMP/SMAD signaling pathway when administered under mild metabolic stress conditions. Tacrolimus 18-28 bone morphogenetic protein 1 Homo sapiens 118-121 31732500-4 2020 Tacrolimus-induced phospho-SMAD1/5 acts in synergy with metabolic stress-activated FOXO1 through formation of a complex. Tacrolimus 0-10 forkhead box O1 Homo sapiens 83-88 31732500-6 2020 Pharmacological inhibition of BMP signaling protects human beta-cells from tacrolimus-induced beta-cell dysfunction in vitro Furthermore, we confirm that BMP/SMAD signaling is activated in protocol pancreas allograft biopsies from recipients on tacrolimus. Tacrolimus 75-85 bone morphogenetic protein 1 Homo sapiens 30-33 31732500-6 2020 Pharmacological inhibition of BMP signaling protects human beta-cells from tacrolimus-induced beta-cell dysfunction in vitro Furthermore, we confirm that BMP/SMAD signaling is activated in protocol pancreas allograft biopsies from recipients on tacrolimus. Tacrolimus 75-85 bone morphogenetic protein 1 Homo sapiens 154-157 31732500-6 2020 Pharmacological inhibition of BMP signaling protects human beta-cells from tacrolimus-induced beta-cell dysfunction in vitro Furthermore, we confirm that BMP/SMAD signaling is activated in protocol pancreas allograft biopsies from recipients on tacrolimus. Tacrolimus 245-255 bone morphogenetic protein 1 Homo sapiens 30-33 31732500-6 2020 Pharmacological inhibition of BMP signaling protects human beta-cells from tacrolimus-induced beta-cell dysfunction in vitro Furthermore, we confirm that BMP/SMAD signaling is activated in protocol pancreas allograft biopsies from recipients on tacrolimus. Tacrolimus 245-255 bone morphogenetic protein 1 Homo sapiens 154-157 32013193-2 2020 Germline mutations in cytochrome P450 isoforms 4 and 5 genes (CYP3A4/5) and the ATP-binding cassette B1 gene (ABCB1) may contribute to interindividual tacrolimus PK variability, which may impact clinical outcomes among allogeneic hematopoietic stem cell transplantation (HSCT) patients. Tacrolimus 151-161 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-70 32010314-0 2020 Tacrolimus reduces atherosclerotic plaque formation in ApoE-/- mice by inhibiting NLRP3 inflammatory corpuscles. Tacrolimus 0-10 apolipoprotein E Mus musculus 55-59 32010314-0 2020 Tacrolimus reduces atherosclerotic plaque formation in ApoE-/- mice by inhibiting NLRP3 inflammatory corpuscles. Tacrolimus 0-10 NLR family, pyrin domain containing 3 Mus musculus 82-87 32010314-10 2020 The protein content of NLRP3, ASC, Casp-1, IL-1beta and IL-18 in the aorta in ApoE-/- mice was remarkably increased, and they were inhibited to some extent after tacrolimus intervention. Tacrolimus 162-172 NLR family, pyrin domain containing 3 Mus musculus 23-28 32010314-10 2020 The protein content of NLRP3, ASC, Casp-1, IL-1beta and IL-18 in the aorta in ApoE-/- mice was remarkably increased, and they were inhibited to some extent after tacrolimus intervention. Tacrolimus 162-172 caspase 1 Mus musculus 35-41 32010314-10 2020 The protein content of NLRP3, ASC, Casp-1, IL-1beta and IL-18 in the aorta in ApoE-/- mice was remarkably increased, and they were inhibited to some extent after tacrolimus intervention. Tacrolimus 162-172 interleukin 1 alpha Mus musculus 43-51 32010314-10 2020 The protein content of NLRP3, ASC, Casp-1, IL-1beta and IL-18 in the aorta in ApoE-/- mice was remarkably increased, and they were inhibited to some extent after tacrolimus intervention. Tacrolimus 162-172 interleukin 18 Mus musculus 56-61 32010314-11 2020 In conclusion, it is speculated that tacrolimus may reduce the formation of AS through inhibiting ROS in macrophages and activation of NLRP3 inflammatory corpuscles and reducing the release of IL-1beta and IL-18. Tacrolimus 37-47 NLR family, pyrin domain containing 3 Mus musculus 135-140 32010314-11 2020 In conclusion, it is speculated that tacrolimus may reduce the formation of AS through inhibiting ROS in macrophages and activation of NLRP3 inflammatory corpuscles and reducing the release of IL-1beta and IL-18. Tacrolimus 37-47 interleukin 1 alpha Mus musculus 193-201 32010314-11 2020 In conclusion, it is speculated that tacrolimus may reduce the formation of AS through inhibiting ROS in macrophages and activation of NLRP3 inflammatory corpuscles and reducing the release of IL-1beta and IL-18. Tacrolimus 37-47 interleukin 18 Mus musculus 206-211 32013193-2 2020 Germline mutations in cytochrome P450 isoforms 4 and 5 genes (CYP3A4/5) and the ATP-binding cassette B1 gene (ABCB1) may contribute to interindividual tacrolimus PK variability, which may impact clinical outcomes among allogeneic hematopoietic stem cell transplantation (HSCT) patients. Tacrolimus 151-161 ATP binding cassette subfamily B member 1 Homo sapiens 110-115 32013193-4 2020 Significant associations were detected between germline variants in CYP3A4/5 and ABCB1 and PK endpoints (e.g., median steady-state tacrolimus concentrations and time to goal tacrolimus concentration). Tacrolimus 131-141 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-76 32013193-4 2020 Significant associations were detected between germline variants in CYP3A4/5 and ABCB1 and PK endpoints (e.g., median steady-state tacrolimus concentrations and time to goal tacrolimus concentration). Tacrolimus 131-141 ATP binding cassette subfamily B member 1 Homo sapiens 81-86 32013193-4 2020 Significant associations were detected between germline variants in CYP3A4/5 and ABCB1 and PK endpoints (e.g., median steady-state tacrolimus concentrations and time to goal tacrolimus concentration). Tacrolimus 174-184 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-76 32013193-4 2020 Significant associations were detected between germline variants in CYP3A4/5 and ABCB1 and PK endpoints (e.g., median steady-state tacrolimus concentrations and time to goal tacrolimus concentration). Tacrolimus 174-184 ATP binding cassette subfamily B member 1 Homo sapiens 81-86 32013193-6 2020 Decreased age and CYP3A5*1/*1 genotype were independently associated with subtherapeutic tacrolimus trough concentrations while CYP3A5*1*3 or CYP3A5*3/*3 genotypes, myeloablative allogeneic HSCT conditioning regimen (MAC) and increased weight were independently associated with supratherapeutic tacrolimus trough concentrations. Tacrolimus 89-99 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 18-24 32657689-0 2020 CYP3A5 polymorphisms and their effects on tacrolimus exposure in an ethnically diverse South African renal transplant population. Tacrolimus 42-52 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 31413313-4 2020 The association of tacrolimus and mTOR inhibitor is supported by pre-clinical data and has been used as GVHD prophylaxis. Tacrolimus 19-29 mechanistic target of rapamycin kinase Homo sapiens 34-38 32657689-3 2020 Cytochrome P-450 3A5 (CYP3A5) is the main enzyme involved in tacrolimus metabolism, and rs776746A>G is the most frequently studied polymorphism in the CYP3A5 gene. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-20 32657689-3 2020 Cytochrome P-450 3A5 (CYP3A5) is the main enzyme involved in tacrolimus metabolism, and rs776746A>G is the most frequently studied polymorphism in the CYP3A5 gene. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 22-28 32657689-3 2020 Cytochrome P-450 3A5 (CYP3A5) is the main enzyme involved in tacrolimus metabolism, and rs776746A>G is the most frequently studied polymorphism in the CYP3A5 gene. Tacrolimus 61-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 151-157 32657689-4 2020 The rs776746A>G (i.e. CYP3A5*3) single-nucleotide polymorphism in CYP3A5 alters tacrolimus predose trough concentration (C0) and may also affect IPV, which may lead to immune- and/or drug-mediated allograft injury. Tacrolimus 80-90 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 22-28 32657689-4 2020 The rs776746A>G (i.e. CYP3A5*3) single-nucleotide polymorphism in CYP3A5 alters tacrolimus predose trough concentration (C0) and may also affect IPV, which may lead to immune- and/or drug-mediated allograft injury. Tacrolimus 80-90 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 66-72 32657689-7 2020 OBJECTIVES: To determine the frequencies and effect of CYP3A5 and adenosine triphosphate-binding cassette subfamily B member 1 (ABCB1) polymorphisms on tacrolimus C0/dose ratios in different ethnic groups attending a tertiary renal transplant clinic in SA, and other factors that may explain inter- and IPV in tacrolimus C0. Tacrolimus 152-162 ATP binding cassette subfamily B member 1 Homo sapiens 128-133 32657689-19 2020 CONCLUSIONS: Compared with global transplant populations, SA renal transplant recipients demonstrated a very high rate of CYP3A5 expression, with a significant impact on tacrolimus pharmacokinetics. Tacrolimus 170-180 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 122-128 32657689-20 2020 Genetic variation in CYP3A5 expression affects tacrolimus dosing requirements, and knowing the CYP3A5 genotype of transplant patients may allow better dose prediction compared with current standard dosing recommendations in a multi-ethnic population. Tacrolimus 47-57 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 21-27 31683063-4 2020 We show here that both an ATP-independent protein chaperone, Spy L32P, and the FK506 binding domain of a prolyl isomerase, FKBP-25 F145A/I223P, are disordered, yet exhibit structures that are distinct from chemically denatured unfolded states in solution, and that they undergo transitions to a more structured state upon ligand binding. Tacrolimus 79-84 FKBP prolyl isomerase 3 Homo sapiens 123-130 31683063-7 2020 The protein-ligand systems studied here (the ATP-independent protein chaperone, Spy L32P, and the FK506 binding domain of a prolyl isomerase, FKBP-25 F145A/I223P) may serve as models for understanding ligand-induced disorder-to-order transitions in proteins. Tacrolimus 98-103 FKBP prolyl isomerase 3 Homo sapiens 142-149 32149187-1 2020 Tacrolimus is a reversible calcineurin inhibitor. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 27-48 32268334-1 2020 BACKGROUND: This study aims to assess outcomes of interleukin-2 (IL-2) receptor blocker induction therapy on allograft and patients" outcomes in standard risk recipients in the tacrolimus era, analysing data form the British Renal Transplant Registry. Tacrolimus 177-187 interleukin 2 Homo sapiens 50-63 32268334-1 2020 BACKGROUND: This study aims to assess outcomes of interleukin-2 (IL-2) receptor blocker induction therapy on allograft and patients" outcomes in standard risk recipients in the tacrolimus era, analysing data form the British Renal Transplant Registry. Tacrolimus 177-187 interleukin 2 Homo sapiens 65-69 32999170-3 2020 Because tacrolimus, which is a well-established immunosuppressant for kidney transplantation, and vonoprazan share the CYP3A4 system for metabolism, drug interactions are anticipated upon simultaneous administration. Tacrolimus 8-18 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 119-125 31941840-3 2020 Both TAC and SIR impaired insulin secretion in fasted and/or stimulated conditions. Tacrolimus 5-8 insulin Homo sapiens 26-33 31941840-4 2020 Treatment with TAC or SIR increased amyloid deposition and islet macrophages, disrupted insulin granule formation, and induced broad transcriptional dysregulation related to peptide processing, ion/calcium flux, and the extracellular matrix; however, it did not affect regulation of beta cell mass. Tacrolimus 15-18 insulin Homo sapiens 88-95 31941840-6 2020 Furthermore, cotreatment with a GLP-1 receptor agonist completely prevented TAC-induced beta cell dysfunction and partially prevented SIR-induced beta cell dysfunction. Tacrolimus 76-79 glucagon like peptide 1 receptor Homo sapiens 32-46 31794606-5 2020 Intriguingly, although NFAT is activated in both DLBCL subtypes, long-term calcineurin inhibition with cyclosporin A or FK506, both clinically approved drugs, triggers potent cytotoxicity specifically in ABC DLBCL cells. Tacrolimus 120-125 ATP binding cassette subfamily B member 6 (Langereis blood group) Homo sapiens 204-207 31919468-8 2020 Consistently, AMPA receptor antagonist CNQX or calcineurin inhibitor FK506 abolished the S-SCAM overexpression-induced loss of GABAA receptors, supporting that GABAergic synapse loss by S-SCAM overexpression is due to the activity-induced dispersal of synaptic GABAA receptors. Tacrolimus 69-74 membrane associated guanylate kinase, WW and PDZ domain containing 2 Homo sapiens 89-95 31919468-8 2020 Consistently, AMPA receptor antagonist CNQX or calcineurin inhibitor FK506 abolished the S-SCAM overexpression-induced loss of GABAA receptors, supporting that GABAergic synapse loss by S-SCAM overexpression is due to the activity-induced dispersal of synaptic GABAA receptors. Tacrolimus 69-74 membrane associated guanylate kinase, WW and PDZ domain containing 2 Homo sapiens 186-192 32640952-5 2020 The recent progress shows that the recommended strategy is to use as maintenance immunosuppressive therapy a combination of a calcineurin inhibitor (preferably tacrolimus) with an antiproliferative drug (preferably mycophenolate mofetil) with steroids that can be withdrawn early or late in low-risk children. Tacrolimus 160-170 calcineurin binding protein 1 Homo sapiens 126-147 32378649-1 2020 Tacrolimus, a calcineurin inhibitor, affects bone metabolism and increases the risk of fracture due to marked bone loss. Tacrolimus 0-10 calcineurin binding protein 1 Rattus norvegicus 14-35 31897105-7 2020 Evaluation of the effectiveness revealed that MMF + GC produced significantly higher overall responses (i.e. complete remission plus partial remission) and that MMF + GC (OR=2.58; 95% CI, 1.67-3.97), CTX + RTX + GC (OR=3.89; 95% CI, 1.60-9.45), CTX + LEF + GC (OR=3.05; 95% CI, 1.05-8.84) and CTX + TAC + GC (OR=6.22; 95% CI, 1.93-20.05) had significantly higher overall responses compared with those to the traditional treatment regimen (CTX + GC). Tacrolimus 299-302 cytochrome P450 family 27 subfamily A member 1 Homo sapiens 200-203 32932253-7 2020 The immunological impact of tacrolimus was revealed by reduced percentages of Tfh, Breg, CD19+BAFF-R+ B cells, and increased percentages of Treg cells as well as down-regulated expression of IL-2, IL-4, IL-10, and IL-13 mRNA levels in PBMCs during the treatment. Tacrolimus 28-38 CD19 molecule Homo sapiens 89-93 32932253-7 2020 The immunological impact of tacrolimus was revealed by reduced percentages of Tfh, Breg, CD19+BAFF-R+ B cells, and increased percentages of Treg cells as well as down-regulated expression of IL-2, IL-4, IL-10, and IL-13 mRNA levels in PBMCs during the treatment. Tacrolimus 28-38 TNF receptor superfamily member 13C Homo sapiens 94-100 32932253-7 2020 The immunological impact of tacrolimus was revealed by reduced percentages of Tfh, Breg, CD19+BAFF-R+ B cells, and increased percentages of Treg cells as well as down-regulated expression of IL-2, IL-4, IL-10, and IL-13 mRNA levels in PBMCs during the treatment. Tacrolimus 28-38 interleukin 2 Homo sapiens 191-195 32932253-7 2020 The immunological impact of tacrolimus was revealed by reduced percentages of Tfh, Breg, CD19+BAFF-R+ B cells, and increased percentages of Treg cells as well as down-regulated expression of IL-2, IL-4, IL-10, and IL-13 mRNA levels in PBMCs during the treatment. Tacrolimus 28-38 interleukin 4 Homo sapiens 197-201 32932253-7 2020 The immunological impact of tacrolimus was revealed by reduced percentages of Tfh, Breg, CD19+BAFF-R+ B cells, and increased percentages of Treg cells as well as down-regulated expression of IL-2, IL-4, IL-10, and IL-13 mRNA levels in PBMCs during the treatment. Tacrolimus 28-38 interleukin 10 Homo sapiens 203-208 32932253-7 2020 The immunological impact of tacrolimus was revealed by reduced percentages of Tfh, Breg, CD19+BAFF-R+ B cells, and increased percentages of Treg cells as well as down-regulated expression of IL-2, IL-4, IL-10, and IL-13 mRNA levels in PBMCs during the treatment. Tacrolimus 28-38 interleukin 13 Homo sapiens 214-219 31897105-8 2020 Ranking probability based on the surface under the cumulative ranking curve indicated that CTX + TAC + GC had the highest probability (80.6%) of being the best treatment for achieving an overall response. Tacrolimus 97-100 cytochrome P450 family 27 subfamily A member 1 Homo sapiens 91-94 31689461-6 2019 At the molecular level, FK506-TKM significantly inhibited infiltration of CD4+ and CD8+ T lymphocytes in colon and differentiation of CD4+ T cells into Th1 and Th17 cells in colon-draining mesenteric lymph nodes via restricting dendritic cell migration from colon. Tacrolimus 24-29 CD4 antigen Mus musculus 74-77 31908154-0 2020 Tacrolimus-induced hypomagnesemia and hypercalciuria requires FKBP12 suggesting a role for calcineurin. Tacrolimus 0-10 FK506 binding protein 1a Mus musculus 62-68 31908154-5 2020 The CNI tacrolimus can inhibit calcineurin only when it binds with the immunophilin, FKBP12; we previously generated mice in which FKBP12 could be deleted along the nephron, to test whether calcineurin inhibition is involved, these mice are normal at baseline. Tacrolimus 8-18 FK506 binding protein 1a Mus musculus 85-91 31908154-6 2020 Here, we confirmed that tacrolimus-treated control mice developed hypomagnesemia and urinary calcium wasting, with decreased protein and mRNA abundance of key magnesium and calcium transport proteins (NCX-1 and Calbindin-D28k ). Tacrolimus 24-34 solute carrier family 8 (sodium/calcium exchanger), member 1 Mus musculus 201-206 31908154-6 2020 Here, we confirmed that tacrolimus-treated control mice developed hypomagnesemia and urinary calcium wasting, with decreased protein and mRNA abundance of key magnesium and calcium transport proteins (NCX-1 and Calbindin-D28k ). Tacrolimus 24-34 calbindin 1 Mus musculus 211-225 31908154-8 2020 In contrast, KS-FKBP12-/- mice treated with tacrolimus were completely protected from these effects. Tacrolimus 44-54 FK506 binding protein 1a Mus musculus 16-22 31914663-9 2020 Inhibition of calcineurin by FK506 results in an increase in the net phosphorylation and a consequent decrease in catalytic activity of sperm GSK3. Tacrolimus 29-34 glycogen synthase kinase 3 beta Mus musculus 142-146 31849280-0 2020 CYP3A5*3 and CYP2C8*3 variants influence exposure and clinical outcomes of tacrolimus-based therapy. Tacrolimus 75-85 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 31849280-4 2020 Main results: CYP3A5*3/*3 genotype influenced increase in TAC concentration from week 1 to month 6 post-transplantation (p < 0.05). Tacrolimus 58-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 31849280-6 2020 Conclusion: The CYP3A5*3 variant is associated with increased early exposure to TAC. Tacrolimus 80-83 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 16-22 31689461-6 2019 At the molecular level, FK506-TKM significantly inhibited infiltration of CD4+ and CD8+ T lymphocytes in colon and differentiation of CD4+ T cells into Th1 and Th17 cells in colon-draining mesenteric lymph nodes via restricting dendritic cell migration from colon. Tacrolimus 24-29 CD4 antigen Mus musculus 134-137 31931404-0 2019 Tacrolimus is effective for neuromyelitis optica spectrum disorders with or without anti-AQP4 antibody. Tacrolimus 0-10 aquaporin 4 Homo sapiens 89-93 31949424-0 2019 Concomitant Treatment with Etanercept and Tacrolimus Synergistically Attenuates Arthritis Progression via Inhibition of Matrix Metalloproteinase-3 Production and Osteoclastogenesis in Human TNF-alpha Transgenic Mice. Tacrolimus 42-52 tumor necrosis factor Homo sapiens 190-199 31931404-14 2019 CONCLUSION: Combined use of TAC with PSL clearly suppressed relapse of both anti-AQP4 antibody-positive and -negative NMOSD. Tacrolimus 28-31 aquaporin 4 Homo sapiens 81-85 31921171-3 2019 The tacrolimus TTR percentage was calculated by linear interpolation with a target range (5-10 ng/ml months 0-3, 4-8 ng/ml months 4-12). Tacrolimus 4-14 transthyretin Homo sapiens 15-18 31921171-14 2019 Conclusions: Increasing the TTR of tacrolimus in the first year was associated with improved long-term outcomes in living kidney transplants, and TTR may be a novel valuable strategy to monitor tacrolimus exposure. Tacrolimus 35-45 transthyretin Homo sapiens 28-31 31921171-14 2019 Conclusions: Increasing the TTR of tacrolimus in the first year was associated with improved long-term outcomes in living kidney transplants, and TTR may be a novel valuable strategy to monitor tacrolimus exposure. Tacrolimus 194-204 transthyretin Homo sapiens 28-31 31921171-14 2019 Conclusions: Increasing the TTR of tacrolimus in the first year was associated with improved long-term outcomes in living kidney transplants, and TTR may be a novel valuable strategy to monitor tacrolimus exposure. Tacrolimus 194-204 transthyretin Homo sapiens 146-149 31836014-0 2019 FXR activation alleviates tacrolimus-induced post-transplant diabetes mellitus by regulating renal gluconeogenesis and glucose uptake. Tacrolimus 26-36 nuclear receptor subfamily 1, group H, member 4 Mus musculus 0-3 31836014-4 2019 The objective of this study was to explore whether FXR is involved in the development of tacrolimus-induced diabetes mellitus. Tacrolimus 89-99 nuclear receptor subfamily 1, group H, member 4 Mus musculus 51-54 31836014-9 2019 RESULTS: FK506 significantly inhibited the mRNA and protein levels of FXR at 48 h and 72 h in HK-2 cells (P < 0.05). Tacrolimus 9-14 nuclear receptor subfamily 1, group H, member 4 Mus musculus 70-73 31836014-5 2019 METHODS: After C57BL/6J mice were treated with tacrolimus (FK506) for 3 months, the fasting blood glucose levels, body weights, renal morphological alterations, and mRNA expression levels of phosphoenolpyruvate carboxykinase (PEPCK) and glucose transporter 2 (GLUT2) among the control group, the FK506 group and the FK506 + GW4064 (a FXR agonist) group (n = 7) were measured. Tacrolimus 47-57 phosphoenolpyruvate carboxykinase 1, cytosolic Mus musculus 191-224 31836014-12 2019 The FXR agonist GW4064 significantly decreased the fasting blood glucose in mice challenged with FK506 for 3 months (P < 0.05), inhibited gluconeogenesis (P < 0.05) and significantly promoted glucose uptake (P < 0.05). Tacrolimus 97-102 nuclear receptor subfamily 1, group H, member 4 Mus musculus 4-7 31836014-14 2019 CONCLUSIONS: FXR activation may mitigate tacrolimus-induced diabetes mellitus by regulating gluconeogenesis as well as glucose uptake of renal cortex proximal tubule epithelial cells in a PGC1alpha/FOXO1-dependent manner, which may be a potential therapeutic strategy for the prevention and treatment of PTDM. Tacrolimus 41-51 nuclear receptor subfamily 1, group H, member 4 Mus musculus 13-16 31836014-14 2019 CONCLUSIONS: FXR activation may mitigate tacrolimus-induced diabetes mellitus by regulating gluconeogenesis as well as glucose uptake of renal cortex proximal tubule epithelial cells in a PGC1alpha/FOXO1-dependent manner, which may be a potential therapeutic strategy for the prevention and treatment of PTDM. Tacrolimus 41-51 peroxisome proliferative activated receptor, gamma, coactivator 1 alpha Mus musculus 188-197 31836014-5 2019 METHODS: After C57BL/6J mice were treated with tacrolimus (FK506) for 3 months, the fasting blood glucose levels, body weights, renal morphological alterations, and mRNA expression levels of phosphoenolpyruvate carboxykinase (PEPCK) and glucose transporter 2 (GLUT2) among the control group, the FK506 group and the FK506 + GW4064 (a FXR agonist) group (n = 7) were measured. Tacrolimus 59-64 phosphoenolpyruvate carboxykinase 1, cytosolic Mus musculus 191-224 31836014-14 2019 CONCLUSIONS: FXR activation may mitigate tacrolimus-induced diabetes mellitus by regulating gluconeogenesis as well as glucose uptake of renal cortex proximal tubule epithelial cells in a PGC1alpha/FOXO1-dependent manner, which may be a potential therapeutic strategy for the prevention and treatment of PTDM. Tacrolimus 41-51 forkhead box O1 Mus musculus 198-203 32047842-0 2020 One-year Outcome of Everolimus With Standard-dose Tacrolimus Immunosuppression in De Novo ABO-incompatible Living Donor Kidney Transplantation: A Retrospective, Single-center, Propensity Score Matching Comparison With Mycophenolate in 42 Transplants. Tacrolimus 50-60 ABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferase Homo sapiens 90-93 31152598-0 2019 Prolonged-Release Tacrolimus Is Less Susceptible to Interaction With the Strong CYP3A Inhibitor Voriconazole in Healthy Volunteers. Tacrolimus 18-28 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 80-85 31696965-11 2019 Notably, inactivation of the Cul3-Klhl18 ligase and calcineurin inhibitors FK506 and cyclosporine A that are known immunosuppressant drugs repressed light-induced photoreceptor damage, suggesting potential therapeutic targets. Tacrolimus 75-80 cullin 3 Mus musculus 29-33 31696965-11 2019 Notably, inactivation of the Cul3-Klhl18 ligase and calcineurin inhibitors FK506 and cyclosporine A that are known immunosuppressant drugs repressed light-induced photoreceptor damage, suggesting potential therapeutic targets. Tacrolimus 75-80 kelch-like 18 Mus musculus 34-40 31152486-0 2019 Inhibition of the mTOR pathway: A new mechanism of beta cell toxicity induced by tacrolimus. Tacrolimus 81-91 mechanistic target of rapamycin kinase Rattus norvegicus 18-22 31152486-2 2019 Tacrolimus (TAC) and rapamycin (Rapa) both bind to FK506-binding protein 12 (FKBP12). Tacrolimus 0-10 FKBP prolyl isomerase 1A Rattus norvegicus 51-75 31152486-2 2019 Tacrolimus (TAC) and rapamycin (Rapa) both bind to FK506-binding protein 12 (FKBP12). Tacrolimus 0-10 FKBP prolyl isomerase 1A Rattus norvegicus 77-83 31152486-2 2019 Tacrolimus (TAC) and rapamycin (Rapa) both bind to FK506-binding protein 12 (FKBP12). Tacrolimus 12-15 FKBP prolyl isomerase 1A Rattus norvegicus 51-75 31152486-2 2019 Tacrolimus (TAC) and rapamycin (Rapa) both bind to FK506-binding protein 12 (FKBP12). Tacrolimus 12-15 FKBP prolyl isomerase 1A Rattus norvegicus 77-83 31152486-4 2019 Because of this similarity, we hypothesized that TAC can also inhibit the mTOR signalling, constituting a possible mechanism of beta cell toxicity. Tacrolimus 49-52 mechanistic target of rapamycin kinase Rattus norvegicus 74-78 31152486-6 2019 TAC and Rapa inhibited the mTOR pathway as reflected by lower levels of phospho-mTOR, phospo-p70S6K, and phospo-S6. Tacrolimus 0-3 mechanistic target of rapamycin kinase Rattus norvegicus 27-31 31152486-6 2019 TAC and Rapa inhibited the mTOR pathway as reflected by lower levels of phospho-mTOR, phospo-p70S6K, and phospo-S6. Tacrolimus 0-3 mechanistic target of rapamycin kinase Rattus norvegicus 80-84 31152486-6 2019 TAC and Rapa inhibited the mTOR pathway as reflected by lower levels of phospho-mTOR, phospo-p70S6K, and phospo-S6. Tacrolimus 0-3 ribosomal protein S6 kinase B1 Rattus norvegicus 93-99 31152486-10 2019 In silico docking and immunoprecipitation experiments confirmed that TAC can form a stable noncovalent interaction with FKBP12-mTOR. Tacrolimus 69-72 FKBP prolyl isomerase 1A Rattus norvegicus 120-126 31152486-10 2019 In silico docking and immunoprecipitation experiments confirmed that TAC can form a stable noncovalent interaction with FKBP12-mTOR. Tacrolimus 69-72 mechanistic target of rapamycin kinase Rattus norvegicus 127-131 31152486-11 2019 Thus, the mTOR inhibition may be a mechanism contributing to the diabetogenic effect of TAC. Tacrolimus 88-91 mechanistic target of rapamycin kinase Rattus norvegicus 10-14 31087280-0 2019 The Effects of CYP3A5 Genetic Polymorphisms on Serum Tacrolimus Dose-Adjusted Concentrations and Long-Term Prognosis in Chinese Heart Transplantation Recipients. Tacrolimus 53-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 31087280-2 2019 We aim to investigate the effects of cytochrome P450 (CYP) 3A5 (rs776746) single nucleotide polymorphisms (SNPs) on serum tacrolimus concentrations/doses (C/Ds, ng/mL per mg/kg) and long-term prognosis in Chinese heart transplant recipients. Tacrolimus 122-132 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-62 31087280-8 2019 RESULTS: In 55 heart transplant recipients (43 males and 12 females), CYP3A5 non-expressors (CYP3A5*3/*3, n = 40) had significantly higher tacrolimus C/Ds than expressors (CYP3A5*1/*3, n = 15) at all time points (P < 0.001). Tacrolimus 139-149 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 70-76 31087280-8 2019 RESULTS: In 55 heart transplant recipients (43 males and 12 females), CYP3A5 non-expressors (CYP3A5*3/*3, n = 40) had significantly higher tacrolimus C/Ds than expressors (CYP3A5*1/*3, n = 15) at all time points (P < 0.001). Tacrolimus 139-149 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 93-99 31087280-8 2019 RESULTS: In 55 heart transplant recipients (43 males and 12 females), CYP3A5 non-expressors (CYP3A5*3/*3, n = 40) had significantly higher tacrolimus C/Ds than expressors (CYP3A5*1/*3, n = 15) at all time points (P < 0.001). Tacrolimus 139-149 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 93-99 31087280-12 2019 CONCLUSIONS: CYP3A5 SNPs affect tacrolimus pharmacokinetics in Chinese heart transplant recipients, and non-expressors have higher tacrolimus C/Ds. Tacrolimus 32-42 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 31087280-12 2019 CONCLUSIONS: CYP3A5 SNPs affect tacrolimus pharmacokinetics in Chinese heart transplant recipients, and non-expressors have higher tacrolimus C/Ds. Tacrolimus 131-141 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 31526865-10 2019 The serum MMP-8 and MMP-9 levels of allograft+FK506 + GM6001 group were significantly lower than those of allograft+FK506 group (P < .05). Tacrolimus 46-51 matrix metallopeptidase 8 Mus musculus 10-15 31638188-0 2019 Tacrolimus increases the expression level of the chemokine receptor CXCR2 to promote renal fibrosis progression. Tacrolimus 0-10 C-X-C motif chemokine receptor 2 Homo sapiens 68-73 31638188-7 2019 The present results demonstrated that compared with the normal control group, the tacrolimus nephrotoxicity group exhibited severe renal fibrosis (P<0.05), upregulated vimentin (P<0.01), downregulated E-cadherin (P<0.05) and upregulated alpha-SMA (P<0.01). Tacrolimus 82-92 vimentin Homo sapiens 171-179 31638188-7 2019 The present results demonstrated that compared with the normal control group, the tacrolimus nephrotoxicity group exhibited severe renal fibrosis (P<0.05), upregulated vimentin (P<0.01), downregulated E-cadherin (P<0.05) and upregulated alpha-SMA (P<0.01). Tacrolimus 82-92 cadherin 1 Homo sapiens 207-217 31638188-9 2019 Moreover, KEGG pathway enrichment analysis identified that tacrolimus increased the expression levels of chemokine (C-X-C) motif ligand (CXCL)1, CXCL2 and CXCL3 and the chemokine receptor C-X-C chemokine receptor type 2 (CXCR2). Tacrolimus 59-69 C-X-C motif chemokine ligand 1 Homo sapiens 116-143 31638188-9 2019 Moreover, KEGG pathway enrichment analysis identified that tacrolimus increased the expression levels of chemokine (C-X-C) motif ligand (CXCL)1, CXCL2 and CXCL3 and the chemokine receptor C-X-C chemokine receptor type 2 (CXCR2). Tacrolimus 59-69 C-X-C motif chemokine ligand 2 Homo sapiens 145-150 31638188-9 2019 Moreover, KEGG pathway enrichment analysis identified that tacrolimus increased the expression levels of chemokine (C-X-C) motif ligand (CXCL)1, CXCL2 and CXCL3 and the chemokine receptor C-X-C chemokine receptor type 2 (CXCR2). Tacrolimus 59-69 C-X-C motif chemokine ligand 3 Homo sapiens 155-160 31638188-9 2019 Moreover, KEGG pathway enrichment analysis identified that tacrolimus increased the expression levels of chemokine (C-X-C) motif ligand (CXCL)1, CXCL2 and CXCL3 and the chemokine receptor C-X-C chemokine receptor type 2 (CXCR2). Tacrolimus 59-69 C-X-C motif chemokine receptor 2 Homo sapiens 221-226 31638188-10 2019 Collectively, the present study suggested that tacrolimus increases the level of chemokine receptor CXCR2 to promote renal fibrosis progression, which is one of the potential mechanisms underlying tacrolimus-induced nephrotoxicity. Tacrolimus 47-57 C-X-C motif chemokine receptor 2 Homo sapiens 100-105 31638188-10 2019 Collectively, the present study suggested that tacrolimus increases the level of chemokine receptor CXCR2 to promote renal fibrosis progression, which is one of the potential mechanisms underlying tacrolimus-induced nephrotoxicity. Tacrolimus 197-207 C-X-C motif chemokine receptor 2 Homo sapiens 100-105 31514576-0 2019 Comparison of Tacrolimus Starting Doses Based on CYP3A5 Phenotype or Genotype in Kidney Transplant Recipients. Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 49-55 31514576-1 2019 BACKGROUND: Selection of expected phenotypes (ie, expressers/non-expressers) is currently used in CYP3A5*3 genotype-based tacrolimus dosing. Tacrolimus 122-132 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 98-104 31514576-8 2019 Based on these results, new tacrolimus starting doses were set at 0.10, 0.20, and 0.30 mg/kg/d for CYP3A5*3/*3, CYP3A5*1/*3, and CYP3A5*1/*1 genotypes, respectively. Tacrolimus 28-38 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 99-105 31514576-8 2019 Based on these results, new tacrolimus starting doses were set at 0.10, 0.20, and 0.30 mg/kg/d for CYP3A5*3/*3, CYP3A5*1/*3, and CYP3A5*1/*1 genotypes, respectively. Tacrolimus 28-38 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 112-118 31514576-8 2019 Based on these results, new tacrolimus starting doses were set at 0.10, 0.20, and 0.30 mg/kg/d for CYP3A5*3/*3, CYP3A5*1/*3, and CYP3A5*1/*1 genotypes, respectively. Tacrolimus 28-38 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 112-118 31514576-9 2019 Tacrolimus overexposure was reduced in the CYP3A5*3/*3 group (63.6% vs 40%, P = .0038). Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 43-49 31514576-12 2019 Our results indicate that the best tacrolimus exposure was obtained for doses of 0.10, 0.20, and 0.20 mg/kg/d for CYP3A5*3/3, CYP3A5*1/*3, and CYP3A5*1/*1, respectively. Tacrolimus 35-45 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 114-120 31514576-12 2019 Our results indicate that the best tacrolimus exposure was obtained for doses of 0.10, 0.20, and 0.20 mg/kg/d for CYP3A5*3/3, CYP3A5*1/*3, and CYP3A5*1/*1, respectively. Tacrolimus 35-45 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 126-132 31514576-12 2019 Our results indicate that the best tacrolimus exposure was obtained for doses of 0.10, 0.20, and 0.20 mg/kg/d for CYP3A5*3/3, CYP3A5*1/*3, and CYP3A5*1/*1, respectively. Tacrolimus 35-45 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 126-132 31733794-7 2019 Therapeutic concentrations of TAC and SRL reduced the percentage of pd1+ and icos+ Tfh cells compared to controls. Tacrolimus 30-33 patr class I histocompatibility antigen, A-126 alpha chain-like Sus scrofa 68-71 31733794-8 2019 In addition, T cells grown in the presence of TAC or SRL expressed less IL-21 and provided less B-cell help. Tacrolimus 46-49 interleukin 21 Homo sapiens 72-77 31526865-10 2019 The serum MMP-8 and MMP-9 levels of allograft+FK506 + GM6001 group were significantly lower than those of allograft+FK506 group (P < .05). Tacrolimus 46-51 matrix metallopeptidase 9 Mus musculus 20-25 31526865-11 2019 MMP-8 and MMP-9 protein expression in the grafts of allograft+FK506 + GM6001 group were lower than those of allograft+FK506 group verified by immunohistochemical staining and western blotting. Tacrolimus 62-67 matrix metallopeptidase 8 Mus musculus 0-5 31526865-11 2019 MMP-8 and MMP-9 protein expression in the grafts of allograft+FK506 + GM6001 group were lower than those of allograft+FK506 group verified by immunohistochemical staining and western blotting. Tacrolimus 62-67 matrix metallopeptidase 9 Mus musculus 10-15 31526865-12 2019 CONCLUSION: FK506 combined with GM6001 could alleviate tracheal obliteration in mouse heterotopic tracheal transplantation model, due to its inhibitory effect on MMPs. Tacrolimus 12-17 matrix metallopeptidase 8 Mus musculus 162-166 31710427-0 2019 Effect of CYP3A5 and ABCB1 Gene Polymorphisms on Tacrolimus Blood Concentration in Renal Transplant Recipients. Tacrolimus 49-59 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 32782938-3 2020 Tacrolimus is topically active, water soluble, and has minimal systemic toxicity when administered rectally; we therefore tested a simple tap water-based enema formulation. Tacrolimus 0-10 nuclear RNA export factor 1 Homo sapiens 138-141 31882422-5 2019 The effects of TSLP-DCs and treatments with FK506, an NFATc1 inhibitor, on naive T cell differentiation were monitored by measuring the interleukin (IL)-4, IL-13, and interferon-gamma (IFN-gamma) expression levels. Tacrolimus 44-49 nuclear factor of activated T cells 1 Homo sapiens 54-60 31271885-2 2019 Sirolimus is a mammalian target of rapamycin inhibitor that has proven effective in GVHD prophylaxis in combination with a calcineurin inhibitor, such as tacrolimus. Tacrolimus 154-164 mechanistic target of rapamycin kinase Homo sapiens 15-44 31710427-0 2019 Effect of CYP3A5 and ABCB1 Gene Polymorphisms on Tacrolimus Blood Concentration in Renal Transplant Recipients. Tacrolimus 49-59 ATP binding cassette subfamily B member 1 Homo sapiens 21-26 31710427-3 2019 CYP3A5 and MDR1 single-nucleotide polymorphisms (SNPs) are the most effective polymorphisms that play an significant role in the pharmacokinetics of Tac. Tacrolimus 149-152 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 31710427-3 2019 CYP3A5 and MDR1 single-nucleotide polymorphisms (SNPs) are the most effective polymorphisms that play an significant role in the pharmacokinetics of Tac. Tacrolimus 149-152 ATP binding cassette subfamily B member 1 Homo sapiens 11-15 31710427-14 2019 CONCLUSIONS: CYP3A5 but not MDR1 genetic polymorphisms affected the Tac pharmacokinetics and dose requirements in renal transplant recipients. Tacrolimus 68-71 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 31401678-0 2019 CYP3A5 and CYP3A7 genetic polymorphisms affect tacrolimus concentration in pediatric patients with nephrotic range proteinuria. Tacrolimus 47-57 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 31600833-17 2019 Addition of bFGF related decapeptide solution to Tacrolimus gave better results than Tacrolimus alone therapy. Tacrolimus 49-59 fibroblast growth factor 2 Homo sapiens 12-16 31600833-17 2019 Addition of bFGF related decapeptide solution to Tacrolimus gave better results than Tacrolimus alone therapy. Tacrolimus 85-95 fibroblast growth factor 2 Homo sapiens 12-16 31401678-0 2019 CYP3A5 and CYP3A7 genetic polymorphisms affect tacrolimus concentration in pediatric patients with nephrotic range proteinuria. Tacrolimus 47-57 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 11-17 31401678-1 2019 PURPOSE: The purpose of this study was to investigate the potential impact of CYP3A4, CYP3A5, and CYP3A7 polymorphisms on the concentration and efficacy of tacrolimus in a cohort of pediatric patients with nephrotic range proteinuria. Tacrolimus 156-166 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 78-84 31401678-1 2019 PURPOSE: The purpose of this study was to investigate the potential impact of CYP3A4, CYP3A5, and CYP3A7 polymorphisms on the concentration and efficacy of tacrolimus in a cohort of pediatric patients with nephrotic range proteinuria. Tacrolimus 156-166 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 86-92 31401678-1 2019 PURPOSE: The purpose of this study was to investigate the potential impact of CYP3A4, CYP3A5, and CYP3A7 polymorphisms on the concentration and efficacy of tacrolimus in a cohort of pediatric patients with nephrotic range proteinuria. Tacrolimus 156-166 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 98-104 31468631-4 2019 Tacrolimus, a calcineurin inhibitor, restored the high- and low-pressure baroreflex control of RSNA following cisplatin-induced renal injury. Tacrolimus 0-10 calcineurin binding protein 1 Rattus norvegicus 14-35 31401678-4 2019 RESULTS: The tacrolimus concentration in patients without CYP3A5*3 A allele was 94% higher than those with A allele (90.7 vs 54.2, P = 0.00006). Tacrolimus 13-23 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 58-64 31401678-5 2019 The CYP3A7 rs2257401 was also associated with the concentration of tacrolimus. Tacrolimus 67-77 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 4-10 31401678-7 2019 In addition, there were significant differences in tacrolimus concentration among CYP3A7 rs10211 G carriers and non-carriers; the latter had an almost twofold C0 of the former (101.8 vs 59.6, P = 0.0004). Tacrolimus 51-61 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 82-88 31401678-8 2019 CONCLUSIONS: Our study demonstrated the associations between CYP3A5*3, CYP3A7 rs2257401 and rs10211, and tacrolimus concentration in pediatric patients with nephrotic range proteinuria. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 61-67 31401678-8 2019 CONCLUSIONS: Our study demonstrated the associations between CYP3A5*3, CYP3A7 rs2257401 and rs10211, and tacrolimus concentration in pediatric patients with nephrotic range proteinuria. Tacrolimus 105-115 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 71-77 31401678-9 2019 Children with CYP3A5*3 A, CYP3A7 rs2257401 C, and rs10211 G alleles might need a higher dose of tacrolimus. Tacrolimus 96-106 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 31401678-9 2019 Children with CYP3A5*3 A, CYP3A7 rs2257401 C, and rs10211 G alleles might need a higher dose of tacrolimus. Tacrolimus 96-106 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 26-32 31488544-3 2019 Although AIPL1 and PDE6 are known to interact via the FK506-binding protein domain of AIPL1, the contribution of the tetratricopeptide repeat (TPR) domain of AIPL1 to its chaperone function is poorly understood. Tacrolimus 54-59 aryl hydrocarbon receptor interacting protein like 1 Homo sapiens 86-91 31880588-0 2019 Association Between CYP3A5 Genetic Polymorphisms with Tacrolimus Dose Requirement and Allograft Outcomes in Iranian Kidney Transplant Recipients. Tacrolimus 54-64 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 31880588-2 2019 This study assessed the relationship between CYP3A5/3A4 polymorphisms and tacrolimus dose requirement as well as 6-month transplant outcomes in Iranian kidney transplant recipients. Tacrolimus 74-84 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-55 31880588-5 2019 RESULTS: The daily tacrolimus dose was significantly higher and tacrolimus dose adjusted trough levels (C/D ratio) was significantly lower in CYP3A5 expressers compared with non-expressers (P < .05). Tacrolimus 19-29 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 142-148 31880588-5 2019 RESULTS: The daily tacrolimus dose was significantly higher and tacrolimus dose adjusted trough levels (C/D ratio) was significantly lower in CYP3A5 expressers compared with non-expressers (P < .05). Tacrolimus 64-74 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 142-148 31880588-7 2019 CONCLUSION: CYP3A5 genetic polymorphism is significantly associated with required tacrolimus dose. Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 12-18 31880588-8 2019 After achieving desired tacrolimus blood level, although some transplant outcomes such as the incidence of clinically suggested acute rejection and time to first rejection were different between CYP3A5 expressers and non-expressers, however, other clinical outcomes did not differ between groups. Tacrolimus 24-34 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 195-201 31770256-7 2019 Moreover, the CYP3A5 genotyping should be considered when WZC is used to increase the blood concentration of FK506. Tacrolimus 109-114 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 14-20 31661769-1 2019 Previous studies demonstrated that the 52-kDa FK506-binding protein (FKBP52) proline-rich loop is functionally relevant in the regulation of steroid hormone receptor activity. Tacrolimus 46-51 FKBP prolyl isomerase 4 Homo sapiens 69-75 31385180-1 2019 PURPOSE: In large observational studies of adult kidney transplant recipients (KTRs) where older adults (65 years old and older) were not well represented, the mammalian target of rapamycin inhibitors (mTOR inhibitors) has poorer outcomes than the standard tacrolimus-mycophenolate-steroids (TAC-MPA-S) regimen. Tacrolimus 257-267 mechanistic target of rapamycin kinase Homo sapiens 202-206 31469511-7 2019 In an in vitro model of tacrolimus-mediated fibrogenesis by tubular cells, we found that tacrolimus treatment significantly induced mRNA and protein expression of CB1 concomitantly to col3a1 and col4a3 up regulation. Tacrolimus 24-34 cannabinoid receptor 1 Homo sapiens 163-166 31469511-7 2019 In an in vitro model of tacrolimus-mediated fibrogenesis by tubular cells, we found that tacrolimus treatment significantly induced mRNA and protein expression of CB1 concomitantly to col3a1 and col4a3 up regulation. Tacrolimus 24-34 collagen type III alpha 1 chain Homo sapiens 184-190 31469511-7 2019 In an in vitro model of tacrolimus-mediated fibrogenesis by tubular cells, we found that tacrolimus treatment significantly induced mRNA and protein expression of CB1 concomitantly to col3a1 and col4a3 up regulation. Tacrolimus 24-34 collagen type IV alpha 3 chain Homo sapiens 195-201 31469511-7 2019 In an in vitro model of tacrolimus-mediated fibrogenesis by tubular cells, we found that tacrolimus treatment significantly induced mRNA and protein expression of CB1 concomitantly to col3a1 and col4a3 up regulation. Tacrolimus 89-99 cannabinoid receptor 1 Homo sapiens 163-166 31469511-7 2019 In an in vitro model of tacrolimus-mediated fibrogenesis by tubular cells, we found that tacrolimus treatment significantly induced mRNA and protein expression of CB1 concomitantly to col3a1 and col4a3 up regulation. Tacrolimus 89-99 collagen type III alpha 1 chain Homo sapiens 184-190 31469511-7 2019 In an in vitro model of tacrolimus-mediated fibrogenesis by tubular cells, we found that tacrolimus treatment significantly induced mRNA and protein expression of CB1 concomitantly to col3a1 and col4a3 up regulation. Tacrolimus 89-99 collagen type IV alpha 3 chain Homo sapiens 195-201 31471736-0 2019 Successful Treatment of PAPA Syndrome with Dual Adalimumab and Tacrolimus Therapy. Tacrolimus 63-73 pappalysin 1 Homo sapiens 24-28 30587068-0 2019 Effects of MDR1 1236C > T-2677G > T-3435C > T polymorphisms on the intracellular accumulation of tacrolimus, cyclosporine A, sirolimus and everolimus. Tacrolimus 106-116 ATP-binding cassette, sub-family B (MDR/TAP), member 1 Sus scrofa 11-15 30587068-9 2019 MDR1 overexpression increased the resistance of LLC-PK1 cells to tacrolimus, cyclosporine A, sirolimus and everolimus. Tacrolimus 65-75 ATP-binding cassette, sub-family B (MDR/TAP), member 1 Sus scrofa 0-4 30587068-15 2019 These findings indicate that wild-type MDR1 exports tacrolimus and sirolimus more efficiency than the MDR1T-T-T, MDR1C-T-T, MDR1C-G-T variant protein. Tacrolimus 52-62 ATP-binding cassette, sub-family B (MDR/TAP), member 1 Sus scrofa 39-43 31488544-3 2019 Although AIPL1 and PDE6 are known to interact via the FK506-binding protein domain of AIPL1, the contribution of the tetratricopeptide repeat (TPR) domain of AIPL1 to its chaperone function is poorly understood. Tacrolimus 54-59 aryl hydrocarbon receptor interacting protein like 1 Homo sapiens 86-91 31616470-0 2019 The Impact of CYP3A4*22 on Tacrolimus Pharmacokinetics and Outcome in Clinical Practice at a Single Kidney Transplant Center. Tacrolimus 27-37 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-20 31442451-5 2019 FK506-mediated inhibition of the calcineurin-NFAT pathway in the HL-1 cells selectively inhibited the stimulatory effect of the conditioned medium derived from ET-1-pre-stimulated endothelial cells on cardiomyocyte fetal gene expression. Tacrolimus 0-5 endothelin 1 Mus musculus 160-164 30953600-6 2019 CYP3A5*3 (rs776746) was associated with higher dose-normalized tacrolimus troughs in all groups but occurred at different allele frequencies and had differing effect sizes. Tacrolimus 63-73 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 31588879-0 2019 Impact of CYP3A4/5 and ABCB1 polymorphisms on tacrolimus exposure and response in pediatric primary nephrotic syndrome. Tacrolimus 46-56 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 10-18 31588879-0 2019 Impact of CYP3A4/5 and ABCB1 polymorphisms on tacrolimus exposure and response in pediatric primary nephrotic syndrome. Tacrolimus 46-56 ATP binding cassette subfamily B member 1 Homo sapiens 23-28 31588879-1 2019 Aim: To evaluate the impact of CYP3A4*1G, CYP3A5*3 and ABCB1-C3435T polymorphisms on tacrolimus concentrations, efficacy and tolerance in pediatric primary nephrotic syndrome. Tacrolimus 85-95 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 31588879-6 2019 Conclusion: The genotypes of CYP3A4*1G and CYP3A5*3 rather than ABCB1-C3435T potentially predicted tacrolimus exposure and clinical response in pediatric primary nephrotic syndrome. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-35 31588879-6 2019 Conclusion: The genotypes of CYP3A4*1G and CYP3A5*3 rather than ABCB1-C3435T potentially predicted tacrolimus exposure and clinical response in pediatric primary nephrotic syndrome. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 43-49 31447189-2 2019 The aim of this study is to analyze the role of mTOR inhibitor add-on in tacrolimus-based kidney transplant recipients. Tacrolimus 73-83 mechanistic target of rapamycin kinase Homo sapiens 48-52 31447189-11 2019 CONCLUSIONS: In our preliminary result, mTOR inhibitor add-on in patients with tacrolimus-based regimen revealed less post-KT UC occurrence. Tacrolimus 79-89 mechanistic target of rapamycin kinase Homo sapiens 40-44 31581670-0 2019 A Low Tacrolimus Concentration/Dose Ratio Increases the Risk for the Development of Acute Calcineurin Inhibitor-Induced Nephrotoxicity. Tacrolimus 6-16 calcineurin binding protein 1 Homo sapiens 90-111 31581670-5 2019 A low C/D ratio (C/D ratio < 1.05 ng/mLx1/mg) was linked with higher C2 tacrolimus blood concentrations (19.2 +- 8.7 microg/L vs. 12.2 +- 5.2 microg/L respectively; p = 0.001) and higher degrees of nephrotoxicity despite comparable trough levels (6.3 +- 2.4 microg/L vs. 6.6 +- 2.2 microg/L respectively; p = 0.669). Tacrolimus 72-85 solute carrier family 22 (organic cation transporter), member 1 Mus musculus 40-44 31581670-7 2019 In renal tubular epithelial cells exposed to tacrolimus according to a fast metabolism pharmacokinetic profile it led to reduced viability and increased Fn14 expression. Tacrolimus 45-55 TNF receptor superfamily member 12A Homo sapiens 153-157 31922058-0 2020 Melding Pharmacogenomic Effect of MDR1 and CYP3A5 Gene Polymorphism on Tacrolimus Dosing in Renal Transplant Recipients in Northern India. Tacrolimus 71-81 ATP binding cassette subfamily B member 1 Homo sapiens 34-38 31922058-0 2020 Melding Pharmacogenomic Effect of MDR1 and CYP3A5 Gene Polymorphism on Tacrolimus Dosing in Renal Transplant Recipients in Northern India. Tacrolimus 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 43-49 31922058-8 2020 We analyzed the blending effect of mutant SNPs of the MDR gene and CYP3A5 for optimized TAC levels. Tacrolimus 88-91 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-73 31922058-9 2020 Results: Among CYP3A5 genotypic variants, the dose-adjusted TAC level was significantly lower, and the TAC dose required to achieve the target level was significantly higher, in CYP3A5*1*1 (expressor) than that of CYP3A5*1*3 and CYP3A5*3*3. Tacrolimus 60-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 31922058-9 2020 Results: Among CYP3A5 genotypic variants, the dose-adjusted TAC level was significantly lower, and the TAC dose required to achieve the target level was significantly higher, in CYP3A5*1*1 (expressor) than that of CYP3A5*1*3 and CYP3A5*3*3. Tacrolimus 60-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 178-184 31922058-9 2020 Results: Among CYP3A5 genotypic variants, the dose-adjusted TAC level was significantly lower, and the TAC dose required to achieve the target level was significantly higher, in CYP3A5*1*1 (expressor) than that of CYP3A5*1*3 and CYP3A5*3*3. Tacrolimus 60-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 178-184 31922058-9 2020 Results: Among CYP3A5 genotypic variants, the dose-adjusted TAC level was significantly lower, and the TAC dose required to achieve the target level was significantly higher, in CYP3A5*1*1 (expressor) than that of CYP3A5*1*3 and CYP3A5*3*3. Tacrolimus 60-63 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 178-184 31922058-9 2020 Results: Among CYP3A5 genotypic variants, the dose-adjusted TAC level was significantly lower, and the TAC dose required to achieve the target level was significantly higher, in CYP3A5*1*1 (expressor) than that of CYP3A5*1*3 and CYP3A5*3*3. Tacrolimus 103-106 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 15-21 31922058-9 2020 Results: Among CYP3A5 genotypic variants, the dose-adjusted TAC level was significantly lower, and the TAC dose required to achieve the target level was significantly higher, in CYP3A5*1*1 (expressor) than that of CYP3A5*1*3 and CYP3A5*3*3. Tacrolimus 103-106 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 178-184 31922058-9 2020 Results: Among CYP3A5 genotypic variants, the dose-adjusted TAC level was significantly lower, and the TAC dose required to achieve the target level was significantly higher, in CYP3A5*1*1 (expressor) than that of CYP3A5*1*3 and CYP3A5*3*3. Tacrolimus 103-106 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 178-184 31922058-9 2020 Results: Among CYP3A5 genotypic variants, the dose-adjusted TAC level was significantly lower, and the TAC dose required to achieve the target level was significantly higher, in CYP3A5*1*1 (expressor) than that of CYP3A5*1*3 and CYP3A5*3*3. Tacrolimus 103-106 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 178-184 31922058-10 2020 Of the MDR1 gene SNPs, only the G2677T/A homozygous mutant was significantly associated with TAC level, and it was strongly correlated with P-gp expression.The daily TAC dose requirement was highest with a combination of CYP3A5*1*1 and homozygous mutant TT+AA genotype of G2677T/A, and was lowest with CYP3A5*3*3 and wild-type GG of the G2677T/A genotype. Tacrolimus 166-169 ATP binding cassette subfamily B member 1 Homo sapiens 7-11 31922058-10 2020 Of the MDR1 gene SNPs, only the G2677T/A homozygous mutant was significantly associated with TAC level, and it was strongly correlated with P-gp expression.The daily TAC dose requirement was highest with a combination of CYP3A5*1*1 and homozygous mutant TT+AA genotype of G2677T/A, and was lowest with CYP3A5*3*3 and wild-type GG of the G2677T/A genotype. Tacrolimus 166-169 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 221-227 31922058-10 2020 Of the MDR1 gene SNPs, only the G2677T/A homozygous mutant was significantly associated with TAC level, and it was strongly correlated with P-gp expression.The daily TAC dose requirement was highest with a combination of CYP3A5*1*1 and homozygous mutant TT+AA genotype of G2677T/A, and was lowest with CYP3A5*3*3 and wild-type GG of the G2677T/A genotype. Tacrolimus 166-169 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 302-308 31922058-11 2020 Conclusion: Both CYP gene and MDR1 gene polymorphism affect TAC dose requirements, and there is a need to look for both in an individual to achieve the target trough concentration. Tacrolimus 60-63 peptidylprolyl isomerase G Homo sapiens 17-20 31922058-11 2020 Conclusion: Both CYP gene and MDR1 gene polymorphism affect TAC dose requirements, and there is a need to look for both in an individual to achieve the target trough concentration. Tacrolimus 60-63 ATP binding cassette subfamily B member 1 Homo sapiens 30-34 31616470-1 2019 Background: Although there is evidence that the CYP3A4*22 variant should be considered in tacrolimus dosing in renal transplantation, its impact beyond tacrolimus dose requirements remains controversial. Tacrolimus 90-100 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-54 31616470-12 2019 Conclusion: At our transplantation center, both CYP3A5*3 and, to a lesser extent, CYP3A4*22 affect tacrolimus pharmacokinetics early after onset of therapy with consequences for steady-state treatment in routine clinical practice. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 48-54 31616470-12 2019 Conclusion: At our transplantation center, both CYP3A5*3 and, to a lesser extent, CYP3A4*22 affect tacrolimus pharmacokinetics early after onset of therapy with consequences for steady-state treatment in routine clinical practice. Tacrolimus 99-109 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 82-88 31583120-0 2019 The splicing FK506-binding protein-51 isoform plays a role in glioblastoma resistance through programmed cell death ligand-1 expression regulation. Tacrolimus 13-18 CD274 antigen Mus musculus 94-124 31537789-0 2019 Harnessing calcineurin-FK506-FKBP12 crystal structures from invasive fungal pathogens to develop antifungal agents. Tacrolimus 23-28 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 29-35 31490997-8 2019 Using FKBP51 as a model system, we show that two amino acids lining the FK506-binding site are essential for binding Glmn and that the FKBP51-Glmn interaction is blocked by FKBP ligands. Tacrolimus 72-77 glomulin, FKBP associated protein Homo sapiens 117-121 31362893-6 2019 In this study, we administered tacrolimus to barttin hypomorphic (Bsndneo/neo) mice, a murine model of type 4 BS that exhibits polyuria, hypokalemia, and metabolic alkalosis. Tacrolimus 31-41 barttin CLCNK type accessory beta subunit Mus musculus 45-52 31362893-8 2019 Furthermore, after treatment with tacrolimus, Bsndneo/neo mice increased levels of phosphorylated NCC and NKCC2. Tacrolimus 34-44 solute carrier family 12, member 1 Mus musculus 106-111 31572289-11 2019 Besides, FK506 also significantly reduced the levels of factors involved in inflammatory response such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), tumor necrosis factor-alpha (TNF-alpha), and Protein Kinase C delta (PKCdelta) that rise after epilepsy. Tacrolimus 9-14 vascular cell adhesion molecule 1 Rattus norvegicus 106-139 31572289-11 2019 Besides, FK506 also significantly reduced the levels of factors involved in inflammatory response such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), tumor necrosis factor-alpha (TNF-alpha), and Protein Kinase C delta (PKCdelta) that rise after epilepsy. Tacrolimus 9-14 vascular cell adhesion molecule 1 Rattus norvegicus 141-147 31572289-11 2019 Besides, FK506 also significantly reduced the levels of factors involved in inflammatory response such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), tumor necrosis factor-alpha (TNF-alpha), and Protein Kinase C delta (PKCdelta) that rise after epilepsy. Tacrolimus 9-14 intercellular adhesion molecule 1 Rattus norvegicus 150-183 31572289-11 2019 Besides, FK506 also significantly reduced the levels of factors involved in inflammatory response such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), tumor necrosis factor-alpha (TNF-alpha), and Protein Kinase C delta (PKCdelta) that rise after epilepsy. Tacrolimus 9-14 intercellular adhesion molecule 1 Rattus norvegicus 185-191 31572289-11 2019 Besides, FK506 also significantly reduced the levels of factors involved in inflammatory response such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), tumor necrosis factor-alpha (TNF-alpha), and Protein Kinase C delta (PKCdelta) that rise after epilepsy. Tacrolimus 9-14 tumor necrosis factor Rattus norvegicus 194-221 31572289-11 2019 Besides, FK506 also significantly reduced the levels of factors involved in inflammatory response such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), tumor necrosis factor-alpha (TNF-alpha), and Protein Kinase C delta (PKCdelta) that rise after epilepsy. Tacrolimus 9-14 tumor necrosis factor Rattus norvegicus 223-232 31572289-11 2019 Besides, FK506 also significantly reduced the levels of factors involved in inflammatory response such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), tumor necrosis factor-alpha (TNF-alpha), and Protein Kinase C delta (PKCdelta) that rise after epilepsy. Tacrolimus 9-14 protein kinase C, delta Rattus norvegicus 239-261 31572289-11 2019 Besides, FK506 also significantly reduced the levels of factors involved in inflammatory response such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), tumor necrosis factor-alpha (TNF-alpha), and Protein Kinase C delta (PKCdelta) that rise after epilepsy. Tacrolimus 9-14 protein kinase C, delta Rattus norvegicus 263-271 31490997-8 2019 Using FKBP51 as a model system, we show that two amino acids lining the FK506-binding site are essential for binding Glmn and that the FKBP51-Glmn interaction is blocked by FKBP ligands. Tacrolimus 72-77 glomulin, FKBP associated protein Homo sapiens 142-146 31102573-11 2019 In conclusion, donor CYP3A5 genotype, time after transplant, and ALT values are associated with tacrolimus disposition between 1 month and 2 years after transplant. Tacrolimus 96-106 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 21-27 31077425-10 2019 CYP3A5*1/*1 patients who have high risks of DGF should be closely monitored because of an increased risk of DGF and reduced glomerular filtration rate with high tacrolimus doses. Tacrolimus 161-171 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 31483851-0 2019 Retraction: Novel Single Nucleotide Polymorphisms in Interleukin 6 Affect Tacrolimus Metabolism in Liver Transplant Patients. Tacrolimus 74-84 interleukin 6 Homo sapiens 53-66 31219197-4 2019 At 3 months post-transplant modelling revealed an association between tacrolimus Css and recipient CASP1 rs580523 genotype (P = 0.005), accounting for 52% Css variance. Tacrolimus 70-80 caspase 1 Homo sapiens 99-104 31219197-6 2019 As this is the first report of CASP1 genetic variability influencing tacrolimus Css , further validation in larger cohorts is required. Tacrolimus 69-79 caspase 1 Homo sapiens 31-36 30597187-0 2019 Sirolimus vs mycophenolate moftile in Tacrolimus based therapy following induction with Antithymocyte globulin promotes regulatory T cell expansion and inhibits RORgammat and T-bet expression in kidney transplantation. Tacrolimus 38-48 T-box transcription factor 21 Homo sapiens 175-180 30597187-8 2019 Patients who received Tac/MMF had significantly higher CD4+ CD25+ FOXP3+ Treg cells compared to patients who received Tac/SRL. Tacrolimus 22-25 CD4 molecule Homo sapiens 55-58 30597187-8 2019 Patients who received Tac/MMF had significantly higher CD4+ CD25+ FOXP3+ Treg cells compared to patients who received Tac/SRL. Tacrolimus 22-25 forkhead box P3 Homo sapiens 66-71 30597187-10 2019 FOXP3 mRNA levels in the patients who received Tac/MMF were increased 4 months after transplantation and the expression was significantly higher than patients who received Tac/SRL. Tacrolimus 47-50 forkhead box P3 Homo sapiens 0-5 31490380-0 2019 Tacrolimus-induced diabetic ketoacidosis with subsequent rapid recovery of endogenous insulin secretion after cessation of tacrolimus: A case report with review of literature. Tacrolimus 0-10 insulin Homo sapiens 86-93 31490380-1 2019 RATIONALE: Immunosuppressive agents such as tacrolimus (TAC) and cyclosporin might cause glycemic disorders by suppressing insulin production. Tacrolimus 44-54 insulin Homo sapiens 123-130 31454358-0 2019 Retraction: Association of MDR1 Gene SNPs and Haplotypes with the Tacrolimus Dose Requirements in Han Chinese Liver Transplant Recipients. Tacrolimus 66-76 ATP binding cassette subfamily B member 1 Homo sapiens 27-31 31402251-0 2019 Insulin Resistance in Nonobese Renal Allograft Recipients on Maintenance Doses of Cyclosporine or Tacrolimus. Tacrolimus 98-108 insulin Homo sapiens 0-7 31321978-2 2019 Seven new FK506 analogues modified at both the FK506-binding protein 12- and the calcineurin-binding regions were biosynthesized. Tacrolimus 10-15 FKBP prolyl isomerase 1A Homo sapiens 47-71 31321978-7 2019 In humans, FK506 binds to FK506-binding protein (FKBP) 12, and the resulting FKBP12-FK506 complex interacts with a Ca2+-calmodulin-dependent phosphatase, calcineurin (CaN). Tacrolimus 11-16 FKBP prolyl isomerase 1A Homo sapiens 26-57 31321978-7 2019 In humans, FK506 binds to FK506-binding protein (FKBP) 12, and the resulting FKBP12-FK506 complex interacts with a Ca2+-calmodulin-dependent phosphatase, calcineurin (CaN). Tacrolimus 11-16 FKBP prolyl isomerase 1A Homo sapiens 77-83 31321978-7 2019 In humans, FK506 binds to FK506-binding protein (FKBP) 12, and the resulting FKBP12-FK506 complex interacts with a Ca2+-calmodulin-dependent phosphatase, calcineurin (CaN). Tacrolimus 26-31 FKBP prolyl isomerase 1A Homo sapiens 77-83 31321978-8 2019 Inactivation of CaN by forming the FKBP12-FK506-CaN ternary complex prevents the activation of nuclear factor of activated T cells (NF-AT), inhibiting the production of interleukin-2 and subsequent T-cell proliferation. Tacrolimus 42-47 FKBP prolyl isomerase 1A Homo sapiens 35-41 31321978-8 2019 Inactivation of CaN by forming the FKBP12-FK506-CaN ternary complex prevents the activation of nuclear factor of activated T cells (NF-AT), inhibiting the production of interleukin-2 and subsequent T-cell proliferation. Tacrolimus 42-47 interleukin 2 Homo sapiens 169-182 31321978-10 2019 Therefore, the synthesis of FK506 analogues that can discriminate human FKBP12/CaN from its fungal counterparts may separate antifungal activity from the immunosuppressive activity, thereby allowing the development of a novel antifungal agent. Tacrolimus 28-33 FKBP prolyl isomerase 1A Homo sapiens 72-78 31400753-0 2019 Ginseng increases Klotho expression by FoxO3-mediated manganese superoxide dismutase in a mouse model of tacrolimus-induced renal injury. Tacrolimus 105-115 klotho Mus musculus 18-24 31531197-0 2019 One-step Heck Reaction Generates Nonimmunosuppressive FK506 Analogs for Pharmacological BMP Activation. Tacrolimus 54-59 bone morphogenetic protein 1 Homo sapiens 88-91 31531197-1 2019 FKBP12 ligands such as FK506 have been shown to activate the BMP signaling pathway and facilitate tissue regeneration. Tacrolimus 23-28 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 0-6 31531197-1 2019 FKBP12 ligands such as FK506 have been shown to activate the BMP signaling pathway and facilitate tissue regeneration. Tacrolimus 23-28 bone morphogenetic protein 1 Homo sapiens 61-64 31409869-0 2019 Model based development of tacrolimus dosing algorithm considering CYP3A5 genotypes and mycophenolate mofetil drug interaction in stable kidney transplant recipients. Tacrolimus 27-37 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-73 31400753-0 2019 Ginseng increases Klotho expression by FoxO3-mediated manganese superoxide dismutase in a mouse model of tacrolimus-induced renal injury. Tacrolimus 105-115 forkhead box O3 Mus musculus 39-44 31400753-3 2019 Although tacrolimus treatment reduced Klotho level in the serum and kidney, ginseng treatment was found to reverse the levels. Tacrolimus 9-19 klotho Mus musculus 38-44 31400753-5 2019 Effect of ginseng on Klotho-induced manganese superoxide dismutase signaling pathway during tacrolimus treatment in mice revealed that ginseng suppressed phosphatidylinositol 3-kinase/serine-threonine kinase Akt-mediated phosphorylation of forkhead box protein O3a and promoted the binding of forkhead box protein O3a to manganese superoxide dismutase promoter. Tacrolimus 92-102 klotho Mus musculus 21-27 31400753-7 2019 These findings together suggested that ginseng attenuated tacrolimus-induced oxidative stress via signaling between Klotho and the phosphatidylinositol 3-kinase/serine-threonine kinase Akt/forkhead box protein O3a-related antioxidant pathway. Tacrolimus 58-68 klotho Mus musculus 116-122 30471066-3 2019 Although cytochrome P450 (CYP) 3A5 and multi-drug resistance 1 (MDR1) polymorphisms influence tacrolimus concentrations, it is unknown if these impact on conversion. Tacrolimus 94-104 ATP binding cassette subfamily B member 1 Homo sapiens 39-62 31087501-0 2019 A Minimal Physiologically-Based Pharmacokinetic Model for Tacrolimus in Living-Donor Liver Transplantation: Perspectives Related to Liver Regeneration and the cytochrome P450 3A5 (CYP3A5) Genotype. Tacrolimus 58-68 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 180-186 31087501-1 2019 In adult patients after living-donor liver transplantation, postoperative days and the cytochrome P450 3A5 (CYP3A5) genotype are known to affect tacrolimus pharmacokinetics. Tacrolimus 145-155 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 87-106 31087501-1 2019 In adult patients after living-donor liver transplantation, postoperative days and the cytochrome P450 3A5 (CYP3A5) genotype are known to affect tacrolimus pharmacokinetics. Tacrolimus 145-155 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 108-114 31087501-2 2019 In this study, we constructed a physiologically-based pharmacokinetic model adapted to the clinical data and evaluated the contribution of liver regeneration as well as hepatic and intestine CYP3A5 genotypes on tacrolimus pharmacokinetics. Tacrolimus 211-221 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 191-197 31087501-5 2019 In conclusion, the constructed physiologically-based pharmacokinetic model clarified that the oral clearance of tacrolimus was affected by the CYP3A5 genotypes in both the liver and intestine to the same extent. Tacrolimus 112-122 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 143-149 31190414-3 2019 Tac is metabolized in the liver and intestine by the cytochrome P450 3A (CYP3A) isoforms CYP3A4 and CYP3A5. Tacrolimus 0-3 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 53-71 31190414-3 2019 Tac is metabolized in the liver and intestine by the cytochrome P450 3A (CYP3A) isoforms CYP3A4 and CYP3A5. Tacrolimus 0-3 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 73-78 31190414-3 2019 Tac is metabolized in the liver and intestine by the cytochrome P450 3A (CYP3A) isoforms CYP3A4 and CYP3A5. Tacrolimus 0-3 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 89-95 31190414-3 2019 Tac is metabolized in the liver and intestine by the cytochrome P450 3A (CYP3A) isoforms CYP3A4 and CYP3A5. Tacrolimus 0-3 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 100-106 30471066-3 2019 Although cytochrome P450 (CYP) 3A5 and multi-drug resistance 1 (MDR1) polymorphisms influence tacrolimus concentrations, it is unknown if these impact on conversion. Tacrolimus 94-104 ATP binding cassette subfamily B member 1 Homo sapiens 64-68 30471066-9 2019 A decrease in tacrolimus exposure (median 21%) was only evident among CYP3A5 expressors [227 (172-366) vs. 180 (104-347) ng h/mL, p = 0.014, n = 18], not among non-expressors [215 (197-290) vs. 217 (129-281) ng h/mL, p = 0.263, n = 8]. Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 70-76 30471066-12 2019 CONCLUSION: The high prevalence of CYP3A5 polymorphism among Asians may lead to a significant reduction in tacrolimus exposure with 1:1 dose conversion of Prograf to Advagraf . Tacrolimus 107-117 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 35-41 31201087-13 2019 There was an inverse relationship between FLT3 expression and tacrolimus levels (-0.029 and -0.176, respectively) in Caucasians and AAs. Tacrolimus 62-72 fms related receptor tyrosine kinase 3 Homo sapiens 42-46 30883749-1 2019 Tacrolimus (TAC) acts as an inhibitor of calcineurin, which inhibits the production of interleukin-2. Tacrolimus 0-10 interleukin 2 Homo sapiens 87-100 30883749-1 2019 Tacrolimus (TAC) acts as an inhibitor of calcineurin, which inhibits the production of interleukin-2. Tacrolimus 12-15 interleukin 2 Homo sapiens 87-100 31201731-0 2019 Influence of Tacrolimus on Depressive-Like Behavior in Diabetic Rats Through Brain-Derived Neurotrophic Factor Regulation in the Hippocampus. Tacrolimus 13-23 brain-derived neurotrophic factor Rattus norvegicus 77-110 31201731-2 2019 The aim of the present study was to investigate the influence of tacrolimus on brain-derived neurotrophic factor (BDNF), the critical protein for maintenance of neuronal functions, in the hippocampus in a diabetic condition. Tacrolimus 65-75 brain-derived neurotrophic factor Rattus norvegicus 79-112 31201731-2 2019 The aim of the present study was to investigate the influence of tacrolimus on brain-derived neurotrophic factor (BDNF), the critical protein for maintenance of neuronal functions, in the hippocampus in a diabetic condition. Tacrolimus 65-75 brain-derived neurotrophic factor Rattus norvegicus 114-118 31201731-6 2019 There was a significant decrease of BDNF expression in the DG and CA3 region in tacrolimus-treated and diabetic rats compared with that of the control group injected with vehicle only. Tacrolimus 80-90 brain-derived neurotrophic factor Rattus norvegicus 36-40 31201731-6 2019 There was a significant decrease of BDNF expression in the DG and CA3 region in tacrolimus-treated and diabetic rats compared with that of the control group injected with vehicle only. Tacrolimus 80-90 carbonic anhydrase 3 Rattus norvegicus 66-69 31201731-8 2019 Tacrolimus treatment in diabetic rats further decreased the BDNF expression level in the DG and CA3 region. Tacrolimus 0-10 brain-derived neurotrophic factor Rattus norvegicus 60-64 31201731-8 2019 Tacrolimus treatment in diabetic rats further decreased the BDNF expression level in the DG and CA3 region. Tacrolimus 0-10 carbonic anhydrase 3 Rattus norvegicus 96-99 31201731-9 2019 Interestingly, mossy fiber sprouting, demonstrated by prominent punctate immunolabeling of BDNF with synaptoporin, was observed in the diabetic group treated with tacrolimus, which localized at the stratum oriens of the CA3 region. Tacrolimus 163-173 brain-derived neurotrophic factor Rattus norvegicus 91-95 31201731-9 2019 Interestingly, mossy fiber sprouting, demonstrated by prominent punctate immunolabeling of BDNF with synaptoporin, was observed in the diabetic group treated with tacrolimus, which localized at the stratum oriens of the CA3 region. Tacrolimus 163-173 carbonic anhydrase 3 Rattus norvegicus 220-223 31201731-10 2019 These data suggest that tacrolimus treatment or a diabetic condition decreases BDNF expression in the hippocampus, and that tacrolimus treatment in the diabetic condition further injures the CA3 region of the hippocampus. Tacrolimus 24-34 brain-derived neurotrophic factor Rattus norvegicus 79-83 31201731-10 2019 These data suggest that tacrolimus treatment or a diabetic condition decreases BDNF expression in the hippocampus, and that tacrolimus treatment in the diabetic condition further injures the CA3 region of the hippocampus. Tacrolimus 124-134 carbonic anhydrase 3 Rattus norvegicus 191-194 31201731-11 2019 In addition to BDNF expression, decreased locomotor activity and evident depressive behavior were observed in tacrolimus-treated diabetic rats. Tacrolimus 110-120 brain-derived neurotrophic factor Rattus norvegicus 15-19 31077643-11 2019 The patients with the homozygous CYP3A5*3 had the higher blood tacrolimus concentration, while CYP3A5*3 allele was not associated with the serum levels of 4beta-OHC and 25-OHD. Tacrolimus 63-73 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 33-39 31230376-1 2019 INTRODUCTION: One factor impacting tacrolimus interpatient variability is the presence of CYP3A5 polymorphisms. Tacrolimus 35-45 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 90-96 31015081-1 2019 We present a case of a 65-year-old African American male, immunosuppressed on Tacrolimus, who initially presented with cerebellar ataxia and rapidly developed Progressive Encephalomyelitis with Rigidity and Myoclonus (PERM) with positive anti-glutamic acid decarboxylase (GAD65) antibodies, no underlying malignancy, and normal neuroimaging. Tacrolimus 78-88 glutamate decarboxylase 1 Homo sapiens 238-270 31015081-1 2019 We present a case of a 65-year-old African American male, immunosuppressed on Tacrolimus, who initially presented with cerebellar ataxia and rapidly developed Progressive Encephalomyelitis with Rigidity and Myoclonus (PERM) with positive anti-glutamic acid decarboxylase (GAD65) antibodies, no underlying malignancy, and normal neuroimaging. Tacrolimus 78-88 glutamate decarboxylase 2 Homo sapiens 272-277 30887101-6 2019 A knockout of the D-lactate dehydrogenase gene, combined with the overexpression of tryptophane synthase and aspartate 1-decarboxylase genes, led to a 29.8% enhancement of tacrolimus production compared to the parent strain. Tacrolimus 172-182 lactate dehydrogenase D Homo sapiens 18-41 30887101-6 2019 A knockout of the D-lactate dehydrogenase gene, combined with the overexpression of tryptophane synthase and aspartate 1-decarboxylase genes, led to a 29.8% enhancement of tacrolimus production compared to the parent strain. Tacrolimus 172-182 glutamate decarboxylase like 1 Homo sapiens 109-134 31124575-0 2019 CYP3A5 genotype affects time to therapeutic tacrolimus level in pediatric kidney transplant recipients. Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 31124575-2 2019 Tacrolimus (TAC) dose requirements are significantly impacted by genetic variation in CYP3A5 polymorphisms, however the impact that genotype has on clinical outcomes in the pediatric kidney transplant population remains unclear. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 86-92 31467711-3 2019 In this study, we investigated the effective dose and injection time of FK506 as an immunophilin ligand for providing a suitable effect on cells of CA2, CA3, and dentate gyrus of the hippocampus. Tacrolimus 72-77 carbonic anhydrase 2 Rattus norvegicus 148-151 31467711-3 2019 In this study, we investigated the effective dose and injection time of FK506 as an immunophilin ligand for providing a suitable effect on cells of CA2, CA3, and dentate gyrus of the hippocampus. Tacrolimus 72-77 carbonic anhydrase 3 Rattus norvegicus 153-156 30831086-9 2019 Compared with the epilepsy group, the levels of NO, NOS (including nNOS and iNOS), and MDA were obviously decreased by FK506 (P < 0.05). Tacrolimus 119-124 nitric oxide synthase 1 Rattus norvegicus 67-71 30831086-9 2019 Compared with the epilepsy group, the levels of NO, NOS (including nNOS and iNOS), and MDA were obviously decreased by FK506 (P < 0.05). Tacrolimus 119-124 nitric oxide synthase 2 Rattus norvegicus 76-80 30831086-11 2019 Besides, compared with the epilepsy group, FK506 significantly increased the AIF level in the mitochondrial, but decreased that in the nuclear fractions, respectively (P < 0.05). Tacrolimus 43-48 apoptosis inducing factor, mitochondria associated 1 Rattus norvegicus 77-80 31423060-0 2019 Influence of CYP3A5 and ABCB1 Polymorphism on Tacrolimus Drug Dosing in South Indian Renal Allograft Recipients. Tacrolimus 46-56 ATP binding cassette subfamily B member 1 Homo sapiens 24-29 31423060-1 2019 Introduction: Tacrolimus blood levels are influenced by polymorphisms involving Cytochrome 3A subfamily (CYP3A5) and P-Glycoprotein (ABCB-1) genes. Tacrolimus 14-24 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 105-111 31423060-1 2019 Introduction: Tacrolimus blood levels are influenced by polymorphisms involving Cytochrome 3A subfamily (CYP3A5) and P-Glycoprotein (ABCB-1) genes. Tacrolimus 14-24 ATP binding cassette subfamily B member 1 Homo sapiens 117-131 31423060-1 2019 Introduction: Tacrolimus blood levels are influenced by polymorphisms involving Cytochrome 3A subfamily (CYP3A5) and P-Glycoprotein (ABCB-1) genes. Tacrolimus 14-24 ATP binding cassette subfamily B member 1 Homo sapiens 133-139 31423060-4 2019 Methods: An analysis of CYP3A5, ABCB1 genotype done in 101 renal transplant recipients by polymerase chain reaction was correlated with blood tacrolimus trough levels (CLIA method), weight, concentration/dose (L/D) ratio, incidence of biopsy proven early acute rejections, and tacrolimus toxicity. Tacrolimus 142-152 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-30 31423060-4 2019 Methods: An analysis of CYP3A5, ABCB1 genotype done in 101 renal transplant recipients by polymerase chain reaction was correlated with blood tacrolimus trough levels (CLIA method), weight, concentration/dose (L/D) ratio, incidence of biopsy proven early acute rejections, and tacrolimus toxicity. Tacrolimus 142-152 ATP binding cassette subfamily B member 1 Homo sapiens 32-37 31423060-4 2019 Methods: An analysis of CYP3A5, ABCB1 genotype done in 101 renal transplant recipients by polymerase chain reaction was correlated with blood tacrolimus trough levels (CLIA method), weight, concentration/dose (L/D) ratio, incidence of biopsy proven early acute rejections, and tacrolimus toxicity. Tacrolimus 277-287 ATP binding cassette subfamily B member 1 Homo sapiens 32-37 31423060-8 2019 Incidence of early acute rejections (30% vs. 9.76%; P 0.016) and tacrolimus-related toxicity (14.6% vs. 5%; P 0.039) were significantly higher in CYP3A5 expressers and nonexpressers, respectively. Tacrolimus 65-75 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 146-152 31423060-11 2019 Conclusions: CYP3A5 polymorphisms correlated with tacrolimus dose requirements and blood levels, incidence of early acute rejection, and tacrolimus nephrotoxicity. Tacrolimus 50-60 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 31423060-11 2019 Conclusions: CYP3A5 polymorphisms correlated with tacrolimus dose requirements and blood levels, incidence of early acute rejection, and tacrolimus nephrotoxicity. Tacrolimus 137-147 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 13-19 31423060-12 2019 CYP3A5 polymorphism analysis prior to renal transplant will aid more precise early tacrolimus dose calculation to balance between rejection and toxicity. Tacrolimus 83-93 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 0-6 30124124-0 2019 Tacrolimus for the treatment of immune-related adverse effects refractory to systemic steroids and anti-tumor necrosis factor alpha therapy. Tacrolimus 0-10 tumor necrosis factor Homo sapiens 104-131 30124124-7 2019 Presented here are three patient cases supporting the use of the calcinuerin inhibitor tacrolimus to treat immune-related adverse effects refractory to corticosteroids and anti-tumor necrosis factor alpha. Tacrolimus 87-97 tumor necrosis factor Homo sapiens 177-204 31087207-0 2019 Axonal Protection by Tacrolimus with Inhibition of NFATc1 in TNF-Induced Optic Nerve Degeneration. Tacrolimus 21-31 tumor necrosis factor Homo sapiens 61-64 31087207-7 2019 A significant increase in TNF protein level was observed in optic nerve 14 days after TNF injection and this increase was prevented by tacrolimus. Tacrolimus 135-145 tumor necrosis factor Homo sapiens 26-29 31087207-7 2019 A significant increase in TNF protein level was observed in optic nerve 14 days after TNF injection and this increase was prevented by tacrolimus. Tacrolimus 135-145 tumor necrosis factor Homo sapiens 86-89 31087207-10 2019 Treatment of tacrolimus significantly ameliorated the TNF-mediated axonal loss. Tacrolimus 13-23 tumor necrosis factor Homo sapiens 54-57 31087207-11 2019 These results suggest that tacrolimus is neuroprotective against axon loss in TNF-induced optic neuropathy and that the effect arises from suppression of the CaN/NFATc1 pathway. Tacrolimus 27-37 tumor necrosis factor Homo sapiens 78-81 31087207-11 2019 These results suggest that tacrolimus is neuroprotective against axon loss in TNF-induced optic neuropathy and that the effect arises from suppression of the CaN/NFATc1 pathway. Tacrolimus 27-37 nuclear factor of activated T cells 1 Homo sapiens 162-168 30058048-0 2019 Drug-metabolizing enzymes CYP3A as a link between tacrolimus and vitamin D in renal transplant recipients: is it relevant in clinical practice? Tacrolimus 50-60 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 26-31 30058048-1 2019 CYP3A enzymes are involved in the metabolism of calcineurin inhibitor tacrolimus as well as vitamin D. Tacrolimus 70-80 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-5 30058048-2 2019 In this review, we summarize the clinical aspects of CYP3A-mediated metabolism of tacrolimus and vitamin D with emphasis on the influence of single-nucleotide polymorphisms on tacrolimus disposition. Tacrolimus 82-92 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 53-58 30058048-2 2019 In this review, we summarize the clinical aspects of CYP3A-mediated metabolism of tacrolimus and vitamin D with emphasis on the influence of single-nucleotide polymorphisms on tacrolimus disposition. Tacrolimus 176-186 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 53-58 31242630-2 2019 The urinary neutrophil gelatinase-associated lipocalin (NGAL) level has been linked to tacrolimus-induced AKI in patients starting tacrolimus treatment the morning after liver transplantation. Tacrolimus 87-97 lipocalin 2 Homo sapiens 12-54 31261624-15 2019 In conclusion, GLP-1R could be implemented for recipients while closely monitoring their tacrolimus levels and any potential side effects. Tacrolimus 89-99 glucagon like peptide 1 receptor Homo sapiens 15-21 31242630-2 2019 The urinary neutrophil gelatinase-associated lipocalin (NGAL) level has been linked to tacrolimus-induced AKI in patients starting tacrolimus treatment the morning after liver transplantation. Tacrolimus 87-97 lipocalin 2 Homo sapiens 56-60 31242630-2 2019 The urinary neutrophil gelatinase-associated lipocalin (NGAL) level has been linked to tacrolimus-induced AKI in patients starting tacrolimus treatment the morning after liver transplantation. Tacrolimus 131-141 lipocalin 2 Homo sapiens 12-54 31242630-2 2019 The urinary neutrophil gelatinase-associated lipocalin (NGAL) level has been linked to tacrolimus-induced AKI in patients starting tacrolimus treatment the morning after liver transplantation. Tacrolimus 131-141 lipocalin 2 Homo sapiens 56-60 31242630-6 2019 HE4 levels after tacrolimus administration were significantly higher in patients who developed AKI (n = 6) than in those who did not (n = 20), whereas NGAL, MCP-1, and L-FABP levels did not differ significantly before or after tacrolimus administration. Tacrolimus 17-27 WAP four-disulfide core domain 2 Homo sapiens 0-3 31181889-2 2019 We have utilized a gene delivery vector containing inducible caspase 9 (iCasp9) gene, which is a synthetic analogue based on the mammalian caspase 9 and fused to a human FK506 binding protein that allows its conditional dimerization to a synthetic, small molecule [chemical inducer of dimerization, AP20187] and results in target cell apoptosis. Tacrolimus 170-175 caspase 9 Homo sapiens 61-70 31005256-6 2019 Inhibition of calcineurin with cyclosporine A, FK506 down-regulates the levels of NFATc3 nuclear translocation and proinflammatory cytokines expression. Tacrolimus 47-52 nuclear factor of activated T cells 3 Homo sapiens 82-88 31200653-11 2019 CL treatment decreased apoptotic cell death (decreased TUNEL-positive cells and reduced expression of active caspase-3) in TAC-treated kidney. Tacrolimus 123-126 caspase 3 Homo sapiens 109-118 30729267-0 2019 IL-2 gene polymorphisms affect tacrolimus response in myasthenia gravis. Tacrolimus 31-41 interleukin 2 Homo sapiens 0-4 31355232-11 2019 Besides, the expression levels of Runx2 and Osterix were up-regulated, and that of PPAR-gamma and C/EBP-alpha were down-regulated in diabetic rats after FK506 treatment. Tacrolimus 153-158 RUNX family transcription factor 2 Rattus norvegicus 34-39 31355232-11 2019 Besides, the expression levels of Runx2 and Osterix were up-regulated, and that of PPAR-gamma and C/EBP-alpha were down-regulated in diabetic rats after FK506 treatment. Tacrolimus 153-158 Sp7 transcription factor Rattus norvegicus 44-51 31355232-11 2019 Besides, the expression levels of Runx2 and Osterix were up-regulated, and that of PPAR-gamma and C/EBP-alpha were down-regulated in diabetic rats after FK506 treatment. Tacrolimus 153-158 peroxisome proliferator-activated receptor gamma Rattus norvegicus 83-93 31355232-11 2019 Besides, the expression levels of Runx2 and Osterix were up-regulated, and that of PPAR-gamma and C/EBP-alpha were down-regulated in diabetic rats after FK506 treatment. Tacrolimus 153-158 CCAAT/enhancer binding protein alpha Rattus norvegicus 98-109 31355232-12 2019 In addition, the nuclear translocation of beta-catenin protein levels were increased in diabetic rats after the treatment of FK506. Tacrolimus 125-130 catenin beta 1 Rattus norvegicus 42-54 30565852-3 2019 Herein we show that recipient C3aR1 deficiency or pharmacological C3aR1 blockade synergizes with tacrolimus to significantly prolong allograft survival versus tacrolimus-treated controls (median survival time 21 vs. 14 days, P < .05). Tacrolimus 159-169 complement component 3a receptor 1 Mus musculus 30-35 30565852-3 2019 Herein we show that recipient C3aR1 deficiency or pharmacological C3aR1 blockade synergizes with tacrolimus to significantly prolong allograft survival versus tacrolimus-treated controls (median survival time 21 vs. 14 days, P < .05). Tacrolimus 159-169 complement component 3a receptor 1 Mus musculus 66-71 30849430-2 2019 Tacrolimus and micelles are probed for the first time by this high spatial resolution technique by element-selective excitation in the C 1s- and O 1s-regimes. Tacrolimus 0-10 complement component 1, s subcomponent 1 Mus musculus 135-146 30729267-2 2019 However, there are no published studies examining the influence of the IL-2 gene polymorphisms on the response of myasthenia gravis (MG) patients to tacrolimus (Tac). Tacrolimus 149-159 interleukin 2 Homo sapiens 71-75 30729267-3 2019 The goal of this study was to investigate the relationship between the polymorphisms of IL-2 and Tac response in MG patients. Tacrolimus 97-100 interleukin 2 Homo sapiens 88-92 30729267-10 2019 CONCLUSION: Myasthenia gravis patients with the rs2069762 variant, rs2069762 G/T and G/G genotype, and TAGG haplotype for IL-2 tended to respond poorly to Tac treatment. Tacrolimus 155-158 interleukin 2 Homo sapiens 122-126 30877873-7 2019 Both DHMEQ and tacrolimus suppress DNCB-induced increase of serum total IgE and attenuate expression of inflammatory factors IL-4, IL-6, IL-13, IL-1beta and interferon (IFN)-gamma in the disrupted ear tissues. Tacrolimus 15-25 interleukin 4 Mus musculus 125-129 29025381-5 2019 Simultaneously, the patient had very low absolute total lymphocyte count of 70 cells/muL during which he received supratherapeutic tacrolimus at whole blood trough levels and mycophenolate mofetil. Tacrolimus 131-141 tripartite motif containing 37 Homo sapiens 85-88 30633591-5 2019 Results: It is found that percentages of CD3+, CD3+CD4+ and CD3+CD8+ T cells in FK506 group are lowered compared to the control group but they are elevated in FK506 + MMF group. Tacrolimus 80-85 CD4 molecule Homo sapiens 51-54 30633591-5 2019 Results: It is found that percentages of CD3+, CD3+CD4+ and CD3+CD8+ T cells in FK506 group are lowered compared to the control group but they are elevated in FK506 + MMF group. Tacrolimus 80-85 CD8a molecule Homo sapiens 64-67 30633591-5 2019 Results: It is found that percentages of CD3+, CD3+CD4+ and CD3+CD8+ T cells in FK506 group are lowered compared to the control group but they are elevated in FK506 + MMF group. Tacrolimus 159-164 CD8a molecule Homo sapiens 64-67 30633591-6 2019 Amount of CD4+CD25+CD127low/-Treg cells in CD3+ CD4+T cells in FK506 + MMF group was higher than that in FK506 group and control group. Tacrolimus 63-68 CD4 molecule Homo sapiens 10-13 30633591-6 2019 Amount of CD4+CD25+CD127low/-Treg cells in CD3+ CD4+T cells in FK506 + MMF group was higher than that in FK506 group and control group. Tacrolimus 63-68 CD4 molecule Homo sapiens 48-51 30633591-6 2019 Amount of CD4+CD25+CD127low/-Treg cells in CD3+ CD4+T cells in FK506 + MMF group was higher than that in FK506 group and control group. Tacrolimus 105-110 CD4 molecule Homo sapiens 10-13 30633591-6 2019 Amount of CD4+CD25+CD127low/-Treg cells in CD3+ CD4+T cells in FK506 + MMF group was higher than that in FK506 group and control group. Tacrolimus 105-110 CD4 molecule Homo sapiens 48-51 30633591-7 2019 The expressions of co-inhibitory receptors (CTLA-4, PD-1, Tim-3, LAG-3 and TIGIT) on Tregs in FK506 + MMF group were significant higher than those in the FK506 group and control group. Tacrolimus 94-99 cytotoxic T-lymphocyte associated protein 4 Homo sapiens 44-50 30633591-7 2019 The expressions of co-inhibitory receptors (CTLA-4, PD-1, Tim-3, LAG-3 and TIGIT) on Tregs in FK506 + MMF group were significant higher than those in the FK506 group and control group. Tacrolimus 94-99 programmed cell death 1 Homo sapiens 52-56 30633591-7 2019 The expressions of co-inhibitory receptors (CTLA-4, PD-1, Tim-3, LAG-3 and TIGIT) on Tregs in FK506 + MMF group were significant higher than those in the FK506 group and control group. Tacrolimus 94-99 hepatitis A virus cellular receptor 2 Homo sapiens 58-63 30633591-7 2019 The expressions of co-inhibitory receptors (CTLA-4, PD-1, Tim-3, LAG-3 and TIGIT) on Tregs in FK506 + MMF group were significant higher than those in the FK506 group and control group. Tacrolimus 94-99 lymphocyte activating 3 Homo sapiens 65-70 30877873-7 2019 Both DHMEQ and tacrolimus suppress DNCB-induced increase of serum total IgE and attenuate expression of inflammatory factors IL-4, IL-6, IL-13, IL-1beta and interferon (IFN)-gamma in the disrupted ear tissues. Tacrolimus 15-25 interleukin 6 Mus musculus 131-135 30633591-7 2019 The expressions of co-inhibitory receptors (CTLA-4, PD-1, Tim-3, LAG-3 and TIGIT) on Tregs in FK506 + MMF group were significant higher than those in the FK506 group and control group. Tacrolimus 94-99 T cell immunoreceptor with Ig and ITIM domains Homo sapiens 75-80 30877873-7 2019 Both DHMEQ and tacrolimus suppress DNCB-induced increase of serum total IgE and attenuate expression of inflammatory factors IL-4, IL-6, IL-13, IL-1beta and interferon (IFN)-gamma in the disrupted ear tissues. Tacrolimus 15-25 interleukin 13 Mus musculus 137-142 30633591-8 2019 The levels of the relative cytokines (TGF-beta and IL-10) in FK506 group are down-regulated compared to the control group. Tacrolimus 61-66 transforming growth factor beta 1 Homo sapiens 38-46 30633591-8 2019 The levels of the relative cytokines (TGF-beta and IL-10) in FK506 group are down-regulated compared to the control group. Tacrolimus 61-66 interleukin 10 Homo sapiens 51-56 30877873-7 2019 Both DHMEQ and tacrolimus suppress DNCB-induced increase of serum total IgE and attenuate expression of inflammatory factors IL-4, IL-6, IL-13, IL-1beta and interferon (IFN)-gamma in the disrupted ear tissues. Tacrolimus 15-25 interleukin 1 alpha Mus musculus 144-152 30877873-7 2019 Both DHMEQ and tacrolimus suppress DNCB-induced increase of serum total IgE and attenuate expression of inflammatory factors IL-4, IL-6, IL-13, IL-1beta and interferon (IFN)-gamma in the disrupted ear tissues. Tacrolimus 15-25 interferon gamma Mus musculus 157-179 30861159-4 2019 In wild-type CYP3A4 rs2242480 (TT) carriers, patients who took calcium channel blockers had lower tacrolimus stable doses than those without the concomitant medications (P < 1 x 10-4 ). Tacrolimus 98-108 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 13-19 30861159-6 2019 Similarly, the tacrolimus stable doses in wild-type CYP3A5 rs776746 carriers who had hypertension were higher than those without hypertension (P = 4.10 x 10-3 ). Tacrolimus 15-25 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 52-58 30861159-8 2019 Our finding suggested that wild-type CYP3A4 rs2242480 (TT) carriers should be more cautious to take tacrolimus when they are coadministrated with calcium channel blockers, and CYP3A5 rs776746 (AA) carriers may need higher tacrolimus dosage when they are in combination with hypertension. Tacrolimus 100-110 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-43 30861159-8 2019 Our finding suggested that wild-type CYP3A4 rs2242480 (TT) carriers should be more cautious to take tacrolimus when they are coadministrated with calcium channel blockers, and CYP3A5 rs776746 (AA) carriers may need higher tacrolimus dosage when they are in combination with hypertension. Tacrolimus 222-232 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-43 30861159-8 2019 Our finding suggested that wild-type CYP3A4 rs2242480 (TT) carriers should be more cautious to take tacrolimus when they are coadministrated with calcium channel blockers, and CYP3A5 rs776746 (AA) carriers may need higher tacrolimus dosage when they are in combination with hypertension. Tacrolimus 222-232 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 176-182 31261526-5 2019 The most commonly reported genotype was CYP3A53/3, which was strongly associated with cyclosporine A (CsA) and tacrolimus (FK506). Tacrolimus 111-121 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-49 30222017-6 2019 For other body locations, a TCI, either pimecrolimus 1% cream, tacrolimus 0.03% ointment in children or 0.1% ointment in adults, should be applied twice daily until clearance. Tacrolimus 63-73 latexin Homo sapiens 28-31 31261526-5 2019 The most commonly reported genotype was CYP3A53/3, which was strongly associated with cyclosporine A (CsA) and tacrolimus (FK506). Tacrolimus 123-128 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-49 31045868-12 2019 The association between CYP3A5 genotype and tacrolimus dose requirement is consistent (Grading A I). Tacrolimus 44-54 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 24-30 31003147-0 2019 Recipient ABCB1, donor and recipient CYP3A5 genotypes influence tacrolimus pharmacokinetics in liver transplant cases. Tacrolimus 64-74 ATP binding cassette subfamily B member 1 Homo sapiens 10-15 31003147-0 2019 Recipient ABCB1, donor and recipient CYP3A5 genotypes influence tacrolimus pharmacokinetics in liver transplant cases. Tacrolimus 64-74 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 31003147-2 2019 METHODS: Meta-analysis was carried out to evaluate how donor/recipient CYP3A5 (n = 678) and recipient ABCB1 (n = 318) genotypes influence tacrolimus pharmacokinetics till one-month of transplantation. Tacrolimus 138-148 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 71-77 31003147-2 2019 METHODS: Meta-analysis was carried out to evaluate how donor/recipient CYP3A5 (n = 678) and recipient ABCB1 (n = 318) genotypes influence tacrolimus pharmacokinetics till one-month of transplantation. Tacrolimus 138-148 ATP binding cassette subfamily B member 1 Homo sapiens 102-107 31003147-7 2019 CONCLUSIONS: The donor and recipient CYP3A5*3 polymorphism influences tacrolimus pharmacokinetics in the first month post-transplantation, whereas the association with recipient ABCB1 3435 C > T is inconclusive. Tacrolimus 70-80 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 37-43 31003147-7 2019 CONCLUSIONS: The donor and recipient CYP3A5*3 polymorphism influences tacrolimus pharmacokinetics in the first month post-transplantation, whereas the association with recipient ABCB1 3435 C > T is inconclusive. Tacrolimus 70-80 ATP binding cassette subfamily B member 1 Homo sapiens 178-183 31045868-16 2019 Population PK models including CYP3A5 and CYP3A4 genotypes will be considered to guide initial tacrolimus dosing. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 31-37 31045868-16 2019 Population PK models including CYP3A5 and CYP3A4 genotypes will be considered to guide initial tacrolimus dosing. Tacrolimus 95-105 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 30827938-5 2019 Further study showed that both FK506 and FKVP activate BMP signaling in multiple cell types through FKBP12 antagonism. Tacrolimus 31-36 bone morphogenetic protein 1 Homo sapiens 55-58 31214251-9 2019 In addition, we found that CYP3A5, which is known to affect the metabolism of tacrolimus, only influenced tacrolimus pharmacokinetics in the SP. Tacrolimus 78-88 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 27-33 31214251-9 2019 In addition, we found that CYP3A5, which is known to affect the metabolism of tacrolimus, only influenced tacrolimus pharmacokinetics in the SP. Tacrolimus 106-116 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 27-33 30827938-5 2019 Further study showed that both FK506 and FKVP activate BMP signaling in multiple cell types through FKBP12 antagonism. Tacrolimus 31-36 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 100-106 30659828-11 2019 Treatment with Mdivi-1 and FK506 upregulated the phosphorylation of Drp1, inhibited Drp1 translocation to mitochondria, and alleviated mitochondrial fragmentation after HIR. Tacrolimus 27-32 dynamin 1-like Mus musculus 68-72 31100260-3 2019 (2019) utilize FKBP12 ligand to demonstrate that wound healing effects of FK506 occur via activation of the BMP (bone morphogenic protein) signaling pathway. Tacrolimus 74-79 FKBP prolyl isomerase 1A pseudogene 4 Homo sapiens 15-21 31100260-3 2019 (2019) utilize FKBP12 ligand to demonstrate that wound healing effects of FK506 occur via activation of the BMP (bone morphogenic protein) signaling pathway. Tacrolimus 74-79 bone morphogenetic protein 1 Homo sapiens 108-111 31100260-3 2019 (2019) utilize FKBP12 ligand to demonstrate that wound healing effects of FK506 occur via activation of the BMP (bone morphogenic protein) signaling pathway. Tacrolimus 74-79 bone morphogenetic protein 1 Homo sapiens 113-137 30659828-11 2019 Treatment with Mdivi-1 and FK506 upregulated the phosphorylation of Drp1, inhibited Drp1 translocation to mitochondria, and alleviated mitochondrial fragmentation after HIR. Tacrolimus 27-32 dynamin 1-like Mus musculus 84-88 30659828-12 2019 What"s more, Mdivi-1 and FK506 restrained cytochrome c release and cleaved caspase-3 expression, ameliorated hippocampal neurons apoptosis, and decreased serum S100beta/NSE concentrations as well. Tacrolimus 25-30 caspase 3 Mus musculus 75-84 30659828-12 2019 What"s more, Mdivi-1 and FK506 restrained cytochrome c release and cleaved caspase-3 expression, ameliorated hippocampal neurons apoptosis, and decreased serum S100beta/NSE concentrations as well. Tacrolimus 25-30 S100 calcium binding protein A1 Mus musculus 160-168 30659828-12 2019 What"s more, Mdivi-1 and FK506 restrained cytochrome c release and cleaved caspase-3 expression, ameliorated hippocampal neurons apoptosis, and decreased serum S100beta/NSE concentrations as well. Tacrolimus 25-30 enolase 2, gamma neuronal Mus musculus 169-172 30387123-5 2019 Mechanically, inhibition of Ca 2+ -calcineurin-NFATc1 signaling by FK506 or 11R-VIVIT abrogated the TRPV4 overexpression-induced osteoclast differentiation and autophagy induction. Tacrolimus 67-72 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 47-53 30928512-6 2019 Compared with 0.05% tacrolimus eye drops, the in vivo immunofluorescence analysis indicated the tacrolimus loaded mPEG-b-PLGA micelles remarkably inhibit the immune rejection after corneal allograft, with the lower expression levels of nuclear factor of activated T cells (NFAT), cluster of differentiation 4 (CD4) and cluster of differentiation 8 (CD8). Tacrolimus 96-106 nuclear factor of activated T-cells 5 Rattus norvegicus 273-277 30928512-6 2019 Compared with 0.05% tacrolimus eye drops, the in vivo immunofluorescence analysis indicated the tacrolimus loaded mPEG-b-PLGA micelles remarkably inhibit the immune rejection after corneal allograft, with the lower expression levels of nuclear factor of activated T cells (NFAT), cluster of differentiation 4 (CD4) and cluster of differentiation 8 (CD8). Tacrolimus 96-106 Cd4 molecule Rattus norvegicus 280-308 30928512-6 2019 Compared with 0.05% tacrolimus eye drops, the in vivo immunofluorescence analysis indicated the tacrolimus loaded mPEG-b-PLGA micelles remarkably inhibit the immune rejection after corneal allograft, with the lower expression levels of nuclear factor of activated T cells (NFAT), cluster of differentiation 4 (CD4) and cluster of differentiation 8 (CD8). Tacrolimus 96-106 Cd4 molecule Rattus norvegicus 310-313 31096684-0 2019 Impact of CYP3A5, POR, and CYP2C19 Polymorphisms on Trough Concentration to Dose Ratio of Tacrolimus in Allogeneic Hematopoietic Stem Cell Transplantation. Tacrolimus 90-100 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 31096684-0 2019 Impact of CYP3A5, POR, and CYP2C19 Polymorphisms on Trough Concentration to Dose Ratio of Tacrolimus in Allogeneic Hematopoietic Stem Cell Transplantation. Tacrolimus 90-100 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 27-34 31096684-2 2019 Here, we investigated the influence of genotypes of CYP3A5, CYP2C19, and POR on the concentration/dose (C/D) ratio of tacrolimus and episodes of acute graft-versus-host disease (GVHD) in Japanese recipients of allogeneic hematopoietic stem cell transplantation (HSCT). Tacrolimus 118-128 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 52-58 31096684-2 2019 Here, we investigated the influence of genotypes of CYP3A5, CYP2C19, and POR on the concentration/dose (C/D) ratio of tacrolimus and episodes of acute graft-versus-host disease (GVHD) in Japanese recipients of allogeneic hematopoietic stem cell transplantation (HSCT). Tacrolimus 118-128 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 60-67 31096684-2 2019 Here, we investigated the influence of genotypes of CYP3A5, CYP2C19, and POR on the concentration/dose (C/D) ratio of tacrolimus and episodes of acute graft-versus-host disease (GVHD) in Japanese recipients of allogeneic hematopoietic stem cell transplantation (HSCT). Tacrolimus 118-128 cytochrome p450 oxidoreductase Homo sapiens 73-76 31096684-4 2019 During continuous intravenous infusion, HSCT recipients carrying the CYP3A5*1 allele, particularly those with at least one POR*28 allele, had a significantly lower tacrolimus C/D ratio throughout all three post-HSCT weeks compared to that in recipients with POR*1/*1 (p < 0.05). Tacrolimus 164-174 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 69-75 31096684-4 2019 During continuous intravenous infusion, HSCT recipients carrying the CYP3A5*1 allele, particularly those with at least one POR*28 allele, had a significantly lower tacrolimus C/D ratio throughout all three post-HSCT weeks compared to that in recipients with POR*1/*1 (p < 0.05). Tacrolimus 164-174 cytochrome p450 oxidoreductase Homo sapiens 123-126 31096684-5 2019 The CYP3A5*3/*3 genotype and the concomitant use of voriconazole were independent predictors of an increased tacrolimus C/D ratio during the switch from continuous intravenous infusion to oral administration (p < 0.05). Tacrolimus 109-119 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 31096684-8 2019 Our results suggest that CYP3A5, POR, and CYP2C19 polymorphisms are useful biomarkers for individualized dosage adjustment of tacrolimus in HSCT recipients. Tacrolimus 126-136 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 25-31 31096684-8 2019 Our results suggest that CYP3A5, POR, and CYP2C19 polymorphisms are useful biomarkers for individualized dosage adjustment of tacrolimus in HSCT recipients. Tacrolimus 126-136 cytochrome p450 oxidoreductase Homo sapiens 33-36 31096684-8 2019 Our results suggest that CYP3A5, POR, and CYP2C19 polymorphisms are useful biomarkers for individualized dosage adjustment of tacrolimus in HSCT recipients. Tacrolimus 126-136 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 42-49 30589990-3 2019 A study in renal transplanted patients disclosed association of CYP3A5*6 and CYP3A5*7 with tacrolimus dosing, which led to addition of these variants to CYP3A5*3 in the CPIC tacrolimus guideline. Tacrolimus 91-101 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 64-70 30589990-3 2019 A study in renal transplanted patients disclosed association of CYP3A5*6 and CYP3A5*7 with tacrolimus dosing, which led to addition of these variants to CYP3A5*3 in the CPIC tacrolimus guideline. Tacrolimus 91-101 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 77-83 30589990-3 2019 A study in renal transplanted patients disclosed association of CYP3A5*6 and CYP3A5*7 with tacrolimus dosing, which led to addition of these variants to CYP3A5*3 in the CPIC tacrolimus guideline. Tacrolimus 91-101 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 77-83 30589990-3 2019 A study in renal transplanted patients disclosed association of CYP3A5*6 and CYP3A5*7 with tacrolimus dosing, which led to addition of these variants to CYP3A5*3 in the CPIC tacrolimus guideline. Tacrolimus 174-184 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 64-70 30589990-3 2019 A study in renal transplanted patients disclosed association of CYP3A5*6 and CYP3A5*7 with tacrolimus dosing, which led to addition of these variants to CYP3A5*3 in the CPIC tacrolimus guideline. Tacrolimus 174-184 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 77-83 30589990-3 2019 A study in renal transplanted patients disclosed association of CYP3A5*6 and CYP3A5*7 with tacrolimus dosing, which led to addition of these variants to CYP3A5*3 in the CPIC tacrolimus guideline. Tacrolimus 174-184 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 77-83 31086126-0 2019 Pharmacokinetic Profile of Prolonged-Release Tacrolimus When Administered via Nasogastric Tube in De Novo Liver Transplantation: A Sub-Study of the DIAMOND Trial. Tacrolimus 45-55 tubulin epsilon 1 Homo sapiens 90-94 31086126-1 2019 BACKGROUND For patients unable to swallow during the immediate post-transplant period, immunosuppressant therapy may be initiated by administering prolonged-release tacrolimus as a suspension via a nasogastric tube. Tacrolimus 165-175 tubulin epsilon 1 Homo sapiens 210-214 31086126-6 2019 The overall mean daily dose of prolonged-release tacrolimus administered via nasogastric tube was higher on Day 1 (0.179 mg/kg) vs. Day 3 (0.140 mg/kg). Tacrolimus 49-59 tubulin epsilon 1 Homo sapiens 89-93 30387123-5 2019 Mechanically, inhibition of Ca 2+ -calcineurin-NFATc1 signaling by FK506 or 11R-VIVIT abrogated the TRPV4 overexpression-induced osteoclast differentiation and autophagy induction. Tacrolimus 67-72 transient receptor potential cation channel, subfamily V, member 4 Mus musculus 100-105 31024070-7 2019 Consistent with the human fertility studies, this investigation reveals a beneficial systemic use of tacrolimus (0.1 mg/kg) in promoting early pregnancy in individuals with PCOS and suggests the need for further research into the selective inhibition of IL17A as a plausibly alternative immunotherapeutic approach in the clinical management of infertile individuals with PCOS. Tacrolimus 101-111 interleukin 17A Homo sapiens 254-259 30753461-5 2019 We further showed that treatment with FK506 (tacrolimus) possibly inhibits the increase in IL-8 levels in RA patients with anti-RANKL Ab, and in vitro assay confirmed that FK506 suppressed IL-8 production in pre-OCLs. Tacrolimus 38-43 C-X-C motif chemokine ligand 8 Homo sapiens 91-95 30753461-5 2019 We further showed that treatment with FK506 (tacrolimus) possibly inhibits the increase in IL-8 levels in RA patients with anti-RANKL Ab, and in vitro assay confirmed that FK506 suppressed IL-8 production in pre-OCLs. Tacrolimus 38-43 TNF superfamily member 11 Homo sapiens 128-133 30753461-5 2019 We further showed that treatment with FK506 (tacrolimus) possibly inhibits the increase in IL-8 levels in RA patients with anti-RANKL Ab, and in vitro assay confirmed that FK506 suppressed IL-8 production in pre-OCLs. Tacrolimus 38-43 C-X-C motif chemokine ligand 8 Homo sapiens 189-193 30753461-5 2019 We further showed that treatment with FK506 (tacrolimus) possibly inhibits the increase in IL-8 levels in RA patients with anti-RANKL Ab, and in vitro assay confirmed that FK506 suppressed IL-8 production in pre-OCLs. Tacrolimus 45-55 C-X-C motif chemokine ligand 8 Homo sapiens 91-95 30753461-5 2019 We further showed that treatment with FK506 (tacrolimus) possibly inhibits the increase in IL-8 levels in RA patients with anti-RANKL Ab, and in vitro assay confirmed that FK506 suppressed IL-8 production in pre-OCLs. Tacrolimus 45-55 TNF superfamily member 11 Homo sapiens 128-133 30753461-5 2019 We further showed that treatment with FK506 (tacrolimus) possibly inhibits the increase in IL-8 levels in RA patients with anti-RANKL Ab, and in vitro assay confirmed that FK506 suppressed IL-8 production in pre-OCLs. Tacrolimus 45-55 C-X-C motif chemokine ligand 8 Homo sapiens 189-193 30753461-5 2019 We further showed that treatment with FK506 (tacrolimus) possibly inhibits the increase in IL-8 levels in RA patients with anti-RANKL Ab, and in vitro assay confirmed that FK506 suppressed IL-8 production in pre-OCLs. Tacrolimus 172-177 C-X-C motif chemokine ligand 8 Homo sapiens 91-95 30753461-5 2019 We further showed that treatment with FK506 (tacrolimus) possibly inhibits the increase in IL-8 levels in RA patients with anti-RANKL Ab, and in vitro assay confirmed that FK506 suppressed IL-8 production in pre-OCLs. Tacrolimus 172-177 TNF superfamily member 11 Homo sapiens 128-133 30753461-5 2019 We further showed that treatment with FK506 (tacrolimus) possibly inhibits the increase in IL-8 levels in RA patients with anti-RANKL Ab, and in vitro assay confirmed that FK506 suppressed IL-8 production in pre-OCLs. Tacrolimus 172-177 C-X-C motif chemokine ligand 8 Homo sapiens 189-193 30707421-4 2019 It has been well established that calcineurin, highly conserved from yeast to mammals, is necessary for invasive fungal disease and is inhibited when in complex with FK506/FKBP12. Tacrolimus 166-171 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 172-178 30324847-9 2019 Notably, Cd treatment increased the activity of PPP3/calcineurin, and pharmacological inhibition of PPP3/calcineurin with FK506 suppressed AKT dephosphorylation and TFE3 activity. Tacrolimus 122-127 AKT serine/threonine kinase 1 Homo sapiens 139-142 30324847-9 2019 Notably, Cd treatment increased the activity of PPP3/calcineurin, and pharmacological inhibition of PPP3/calcineurin with FK506 suppressed AKT dephosphorylation and TFE3 activity. Tacrolimus 122-127 transcription factor binding to IGHM enhancer 3 Homo sapiens 165-169 30597277-0 2019 Effect of CYP3A4, CYP3A5, and ABCB1 Polymorphisms on Intravenous Tacrolimus Exposure and Adverse Events in Adult Allogeneic Stem Cell Transplant Patients. Tacrolimus 65-75 ATP binding cassette subfamily B member 1 Homo sapiens 30-35 30597277-10 2019 ABCB1 C2677T was significantly associated with concentrations >15ng/mL (odds ratio, 6.2; 95% confidence interval, 1.8 to 23.6; P = .004) and tacrolimus-related toxicities (odds ratio, 7.5; 95% confidence interval, 1.6 to 55.2; P = .02). Tacrolimus 144-154 ATP binding cassette subfamily B member 1 Homo sapiens 0-5 30597277-12 2019 tacrolimus exposure, whereas ABCB1 C2677T also impacts tacrolimus-related toxicities in stem cell transplants. Tacrolimus 55-65 ATP binding cassette subfamily B member 1 Homo sapiens 29-34 30983820-3 2019 The performance of the calcineurin inhibitor tacrolimus in this clinical setting is insufficiently elucidated. Tacrolimus 45-55 calcineurin binding protein 1 Homo sapiens 23-44 30983820-19 2019 In all, tacrolimus therapy appears to be a viable option for short-term treatment of steroid-refractory acute severe ulcerative colitis besides ciclosporin and anti-tumor necrosis factor alpha treatment. Tacrolimus 8-18 tumor necrosis factor Homo sapiens 165-192 30707421-3 2019 One potential antifungal drug, FK506, establishes a ternary complex between the phosphatase, calcineurin, and the 12-kDa peptidyl-prolyl isomerase FK506-binding protein, FKBP12. Tacrolimus 31-36 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 170-176 30707421-7 2019 Here we report the backbone and sidechain NMR assignments of recombinant FKBP12 proteins from the pathogenic fungi Mucor circinelloides and Aspergillus fumigatus in the apo form and compare these to the backbone assignments of the FK506 bound form. Tacrolimus 231-236 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 73-79 30707421-8 2019 In addition, we report the backbone assignments of the apo and FK506 bound forms of the Homo sapiens FKBP12 protein for evaluation against the fungal forms. Tacrolimus 63-68 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 101-107 30707421-9 2019 These data are the first steps towards defining, at a residue specific level, the impacts of FK506 binding to fungal and mammalian FKBP12 proteins. Tacrolimus 93-98 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 131-137 30251118-10 2019 As calcineurin may dephosphorylate Drp1, we determined the effect of a calcineurin inhibitor, FK506, which prevented leptin-induced hypertrophy as well as mitochondrial fission and mitochondrial dysfunction. Tacrolimus 94-99 leptin Homo sapiens 117-123 30713033-10 2019 RESULTS: In rats pre-treated with FK506, the levels of transaminases, TNF-alpha and IL-1beta were reduced significantly and also liver damage was dramatically mitigated compared to those without FK506 pre-treatment. Tacrolimus 34-39 tumor necrosis factor Rattus norvegicus 70-79 30713033-10 2019 RESULTS: In rats pre-treated with FK506, the levels of transaminases, TNF-alpha and IL-1beta were reduced significantly and also liver damage was dramatically mitigated compared to those without FK506 pre-treatment. Tacrolimus 34-39 interleukin 1 beta Rattus norvegicus 84-92 30713033-11 2019 Moreover, the expression of HO-1 at the level of both transcription and translation increased clearly and the activation of the HIF-1alpha was found in FK506 pre-treated livers. Tacrolimus 152-157 heme oxygenase 1 Rattus norvegicus 28-32 30713033-11 2019 Moreover, the expression of HO-1 at the level of both transcription and translation increased clearly and the activation of the HIF-1alpha was found in FK506 pre-treated livers. Tacrolimus 152-157 hypoxia inducible factor 1 subunit alpha Rattus norvegicus 128-138 30713033-12 2019 Moreover, NFAT3 protein transportation to the nucleus was reduced and Bax protein expression was decreased, but the expression of Bcl-2 protein was markedly increased after FK506 pre-treatment. Tacrolimus 173-178 BCL2, apoptosis regulator Rattus norvegicus 130-135 30713033-13 2019 CONCLUSION: FK506 pre-treatment could lessen hepatic ischemia-reperfusion injury through up-regulating the expression of HIF-1alpha and HO-1, and inhibiting nuclear translocation of NFAT3 in liver tissues. Tacrolimus 12-17 hypoxia inducible factor 1 subunit alpha Rattus norvegicus 121-131 30713033-13 2019 CONCLUSION: FK506 pre-treatment could lessen hepatic ischemia-reperfusion injury through up-regulating the expression of HIF-1alpha and HO-1, and inhibiting nuclear translocation of NFAT3 in liver tissues. Tacrolimus 12-17 heme oxygenase 1 Rattus norvegicus 136-140 30704156-0 2019 Individualized treatment based on CYP3A5 single-nucleotide polymorphisms with tacrolimus in ulcerative colitis. Tacrolimus 78-88 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 34-40 30704156-1 2019 BACKGROUND/AIMS: The pharmacokinetics of tacrolimus (TAC) is known to be largely influenced by single-nucleotide polymorphisms (SNPs) in CYP3A5. Tacrolimus 41-51 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 137-143 30879170-2 2019 Human FKBP12 is a prototype of this family and it is involved in many diseases due to its interaction with the immunosuppressive drugs FK506 and rapamycin. Tacrolimus 135-140 FKBP prolyl isomerase 1A pseudogene 1 Homo sapiens 6-12 30294901-3 2019 The immunophilin FKBP12 binding ligand FK506 is well known as an immunosuppressive agent by inhibiting the calcineurin expression. Tacrolimus 39-44 FK506 binding protein 1a Mus musculus 4-23 30294901-4 2019 In this study, we synthesized a series of modified compounds based on the FKBP12 binding moiety to as same as the binding structure of rapamycin and FK506 particularly. Tacrolimus 149-154 FK506 binding protein 1a Mus musculus 74-80 30457174-6 2019 Treatment with calcineurin inhibitor cyclosporin A (CsA) or FK506 suppressed cell proliferation and induced apoptotic cell death in both MYCN-amplified and MYCN-non-amplified NB cell lines. Tacrolimus 60-65 MYCN proto-oncogene, bHLH transcription factor Homo sapiens 137-141 30983501-0 2019 Impact of CYP3A5 phenotype on tacrolimus concentrations after sublingual and oral administration in lung transplant. Tacrolimus 30-40 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 30983501-6 2019 Conclusion: This study suggests personalized dosing strategies for tacrolimus likely need to consider characteristics beyond CYP3A5 genotype. Tacrolimus 67-77 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 125-131 30979494-7 2019 CONCLUSION: Wild-type MDR1 gene product P-glycoprotein expressed in the intestine reduces drug absorption from the gastrointestinal tract and may have contributed to low blood levels of tacrolimus in this patient when tacrolimus was orally administered. Tacrolimus 186-196 ATP binding cassette subfamily B member 1 Homo sapiens 22-26 30979494-7 2019 CONCLUSION: Wild-type MDR1 gene product P-glycoprotein expressed in the intestine reduces drug absorption from the gastrointestinal tract and may have contributed to low blood levels of tacrolimus in this patient when tacrolimus was orally administered. Tacrolimus 186-196 ATP binding cassette subfamily B member 1 Homo sapiens 40-54 30979494-7 2019 CONCLUSION: Wild-type MDR1 gene product P-glycoprotein expressed in the intestine reduces drug absorption from the gastrointestinal tract and may have contributed to low blood levels of tacrolimus in this patient when tacrolimus was orally administered. Tacrolimus 218-228 ATP binding cassette subfamily B member 1 Homo sapiens 22-26 30979494-7 2019 CONCLUSION: Wild-type MDR1 gene product P-glycoprotein expressed in the intestine reduces drug absorption from the gastrointestinal tract and may have contributed to low blood levels of tacrolimus in this patient when tacrolimus was orally administered. Tacrolimus 218-228 ATP binding cassette subfamily B member 1 Homo sapiens 40-54 30371942-1 2019 Tacrolimus, a calcineurin inhibitor, is a common immunosuppressant prescribed after organ transplantation and has notable inter- and intrapatient pharmacokinetic variability. Tacrolimus 0-10 calcineurin binding protein 1 Homo sapiens 14-35 30371942-8 2019 The CYP3A5 genotype, as a covariate, consistently impacted tacrolimus clearance, and dosing adjustments were required to achieve similar drug exposure among patients. Tacrolimus 59-69 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 4-10 30478741-0 2019 FK506 (tacrolimus) causes pain sensation through the activation of transient receptor potential ankyrin 1 (TRPA1) channels. Tacrolimus 0-5 transient receptor potential cation channel subfamily A member 1 Homo sapiens 67-105 30478741-0 2019 FK506 (tacrolimus) causes pain sensation through the activation of transient receptor potential ankyrin 1 (TRPA1) channels. Tacrolimus 0-5 transient receptor potential cation channel subfamily A member 1 Homo sapiens 107-112 30478741-0 2019 FK506 (tacrolimus) causes pain sensation through the activation of transient receptor potential ankyrin 1 (TRPA1) channels. Tacrolimus 7-17 transient receptor potential cation channel subfamily A member 1 Homo sapiens 67-105 30478741-0 2019 FK506 (tacrolimus) causes pain sensation through the activation of transient receptor potential ankyrin 1 (TRPA1) channels. Tacrolimus 7-17 transient receptor potential cation channel subfamily A member 1 Homo sapiens 107-112 30478741-3 2019 In this study, we found that FK506 activates transient receptor potential ankyrin 1 (TRPA1) channels. Tacrolimus 29-34 transient receptor potential cation channel subfamily A member 1 Homo sapiens 45-83 30478741-3 2019 In this study, we found that FK506 activates transient receptor potential ankyrin 1 (TRPA1) channels. Tacrolimus 29-34 transient receptor potential cation channel subfamily A member 1 Homo sapiens 85-90 30478741-4 2019 In Ca2+-imaging experiments, increases in intracellular Ca2+ concentrations ([Ca2+]i) by FK506 were observed in HEK293T cells expressing hTRPA1 or hTRPM8. Tacrolimus 89-94 transient receptor potential cation channel subfamily A member 1 Homo sapiens 137-143 30478741-4 2019 In Ca2+-imaging experiments, increases in intracellular Ca2+ concentrations ([Ca2+]i) by FK506 were observed in HEK293T cells expressing hTRPA1 or hTRPM8. Tacrolimus 89-94 transient receptor potential cation channel subfamily M member 8 Homo sapiens 147-153 30478741-5 2019 FK506-induced currents were observed in HEK293T cells expressing hTRPA1 or mTRPA1, but less or not at all in cells expressing hTRPV1 or hTRPM8 using a patch-clamp technique. Tacrolimus 0-5 transient receptor potential cation channel subfamily A member 1 Homo sapiens 65-71 30478741-5 2019 FK506-induced currents were observed in HEK293T cells expressing hTRPA1 or mTRPA1, but less or not at all in cells expressing hTRPV1 or hTRPM8 using a patch-clamp technique. Tacrolimus 0-5 transient receptor potential cation channel, subfamily A, member 1 Mus musculus 75-81 30478741-6 2019 FK506 also evoked single-channel opening of hTRPA1 in an inside-out configuration. Tacrolimus 0-5 transient receptor potential cation channel subfamily A member 1 Homo sapiens 44-50 30478741-7 2019 FK506-induced [Ca2+]i increases were also observed in TRPA1-expressing mouse primary sensory neurons. Tacrolimus 0-5 transient receptor potential cation channel, subfamily A, member 1 Mus musculus 54-59 30478741-8 2019 Furthermore, injection of FK506 evoked licking or biting behaviors and these behaviors were almost abolished in TRPA1 knockout mice. Tacrolimus 26-31 transient receptor potential cation channel, subfamily A, member 1 Mus musculus 112-117 30478741-9 2019 These results indicate that FK506 might cause pain sensations through TRPA1 activation. Tacrolimus 28-33 transient receptor potential cation channel subfamily A member 1 Homo sapiens 70-75 30457174-8 2019 c-Myc, MYCN, and beta-catenin were downregulated at the mRNA and protein levels in CsA or FK506-treated NB cells. Tacrolimus 90-95 MYC proto-oncogene, bHLH transcription factor Homo sapiens 0-5 30457174-8 2019 c-Myc, MYCN, and beta-catenin were downregulated at the mRNA and protein levels in CsA or FK506-treated NB cells. Tacrolimus 90-95 MYCN proto-oncogene, bHLH transcription factor Homo sapiens 7-11 30457174-8 2019 c-Myc, MYCN, and beta-catenin were downregulated at the mRNA and protein levels in CsA or FK506-treated NB cells. Tacrolimus 90-95 catenin beta 1 Homo sapiens 17-29 30260738-5 2019 Recently, it was shown that tacrolimus increased ALK-1 signaling and had beneficial effects in selected end-stage PAH patients. Tacrolimus 28-38 activin A receptor like type 1 Homo sapiens 49-54 30592973-20 2019 On TLR-day an association of renal function and Tac concentration with TLR4ex was clear only in the tacrolimus population. Tacrolimus 100-110 toll like receptor 4 Homo sapiens 71-75 30592973-25 2019 Evaluation of the averaged TLR4ex can be used to assess the efficacy of immunosuppression in the treatment with tacrolimus and to estimate the likelihood of deterioration in renal function. Tacrolimus 112-122 toll like receptor 4 Homo sapiens 27-31 30260084-5 2019 Tacrolimus clearances were 26, 18.8 and 12.3 L/h, for patients with genetic polymorphisms CYP3A5*1*1, *1*3 and *3*3, respectively. Tacrolimus 0-10 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 90-96 30983748-0 2019 Loss of Subpodocytic Space Predicts Poor Response to Tacrolimus in Steroid-Resistant Calcineurin Inhibitor-Naive Adult-Onset Primary Focal Segmental Glomerulosclerosis. Tacrolimus 53-63 calcineurin binding protein 1 Homo sapiens 85-106 30983748-8 2019 The tacrolimus-resistant patients were of older age, had a longer duration of illness, and a lower eGFR as compared to tacrolimus responsive cases. Tacrolimus 4-14 epidermal growth factor receptor Homo sapiens 99-103 30457174-6 2019 Treatment with calcineurin inhibitor cyclosporin A (CsA) or FK506 suppressed cell proliferation and induced apoptotic cell death in both MYCN-amplified and MYCN-non-amplified NB cell lines. Tacrolimus 60-65 MYCN proto-oncogene, bHLH transcription factor Homo sapiens 156-160 30532015-1 2019 Rapamycin and FK506 are macrocyclic natural products with an extraordinary mode of action, in which they form binary complexes with FK506-binding protein (FKBP) through a shared FKBP-binding domain before forming ternary complexes with their respective targets, mechanistic target of rapamycin (mTOR) and calcineurin, respectively. Tacrolimus 14-19 mechanistic target of rapamycin kinase Homo sapiens 295-299 30931336-14 2019 Furthermore, they could demonstrate that treatment with the immunosuppressive drug tacrolimus resulted in decreased CMV-specific IFN-gamma and of IL-21 production. Tacrolimus 83-93 interferon gamma Homo sapiens 129-138 30907352-0 2019 [Clinical effect of tacrolimus combined with glucocorticoid in the treatment of IgA nephropathy in children]. Tacrolimus 20-30 IGAN1 Homo sapiens 80-95 30946720-5 2019 Some studies suggest that post-transplant immunosuppression with tacrolimus is linked to an increased occurrence of IgE-mediated sensitization and manifestation of allergic disease. Tacrolimus 65-75 immunoglobulin heavy constant epsilon Homo sapiens 116-119 30879580-13 2019 CONCLUSION: Rituximab and switching tacrolimus to cyclosporine, in conjunction with plasmapheresis, appeared to be effective and safe in children with recurrent FSGS. Tacrolimus 36-46 actinin alpha 4 Homo sapiens 161-165 30931336-14 2019 Furthermore, they could demonstrate that treatment with the immunosuppressive drug tacrolimus resulted in decreased CMV-specific IFN-gamma and of IL-21 production. Tacrolimus 83-93 interleukin 21 Homo sapiens 146-151 30719576-6 2019 The CL/F of tacrolimus in Wuzhi tablets co-administration and CYP3A5 non-expresser groups were 19.3% and 19.1% lower than that of the non-Wuzhi tablets and CYP3A5 expresser groups, respectively. Tacrolimus 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 62-68 30718414-10 2019 In vivo, tacrolimus increases levels of KLHL3S433-P, resulting in increased levels of WNK4, phosphorylated SPAK, and NCC. Tacrolimus 9-19 WNK lysine deficient protein kinase 4 Homo sapiens 86-90 30718414-10 2019 In vivo, tacrolimus increases levels of KLHL3S433-P, resulting in increased levels of WNK4, phosphorylated SPAK, and NCC. Tacrolimus 9-19 serine/threonine kinase 39 Homo sapiens 107-111 30718414-11 2019 Moreover, tacrolimus attenuates KLHL3-mediated WNK4 ubiquitylation and degradation, while this effect is absent in KLHL3 with S433A substitution. Tacrolimus 10-20 kelch like family member 3 Homo sapiens 32-37 30718414-11 2019 Moreover, tacrolimus attenuates KLHL3-mediated WNK4 ubiquitylation and degradation, while this effect is absent in KLHL3 with S433A substitution. Tacrolimus 10-20 WNK lysine deficient protein kinase 4 Homo sapiens 47-51 30718414-12 2019 Additionally, increased extracellular K+ induced calcineurin-dependent dephosphorylation of KLHL3S433-P These findings demonstrate that KLHL3S433-P is a calcineurin substrate and implicate increased KLHL3 phosphorylation in tacrolimus-induced pathologies. Tacrolimus 224-234 kelch like family member 3 Homo sapiens 92-97 30997048-1 2019 We report the initiation of CFTR modulator lumacaftor/ivacaftor combination (LUM/IVA) in two adolescents with cystic fibrosis who were treated with antifungal azoles (AZO) and tacrolimus (TCS) for liver transplantation. Tacrolimus 176-186 CF transmembrane conductance regulator Homo sapiens 28-32 30997048-1 2019 We report the initiation of CFTR modulator lumacaftor/ivacaftor combination (LUM/IVA) in two adolescents with cystic fibrosis who were treated with antifungal azoles (AZO) and tacrolimus (TCS) for liver transplantation. Tacrolimus 176-186 lumican Homo sapiens 77-80 30997048-1 2019 We report the initiation of CFTR modulator lumacaftor/ivacaftor combination (LUM/IVA) in two adolescents with cystic fibrosis who were treated with antifungal azoles (AZO) and tacrolimus (TCS) for liver transplantation. Tacrolimus 188-191 CF transmembrane conductance regulator Homo sapiens 28-32 30997048-1 2019 We report the initiation of CFTR modulator lumacaftor/ivacaftor combination (LUM/IVA) in two adolescents with cystic fibrosis who were treated with antifungal azoles (AZO) and tacrolimus (TCS) for liver transplantation. Tacrolimus 188-191 lumican Homo sapiens 77-80 30794705-0 2019 Correction: Alteration of urinary neutrophil gelatinase-associated lipocalin as a predictor of tacrolimus-induced chronic renal allograft fibrosis in tacrolimus dose adjustments following kidney transplantation. Tacrolimus 95-105 lipocalin 2 Homo sapiens 34-76 30794705-0 2019 Correction: Alteration of urinary neutrophil gelatinase-associated lipocalin as a predictor of tacrolimus-induced chronic renal allograft fibrosis in tacrolimus dose adjustments following kidney transplantation. Tacrolimus 150-160 lipocalin 2 Homo sapiens 34-76 30545944-0 2019 The Immunosuppressant Macrolide Tacrolimus Activates Cold-Sensing TRPM8 Channels. Tacrolimus 32-42 transient receptor potential cation channel subfamily M member 8 Homo sapiens 66-71 30545944-2 2019 We found that TRPM8 is a pharmacological target of tacrolimus (FK506), a macrolide immunosuppressant with several clinical uses, including the treatment of organ rejection following transplants, treatment of atopic dermatitis, and dry eye disease. Tacrolimus 51-61 transient receptor potential cation channel subfamily M member 8 Homo sapiens 14-19 30545944-2 2019 We found that TRPM8 is a pharmacological target of tacrolimus (FK506), a macrolide immunosuppressant with several clinical uses, including the treatment of organ rejection following transplants, treatment of atopic dermatitis, and dry eye disease. Tacrolimus 63-68 transient receptor potential cation channel subfamily M member 8 Homo sapiens 14-19 30545944-4 2019 Tacrolimus activates TRPM8 channels in different species, including humans, and sensitizes their response to cold temperature by inducing a leftward shift in the voltage-dependent activation curve. Tacrolimus 0-10 transient receptor potential cation channel subfamily M member 8 Homo sapiens 21-26 30545944-5 2019 The effects of tacrolimus on purified TRPM8 in lipid bilayers demonstrates conclusively that it has a direct gating effect. Tacrolimus 15-25 transient receptor potential cation channel subfamily M member 8 Homo sapiens 38-43 30545944-7 2019 Menthol (TRPM8-Y745H)- and icilin (TRPM8-N799A)-insensitive mutants were also activated by tacrolimus, suggesting a different binding site. Tacrolimus 91-101 transient receptor potential cation channel subfamily M member 8 Homo sapiens 9-14 30545944-7 2019 Menthol (TRPM8-Y745H)- and icilin (TRPM8-N799A)-insensitive mutants were also activated by tacrolimus, suggesting a different binding site. Tacrolimus 91-101 transient receptor potential cation channel subfamily M member 8 Homo sapiens 35-40 30545944-8 2019 In cultured mouse DRG neurons, tacrolimus evokes an increase in intracellular calcium almost exclusively in cold-sensitive neurons, and these responses were drastically blunted in Trpm8 KO mice or after the application of TRPM8 antagonists. Tacrolimus 31-41 transient receptor potential cation channel, subfamily M, member 8 Mus musculus 180-185 30545944-8 2019 In cultured mouse DRG neurons, tacrolimus evokes an increase in intracellular calcium almost exclusively in cold-sensitive neurons, and these responses were drastically blunted in Trpm8 KO mice or after the application of TRPM8 antagonists. Tacrolimus 31-41 transient receptor potential cation channel, subfamily M, member 8 Mus musculus 222-227 30545944-10 2019 Together, our results identify TRPM8 channels in sensory neurons as molecular targets of the immunosuppressant tacrolimus. Tacrolimus 111-121 transient receptor potential cation channel, subfamily M, member 8 Mus musculus 31-36 30545944-11 2019 The actions of tacrolimus on TRPM8 resemble those of menthol but likely involve interactions with other channel residues.SIGNIFICANCE STATEMENT TRPM8 is a polymodal TRP channel involved in cold temperature sensing, thermoregulation, and cold pain. Tacrolimus 15-25 transient receptor potential cation channel, subfamily M, member 8 Mus musculus 29-34 30545944-11 2019 The actions of tacrolimus on TRPM8 resemble those of menthol but likely involve interactions with other channel residues.SIGNIFICANCE STATEMENT TRPM8 is a polymodal TRP channel involved in cold temperature sensing, thermoregulation, and cold pain. Tacrolimus 15-25 transient receptor potential cation channel, subfamily M, member 8 Mus musculus 144-149 30545944-13 2019 We report the direct agonist effect of tacrolimus, a potent natural immunosuppressant with multiple clinical applications, on TRPM8 activity. Tacrolimus 39-49 transient receptor potential cation channel, subfamily M, member 8 Mus musculus 126-131 30719576-8 2019 Monte Carlo simulation showed that the nephrotic syndrome patients that were CYP3A5 non-expressers or co-administered Wuzhi tablets received 50% or 33.3% lower dose of tacrolimus to reach the target concentration. Tacrolimus 168-178 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 77-83 30719576-9 2019 In contrast, the NR1I2 rs227707 TT genotype carriers were administered a 33.3% higher dose of tacrolimus than the NR1I2 rs227707 CC/CT genotype carriers. Tacrolimus 94-104 nuclear receptor subfamily 1 group I member 2 Homo sapiens 17-22 30307767-0 2019 Effect of Klotho on autophagy clearance in tacrolimus-induced renal injury. Tacrolimus 43-53 klotho Mus musculus 10-16 30307767-1 2019 Recently, we showed that tacrolimus-induced renal injury was closely associated with impairment of autophagy clearance, and Klotho deficiency aggravated tacrolimus-induced renal injury. Tacrolimus 153-163 klotho Mus musculus 124-130 30307767-2 2019 In this study, we evaluated the effect of Klotho treatment on autophagy clearance in tacrolimus-induced renal injury. Tacrolimus 85-95 klotho Mus musculus 42-48 30307767-3 2019 We evaluated the effect of Klotho on tacrolimus-induced renal injury in an experimental mouse model and in vitro by treatment with tacrolimus and/or recombinant mouse Klotho. Tacrolimus 37-47 klotho Mus musculus 27-33 30472069-0 2019 Tacrolimus inhibits Th1 and Th17 responses in MuSK-antibody positive myasthenia gravis patients. Tacrolimus 0-10 muscle associated receptor tyrosine kinase Homo sapiens 46-50 30307767-4 2019 In vivo and in vitro studies showed that tacrolimus treatment impaired lysosomal acidification and decreased cathepsin B activity, expression of lysosome-associated membrane protein 2, and expression of transcription factor EB (TFEB), a master regulator for lysosomal biogenesis. Tacrolimus 41-51 cathepsin B Mus musculus 109-120 30472069-3 2019 Tacrolimus, an immunosuppressant used in AChR-MG and transplantation patients, inhibits T cell responses through interference with IL-2 transcription. Tacrolimus 0-10 interleukin 2 Homo sapiens 131-135 30307767-4 2019 In vivo and in vitro studies showed that tacrolimus treatment impaired lysosomal acidification and decreased cathepsin B activity, expression of lysosome-associated membrane protein 2, and expression of transcription factor EB (TFEB), a master regulator for lysosomal biogenesis. Tacrolimus 41-51 transcription factor EB Mus musculus 203-226 30472069-6 2019 We determined that tacrolimus profoundly suppressed CD4 and CD8 T cell proliferation and significantly suppressed Th1 and Th17 responses, as demonstrated by a reduced frequency of IFN-gamma, IL-2, and IL-17 producing CD4 T cells and reduced frequencies of IFN-gamma and IL-2 producing CD8 T cells. Tacrolimus 19-29 CD4 molecule Homo sapiens 52-55 30307767-4 2019 In vivo and in vitro studies showed that tacrolimus treatment impaired lysosomal acidification and decreased cathepsin B activity, expression of lysosome-associated membrane protein 2, and expression of transcription factor EB (TFEB), a master regulator for lysosomal biogenesis. Tacrolimus 41-51 transcription factor EB Mus musculus 228-232 30472069-6 2019 We determined that tacrolimus profoundly suppressed CD4 and CD8 T cell proliferation and significantly suppressed Th1 and Th17 responses, as demonstrated by a reduced frequency of IFN-gamma, IL-2, and IL-17 producing CD4 T cells and reduced frequencies of IFN-gamma and IL-2 producing CD8 T cells. Tacrolimus 19-29 CD8a molecule Homo sapiens 60-63 30472069-6 2019 We determined that tacrolimus profoundly suppressed CD4 and CD8 T cell proliferation and significantly suppressed Th1 and Th17 responses, as demonstrated by a reduced frequency of IFN-gamma, IL-2, and IL-17 producing CD4 T cells and reduced frequencies of IFN-gamma and IL-2 producing CD8 T cells. Tacrolimus 19-29 interferon gamma Homo sapiens 180-189 30307767-8 2019 Collectively, our data suggest that Klotho improves autophagy clearance via activation of lysosomal function in tacrolimus-induced nephrotoxicity.-Lim, S. W., Shin, Y. J., Luo, K., Quan, Y., Ko, E. J., Chung, B. H., Yang, C. W. Effect of Klotho on autophagy clearance in tacrolimus-induced renal injury. Tacrolimus 112-122 klotho Mus musculus 36-42 30472069-6 2019 We determined that tacrolimus profoundly suppressed CD4 and CD8 T cell proliferation and significantly suppressed Th1 and Th17 responses, as demonstrated by a reduced frequency of IFN-gamma, IL-2, and IL-17 producing CD4 T cells and reduced frequencies of IFN-gamma and IL-2 producing CD8 T cells. Tacrolimus 19-29 interleukin 2 Homo sapiens 191-195 30307767-8 2019 Collectively, our data suggest that Klotho improves autophagy clearance via activation of lysosomal function in tacrolimus-induced nephrotoxicity.-Lim, S. W., Shin, Y. J., Luo, K., Quan, Y., Ko, E. J., Chung, B. H., Yang, C. W. Effect of Klotho on autophagy clearance in tacrolimus-induced renal injury. Tacrolimus 271-281 klotho Mus musculus 36-42 30472069-6 2019 We determined that tacrolimus profoundly suppressed CD4 and CD8 T cell proliferation and significantly suppressed Th1 and Th17 responses, as demonstrated by a reduced frequency of IFN-gamma, IL-2, and IL-17 producing CD4 T cells and reduced frequencies of IFN-gamma and IL-2 producing CD8 T cells. Tacrolimus 19-29 interleukin 17A Homo sapiens 201-206 30472069-6 2019 We determined that tacrolimus profoundly suppressed CD4 and CD8 T cell proliferation and significantly suppressed Th1 and Th17 responses, as demonstrated by a reduced frequency of IFN-gamma, IL-2, and IL-17 producing CD4 T cells and reduced frequencies of IFN-gamma and IL-2 producing CD8 T cells. Tacrolimus 19-29 CD4 molecule Homo sapiens 217-220 30323313-0 2019 Impact of single nucleotide polymorphisms on P450 oxidoreductase and peroxisome proliferator-activated receptor alpha on tacrolimus pharmacokinetics in renal transplant recipients. Tacrolimus 121-131 peroxisome proliferator activated receptor alpha Homo sapiens 69-117 30472069-6 2019 We determined that tacrolimus profoundly suppressed CD4 and CD8 T cell proliferation and significantly suppressed Th1 and Th17 responses, as demonstrated by a reduced frequency of IFN-gamma, IL-2, and IL-17 producing CD4 T cells and reduced frequencies of IFN-gamma and IL-2 producing CD8 T cells. Tacrolimus 19-29 interferon gamma Homo sapiens 256-265 30472069-6 2019 We determined that tacrolimus profoundly suppressed CD4 and CD8 T cell proliferation and significantly suppressed Th1 and Th17 responses, as demonstrated by a reduced frequency of IFN-gamma, IL-2, and IL-17 producing CD4 T cells and reduced frequencies of IFN-gamma and IL-2 producing CD8 T cells. Tacrolimus 19-29 interleukin 2 Homo sapiens 270-274 30472069-6 2019 We determined that tacrolimus profoundly suppressed CD4 and CD8 T cell proliferation and significantly suppressed Th1 and Th17 responses, as demonstrated by a reduced frequency of IFN-gamma, IL-2, and IL-17 producing CD4 T cells and reduced frequencies of IFN-gamma and IL-2 producing CD8 T cells. Tacrolimus 19-29 CD8a molecule Homo sapiens 285-288 30472069-7 2019 Tacrolimus also inhibits pathogenic Th17 cells coproducing IL-17 and IFN-gamma. Tacrolimus 0-10 interleukin 17A Homo sapiens 59-64 30472069-7 2019 Tacrolimus also inhibits pathogenic Th17 cells coproducing IL-17 and IFN-gamma. Tacrolimus 0-10 interferon gamma Homo sapiens 69-78 30446894-1 2019 Background Dosage quantities of tacrolimus (TAC) vary according to cytochrome P450 3A5 (CYP3A5) genotype. Tacrolimus 32-42 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 67-86 30446894-1 2019 Background Dosage quantities of tacrolimus (TAC) vary according to cytochrome P450 3A5 (CYP3A5) genotype. Tacrolimus 32-42 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 88-94 30323313-2 2019 We aimed to investigate the impact of single nucleotide polymorphisms (SNPs) in the POR and PPARA genes on the pharmacokinetics of tacrolimus (TAC) in renal transplant recipients. Tacrolimus 131-141 cytochrome p450 oxidoreductase Homo sapiens 84-87 30323313-2 2019 We aimed to investigate the impact of single nucleotide polymorphisms (SNPs) in the POR and PPARA genes on the pharmacokinetics of tacrolimus (TAC) in renal transplant recipients. Tacrolimus 131-141 peroxisome proliferator activated receptor alpha Homo sapiens 92-97 30295407-1 2019 INTRODUCTION: Isavuconazole, a triazole antifungal, is an inhibitor of cytochrome P450 3A4, which also metabolizes tacrolimus and sirolimus. Tacrolimus 115-125 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 71-90 30520827-0 2019 Switching Immunosuppression From Cyclosporine to Tacrolimus in Kidney Transplant Recipients Based on CYP3A5 Genotyping. Tacrolimus 49-59 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 101-107 31384453-0 2019 Implication of interleukin-2 receptor antibody induction therapy in standard risk renal transplant in the tacrolimus era: a meta-analysis. Tacrolimus 106-116 interleukin 2 Homo sapiens 15-28 31384453-3 2019 The aim of our study and meta-analysis is to explore the effect of IL-2 induction therapy on the rate of rejection and patient and graft survival in standard-risk renal transplant patients with tacrolimus-based maintenance immunotherapy. Tacrolimus 194-204 interleukin 2 Homo sapiens 67-71 30643171-7 2019 Moreover, Tacrolimus reduced TLR5 expression in bladder macrophages during UTI. Tacrolimus 10-20 toll-like receptor 5 Mus musculus 29-33 30643171-8 2019 This immunosuppressive state can be explained by the upregulation of TLR-signaling negative regulators (A20, ATF3, IRAK-M and SOCS1) and parallel downregulation of TLR5 as observed in Tacrolimus treated granulocytes and macrophages. Tacrolimus 184-194 toll-like receptor 5 Mus musculus 164-168 30521345-11 2019 A model drug, tacrolimus, was encapsulated in MNP (T-RNP) and significantly suppressed the progression of RA in mice. Tacrolimus 14-24 modifier of Niemann Pick type C1 Mus musculus 46-49