PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 1718419-5 1991 In rifampicin-treated cells, metabolically labeled with [35S]methionine and induced by IPTG, the P68 kinase was the predominant labeled product. Rifampin 3-13 KH RNA binding domain containing, signal transduction associated 1 Homo sapiens 97-100 2015817-6 1991 Onset of CYP3A6 gene expression occurred at day 30 of gestation and both transcription and mRNA accumulation were transplacentally inducible by rifampicin only shortly before birth, i.e. after treatment of the females between days 28 and 30 of gestation. Rifampin 144-154 cytochrome P450 3A6 Oryctolagus cuniculus 9-15 1863618-11 1991 Over the entire outbreak period, the MRSA strain developed resistance to rifampin, imipenem-cilastatin and ciprofloxacin. Rifampin 73-81 solute carrier family 9 member A6 Homo sapiens 37-41 2388264-12 1990 The rifampicin resistance of uvsW-repressed replication suggests that it involves either tertiary initiation or some novel mode of initiation. Rifampin 4-14 UvsW helicase Escherichia phage T4 29-33 1991139-9 1991 In contrast, the transport of a typical substrate of the bilirubin carrier (rifampicin), of amino acids (alpha-aminoisobutyric acid), long chain fatty acids (oleic acid) and cationic compounds (thiamin hydrochloride) was not inhibited by the same renin inhibitors. Rifampin 76-86 renin Homo sapiens 247-252 2013969-7 1991 Treatment of the mice with the new rifamycin derivatives or rifampicin (RFP: as a control drug) was performed by daily oral administration of 10 mg/kg of the drugs, starting at the 24th hour of infection and continuing until the 40th day of infection. Rifampin 60-70 tripartite motif-containing 27 Mus musculus 72-75 2015817-8 1991 CYP3A6 gene expression was also induced by rifampicin in 1-week-old and 2-week-old animals. Rifampin 43-53 cytochrome P450 3A6 Oryctolagus cuniculus 0-6 2015817-15 1991 It is concluded that (a) the onset of CYP3A6 gene expression in the fetus occurs at day 30 of gestation, (b) expression of this gene may be induced transplacentally by rifampicin, (c) CYP1A1, CYP1A2 and CYP3A6 gene expression is sharply activated at weaning, and (d) thyroid hormones appear not to be responsible for the pattern of developmental expression of these genes in the rabbit. Rifampin 168-178 cytochrome P450 3A6 Oryctolagus cuniculus 38-44 2015817-15 1991 It is concluded that (a) the onset of CYP3A6 gene expression in the fetus occurs at day 30 of gestation, (b) expression of this gene may be induced transplacentally by rifampicin, (c) CYP1A1, CYP1A2 and CYP3A6 gene expression is sharply activated at weaning, and (d) thyroid hormones appear not to be responsible for the pattern of developmental expression of these genes in the rabbit. Rifampin 168-178 cytochrome P450 1A2 Oryctolagus cuniculus 192-198 2015817-15 1991 It is concluded that (a) the onset of CYP3A6 gene expression in the fetus occurs at day 30 of gestation, (b) expression of this gene may be induced transplacentally by rifampicin, (c) CYP1A1, CYP1A2 and CYP3A6 gene expression is sharply activated at weaning, and (d) thyroid hormones appear not to be responsible for the pattern of developmental expression of these genes in the rabbit. Rifampin 168-178 cytochrome P450 3A6 Oryctolagus cuniculus 203-209 2200675-1 1990 Hepatocytes from adult and newborn humans were put into primary culture and exposed to phenobarbital, 3-methylcholanthrene, or rifampicin, three well-known inducers of cytochrome P-450 in animals. Rifampin 127-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-184 2200675-5 1990 Phenobarbital and rifampicin increased P-450 IIC8/9/10 mRNA transcripts and the corresponding protein, while 3-methylcholanthrene was ineffective. Rifampin 18-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-54 1974199-5 1990 Only pretreatment of rabbits with rifampicin, which induces cytochrome P-450 form 3c (P-450IIIA6), significantly increased the microsomal hydroxylation of tolbutamide. Rifampin 34-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 2103248-3 1990 We consider that the association of two antibiotics (rifampin, doxycycline or trimethoprim/sulfamethoxazole) are valid and should be administered until the symptoms disappear and the CSF becomes normal. Rifampin 53-61 colony stimulating factor 2 Homo sapiens 183-186 2185231-7 1990 Rifampin challenge, end-labeling with [gamma-32P]ATP, and selective initiation with a dinucleotide all indicate that the decreased in vitro activity of the wild-type polymerase relative to that of the alc mutants was due to inhibition of elongation, not to any difference in initiation rates. Rifampin 0-8 Alc inhibitor of host transcription Escherichia phage T4 201-204 20702214-3 1990 Phenobarbital and rifampicin were found to increase the levels of P-450 IIC8, 9, 10 mRNA and protein while troleandomycin and 3-methylcholanthrene were ineffective. Rifampin 18-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-79 33806634-6 2021 DDI model performance was evaluated by prediction of clinical studies with rifampicin (CYP2B6 and CYP2C19 inducer), fluvoxamine (CYP2C19 inhibitor) and voriconazole (CYP2B6 and CYP2C19 inhibitor). Rifampin 75-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 33590212-8 2021 Additionally, while rifampin induced CYP3A4 in HepaRG-MPCCs and HepaRG-CCs, only HepaRG-MPCCs showed the dual omeprazole-mediated CYP1A2/3A4 induction as with PHHs. Rifampin 20-28 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-43 33945104-8 2021 These results suggested that TIR8 exerts a protective role in IL-1beta-mediated EMT and potentially represents a new target for RIF treatment. Rifampin 128-131 single Ig and TIR domain containing Rattus norvegicus 29-33 33945104-8 2021 These results suggested that TIR8 exerts a protective role in IL-1beta-mediated EMT and potentially represents a new target for RIF treatment. Rifampin 128-131 interleukin 1 alpha Rattus norvegicus 62-70 29368402-0 2018 Comprehensive PBPK Model of Rifampicin for Quantitative Prediction of Complex Drug-Drug Interactions: CYP3A/2C9 Induction and OATP Inhibition Effects. Rifampin 28-38 solute carrier organic anion transporter family member 1A2 Homo sapiens 126-130 33232732-8 2021 The first pattern (amoxicillin-clavulanate, ceftriaxone) is characterised by high rate of UAP in every department, the second (cloxacillin, rifampin) by high rate of SAP in every department and the third (broad-spectrum beta-lactams) by heterogeneous distribution of SAP/UAP among departments. Rifampin 140-148 SH2 domain containing 1A Homo sapiens 166-169 33763207-2 2020 Drugs such as rifampicin and glibenclamide inhibit BSEP. Rifampin 14-24 ATP binding cassette subfamily B member 11 Homo sapiens 51-55 29368402-3 2018 The parameters for cytochrome P450 (CYP) 3A and CYP2C9 induction by rifampicin were similarly optimized using clinical DDI data with midazolam and tolbutamide as probe substrates, respectively. Rifampin 68-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 29368402-2 2018 Using in silico and in vitro parameters, and reported clinical pharmacokinetic data, rifampicin PBPK model was built and relevant parameters for saturable hepatic uptake and UDP-glucuronosyltransferase (UGT) auto-induction were optimized by fitting. Rifampin 85-95 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 174-201 29368402-6 2018 Altogether, our constructed PBPK model of rifampicin demonstrates the robustness and utility in quantitatively predicting CYP3A/2C9 induction-mediated and/or OATP inhibition-mediated DDIs with victim drugs. Rifampin 42-52 solute carrier organic anion transporter family member 1A2 Homo sapiens 158-162 29368402-2 2018 Using in silico and in vitro parameters, and reported clinical pharmacokinetic data, rifampicin PBPK model was built and relevant parameters for saturable hepatic uptake and UDP-glucuronosyltransferase (UGT) auto-induction were optimized by fitting. Rifampin 85-95 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 203-206 29368402-3 2018 The parameters for cytochrome P450 (CYP) 3A and CYP2C9 induction by rifampicin were similarly optimized using clinical DDI data with midazolam and tolbutamide as probe substrates, respectively. Rifampin 68-78 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-43 16847425-0 2006 Effect of SLCO1B1 polymorphism on induction of CYP3A4 by rifampicin. Rifampin 57-67 solute carrier organic anion transporter family member 1B1 Homo sapiens 10-17 18096695-5 2008 In this study, we showed that activation of PXR by genetic (using a constitutively activated PXR) or pharmacological (using PXR agonist rifampicin) means protected the PXR-overexpressing colon cancer HCT116 cells from deoxycholic acid-induced apoptosis. Rifampin 136-146 nuclear receptor subfamily 1 group I member 2 Homo sapiens 44-47 18096695-9 2008 Treatment with rifampicin in colon cancer LS180 cells, a cell line known to express endogenous PXR, also inhibited apoptosis. Rifampin 15-25 nuclear receptor subfamily 1 group I member 2 Homo sapiens 95-98 16847425-0 2006 Effect of SLCO1B1 polymorphism on induction of CYP3A4 by rifampicin. Rifampin 57-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-53 16847425-1 2006 Rifampicin (rifampin) is a potent inducer of cytochrome P450 (CYP) 3A4. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 45-70 16847425-1 2006 Rifampicin (rifampin) is a potent inducer of cytochrome P450 (CYP) 3A4. Rifampin 12-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 45-70 16847425-3 2006 The present study aimed to investigate the possible association of single nucleotide polymorphisms (SNP) in the SLCO1B1 gene encoding for OATP1B1 with the inducing effect of rifampicin on hepatic CYP3A4. Rifampin 174-184 solute carrier organic anion transporter family member 1B1 Homo sapiens 112-119 16847425-3 2006 The present study aimed to investigate the possible association of single nucleotide polymorphisms (SNP) in the SLCO1B1 gene encoding for OATP1B1 with the inducing effect of rifampicin on hepatic CYP3A4. Rifampin 174-184 solute carrier organic anion transporter family member 1B1 Homo sapiens 138-145 16847425-3 2006 The present study aimed to investigate the possible association of single nucleotide polymorphisms (SNP) in the SLCO1B1 gene encoding for OATP1B1 with the inducing effect of rifampicin on hepatic CYP3A4. Rifampin 174-184 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 196-202 16847425-7 2006 A large intersubject variability existed in the induction of CYP3A4 by rifampicin, but no associations were observed between the variability in induction and any of the polymorphisms studied. Rifampin 71-81 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 61-67 16847425-9 2006 However, the present study had sufficient power to detect only a considerably smaller rifampicin-induced increase in 4beta-hydroxycholesterol in carriers of the SLCO1B1 c.521C allele compared to subjects with the reference genotype. Rifampin 86-96 solute carrier organic anion transporter family member 1B1 Homo sapiens 161-168 34949673-12 2022 Increased susceptibility to RTV-induced liver injury caused by the PXR activator rifampicin was attenuated by HNF4A-AS1 overexpression or HNF1A-AS1 knockdown. Rifampin 81-91 nuclear receptor subfamily 1 group I member 2 Homo sapiens 67-70 34623637-8 2022 For the first time, it was possible to deploy plasma-derived sEV to study CYP3A4 induction beyond rifampicin, a more potent CYP3A4 inducer. Rifampin 98-108 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 124-130 34689256-4 2022 We used 3D PHHs to assess time-dependent expression profiles of 12 prototypic PXR-controlled genes in the time course of 168 h of rifampicin treatment (1 or 10 microM). Rifampin 130-140 nuclear receptor subfamily 1 group I member 2 Homo sapiens 78-81 34742755-7 2022 Immunofluorescent staining of centromere protein B with the micronuclei formed by BDE-47 in HepG2 cells pretreated with ethanol or rifampicin demonstrated selective formation of centromere-containing micronuclei. Rifampin 131-141 centromere protein B Homo sapiens 30-50 34949673-14 2022 Significance Statement HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as lncRNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. Rifampin 290-300 HNF4A antisense RNA 1 Homo sapiens 23-32 34949673-14 2022 Significance Statement HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as lncRNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. Rifampin 290-300 HNF1A antisense RNA 1 Homo sapiens 37-46 34949673-12 2022 Increased susceptibility to RTV-induced liver injury caused by the PXR activator rifampicin was attenuated by HNF4A-AS1 overexpression or HNF1A-AS1 knockdown. Rifampin 81-91 HNF4A antisense RNA 1 Homo sapiens 110-119 34949673-14 2022 Significance Statement HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as lncRNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. Rifampin 290-300 hepatocyte nuclear factor 4 alpha Homo sapiens 144-149 34949673-12 2022 Increased susceptibility to RTV-induced liver injury caused by the PXR activator rifampicin was attenuated by HNF4A-AS1 overexpression or HNF1A-AS1 knockdown. Rifampin 81-91 HNF1A antisense RNA 1 Homo sapiens 138-147 34949673-14 2022 Significance Statement HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as lncRNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. Rifampin 290-300 HNF1 homeobox A Homo sapiens 154-159 34966254-2 2021 Previously we demonstrated that oral or intranasal rifampicin improved the cognition of APP-, tau-, and alpha-synuclein-transgenic mice by reducing the amount of Abeta, tau, and alpha-synuclein oligomers in the brain. Rifampin 51-61 synuclein, alpha Mus musculus 104-119 34949673-14 2022 Significance Statement HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as lncRNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. Rifampin 290-300 nuclear receptor subfamily 1 group I member 2 Homo sapiens 374-377 34949673-14 2022 Significance Statement HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as lncRNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. Rifampin 290-300 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 428-434 34762904-9 2021 Furthermore, we found that CYP3A4 induction by rifampicin augmented NBP-induced cell toxicity and supplementing with GSH or NAC alleviated the oxidative stresses and reactive metabolites caused by 3-OH-NBP. Rifampin 47-57 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 27-33 34888851-2 2022 Rifampin inhibits transporters including organic-anion-transporting polypeptide (OATP)1B and P-glycoprotein (P-gp), and induces enzymes and transporters including cytochrome P450 3A, UDP-glucuronosyltransferase (UGT)1A, and P-gp. Rifampin 0-8 ATP binding cassette subfamily B member 1 Homo sapiens 93-107 34888851-2 2022 Rifampin inhibits transporters including organic-anion-transporting polypeptide (OATP)1B and P-glycoprotein (P-gp), and induces enzymes and transporters including cytochrome P450 3A, UDP-glucuronosyltransferase (UGT)1A, and P-gp. Rifampin 0-8 ATP binding cassette subfamily B member 1 Homo sapiens 109-113 34888851-2 2022 Rifampin inhibits transporters including organic-anion-transporting polypeptide (OATP)1B and P-glycoprotein (P-gp), and induces enzymes and transporters including cytochrome P450 3A, UDP-glucuronosyltransferase (UGT)1A, and P-gp. Rifampin 0-8 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 183-189 34888851-2 2022 Rifampin inhibits transporters including organic-anion-transporting polypeptide (OATP)1B and P-glycoprotein (P-gp), and induces enzymes and transporters including cytochrome P450 3A, UDP-glucuronosyltransferase (UGT)1A, and P-gp. Rifampin 0-8 ATP binding cassette subfamily B member 1 Homo sapiens 224-228 34988253-1 2022 Clarithromycin (CYP inhibitor) can be used instead of azithromycin for nontuberculous mycobacteria therapy in patients requiring CYP substrates to mitigate rifampin"s CYP induction. Rifampin 156-164 peptidylprolyl isomerase G Homo sapiens 129-132 34988253-1 2022 Clarithromycin (CYP inhibitor) can be used instead of azithromycin for nontuberculous mycobacteria therapy in patients requiring CYP substrates to mitigate rifampin"s CYP induction. Rifampin 156-164 peptidylprolyl isomerase G Homo sapiens 167-170 34966254-2 2021 Previously we demonstrated that oral or intranasal rifampicin improved the cognition of APP-, tau-, and alpha-synuclein-transgenic mice by reducing the amount of Abeta, tau, and alpha-synuclein oligomers in the brain. Rifampin 51-61 amyloid beta (A4) precursor protein Mus musculus 162-167 34966254-2 2021 Previously we demonstrated that oral or intranasal rifampicin improved the cognition of APP-, tau-, and alpha-synuclein-transgenic mice by reducing the amount of Abeta, tau, and alpha-synuclein oligomers in the brain. Rifampin 51-61 synuclein, alpha Mus musculus 178-193 34966254-5 2021 Compared with rifampicin and resveratrol alone, the combinatorial medicine significantly improved mouse cognition, reduced amyloid oligomer accumulation, and recovered synaptophysin levels in the hippocampus. Rifampin 14-24 synaptophysin Mus musculus 168-181 34966254-7 2021 Notably, resveratrol alone and the combinatorial medicine, but not rifampicin alone, enhanced the levels of brain-derived neurotrophic factor (BDNF) and its precursor, pro-BDNF, in the hippocampus. Rifampin 67-77 brain derived neurotrophic factor Mus musculus 108-141 34966254-7 2021 Notably, resveratrol alone and the combinatorial medicine, but not rifampicin alone, enhanced the levels of brain-derived neurotrophic factor (BDNF) and its precursor, pro-BDNF, in the hippocampus. Rifampin 67-77 brain derived neurotrophic factor Mus musculus 143-147 34694920-2 2021 Mutation analysis was done by sequencing the rpoB gene encoding RNA polymerase beta-subunit of the rifampicin-resistant mutants. Rifampin 99-109 rpoB Deinococcus radiodurans R1 45-49 34871095-8 2021 MALDI MS imaging analysis of clofazimine, pyrazinamide and rifampicin revealed a drug-specific distribution within different lesion types including cellular granulomas, developing in BALB/c wild-type mice, and necrotic granulomas of BALB/c IL-13tg animals, emphasizing the necessity of pre-clinical models reflecting human pathology. Rifampin 59-69 interleukin 13 Mus musculus 240-245 34562427-1 2021 Post-exposure prophylaxis (PEP) with single-dose rifampicin (SDR) reduces the risk of developing leprosy among contacts of leprosy patients. Rifampin 49-59 prolyl endopeptidase Homo sapiens 27-30 34872459-11 2021 Patients with tuberculosis with older age and genetic polymorphism of CYP3A4, CYP2C9, and CYP2C19 who received long-term rifampicin treatment were more likely to have antituberculosis drug-induced liver injury. Rifampin 121-131 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 70-76 34872459-11 2021 Patients with tuberculosis with older age and genetic polymorphism of CYP3A4, CYP2C9, and CYP2C19 who received long-term rifampicin treatment were more likely to have antituberculosis drug-induced liver injury. Rifampin 121-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 34872459-11 2021 Patients with tuberculosis with older age and genetic polymorphism of CYP3A4, CYP2C9, and CYP2C19 who received long-term rifampicin treatment were more likely to have antituberculosis drug-induced liver injury. Rifampin 121-131 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 90-97 34729944-1 2021 Rifampicin induces both P-glycoprotein (P-gp) and cytochrome P450 3A4 (CYP3A4) through regulating common nuclear receptors (e.g., pregnane X receptor). Rifampin 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 24-38 34729944-1 2021 Rifampicin induces both P-glycoprotein (P-gp) and cytochrome P450 3A4 (CYP3A4) through regulating common nuclear receptors (e.g., pregnane X receptor). Rifampin 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 40-44 34729944-1 2021 Rifampicin induces both P-glycoprotein (P-gp) and cytochrome P450 3A4 (CYP3A4) through regulating common nuclear receptors (e.g., pregnane X receptor). Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-69 34729944-1 2021 Rifampicin induces both P-glycoprotein (P-gp) and cytochrome P450 3A4 (CYP3A4) through regulating common nuclear receptors (e.g., pregnane X receptor). Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 71-77 34729944-1 2021 Rifampicin induces both P-glycoprotein (P-gp) and cytochrome P450 3A4 (CYP3A4) through regulating common nuclear receptors (e.g., pregnane X receptor). Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 130-149 34729944-4 2021 In this study, we used a PBPK modeling approach for the assessment of DDIs between rifampicin and 12 drugs: three sensitive P-gp substrates, seven P-gp-CYP3A4 dual substrates, and two P-gp-CYP3A4 dual substrates and inhibitors. Rifampin 83-93 ATP binding cassette subfamily B member 1 Homo sapiens 124-128 34729944-4 2021 In this study, we used a PBPK modeling approach for the assessment of DDIs between rifampicin and 12 drugs: three sensitive P-gp substrates, seven P-gp-CYP3A4 dual substrates, and two P-gp-CYP3A4 dual substrates and inhibitors. Rifampin 83-93 ATP binding cassette subfamily B member 1 Homo sapiens 147-151 34729944-4 2021 In this study, we used a PBPK modeling approach for the assessment of DDIs between rifampicin and 12 drugs: three sensitive P-gp substrates, seven P-gp-CYP3A4 dual substrates, and two P-gp-CYP3A4 dual substrates and inhibitors. Rifampin 83-93 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 152-158 34729944-4 2021 In this study, we used a PBPK modeling approach for the assessment of DDIs between rifampicin and 12 drugs: three sensitive P-gp substrates, seven P-gp-CYP3A4 dual substrates, and two P-gp-CYP3A4 dual substrates and inhibitors. Rifampin 83-93 ATP binding cassette subfamily B member 1 Homo sapiens 184-188 34729944-4 2021 In this study, we used a PBPK modeling approach for the assessment of DDIs between rifampicin and 12 drugs: three sensitive P-gp substrates, seven P-gp-CYP3A4 dual substrates, and two P-gp-CYP3A4 dual substrates and inhibitors. Rifampin 83-93 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 189-195 34729944-5 2021 A 3.5-fold increase of intestinal P-gp abundance was incorporated in the PBPK models to account for rifampicin-mediated P-gp induction at steady state. Rifampin 100-110 ATP binding cassette subfamily B member 1 Homo sapiens 34-38 34729944-5 2021 A 3.5-fold increase of intestinal P-gp abundance was incorporated in the PBPK models to account for rifampicin-mediated P-gp induction at steady state. Rifampin 100-110 ATP binding cassette subfamily B member 1 Homo sapiens 120-124 34282472-2 2021 Two drug-drug interaction studies, one with acid-reducing agents, esomeprazole (proton pump inhibitor (PPI)) and ranitidine (histamine-2 (H2) antagonist), and the other with potent CYP3A-modulating agents, itraconazole (CYP3A inhibitor) and rifampin (CYP3A inducer), were performed. Rifampin 241-249 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 251-256 34722780-5 2021 Moreover, IFI44L was significantly upregulated, and it restricted the intracellular survival of M. tuberculosis H37Rv strains at 72 h after rifampicin treatment. Rifampin 140-150 interferon induced protein 44 like Homo sapiens 10-16 34529779-0 2021 Pharmacokinetics and safety of high-dose rifampicin in children with TB: the Opti-Rif trial. Rifampin 41-51 ras homolog family member F, filopodia associated Homo sapiens 82-85 34278601-16 2021 Most PHSF patients have UGT1A1 deficiency, which may be the target of rifampicin. Rifampin 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 34664792-3 2021 Phenotyping studies indicated cytochrome P450 (CYP) 3A are the major CYP isoform responsible for zanubrutinib metabolism, which was confirmed by a clinical DDI study with itraconazole and rifampin. Rifampin 188-196 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 30-54 34686228-0 2021 Impact of the bacillary load on the accuracy of rifampicin resistance results by Xpert MTB/RIF. Rifampin 48-58 ras homolog family member F, filopodia associated Homo sapiens 92-95 34722780-6 2021 Individuals with cutaneous tuberculosis (CTB) were found to have significantly higher IFI44L expression after 6 months of rifampicin therapy than after only 1 month. Rifampin 122-132 interferon induced protein 44 like Homo sapiens 86-92 34691435-0 2021 The role of C-Reactive protein as an inflammatory marker to predict prolonged QTc interval in rifampicin-resistant tuberculosis patients: A case-control study. Rifampin 94-104 C-reactive protein Homo sapiens 12-30 34491807-6 2021 Through microcalorimetric measurements, adjunctive rifampicin resulted in decreased metabolic activity and extended lag phase for all clinical GAS strains tested (p<0.05). Rifampin 51-61 gastrin Homo sapiens 143-146 34491807-7 2021 In addition, a case report is presented of adjunctive rifampicin treatment in an NSTI case with persistent GAS tissue infection. Rifampin 54-64 gastrin Homo sapiens 107-110 34491807-8 2021 Conclusion: The findings of this study demonstrate that adjunctive rifampicin enhances clearance of GAS biofilm in an in vitro tissue infection model. Rifampin 67-77 gastrin Homo sapiens 100-103 34691435-1 2021 Background: long-term use of anti-tuberculosis drugs (ATD) increases the risk of QTc prolongation, while C-reactive protein (CRP) can be used as an inflammatory marker of Mycobacterium tuberculosis infection.Objective: correlation of CRP on the QTc interval in Rifampicin-resistant tuberculosis (RR-TB) patients with the short regimen. Rifampin 261-271 C-reactive protein Homo sapiens 105-123 34691435-1 2021 Background: long-term use of anti-tuberculosis drugs (ATD) increases the risk of QTc prolongation, while C-reactive protein (CRP) can be used as an inflammatory marker of Mycobacterium tuberculosis infection.Objective: correlation of CRP on the QTc interval in Rifampicin-resistant tuberculosis (RR-TB) patients with the short regimen. Rifampin 261-271 C-reactive protein Homo sapiens 125-128 34691435-1 2021 Background: long-term use of anti-tuberculosis drugs (ATD) increases the risk of QTc prolongation, while C-reactive protein (CRP) can be used as an inflammatory marker of Mycobacterium tuberculosis infection.Objective: correlation of CRP on the QTc interval in Rifampicin-resistant tuberculosis (RR-TB) patients with the short regimen. Rifampin 261-271 C-reactive protein Homo sapiens 234-237 34273250-9 2021 Other than a simulated ~60% exposure reduction with strong CYP3A/2C inducers such as rifampin, other DDI liabilities were minimal and considered not clinically relevant. Rifampin 85-93 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 59-66 34619012-6 2021 Furthermore, when (14 C)epyrifenacil was administered with OATP inhibitors, the liver/plasma ratio of S-3100-CA was significantly decreased by rifampicin, an Oatp1a1/Oatp1a4 inhibitor in mice, but not by digoxin, an Oatp1a4-specific inhibitor. Rifampin 143-153 solute carrier organic anion transporter family member 1A2 Homo sapiens 59-63 34619012-6 2021 Furthermore, when (14 C)epyrifenacil was administered with OATP inhibitors, the liver/plasma ratio of S-3100-CA was significantly decreased by rifampicin, an Oatp1a1/Oatp1a4 inhibitor in mice, but not by digoxin, an Oatp1a4-specific inhibitor. Rifampin 143-153 solute carrier organic anion transporter family, member 1a1 Mus musculus 158-165 34619012-6 2021 Furthermore, when (14 C)epyrifenacil was administered with OATP inhibitors, the liver/plasma ratio of S-3100-CA was significantly decreased by rifampicin, an Oatp1a1/Oatp1a4 inhibitor in mice, but not by digoxin, an Oatp1a4-specific inhibitor. Rifampin 143-153 solute carrier organic anion transporter family, member 1a4 Mus musculus 166-173 34581255-0 2022 Treatment of spinal tuberculosis in rabbits using bovine serum albumin nanoparticles loaded with isoniazid and rifampicin. Rifampin 111-121 albumin Oryctolagus cuniculus 57-70 34581255-1 2022 OBJECTIVE: To evaluate the clinical efficacy of bovine serum albumin nanoparticles loaded with isoniazid and rifampicin (INH-RFP-BSA-NPs) in the treatment of spinal tuberculosis in rabbits. Rifampin 109-119 albumin Oryctolagus cuniculus 55-68 34498807-2 2022 Three drug-drug interactions studies were conducted in healthy subjects to evaluate the effect of P-glycoprotein (P-gp) modulation (Study 1: P-gp inhibition by itraconazole; Study 2: P-gp induction by rifampin) on filgotinib pharmacokinetics and the potential of filgotinib to impact exposure of metformin, an organic cation transporter (OCT) 2 and multidrug and toxin extrusion (MATE) 1/2K substrate (Study 3). Rifampin 201-209 ATP binding cassette subfamily B member 1 Homo sapiens 183-187 34145979-6 2021 Based on these results, the strong CYP3A inhibitor itraconazole and inducer rifampin significantly influenced the PK of mobocertinib and its active metabolites. Rifampin 76-84 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-40 34518943-5 2021 However, CYP inhibitory effect of INH gets masked by opposite enzyme-inducing effect of rifampicin, when used in combination. Rifampin 88-98 peptidylprolyl isomerase G Homo sapiens 9-12 34171773-7 2021 ECg and EGCg also strongly inhibited AADAC-mediated rifampicin hydrolase activity in human liver microsomes with IC50 values of 2.2 +- 1.4 and 1.7 +- 0.4 muM, respectively, whereas it weakly inhibited p-nitrophenyl acetate hydrolase activity, which is catalyzed by AADAC, CES1, and CES2. Rifampin 52-62 arylacetamide deacetylase Homo sapiens 37-42 34485328-1 2021 Background: The Xpert Mycobacterium tuberculosis/rifampin (MTB/RIF) assay has shown good diagnostic efficacy in brushing and biopsy tissue samples from patients with tracheobronchial tuberculosis (TBTB). Rifampin 49-57 ras homolog family member F, filopodia associated Homo sapiens 63-66 34447304-6 2021 We used the PBMCs collected from three healthy control participants with no known history of adverse drug reaction and two patients with a rifampicin-associated drug reaction with eosinophilia and systemic symptoms (DRESS), confirmed on IFN-gamma ELISpot assay. Rifampin 139-149 interferon gamma Homo sapiens 237-246 34447304-8 2021 Results: In a human immunodeficiency virus (HIV) negative and a positive rifampicin-associated DRESS with positive ex vivo IFN-gamma ELISpot assay, use of 10- and 100-fold Cmax drug concentrations decreased spot forming units/million cells by 32-100%, and this corresponded to cell cytotoxicity of more than 40 and 20% using an LDH assay and 7-AAD cell viability staining, respectively. Rifampin 73-83 interferon gamma Homo sapiens 123-132 34446483-3 2021 Post-exposure prophylaxis with single-dose rifampicin (SDR-PEP) reduces the risk of developing leprosy when administered to screened contacts of patients. Rifampin 43-53 caveolae associated protein 2 Homo sapiens 55-58 34446483-3 2021 Post-exposure prophylaxis with single-dose rifampicin (SDR-PEP) reduces the risk of developing leprosy when administered to screened contacts of patients. Rifampin 43-53 prolyl endopeptidase Homo sapiens 59-62 34445158-0 2021 Nasal Rifampicin Improves Cognition in a Mouse Model of Dementia with Lewy Bodies by Reducing alpha-Synuclein Oligomers. Rifampin 6-16 synuclein, alpha Mus musculus 94-109 34445158-3 2021 Previously, we demonstrated that in APP- and tau-transgenic mice, oral or intranasal rifampicin reduced brain Abeta and tau oligomers and improved mouse cognition. Rifampin 85-95 amyloid beta (A4) precursor protein Mus musculus 110-115 34445158-5 2021 Rifampicin was intranasally administered to 6-month-old A53T-mutant alpha-synuclein-transgenic mice at 0.1 mg/day for 1 month. Rifampin 0-10 synuclein, alpha Mus musculus 68-83 34445158-9 2021 Intranasal rifampicin significantly reduced the levels of (pSer129) alpha-synuclein in the hippocampus and alpha-synuclein oligomers in the visual cortex and hippocampus. Rifampin 11-21 synuclein, alpha Mus musculus 68-83 34445158-9 2021 Intranasal rifampicin significantly reduced the levels of (pSer129) alpha-synuclein in the hippocampus and alpha-synuclein oligomers in the visual cortex and hippocampus. Rifampin 11-21 synuclein, alpha Mus musculus 107-122 34444675-7 2021 Rifampicin is a well-known human PXR ligand that has been used to treat intractable pruritus in severe cholestasis. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 33-36 34386782-4 2021 We recruited patients with sputum Xpert MTB/RIF-positive rifampicin-susceptible pulmonary tuberculosis. Rifampin 57-67 ras homolog family member F, filopodia associated Homo sapiens 44-47 34267345-0 2022 Itraconazole and rifampicin, as CYP3A modulators but not P-gp modulators, affect the pharmacokinetics of almonertinib and active metabolite HAS-719 in healthy volunteers. Rifampin 17-27 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-37 34302035-8 2021 The MP-evolved strains showed cross-resistance to rifampicin and clarithromycin owing to the acquisition of a mutation in the intergenic region of the Rv2864c ortholog, which encodes a penicillin-binding protein, at an early stage. Rifampin 50-60 penicillin-binding lipoprotein Mycobacterium tuberculosis H37Rv 151-158 34267345-3 2022 In this study, we investigated the effects of CYP3A inhibitor itraconazole and CYP3A inducer rifampicin on the pharmacokinetics of almonertinib and HAS-719 in 64 healthy volunteers. Rifampin 93-103 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 79-84 34267345-7 2022 Co-administration of rifampicin in Beagle dogs reduced the fecal recovery of almonertinib and HAS-719, and markedly increased the levels of metabolites derived from further metabolism of HAS-719, which was consistent with human plasma data, suggesting that although rifampicin was also a potent inducer of P-gp, the pharmacokinetic alternation of HAS-719 was mainly due to its further metabolism but not excretion changes. Rifampin 21-31 phosphoglycolate phosphatase Homo sapiens 306-310 34267345-7 2022 Co-administration of rifampicin in Beagle dogs reduced the fecal recovery of almonertinib and HAS-719, and markedly increased the levels of metabolites derived from further metabolism of HAS-719, which was consistent with human plasma data, suggesting that although rifampicin was also a potent inducer of P-gp, the pharmacokinetic alternation of HAS-719 was mainly due to its further metabolism but not excretion changes. Rifampin 266-276 phosphoglycolate phosphatase Homo sapiens 306-310 34217369-0 2021 Hepatoprotective activity of melittin on isoniazid- and rifampicin-induced liver injuries in male albino rats. Rifampin 56-66 melittin Apis mellifera 29-37 34217369-1 2021 BACKGROUND: The present study investigated the ameliorative effect of melittin, a major polypeptide in the venom of honeybee (Apis mellifera), on isoniazid-(INH) and rifampicin-(RIF) induced hepatotoxicity in male albino rats. Rifampin 166-176 melittin Apis mellifera 70-78 34217369-1 2021 BACKGROUND: The present study investigated the ameliorative effect of melittin, a major polypeptide in the venom of honeybee (Apis mellifera), on isoniazid-(INH) and rifampicin-(RIF) induced hepatotoxicity in male albino rats. Rifampin 178-181 melittin Apis mellifera 70-78 34143391-8 2021 The simulations of CYP inducers predicted that the strongest interaction would occur with rifampin, with the AUC decreasing to 0.36 of the control value, whereas the simulations of CYP inhibitors predicted that the greatest effect would occur with fluconazole, with a 1.43-fold increase in AUC. Rifampin 90-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 34177592-0 2021 Rifampicin Induces Gene, Protein, and Activity of P-Glycoprotein (ABCB1) in Human Precision-Cut Intestinal Slices. Rifampin 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 50-64 34267886-3 2021 Subsequent assay of this library led to the identification of a series of potent hPXR agonists, showing better efficacy than that of typical hPXR agonist, rifampicin. Rifampin 155-165 nuclear receptor subfamily 1 group I member 2 Homo sapiens 81-85 34267886-3 2021 Subsequent assay of this library led to the identification of a series of potent hPXR agonists, showing better efficacy than that of typical hPXR agonist, rifampicin. Rifampin 155-165 nuclear receptor subfamily 1 group I member 2 Homo sapiens 141-145 34177592-0 2021 Rifampicin Induces Gene, Protein, and Activity of P-Glycoprotein (ABCB1) in Human Precision-Cut Intestinal Slices. Rifampin 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 66-71 34177592-7 2021 Rifampicin significantly increased gene expression, protein levels, and efflux activity of ABCB1. Rifampin 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 91-96 34154362-12 2021 CONCLUSIONS: Our meta-analysis has uncovered an association between CYP2E1 RsaI/PstI polymorphisms and ATDILI, especially among patients who receive a four-drug (INH + RIF + PZA + EMB) or three-drug (INH + RIF + PZA) anti-TB treatment regimen. Rifampin 206-209 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 68-74 34154362-12 2021 CONCLUSIONS: Our meta-analysis has uncovered an association between CYP2E1 RsaI/PstI polymorphisms and ATDILI, especially among patients who receive a four-drug (INH + RIF + PZA + EMB) or three-drug (INH + RIF + PZA) anti-TB treatment regimen. Rifampin 206-209 serine peptidase inhibitor Kazal type 1 Homo sapiens 80-84 34458006-3 2021 Combined administrations of 2-allyl-2-isopropylacetamide and rifampicin in rabbits halved hepatic PPOX activity, resulting in increased accumulation of a potentially neurotoxic heme precursor, lipid peroxidation, inflammation, and hepatocyte cytoplasmic stress. Rifampin 61-71 protoporphyrinogen oxidase Oryctolagus cuniculus 98-102 34223120-0 2021 TPR1, a novel rifampicin derivative, demonstrates efficacy alone and in combination with doxycycline against the NIAID Category A priority pathogen Francisella tularensis. Rifampin 14-24 tetratricopeptide repeat domain 1 Mus musculus 0-4 34069974-7 2021 In addition, we showed that the expression of MDR1 and CYP3A4 genes increased synergistically after 24 h of rifampicin and MK-4 co-treatment. Rifampin 108-118 ATP binding cassette subfamily B member 1 Homo sapiens 46-50 34069974-7 2021 In addition, we showed that the expression of MDR1 and CYP3A4 genes increased synergistically after 24 h of rifampicin and MK-4 co-treatment. Rifampin 108-118 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 55-61 34223120-2 2021 Methods: We utilized a systematic analysis of antibacterial potency, extent of dissemination by analysis of bacterial burden in a secondary vital organ, and survival rates to assess the efficacy of a novel rifampicin derivative, TPR1. Rifampin 206-216 tetratricopeptide repeat domain 1 Mus musculus 229-233 35460165-2 2022 This phase 1, open-label, randomized, two-way crossover study (NCT04121078) evaluated the effect of single-dose intravenous rifampin, a potent inhibitor of the organic anion transporting polypeptides (OATP) 1B1 and 1B3, on the pharmacokinetics and safety of trazpiroben in healthy adults. Rifampin 124-132 solute carrier organic anion transporter family member 1B1 Homo sapiens 160-218 34345857-9 2021 For the CYP2B/C/D"s, phenobarbital and rifampicin caused increases in expression. Rifampin 39-49 cytochrome P450 family 2 subfamily B member 7, pseudogene Homo sapiens 8-13 34345857-11 2021 Both phenobarbital, rifampicin and omeprazole increased CYP3A expression in PHH and PPH. Rifampin 20-30 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 56-61 35370222-1 2022 We encountered cases in which the anticoagulant effects of warfarin (CYP2C9 substrate) were reversibly attenuated by the concomitant administration of rifampicin or bosentan, which are potent pregnane X receptor (PXR) ligands. Rifampin 151-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 35370222-1 2022 We encountered cases in which the anticoagulant effects of warfarin (CYP2C9 substrate) were reversibly attenuated by the concomitant administration of rifampicin or bosentan, which are potent pregnane X receptor (PXR) ligands. Rifampin 151-161 nuclear receptor subfamily 1 group I member 2 Homo sapiens 192-211 35370222-1 2022 We encountered cases in which the anticoagulant effects of warfarin (CYP2C9 substrate) were reversibly attenuated by the concomitant administration of rifampicin or bosentan, which are potent pregnane X receptor (PXR) ligands. Rifampin 151-161 nuclear receptor subfamily 1 group I member 2 Homo sapiens 213-216 35292967-7 2022 Correlation between AUCR of pitavastatin, and Cmax R or AUCR of CP-I were consistent between this study and our previous study using rifampicin as an OATP1B1/1B3 inhibitor. Rifampin 133-143 solute carrier organic anion transporter family member 1B1 Homo sapiens 150-161 35460165-9 2022 This is also the first assessment of the utility of CPI and CPIII as endogenous biomarkers of OATP1B1/1B3 inhibition after a single intravenous dose of rifampin. Rifampin 152-160 solute carrier organic anion transporter family member 1B1 Homo sapiens 94-105 35636769-7 2022 In contrast, multiple doses of 15 mg/kg rifampin (RIF), a pregnane X receptor (PXR) agonist, had no effect on hepatic OATP1B protein although it induced the intestinal P-gp and MR2 proteins by ~2-fold. Rifampin 40-48 glycerol-3-phosphate phosphatase Macaca fascicularis 168-172 35238961-8 2022 Rifampicin increased CYP2C19-mediated omeprazole metabolism. Rifampin 0-10 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 21-28 35238961-10 2022 Both fluconazole and rifampicin altered CYP3A4 activity whereas no change of CYP2D6 activity was observed at all. Rifampin 21-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-46 35636769-7 2022 In contrast, multiple doses of 15 mg/kg rifampin (RIF), a pregnane X receptor (PXR) agonist, had no effect on hepatic OATP1B protein although it induced the intestinal P-gp and MR2 proteins by ~2-fold. Rifampin 50-53 glycerol-3-phosphate phosphatase Macaca fascicularis 168-172 35616006-7 2022 The AUC of asciminib was marginally decreased by concomitant administration with the strong CYP3A inducer rifampicin (by ~13-15%) and the strong P-gp inhibitor quinidine (by ~13-16%). Rifampin 106-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 92-97 35570332-0 2022 Robust PBPK Model of Rifampicin for Predicting Drug-Drug Interactions via P-gp Induction and Inhibition in Intestine, Liver, and Kidney. Rifampin 21-31 ATP binding cassette subfamily B member 1 Homo sapiens 74-78 35579824-6 2022 The relative contributions of different cytochromes P450 (CYPs), mainly CYP3A4 and CYP2B6, involved in esketamine/noresketamine clearance was captured correctly in the IN-PBPK model using the DDI clinical studies of intranasal esketamine with clarithromycin and rifampicin and a published DDI study of oral esketamine with ticlopidine. Rifampin 262-272 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 72-78 35570332-1 2022 P-glycoprotein (P-gp) is an efflux transporter that plays an important role in pharmacokinetics of its substrate, and P-gp activities can be altered by induction and inhibition effects of rifampicin. Rifampin 188-198 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 35570332-1 2022 P-glycoprotein (P-gp) is an efflux transporter that plays an important role in pharmacokinetics of its substrate, and P-gp activities can be altered by induction and inhibition effects of rifampicin. Rifampin 188-198 ATP binding cassette subfamily B member 1 Homo sapiens 16-20 35570332-1 2022 P-glycoprotein (P-gp) is an efflux transporter that plays an important role in pharmacokinetics of its substrate, and P-gp activities can be altered by induction and inhibition effects of rifampicin. Rifampin 188-198 ATP binding cassette subfamily B member 1 Homo sapiens 118-122 35570332-2 2022 This study aimed to establish a physiologically based pharmacokinetic (PBPK) model of rifampicin to predict the P-gp-mediated drug-drug interactions (DDIs) and assess the DDI impact in the intestine, liver, and kidney. Rifampin 86-96 ATP binding cassette subfamily B member 1 Homo sapiens 112-116 35570332-3 2022 The induction and inhibition parameters of rifampicin for P-gp were estimated using 2 of 7 DDI cases of rifampicin and digoxin and incorporated into our previously constructed PBPK model of rifampicin. Rifampin 43-53 ATP binding cassette subfamily B member 1 Homo sapiens 58-62 35570332-3 2022 The induction and inhibition parameters of rifampicin for P-gp were estimated using 2 of 7 DDI cases of rifampicin and digoxin and incorporated into our previously constructed PBPK model of rifampicin. Rifampin 104-114 ATP binding cassette subfamily B member 1 Homo sapiens 58-62 35570332-3 2022 The induction and inhibition parameters of rifampicin for P-gp were estimated using 2 of 7 DDI cases of rifampicin and digoxin and incorporated into our previously constructed PBPK model of rifampicin. Rifampin 190-200 ATP binding cassette subfamily B member 1 Homo sapiens 58-62 35570332-5 2022 Based on the established PBPK model, following repeated dosing of 600 mg rifampicin, the deduced net effect was a ~3-fold induction in P-gp activities in the intestine, liver, and kidney. Rifampin 73-83 ATP binding cassette subfamily B member 1 Homo sapiens 135-139 35570332-9 2022 These findings demonstrate that our rifampicin model can be applicable to quantitatively predict the net impact of P-gp induction and/or inhibition on diverse P-gp substrates and investigate the magnitude of DDIs in each tissue. Rifampin 36-46 ATP binding cassette subfamily B member 1 Homo sapiens 115-119 35569070-0 2022 Long-term efficacy and safety of rifampin in the treatment of a patient carrying a CYP24A1 loss-of-function variant. Rifampin 33-41 cytochrome P450 family 24 subfamily A member 1 Homo sapiens 83-90 35570332-9 2022 These findings demonstrate that our rifampicin model can be applicable to quantitatively predict the net impact of P-gp induction and/or inhibition on diverse P-gp substrates and investigate the magnitude of DDIs in each tissue. Rifampin 36-46 ATP binding cassette subfamily B member 1 Homo sapiens 159-163 35569070-2 2022 Rifampin is an antituberculosis drug that is a potent inducer of cytochrome P450 3 subfamily A member 4 (CYP3A4), involved in an alternative catabolic pathway of vitamin D. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-103 35452411-7 2022 In 100% of rifampin-resistant isolates, mutations were found in the rifampin resistance-determining region (RRDR) of the rpoB, with S450L substitution being the most common, especially in MDRs (77.8%, 7/9). Rifampin 11-19 DNA-directed RNA polymerase subunit beta Mycobacterium tuberculosis H37Rv 121-125 35569070-2 2022 Rifampin is an antituberculosis drug that is a potent inducer of cytochrome P450 3 subfamily A member 4 (CYP3A4), involved in an alternative catabolic pathway of vitamin D. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-111 35536601-8 2022 Activation of the nuclear receptors PXR and AHR was demonstrated via the induction of specific CYP isoenzymes by rifampicin, pregnenolone-16a-carbonitrile and beta-naphthoflavone. Rifampin 113-123 nuclear receptor subfamily 1 group I member 2 Homo sapiens 36-39 35536601-8 2022 Activation of the nuclear receptors PXR and AHR was demonstrated via the induction of specific CYP isoenzymes by rifampicin, pregnenolone-16a-carbonitrile and beta-naphthoflavone. Rifampin 113-123 aryl hydrocarbon receptor Homo sapiens 44-47 35536601-8 2022 Activation of the nuclear receptors PXR and AHR was demonstrated via the induction of specific CYP isoenzymes by rifampicin, pregnenolone-16a-carbonitrile and beta-naphthoflavone. Rifampin 113-123 peptidylprolyl isomerase G Homo sapiens 95-98 35625816-0 2022 C9orf72 Hexanucleotide Repeat Expansion-Related Neuropathology Is Attenuated by Nasal Rifampicin in Mice. Rifampin 86-96 C9orf72-SMCR8 complex subunit Homo sapiens 0-7 35625816-5 2022 Rifampicin treatment reduced the formation of RNA foci and cytoplasmic inclusions containing DPRs or phosphorylated TDP-43, and furthermore, the levels of phosphorylated double-strand RNA-dependent protein kinase (PKR) that regulates repeat-associated non-ATG (RAN) translation. Rifampin 0-10 TAR DNA binding protein Mus musculus 116-122 35625816-5 2022 Rifampicin treatment reduced the formation of RNA foci and cytoplasmic inclusions containing DPRs or phosphorylated TDP-43, and furthermore, the levels of phosphorylated double-strand RNA-dependent protein kinase (PKR) that regulates repeat-associated non-ATG (RAN) translation. Rifampin 0-10 eukaryotic translation initiation factor 2-alpha kinase 2 Mus musculus 214-217 35625816-5 2022 Rifampicin treatment reduced the formation of RNA foci and cytoplasmic inclusions containing DPRs or phosphorylated TDP-43, and furthermore, the levels of phosphorylated double-strand RNA-dependent protein kinase (PKR) that regulates repeat-associated non-ATG (RAN) translation. Rifampin 0-10 RAN, member RAS oncogene family Mus musculus 234-259 35625816-5 2022 Rifampicin treatment reduced the formation of RNA foci and cytoplasmic inclusions containing DPRs or phosphorylated TDP-43, and furthermore, the levels of phosphorylated double-strand RNA-dependent protein kinase (PKR) that regulates repeat-associated non-ATG (RAN) translation. Rifampin 0-10 RAN, member RAS oncogene family Mus musculus 261-264 35625816-7 2022 Our findings suggest a therapeutic potential of nasal rifampicin in the prevention of C9orf72-linked neurodegenerative disorders. Rifampin 54-64 C9orf72-SMCR8 complex subunit Homo sapiens 86-93 35506332-5 2022 The model under-predicted rifampin induction by 100% (approximate 6.7-fold decrease in AUC and approximate 2.6-fold decrease in Cmax in DDI study), presumably reflecting non-CYP3A4 mechanisms being impacted. Rifampin 26-34 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 174-180 35428895-8 2022 Additionally, in the presence of strong or moderate CYP3A4 inducers, rifampicin and efavirenz, ipatasertib exposures were predicted to decrease by 86% and 74%, respectively. Rifampin 69-79 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 52-58 35241487-1 2022 Cyclosporine A (CsA) and rifampin are potent inhibitors of organic anion transporting polypeptide (OATP) 1B1 and are widely used to assess the risk for drug-drug interactions. Rifampin 25-33 solute carrier organic anion transporter family member 1B1 Homo sapiens 59-108 35241487-8 2022 Rifampin also displayed preincubation time-dependent inhibition of OATP1B1, albeit the extent of enhancement was only 2-fold. Rifampin 0-8 solute carrier organic anion transporter family member 1B1 Homo sapiens 67-74 35435627-2 2022 Rifampicin (RIF) is a strong index CYP3A4 inducer. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-41 35435627-2 2022 Rifampicin (RIF) is a strong index CYP3A4 inducer. Rifampin 12-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-41 35510231-6 2022 Furthermore, although pretreatment with the CYP3A inducer rifampicin or the substitution of glucose with galactose in the growth media significantly augmented the loss of cell viability elicited by dronedarone and poyendarone, a lower loss of cell viability was consistently observed in poyendarone across all concentrations. Rifampin 58-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 44-49 35455390-10 2022 Rifampicin effectively blocked both the Oatp-mediated influx and the Mrp2/3-related efflux of 99mTc-mebrofenin. Rifampin 0-10 ATP binding cassette subfamily C member 2 Rattus norvegicus 69-75 35229613-2 2022 We showed previously that activation of pregnane X receptor (PXR) by rifampicin elevates 24-hour blood pressure (BP) and plasma 4beta-hydroxycholesterol (4betaHC), agonist for liver X receptor (LXR). Rifampin 69-79 nuclear receptor subfamily 1 group I member 2 Homo sapiens 40-59 35229613-2 2022 We showed previously that activation of pregnane X receptor (PXR) by rifampicin elevates 24-hour blood pressure (BP) and plasma 4beta-hydroxycholesterol (4betaHC), agonist for liver X receptor (LXR). Rifampin 69-79 nuclear receptor subfamily 1 group I member 2 Homo sapiens 61-64 35230741-6 2022 Consistent with the proposed metabolic pathway of belumosudil, the strong CYP3A4 inducer rifampicin significantly decreased exposure of belumosudil and KD025m2 and increased KD025m1 exposure. Rifampin 89-99 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 74-80 35146537-7 2022 We also identified other variants which were associated with adverse effects in isoniazid and rifampicin (CYP2E1; hepatotoxicity). Rifampin 94-104 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 106-112 35605912-3 2022 Then two clinical studies were performed to investigate the effects of itraconazole and rifampicin (potent CYP3A4/5 inhibitor and inducer, respectively) on the pharmacokinetics of youkenafil and its main metabolite, N-desethyl youkenafil (M1). Rifampin 88-98 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 107-115 35229613-3 2022 Methods and Results In combined "PXR activation data set" (n=62) of 4 clinical trials, 1 week rifampicin dosing increased office systolic BP (SBP) by 3.1 mm Hg, DBP 1.8 mm Hg, and mean arterial pressure 2.2 mm Hg in comparison with placebo (P<0.01). Rifampin 94-104 nuclear receptor subfamily 1 group I member 2 Homo sapiens 33-36 35165925-1 2022 AIMS: Clinical drug interaction studies with itraconazole and rifampicin demonstrated acalabrutinib is a sensitive substrate of CYP3A. Rifampin 62-72 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 128-133 35452411-7 2022 In 100% of rifampin-resistant isolates, mutations were found in the rifampin resistance-determining region (RRDR) of the rpoB, with S450L substitution being the most common, especially in MDRs (77.8%, 7/9). Rifampin 68-76 DNA-directed RNA polymerase subunit beta Mycobacterium tuberculosis H37Rv 121-125 35045794-0 2022 4-Phenylbutyrate protects against rifampin-induced liver injury via regulating MRP2 ubiquitination through inhibiting endoplasmic reticulum stress. Rifampin 34-42 ATP binding cassette subfamily C member 2 Homo sapiens 79-83 35544702-0 2022 Tanshinone IIA alleviate rifampicin-induced cholestasis by regulating the expression and function of bile salt export pump. Rifampin 25-35 ATP binding cassette subfamily B member 11 Rattus norvegicus 101-122 35062050-13 2022 In the NAFLD model, Omeprazole maintained an expected induction of CYP1A1, however Phenytoin and Rifampicin showed elevated induction of CYP2B6 and CYP2C9 compared to healthy cultures. Rifampin 97-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 35062050-12 2022 We then exposed the NAFLD model to the drug inducers rifampicin, Omeprazole, and Phenytoin as activators of transcription factors pregnane X receptor (PXR), aryl hydrocarbon receptor (AHR) and constitutive androstane receptor (CAR), respectively. Rifampin 53-63 nuclear receptor subfamily 1 group I member 2 Homo sapiens 130-149 35062050-12 2022 We then exposed the NAFLD model to the drug inducers rifampicin, Omeprazole, and Phenytoin as activators of transcription factors pregnane X receptor (PXR), aryl hydrocarbon receptor (AHR) and constitutive androstane receptor (CAR), respectively. Rifampin 53-63 nuclear receptor subfamily 1 group I member 2 Homo sapiens 151-154 35062050-12 2022 We then exposed the NAFLD model to the drug inducers rifampicin, Omeprazole, and Phenytoin as activators of transcription factors pregnane X receptor (PXR), aryl hydrocarbon receptor (AHR) and constitutive androstane receptor (CAR), respectively. Rifampin 53-63 aryl hydrocarbon receptor Homo sapiens 157-182 35062050-12 2022 We then exposed the NAFLD model to the drug inducers rifampicin, Omeprazole, and Phenytoin as activators of transcription factors pregnane X receptor (PXR), aryl hydrocarbon receptor (AHR) and constitutive androstane receptor (CAR), respectively. Rifampin 53-63 aryl hydrocarbon receptor Homo sapiens 184-187 35062050-12 2022 We then exposed the NAFLD model to the drug inducers rifampicin, Omeprazole, and Phenytoin as activators of transcription factors pregnane X receptor (PXR), aryl hydrocarbon receptor (AHR) and constitutive androstane receptor (CAR), respectively. Rifampin 53-63 nuclear receptor subfamily 1 group I member 3 Homo sapiens 193-225 35062050-12 2022 We then exposed the NAFLD model to the drug inducers rifampicin, Omeprazole, and Phenytoin as activators of transcription factors pregnane X receptor (PXR), aryl hydrocarbon receptor (AHR) and constitutive androstane receptor (CAR), respectively. Rifampin 53-63 nuclear receptor subfamily 1 group I member 3 Homo sapiens 227-230 35062050-13 2022 In the NAFLD model, Omeprazole maintained an expected induction of CYP1A1, however Phenytoin and Rifampicin showed elevated induction of CYP2B6 and CYP2C9 compared to healthy cultures. Rifampin 97-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 34985190-0 2022 Two novel ATP8B1 mutations involved in progressive familial intrahepatic cholestasis type 1 that is ameliorated by rifampicin: a case report. Rifampin 115-125 ATPase phospholipid transporting 8B1 Homo sapiens 10-16 34985190-0 2022 Two novel ATP8B1 mutations involved in progressive familial intrahepatic cholestasis type 1 that is ameliorated by rifampicin: a case report. Rifampin 115-125 ATPase phospholipid transporting 8B1 Homo sapiens 51-91 35203506-0 2022 Nasal Rifampicin Halts the Progression of Tauopathy by Inhibiting Tau Oligomer Propagation in Alzheimer Brain Extract-Injected Mice. Rifampin 6-16 microtubule associated protein tau Homo sapiens 66-69 35203506-3 2022 We previously showed that intranasal rifampicin inhibits tau oligomer accumulation and improves cognition in tauopathy mice. Rifampin 37-47 microtubule associated protein tau Homo sapiens 57-60 35203506-8 2022 Rifampicin treatment inhibited the spreading of tau oligomers from the injection site to other brain regions and neurofibrillary tangle formation in the entorhinal cortex. Rifampin 0-10 microtubule associated protein tau Homo sapiens 48-51 35203506-10 2022 These results suggest that intranasal rifampicin could be a promising remedy that halts the progression of tauopathy by inhibiting tau oligomer propagation. Rifampin 38-48 microtubule associated protein tau Homo sapiens 131-134 35048417-6 2022 Analysis of cell culture medium of rifampicin treated HS explants revealed an anti-inflammatory effect of rifampicin that significantly inhibiting interleukin (IL)-1beta, IL-6, IL-8, IL-10, and tumour necrosis factor (TNF) -alpha production. Rifampin 35-45 interleukin 1 alpha Homo sapiens 147-169 35048417-6 2022 Analysis of cell culture medium of rifampicin treated HS explants revealed an anti-inflammatory effect of rifampicin that significantly inhibiting interleukin (IL)-1beta, IL-6, IL-8, IL-10, and tumour necrosis factor (TNF) -alpha production. Rifampin 35-45 interleukin 6 Homo sapiens 171-175 35048417-6 2022 Analysis of cell culture medium of rifampicin treated HS explants revealed an anti-inflammatory effect of rifampicin that significantly inhibiting interleukin (IL)-1beta, IL-6, IL-8, IL-10, and tumour necrosis factor (TNF) -alpha production. Rifampin 35-45 interleukin 10 Homo sapiens 183-188 35048417-6 2022 Analysis of cell culture medium of rifampicin treated HS explants revealed an anti-inflammatory effect of rifampicin that significantly inhibiting interleukin (IL)-1beta, IL-6, IL-8, IL-10, and tumour necrosis factor (TNF) -alpha production. Rifampin 106-116 interleukin 1 alpha Homo sapiens 147-169 35048417-6 2022 Analysis of cell culture medium of rifampicin treated HS explants revealed an anti-inflammatory effect of rifampicin that significantly inhibiting interleukin (IL)-1beta, IL-6, IL-8, IL-10, and tumour necrosis factor (TNF) -alpha production. Rifampin 106-116 interleukin 6 Homo sapiens 171-175 35048417-6 2022 Analysis of cell culture medium of rifampicin treated HS explants revealed an anti-inflammatory effect of rifampicin that significantly inhibiting interleukin (IL)-1beta, IL-6, IL-8, IL-10, and tumour necrosis factor (TNF) -alpha production. Rifampin 106-116 C-X-C motif chemokine ligand 8 Homo sapiens 177-181 35048417-6 2022 Analysis of cell culture medium of rifampicin treated HS explants revealed an anti-inflammatory effect of rifampicin that significantly inhibiting interleukin (IL)-1beta, IL-6, IL-8, IL-10, and tumour necrosis factor (TNF) -alpha production. Rifampin 106-116 interleukin 10 Homo sapiens 183-188 35048417-6 2022 Analysis of cell culture medium of rifampicin treated HS explants revealed an anti-inflammatory effect of rifampicin that significantly inhibiting interleukin (IL)-1beta, IL-6, IL-8, IL-10, and tumour necrosis factor (TNF) -alpha production. Rifampin 106-116 tumor necrosis factor Homo sapiens 194-229 35048417-7 2022 Immunohistochemistry of the rifampicin-treated explants suggested a tendency for it to reduce the expression of TLR2 while not affecting the number of immune cells. Rifampin 28-38 toll like receptor 2 Homo sapiens 112-116 2531681-2 1989 Seminal plasmin, which is known to be a stronger inhibitor than rifampicin binds at the same site as rifampicin to RNA polymerase. Rifampin 64-74 plasminogen Bos taurus 8-15 32795137-5 2022 In our study, we have investigated the role of these major tuberculosis drugs namely Rifampicin, Streptomycin, Isoniazid, Pyrazinamide, and Ethambutol on actin polymerization which are famously known to be a central player in the sarcomere region of the muscle in human body. Rifampin 85-95 actin Saccharomyces cerevisiae S288C 154-159 2531681-2 1989 Seminal plasmin, which is known to be a stronger inhibitor than rifampicin binds at the same site as rifampicin to RNA polymerase. Rifampin 101-111 plasminogen Bos taurus 8-15 2677927-5 1989 Against enterococci, all combinations showed a synergistic or additive effect, the most pronounced effect being demonstrated with rifampicin (FIC Index value around 0.3). Rifampin 130-140 Fic Escherichia coli 142-145 2590599-6 1989 Cortisol 6 beta-hydroxylase activity increased from 15 +/- 6 pmol min-1 mg-1 in organ donors (considered as "control subjects") to 87 +/- 31 pmol min-1 mg-1 in rifampicin treated patients. Rifampin 160-170 CD59 molecule (CD59 blood group) Homo sapiens 146-156 2777787-3 1989 In this report, we demonstrate that in immature rabbits, rifampicin activates the transcription of the Cyp3A6 gene which encodes P450IIIA6 (cytochrome P-450 3c). Rifampin 57-67 cytochrome P450 3A6 Oryctolagus cuniculus 103-109 2777787-3 1989 In this report, we demonstrate that in immature rabbits, rifampicin activates the transcription of the Cyp3A6 gene which encodes P450IIIA6 (cytochrome P-450 3c). Rifampin 57-67 cytochrome P450 3A6 Oryctolagus cuniculus 140-159 2806536-1 1989 In experiments of rats it was established that administration of isoniazid and rifampicin in equimolecular doses (50 and 250 mg/kg) for 14 days increased the activities of alanine aminotransferase, aspartate aminotransferase and alkaline phosphatase in the serum, enhanced lipid peroxidation in hepatocytic membranes, caused significant disturbances of bile excretion. Rifampin 79-89 glutamic-oxaloacetic transaminase 2 Rattus norvegicus 198-224 2811375-0 1989 Plasma levels of estradiol, estrone, estrone sulfate and sex hormone binding globulin in patients receiving rifampicin. Rifampin 108-118 sex hormone binding globulin Homo sapiens 57-85 2811375-5 1989 In eight patients, from whom all tuberculostatic treatment except rifampicin had been withdrawn, plasma sex hormone binding globulin was found to be increased by 75% by rifampicin treatment. Rifampin 66-76 sex hormone binding globulin Homo sapiens 104-132 2811375-5 1989 In eight patients, from whom all tuberculostatic treatment except rifampicin had been withdrawn, plasma sex hormone binding globulin was found to be increased by 75% by rifampicin treatment. Rifampin 169-179 sex hormone binding globulin Homo sapiens 104-132 2682943-3 1989 Rifampin and perhaps nafcillin induce cytochrome P-450-dependent isoenzyme metabolism of cyclosporine. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 2565211-2 1989 Purification and identification of the rifampicin-inducible human liver cytochrome P-450 (cyclosporin A oxidase) as a product of P450IIIA gene subfamily. Rifampin 39-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 16347898-5 1989 Rifampin-resistant strain D10 and 2-bromobutyrate mutant R were found to be deficient in only CoA-transferase, while several other mutants exhibited reduced butyraldehyde dehydrogenase and butanol dehydrogenase activities as well. Rifampin 0-8 CA_C1423 Clostridium acetobutylicum ATCC 824 171-184 16347898-5 1989 Rifampin-resistant strain D10 and 2-bromobutyrate mutant R were found to be deficient in only CoA-transferase, while several other mutants exhibited reduced butyraldehyde dehydrogenase and butanol dehydrogenase activities as well. Rifampin 0-8 CA_C1423 Clostridium acetobutylicum ATCC 824 197-210 2744145-2 1989 The use of GABA under induction of cytochrome P-450 of the liver by phenobarbital or rifampicin was not followed by immunodepression. Rifampin 85-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 2565211-10 1989 Moreover, these monoxygenase activities and the hepatic level of P-450(CsA) were simultaneously increased in the liver of patients treated for 4 days with 600 mg of rifampicin per day. Rifampin 165-175 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 65-75 2565211-12 1989 We conclude that P-450(CsA) is responsible for most (80%) of CsA oxidase activity in human liver, is encoded by gene P450IIIA3, as is NF oxidase, or a very closely related gene, and is strongly inducible by rifampicin pretreatment. Rifampin 207-217 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 17-27 2792166-6 1989 In these 6 volunteers, intake of diltiazem (240 mg daily), concurrently with rifampicin for a week, significantly elevated theophylline half-life to 6.2 h as well as reduced its clearance to 1.03 ml.min-1.kg-1. Rifampin 77-87 CD59 molecule (CD59 blood group) Homo sapiens 199-204 3175736-3 1988 After cessation of rifampin and isoniazid, prothrombin time was maintained within the same range by a 50% reduction of warfarin doses, despite a twofold rise in the plasma warfarin concentration. Rifampin 19-27 coagulation factor II, thrombin Homo sapiens 43-54 2559042-3 1989 Nondirected and FMLP-directed migration was significantly (P less than 0.01) inhibited by rifamycin SV and rifampin at assay concentrations above 0.31 and 5.0 micrograms/ml respectively thus confirming the low dose rifamycin effect observed by others using human neutrophils. Rifampin 107-115 formyl peptide receptor 1 Homo sapiens 16-20 3182866-5 1988 The antibiotic rifampicin induces cytochrome P-450 3c in rabbit liver microsomes, and the benzo(a)pyrene hydroxylase, estradiol 2-hydroxylase, and progesterone 6 beta-hydroxylase activities of these microsomes are stimulated by alpha-naphthoflavone. Rifampin 15-25 cytochrome P450 3A6 Oryctolagus cuniculus 34-53 3412203-2 1988 Further evidence is given of by time rifampicin induction of beta-glucuronidase and beta-N acetylglucosaminidase and its possible relation to hepatitis and pancreatitis. Rifampin 37-47 glucuronidase beta Homo sapiens 61-79 3147199-2 1988 In fact, the antibacterial activity of SPA-S-565 against numerous Gram-positive cocci belonging to Staphylococcus and Streptococcus species as well as against 20 strains of Mycobacterium tuberculosis, was similar to that of rifampicin. Rifampin 224-234 surfactant associated protein A1 Mus musculus 39-42 3412203-2 1988 Further evidence is given of by time rifampicin induction of beta-glucuronidase and beta-N acetylglucosaminidase and its possible relation to hepatitis and pancreatitis. Rifampin 37-47 O-GlcNAcase Homo sapiens 84-112 3367901-1 1988 Rifampicin induces cytochrome P-450 3c, progesterone 16 alpha- and 6 beta-hydroxylation, 17 beta-estradiol 2-hydroxylation, benzo[a] pyrene hydroxylation, and erythromycin N-demethylation in rabbit liver microsomes. Rifampin 0-10 cytochrome P450 3A6 Oryctolagus cuniculus 19-38 3259880-2 1988 This study was undertaken into liver microsomal fractions prepared from untreated rabbits or animals treated with drugs known to specifically induce various cytochrome P-450 isozymes such as form LM2 by phenobarbital, LM4 and LM6 by 3-methylcholanthrene and beta-naphthoflavone, LM3a by ethyl alcohol and acetone, and LM3c by macrolide antibiotics (rifampicin, erythromycin and triacetyloleandomycin). Rifampin 349-359 cytochrome P-450 Oryctolagus cuniculus 157-173 3665337-6 1987 Based on formation clearance values estimated for 6-, 7-, and 8-hydroxywarfarin, rifampin appears to increase the clearance of the parent drug by induction of the cytochrome P-450 isozyme(s) responsible for aromatic hydroxylation. Rifampin 81-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 3335807-5 1988 However, an increase in adherence of 65% was observed for one strain, RP14 (ATCC 35981), with rifampin treatment. Rifampin 94-102 TUB like protein 1 Homo sapiens 70-74 3573148-2 1987 Results of in vivo studies showed that the transcription of the Xp10 genome in Xp10-infected cells shifted from rifampin sensitivity to rifampin resistance. Rifampin 112-120 xenotropic-MCF leukemia virus 51 Mus musculus 64-68 3608348-5 1987 Therefore rifampin preferentially affects norantipyrine or desmethyl- and 3-hydroxydiazepam metabolic formation, suggesting induction of different (iso)zymes of cytochrome P-450. Rifampin 10-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-177 3573148-2 1987 Results of in vivo studies showed that the transcription of the Xp10 genome in Xp10-infected cells shifted from rifampin sensitivity to rifampin resistance. Rifampin 112-120 xenotropic-MCF leukemia virus 51 Mus musculus 79-83 3573148-2 1987 Results of in vivo studies showed that the transcription of the Xp10 genome in Xp10-infected cells shifted from rifampin sensitivity to rifampin resistance. Rifampin 136-144 xenotropic-MCF leukemia virus 51 Mus musculus 64-68 3573148-2 1987 Results of in vivo studies showed that the transcription of the Xp10 genome in Xp10-infected cells shifted from rifampin sensitivity to rifampin resistance. Rifampin 136-144 xenotropic-MCF leukemia virus 51 Mus musculus 79-83 3091058-0 1986 Recombinant interferon-gamma and chemotherapy with isoniazid and rifampicin in experimental murine tuberculosis. Rifampin 65-75 interferon gamma Mus musculus 12-28 3919717-0 1985 Isolation and partial characterization of a rifampicin induced rabbit liver microsomal cytochrome P-450. Rifampin 44-54 cytochrome P-450 Oryctolagus cuniculus 87-103 3707122-11 1986 Reversal of the delayed-nodulation phenotype of HS111 through lectin pretreatment was prevented by chloramphenicol or rifampin. Rifampin 118-126 LOW QUALITY PROTEIN: lectin Glycine max 62-68 4074601-4 1985 Clearance of phenytoin in patients receiving only rifampicin increased from 46.7 ml min-1 +/- 20.6 ml min-1 to 97.8 ml min-1 +/- 33.4 ml min-1 (P less than 0.01), while clearance in patients on three drugs increased from 47.1 +/- 23.4 ml min-1 to 81.3 ml min-1 +/- 41.6 ml min-1 (P less than 0.01). Rifampin 50-60 CD59 molecule (CD59 blood group) Homo sapiens 84-89 4074601-4 1985 Clearance of phenytoin in patients receiving only rifampicin increased from 46.7 ml min-1 +/- 20.6 ml min-1 to 97.8 ml min-1 +/- 33.4 ml min-1 (P less than 0.01), while clearance in patients on three drugs increased from 47.1 +/- 23.4 ml min-1 to 81.3 ml min-1 +/- 41.6 ml min-1 (P less than 0.01). Rifampin 50-60 CD59 molecule (CD59 blood group) Homo sapiens 102-115 4074601-4 1985 Clearance of phenytoin in patients receiving only rifampicin increased from 46.7 ml min-1 +/- 20.6 ml min-1 to 97.8 ml min-1 +/- 33.4 ml min-1 (P less than 0.01), while clearance in patients on three drugs increased from 47.1 +/- 23.4 ml min-1 to 81.3 ml min-1 +/- 41.6 ml min-1 (P less than 0.01). Rifampin 50-60 CD59 molecule (CD59 blood group) Homo sapiens 102-107 4074601-4 1985 Clearance of phenytoin in patients receiving only rifampicin increased from 46.7 ml min-1 +/- 20.6 ml min-1 to 97.8 ml min-1 +/- 33.4 ml min-1 (P less than 0.01), while clearance in patients on three drugs increased from 47.1 +/- 23.4 ml min-1 to 81.3 ml min-1 +/- 41.6 ml min-1 (P less than 0.01). Rifampin 50-60 CD59 molecule (CD59 blood group) Homo sapiens 102-107 4074601-4 1985 Clearance of phenytoin in patients receiving only rifampicin increased from 46.7 ml min-1 +/- 20.6 ml min-1 to 97.8 ml min-1 +/- 33.4 ml min-1 (P less than 0.01), while clearance in patients on three drugs increased from 47.1 +/- 23.4 ml min-1 to 81.3 ml min-1 +/- 41.6 ml min-1 (P less than 0.01). Rifampin 50-60 CD59 molecule (CD59 blood group) Homo sapiens 102-107 4074601-4 1985 Clearance of phenytoin in patients receiving only rifampicin increased from 46.7 ml min-1 +/- 20.6 ml min-1 to 97.8 ml min-1 +/- 33.4 ml min-1 (P less than 0.01), while clearance in patients on three drugs increased from 47.1 +/- 23.4 ml min-1 to 81.3 ml min-1 +/- 41.6 ml min-1 (P less than 0.01). Rifampin 50-60 CD59 molecule (CD59 blood group) Homo sapiens 102-107 4074601-4 1985 Clearance of phenytoin in patients receiving only rifampicin increased from 46.7 ml min-1 +/- 20.6 ml min-1 to 97.8 ml min-1 +/- 33.4 ml min-1 (P less than 0.01), while clearance in patients on three drugs increased from 47.1 +/- 23.4 ml min-1 to 81.3 ml min-1 +/- 41.6 ml min-1 (P less than 0.01). Rifampin 50-60 CD59 molecule (CD59 blood group) Homo sapiens 102-107 3720370-2 1986 Short-course chemotherapy (SCC) for pulmonary tuberculosis for nine months with isoniazid (INH) and rifampin (RIF) is well established. Rifampin 100-108 ras homolog family member F, filopodia associated Homo sapiens 110-113 2581616-2 1985 We have reported that a protein newly synthesized in the presence of rifampin might be a product of the srnB gene required for stable RNA degradation (Ito, R. and Ohnishi, Y. Rifampin 69-77 modulator of post-segregation killing protein SrnB Escherichia coli 104-108 3161772-0 1985 Induction of rat liver bilirubin-conjugating enzymes and glutathione S-transferase by rifampicin. Rifampin 86-96 hematopoietic prostaglandin D synthase Rattus norvegicus 57-82 3161772-2 1985 Rifampicin induced bilirubin UDP-glucuronyltransferase, bilirubin UDP-glucosyltransferase, bilirubin UDP-xylosyltransferase and glutathione S-transferase activities, but did not induce mixed function oxidase activities. Rifampin 0-10 hematopoietic prostaglandin D synthase Rattus norvegicus 128-153 3161772-6 1985 Induction of bilirubin-conjugating enzymes and glutathione S-transferase by rifampicin in rats was different from that in humans, in which selective induction of mixed function oxidase is reported to occur. Rifampin 76-86 hematopoietic prostaglandin D synthase Rattus norvegicus 47-72 3882459-0 1985 Interaction of bovine seminalplasmin with Escherichia coli RNA polymerase in the presence of rifampicin. Rifampin 93-103 caltrin Bos taurus 22-36 3882459-3 1985 Rifampicin quenced the intrinsic fluorescence of RNA polymerase and seminalplasmin when excited at 280 nm. Rifampin 0-10 caltrin Bos taurus 68-82 3882459-4 1985 However, excess of seminalplasmin reversed the quenching of RNA polymerase fluorescence by rifampicin. Rifampin 91-101 caltrin Bos taurus 19-33 3882459-5 1985 Upon addition of rifampicin to the seminalplasmin-RNA polymerase complex, no change in fluorescence spectrum was observed. Rifampin 17-27 caltrin Bos taurus 35-49 3882459-6 1985 It appeared that although rifampicin could form complexes with RNA polymerase and seminalplasmin alone, no binding domain was available for rifampicin in the RNA polymerase-seminalplasmin complex. Rifampin 26-36 caltrin Bos taurus 82-96 3919717-1 1985 Rifampicin administration to New Zealand male rabbits increased the concentration of an LM3 form of cytochrome P-450 to up to 30% of the microsomal P-450 concentration. Rifampin 0-10 cytochrome P-450 Oryctolagus cuniculus 100-116 6318656-2 1983 First, rifampin inhibited chemotaxis induced with FMLP but reversed the immobilization of PMLs that occurred at high FMLP concentrations. Rifampin 7-15 formyl peptide receptor 1 Homo sapiens 50-54 6508979-5 1984 Rifampicin seems to induce preferentially the cytochrome P-450 (iso-) enzyme(s) involved in the demethylation of antipyrine to norantipyrine. Rifampin 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 6466384-0 1984 Inductive and repressive effects of rifampicin on rabbit liver microsomal cytochrome P-450. Rifampin 36-46 cytochrome P-450 Oryctolagus cuniculus 74-90 6466384-6 1984 The concomitant inductive/repressive effect of rifampicine on two cytochrome P-450 isoenzymes makes this drug a very atypical inducer, at least in the rabbit. Rifampin 47-58 cytochrome P-450 Oryctolagus cuniculus 66-82 6318656-1 1983 Three independent experimental approaches support the hypothesis that rifampin competes for receptors on polymorphonuclear leukocytes (PMLs) with small peptide chemoattractants, e.g., N-formylmethionylleucylphenylalanine (FMLP), but not with serum-derived chemoattractants (C5a). Rifampin 70-78 formyl peptide receptor 1 Homo sapiens 222-226 6318656-1 1983 Three independent experimental approaches support the hypothesis that rifampin competes for receptors on polymorphonuclear leukocytes (PMLs) with small peptide chemoattractants, e.g., N-formylmethionylleucylphenylalanine (FMLP), but not with serum-derived chemoattractants (C5a). Rifampin 70-78 complement C5a receptor 1 Homo sapiens 274-277 6494651-5 1984 Not only the microsomal monooxygenase but also amidase was strongly induced by pretreatment with phenobarbital, 3-methylcholanthrene and rifampicin, respectively. Rifampin 137-147 cytochrome P450, family 1, subfamily a, polypeptide 1 Rattus norvegicus 13-37 6639842-5 1983 Rifampicin (600 mg/day) caused a large increase in propranolol"s oral clearance (35.7 +/- 16.3 vs 96.1 +/- 26.9 ml min-1 kg-1, mean +/- s.d. Rifampin 0-10 CD59 molecule (CD59 blood group) Homo sapiens 115-125 6318656-2 1983 First, rifampin inhibited chemotaxis induced with FMLP but reversed the immobilization of PMLs that occurred at high FMLP concentrations. Rifampin 7-15 formyl peptide receptor 1 Homo sapiens 117-121 6318656-3 1983 Second, rifampin competed with radiolabeled FMLP for binding sites on PMLs and displaced already-bound radiolabeled FMLP. Rifampin 8-16 formyl peptide receptor 1 Homo sapiens 116-120 6318656-4 1983 Third, rifampin blocked and reversed the bipolar shape changes induced in PMLs by FMLP. Rifampin 7-15 formyl peptide receptor 1 Homo sapiens 82-86 6318656-7 1983 The evidence suggests, therefore, that rifampin is a ligand for FMLP-type receptors on PMLs. Rifampin 39-47 formyl peptide receptor 1 Homo sapiens 64-68 6854335-0 1983 Non-competitive inhibition of hepatic and intestinal aryl hydrocarbon hydroxylase activities from rats by rifampin. Rifampin 106-114 cytochrome P450, family 1, subfamily a, polypeptide 1 Rattus norvegicus 53-81 6357197-4 1983 Since the resistance to nuclease was not observed in the presence of rifampicin or by preincubation without UTP, both RNA chain initiation and elongation were considered to be essential for the manifestation of resistance. Rifampin 69-79 nuclease Escherichia coli 24-32 6222344-1 1983 In a 48-years old woman, intermittent rifampicin treatment induced an immunoallergic reaction with digestive disorders, haemolysis, acute renal failure and prolonged prothrombin time. Rifampin 38-48 coagulation factor II, thrombin Homo sapiens 166-177 6196945-3 1983 The effect of different inhibitors like actinomycin D, chloramphenicol, tetracycline, nitrofurantoin and rifampicin on histidase induction was also studied. Rifampin 105-115 histidine ammonia-lyase Homo sapiens 119-128 6854335-1 1983 Rifampin is a semisynthetic antibiotic which is known to alter hepatic cytochrome P-450 mediated drug metabolizing enzymes. Rifampin 0-8 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 71-87 6854335-2 1983 Using benzo(a)pyrene as the substrate we have shown that rifampin acts as a non-competitive inhibitor of hepatic microsomal aryl hydrocarbon hydroxylase in vitro at or below 0.10 mM and that it is also a non-competitive inhibitor of intestinal aryl hydrocarbon hydroxylase at or below 0.075 mM. Rifampin 57-65 cytochrome P450, family 1, subfamily a, polypeptide 1 Rattus norvegicus 124-152 6854335-2 1983 Using benzo(a)pyrene as the substrate we have shown that rifampin acts as a non-competitive inhibitor of hepatic microsomal aryl hydrocarbon hydroxylase in vitro at or below 0.10 mM and that it is also a non-competitive inhibitor of intestinal aryl hydrocarbon hydroxylase at or below 0.075 mM. Rifampin 57-65 cytochrome P450, family 1, subfamily a, polypeptide 1 Rattus norvegicus 244-272 7039435-1 1982 Early phase hyperglycemia, associated with increased rates of insulin and C-peptide secretion after oral administration of 100 g glucose, was observed among patients with pulmonary tuberculosis who were taking rifampicin. Rifampin 210-220 insulin Homo sapiens 62-69 7154665-6 1982 Rifampicin treatment gives a variable enzyme induction of antipyrine metabolism, cytochrome P450 and aniline hydroxylase activity; aminopyrine N-demethylase activity is significantly inhibited. Rifampin 0-10 cytochrome P-450 Oryctolagus cuniculus 81-156 6189376-1 1982 Alkaline phosphate, catalase and beta-galactosidase activities of Vibrio et tor were decreased after acquisition of resistance towards rifampicin. Rifampin 135-145 catalase Homo sapiens 20-28 6189376-1 1982 Alkaline phosphate, catalase and beta-galactosidase activities of Vibrio et tor were decreased after acquisition of resistance towards rifampicin. Rifampin 135-145 galactosidase beta 1 Homo sapiens 33-51 6845400-2 1983 The maximal plasma concentrations (Cmax) and the elimination half-lives (t 1/2 beta) of RMP are 8.83 +/- 1.72 mg L-1 and 4.09 +/- 2.59 h, respectively. Rifampin 88-91 interleukin 1 receptor like 1 Homo sapiens 73-83 7039435-1 1982 Early phase hyperglycemia, associated with increased rates of insulin and C-peptide secretion after oral administration of 100 g glucose, was observed among patients with pulmonary tuberculosis who were taking rifampicin. Rifampin 210-220 insulin Homo sapiens 74-83 7042603-7 1981 This trial revealed the satisfactory efficacy, good tolerability, and practicability of a supervised once-monthly 1200 mg single oral dose rifampin schedule as a component of combination regimes for the initial treatment of patients with lepromatous (LLs and LLp) leprosy. Rifampin 139-147 LLP homolog, long-term synaptic facilitation factor Homo sapiens 259-262 6277242-5 1982 It should be noted that the inhibitory effect of rifampicin on the activity of SDH, TP and AP was less pronounced than that of lincomycin. Rifampin 49-59 aminoadipate-semialdehyde synthase Mus musculus 79-82 7278139-1 1981 Lepromatous patients of "Introductory Rifampicin therapy" (Lepr. Rifampin 38-48 leptin receptor Homo sapiens 0-4 7416733-1 1980 The effect of semisynthetic antibiotics, such as ampicillin and rifampicin on activity of succinate dehydrogenase (SDH) in the liver and spleen of mice immunized with heated typhoid vaccine was studied. Rifampin 64-74 aminoadipate-semialdehyde synthase Mus musculus 90-113 7416733-1 1980 The effect of semisynthetic antibiotics, such as ampicillin and rifampicin on activity of succinate dehydrogenase (SDH) in the liver and spleen of mice immunized with heated typhoid vaccine was studied. Rifampin 64-74 aminoadipate-semialdehyde synthase Mus musculus 115-118 6110213-0 1980 [The activity of transaminases (GOT and GPT), alkaline phosphatase and gamma-glutamyl transferases in the serum of patients treated with Rifampicin (author"s transl)]. Rifampin 137-147 glutamic--pyruvic transaminase Homo sapiens 40-43 7389010-3 1980 PB, Rifa and PCN induced the aryl hydrocarbon hydroxylase (AHH) in the skin though to a lesser extent than MC. Rifampin 4-8 cytochrome P450, family 1, subfamily a, polypeptide 1 Rattus norvegicus 29-57 7389010-3 1980 PB, Rifa and PCN induced the aryl hydrocarbon hydroxylase (AHH) in the skin though to a lesser extent than MC. Rifampin 4-8 cytochrome P450, family 1, subfamily a, polypeptide 1 Rattus norvegicus 59-62 7359301-0 1980 Rifampin for CSF shunt infection. Rifampin 0-8 colony stimulating factor 2 Homo sapiens 13-16 702642-6 1978 Results obtained from an addition of rifampin at the prereplicative cycle after infection indicated that mRNA from genes 43, rIIA, 46, 39, 52, and 63 are more stable in T4amC5 (gene 59) than in wild-type-infected cells. Rifampin 37-45 protein kinase cAMP-dependent type I regulatory subunit alpha Rattus norvegicus 125-129 6449961-1 1980 In mitomycin C-treated lambda lysogens, even though the rate of synthesis of RecA protein was greatly reduced by a low concentration of rifampicin (4 microgram/ml), induction of prophage lambda occurred readily as assessed by (i) cell lysis of the lysogens, (ii) production of progeny phage, and (iii) extensive cleavage of lambda repressor. Rifampin 136-146 RAD51 recombinase Homo sapiens 77-81 477551-3 1979 During and after treatment with rifampicin optimal levels of prothrombin time were difficult or even impossible to achieve in two of the three patients. Rifampin 32-42 coagulation factor II, thrombin Homo sapiens 61-72 6244557-4 1980 EGF also induced a perturbation of chromatin structure in GH3 cell nuclei that was detected by an increase (40%) in the number of rifampicin-resistant initiation sites for bacterial RNA polymerase. Rifampin 130-140 epidermal growth factor Rattus norvegicus 0-3 924280-0 1977 Effect of rifampicin on tryptophanase induction in normal and rifampicin-resistant Vibrio el tor. Rifampin 10-20 tryptophan 2,3-dioxygenase Homo sapiens 24-37 657879-4 1978 From the beginning of combination therapy with synergistic-acting substances (rifampicin + isoprodian (INH + PTH + DDS) the logarithms of the number of bacteria in the homogenates decreased, both during treatment period and during treatment-free observation period (Figs. Rifampin 78-88 parathyroid hormone Homo sapiens 109-112 856862-0 1977 Protein binding of rifampicin to bovine serum albumin as measured by gel filtration. Rifampin 19-29 albumin Homo sapiens 40-53 322700-0 1977 Treatment of TRIC infection of the eye with rifampicin or chloramphenicol. Rifampin 44-54 MARVEL domain containing 2 Homo sapiens 13-17 930514-3 1977 Rifampicin induced solely lun mutation. Rifampin 0-10 TOP1 binding arginine/serine rich protein, E3 ubiquitin ligase Homo sapiens 26-29 140033-4 1977 Rifampicin seems to be a potent inducer of drug metabolism in humans and it causes a proliferation of the smooth endoplasmatic reticulum and an increase of cytochrome P450 content in the liver. Rifampin 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-171 33307820-0 2021 Tanshinone IIA may alleviate rifampin-induced cholestasis by regulating the expression and function of NTCP. Rifampin 29-37 solute carrier family 10 member 1 Rattus norvegicus 103-107 1100115-1 1975 The influence of temperature and KCl concentration on the formation of rifampicin-resistant preinitiation complexes by holo RNA polymerase has been compared for T4 DNA and Azotobacter phage A21 DNA. Rifampin 71-81 immunoglobulin kappa variable 2-26 (pseudogene) Homo sapiens 190-193 1100115-7 1975 Both holo and core RNA polymerases are able to form complexes with A21 DNA that are resistant to attack by rifampicin. Rifampin 107-117 immunoglobulin kappa variable 2-26 (pseudogene) Homo sapiens 67-70 775309-0 1976 A bacterial RNA polymerase mutant that renders lambda growth independent of the N and cro functions at 42 degrees C. We describe a bacterial RNA polymerase mutation, rif 501, which confers rifampicin resistance and thermosensitivity to E. coli K 12. Rifampin 189-199 cro Escherichia virus Lambda 86-89 1233229-5 1975 The acceleration of oestrogen hydroxylation by rifampicin was paralleled by an increase in microsomal cytochrome P-450, and also by microsomal reduction of rifampicin-quinone, a reactive metabolite of rifampicin. Rifampin 47-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-118 4584805-0 1973 Conjugal deoxyribonucleic acid replication by Excherichia coli K-12: effect of chloramphenicol and rifampin. Rifampin 99-107 keratin 12 Homo sapiens 63-67 33405191-3 2021 METHOD: P-gp modulation in rats was performed by using P-gp inducer (150 mg/kg rifampicin) and P-gp inhibitor (10 mg/kg cyclosporine A) for 14 days prior to be infected with Helicobacter pylori (H. pylori). Rifampin 79-89 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 8-12 33405191-10 2021 The use of rifampicin to induce P-gp level was also shown to be effective in eradicating the H. pylori infection. Rifampin 11-21 ATP binding cassette subfamily B member 1 Homo sapiens 32-36 33629115-5 2021 Rifampicin (RIF), a first line anti-TB drug, is a human specific activator of pregnane X receptor (PXR). Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 78-97 33674270-2 2021 In the current study, the hepatocyte nuclear factor 4 alpha (HNF4A) antisense RNA 1 (HNF4A-AS1), an antisense lncRNA of HNF4A, was found to be a negative regulator of the basal and rifampicin (RIF)-induced expression of nuclear receptors and downstream CYPs. Rifampin 181-191 hepatocyte nuclear factor 4 alpha Homo sapiens 26-59 33674270-2 2021 In the current study, the hepatocyte nuclear factor 4 alpha (HNF4A) antisense RNA 1 (HNF4A-AS1), an antisense lncRNA of HNF4A, was found to be a negative regulator of the basal and rifampicin (RIF)-induced expression of nuclear receptors and downstream CYPs. Rifampin 181-191 hepatocyte nuclear factor 4 alpha Homo sapiens 61-66 33674270-2 2021 In the current study, the hepatocyte nuclear factor 4 alpha (HNF4A) antisense RNA 1 (HNF4A-AS1), an antisense lncRNA of HNF4A, was found to be a negative regulator of the basal and rifampicin (RIF)-induced expression of nuclear receptors and downstream CYPs. Rifampin 181-191 HNF4A antisense RNA 1 Homo sapiens 85-94 33674270-2 2021 In the current study, the hepatocyte nuclear factor 4 alpha (HNF4A) antisense RNA 1 (HNF4A-AS1), an antisense lncRNA of HNF4A, was found to be a negative regulator of the basal and rifampicin (RIF)-induced expression of nuclear receptors and downstream CYPs. Rifampin 181-191 hepatocyte nuclear factor 4 alpha Homo sapiens 85-90 33749522-0 2021 Rifampicin ameliorates lipopolysaccharide-induced cognitive and motor impairments via inhibition of the TLR4/MyD88/NF-kappaB signaling pathway in mice. Rifampin 0-10 toll-like receptor 4 Mus musculus 104-108 33749522-0 2021 Rifampicin ameliorates lipopolysaccharide-induced cognitive and motor impairments via inhibition of the TLR4/MyD88/NF-kappaB signaling pathway in mice. Rifampin 0-10 myeloid differentiation primary response gene 88 Mus musculus 109-114 33749522-0 2021 Rifampicin ameliorates lipopolysaccharide-induced cognitive and motor impairments via inhibition of the TLR4/MyD88/NF-kappaB signaling pathway in mice. Rifampin 0-10 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 115-124 33749522-9 2021 Rifampicin suppressed the release of pro-inflammatory mediators, including tumor necrosis factor-alpha, interleukin-1beta, and prostaglandin E2 in the serum and nitric oxide (NO) in brain tissue, and cyclooxygenase-2 and inducible nitric oxide synthase levels. Rifampin 0-10 tumor necrosis factor Mus musculus 75-102 33749522-9 2021 Rifampicin suppressed the release of pro-inflammatory mediators, including tumor necrosis factor-alpha, interleukin-1beta, and prostaglandin E2 in the serum and nitric oxide (NO) in brain tissue, and cyclooxygenase-2 and inducible nitric oxide synthase levels. Rifampin 0-10 interleukin 1 beta Mus musculus 104-121 33749522-9 2021 Rifampicin suppressed the release of pro-inflammatory mediators, including tumor necrosis factor-alpha, interleukin-1beta, and prostaglandin E2 in the serum and nitric oxide (NO) in brain tissue, and cyclooxygenase-2 and inducible nitric oxide synthase levels. Rifampin 0-10 prostaglandin-endoperoxide synthase 2 Mus musculus 200-216 33749522-9 2021 Rifampicin suppressed the release of pro-inflammatory mediators, including tumor necrosis factor-alpha, interleukin-1beta, and prostaglandin E2 in the serum and nitric oxide (NO) in brain tissue, and cyclooxygenase-2 and inducible nitric oxide synthase levels. Rifampin 0-10 nitric oxide synthase 2, inducible Mus musculus 221-252 34035488-6 2022 In addition, inconsistencies were observed between the protein expression and enzyme activities of CYP3A4 in cells induced by rifampin but not in groups treated with furmonertinib. Rifampin 126-134 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-105 34035488-9 2022 The fold change in the CYP3A4 enzyme activity in the cells treated with rifampin was 5.22 +- 1.13, which was similar to that of 0.5 microM furmonertinib (3.79 +- 0.52). Rifampin 72-80 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 23-29 33970450-7 2021 RESULTS: Based on the findings, Rifampicin and Letermovir appeared as the most promising drug showing a very good binding affinity with the main protease of SARS-CoV-2 and TNF-alpha, IL-6, and IL-1beta. Rifampin 32-42 tumor necrosis factor Homo sapiens 172-181 33970450-7 2021 RESULTS: Based on the findings, Rifampicin and Letermovir appeared as the most promising drug showing a very good binding affinity with the main protease of SARS-CoV-2 and TNF-alpha, IL-6, and IL-1beta. Rifampin 32-42 interleukin 6 Homo sapiens 183-187 33970450-7 2021 RESULTS: Based on the findings, Rifampicin and Letermovir appeared as the most promising drug showing a very good binding affinity with the main protease of SARS-CoV-2 and TNF-alpha, IL-6, and IL-1beta. Rifampin 32-42 interleukin 1 alpha Homo sapiens 193-201 33963620-2 2021 Isavuconazole is metabolized by CYP3A4 and CYP3A5 and it has been shown that the CYP3A inducer rifampin reduces isavuconazole exposure. Rifampin 95-103 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-38 33963620-2 2021 Isavuconazole is metabolized by CYP3A4 and CYP3A5 and it has been shown that the CYP3A inducer rifampin reduces isavuconazole exposure. Rifampin 95-103 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 43-49 33963620-2 2021 Isavuconazole is metabolized by CYP3A4 and CYP3A5 and it has been shown that the CYP3A inducer rifampin reduces isavuconazole exposure. Rifampin 95-103 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-37 33749522-11 2021 Rifampicin inhibited the activation of the toll-like receptor 4 (TLR4)/myeloid differentiation primary response 88 (MyD88)/nuclear factor kappa B (NF-kappaB) signaling pathway. Rifampin 0-10 toll-like receptor 4 Mus musculus 43-63 33749522-11 2021 Rifampicin inhibited the activation of the toll-like receptor 4 (TLR4)/myeloid differentiation primary response 88 (MyD88)/nuclear factor kappa B (NF-kappaB) signaling pathway. Rifampin 0-10 toll-like receptor 4 Mus musculus 65-69 33749522-11 2021 Rifampicin inhibited the activation of the toll-like receptor 4 (TLR4)/myeloid differentiation primary response 88 (MyD88)/nuclear factor kappa B (NF-kappaB) signaling pathway. Rifampin 0-10 myeloid differentiation primary response gene 88 Mus musculus 116-121 33749522-11 2021 Rifampicin inhibited the activation of the toll-like receptor 4 (TLR4)/myeloid differentiation primary response 88 (MyD88)/nuclear factor kappa B (NF-kappaB) signaling pathway. Rifampin 0-10 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 147-156 33749522-12 2021 Rifampicin downregulated TLR4 and MyD88 protein levels and inhibited NF-kappaB inhibitor alpha and NF-kappaB inhibitor kinase beta phosphorylation, thus reducing p65 nuclear transfer by inhibiting NF-kappaB signaling activation in LPS-treated mice. Rifampin 0-10 toll-like receptor 4 Mus musculus 25-29 33749522-12 2021 Rifampicin downregulated TLR4 and MyD88 protein levels and inhibited NF-kappaB inhibitor alpha and NF-kappaB inhibitor kinase beta phosphorylation, thus reducing p65 nuclear transfer by inhibiting NF-kappaB signaling activation in LPS-treated mice. Rifampin 0-10 myeloid differentiation primary response gene 88 Mus musculus 34-39 33749522-12 2021 Rifampicin downregulated TLR4 and MyD88 protein levels and inhibited NF-kappaB inhibitor alpha and NF-kappaB inhibitor kinase beta phosphorylation, thus reducing p65 nuclear transfer by inhibiting NF-kappaB signaling activation in LPS-treated mice. Rifampin 0-10 nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha Mus musculus 69-94 33749522-12 2021 Rifampicin downregulated TLR4 and MyD88 protein levels and inhibited NF-kappaB inhibitor alpha and NF-kappaB inhibitor kinase beta phosphorylation, thus reducing p65 nuclear transfer by inhibiting NF-kappaB signaling activation in LPS-treated mice. Rifampin 0-10 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 69-78 33749522-12 2021 Rifampicin downregulated TLR4 and MyD88 protein levels and inhibited NF-kappaB inhibitor alpha and NF-kappaB inhibitor kinase beta phosphorylation, thus reducing p65 nuclear transfer by inhibiting NF-kappaB signaling activation in LPS-treated mice. Rifampin 0-10 v-rel reticuloendotheliosis viral oncogene homolog A (avian) Mus musculus 162-165 33749522-12 2021 Rifampicin downregulated TLR4 and MyD88 protein levels and inhibited NF-kappaB inhibitor alpha and NF-kappaB inhibitor kinase beta phosphorylation, thus reducing p65 nuclear transfer by inhibiting NF-kappaB signaling activation in LPS-treated mice. Rifampin 0-10 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 99-108 33749522-13 2021 CONCLUSION: Rifampicin protects against LPS-induced neuroinflammation and attenuates cognitive and motor impairments by inhibiting the TLR4/MyD88/NF-kappaB signaling pathway. Rifampin 12-22 toll-like receptor 4 Mus musculus 135-139 33749522-13 2021 CONCLUSION: Rifampicin protects against LPS-induced neuroinflammation and attenuates cognitive and motor impairments by inhibiting the TLR4/MyD88/NF-kappaB signaling pathway. Rifampin 12-22 myeloid differentiation primary response gene 88 Mus musculus 140-145 33749522-13 2021 CONCLUSION: Rifampicin protects against LPS-induced neuroinflammation and attenuates cognitive and motor impairments by inhibiting the TLR4/MyD88/NF-kappaB signaling pathway. Rifampin 12-22 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 146-155 33629115-5 2021 Rifampicin (RIF), a first line anti-TB drug, is a human specific activator of pregnane X receptor (PXR). Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 99-102 33629115-5 2021 Rifampicin (RIF), a first line anti-TB drug, is a human specific activator of pregnane X receptor (PXR). Rifampin 12-15 nuclear receptor subfamily 1 group I member 2 Homo sapiens 78-97 33629115-5 2021 Rifampicin (RIF), a first line anti-TB drug, is a human specific activator of pregnane X receptor (PXR). Rifampin 12-15 nuclear receptor subfamily 1 group I member 2 Homo sapiens 99-102 33880760-1 2021 The objective of this study was to determine the effects of the OATP inhibitor rifampin on pharmacokinetic of Biopharmaceutics Drug Disposition Classification System (BDDCS) Class 1 compound fluvastatin. Rifampin 79-87 solute carrier organic anion transporter family member 1A2 Homo sapiens 64-68 33907428-2 2021 We presented a case of a household contact of rifampicin-resistant TB revealing reactive IFN-gamma release assay with unsuspicious clinical and radiologic examinations. Rifampin 46-56 interferon gamma Homo sapiens 89-98 33759451-7 2021 Subsequently, the model was used to explore UGT1A1-related DDI risk with atazanavir and rifampicin along with the effect of enzyme genotype on raltegravir apparent clearance. Rifampin 88-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 33937284-13 2021 Cirrhotic patients displayed higher concentrations of MOR agonist Leu-enkephalin and KOR agonist dynorphin A. Endogenous opioid levels remained largely unchanged after successful treatment with the potent anti-pruritic drugs rifampicin and bezafibrate. Rifampin 225-235 opioid receptor mu 1 Homo sapiens 54-57 33851518-5 2022 Rifampicin treatment increased ALP activity and mRNA level of bone biomarker genes (ALP, MGP, OPN, and OPG). Rifampin 0-10 secreted phosphoprotein 1 Homo sapiens 94-97 33851518-5 2022 Rifampicin treatment increased ALP activity and mRNA level of bone biomarker genes (ALP, MGP, OPN, and OPG). Rifampin 0-10 basic transcription factor 3 pseudogene 11 Homo sapiens 103-106 33851518-9 2022 In conclusion, rifampicin treatment induces the bone form of ALP in the serum of healthy human volunteers. Rifampin 15-25 alkaline phosphatase, placental Homo sapiens 61-64 33863817-10 2021 Interestingly, with rifampin, in vivo CYP3A activity was approximately 4-fold higher than in vitro activity. Rifampin 20-28 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 38-43 33851518-0 2022 Rifampicin induces the bone form of alkaline phosphatase in humans. Rifampin 0-10 alkaline phosphatase, placental Homo sapiens 36-56 33851518-2 2022 In our clinical trials on healthy volunteers to discover novel metabolic functions of PXR activation, we observed that rifampicin, a well-established ligand for human PXR, 600 mg daily for a week, increased the plasma alkaline phosphatase (ALP) significantly compared with the placebo. Rifampin 119-129 nuclear receptor subfamily 1 group I member 2 Homo sapiens 86-89 33851518-2 2022 In our clinical trials on healthy volunteers to discover novel metabolic functions of PXR activation, we observed that rifampicin, a well-established ligand for human PXR, 600 mg daily for a week, increased the plasma alkaline phosphatase (ALP) significantly compared with the placebo. Rifampin 119-129 nuclear receptor subfamily 1 group I member 2 Homo sapiens 167-170 33851518-2 2022 In our clinical trials on healthy volunteers to discover novel metabolic functions of PXR activation, we observed that rifampicin, a well-established ligand for human PXR, 600 mg daily for a week, increased the plasma alkaline phosphatase (ALP) significantly compared with the placebo. Rifampin 119-129 alkaline phosphatase, placental Homo sapiens 218-238 33851518-2 2022 In our clinical trials on healthy volunteers to discover novel metabolic functions of PXR activation, we observed that rifampicin, a well-established ligand for human PXR, 600 mg daily for a week, increased the plasma alkaline phosphatase (ALP) significantly compared with the placebo. Rifampin 119-129 alkaline phosphatase, placental Homo sapiens 240-243 33851518-5 2022 Rifampicin treatment increased ALP activity and mRNA level of bone biomarker genes (ALP, MGP, OPN, and OPG). Rifampin 0-10 alkaline phosphatase, placental Homo sapiens 31-34 33851518-5 2022 Rifampicin treatment increased ALP activity and mRNA level of bone biomarker genes (ALP, MGP, OPN, and OPG). Rifampin 0-10 alkaline phosphatase, placental Homo sapiens 84-87 33851518-5 2022 Rifampicin treatment increased ALP activity and mRNA level of bone biomarker genes (ALP, MGP, OPN, and OPG). Rifampin 0-10 matrix Gla protein Homo sapiens 89-92 33759451-8 2021 Simulations of pregnancy-related induction of UGT1A1 either exacerbated UGT1A1 induction by rifampicin or negated atazanavir UGT1A1 inhibition. Rifampin 92-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 33759451-8 2021 Simulations of pregnancy-related induction of UGT1A1 either exacerbated UGT1A1 induction by rifampicin or negated atazanavir UGT1A1 inhibition. Rifampin 92-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 33759451-8 2021 Simulations of pregnancy-related induction of UGT1A1 either exacerbated UGT1A1 induction by rifampicin or negated atazanavir UGT1A1 inhibition. Rifampin 92-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 33790071-0 2021 Real-World Performance Assessment of Xpert MTB/RIF Assay for Detecting Pulmonary Tuberculosis and Rifampin Resistance in a Single Tertiary Care Hospital in Korea. Rifampin 98-106 ras homolog family member F, filopodia associated Homo sapiens 47-50 33446525-10 2021 Similar tendencies were observed in the other 4 substrates of AADAC; that is, p-nitrophenyl acetate, ketoconazole, phenacetin, and rifampicin. Rifampin 131-141 arylacetamide deacetylase Homo sapiens 62-67 33412187-0 2021 Rifampicin impairs adipogenesis by suppressing NRF2-ARE activity in mice fed a high-fat diet. Rifampin 0-10 nuclear factor, erythroid derived 2, like 2 Mus musculus 47-51 33263786-6 2021 Furthermore, we showed that CYP1A1 and CYP1B1 were induced by beta-naphtoflavone in all three models, whereas CYP3A4 was induced by phenobarbital and rifampicin in HIOs, in the IEC-based model (although not statistically significant), but not in Caco-2 cells. Rifampin 150-160 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 110-116 33355210-9 2021 Physiologically based pharmacokinetic (PBPK) models built upon two independent bottom-up approaches predicted elevation of E7766 plasma exposure when administered with Rifampicin, a clinical OATP inhibitor. Rifampin 168-178 solute carrier organic anion transporter family member 1A2 Homo sapiens 191-195 33355212-7 2021 Rifampin induced CYP3A4; estimated EC50 and Emax were 3.75 microM and 8.96-fold, respectively for ileal organoids, and 1.40 microM and 11.3-fold, respectively, for colon organoids. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 17-23 33355212-8 2021 Ileal, but not colon organoids exhibited nifedipine oxidase activity, which was induced by rifampin up to 14-fold. Rifampin 91-99 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-59 33483427-7 2021 Impaired co-activator recruitment by nelfinavir, as compared to the full agonist rifampin, proved to be the underlying mechanism of both effects on PXR. Rifampin 81-89 nuclear receptor subfamily 1 group I member 2 Homo sapiens 148-151 33483427-8 2021 Physiological relevance of nelfinavir-dependent modulation of PXR activity was investigated in respectively treated primary human hepatocytes, which showed differential induction of PXR target genes and antagonism of rifampin-induced ABCB1 and CYP3A4 gene expression. Rifampin 217-225 nuclear receptor subfamily 1 group I member 2 Homo sapiens 62-65 33483427-8 2021 Physiological relevance of nelfinavir-dependent modulation of PXR activity was investigated in respectively treated primary human hepatocytes, which showed differential induction of PXR target genes and antagonism of rifampin-induced ABCB1 and CYP3A4 gene expression. Rifampin 217-225 ATP binding cassette subfamily B member 1 Homo sapiens 234-239 33483427-8 2021 Physiological relevance of nelfinavir-dependent modulation of PXR activity was investigated in respectively treated primary human hepatocytes, which showed differential induction of PXR target genes and antagonism of rifampin-induced ABCB1 and CYP3A4 gene expression. Rifampin 217-225 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 244-250 33616229-1 2021 BACKGROUND: Xpert MTB/RIF and Xpert MTB/RIF Ultra (Xpert Ultra) are World Health Organization (WHO)-recommended rapid tests that simultaneously detect tuberculosis and rifampicin resistance in people with signs and symptoms of tuberculosis. Rifampin 168-178 ras homolog family member F, filopodia associated Homo sapiens 40-43 33616229-30 2021 Rifampicin resistance detection Pooled sensitivity and specificity were 94.9% (88.9 to 97.9) and 99.1% (97.7 to 99.8) (5 studies, 921 participants; high-certainty evidence) for Xpert Ultra versus 95.3% (90.0 to 98.1) and 98.8% (97.2 to 99.6) (5 studies, 930 participants; high-certainty evidence) for Xpert MTB/RIF. Rifampin 0-10 ras homolog family member F, filopodia associated Homo sapiens 311-314 33547469-4 2021 As absorption of temsavir is not altered with increased gastric pH, patients may take acid suppressive agents such as famotidine during fostemsavir therapy.Temsavir is primarily metabolized through hydrolysis but also via cytochrome P-450 (CYP) oxidation; therefore, coadministration of fostemsavir with strong CYP3A inducers such as rifampin, carbamazepine, phenytoin, mitotane, enzalutamide, or St John"s wort is contraindicated because it may result in significantly lower temsavir exposure, which can ultimately impair virologic response. Rifampin 334-342 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-238 33688304-0 2021 Antituberculosis Drugs (Rifampicin and Isoniazid) Induce Liver Injury by Regulating NLRP3 Inflammasomes. Rifampin 24-34 NLR family, pyrin domain containing 3 Rattus norvegicus 84-89 33616229-33 2021 The number of people wrongly diagnosed with rifampicin resistance would be 8 with Xpert Ultra and 11 with Xpert MTB/RIF. Rifampin 44-54 ras homolog family member F, filopodia associated Homo sapiens 116-119 33617360-0 2021 Quercetin Modulates Nrf2 and NF-kappaB/TLR-4 Pathways to Protect against Isoniazid and Rifampicin Induced Hepatotoxicity in vivo. Rifampin 87-97 toll-like receptor 4 Rattus norvegicus 39-44 33672438-5 2021 In addition, the modulation of CYP3A was reflected in the 1beta-OH-DCA/DCA UMR after the intake of rifampicin (induction ratio = 11.4, p < 0.01). Rifampin 99-109 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-36 32705692-2 2021 We assessed the impact of RI and single dose oral rifampin (RIF) on the pharmacokinetics of CYP3A, OATP1B, P-gp, and BCRP substrates using a microdose cocktail and OATP1B endogenous biomarkers. Rifampin 50-58 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 92-97 33583375-0 2021 Quality by Design Approach for Development and Optimization of Rifampicin Loaded Bovine Serum Albumin Nanoparticles and Characterization. Rifampin 63-73 albumin Mus musculus 88-101 33583375-1 2021 AIM: This study is focused on preparing rifampicin loaded bovine serum albumin nanoparticles (RIF BSA NPs) suitable for intravenous application using systematic quality by design (QbD) approach. Rifampin 40-50 albumin Mus musculus 65-78 33583375-5 2021 OBJECTIVES: The main objective of this study is optimizing particle size and entrapment efficiency of rifampicin loaded bovine serum albumin nanoparticles (RIF BSA NPs) and making it suitable for intravenous application using QbD approach. Rifampin 102-112 albumin Mus musculus 127-140 33538008-3 2021 We aimed to investigate relationship between genetic polymorphism of SLCO1B1 and rifampin exposure and its impact on clinical outcomes in Korean patients with active pulmonary TB. Rifampin 81-89 solute carrier organic anion transporter family member 1B1 Homo sapiens 69-76 32705692-2 2021 We assessed the impact of RI and single dose oral rifampin (RIF) on the pharmacokinetics of CYP3A, OATP1B, P-gp, and BCRP substrates using a microdose cocktail and OATP1B endogenous biomarkers. Rifampin 50-58 phosphoglycolate phosphatase Homo sapiens 107-111 32705692-2 2021 We assessed the impact of RI and single dose oral rifampin (RIF) on the pharmacokinetics of CYP3A, OATP1B, P-gp, and BCRP substrates using a microdose cocktail and OATP1B endogenous biomarkers. Rifampin 50-58 BCR pseudogene 1 Homo sapiens 117-121 32705692-2 2021 We assessed the impact of RI and single dose oral rifampin (RIF) on the pharmacokinetics of CYP3A, OATP1B, P-gp, and BCRP substrates using a microdose cocktail and OATP1B endogenous biomarkers. Rifampin 60-63 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 92-97 32705692-2 2021 We assessed the impact of RI and single dose oral rifampin (RIF) on the pharmacokinetics of CYP3A, OATP1B, P-gp, and BCRP substrates using a microdose cocktail and OATP1B endogenous biomarkers. Rifampin 60-63 phosphoglycolate phosphatase Homo sapiens 107-111 32705692-2 2021 We assessed the impact of RI and single dose oral rifampin (RIF) on the pharmacokinetics of CYP3A, OATP1B, P-gp, and BCRP substrates using a microdose cocktail and OATP1B endogenous biomarkers. Rifampin 60-63 BCR pseudogene 1 Homo sapiens 117-121 33130504-1 2021 The Xpert MTB/RIF assay is a molecular assay that has improved the detection of tuberculosis and rifampicin resistance. Rifampin 97-107 ras homolog family member F, filopodia associated Homo sapiens 14-17 33509114-1 2021 BACKGROUND: Xpert MTB/RIF (Xpert) has been recommended by WHO as the initial diagnostic test for TB and rifampicin-resistance detection. Rifampin 104-114 ras homolog family member F, filopodia associated Homo sapiens 22-25 32759993-0 2021 New paradigms of USP53 disease: normal GGT cholestasis, BRIC, cholangiopathy, and responsiveness to rifampicin. Rifampin 100-110 ubiquitin specific peptidase 53 Homo sapiens 17-22 33155329-2 2021 Since rifampicin is a hepatic enzyme (CYP3A4) inducer, in a post-renal transplant recipient, the dose of calcineurin inhibitors needs to be up-regulated and frequently monitored. Rifampin 6-16 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 38-44 33506323-4 2021 The aim of this study was to investigate the effects of rifampicin (a strong CYP3A4 inducer) on the pharmacokinetics of alflutinib and AST5902 in healthy volunteers, thus providing important information for drug-drug interaction evaluation and guiding clinical usage. Rifampin 56-66 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 77-83 33506323-11 2021 Conclusions As a strong CYP3A4 inducer, rifampicin exerted significant effects on the pharmacokinetics of alflutinib and the total active ingredients (alflutinib and AST5902). Rifampin 40-50 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-30 33542643-2 2021 Due to rifampin"s strong induction of CYP2C9, most cases could not attain the target international normalized ratio (INR) despite warfarin dose escalation. Rifampin 7-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 33614819-0 2021 Low Cycle Threshold Value in Xpert MTB/RIF Assay May Herald False Detection of Tuberculosis and Rifampicin Resistance: A Study of Two Cases. Rifampin 96-106 ras homolog family member F, filopodia associated Homo sapiens 39-42 33614819-1 2021 We report 2 cases for whom Xpert MTB/RIF falsely signaled rifampicin-resistant tuberculosis, based on unusually low cycle threshold and 3 of 5 probes missing. Rifampin 58-68 ras homolog family member F, filopodia associated Homo sapiens 37-40 32460379-3 2021 Current data indicate that intestinal P-gp protein levels can be induced <=3-4 fold in humans primarily with pregnane X receptor (PXR) activators, and that this induction can decrease the systemic exposure of drugs with P-gp efflux-limited absorption (e.g., <=67% decrease in the exposure of total dabigatran following rifampin multiple oral dosing). Rifampin 319-327 ATP binding cassette subfamily B member 1 Homo sapiens 38-42 33448348-37 2021 Xpert Ultra and Xpert MTB/RIF had similar sensitivity and specificity for rifampicin resistance. Rifampin 74-84 ras homolog family member F, filopodia associated Homo sapiens 26-29 33443639-1 2021 Diagnostic yield of an automated molecular test, Gene Xpert Mycobacterium tuberculosis/rifampicin (MTB/RIF), was evaluated in this study to simultaneously detect the MTB gene and resistance to rifampicin (RIF) on cytology samples acquired via endoscopic ultrasound (EUS)-guided fine-needle aspiration (FNAC) in suspected tubercular lymphadenitis. Rifampin 87-97 metallothionein 1J, pseudogene Homo sapiens 99-102 33443639-1 2021 Diagnostic yield of an automated molecular test, Gene Xpert Mycobacterium tuberculosis/rifampicin (MTB/RIF), was evaluated in this study to simultaneously detect the MTB gene and resistance to rifampicin (RIF) on cytology samples acquired via endoscopic ultrasound (EUS)-guided fine-needle aspiration (FNAC) in suspected tubercular lymphadenitis. Rifampin 87-97 metallothionein 1J, pseudogene Homo sapiens 166-169 33443639-1 2021 Diagnostic yield of an automated molecular test, Gene Xpert Mycobacterium tuberculosis/rifampicin (MTB/RIF), was evaluated in this study to simultaneously detect the MTB gene and resistance to rifampicin (RIF) on cytology samples acquired via endoscopic ultrasound (EUS)-guided fine-needle aspiration (FNAC) in suspected tubercular lymphadenitis. Rifampin 193-203 metallothionein 1J, pseudogene Homo sapiens 99-102 32460379-3 2021 Current data indicate that intestinal P-gp protein levels can be induced <=3-4 fold in humans primarily with pregnane X receptor (PXR) activators, and that this induction can decrease the systemic exposure of drugs with P-gp efflux-limited absorption (e.g., <=67% decrease in the exposure of total dabigatran following rifampin multiple oral dosing). Rifampin 319-327 ATP binding cassette subfamily B member 1 Homo sapiens 220-224 33305020-6 2020 The GeneXpert Mtuberculosis/RIF assay is a novel molecular diagnostic method for rapid diagnosis of tuberculosis and rifampicin resistance in clinical specimens. Rifampin 117-127 ras homolog family member F, filopodia associated Homo sapiens 28-31 32990533-10 2021 Rifampin is reported to perpetrate drug interactions by inducing CYP3A or inhibiting OATP1B; it is also reported to be a victim of drug interactions, due to the dual inhibition of CYP3A4 and OATP1B by indinavir. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-70 32990533-10 2021 Rifampin is reported to perpetrate drug interactions by inducing CYP3A or inhibiting OATP1B; it is also reported to be a victim of drug interactions, due to the dual inhibition of CYP3A4 and OATP1B by indinavir. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 180-186 32806986-5 2021 With the introduction of Xpert MTB/RIF and more recently Xpert Ultra, this has changed TB diagnostics by providing a rapid accessible platform to diagnose TB and identify rifampicin resistance within 2 hours. Rifampin 171-181 ras homolog family member F, filopodia associated Homo sapiens 35-38 33159914-6 2021 The doubly transfected MDCKII cells were also used to kinetically analyze the inhibitory effects of rifampicin on BSEP and NTCP. Rifampin 100-110 ATP binding cassette subfamily B member 11 Homo sapiens 114-118 33139294-7 2020 CSF penetration of rifampicin was relatively poor and increased with increasing protein levels in CSF, a marker of CSF inflammation. Rifampin 19-29 colony stimulating factor 2 Homo sapiens 0-3 33139294-7 2020 CSF penetration of rifampicin was relatively poor and increased with increasing protein levels in CSF, a marker of CSF inflammation. Rifampin 19-29 colony stimulating factor 2 Homo sapiens 98-101 33322603-5 2020 Whole-mount in situ hybridization revealed cyp2y3, 2r1, and 3a65 transcripts in larvae at 55 hpf after exposure to rifampicin, phenobarbital, or 2,3,7,8-tetrachlorodibenzo-p-dioxin from 30 hpf onward. Rifampin 115-125 cytochrome P450, family 2, subfamily Y, polypeptide 3 Danio rerio 43-49 33317678-0 2020 Systematic rifampicin resistance errors with Xpert MTB/RIF Ultra: implications for regulation of genotypic assays. Rifampin 11-21 ras homolog family member F, filopodia associated Homo sapiens 56-59 33361721-8 2020 Xpert MTB/RIF was able to detect rifampicin resistance with a sensitivity and specificity of 87.5% and 98.9%, respectively. Rifampin 33-43 ras homolog family member F, filopodia associated Homo sapiens 10-13 33361721-13 2020 Xpert MTB/RIF can be employed in urban district health facilities not only to diagnose PTB in smear-positive patients, but also to detect rifampicin resistance with good sensitivity and specificity. Rifampin 138-148 ras homolog family member F, filopodia associated Homo sapiens 10-13 32415528-7 2020 The induction of neural cell death by duloxetine was not affected by classic cell death inhibitors but was promoted by the CYP inducer rifampicin. Rifampin 135-145 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 123-126 33127565-0 2021 Hepatocyte-derived MANF is protective for rifampicin-induced cholestatic hepatic injury via inhibiting ATF4-CHOP signal activation. Rifampin 42-52 mesencephalic astrocyte-derived neurotrophic factor Mus musculus 19-23 33127565-0 2021 Hepatocyte-derived MANF is protective for rifampicin-induced cholestatic hepatic injury via inhibiting ATF4-CHOP signal activation. Rifampin 42-52 activating transcription factor 4 Mus musculus 103-107 33127565-0 2021 Hepatocyte-derived MANF is protective for rifampicin-induced cholestatic hepatic injury via inhibiting ATF4-CHOP signal activation. Rifampin 42-52 DNA-damage inducible transcript 3 Mus musculus 108-112 33127565-2 2021 Here we found that mesencephalic astrocyte-derived neurotrophic factor (MANF), an endoplasmic reticulum (ER) stress inducible protein, is a protector in RFP-induced liver injury. Rifampin 153-156 mesencephalic astrocyte-derived neurotrophic factor Mus musculus 19-70 33127565-2 2021 Here we found that mesencephalic astrocyte-derived neurotrophic factor (MANF), an endoplasmic reticulum (ER) stress inducible protein, is a protector in RFP-induced liver injury. Rifampin 153-156 mesencephalic astrocyte-derived neurotrophic factor Mus musculus 72-76 33386800-4 2021 OBJECTIVE: Based on our previous findings that rifampicin possesses neuroprotective effects on improving cognitive function after neuroinflammation, we aimed to examine in this study whether rifampicin can inhibit Abeta accumulation by enhancing autophagy in a mouse model of lipopolysaccharide (LPS)-induced cognitive impairment. Rifampin 191-201 amyloid beta (A4) precursor protein Mus musculus 214-219 33386800-9 2021 Our results further demonstrated that rifampicin improved the neurological function by promoting autophagy through the inhibition of Akt/mTOR/p70S6K signaling pathway in the hippocampus of LPS-induced mice. Rifampin 38-48 thymoma viral proto-oncogene 1 Mus musculus 133-136 33386800-9 2021 Our results further demonstrated that rifampicin improved the neurological function by promoting autophagy through the inhibition of Akt/mTOR/p70S6K signaling pathway in the hippocampus of LPS-induced mice. Rifampin 38-48 mechanistic target of rapamycin kinase Mus musculus 137-141 33386800-9 2021 Our results further demonstrated that rifampicin improved the neurological function by promoting autophagy through the inhibition of Akt/mTOR/p70S6K signaling pathway in the hippocampus of LPS-induced mice. Rifampin 38-48 ribosomal protein S6 kinase, polypeptide 1 Mus musculus 142-148 33386800-10 2021 CONCLUSION: Rifampicin ameliorates cognitive impairment by suppression of Abeta1-42 accumulation through inhibition of Akt/mTOR/p70S6K signaling and enhancement of autophagy in the hippocampus of LPS-induced mice. Rifampin 12-22 thymoma viral proto-oncogene 1 Mus musculus 119-122 33386800-10 2021 CONCLUSION: Rifampicin ameliorates cognitive impairment by suppression of Abeta1-42 accumulation through inhibition of Akt/mTOR/p70S6K signaling and enhancement of autophagy in the hippocampus of LPS-induced mice. Rifampin 12-22 mechanistic target of rapamycin kinase Mus musculus 123-127 33386800-10 2021 CONCLUSION: Rifampicin ameliorates cognitive impairment by suppression of Abeta1-42 accumulation through inhibition of Akt/mTOR/p70S6K signaling and enhancement of autophagy in the hippocampus of LPS-induced mice. Rifampin 12-22 ribosomal protein S6 kinase, polypeptide 1 Mus musculus 128-134 33268331-6 2020 Importantly, both fresh and cryopreserved Definitive Endoderm and Hepatoblasts successfully differentiated to pure and functional hepatocytes with increased CYP3A4 activity in response to rifampicin and lipid accumulation upon fatty acid (FA) treatment. Rifampin 188-198 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 157-163 33305020-16 2020 GeneXpert Mtuberculosis/RIF showed 2 cases of rifampicin resistance out of 67 cases. Rifampin 46-56 ras homolog family member F, filopodia associated Homo sapiens 24-27 32788213-2 2020 An anti-epileptic drug, phenobarbital (PB), activates CAR and its target CYP2B genes, while PXR is activated by drugs such as rifampicin and statins for the CYP3A genes. Rifampin 126-136 nuclear receptor subfamily 1 group I member 2 Homo sapiens 92-95 32788213-2 2020 An anti-epileptic drug, phenobarbital (PB), activates CAR and its target CYP2B genes, while PXR is activated by drugs such as rifampicin and statins for the CYP3A genes. Rifampin 126-136 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 157-162 33147218-0 2020 Frequency of MTB and rifampicin resistance MTB using Xpert-MTB/RIF assay among adult presumptive tuberculosis patients in Tigray, Northern Ethiopia: A cross sectional study. Rifampin 21-31 metallothionein 1J, pseudogene Homo sapiens 43-46 31665299-8 2020 Rifampicin CSF concentrations were described by a partition coefficient (5.5% [95%CI 4.4-6.4]) and half-life for distribution plasma to CSF (2.1 h [1.3-2.9]). Rifampin 0-10 colony stimulating factor 2 Homo sapiens 11-14 31665299-8 2020 Rifampicin CSF concentrations were described by a partition coefficient (5.5% [95%CI 4.4-6.4]) and half-life for distribution plasma to CSF (2.1 h [1.3-2.9]). Rifampin 0-10 colony stimulating factor 2 Homo sapiens 136-139 33147218-0 2020 Frequency of MTB and rifampicin resistance MTB using Xpert-MTB/RIF assay among adult presumptive tuberculosis patients in Tigray, Northern Ethiopia: A cross sectional study. Rifampin 21-31 metallothionein 1J, pseudogene Homo sapiens 43-46 33147218-2 2020 Data on rifampicin resistance MTB using Xpert- MTB/RIF assay in Ethiopia, particularly in the study area is limited. Rifampin 8-18 metallothionein 1J, pseudogene Homo sapiens 30-33 33147218-2 2020 Data on rifampicin resistance MTB using Xpert- MTB/RIF assay in Ethiopia, particularly in the study area is limited. Rifampin 8-18 metallothionein 1J, pseudogene Homo sapiens 47-50 33147218-3 2020 The aim of this study was to determine the frequency of MTB and rifampicin resistant-MTB among presumptive tuberculosis patients in Tigray, Northern Ethiopia. Rifampin 64-74 metallothionein 1J, pseudogene Homo sapiens 85-88 33147218-13 2020 Likewise, rifampicin resistant-MTB was more prevalent among relapse (p < 0.001) and failure cases (p = 0.025); while age group 30-39 years was significantly associated with lower frequency of rifampicin resistant-MTB (p = 0.008). Rifampin 10-20 metallothionein 1J, pseudogene Homo sapiens 31-34 33147218-13 2020 Likewise, rifampicin resistant-MTB was more prevalent among relapse (p < 0.001) and failure cases (p = 0.025); while age group 30-39 years was significantly associated with lower frequency of rifampicin resistant-MTB (p = 0.008). Rifampin 10-20 metallothionein 1J, pseudogene Homo sapiens 213-216 33147218-13 2020 Likewise, rifampicin resistant-MTB was more prevalent among relapse (p < 0.001) and failure cases (p = 0.025); while age group 30-39 years was significantly associated with lower frequency of rifampicin resistant-MTB (p = 0.008). Rifampin 192-202 metallothionein 1J, pseudogene Homo sapiens 31-34 33147218-13 2020 Likewise, rifampicin resistant-MTB was more prevalent among relapse (p < 0.001) and failure cases (p = 0.025); while age group 30-39 years was significantly associated with lower frequency of rifampicin resistant-MTB (p = 0.008). Rifampin 192-202 metallothionein 1J, pseudogene Homo sapiens 213-216 33147218-14 2020 CONCLUSION: Frequency of MTB among tuberculosis presumptive patients was low; however, the problem of rifampicin resistant-MTB among the tuberculosis confirmed patients was high. Rifampin 102-112 metallothionein 1J, pseudogene Homo sapiens 123-126 32779277-9 2020 While broadly inhibiting plastid transcription with rifampicin was also able to suppress cell death in fc2 mutants, specific reductions in plastid gene expression using other mutations was not always sufficient. Rifampin 52-62 ferrochelatase 2 Arabidopsis thaliana 103-106 33030266-3 2020 Physiologically-based pharmacokinetic (PBPK) models for apixaban and rivaroxaban were developed to estimate the net effect of CYP3A induction, P-gp inhibition and P-gp induction by rifampicin. Rifampin 181-191 ATP binding cassette subfamily B member 1 Homo sapiens 163-167 33187034-2 2020 Xpert MTB/ RIF assay is a fully automated cartridge-based real-time PCR to detect MTB and resistance to rifampicin within two hours using three specific primers and five unique molecular probes to target the rpoB gene. Rifampin 104-114 ras homolog family member F, filopodia associated Homo sapiens 11-14 32935287-7 2020 Rifampicin 600 mg/day resulted in ~ 90% reductions in both the Cmax (Day 10 GMR [90% CI], 0.14 [0.12-0.15]) and AUC (0.07 [0.06-0.07]) of balovaptan 10 mg/day. Rifampin 0-10 colony stimulating factor 2 receptor subunit alpha Homo sapiens 76-79 32978634-7 2020 Co-administration of the strong CYP3A4 inducer, rifampin, predicted a greater DDI effect on a single dose of ivosidenib than on multiple doses (AUC ratios 0.35 and 0.67, Cmax ratios 0.91 and 0.81, respectively). Rifampin 48-56 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-38 32748370-9 2020 Significantly accelerated GR nuclear translocation was determined in EPI-ECs following treatment with GR modulators/ligands dexamethasone, rifampicin, or phenytoin. Rifampin 139-149 nuclear receptor subfamily 3 group C member 1 Homo sapiens 26-28 32748370-9 2020 Significantly accelerated GR nuclear translocation was determined in EPI-ECs following treatment with GR modulators/ligands dexamethasone, rifampicin, or phenytoin. Rifampin 139-149 nuclear receptor subfamily 3 group C member 1 Homo sapiens 102-104 33879905-1 2020 OBJECTIVE: To systematically review the diagnostic accuracy of Xpert Mycobacterium tuberculosis/rifampicin (Xpert MTB/RIF) for the detection of active tuberculosis (TB) and rifampicin-resistance TB in Chinese patients. Rifampin 97-107 ras homolog family member F, filopodia associated Homo sapiens 120-123 33879905-16 2020 CONCLUSION: Xpert MTB/RIF has good sensitivity and specificity in detecting TB and rifampicin-resistant TB in Chinese people. Rifampin 84-94 ras homolog family member F, filopodia associated Homo sapiens 23-26 32320483-7 2020 Rifampicin requires solely PXR to induce CYP3A4 and CYP2B6, while phenobarbital-mediated induction of these CYPs did not show absolute dependency on either PXR or CAR suggesting its ability to switch nuclear receptor activation. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 27-30 33060611-8 2020 HepaMN cells showed increased CYP3A4 expression after exposure to rifampicin, implying that their close resemblance to normal human hepatocytes makes them suitable for research applications including drug metabolism studies. Rifampin 66-76 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 30-36 32235864-6 2020 Rifampin is a known strong CYP3A4 inducer and is recommended by the FDA as a positive control in the CYP3A4 induction assay. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 27-33 32235864-6 2020 Rifampin is a known strong CYP3A4 inducer and is recommended by the FDA as a positive control in the CYP3A4 induction assay. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 101-107 32250463-1 2020 AIMS: To investigate the effects of the strong cytochrome P450 (CYP) 3A inhibitor itraconazole and the strong CYP3A inducer rifampicin on the pharmacokinetics (PK) of single-dose esaxerenone, a non-steroidal mineralocorticoid receptor blocker, in healthy Japanese subjects. Rifampin 124-134 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 110-115 32857315-11 2020 Rifampin reduced the AUC for CC112273 and CC1084037, primarily via CYP2C8 induction, by approximately 60% and 55%, respectively. Rifampin 0-8 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 67-73 32344455-8 2020 In conclusion, rifampin stimulates renin activity and has a hypertensive effect. Rifampin 15-23 renin Homo sapiens 35-40 32320483-7 2020 Rifampicin requires solely PXR to induce CYP3A4 and CYP2B6, while phenobarbital-mediated induction of these CYPs did not show absolute dependency on either PXR or CAR suggesting its ability to switch nuclear receptor activation. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-47 32320483-7 2020 Rifampicin requires solely PXR to induce CYP3A4 and CYP2B6, while phenobarbital-mediated induction of these CYPs did not show absolute dependency on either PXR or CAR suggesting its ability to switch nuclear receptor activation. Rifampin 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 32344455-0 2020 Pregnane X receptor activator rifampin increases blood pressure and stimulates plasma renin activity. Rifampin 30-38 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-19 32344455-0 2020 Pregnane X receptor activator rifampin increases blood pressure and stimulates plasma renin activity. Rifampin 30-38 renin Homo sapiens 86-91 32344455-1 2020 We conducted a clinical trial with 22 healthy volunteers to investigate the effects of pregnane X receptor (PXR) agonist rifampin on blood pressure (BP). Rifampin 121-129 nuclear receptor subfamily 1 group I member 2 Homo sapiens 108-111 32719071-3 2020 This study investigated the in vivo effects of the common inducer rifampin (RIF) on the activity and expression of cynomolgus monkey OATP1B1 and OATP1B3 transporters, which are structurally and functionally similar their human OATP1B counterparts. Rifampin 66-74 solute carrier organic anion transporter family member 1B1 Homo sapiens 133-140 32275771-0 2020 The effects of weak and strong CYP3A induction by rifampicin on the pharmacokinetics of five progestins and ethinylestradiol compared to midazolam. Rifampin 50-60 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-36 32275771-3 2020 Rifampicin was used to induce CYP3A. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 30-35 32719071-3 2020 This study investigated the in vivo effects of the common inducer rifampin (RIF) on the activity and expression of cynomolgus monkey OATP1B1 and OATP1B3 transporters, which are structurally and functionally similar their human OATP1B counterparts. Rifampin 66-74 solute carrier organic anion transporter family member 1B3 Homo sapiens 145-152 32719071-3 2020 This study investigated the in vivo effects of the common inducer rifampin (RIF) on the activity and expression of cynomolgus monkey OATP1B1 and OATP1B3 transporters, which are structurally and functionally similar their human OATP1B counterparts. Rifampin 76-79 solute carrier organic anion transporter family member 1B1 Homo sapiens 133-140 32719071-3 2020 This study investigated the in vivo effects of the common inducer rifampin (RIF) on the activity and expression of cynomolgus monkey OATP1B1 and OATP1B3 transporters, which are structurally and functionally similar their human OATP1B counterparts. Rifampin 76-79 solute carrier organic anion transporter family member 1B3 Homo sapiens 145-152 32719071-9 2020 Rifampin treatment showed 2.0- to 3.3-fold increases in P-gp, BCRP, and MRP2 expression in the small intestine. Rifampin 0-8 glycerol-3-phosphate phosphatase Macaca fascicularis 56-60 32690641-0 2020 Rifampicin transport by OATP1B1 variants. Rifampin 0-10 solute carrier organic anion transporter family member 1B1 Homo sapiens 24-31 32029904-6 2020 Using this system, we validated the association of hypermethylation of CYP2D6 and CYP2E1 with rifampin-induced DILI. Rifampin 94-102 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 71-77 32029904-6 2020 Using this system, we validated the association of hypermethylation of CYP2D6 and CYP2E1 with rifampin-induced DILI. Rifampin 94-102 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 82-88 32029904-7 2020 Our results revealed that, following treatment of HepaRG cells with rifampin, the methylation levels of CYP2D6 and CYP2E1 were inversely proportional to cell viability and glutathione content, and directly proportional to caspase 3/7 activity. Rifampin 68-76 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 104-110 32029904-7 2020 Our results revealed that, following treatment of HepaRG cells with rifampin, the methylation levels of CYP2D6 and CYP2E1 were inversely proportional to cell viability and glutathione content, and directly proportional to caspase 3/7 activity. Rifampin 68-76 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 115-121 32029904-7 2020 Our results revealed that, following treatment of HepaRG cells with rifampin, the methylation levels of CYP2D6 and CYP2E1 were inversely proportional to cell viability and glutathione content, and directly proportional to caspase 3/7 activity. Rifampin 68-76 caspase 3 Homo sapiens 222-231 33061879-4 2020 Hepatotoxicity is the most commonly reported ADR in patients treated with anti-tubercular drugs such as isoniazid, rifampicin and pyrazinamide. Rifampin 115-125 aldo-keto reductase family 1 member B Homo sapiens 45-48 33117645-2 2020 Xpert MTB/RIF was positive after his second rifampicin-susceptible TB treatment. Rifampin 44-54 ras homolog family member F, filopodia associated Homo sapiens 10-13 32690641-1 2020 Single nucleotide polymorphisms in the OATP1B1 transporter have been suggested to partially explain the large interindividual variation in rifampicin exposure. Rifampin 139-149 solute carrier organic anion transporter family member 1B1 Homo sapiens 39-46 32100958-4 2020 SLCO1B1 rs4149056 was associated with reduction in rifampin exposure, which has implications for dosing this important anti-tuberculosis therapy. Rifampin 51-59 solute carrier organic anion transporter family member 1B1 Homo sapiens 0-7 33041817-8 2020 Activation of PXR and CAR in cells treated with 6-(4-chlorophenyl)-imidazo (2,1-b) thiazole-5-carbaldehyde (CITCO) and rifampicin (Rif) resulted in the enhancement of CYP450 isoforms, UGT1A1, PXR, and CAR. Rifampin 119-129 nuclear receptor subfamily 1 group I member 2 Homo sapiens 14-17 33041817-8 2020 Activation of PXR and CAR in cells treated with 6-(4-chlorophenyl)-imidazo (2,1-b) thiazole-5-carbaldehyde (CITCO) and rifampicin (Rif) resulted in the enhancement of CYP450 isoforms, UGT1A1, PXR, and CAR. Rifampin 119-129 nuclear receptor subfamily 1 group I member 3 Homo sapiens 22-25 33041817-8 2020 Activation of PXR and CAR in cells treated with 6-(4-chlorophenyl)-imidazo (2,1-b) thiazole-5-carbaldehyde (CITCO) and rifampicin (Rif) resulted in the enhancement of CYP450 isoforms, UGT1A1, PXR, and CAR. Rifampin 131-134 nuclear receptor subfamily 1 group I member 2 Homo sapiens 14-17 33041817-8 2020 Activation of PXR and CAR in cells treated with 6-(4-chlorophenyl)-imidazo (2,1-b) thiazole-5-carbaldehyde (CITCO) and rifampicin (Rif) resulted in the enhancement of CYP450 isoforms, UGT1A1, PXR, and CAR. Rifampin 131-134 nuclear receptor subfamily 1 group I member 3 Homo sapiens 22-25 32936818-0 2020 Post-exposure prophylaxis (PEP) efficacy of rifampin, rifapentine, moxifloxacin, minocycline, and clarithromycin in a susceptible-subclinical model of leprosy. Rifampin 44-52 prolyl endopeptidase Mus musculus 27-30 32557601-3 2020 Ritonavir (a strong CYP3A inhibitor) increased ribociclib 400 mg single-dose AUC by 3.2-fold, whereas rifampin (a strong CYP3A inducer) decreased ribociclib AUC by 89% in healthy volunteers (HV). Rifampin 102-110 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 121-126 32166864-0 2020 Effect of rifampin-mediated OATP1B1 and OATP1B3 transporter inhibition on the pharmacokinetics of the P2Y12 receptor antagonist selatogrel. Rifampin 10-18 solute carrier organic anion transporter family member 1B1 Homo sapiens 28-35 32166864-0 2020 Effect of rifampin-mediated OATP1B1 and OATP1B3 transporter inhibition on the pharmacokinetics of the P2Y12 receptor antagonist selatogrel. Rifampin 10-18 purinergic receptor P2Y12 Homo sapiens 102-107 32166864-3 2020 Therefore, the study aim was to investigate the effect of rifampin-mediated OATP1B1 and OATP1B3 inhibition on the pharmacokinetics (PK) of selatogrel. Rifampin 58-66 solute carrier organic anion transporter family member 1B1 Homo sapiens 76-83 32824985-8 2020 Previously, mutation of the intracellular domain of TLR2 has demonstrated its role in determining the susceptibility to LL, though LL was successfully treated using a combination of DDS with rifampicin and clofazimine. Rifampin 191-201 toll like receptor 2 Homo sapiens 52-56 32387752-3 2020 Rifampicin and isoniazid co-therapy targets porphyrin biosynthesis via PXR and results in hepatic protoporphyrin IX accumulation and subsequent liver injury. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 71-74 31900032-3 2020 Values for the effective permeability and the intrinsic clearance of hepatic uptake by organic anion transporting polypeptide (OATP) 1B1 were optimized in a simulator platform.This PBPK model was subsequently validated using reported maximum pemafibrate plasma concentration and area under the curve values in reported interaction studies in healthy subjects co-administered with rifampicin.For subjects with Child-Pugh A and B liver cirrhosis, the intrinsic clearance of hepatic uptake of pemafibrate by OATP1B1 were modeled using 53% and 31% of that of healthy subjects, respectively. Rifampin 380-390 solute carrier organic anion transporter family member 1B1 Homo sapiens 87-136 32387752-2 2020 Pregnane X receptor (PXR, encoded by the NR1I2 gene) is a ligand-dependent transcription factor, and rifampicin is a human PXR-specific activator. Rifampin 101-111 nuclear receptor subfamily 1 group I member 2 Homo sapiens 123-126 32918434-6 2020 We postulate that rifampicin has attenuated the therapeutic effect of amlodipine via potent induction of hepatic CYP3A4 but the failure to control the blood pressure even with medications unrelated to cytochrome P450 pathways raises the spectre of an additional interaction. Rifampin 18-28 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 113-119 32853411-25 2020 Xpert MTB/RIF was accurate for detection of rifampicin resistance. Rifampin 44-54 ras homolog family member F, filopodia associated Homo sapiens 10-13 32750059-2 2020 Following a recent WHO guideline, the Indian National Leprosy Programme is introducing post-exposure prophylaxis with single-dose rifampicin (SDR-PEP) in all high-endemic districts of the country. Rifampin 130-140 caveolae associated protein 2 Homo sapiens 142-145 32750059-2 2020 Following a recent WHO guideline, the Indian National Leprosy Programme is introducing post-exposure prophylaxis with single-dose rifampicin (SDR-PEP) in all high-endemic districts of the country. Rifampin 130-140 progestagen associated endometrial protein Homo sapiens 146-149 32492461-6 2020 These data suggested that rifampin increases TG and TC levels in the liver may be related to activate HMGCR, CYP7A1, PXR and FXR, theses toxic actions of rifampin were alleviated by pyrazinamide may be due to inhibite the activity of CYP7A1, PXR and FAS, and increasing the LPL protein expression and activity. Rifampin 26-34 3-hydroxy-3-methylglutaryl-Coenzyme A reductase Mus musculus 102-107 32492461-6 2020 These data suggested that rifampin increases TG and TC levels in the liver may be related to activate HMGCR, CYP7A1, PXR and FXR, theses toxic actions of rifampin were alleviated by pyrazinamide may be due to inhibite the activity of CYP7A1, PXR and FAS, and increasing the LPL protein expression and activity. Rifampin 26-34 cytochrome P450, family 7, subfamily a, polypeptide 1 Mus musculus 109-115 32492461-0 2020 Pyrazinamide alleviates rifampin-induced steatohepatitis in mice by regulating the activities of cholesterol-activated 7alpha-hydroxylase and lipoprotein lipase. Rifampin 24-32 lipoprotein lipase Mus musculus 142-160 32492461-3 2020 The results showed that rifampin significantly increased transaminases, TBIL and TBA levels in serum, increased TG, TC content, HMGCR and CYP7A1 protein, CYP7A1, FGFR4, PXR, FAS and FXR mRNA expression, but decreased the level of SREBP-1c mRNA and induced severe steatohepatitis and hepatocyte necrosis in liver in mice. Rifampin 24-32 3-hydroxy-3-methylglutaryl-Coenzyme A reductase Mus musculus 128-133 32492461-6 2020 These data suggested that rifampin increases TG and TC levels in the liver may be related to activate HMGCR, CYP7A1, PXR and FXR, theses toxic actions of rifampin were alleviated by pyrazinamide may be due to inhibite the activity of CYP7A1, PXR and FAS, and increasing the LPL protein expression and activity. Rifampin 26-34 nuclear receptor subfamily 1, group I, member 2 Mus musculus 117-120 32492461-3 2020 The results showed that rifampin significantly increased transaminases, TBIL and TBA levels in serum, increased TG, TC content, HMGCR and CYP7A1 protein, CYP7A1, FGFR4, PXR, FAS and FXR mRNA expression, but decreased the level of SREBP-1c mRNA and induced severe steatohepatitis and hepatocyte necrosis in liver in mice. Rifampin 24-32 cytochrome P450, family 7, subfamily a, polypeptide 1 Mus musculus 138-144 32492461-3 2020 The results showed that rifampin significantly increased transaminases, TBIL and TBA levels in serum, increased TG, TC content, HMGCR and CYP7A1 protein, CYP7A1, FGFR4, PXR, FAS and FXR mRNA expression, but decreased the level of SREBP-1c mRNA and induced severe steatohepatitis and hepatocyte necrosis in liver in mice. Rifampin 24-32 cytochrome P450, family 7, subfamily a, polypeptide 1 Mus musculus 154-160 32492461-6 2020 These data suggested that rifampin increases TG and TC levels in the liver may be related to activate HMGCR, CYP7A1, PXR and FXR, theses toxic actions of rifampin were alleviated by pyrazinamide may be due to inhibite the activity of CYP7A1, PXR and FAS, and increasing the LPL protein expression and activity. Rifampin 26-34 nuclear receptor subfamily 1, group H, member 4 Mus musculus 125-128 32492461-3 2020 The results showed that rifampin significantly increased transaminases, TBIL and TBA levels in serum, increased TG, TC content, HMGCR and CYP7A1 protein, CYP7A1, FGFR4, PXR, FAS and FXR mRNA expression, but decreased the level of SREBP-1c mRNA and induced severe steatohepatitis and hepatocyte necrosis in liver in mice. Rifampin 24-32 fibroblast growth factor receptor 4 Mus musculus 162-167 32492461-6 2020 These data suggested that rifampin increases TG and TC levels in the liver may be related to activate HMGCR, CYP7A1, PXR and FXR, theses toxic actions of rifampin were alleviated by pyrazinamide may be due to inhibite the activity of CYP7A1, PXR and FAS, and increasing the LPL protein expression and activity. Rifampin 26-34 cytochrome P450, family 7, subfamily a, polypeptide 1 Mus musculus 234-240 32492461-3 2020 The results showed that rifampin significantly increased transaminases, TBIL and TBA levels in serum, increased TG, TC content, HMGCR and CYP7A1 protein, CYP7A1, FGFR4, PXR, FAS and FXR mRNA expression, but decreased the level of SREBP-1c mRNA and induced severe steatohepatitis and hepatocyte necrosis in liver in mice. Rifampin 24-32 nuclear receptor subfamily 1, group I, member 2 Mus musculus 169-172 32492461-3 2020 The results showed that rifampin significantly increased transaminases, TBIL and TBA levels in serum, increased TG, TC content, HMGCR and CYP7A1 protein, CYP7A1, FGFR4, PXR, FAS and FXR mRNA expression, but decreased the level of SREBP-1c mRNA and induced severe steatohepatitis and hepatocyte necrosis in liver in mice. Rifampin 24-32 nuclear receptor subfamily 1, group H, member 4 Mus musculus 182-185 32492461-6 2020 These data suggested that rifampin increases TG and TC levels in the liver may be related to activate HMGCR, CYP7A1, PXR and FXR, theses toxic actions of rifampin were alleviated by pyrazinamide may be due to inhibite the activity of CYP7A1, PXR and FAS, and increasing the LPL protein expression and activity. Rifampin 26-34 nuclear receptor subfamily 1, group I, member 2 Mus musculus 242-245 32492461-6 2020 These data suggested that rifampin increases TG and TC levels in the liver may be related to activate HMGCR, CYP7A1, PXR and FXR, theses toxic actions of rifampin were alleviated by pyrazinamide may be due to inhibite the activity of CYP7A1, PXR and FAS, and increasing the LPL protein expression and activity. Rifampin 26-34 lipoprotein lipase Mus musculus 274-277 32492461-3 2020 The results showed that rifampin significantly increased transaminases, TBIL and TBA levels in serum, increased TG, TC content, HMGCR and CYP7A1 protein, CYP7A1, FGFR4, PXR, FAS and FXR mRNA expression, but decreased the level of SREBP-1c mRNA and induced severe steatohepatitis and hepatocyte necrosis in liver in mice. Rifampin 24-32 sterol regulatory element binding transcription factor 1 Mus musculus 230-238 32492461-6 2020 These data suggested that rifampin increases TG and TC levels in the liver may be related to activate HMGCR, CYP7A1, PXR and FXR, theses toxic actions of rifampin were alleviated by pyrazinamide may be due to inhibite the activity of CYP7A1, PXR and FAS, and increasing the LPL protein expression and activity. Rifampin 154-162 3-hydroxy-3-methylglutaryl-Coenzyme A reductase Mus musculus 102-107 32492461-6 2020 These data suggested that rifampin increases TG and TC levels in the liver may be related to activate HMGCR, CYP7A1, PXR and FXR, theses toxic actions of rifampin were alleviated by pyrazinamide may be due to inhibite the activity of CYP7A1, PXR and FAS, and increasing the LPL protein expression and activity. Rifampin 154-162 cytochrome P450, family 7, subfamily a, polypeptide 1 Mus musculus 109-115 32492461-6 2020 These data suggested that rifampin increases TG and TC levels in the liver may be related to activate HMGCR, CYP7A1, PXR and FXR, theses toxic actions of rifampin were alleviated by pyrazinamide may be due to inhibite the activity of CYP7A1, PXR and FAS, and increasing the LPL protein expression and activity. Rifampin 154-162 nuclear receptor subfamily 1, group I, member 2 Mus musculus 117-120 32492461-6 2020 These data suggested that rifampin increases TG and TC levels in the liver may be related to activate HMGCR, CYP7A1, PXR and FXR, theses toxic actions of rifampin were alleviated by pyrazinamide may be due to inhibite the activity of CYP7A1, PXR and FAS, and increasing the LPL protein expression and activity. Rifampin 154-162 nuclear receptor subfamily 1, group H, member 4 Mus musculus 125-128 32492461-6 2020 These data suggested that rifampin increases TG and TC levels in the liver may be related to activate HMGCR, CYP7A1, PXR and FXR, theses toxic actions of rifampin were alleviated by pyrazinamide may be due to inhibite the activity of CYP7A1, PXR and FAS, and increasing the LPL protein expression and activity. Rifampin 154-162 cytochrome P450, family 7, subfamily a, polypeptide 1 Mus musculus 234-240 32492461-6 2020 These data suggested that rifampin increases TG and TC levels in the liver may be related to activate HMGCR, CYP7A1, PXR and FXR, theses toxic actions of rifampin were alleviated by pyrazinamide may be due to inhibite the activity of CYP7A1, PXR and FAS, and increasing the LPL protein expression and activity. Rifampin 154-162 nuclear receptor subfamily 1, group I, member 2 Mus musculus 242-245 32492461-6 2020 These data suggested that rifampin increases TG and TC levels in the liver may be related to activate HMGCR, CYP7A1, PXR and FXR, theses toxic actions of rifampin were alleviated by pyrazinamide may be due to inhibite the activity of CYP7A1, PXR and FAS, and increasing the LPL protein expression and activity. Rifampin 154-162 lipoprotein lipase Mus musculus 274-277 32521634-5 2020 In this narrative review, we aim to identify literature that has explored the influence of single nucleotide polymorphisms (SNPs) of genes encoding drug transporters and their transcriptional regulators (SLCO1B1, ABCB1, PXR and CAR), metabolizing enzymes (CES1, CES2 and AADAC) and VDR and its pathway regulators (VDR, CYP27B1 and CYP24A1) on plasma RF concentrations in TB patients on antitubercular therapy. Rifampin 350-352 solute carrier organic anion transporter family member 1B1 Homo sapiens 204-211 32474202-9 2020 The HbA1c<7% group had a lower risk of developing rifampicin resistance, isoniazid resistance and MDR, with odd ratios (ORs) of 1.904 (p=0.001), 2.896 (p<0.001) and 3.228 (p<0.001), respectively. Rifampin 50-60 hemoglobin subunit alpha 1 Homo sapiens 4-8 32660103-0 2020 Toxicoproteomic Profiling of hPXR Transgenic Mice Treated with Rifampicin and Isoniazid. Rifampin 63-73 nuclear receptor subfamily 1 group I member 2 Homo sapiens 29-33 32350061-8 2020 Induction of CYP3A4 using 10 muM rifampicin was 12-fold for RNA, 8-fold for protein, and 3-fold for activity; for CYP1A2 with 50 muM omeprazole induction was 30-fold for RNA, 13-fold for protein, and 17-fold for activity; for CYP2B6 with 50 muM phenytoin induction was 23-fold for RNA, 2-fold for protein, and 5-fold for activity. Rifampin 33-43 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 13-19 32574211-4 2020 We discovered an aminopyrimidine small molecule, 2062, that had no direct antimycobacterial activity, but synergized with rifampin to reduce bacterial burden in Mtb infected macrophages and mice and also dampened lung immunopathology. Rifampin 122-130 non-SMC condensin II complex, subunit G2 Mus musculus 161-164 32361205-8 2020 Dose adjustment should be considered when brivaracetam is coadministered with the more potent CYP inducer, rifampin. Rifampin 107-115 peptidylprolyl isomerase G Homo sapiens 94-97 32002554-1 2020 BACKGROUND: Mutations in the genes inhA, katG, and rpoB confer resistance to anti-tuberculosis (TB) drugs isoniazid and rifampin. Rifampin 120-128 inhibin subunit alpha Homo sapiens 35-39 32521634-5 2020 In this narrative review, we aim to identify literature that has explored the influence of single nucleotide polymorphisms (SNPs) of genes encoding drug transporters and their transcriptional regulators (SLCO1B1, ABCB1, PXR and CAR), metabolizing enzymes (CES1, CES2 and AADAC) and VDR and its pathway regulators (VDR, CYP27B1 and CYP24A1) on plasma RF concentrations in TB patients on antitubercular therapy. Rifampin 350-352 CXADR pseudogene 1 Homo sapiens 228-231 32440172-0 2020 Successful Treatment of a Multidrug-Resistant Tuberculosis Patient with a Negative Xpert MTB/RIF Test for Rifampicin-Resistant Tuberculosis in Guizhou Province of China: A Case Report. Rifampin 106-116 ras homolog family member F, filopodia associated Homo sapiens 93-96 32052379-9 2020 Rifampin is the most potent P-gp inducer, resulting in an average reduction in substrate exposure ranging between 20 and 67%. Rifampin 0-8 ATP binding cassette subfamily B member 1 Homo sapiens 28-32 32538031-10 2020 Thirty samples (8.5%) were detected as rifampicin resistance by Xpert MTB/RIF assay. Rifampin 39-49 ras homolog family member F, filopodia associated Homo sapiens 74-77 32538031-12 2020 Xpert MTB/RIF assay is the most rapid, sensitive, and specific method for microbiological confirmation and rifampicin resistance detection in pediatric tuberculosis. Rifampin 107-117 ras homolog family member F, filopodia associated Homo sapiens 10-13 32303457-3 2020 Omeprazole and rifampicin respectively induced CYP1A1 and CYP3A8 mRNAs, while phenobarbital induced CYP2C43, CYP2C75, and CYP3A8, and slightly induced CYP2B6. Rifampin 15-25 cytochrome P450 family 1 subfamily A member 1 Macaca fascicularis 47-53 32303457-3 2020 Omeprazole and rifampicin respectively induced CYP1A1 and CYP3A8 mRNAs, while phenobarbital induced CYP2C43, CYP2C75, and CYP3A8, and slightly induced CYP2B6. Rifampin 15-25 cytochrome P450 family 3 subfamily A member 4 Macaca fascicularis 58-64 32303457-3 2020 Omeprazole and rifampicin respectively induced CYP1A1 and CYP3A8 mRNAs, while phenobarbital induced CYP2C43, CYP2C75, and CYP3A8, and slightly induced CYP2B6. Rifampin 15-25 cytochrome P450 family 2 subfamily C member 19 Macaca fascicularis 109-116 32303457-3 2020 Omeprazole and rifampicin respectively induced CYP1A1 and CYP3A8 mRNAs, while phenobarbital induced CYP2C43, CYP2C75, and CYP3A8, and slightly induced CYP2B6. Rifampin 15-25 cytochrome P450 family 3 subfamily A member 4 Macaca fascicularis 122-128 32303457-3 2020 Omeprazole and rifampicin respectively induced CYP1A1 and CYP3A8 mRNAs, while phenobarbital induced CYP2C43, CYP2C75, and CYP3A8, and slightly induced CYP2B6. Rifampin 15-25 cytochrome P450 family 2 subfamily B member 6 Macaca fascicularis 151-157 32303457-4 2020 The nuclear receptors AHR, PXR, and CAR mRNA levels, which were activated by omeprazole, rifampicin, and phenobarbital, respectively, tended to decrease via exposure to inducers despite the increase in CYP mRNA levels. Rifampin 89-99 aryl hydrocarbon receptor Macaca fascicularis 22-25 32303457-4 2020 The nuclear receptors AHR, PXR, and CAR mRNA levels, which were activated by omeprazole, rifampicin, and phenobarbital, respectively, tended to decrease via exposure to inducers despite the increase in CYP mRNA levels. Rifampin 89-99 nuclear receptor subfamily 1 group I member 3 Macaca fascicularis 36-39 31733411-2 2020 OBJECTIVE: Rifampicin resistant (RR) tuberculosis (TB) on X-pert MTB/Rif is assumed a surrogate for MDR TB. Rifampin 11-21 metallothionein 1J, pseudogene Homo sapiens 65-68 32440172-1 2020 The Xpert MTB/RIF (Xpert) assay recommended by the World Health Organization (WHO) can be used to simultaneously detect Mycobacterium tuberculosis complex (MTBC) and rifampicin (RIF) resistance associated mutations. Rifampin 166-176 ras homolog family member F, filopodia associated Homo sapiens 14-17 32267374-1 2020 The aim was to analyze the acceptability of chemoprophylaxis with single-dose rifampicin (PEP) in contacts, index leprosy cases, and health professionals and related factors that can influence adherence. Rifampin 78-88 prolyl endopeptidase Homo sapiens 90-93 32291610-8 2020 Further, the model was able to accurately predict the impact of a strong CYP3A inducer (rifampicin) and inhibitor (ketoconazole) on bortezomib exposure. Rifampin 88-98 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 73-78 31884012-3 2020 In hiPSC-IECs, gene expression levels of cytochrome P450 3A4 (CYP3A4) and P-glycoprotein (P-gp) are enhanced by rifampicin and 1,25-dihydroxyvitamin D3. Rifampin 112-122 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-60 31956998-5 2020 Rifampin 15mg/kg increased plasma and CSF exposures compared to 10mg/kg: day 14 plasma AUC0-24 increased from 48.2h mg/L (range 18.2-93.8) to 82.5h mg/L (range 8.7-161.0) and CSF AUC0-24 from 3.5h mg/L (range 1.2-9.6) to 6.0h mg/L (range 0.7-15.1). Rifampin 0-8 colony stimulating factor 2 Homo sapiens 38-41 31956998-5 2020 Rifampin 15mg/kg increased plasma and CSF exposures compared to 10mg/kg: day 14 plasma AUC0-24 increased from 48.2h mg/L (range 18.2-93.8) to 82.5h mg/L (range 8.7-161.0) and CSF AUC0-24 from 3.5h mg/L (range 1.2-9.6) to 6.0h mg/L (range 0.7-15.1). Rifampin 0-8 colony stimulating factor 2 Homo sapiens 175-178 31770456-2 2020 This open-label, two-arm, drug-drug interaction Phase 1 study in patients with advanced solid tumors assessed the effect of a P-gp inhibitor (itraconazole) and a P-gp inducer (rifampin) on the pharmacokinetics of a single dose of talazoparib. Rifampin 176-184 ATP binding cassette subfamily B member 1 Homo sapiens 162-166 32553308-0 2020 Prevalence of p-glycoprotein (PGP) expression, function and its effect on efficacy of rifampicin in patients with lymph node tuberculosis. Rifampin 86-96 ATP binding cassette subfamily B member 1 Homo sapiens 14-28 32553308-0 2020 Prevalence of p-glycoprotein (PGP) expression, function and its effect on efficacy of rifampicin in patients with lymph node tuberculosis. Rifampin 86-96 ATP binding cassette subfamily B member 1 Homo sapiens 30-33 32553308-3 2020 In this study we aim to study the prevalence of PGP expression and function and its relationship with serum concentrations of Rifampicin in consecutive patients with lymph node tuberculosis. Rifampin 126-136 ATP binding cassette subfamily B member 1 Homo sapiens 48-51 32553308-6 2020 The mean net PGP expression expressed as percent and relative fluorescence indices (RFI) of PGP expression and function respectively was compared at baseline at 2 months and was also correlated with serum rifampicin levels. Rifampin 205-215 ATP binding cassette subfamily B member 1 Homo sapiens 13-16 32553308-10 2020 CONCLUSIONS: Overexpression of PGP is common in patients with lymph node tuberculosis and leads to lower concentrations of Rifampicin in blood which subsequently may give rise to development of drug resistance. Rifampin 123-133 ATP binding cassette subfamily B member 1 Homo sapiens 31-34 31884012-3 2020 In hiPSC-IECs, gene expression levels of cytochrome P450 3A4 (CYP3A4) and P-glycoprotein (P-gp) are enhanced by rifampicin and 1,25-dihydroxyvitamin D3. Rifampin 112-122 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 31884012-3 2020 In hiPSC-IECs, gene expression levels of cytochrome P450 3A4 (CYP3A4) and P-glycoprotein (P-gp) are enhanced by rifampicin and 1,25-dihydroxyvitamin D3. Rifampin 112-122 ATP binding cassette subfamily B member 1 Homo sapiens 74-88 31884012-3 2020 In hiPSC-IECs, gene expression levels of cytochrome P450 3A4 (CYP3A4) and P-glycoprotein (P-gp) are enhanced by rifampicin and 1,25-dihydroxyvitamin D3. Rifampin 112-122 ATP binding cassette subfamily B member 1 Homo sapiens 90-94 31368836-5 2020 Rifampicin (OATP inhibitor) for OATP1B3-HEK293 cells and prazosin for OCT1-HEK293 cells could inhibit the uptake of jatrorrhizine with the IC50 of 5.49 +- 1.05 and 2.77 +- 0.72 muM, respectively.The above data indicate that hepatic uptake of jatrorrhizine is involved in both OATP and OCT, which may have important roles in jatrorrhizine liver disposition and potential drug-drug interactions. Rifampin 0-10 solute carrier organic anion transporter family member 1A2 Homo sapiens 12-16 31368836-5 2020 Rifampicin (OATP inhibitor) for OATP1B3-HEK293 cells and prazosin for OCT1-HEK293 cells could inhibit the uptake of jatrorrhizine with the IC50 of 5.49 +- 1.05 and 2.77 +- 0.72 muM, respectively.The above data indicate that hepatic uptake of jatrorrhizine is involved in both OATP and OCT, which may have important roles in jatrorrhizine liver disposition and potential drug-drug interactions. Rifampin 0-10 solute carrier organic anion transporter family member 1A2 Homo sapiens 32-36 32292343-4 2020 We measured oxysterols in serum of volunteers dosed with PXR agonist rifampicin 600 mg/day versus placebo for a week and analyzed the expression of cholesterol transporters in mononuclear cells. Rifampin 69-79 nuclear receptor subfamily 1 group I member 2 Homo sapiens 57-60 32376574-8 2020 The PXR target gene MDR1 also increased significantly in the rifampicin- and SR12813-treated cells compared with the control cells (P=0.020 and 0.01, respectively). Rifampin 61-71 nuclear receptor subfamily 1 group I member 2 Homo sapiens 4-7 32376574-8 2020 The PXR target gene MDR1 also increased significantly in the rifampicin- and SR12813-treated cells compared with the control cells (P=0.020 and 0.01, respectively). Rifampin 61-71 ATP binding cassette subfamily B member 1 Homo sapiens 20-24 32376574-10 2020 The protein level of PDCD4 increased significantly in both rifampicin (t= 2.779, P=0.025) group and VP- PXR group (t=3.066, P=0.012). Rifampin 59-69 programmed cell death 4 Homo sapiens 21-26 32376574-6 2020 RESULTS: The expressions of PDCD5 and PDCD6 mRNA did not differ significantly between rifampicin-treated and the control cells, while PDCD4 mRNA expression increased (t=4.209, P=0.008) and PDCD2 mRNA decreased significantly (t=-2.875, P=0.017) in rifampicin-treated cells. Rifampin 86-96 programmed cell death 2 Homo sapiens 189-194 32106230-7 2020 In addition, an increase in PXR expression levels was observed in both hepatic cell lines and iPSC derived hepatocytes upon rifampicin treatment, whereas a reproducible increase in PXR expression was not achieved in PHHs. Rifampin 124-134 nuclear receptor subfamily 1 group I member 2 Homo sapiens 28-31 32265867-5 2020 Deletion of greA results in growth retardation and poor survival in response to adverse stress, besides rendering M. tuberculosis more susceptible to vancomycin and rifampicin. Rifampin 165-175 transcription elongation factor GreA Mycobacterium tuberculosis H37Rv 12-16 31605489-7 2020 Simvastatin, fluvastatin and pravastatin showed the most favorable therapeutic index, and enhanced the anti-tubercular activity of the first-line drugs isoniazid, rifampin and pyrazinamide in THP-1 cells. Rifampin 163-171 GLI family zinc finger 2 Homo sapiens 192-197 31877331-5 2020 This priming effect persisted up to 48 h. Mechanistically, DMSO pretreatment reduced H4K12 acetylation and therefore enhanced the subsequent rifampicin stimulated histone H4R3 methylation on the regulatory region of PXR target gene CYP3A4. Rifampin 141-151 nuclear receptor subfamily 1 group I member 2 Homo sapiens 216-219 31877331-5 2020 This priming effect persisted up to 48 h. Mechanistically, DMSO pretreatment reduced H4K12 acetylation and therefore enhanced the subsequent rifampicin stimulated histone H4R3 methylation on the regulatory region of PXR target gene CYP3A4. Rifampin 141-151 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 232-238 32233619-0 2020 Effect of Xpert MTB/RIF on the treatment of multi-drug-resistant or rifampicin-resistant tuberculosis screened out from re-treatment pulmonary tuberculosis patients, a prospective cohort study. Rifampin 68-78 ras homolog family member F, filopodia associated Homo sapiens 20-23 32233619-2 2020 The Xpert MTB/RIF assay possesses high efficacy for the evaluation of rifampicin resistance. Rifampin 70-80 ras homolog family member F, filopodia associated Homo sapiens 14-17 31879282-12 2020 Although disparate sets of tissue biopsy (atorvastatin and carbamazepine) and in vitro hepatocyte (phenobarbital, chenodeoxycholate, and amprenavir) data present OATP messenger RNA induction (>=2-fold) by agents beyond RIF, the clinical relevance of such data needs to be determined. Rifampin 219-222 solute carrier organic anion transporter family member 1A2 Homo sapiens 162-166 31648047-5 2020 We found that rifampicin pretreatment suppressed the gene expression of IL-1beta and IL-6 via inhibiting activation of JNK after rotenone induction, but the anti-inflammatory effect of rifampicin was reversed by chloroquine. Rifampin 14-24 interleukin 1 alpha Homo sapiens 72-80 31648047-5 2020 We found that rifampicin pretreatment suppressed the gene expression of IL-1beta and IL-6 via inhibiting activation of JNK after rotenone induction, but the anti-inflammatory effect of rifampicin was reversed by chloroquine. Rifampin 14-24 interleukin 6 Homo sapiens 85-89 31648047-5 2020 We found that rifampicin pretreatment suppressed the gene expression of IL-1beta and IL-6 via inhibiting activation of JNK after rotenone induction, but the anti-inflammatory effect of rifampicin was reversed by chloroquine. Rifampin 14-24 mitogen-activated protein kinase 8 Homo sapiens 119-122 31648047-6 2020 Moreover, rifampicin pretreatment not only improved the ratio of LC3-II/LC3-I in rotenone-treated cells, but also increased autolysosomes and decreased autophagosomes in RFP-GFP-LC3B transfected HM cells exposed to rotenone, thus indicating rifampicin improves autophagy flux in rotenone-treated HM cells. Rifampin 10-20 tripartite motif containing 27 Homo sapiens 170-173 31648047-6 2020 Moreover, rifampicin pretreatment not only improved the ratio of LC3-II/LC3-I in rotenone-treated cells, but also increased autolysosomes and decreased autophagosomes in RFP-GFP-LC3B transfected HM cells exposed to rotenone, thus indicating rifampicin improves autophagy flux in rotenone-treated HM cells. Rifampin 10-20 microtubule associated protein 1 light chain 3 beta Homo sapiens 178-182 31648047-7 2020 Finally, we verified rifampicin pretreatment enhanced ATP6V0A1 expression when compared to that exposed to rotenone alone. Rifampin 21-31 ATPase H+ transporting V0 subunit a1 Homo sapiens 54-62 31648047-8 2020 ATP6V0A1 knockdown inhibited the effect of rifampicin on maintaining lysosome acidification and autophagosome-lysosome fusion in rotenone-treated microglia. Rifampin 43-53 ATPase H+ transporting V0 subunit a1 Homo sapiens 0-8 31648047-9 2020 Taken together, our results indicated that rifampicin attenuates rotenone-induced microglia inflammation partially via elevating ATP6V0A1. Rifampin 43-53 ATPase H+ transporting V0 subunit a1 Homo sapiens 129-137 31863284-2 2020 METHODS: This phase 1, open-label, two-period, crossover study estimated the effect of oral rifampin (a strong CYP3A inducer) on the pharmacokinetics and safety of oral lorlatinib (NCT02804399). Rifampin 92-100 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 111-116 31901518-4 2020 Here, we demonstrate that in the context of therapy, the AhR binds several TB drugs, including front line drugs rifampicin (RIF) and rifabutin (RFB), resulting in altered host defense and drug metabolism. Rifampin 112-122 aryl hydrocarbon receptor Homo sapiens 57-60 31901518-4 2020 Here, we demonstrate that in the context of therapy, the AhR binds several TB drugs, including front line drugs rifampicin (RIF) and rifabutin (RFB), resulting in altered host defense and drug metabolism. Rifampin 124-127 aryl hydrocarbon receptor Homo sapiens 57-60 32123755-1 2020 Rationale: Previously treated persons with bacteriologically confirmed pulmonary tuberculosis (BC-PTB) have increased risk of developing multi-drug resistant or rifampicin resistant tuberculosis (MDR/RR-TB). Rifampin 161-171 polypyrimidine tract binding protein 1 Homo sapiens 98-101 31993042-0 2019 Linezolid and Rifampicin Combination to Combat cfr-Positive Multidrug-Resistant MRSA in Murine Models of Bacteremia and Skin and Skin Structure Infection. Rifampin 14-24 solute carrier family 9 member A6 Homo sapiens 80-84 31993325-6 2020 Co-administration of eliglustat with oral doses of rifampin reduced eliglustat exposure by >85% due to induction of CYP3A4/P-gp by rifampin, while a single intravenous dose of rifampin had no effect on eliglustat, confirming that eliglustat is not an OATP substrate. Rifampin 51-59 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-122 31993325-6 2020 Co-administration of eliglustat with oral doses of rifampin reduced eliglustat exposure by >85% due to induction of CYP3A4/P-gp by rifampin, while a single intravenous dose of rifampin had no effect on eliglustat, confirming that eliglustat is not an OATP substrate. Rifampin 51-59 ATP binding cassette subfamily B member 1 Homo sapiens 123-127 31993325-6 2020 Co-administration of eliglustat with oral doses of rifampin reduced eliglustat exposure by >85% due to induction of CYP3A4/P-gp by rifampin, while a single intravenous dose of rifampin had no effect on eliglustat, confirming that eliglustat is not an OATP substrate. Rifampin 131-139 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-122 31993325-6 2020 Co-administration of eliglustat with oral doses of rifampin reduced eliglustat exposure by >85% due to induction of CYP3A4/P-gp by rifampin, while a single intravenous dose of rifampin had no effect on eliglustat, confirming that eliglustat is not an OATP substrate. Rifampin 131-139 ATP binding cassette subfamily B member 1 Homo sapiens 123-127 31993325-6 2020 Co-administration of eliglustat with oral doses of rifampin reduced eliglustat exposure by >85% due to induction of CYP3A4/P-gp by rifampin, while a single intravenous dose of rifampin had no effect on eliglustat, confirming that eliglustat is not an OATP substrate. Rifampin 131-139 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-122 31993325-6 2020 Co-administration of eliglustat with oral doses of rifampin reduced eliglustat exposure by >85% due to induction of CYP3A4/P-gp by rifampin, while a single intravenous dose of rifampin had no effect on eliglustat, confirming that eliglustat is not an OATP substrate. Rifampin 131-139 ATP binding cassette subfamily B member 1 Homo sapiens 123-127 32019201-1 2020 The purpose of this study is to analyze the polyphenolic rich extract of Crocus sativus L. petals (CSP) in modulating liver oxidative stress and inflammatory response status against rifampicin isoniazid (INH-RIF) drug-induced liver injury. Rifampin 182-202 DnaJ heat shock protein family (Hsp40) member C5 Rattus norvegicus 99-102 31993042-8 2019 Altogether, these data suggest that the use of linezolid in combination with rifampicin poses a viable therapeutic alternative for bacteremia and SSSI caused by cfr-positive multidrug resistant MRSA. Rifampin 77-87 solute carrier family 9 member A6 Homo sapiens 194-198 31678598-0 2020 Rifampicin activates AMPK and alleviates oxidative stress in the liver as mediated with Nrf2 signaling. Rifampin 0-10 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 21-25 31678598-0 2020 Rifampicin activates AMPK and alleviates oxidative stress in the liver as mediated with Nrf2 signaling. Rifampin 0-10 NFE2 like bZIP transcription factor 2 Homo sapiens 88-92 31678598-4 2020 In vivo, oral administration of rifampicin (100 or 200 mg/kg) attenuated elevation of serum alanine aminotransferase (ALT) and aspartate transaminase (AST), serum liver injury markers, against APAP treatment and, histologically, ameliorated tissue damage. Rifampin 32-42 glutamic--pyruvic transaminase Homo sapiens 92-116 31678598-4 2020 In vivo, oral administration of rifampicin (100 or 200 mg/kg) attenuated elevation of serum alanine aminotransferase (ALT) and aspartate transaminase (AST), serum liver injury markers, against APAP treatment and, histologically, ameliorated tissue damage. Rifampin 32-42 solute carrier family 17 member 5 Homo sapiens 127-149 31678598-4 2020 In vivo, oral administration of rifampicin (100 or 200 mg/kg) attenuated elevation of serum alanine aminotransferase (ALT) and aspartate transaminase (AST), serum liver injury markers, against APAP treatment and, histologically, ameliorated tissue damage. Rifampin 32-42 solute carrier family 17 member 5 Homo sapiens 151-154 31678598-7 2020 Further, we explored whether rifampicin treatment enhanced phosphorylation of AMP-activated protein kinase (AMPK) by activation of liver kinase B1 (LKB1), the upstream kinase of AMPKalpha. Rifampin 29-39 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 78-106 31678598-7 2020 Further, we explored whether rifampicin treatment enhanced phosphorylation of AMP-activated protein kinase (AMPK) by activation of liver kinase B1 (LKB1), the upstream kinase of AMPKalpha. Rifampin 29-39 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 108-112 31678598-7 2020 Further, we explored whether rifampicin treatment enhanced phosphorylation of AMP-activated protein kinase (AMPK) by activation of liver kinase B1 (LKB1), the upstream kinase of AMPKalpha. Rifampin 29-39 serine/threonine kinase 11 Homo sapiens 131-146 31678598-7 2020 Further, we explored whether rifampicin treatment enhanced phosphorylation of AMP-activated protein kinase (AMPK) by activation of liver kinase B1 (LKB1), the upstream kinase of AMPKalpha. Rifampin 29-39 serine/threonine kinase 11 Homo sapiens 148-152 31678598-9 2020 Moreover, we confirmed a reversed cell protective effect of rifampicin under compound C (an AMPK inhibitor) treatment. Rifampin 60-70 protein kinase AMP-activated catalytic subunit alpha 2 Homo sapiens 92-96 31678598-10 2020 Overall, our data demonstrate that rifampicin effectively protects the liver against cellular oxidative stress through AMPKalpha and Nrf2 pathway. Rifampin 35-45 NFE2 like bZIP transcription factor 2 Homo sapiens 133-137 31316180-0 2020 Rifampicin induces clathrin-dependent endocytosis and ubiquitin-proteasome degradation of MRP2 via oxidative stress-activated PKC-ERK/JNK/p38 and PI3K signaling pathways in HepG2 cells. Rifampin 0-10 ATP binding cassette subfamily C member 2 Homo sapiens 90-94 31998607-9 2020 Importantly, we observed significant upregulation of miR-18a-5p expression 6 h after treatment with the PXR ligand rifampicin, which indicates a putative mechanism underlying NR1I2 negative feed-back regulation in hepatic cells. Rifampin 115-125 nuclear receptor subfamily 1, group I, member 2 Mus musculus 104-107 31316180-0 2020 Rifampicin induces clathrin-dependent endocytosis and ubiquitin-proteasome degradation of MRP2 via oxidative stress-activated PKC-ERK/JNK/p38 and PI3K signaling pathways in HepG2 cells. Rifampin 0-10 mitogen-activated protein kinase 1 Homo sapiens 138-141 31316180-2 2020 In this study, we investigated the effects of rifampicin (RFP) on the membrane expression of multidrug resistance-associated protein 2 (MRP2) and the relationship between oxidative stress and RFP-induced endocytosis of MRP2 in HepG2 cells. Rifampin 46-56 ATP binding cassette subfamily C member 2 Homo sapiens 93-134 31316180-2 2020 In this study, we investigated the effects of rifampicin (RFP) on the membrane expression of multidrug resistance-associated protein 2 (MRP2) and the relationship between oxidative stress and RFP-induced endocytosis of MRP2 in HepG2 cells. Rifampin 46-56 ATP binding cassette subfamily C member 2 Homo sapiens 136-140 31571363-12 2020 Finally, in vivo, Nrf2 played an important role in RFP-induced liver injury (more serious liver injury in Nrf2-/- mice), and TAN IIA prevented it. Rifampin 51-54 nuclear factor, erythroid derived 2, like 2 Mus musculus 18-22 31604032-0 2020 Rifampicin, Not Vitamin E, Suppresses Parenteral Nutrition Associated Liver Disease Development through Pregnane X Receptor Pathway in Piglets. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 104-123 31432507-3 2020 Most importantly, we found that YPAC allows PHHs to retain enzymatic activities of CYP1A2, CYP2B6, and CYP3A4 even after 40 days of culture, and that inducibility of CYP3A4 activity in response to the prototypical inducers rifampicin and phenobarbital is also maintained. Rifampin 223-233 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 166-172 32407272-2 2020 Rifampicin is a nonselective inducer of P-glycoprotein (P-gp) and CYP enzymes such as CYP3A4 and others. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 86-92 31571363-0 2020 Tanshinone IIA prevents rifampicin-induced liver injury by regulating BSEP/NTCP expression via epigenetic activation of NRF2. Rifampin 24-34 ATP binding cassette subfamily B member 11 Homo sapiens 70-74 31571363-0 2020 Tanshinone IIA prevents rifampicin-induced liver injury by regulating BSEP/NTCP expression via epigenetic activation of NRF2. Rifampin 24-34 solute carrier family 10 member 1 Homo sapiens 75-79 31571363-12 2020 Finally, in vivo, Nrf2 played an important role in RFP-induced liver injury (more serious liver injury in Nrf2-/- mice), and TAN IIA prevented it. Rifampin 51-54 nuclear factor, erythroid derived 2, like 2 Mus musculus 106-110 31571363-0 2020 Tanshinone IIA prevents rifampicin-induced liver injury by regulating BSEP/NTCP expression via epigenetic activation of NRF2. Rifampin 24-34 NFE2 like bZIP transcription factor 2 Homo sapiens 120-124 31571363-14 2020 Pharmacological activation of NRF2 by TAN IIA may be beneficial for RFP-induced liver injury. Rifampin 68-71 NFE2 like bZIP transcription factor 2 Homo sapiens 30-34 31247120-7 2019 Furthermore, we applied a CRISPR/Cas9-mediated methylation modifiable cell model and demonstrated that four CpGs in or near the gene region of AK2, SLC8A2, and PSTPIP2 affected the cellular response to rifampicin treatment. Rifampin 202-212 adenylate kinase 2 Homo sapiens 143-146 31856744-13 2019 Rifampicin resistance rate found by GeneXpert MTB/RIF was 0.84%. Rifampin 0-10 ras homolog family member F, filopodia associated Homo sapiens 50-53 31856744-14 2019 Comparison of Rifampicin resistance obtained by GeneXpert MTB/RIF and Genotype MTBDRplus, showed 100% agreement between the two techniques for studied samples. Rifampin 14-24 ras homolog family member F, filopodia associated Homo sapiens 62-65 31805862-3 2019 Post-exposure prophylaxis (PEP) with a single dose of Rifampicin (SDR) has conditionally been recommended by the World Health Organization (WHO), based on a randomized-controlled-trial in Bangladesh. Rifampin 54-64 prolyl endopeptidase Homo sapiens 27-30 31129789-0 2019 Physiologically Based Pharmacokinetic Models for Prediction of Complex CYP2C8 and OATP1B1 (SLCO1B1) Drug-Drug-Gene Interactions: A Modeling Network of Gemfibrozil, Repaglinide, Pioglitazone, Rifampicin, Clarithromycin and Itraconazole. Rifampin 191-201 solute carrier organic anion transporter family member 1B1 Homo sapiens 82-89 31129789-0 2019 Physiologically Based Pharmacokinetic Models for Prediction of Complex CYP2C8 and OATP1B1 (SLCO1B1) Drug-Drug-Gene Interactions: A Modeling Network of Gemfibrozil, Repaglinide, Pioglitazone, Rifampicin, Clarithromycin and Itraconazole. Rifampin 191-201 solute carrier organic anion transporter family member 1B1 Homo sapiens 91-98 31247120-7 2019 Furthermore, we applied a CRISPR/Cas9-mediated methylation modifiable cell model and demonstrated that four CpGs in or near the gene region of AK2, SLC8A2, and PSTPIP2 affected the cellular response to rifampicin treatment. Rifampin 202-212 solute carrier family 8 member A2 Homo sapiens 148-154 31247120-7 2019 Furthermore, we applied a CRISPR/Cas9-mediated methylation modifiable cell model and demonstrated that four CpGs in or near the gene region of AK2, SLC8A2, and PSTPIP2 affected the cellular response to rifampicin treatment. Rifampin 202-212 proline-serine-threonine phosphatase interacting protein 2 Homo sapiens 160-167 31388941-12 2019 Conversely, strong CYP3A inducers, such as rifampin, are contraindicated with doravirine owing to a significant reduction in exposure with potential for impaired virological efficacy. Rifampin 43-51 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-24 31582395-3 2019 Three-day treatment of RIF significantly induced CYP3A4 (~60-fold induction), but not CYP1A2 and CYP2D6 genes. Rifampin 23-26 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 49-55 31571146-4 2019 A phase I drug-interaction study in healthy volunteers evaluated the impact of co-administering rifampicin [CYP3A4 and P-glycoprotein (P-gp) inducer] and itraconazole [CYP3A4, P-gp and breast cancer resistance protein (BCRP) inhibitor] on the pharmacokinetics of darolutamide. Rifampin 96-106 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 108-114 31686406-2 2019 Concomitant use of rifampicin, an inducer of these enzymes, with FXa-DOACs, has been shown to decrease FXa-DOAC concentrations in healthy subjects. Rifampin 19-29 coagulation factor X Homo sapiens 65-68 31686406-2 2019 Concomitant use of rifampicin, an inducer of these enzymes, with FXa-DOACs, has been shown to decrease FXa-DOAC concentrations in healthy subjects. Rifampin 19-29 coagulation factor X Homo sapiens 103-106 31571146-4 2019 A phase I drug-interaction study in healthy volunteers evaluated the impact of co-administering rifampicin [CYP3A4 and P-glycoprotein (P-gp) inducer] and itraconazole [CYP3A4, P-gp and breast cancer resistance protein (BCRP) inhibitor] on the pharmacokinetics of darolutamide. Rifampin 96-106 ATP binding cassette subfamily B member 1 Homo sapiens 135-139 31582395-9 2019 To investigate in vivo OATP induction, RIF (18 mg/kg per day) was orally dosed to cynomolgus monkeys for 7 days. Rifampin 39-42 solute carrier organic anion transporter family member 1A2 Homo sapiens 23-27 31172535-7 2019 Avadomide exposures when coadministered with the CYP3A inducer rifampin were 62.83% and 88.17% of that when administered alone, for AUC0-inf and Cmax , respectively. Rifampin 63-71 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 49-54 31976003-10 2019 Additionally, sesamin reduced valproate- and rifampin-induced LXRalpha and pregnane X receptor (PXR) transactivation. Rifampin 45-53 nuclear receptor subfamily 1 group H member 3 Homo sapiens 62-70 31976003-10 2019 Additionally, sesamin reduced valproate- and rifampin-induced LXRalpha and pregnane X receptor (PXR) transactivation. Rifampin 45-53 nuclear receptor subfamily 1 group I member 2 Homo sapiens 75-94 31976003-10 2019 Additionally, sesamin reduced valproate- and rifampin-induced LXRalpha and pregnane X receptor (PXR) transactivation. Rifampin 45-53 nuclear receptor subfamily 1 group I member 2 Homo sapiens 96-99 31676845-7 2019 We also demonstrated that matured Huh7s can be used for drug induction studies, where CYP3A4, CYP1A2, CYP2C9, and CYP2C19 inductions were achieved following rifampicin treatment. Rifampin 157-167 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 86-92 31754620-3 2019 Rifampicin-resistant TB (RR-TB) or multidrug-resistant TB (MDR-TB) was diagnosed upon the detection of rifampicin resistance by Xpert MTB/RIF or resistance to rifampicin and isoniazid by phenotypic drug susceptibility testing (DST). Rifampin 0-10 ras homolog family member F, filopodia associated Homo sapiens 138-141 31420941-2 2019 We previously constructed a physiologically-based pharmacokinetic (PBPK) model of rifampicin accounting for the components for the induction of cytochrome P450 (CYP) 3A/CYP2C9 and the inhibition of organic anion transporting polypeptide 1B (OATP1B). Rifampin 82-92 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 144-168 31420941-2 2019 We previously constructed a physiologically-based pharmacokinetic (PBPK) model of rifampicin accounting for the components for the induction of cytochrome P450 (CYP) 3A/CYP2C9 and the inhibition of organic anion transporting polypeptide 1B (OATP1B). Rifampin 82-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 31420941-3 2019 This study aimed to expand and verify the PBPK model for rifampicin by incorporating additional components for the induction of OATP1B and CYP2C8 and the inhibition of multidrug resistance protein 2. Rifampin 57-67 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 139-145 31455676-4 2019 Oral administration of the human PXR activator rifampicin increased hepatic expression of CYP3A4 mRNA and triazolam (TRZ) 1"- and 4-hydroxylation activities, CYP3A probe activities, in the liver and intestine microsomes of hCYP3A-MAC/hPXR mice. Rifampin 47-57 nuclear receptor subfamily 1 group I member 2 Homo sapiens 33-36 31455676-4 2019 Oral administration of the human PXR activator rifampicin increased hepatic expression of CYP3A4 mRNA and triazolam (TRZ) 1"- and 4-hydroxylation activities, CYP3A probe activities, in the liver and intestine microsomes of hCYP3A-MAC/hPXR mice. Rifampin 47-57 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-96 31455676-4 2019 Oral administration of the human PXR activator rifampicin increased hepatic expression of CYP3A4 mRNA and triazolam (TRZ) 1"- and 4-hydroxylation activities, CYP3A probe activities, in the liver and intestine microsomes of hCYP3A-MAC/hPXR mice. Rifampin 47-57 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-95 31455676-4 2019 Oral administration of the human PXR activator rifampicin increased hepatic expression of CYP3A4 mRNA and triazolam (TRZ) 1"- and 4-hydroxylation activities, CYP3A probe activities, in the liver and intestine microsomes of hCYP3A-MAC/hPXR mice. Rifampin 47-57 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 223-229 31455676-5 2019 The plasma concentration of TRZ after oral dosing was significantly decreased by rifampicin treatment in hCYP3A-MAC/hPXR mice but not in hCYP3A-MAC mice. Rifampin 81-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-111 31455676-6 2019 In addition, mass spectrometry imaging analysis showed that rifampicin treatment increased the formation of hydroxy TRZ in the intestine of hCYP3A-MAC/hPXR mice after oral dosing of TRZ. Rifampin 60-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 140-146 31455676-7 2019 The plasma concentration of 1"- and 4-hydroxy TRZ in portal blood was also increased by rifampicin treatment in hCYP3A-MAC/hPXR mice. Rifampin 88-98 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 112-118 31445882-0 2019 MicroRNAs hsa-miR-495-3p and hsa-miR-486-5p suppress basal and rifampicin-induced expression of human sulfotransferase 2A1 (SULT2A1) by facilitating mRNA degradation. Rifampin 63-73 sulfotransferase family 2A member 1 Homo sapiens 102-122 31445882-0 2019 MicroRNAs hsa-miR-495-3p and hsa-miR-486-5p suppress basal and rifampicin-induced expression of human sulfotransferase 2A1 (SULT2A1) by facilitating mRNA degradation. Rifampin 63-73 sulfotransferase family 2A member 1 Homo sapiens 124-131 31445882-8 2019 Furthermore, gain- and loss-of-function assays demonstrated that hsa-miR-486-5p and hsa-miR-495-3p negatively modulate basal and rifampicin-induced expression of SULT2A1 in HepG2 cells by decreasing mRNA stability. Rifampin 129-139 sulfotransferase family 2A, dehydroepiandrosterone (DHEA)-preferring, member 1 Mus musculus 162-169 31090092-7 2019 5-HMT fractional clearance by CYP3A and CYP2D6 was verified from clinical DDI studies with a potent CYP3A4 inhibitor (ketoconazole) and inducer (rifampicin) in CYP2D6 extensive and poor metabolizers and with a moderate CYP3A inhibitor (fluconazole) in healthy volunteers. Rifampin 145-155 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 30-35 31090092-7 2019 5-HMT fractional clearance by CYP3A and CYP2D6 was verified from clinical DDI studies with a potent CYP3A4 inhibitor (ketoconazole) and inducer (rifampicin) in CYP2D6 extensive and poor metabolizers and with a moderate CYP3A inhibitor (fluconazole) in healthy volunteers. Rifampin 145-155 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 40-46 31090092-7 2019 5-HMT fractional clearance by CYP3A and CYP2D6 was verified from clinical DDI studies with a potent CYP3A4 inhibitor (ketoconazole) and inducer (rifampicin) in CYP2D6 extensive and poor metabolizers and with a moderate CYP3A inhibitor (fluconazole) in healthy volunteers. Rifampin 145-155 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 160-166 31455676-11 2019 Expression of CYP3A4 and metabolism of triazolam, a typical CYP3A substrate, in the liver of CYP3A/PXR-humanized mice were enhanced in response to rifampicin, a typical human PXR activator. Rifampin 147-157 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-20 31455676-11 2019 Expression of CYP3A4 and metabolism of triazolam, a typical CYP3A substrate, in the liver of CYP3A/PXR-humanized mice were enhanced in response to rifampicin, a typical human PXR activator. Rifampin 147-157 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 14-19 31455676-11 2019 Expression of CYP3A4 and metabolism of triazolam, a typical CYP3A substrate, in the liver of CYP3A/PXR-humanized mice were enhanced in response to rifampicin, a typical human PXR activator. Rifampin 147-157 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 60-65 31455676-11 2019 Expression of CYP3A4 and metabolism of triazolam, a typical CYP3A substrate, in the liver of CYP3A/PXR-humanized mice were enhanced in response to rifampicin, a typical human PXR activator. Rifampin 147-157 nuclear receptor subfamily 1, group I, member 2 Mus musculus 99-102 31455676-11 2019 Expression of CYP3A4 and metabolism of triazolam, a typical CYP3A substrate, in the liver of CYP3A/PXR-humanized mice were enhanced in response to rifampicin, a typical human PXR activator. Rifampin 147-157 nuclear receptor subfamily 1 group I member 2 Homo sapiens 175-178 31676845-7 2019 We also demonstrated that matured Huh7s can be used for drug induction studies, where CYP3A4, CYP1A2, CYP2C9, and CYP2C19 inductions were achieved following rifampicin treatment. Rifampin 157-167 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 94-100 31676845-7 2019 We also demonstrated that matured Huh7s can be used for drug induction studies, where CYP3A4, CYP1A2, CYP2C9, and CYP2C19 inductions were achieved following rifampicin treatment. Rifampin 157-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 31676845-7 2019 We also demonstrated that matured Huh7s can be used for drug induction studies, where CYP3A4, CYP1A2, CYP2C9, and CYP2C19 inductions were achieved following rifampicin treatment. Rifampin 157-167 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 114-121 31581194-6 2019 PXR, activated by rifampicin, is rate-limiting for mucosal NF-kappaB activation in IBD. Rifampin 18-28 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-3 31581123-8 2019 The tuberculostatic drug rifampicin is a CYP3A4 inducer, increasing the metabolism of hydrocortisone. Rifampin 25-35 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-47 31581194-6 2019 PXR, activated by rifampicin, is rate-limiting for mucosal NF-kappaB activation in IBD. Rifampin 18-28 nuclear factor kappa B subunit 1 Homo sapiens 59-68 30991038-0 2019 Rifampin Induces Expression of P-glycoprotein on the THP1 Cell-Derived Macrophages, Causing Decrease Intramacrophage Concentration of Prothionamide. Rifampin 0-8 ATP binding cassette subfamily B member 1 Homo sapiens 31-45 31526233-6 2019 On the other hand, treatment of HepaRG cells with RMP resulted in a significant increase in mRNA expression for CYP2B6 (6.68-fold, p = 0.007) and CYP3A4 (111.96-fold, p = 0.001), whereas NR1I3 expression decreased (0.46-fold, p = 0.033). Rifampin 50-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-118 31526233-6 2019 On the other hand, treatment of HepaRG cells with RMP resulted in a significant increase in mRNA expression for CYP2B6 (6.68-fold, p = 0.007) and CYP3A4 (111.96-fold, p = 0.001), whereas NR1I3 expression decreased (0.46-fold, p = 0.033). Rifampin 50-53 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 146-152 31526233-6 2019 On the other hand, treatment of HepaRG cells with RMP resulted in a significant increase in mRNA expression for CYP2B6 (6.68-fold, p = 0.007) and CYP3A4 (111.96-fold, p = 0.001), whereas NR1I3 expression decreased (0.46-fold, p = 0.033). Rifampin 50-53 nuclear receptor subfamily 1 group I member 3 Homo sapiens 187-192 30759187-0 2019 Prevalence, Predictors, and Successful Treatment Outcomes of Xpert MTB/RIF-identified Rifampicin-resistant Tuberculosis in Post-conflict Eastern Democratic Republic of the Congo, 2012-2017: A Retrospective Province-Wide Cohort Study. Rifampin 86-96 metallothionein 1J, pseudogene Homo sapiens 67-70 31361500-3 2019 Rifampin significantly induced the transcription of ABCB1 and ABCC2 and protein abundance of P-gp but not of MRP2. Rifampin 0-8 ATP binding cassette subfamily B member 1 Homo sapiens 52-57 31361500-3 2019 Rifampin significantly induced the transcription of ABCB1 and ABCC2 and protein abundance of P-gp but not of MRP2. Rifampin 0-8 ATP binding cassette subfamily C member 2 Homo sapiens 62-67 31361500-3 2019 Rifampin significantly induced the transcription of ABCB1 and ABCC2 and protein abundance of P-gp but not of MRP2. Rifampin 0-8 ATP binding cassette subfamily B member 1 Homo sapiens 93-97 31361500-3 2019 Rifampin significantly induced the transcription of ABCB1 and ABCC2 and protein abundance of P-gp but not of MRP2. Rifampin 0-8 ATP binding cassette subfamily C member 2 Homo sapiens 109-113 31119275-11 2019 Rifampicin co-administration showed the largest impact on CL/F. Rifampin 0-10 crooked neck pre-mRNA splicing factor 1 Homo sapiens 58-62 31569378-4 2019 Validation of RGs was performed by an expression analysis of CYP3A4 and CYP1A2 on rifampicin and beta-naphthoflavone induction, respectively. Rifampin 82-92 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 61-67 31569378-4 2019 Validation of RGs was performed by an expression analysis of CYP3A4 and CYP1A2 on rifampicin and beta-naphthoflavone induction, respectively. Rifampin 82-92 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 72-78 31527775-6 2019 Uptake of NP-59 into HEK293 cells expressing organic anion transporting polypeptide (OATP)1B1 and OATP1B3 was significantly higher than that into mock cells and was inhibited by rifampicin. Rifampin 178-188 solute carrier organic anion transporter family member 1B1 Homo sapiens 85-93 31527775-6 2019 Uptake of NP-59 into HEK293 cells expressing organic anion transporting polypeptide (OATP)1B1 and OATP1B3 was significantly higher than that into mock cells and was inhibited by rifampicin. Rifampin 178-188 solute carrier organic anion transporter family member 1B3 Homo sapiens 98-105 31361500-0 2019 Transcriptional and Post-Transcriptional Regulation of Duodenal P-Glycoprotein and MRP2 in Healthy Human Subjects after Chronic Treatment with Rifampin and Carbamazepine. Rifampin 143-151 ATP binding cassette subfamily B member 1 Homo sapiens 64-78 31361500-0 2019 Transcriptional and Post-Transcriptional Regulation of Duodenal P-Glycoprotein and MRP2 in Healthy Human Subjects after Chronic Treatment with Rifampin and Carbamazepine. Rifampin 143-151 ATP binding cassette subfamily C member 2 Homo sapiens 83-87 31420942-0 2019 Physiologically-Based Pharmacokinetic Modeling Approach to Predict Rifampin-Mediated Intestinal P-Glycoprotein Induction. Rifampin 67-75 ATP binding cassette subfamily B member 1 Homo sapiens 96-110 31420942-2 2019 This study aimed to predict effects of rifampin-mediated intestinal P-glycoprotein (Pgp) induction on pharmacokinetics of Pgp substrates via PBPK modeling. Rifampin 39-47 ATP binding cassette subfamily B member 1 Homo sapiens 68-82 31420942-2 2019 This study aimed to predict effects of rifampin-mediated intestinal P-glycoprotein (Pgp) induction on pharmacokinetics of Pgp substrates via PBPK modeling. Rifampin 39-47 ATP binding cassette subfamily B member 1 Homo sapiens 84-87 31420942-2 2019 This study aimed to predict effects of rifampin-mediated intestinal P-glycoprotein (Pgp) induction on pharmacokinetics of Pgp substrates via PBPK modeling. Rifampin 39-47 ATP binding cassette subfamily B member 1 Homo sapiens 122-125 31420942-5 2019 Next, we predicted rifampin-mediated fold increases in intestinal Pgp abundances to reasonably recover clinically observed DDI results. Rifampin 19-27 ATP binding cassette subfamily B member 1 Homo sapiens 66-69 31420942-6 2019 The modeling results suggested that threefold to fourfold increases in intestinal Pgp abundances could sufficiently reproduce the DDI results of these Pgp substrates with rifampin. Rifampin 171-179 ATP binding cassette subfamily B member 1 Homo sapiens 82-85 31420942-6 2019 The modeling results suggested that threefold to fourfold increases in intestinal Pgp abundances could sufficiently reproduce the DDI results of these Pgp substrates with rifampin. Rifampin 171-179 ATP binding cassette subfamily B member 1 Homo sapiens 151-154 30991038-9 2019 Further studies would be necessary to know the influence of RIF-induced P-gp induction on the treatment outcome of patients with TB. Rifampin 60-63 phosphoglycolate phosphatase Homo sapiens 72-76 31656654-12 2019 Conclusions: Coadministration of rifampicin and clarithromycin may increase CYP3A enzymatic activity. Rifampin 33-43 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 76-81 30991038-0 2019 Rifampin Induces Expression of P-glycoprotein on the THP1 Cell-Derived Macrophages, Causing Decrease Intramacrophage Concentration of Prothionamide. Rifampin 0-8 GLI family zinc finger 2 Homo sapiens 53-57 30991038-3 2019 The present study determined the effect of RIF on MDR1 gene (P-glycoprotein, P-gp) expression in THP1 macrophages and analyzed the intracellular concentration of the anti-TB drug prothionamide in the presence of RIF. Rifampin 43-46 ATP binding cassette subfamily B member 1 Homo sapiens 50-54 30991038-3 2019 The present study determined the effect of RIF on MDR1 gene (P-glycoprotein, P-gp) expression in THP1 macrophages and analyzed the intracellular concentration of the anti-TB drug prothionamide in the presence of RIF. Rifampin 43-46 ATP binding cassette subfamily B member 1 Homo sapiens 61-75 31656654-0 2019 Effect of rifampicin and clarithromycin on the CYP3A activity in patients with Mycobacterium avium complex. Rifampin 10-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-52 31656654-3 2019 The cytochrome P450 (CYP) enzyme inducer, rifampicin, and the CYP inhibitor, clarithromycin, have clinical activity against MAC and key drugs in the treatment of MAC infection. Rifampin 42-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 30991038-3 2019 The present study determined the effect of RIF on MDR1 gene (P-glycoprotein, P-gp) expression in THP1 macrophages and analyzed the intracellular concentration of the anti-TB drug prothionamide in the presence of RIF. Rifampin 43-46 phosphoglycolate phosphatase Homo sapiens 77-81 30991038-3 2019 The present study determined the effect of RIF on MDR1 gene (P-glycoprotein, P-gp) expression in THP1 macrophages and analyzed the intracellular concentration of the anti-TB drug prothionamide in the presence of RIF. Rifampin 43-46 GLI family zinc finger 2 Homo sapiens 97-101 31404338-0 2019 Xpert MTB/RIF performance to diagnose tuberculosis and rifampicin resistance in a reference centre in southern Brazil. Rifampin 55-65 ras homolog family member F, filopodia associated Homo sapiens 10-13 31382894-0 2019 Xpert MTB/RIF assay for the diagnosis of rifampicin resistance in different regions: a meta-analysis. Rifampin 41-51 ras homolog family member F, filopodia associated Homo sapiens 10-13 31382894-1 2019 BACKGROUND: To estimate the diagnostic accuracy of Xpert MTB/RIF for rifampicin resistance in different regions, a meta-analysis was carried out. Rifampin 69-79 ras homolog family member F, filopodia associated Homo sapiens 61-64 31382894-5 2019 The pooled sensitivity, specificity and AUC of Xpert MTB/RIF for rifampicin resistance detection were 0.93 (95% CI 0.90-0.95), 0.98 (95% CI 0.96-0.98) and 0.99 (95% CI 0.97-0.99), respectively. Rifampin 65-75 ras homolog family member F, filopodia associated Homo sapiens 57-60 31382894-8 2019 CONCLUSIONS: The diagnostic accuracy of Xpert MTB/RIF for rifampicin resistance detection was excellent. Rifampin 58-68 ras homolog family member F, filopodia associated Homo sapiens 50-53 31404338-2 2019 Xpert MTB/RIF has been implemented in many countries to reduce the time to TB diagnosis and to rapidly detect rifampicin resistance. Rifampin 110-120 metallothionein 1J, pseudogene Homo sapiens 6-9 31404338-2 2019 Xpert MTB/RIF has been implemented in many countries to reduce the time to TB diagnosis and to rapidly detect rifampicin resistance. Rifampin 110-120 ras homolog family member F, filopodia associated Homo sapiens 10-13 31404338-3 2019 The study aimed to describe and evaluate Xpert MTB/RIF performance in diagnosing pulmonary TB and rifampicin resistance in a tertiary healthcare facility in Brazil. Rifampin 98-108 metallothionein 1J, pseudogene Homo sapiens 47-50 31404338-3 2019 The study aimed to describe and evaluate Xpert MTB/RIF performance in diagnosing pulmonary TB and rifampicin resistance in a tertiary healthcare facility in Brazil. Rifampin 98-108 ras homolog family member F, filopodia associated Homo sapiens 51-54 31349736-0 2019 Rifampicin and Its Derivative Rifampicin Quinone Reduce Microglial Inflammatory Responses and Neurodegeneration Induced In Vitro by alpha-Synuclein Fibrillary Aggregates. Rifampin 0-10 synuclein alpha Homo sapiens 132-147 30570831-3 2019 This phase 1, open-label, multicenter study evaluated the effect of rifampicin, a CYP3A4 inducer, on the pharmacokinetics of single-dose vemurafenib in 27 patients with BRAFV600 mutation-positive metastatic malignancy. Rifampin 68-78 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 82-88 30583052-10 2019 Xpert MTB/RIF in addition detected rifampicin resistance in 13 patients, in perfect agreement with results from the line probe assay. Rifampin 35-45 ras homolog family member F, filopodia associated Homo sapiens 10-13 31349736-3 2019 Pretreatments with Rif and RifQ reduced the secretion of prototypical inflammatory cytokines (TNF- , IL-6) and the burst of oxidative stress in microglial cells activated with alphaS fibrillary aggregates. Rifampin 19-22 tumor necrosis factor Homo sapiens 94-97 31349736-3 2019 Pretreatments with Rif and RifQ reduced the secretion of prototypical inflammatory cytokines (TNF- , IL-6) and the burst of oxidative stress in microglial cells activated with alphaS fibrillary aggregates. Rifampin 19-22 interleukin 6 Homo sapiens 101-105 31332230-8 2019 Treatment of spheroids with rifampicin increased CYP3A4 activity, demonstrating the metabolic capacity of HepaRG spheroids. Rifampin 28-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 49-55 31262885-6 2019 Rifampicin, a PXR agonist, reduced the accumulation of CDDP and suppressed growth inhibition and caspase-3 activation in HepG2 after CDDP exposure. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 14-17 30903956-8 2019 The developed sensor possessed a high sensitivity toward rifampicin with a detection limit of 0.16 muM and excellent specificity, as compared to rifabutin and rifapentine. Rifampin 57-67 latexin Homo sapiens 99-102 31334131-6 2019 This compound also synergized with rifampicin and azithromycin at sub-micromolar concentrations (0.25-0.5 muM), thereby inducing susceptibility to these antibiotics at clinically relevant concentrations in clinical MDR isolates. Rifampin 35-45 latexin Homo sapiens 106-109 31262885-6 2019 Rifampicin, a PXR agonist, reduced the accumulation of CDDP and suppressed growth inhibition and caspase-3 activation in HepG2 after CDDP exposure. Rifampin 0-10 caspase 3 Homo sapiens 97-106 31260434-10 2019 Polymyxins are useful in the treatment of patients infected with Klebsiella pneumoniae NDM (New Delhi metallo-beta-lactamase), where the effectiveness of therapy is greater when colistin is used together with carbapenem or rifampicin or tigecycline. Rifampin 223-233 NDM-1 Klebsiella pneumoniae 92-124 30975793-4 2019 Data implied rifamycin SV (20 microM) inhibits three OATPs, while rifampicin (5 microM) inhibits OATP1B1/1B3 only. Rifampin 66-76 solute carrier organic anion transporter family member 1B1 Homo sapiens 97-108 31032736-0 2019 Peppermint (Mentha piperita L.) extract effectively inhibits cytochrome P450 3A4 (CYP3A4) mRNA induction in rifampicin-treated HepG2 cells. Rifampin 108-118 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 82-88 31032736-2 2019 Therefore, here, we examined the suppressive effects of the extracts from seven edible herbs on the induction of CYP3A4 gene expression in rifampicin-treated HepG2 cells. Rifampin 139-149 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 113-119 31032736-5 2019 It suppressed the induction of CYP3A4 mRNA expression by rifampicin in a dose-dependent manner. Rifampin 57-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 31032736-7 2019 These findings demonstrate that pheophorbide a suppresses the induction of CYP3A4 mRNA expression in rifampicin-treated HepG2 cells. Rifampin 101-111 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 75-81 30907428-0 2019 KITTEN following CAT on the long-term use of rifampicin in hidradenitis suppurativa and effectiveness of oral contraceptives. Rifampin 45-55 catalase Homo sapiens 17-20 31173647-19 2019 AUTHORS" CONCLUSIONS: We found Xpert MTB/RIF to be sensitive and specific for diagnosing PTB and rifampicin resistance, consistent with findings reported previously. Rifampin 97-107 ras homolog family member F, filopodia associated Homo sapiens 41-44 31218462-0 2019 Guidance for Rifampin and Midazolam Dosing Protocols To Study Intestinal and Hepatic Cytochrome P450 (CYP) 3A4 Induction and De-induction. Rifampin 13-21 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 85-110 31218462-3 2019 To collate published data regarding induction of CYP3A4 expression by rifampin and identify an optimal protocol to study DDIs using physiologically based pharmacokinetic (PBPK) modelling. Rifampin 70-78 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 49-55 31218462-4 2019 The University of Washington Drug Interaction Database was searched for published data regarding induction of CYP3A4 by rifampin. Rifampin 120-128 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 110-116 31218462-5 2019 A verified PBPK model was used to define the optimal dose, duration, timing and route of administration of rifampin and midazolam to assess induction of intestinal and hepatic CYP3A4 by rifampin. Rifampin 186-194 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 176-182 31218462-9 2019 Intestinal CYP3A4 expression returned to baseline in > 90% of participants within 7 days of rifampin cessation, whereas induction of hepatic CYP3A4 persisted for greater than 7 days in > 50% of participants. Rifampin 92-100 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 11-17 31047967-7 2019 Contribution of both CYP3A and P-GP induction in intestine and liver by rifampicin to pharmacokinetic profiles of victim drugs was investigated. Rifampin 72-82 phosphoglycolate phosphatase Homo sapiens 31-35 31047967-11 2019 DDIs of rifampicin with CYP3A or P-GP substrates following oral versus intravenous administration to human were successfully predicted using the developed semi-PBPK model. Rifampin 8-18 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-29 31047967-11 2019 DDIs of rifampicin with CYP3A or P-GP substrates following oral versus intravenous administration to human were successfully predicted using the developed semi-PBPK model. Rifampin 8-18 phosphoglycolate phosphatase Homo sapiens 33-37 31047967-0 2019 Simultaneously predict pharmacokinetic interaction of rifampicin with oral versus intravenous substrates of cytochrome P450 3A/P-glycoprotein to healthy human using a semi-physiologically based pharmacokinetic model involving both enzyme and transporter turnover. Rifampin 54-64 ATP binding cassette subfamily B member 1 Homo sapiens 127-141 31047967-2 2019 We aimed to establish a semi-physiologically based pharmacokinetic (semi-PBPK) model involving both enzyme and transporter turnover to simultaneously predict pharmacokinetic interaction of rifampicin with oral versus intravenous substrates of cytochrome P450 (CYP) 3A4/P-glycoprotein (P-GP) in human. Rifampin 189-199 ATP binding cassette subfamily B member 1 Homo sapiens 269-283 31047967-2 2019 We aimed to establish a semi-physiologically based pharmacokinetic (semi-PBPK) model involving both enzyme and transporter turnover to simultaneously predict pharmacokinetic interaction of rifampicin with oral versus intravenous substrates of cytochrome P450 (CYP) 3A4/P-glycoprotein (P-GP) in human. Rifampin 189-199 phosphoglycolate phosphatase Homo sapiens 285-289 31047967-3 2019 Rifampicin was chosen as the CYP3A /P-GP inducer. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-34 31047967-3 2019 Rifampicin was chosen as the CYP3A /P-GP inducer. Rifampin 0-10 phosphoglycolate phosphatase Homo sapiens 36-40 30964656-11 2019 Baicalein, curcumin, and rifampicin showed concentration-dependent inhibition of the alpha-synuclein aggregation and the IC50 (the concentration of the compound at which the maxiumum intensity was reduced by one-half) were calculated. Rifampin 25-35 synuclein alpha Homo sapiens 85-100 31122209-13 2019 The number of live colonies colonized by the two mutants in the duckling brain and trachea were lower than that of the parental strain within 24 h. CONCLUSIONS: Mutations of rpoB gene in R. anatipestifer mediate rifampin resistance and result in fitness costs. Rifampin 212-220 DNA-directed RNA polymerase subunit beta Riemerella anatipestifer ATCC 11845 = DSM 15868 174-178 31005730-7 2019 Including the blaNDM-1 gene, in total 13 resistance genes were identified in E. cloacae EC32 conferring resistance to beta-lactams, rifampicin, phenicols, fosfomycin, macrolides, quinolones, sulfonamides and tetracycline. Rifampin 132-142 metallo-beta-lactamase NDM-1 Enterobacter cloacae 14-22 31145690-7 2019 The estimated AUC ratios of ruxolitinib by co-administration of the moderate CYP3A4 inhibitor erythromycin (500 mg) and the strong CYP3A4 inducer rifampicin (600 mg) were within a 20% error compared with the clinically observed values. Rifampin 146-156 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 131-137 30888624-7 2019 After a 14-day washout, 600 mg of rifampin (rifampicin), a strong CYP3A4 inducer, as well as an inducer of UGT enzymes and a weak inducer of CYP1A2, was administered once daily on days 15-21. Rifampin 34-42 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 66-72 30748026-3 2019 Multiple studies in humans have shown that 4beta-OHC can qualitatively differentiate among weak, moderate, and potent CYP3A induction when an inducer, typically rifampin, is administered for up to 2 weeks. Rifampin 161-169 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 118-123 30762305-0 2019 Physiologically-Based Pharmacokinetic Models for CYP1A2 Drug-Drug Interaction Prediction: A Modeling Network of Fluvoxamine, Theophylline, Caffeine, Rifampicin, and Midazolam. Rifampin 149-159 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 49-55 30755272-4 2019 A recent rifampicin exposure 1 week earlier had resulted in a massive rise of CRP levels without organ manifestations. Rifampin 9-19 C-reactive protein Homo sapiens 78-81 31206055-2 2019 As a result of the Global Fund, some facilities in the country have access to improved diagnostics, including Xpert MTB/RIF testing, of particular use in diagnosing those at risk of drug resistance, in the form of rifampicin-resistant (RR) TB. Rifampin 214-224 ras homolog family member F, filopodia associated Homo sapiens 120-123 30944818-0 2019 Mitochondrial Damage and Drp1 Overexpression in Rifampicin- and Isoniazid-induced Liver Injury Cell Model. Rifampin 48-58 dynamin 1 like Homo sapiens 25-29 30682622-10 2019 Among tested xenobiotics tributyltin chloride and rifampicin non-enzymatically bound Gstr1 enzyme (the calculated Ki values are 0.26 muM and 65 muM, respectively) and inhibited its activity, showing that these compounds are reversible noncompetitive inhibitors of zebrafish Gstr1. Rifampin 50-60 glutathione S-transferase rho Danio rerio 85-90 30682622-10 2019 Among tested xenobiotics tributyltin chloride and rifampicin non-enzymatically bound Gstr1 enzyme (the calculated Ki values are 0.26 muM and 65 muM, respectively) and inhibited its activity, showing that these compounds are reversible noncompetitive inhibitors of zebrafish Gstr1. Rifampin 50-60 glutathione S-transferase rho Danio rerio 274-279 30602592-7 2019 Moreover, the rifampicin-induced expression of P450s was also affected by HNF1alpha and HNF1alpha-AS1. Rifampin 14-24 HNF1 homeobox A Homo sapiens 74-83 30602592-7 2019 Moreover, the rifampicin-induced expression of P450s was also affected by HNF1alpha and HNF1alpha-AS1. Rifampin 14-24 HNF1 homeobox A Homo sapiens 88-97 30602592-7 2019 Moreover, the rifampicin-induced expression of P450s was also affected by HNF1alpha and HNF1alpha-AS1. Rifampin 14-24 prostaglandin D2 receptor Homo sapiens 98-101 30622215-0 2019 Belinostat, at Its Clinically Relevant Concentrations, Inhibits Rifampicin-Induced CYP3A4 and MDR1 Gene Expression. Rifampin 64-74 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-89 30622215-0 2019 Belinostat, at Its Clinically Relevant Concentrations, Inhibits Rifampicin-Induced CYP3A4 and MDR1 Gene Expression. Rifampin 64-74 ATP binding cassette subfamily B member 1 Homo sapiens 94-98 30622215-8 2019 BEL repressed rifampicin-induced gene expression of CYP3A4 and multidrug resistance protein 1, as well as their respective protein activities. Rifampin 14-24 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 52-58 30622215-8 2019 BEL repressed rifampicin-induced gene expression of CYP3A4 and multidrug resistance protein 1, as well as their respective protein activities. Rifampin 14-24 ATP binding cassette subfamily B member 1 Homo sapiens 63-93 31039134-3 2019 Both rifampicin and efavirenz are activators of the pregnane X receptor (PXR), a transcription factor with significant inter-species differences in ligand-dependent activation. Rifampin 5-15 nuclear receptor subfamily 1 group I member 2 Homo sapiens 52-71 31039134-3 2019 Both rifampicin and efavirenz are activators of the pregnane X receptor (PXR), a transcription factor with significant inter-species differences in ligand-dependent activation. Rifampin 5-15 nuclear receptor subfamily 1 group I member 2 Homo sapiens 73-76 30987227-11 2019 When MA5 was screened for antibiotic resistance, broad resistance against penicillin, streptomycin, tetracycline, ampicillin, rifampicin, and erythromycin was found; this correlated with the presence of multiple gene determinants for antibiotic resistance within the whole genome sequence of MA5. Rifampin 126-136 PNMA family member 3 Homo sapiens 5-8 30902567-7 2019 Rifampicin increased omeprazole metabolism in all subjects irrespective of genotype, which suggested that CYP2C19 enzymatic activity was induced by rifampicin administration for all genotypes. Rifampin 0-10 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 106-113 30902567-7 2019 Rifampicin increased omeprazole metabolism in all subjects irrespective of genotype, which suggested that CYP2C19 enzymatic activity was induced by rifampicin administration for all genotypes. Rifampin 148-158 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 106-113 31084692-1 2019 BACKGROUND: Xpert MTB/RIF assay rapidly diagnoses rifampicin resistance, enabling early initiation of second-line tuberculosis (TB) treatment. Rifampin 50-60 ras homolog family member F, filopodia associated Homo sapiens 22-25 30949526-0 2019 Surveillance of Rifampicin Resistance With GeneXpert MTB/RIF in the National Reference Laboratory for Tuberculosis at the Institut Pasteur in Bangui, 2015-2017. Rifampin 16-26 ras homolog family member F, filopodia associated Homo sapiens 57-60 30949526-2 2019 Surveillance of resistance to rifampicin with GeneXpert MTB/RIF was instituted in the National TB Reference Laboratory in 2015. Rifampin 30-40 ras homolog family member F, filopodia associated Homo sapiens 60-63 30342195-17 2019 To further investigate the regulatory mechanisms, we found that ginsenosides enhanced the rifampicin-induced pregnane X receptor (PXR) transactivation of the CYP3A4 promoter. Rifampin 90-100 nuclear receptor subfamily 1 group I member 2 Homo sapiens 109-128 30758392-9 2019 However, rifampicin-treated patients had higher CD4 counts and more frequently undetectable HIV viral load by the end of treatment (67% versus 18%, p < 0.001) compared to rifabutin-treated patients, even when only ART-experienced patients were evaluated (66,6% versus 36,3%, p = 0.039). Rifampin 9-19 CD4 molecule Homo sapiens 48-51 30496848-1 2019 INTRODUCTION: Xpert MTB/RIF is recommended for the simultaneous detection of tuberculosis (TB) and rifampicin resistance directly from sputum specimens. Rifampin 99-109 ras homolog family member F, filopodia associated Homo sapiens 24-27 32032019-0 2019 Rifampicin resistant tuberculosis in presumptive pulmonary tuberculosis cases in Dubti Hospital, Afar, Ethiopia. Rifampin 0-10 aldo-keto reductase family 7 member A2 Homo sapiens 97-101 30227201-0 2019 Inhibitory kinetics and mechanism of rifampicin on alpha-glucosidase: Insights from spectroscopic and molecular docking analyses. Rifampin 37-47 sucrase-isomaltase Homo sapiens 51-68 30227201-2 2019 In this study, the inhibitory effect and mechanism of rifampicin on alpha-glucosidase were investigated by multispectroscopic methods along with molecular docking technique. Rifampin 54-64 sucrase-isomaltase Homo sapiens 68-85 30227201-3 2019 The results showed that rifampicin inhibited alpha-glucosidase activity prominently (IC50 = 135 +- 1.2 muM) in a reversible and competitive-type manner. Rifampin 24-34 sucrase-isomaltase Homo sapiens 45-62 30227201-4 2019 The fluorescence intensity of alpha-glucosidase was quenched by rifampicin through forming rifampicin-alpha-glucosidase complex in a static procedure. Rifampin 64-74 sucrase-isomaltase Homo sapiens 30-47 30227201-4 2019 The fluorescence intensity of alpha-glucosidase was quenched by rifampicin through forming rifampicin-alpha-glucosidase complex in a static procedure. Rifampin 64-74 sucrase-isomaltase Homo sapiens 102-119 30227201-7 2019 Moreover, it was found that the binding of rifampicin to alpha-glucosidase could alter the conformation of the enzyme to make it steady, and the binding distance was estimated to be 1.02 nm. Rifampin 43-53 sucrase-isomaltase Homo sapiens 57-74 29405807-8 2019 Atorvastatin and rifampicin could inhibit the uptake of deoxyschizandrin and schizandrin B mediated by OATP1B1. Rifampin 17-27 solute carrier organic anion transporter family member 1B1 Homo sapiens 103-110 30342195-17 2019 To further investigate the regulatory mechanisms, we found that ginsenosides enhanced the rifampicin-induced pregnane X receptor (PXR) transactivation of the CYP3A4 promoter. Rifampin 90-100 nuclear receptor subfamily 1 group I member 2 Homo sapiens 130-133 30342195-17 2019 To further investigate the regulatory mechanisms, we found that ginsenosides enhanced the rifampicin-induced pregnane X receptor (PXR) transactivation of the CYP3A4 promoter. Rifampin 90-100 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 158-164 30342195-18 2019 Treatment of hPXR-over-expressed cells with ginsenosides increased the rifampicin-inducible expression of CYP3A4 in a concentration-dependent manner. Rifampin 71-81 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 106-112 30555014-6 2019 RESULTS: Intoxication with RIF and INH markedly reduced the hematological indices and elevated the biochemical enzyme markers (AST, ALT and ALP, P < 0.001) and lipid profile (P < 0.001), while antioxidant biomarkers were significantly (P < 0.01) depressed and MDA was elevated. Rifampin 27-30 glutamic-oxaloacetic transaminase 2 Rattus norvegicus 127-130 30503582-7 2019 In addition, treatment with 3-methylcholanthrene, phenobarbital, and rifampin led to the induction of cytochrome P-450 (cyp) 1a1 and cyp1a2, cyp2b10, cyp3a11. Rifampin 69-77 cytochrome P450, family 1, subfamily a, polypeptide 1 Mus musculus 102-128 30503582-7 2019 In addition, treatment with 3-methylcholanthrene, phenobarbital, and rifampin led to the induction of cytochrome P-450 (cyp) 1a1 and cyp1a2, cyp2b10, cyp3a11. Rifampin 69-77 cytochrome P450, family 1, subfamily a, polypeptide 2 Mus musculus 133-139 30503582-7 2019 In addition, treatment with 3-methylcholanthrene, phenobarbital, and rifampin led to the induction of cytochrome P-450 (cyp) 1a1 and cyp1a2, cyp2b10, cyp3a11. Rifampin 69-77 cytochrome P450, family 2, subfamily b, polypeptide 10 Mus musculus 141-148 30503582-7 2019 In addition, treatment with 3-methylcholanthrene, phenobarbital, and rifampin led to the induction of cytochrome P-450 (cyp) 1a1 and cyp1a2, cyp2b10, cyp3a11. Rifampin 69-77 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 150-157 30340248-5 2019 Mean (95% confidence interval) exosome-derived CYP3A4 protein expression pre- and post-rifampicin dosing was 0.24 (0.2-0.28) and 0.42 (0.21-0.65) ng ml-1 exosome concentrate. Rifampin 87-97 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-53 30340248-6 2019 Mean (95% confidence interval) exosome CYP3A4 mRNA expression pre- and post-rifampicin dosing was 6.0 (1.1-32.7) and 48.3 (11.3-104) x 10-11 2-DeltaDeltaCt , respectively. Rifampin 76-86 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 39-45 30804707-5 2019 The protective roles of rifaximin and rifampicin partly depend on promoting pregnane X receptor (PXR) expression. Rifampin 38-48 nuclear receptor subfamily 1 group I member 2 Homo sapiens 76-95 30804707-5 2019 The protective roles of rifaximin and rifampicin partly depend on promoting pregnane X receptor (PXR) expression. Rifampin 38-48 nuclear receptor subfamily 1 group I member 2 Homo sapiens 97-100 31366871-0 2019 Changes in Bile Acid Concentrations after Administration of Ketoconazole or Rifampicin to Chimeric Mice with Humanized Liver. Rifampin 76-86 glucosidase beta 2 Mus musculus 11-20 31366871-5 2019 Administration of a single dose of ketoconazole or rifampicin, known to potentially cause cholestasis by inhibiting BSEP, did not result in elevated levels of alkaline phosphatase (ALP), which are known hepatic biomarkers. Rifampin 51-61 ATP-binding cassette, sub-family B (MDR/TAP), member 11 Mus musculus 116-120 31366871-8 2019 The mRNA levels of bile acid-related genes were also altered after treatment with ketoconazole or rifampicin. Rifampin 98-108 glucosidase beta 2 Mus musculus 19-28 31228606-8 2019 Moreover, the drug-drug interaction and drug-food interaction could be observed by using our human iPS-IECM in the presence of an inducer and inhibitor of CYP3A4, i.e., rifampicin and grape fruit juice, respectively. Rifampin 169-179 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 155-161 30539784-8 2019 In line with this, rifampicin decreased the number of apoptotic cells in the corpus callosum thereby diminishing the expression of cleaved caspase-3 and Bax, as well as increasing Bcl-2. Rifampin 19-29 BCL2-associated X protein Mus musculus 153-156 30539784-8 2019 In line with this, rifampicin decreased the number of apoptotic cells in the corpus callosum thereby diminishing the expression of cleaved caspase-3 and Bax, as well as increasing Bcl-2. Rifampin 19-29 B cell leukemia/lymphoma 2 Mus musculus 180-185 30539784-9 2019 Moreover, rifampicin significantly lowered the levels of interleukin-6, interleukin-1beta, caspase-12 activity, heme oxygenase-1(HO-1), nitric oxide (NO), and malondialdehyde (MDA) in mice treated with cuprizone. Rifampin 10-20 interleukin 6 Mus musculus 57-70 30539784-9 2019 Moreover, rifampicin significantly lowered the levels of interleukin-6, interleukin-1beta, caspase-12 activity, heme oxygenase-1(HO-1), nitric oxide (NO), and malondialdehyde (MDA) in mice treated with cuprizone. Rifampin 10-20 interleukin 1 beta Mus musculus 72-89 30539784-9 2019 Moreover, rifampicin significantly lowered the levels of interleukin-6, interleukin-1beta, caspase-12 activity, heme oxygenase-1(HO-1), nitric oxide (NO), and malondialdehyde (MDA) in mice treated with cuprizone. Rifampin 10-20 heme oxygenase 1 Mus musculus 112-128 30539784-10 2019 Conversely, the activity of glutathione peroxidase (GPx) and the level of ferric reducing ability of plasma (FRAP) were increased in response to the treatment with rifampicin. Rifampin 164-174 mechanistic target of rapamycin kinase Mus musculus 109-113 30797272-2 2019 OBJECTIVE: To determine the common mutations responsible for rifampicin resistance in TB cases detected by Xpert MTB/RIF assay. Rifampin 61-71 ras homolog family member F, filopodia associated Homo sapiens 117-120 30797272-10 2019 CONCLUSION: Xpert MTB/RIF assay uses various combinations of probe to detect MTB along with rifampicin resistance and is a valuable diagnostic tool. Rifampin 92-102 ras homolog family member F, filopodia associated Homo sapiens 22-25 30775278-1 2018 Setting: Myanmar"s National Tuberculosis Programme (NTP) uses the Xpert MTB/RIF assay to diagnose rifampicin (RMP) resistance in sputum smear-positive (Sm+) pulmonary tuberculosis (TB) patients. Rifampin 99-109 metallothionein 1J, pseudogene Homo sapiens 73-76 29503005-11 2019 As rifampin loosely binds to Dacron ESGs by weak intermolecular forces, a rifampin-coated ESG would need to be inserted in a timely fashion to treat the diseased aorta and to deliver its antibiotic affect. Rifampin 3-11 TLE family member 2, transcriptional corepressor Homo sapiens 36-39 30775278-1 2018 Setting: Myanmar"s National Tuberculosis Programme (NTP) uses the Xpert MTB/RIF assay to diagnose rifampicin (RMP) resistance in sputum smear-positive (Sm+) pulmonary tuberculosis (TB) patients. Rifampin 99-109 ras homolog family member F, filopodia associated Homo sapiens 77-80 30775278-1 2018 Setting: Myanmar"s National Tuberculosis Programme (NTP) uses the Xpert MTB/RIF assay to diagnose rifampicin (RMP) resistance in sputum smear-positive (Sm+) pulmonary tuberculosis (TB) patients. Rifampin 111-114 metallothionein 1J, pseudogene Homo sapiens 73-76 30775278-1 2018 Setting: Myanmar"s National Tuberculosis Programme (NTP) uses the Xpert MTB/RIF assay to diagnose rifampicin (RMP) resistance in sputum smear-positive (Sm+) pulmonary tuberculosis (TB) patients. Rifampin 111-114 ras homolog family member F, filopodia associated Homo sapiens 77-80 30171692-0 2018 CYP3A4-mediated effects of rifampicin on the pharmacokinetics of vilaprisan and its UGT1A1-mediated effects on bilirubin glucuronidation in humans. Rifampin 27-37 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 30325673-4 2018 This study aimed to develop a nanoemulsion (NE) containing rifampicin (RIF-NE) and evaluate its in vitro antimycobacterial activity using Resazurin Microtiter Assay against three Mycobacterium tuberculosis strains: two susceptible and a multidrug-resistant. Rifampin 59-69 alpha fetoprotein regulation 2, inducibility Mus musculus 71-74 30558670-1 2018 BACKGROUND: In 2011, South Africa improved its ability to test for rifampicin-resistant TB (RR-TB) by introducing GeneXpert MTB/RIF. Rifampin 67-77 ras homolog family member F, filopodia associated Homo sapiens 128-131 30171692-1 2018 AIMS: The primary aim of the present study was to quantify the effects of rifampicin, a strong cytochrome P450 (CYP) 3A4 inducer, on the pharmacokinetics of the new selective progesterone receptor modulator, vilaprisan. Rifampin 74-84 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 95-120 30171692-1 2018 AIMS: The primary aim of the present study was to quantify the effects of rifampicin, a strong cytochrome P450 (CYP) 3A4 inducer, on the pharmacokinetics of the new selective progesterone receptor modulator, vilaprisan. Rifampin 74-84 progesterone receptor Homo sapiens 175-196 29700814-3 2018 The MTP model predicted rifampicin biomarker response observed in 1) a hollow-fiber infection model, 2) a murine study to determine pharmacokinetic/pharmacodynamic indices, and 3) several clinical phase IIa early bactericidal activity (EBA) studies. Rifampin 24-34 ATPase, class II, type 9A Mus musculus 203-206 29569712-1 2018 Rifampin demonstrated dose-dependent relative induction between cytochrome P (CYP)3A and P-glycoprotein (P-gp), organic anion transporting polypeptides (OATPs), or CYP2C9; P-gp, OATP, and CYP2C9 induction was one drug-drug interaction (DDI) category lower than that observed for CYP3A across a wide range of pregnane X receptor (PXR) agonism. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 64-84 29569712-1 2018 Rifampin demonstrated dose-dependent relative induction between cytochrome P (CYP)3A and P-glycoprotein (P-gp), organic anion transporting polypeptides (OATPs), or CYP2C9; P-gp, OATP, and CYP2C9 induction was one drug-drug interaction (DDI) category lower than that observed for CYP3A across a wide range of pregnane X receptor (PXR) agonism. Rifampin 0-8 ATP binding cassette subfamily B member 1 Homo sapiens 89-103 29569712-1 2018 Rifampin demonstrated dose-dependent relative induction between cytochrome P (CYP)3A and P-glycoprotein (P-gp), organic anion transporting polypeptides (OATPs), or CYP2C9; P-gp, OATP, and CYP2C9 induction was one drug-drug interaction (DDI) category lower than that observed for CYP3A across a wide range of pregnane X receptor (PXR) agonism. Rifampin 0-8 ATP binding cassette subfamily B member 1 Homo sapiens 105-109 29569712-1 2018 Rifampin demonstrated dose-dependent relative induction between cytochrome P (CYP)3A and P-glycoprotein (P-gp), organic anion transporting polypeptides (OATPs), or CYP2C9; P-gp, OATP, and CYP2C9 induction was one drug-drug interaction (DDI) category lower than that observed for CYP3A across a wide range of pregnane X receptor (PXR) agonism. Rifampin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 164-170 29569712-1 2018 Rifampin demonstrated dose-dependent relative induction between cytochrome P (CYP)3A and P-glycoprotein (P-gp), organic anion transporting polypeptides (OATPs), or CYP2C9; P-gp, OATP, and CYP2C9 induction was one drug-drug interaction (DDI) category lower than that observed for CYP3A across a wide range of pregnane X receptor (PXR) agonism. Rifampin 0-8 ATP binding cassette subfamily B member 1 Homo sapiens 172-176 30340067-10 2018 The method was successfully used for the determination of 4beta-OHC and 4alpha-OHC concentrations in clinical plasma and serum samples collected before and after treatment with a known CYP3A4 inducer rifampicin. Rifampin 200-210 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 185-191 29569712-1 2018 Rifampin demonstrated dose-dependent relative induction between cytochrome P (CYP)3A and P-glycoprotein (P-gp), organic anion transporting polypeptides (OATPs), or CYP2C9; P-gp, OATP, and CYP2C9 induction was one drug-drug interaction (DDI) category lower than that observed for CYP3A across a wide range of pregnane X receptor (PXR) agonism. Rifampin 0-8 solute carrier organic anion transporter family member 1A2 Homo sapiens 153-157 29569712-1 2018 Rifampin demonstrated dose-dependent relative induction between cytochrome P (CYP)3A and P-glycoprotein (P-gp), organic anion transporting polypeptides (OATPs), or CYP2C9; P-gp, OATP, and CYP2C9 induction was one drug-drug interaction (DDI) category lower than that observed for CYP3A across a wide range of pregnane X receptor (PXR) agonism. Rifampin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 188-194 29569712-1 2018 Rifampin demonstrated dose-dependent relative induction between cytochrome P (CYP)3A and P-glycoprotein (P-gp), organic anion transporting polypeptides (OATPs), or CYP2C9; P-gp, OATP, and CYP2C9 induction was one drug-drug interaction (DDI) category lower than that observed for CYP3A across a wide range of pregnane X receptor (PXR) agonism. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 279-284 29569712-1 2018 Rifampin demonstrated dose-dependent relative induction between cytochrome P (CYP)3A and P-glycoprotein (P-gp), organic anion transporting polypeptides (OATPs), or CYP2C9; P-gp, OATP, and CYP2C9 induction was one drug-drug interaction (DDI) category lower than that observed for CYP3A across a wide range of pregnane X receptor (PXR) agonism. Rifampin 0-8 nuclear receptor subfamily 1 group I member 2 Homo sapiens 308-327 29569712-1 2018 Rifampin demonstrated dose-dependent relative induction between cytochrome P (CYP)3A and P-glycoprotein (P-gp), organic anion transporting polypeptides (OATPs), or CYP2C9; P-gp, OATP, and CYP2C9 induction was one drug-drug interaction (DDI) category lower than that observed for CYP3A across a wide range of pregnane X receptor (PXR) agonism. Rifampin 0-8 nuclear receptor subfamily 1 group I member 2 Homo sapiens 329-332 29569723-2 2018 The objective of this study was to establish relative induction relationships between CYP3A and drug transporters (P-glycoprotein (P-gp), organic anion transporting polypeptide (OATP), and breast cancer resistance protein (BCRP)) or other P450s (CYP2C9 and CYP1A2) using ascending doses of the prototypical pregnane xenobiotic receptor (PXR) agonist, rifampin, to elicit weak, moderate, and strong PXR agonism. Rifampin 351-359 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 86-91 29569723-2 2018 The objective of this study was to establish relative induction relationships between CYP3A and drug transporters (P-glycoprotein (P-gp), organic anion transporting polypeptide (OATP), and breast cancer resistance protein (BCRP)) or other P450s (CYP2C9 and CYP1A2) using ascending doses of the prototypical pregnane xenobiotic receptor (PXR) agonist, rifampin, to elicit weak, moderate, and strong PXR agonism. Rifampin 351-359 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 189-221 29569723-2 2018 The objective of this study was to establish relative induction relationships between CYP3A and drug transporters (P-glycoprotein (P-gp), organic anion transporting polypeptide (OATP), and breast cancer resistance protein (BCRP)) or other P450s (CYP2C9 and CYP1A2) using ascending doses of the prototypical pregnane xenobiotic receptor (PXR) agonist, rifampin, to elicit weak, moderate, and strong PXR agonism. Rifampin 351-359 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 223-227 30105453-1 2018 PURPOSE: To assess the potential pharmacokinetic (PK) interactions between siponimod and rifampin, a strong CYP3A4/moderate CYP2C9 inducer, in healthy subjects. Rifampin 89-97 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 108-114 30105453-1 2018 PURPOSE: To assess the potential pharmacokinetic (PK) interactions between siponimod and rifampin, a strong CYP3A4/moderate CYP2C9 inducer, in healthy subjects. Rifampin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 30105453-14 2018 CONCLUSIONS: The study findings suggest that in the presence of rifampin, a strong CYP3A4/moderate CYP2C9 inducer, siponimod showed significant decrease in Cmax,ss (45%) and AUCtau,ss (57%) in healthy subjects. Rifampin 64-72 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-89 30105453-14 2018 CONCLUSIONS: The study findings suggest that in the presence of rifampin, a strong CYP3A4/moderate CYP2C9 inducer, siponimod showed significant decrease in Cmax,ss (45%) and AUCtau,ss (57%) in healthy subjects. Rifampin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 31720410-13 2019 Three of the 31 Xpert MTB/RIF positive patients were detected as resistance to rifampicin (RR). Rifampin 79-89 ras homolog family member F, filopodia associated Homo sapiens 26-29 30178313-3 2018 Xpert MTB/RIF (Mycobacterium tuberculosis/Rifampicin) is increasingly used in many countries as the initial diagnostic test for TB. Rifampin 42-52 ras homolog family member F, filopodia associated Homo sapiens 10-13 30179260-6 2018 In the clinical setting, exposure to nilotinib was significantly reduced by the induction of CYP3A4 with rifampicin and significantly increased by the inhibition of CYP3A with ketoconazole. Rifampin 105-115 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-99 30179260-6 2018 In the clinical setting, exposure to nilotinib was significantly reduced by the induction of CYP3A4 with rifampicin and significantly increased by the inhibition of CYP3A with ketoconazole. Rifampin 105-115 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-98 30382126-7 2018 Co-treatment of HepaRG cells by the cAMP analog Sp-5,6-DCl-cBIMPS and the reference PXR agonist rifampicin reproduced the polarizing effects of FSK. Rifampin 96-106 nuclear receptor subfamily 1 group I member 2 Homo sapiens 84-87 30175555-2 2018 The in vivo inhibition constants of rifampicin used as initial input parameters for OATP1Bs (Ki,u,OATP1Bs ) and multidrug resistance-associated protein two-mediated biliary excretion were estimated as 0.23 and 0.87 muM, respectively, from previous reports. Rifampin 36-46 ornithine aminotransferase pseudogene 1 Homo sapiens 84-89 30175555-2 2018 The in vivo inhibition constants of rifampicin used as initial input parameters for OATP1Bs (Ki,u,OATP1Bs ) and multidrug resistance-associated protein two-mediated biliary excretion were estimated as 0.23 and 0.87 muM, respectively, from previous reports. Rifampin 36-46 ATP binding cassette subfamily C member 3 Homo sapiens 112-151 29095659-0 2018 Indirect activation of pregnane X receptor in the induction of hepatic CYP3A11 by high-dose rifampicin in mice. Rifampin 92-102 nuclear receptor subfamily 1, group I, member 2 Mus musculus 23-42 29095659-0 2018 Indirect activation of pregnane X receptor in the induction of hepatic CYP3A11 by high-dose rifampicin in mice. Rifampin 92-102 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 71-78 29095659-1 2018 Rifampicin (RIF), a typical ligand of human pregnane X receptor (PXR), powerfully induces the expression of cytochrome P450 3A4 (CYP3A4) in humans. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 44-63 29095659-1 2018 Rifampicin (RIF), a typical ligand of human pregnane X receptor (PXR), powerfully induces the expression of cytochrome P450 3A4 (CYP3A4) in humans. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 65-68 29095659-1 2018 Rifampicin (RIF), a typical ligand of human pregnane X receptor (PXR), powerfully induces the expression of cytochrome P450 3A4 (CYP3A4) in humans. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 108-127 29095659-1 2018 Rifampicin (RIF), a typical ligand of human pregnane X receptor (PXR), powerfully induces the expression of cytochrome P450 3A4 (CYP3A4) in humans. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 129-135 29095659-1 2018 Rifampicin (RIF), a typical ligand of human pregnane X receptor (PXR), powerfully induces the expression of cytochrome P450 3A4 (CYP3A4) in humans. Rifampin 12-15 nuclear receptor subfamily 1 group I member 2 Homo sapiens 44-63 29095659-1 2018 Rifampicin (RIF), a typical ligand of human pregnane X receptor (PXR), powerfully induces the expression of cytochrome P450 3A4 (CYP3A4) in humans. Rifampin 12-15 nuclear receptor subfamily 1 group I member 2 Homo sapiens 65-68 29095659-1 2018 Rifampicin (RIF), a typical ligand of human pregnane X receptor (PXR), powerfully induces the expression of cytochrome P450 3A4 (CYP3A4) in humans. Rifampin 12-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 108-127 29095659-1 2018 Rifampicin (RIF), a typical ligand of human pregnane X receptor (PXR), powerfully induces the expression of cytochrome P450 3A4 (CYP3A4) in humans. Rifampin 12-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 129-135 29095659-2 2018 Although it is thought that RIF is not a ligand of rodent PXR, treatment with high-dose RIF (e.g. more than 20 mg/kg) increases the expression of CYP3A in the mouse liver. Rifampin 88-91 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 146-151 29095659-6 2018 In addition, high-dose RIF stimulated nuclear accumulation of mPXR in the mouse liver, and geldanamycin and okadaic acid attenuated the induction of Cyp3a11 and other PXR-target genes in primary hepatocytes, suggesting that high-dose RIF triggers nuclear translocation of mPXR. Rifampin 23-26 nuclear receptor subfamily 1, group I, member 2 Mus musculus 62-66 29095659-6 2018 In addition, high-dose RIF stimulated nuclear accumulation of mPXR in the mouse liver, and geldanamycin and okadaic acid attenuated the induction of Cyp3a11 and other PXR-target genes in primary hepatocytes, suggesting that high-dose RIF triggers nuclear translocation of mPXR. Rifampin 23-26 nuclear receptor subfamily 1, group I, member 2 Mus musculus 63-66 29095659-6 2018 In addition, high-dose RIF stimulated nuclear accumulation of mPXR in the mouse liver, and geldanamycin and okadaic acid attenuated the induction of Cyp3a11 and other PXR-target genes in primary hepatocytes, suggesting that high-dose RIF triggers nuclear translocation of mPXR. Rifampin 23-26 nuclear receptor subfamily 1, group I, member 2 Mus musculus 272-276 29095659-6 2018 In addition, high-dose RIF stimulated nuclear accumulation of mPXR in the mouse liver, and geldanamycin and okadaic acid attenuated the induction of Cyp3a11 and other PXR-target genes in primary hepatocytes, suggesting that high-dose RIF triggers nuclear translocation of mPXR. Rifampin 234-237 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 149-156 30283337-6 2018 Furthermore, the toxicity was increased in CYP3A4-induced PHH by rifampicin, and CYP3A4 over-expressed (OE) HepG2 and L02 cells. Rifampin 65-75 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 43-49 30351048-14 2018 Furthermore, it reduced valproate- and rifampin-induced LXRalpha- and pregnane X receptor-mediated lipogenesis, respectively, which indicates its potential benefit in treating drug-induced hepatic steatosis. Rifampin 39-47 nuclear receptor subfamily 1 group H member 3 Homo sapiens 56-64 30351048-14 2018 Furthermore, it reduced valproate- and rifampin-induced LXRalpha- and pregnane X receptor-mediated lipogenesis, respectively, which indicates its potential benefit in treating drug-induced hepatic steatosis. Rifampin 39-47 nuclear receptor subfamily 1 group I member 2 Homo sapiens 70-89 30091221-0 2018 PBPK Models for CYP3A4 and P-gp DDI Prediction: A Modeling Network of Rifampicin, Itraconazole, Clarithromycin, Midazolam, Alfentanil, and Digoxin. Rifampin 70-80 phosphoglycolate phosphatase Homo sapiens 27-31 30180590-5 2018 To assess if the prototypical OATP inhibitor rifampicin can inhibit liver uptake of [11C]erlotinib in vivo, we performed [11C]erlotinib PET scans in six healthy volunteers without and with intravenous pretreatment with rifampicin (600 mg). Rifampin 45-55 solute carrier organic anion transporter family member 1A2 Homo sapiens 30-34 30180590-10 2018 Furthermore, our data suggest that a standard clinical dose of rifampicin may exert in vivo a moderate inhibitory effect on hepatic OATP2B1. Rifampin 63-73 solute carrier organic anion transporter family member 2B1 Homo sapiens 132-139 29908302-3 2018 Rifampicin, a PXR-activating agent, elicits a similar effect as PXR+ cells. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 14-17 29908302-3 2018 Rifampicin, a PXR-activating agent, elicits a similar effect as PXR+ cells. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 64-67 30195730-11 2018 Rifampicin pretreatment attenuated SE-induced neuroinflammation and decreased the hippocampal expression of interleukin-1beta, tumor necrosis factor-alpha, nuclear factor kappa-B, and cyclooxygenase-2. Rifampin 0-10 interleukin 1 beta Rattus norvegicus 108-178 30195730-11 2018 Rifampicin pretreatment attenuated SE-induced neuroinflammation and decreased the hippocampal expression of interleukin-1beta, tumor necrosis factor-alpha, nuclear factor kappa-B, and cyclooxygenase-2. Rifampin 0-10 prostaglandin-endoperoxide synthase 2 Rattus norvegicus 184-200 30195730-12 2018 Moreover, rifampicin mitigated SE-induced neuronal apoptosis as indicated by fewer positive cytochrome c immunostained cells and lower caspase-3 activity in the hippocampus. Rifampin 10-20 caspase 3 Rattus norvegicus 135-144 30170758-3 2018 Rifampin induces UGT1A9, UGT2B7, CYP3A4, and CYP3A5. Rifampin 0-8 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 17-23 30025298-1 2018 Rifampicin (RIF) and clindamycin phosphate (CDM) are the main drugs currently used in combination to treat severe infectious diseases in hair follicles. Rifampin 0-10 ras homolog family member F, filopodia associated Homo sapiens 12-15 29243231-2 2018 Plasma and urine CPI data in the presence/absence of rifampicin were modeled to describe CPI synthesis, elimination clearances, and obtain rifampicin in vivo OATP Ki. Rifampin 139-149 solute carrier organic anion transporter family member 1A2 Homo sapiens 158-162 29243231-5 2018 Estimated rifampicin in vivo unbound OATP Ki (0.13 muM) using CPI data was 2-fold lower relative to rosuvastatin. Rifampin 10-20 solute carrier organic anion transporter family member 1A2 Homo sapiens 37-41 30170758-3 2018 Rifampin induces UGT1A9, UGT2B7, CYP3A4, and CYP3A5. Rifampin 0-8 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 25-31 30170758-3 2018 Rifampin induces UGT1A9, UGT2B7, CYP3A4, and CYP3A5. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 33-39 30170758-3 2018 Rifampin induces UGT1A9, UGT2B7, CYP3A4, and CYP3A5. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 45-51 30148542-0 2018 Xpert MTB/RIF assay for extrapulmonary tuberculosis and rifampicin resistance. Rifampin 57-67 ras homolog family member F, filopodia associated Homo sapiens 11-14 30148542-3 2018 Xpert MTB/RIF (Xpert) is a World Health Organization (WHO)-recommended, rapid, automated, nucleic acid amplification assay that is used widely for simultaneous detection of Mycobacterium tuberculosis complex and rifampicin resistance in sputum specimens. Rifampin 213-223 ras homolog family member F, filopodia associated Homo sapiens 11-14 30087611-1 2018 Metoprolol is used for phenotyping of cytochrome P450 (CYP) 2D6, a CYP isoform considered not to be inducible by inducers of the CYP2C, CYP2B, and CYP3A families such as rifampicin. Rifampin 170-180 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 38-63 29742964-9 2018 In addition, in the organoids generated, cytochrome P450 3A8 (CYP3A8) activity was inhibited by the specific inhibitor ketoconazole and was induced by rifampicin. Rifampin 151-161 cytochrome P450 family 3 subfamily A member 4 Macaca fascicularis 41-60 29742964-9 2018 In addition, in the organoids generated, cytochrome P450 3A8 (CYP3A8) activity was inhibited by the specific inhibitor ketoconazole and was induced by rifampicin. Rifampin 151-161 cytochrome P450 family 3 subfamily A member 4 Macaca fascicularis 62-68 30068608-7 2018 This interaction is the target of rifampicin, which prevents A17 binding, explaining the inhibition of viral morphogenesis. Rifampin 34-44 immunoglobulin kappa variable 2-30 Homo sapiens 61-64 30087611-2 2018 While assessing CYP2D6 activity under basal conditions and after pre-treatment with rifampicin in vivo, we surprisingly observed a drop in the metoprolol/alpha-OH-metoprolol clearance ratio, suggesting CYP2D6 induction. Rifampin 84-94 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 16-22 30087611-2 2018 While assessing CYP2D6 activity under basal conditions and after pre-treatment with rifampicin in vivo, we surprisingly observed a drop in the metoprolol/alpha-OH-metoprolol clearance ratio, suggesting CYP2D6 induction. Rifampin 84-94 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 202-208 30087611-9 2018 CYP3A4, 2B6, and 2C9, which are inducible by rifampicin, contribute to alpha-hydroxylation, O-demethylation, and N-dealkylation of metoprolol. Rifampin 45-55 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-11 28685622-8 2018 TaqMan gene expression analysis of human hepatocytes treated with PF-06282999 and the prototypical PXR agonist rifampin demonstrated increases in mRNA for CYP3A4 and related CYPs that are regulated by PXR. Rifampin 112-120 nuclear receptor subfamily 1 group I member 2 Homo sapiens 100-103 30094330-1 2018 Introduction: Oral rifampicin has been shown to significantly reduce amyloid beta (Abeta) and tau pathologies in mice. Rifampin 19-29 histocompatibility 2, class II antigen A, beta 1 Mus musculus 83-88 29666154-8 2018 The ft values were then used for evaluating in vitro-in vivo correlations of hepatic uptake inhibition with OATP inhibitors rifampicin and cyclosporine. Rifampin 124-134 solute carrier organic anion transporter family member 1A2 Homo sapiens 108-112 29940951-9 2018 Compared with drug susceptibility testing (DST), the pooled accordance rate of Xpert MTB/RIF in detecting rifampicin-susceptible cases and rifampicin-resistant cases was 99% (95% CI: 95-104%, I2 = 8.7%) and 94% (95% CI: 86-102%), respectively. Rifampin 106-116 metallothionein 1J, pseudogene Homo sapiens 85-88 29940951-9 2018 Compared with drug susceptibility testing (DST), the pooled accordance rate of Xpert MTB/RIF in detecting rifampicin-susceptible cases and rifampicin-resistant cases was 99% (95% CI: 95-104%, I2 = 8.7%) and 94% (95% CI: 86-102%), respectively. Rifampin 106-116 ras homolog family member F, filopodia associated Homo sapiens 89-92 28685622-8 2018 TaqMan gene expression analysis of human hepatocytes treated with PF-06282999 and the prototypical PXR agonist rifampin demonstrated increases in mRNA for CYP3A4 and related CYPs that are regulated by PXR. Rifampin 112-120 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 156-162 28685622-8 2018 TaqMan gene expression analysis of human hepatocytes treated with PF-06282999 and the prototypical PXR agonist rifampin demonstrated increases in mRNA for CYP3A4 and related CYPs that are regulated by PXR. Rifampin 112-120 nuclear receptor subfamily 1 group I member 2 Homo sapiens 202-205 28686070-0 2018 Effects of aging and rifampicin pretreatment on the pharmacokinetics of human cytochrome P450 probes caffeine, warfarin, omeprazole, metoprolol and midazolam in common marmosets genotyped for cytochrome P450 2C19. Rifampin 21-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 29343610-5 2018 CYP2J2 has been shown to be resistant to induction by canonical CYP inducers such as phenytoin and rifampin. Rifampin 99-107 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 0-6 29412463-4 2018 This study enrolled 64 healthy subjects to evaluate the effect of cyclosporine, a P-gp, BCRP, and OATP1B1/1B3 inhibitor; rifampin, a strong CYP3A4 and P-gp inducer; efavirenz, a moderate CYP3A4 inducer; and cobicistat, a potent CYP3A inhibitor, on presatovir pharmacokinetics. Rifampin 121-129 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 140-146 29412463-4 2018 This study enrolled 64 healthy subjects to evaluate the effect of cyclosporine, a P-gp, BCRP, and OATP1B1/1B3 inhibitor; rifampin, a strong CYP3A4 and P-gp inducer; efavirenz, a moderate CYP3A4 inducer; and cobicistat, a potent CYP3A inhibitor, on presatovir pharmacokinetics. Rifampin 121-129 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 187-193 29440451-2 2018 The goal of this research was to establish PXR-/CAR-/AhR-specific drug-metabolizing enzyme (DME) and transporter gene expression signatures in sandwich-cultured cryopreserved human hepatocytes using selective activators of PXR (rifampin), CAR (CITCO), and AhR (omeprazole). Rifampin 228-236 nuclear receptor subfamily 1 group I member 2 Homo sapiens 43-46 29440451-2 2018 The goal of this research was to establish PXR-/CAR-/AhR-specific drug-metabolizing enzyme (DME) and transporter gene expression signatures in sandwich-cultured cryopreserved human hepatocytes using selective activators of PXR (rifampin), CAR (CITCO), and AhR (omeprazole). Rifampin 228-236 nuclear receptor subfamily 1 group I member 3 Homo sapiens 48-51 29440451-2 2018 The goal of this research was to establish PXR-/CAR-/AhR-specific drug-metabolizing enzyme (DME) and transporter gene expression signatures in sandwich-cultured cryopreserved human hepatocytes using selective activators of PXR (rifampin), CAR (CITCO), and AhR (omeprazole). Rifampin 228-236 aryl hydrocarbon receptor Homo sapiens 53-56 29440451-2 2018 The goal of this research was to establish PXR-/CAR-/AhR-specific drug-metabolizing enzyme (DME) and transporter gene expression signatures in sandwich-cultured cryopreserved human hepatocytes using selective activators of PXR (rifampin), CAR (CITCO), and AhR (omeprazole). Rifampin 228-236 nuclear receptor subfamily 1 group I member 2 Homo sapiens 223-226 29469947-6 2018 The inducers omeprazole, phenobarbital and rifampicin increased the levels of CYP1A2, 2B6 and 3A4 mRNAs by 110-fold, 12.5-fold and 5.4-fold, respectively, at day 2, compared with control human hepatocytes. Rifampin 43-53 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 78-84 29872095-6 2018 GM-CSF neutralization compromised the bacterial control under sub-optimal isoniazid/rifampicin treatment in TNFalpha-deficient mice, leading to exacerbated lung inflammation with necrotic granulomatous structures and high numbers of intracellular M. tuberculosis bacilli. Rifampin 84-94 colony stimulating factor 2 (granulocyte-macrophage) Mus musculus 0-6 29867823-6 2018 In vitro, colistin plus rifampin was synergistic against all the strains at 24 h. In vivo, compared to the controls, rifampin alone reduced tissue bacterial concentrations against VIM-1 and OXA-48 plus CTX-M-15 strains; CMS plus rifampin reduced tissue bacterial concentrations of these two CP-Kp and of the KPC-3 strain. Rifampin 24-32 OXA-48 Klebsiella pneumoniae 190-196 29867823-6 2018 In vitro, colistin plus rifampin was synergistic against all the strains at 24 h. In vivo, compared to the controls, rifampin alone reduced tissue bacterial concentrations against VIM-1 and OXA-48 plus CTX-M-15 strains; CMS plus rifampin reduced tissue bacterial concentrations of these two CP-Kp and of the KPC-3 strain. Rifampin 24-32 cystin 1 Mus musculus 291-296 29867823-6 2018 In vitro, colistin plus rifampin was synergistic against all the strains at 24 h. In vivo, compared to the controls, rifampin alone reduced tissue bacterial concentrations against VIM-1 and OXA-48 plus CTX-M-15 strains; CMS plus rifampin reduced tissue bacterial concentrations of these two CP-Kp and of the KPC-3 strain. Rifampin 24-32 KPC-3 Klebsiella pneumoniae 308-313 29867823-6 2018 In vitro, colistin plus rifampin was synergistic against all the strains at 24 h. In vivo, compared to the controls, rifampin alone reduced tissue bacterial concentrations against VIM-1 and OXA-48 plus CTX-M-15 strains; CMS plus rifampin reduced tissue bacterial concentrations of these two CP-Kp and of the KPC-3 strain. Rifampin 117-125 OXA-48 Klebsiella pneumoniae 190-196 29867823-6 2018 In vitro, colistin plus rifampin was synergistic against all the strains at 24 h. In vivo, compared to the controls, rifampin alone reduced tissue bacterial concentrations against VIM-1 and OXA-48 plus CTX-M-15 strains; CMS plus rifampin reduced tissue bacterial concentrations of these two CP-Kp and of the KPC-3 strain. Rifampin 117-125 cystin 1 Mus musculus 291-296 29867823-6 2018 In vitro, colistin plus rifampin was synergistic against all the strains at 24 h. In vivo, compared to the controls, rifampin alone reduced tissue bacterial concentrations against VIM-1 and OXA-48 plus CTX-M-15 strains; CMS plus rifampin reduced tissue bacterial concentrations of these two CP-Kp and of the KPC-3 strain. Rifampin 117-125 KPC-3 Klebsiella pneumoniae 308-313 29867823-6 2018 In vitro, colistin plus rifampin was synergistic against all the strains at 24 h. In vivo, compared to the controls, rifampin alone reduced tissue bacterial concentrations against VIM-1 and OXA-48 plus CTX-M-15 strains; CMS plus rifampin reduced tissue bacterial concentrations of these two CP-Kp and of the KPC-3 strain. Rifampin 117-125 OXA-48 Klebsiella pneumoniae 190-196 29867823-6 2018 In vitro, colistin plus rifampin was synergistic against all the strains at 24 h. In vivo, compared to the controls, rifampin alone reduced tissue bacterial concentrations against VIM-1 and OXA-48 plus CTX-M-15 strains; CMS plus rifampin reduced tissue bacterial concentrations of these two CP-Kp and of the KPC-3 strain. Rifampin 117-125 cystin 1 Mus musculus 291-296 29867823-6 2018 In vitro, colistin plus rifampin was synergistic against all the strains at 24 h. In vivo, compared to the controls, rifampin alone reduced tissue bacterial concentrations against VIM-1 and OXA-48 plus CTX-M-15 strains; CMS plus rifampin reduced tissue bacterial concentrations of these two CP-Kp and of the KPC-3 strain. Rifampin 117-125 KPC-3 Klebsiella pneumoniae 308-313 29867823-7 2018 Rifampin and the combination increased the survival against the KPC-3 strain; in the pneumonia model, the combination also improved the survival. Rifampin 0-8 KPC-3 Klebsiella pneumoniae 64-69 29488691-0 2018 Sensitivity of ivacaftor to drug-drug interactions with rifampin, a cytochrome P450 3A4 inducer. Rifampin 56-64 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-87 29488691-2 2018 Because it is a substrate of the cytochrome P450 system, specifically CYP3A4/5, ivacaftor is subject to significant drug-drug interactions, including due to commonly used antimicrobials such as rifampin. Rifampin 194-202 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 70-76 29356861-12 2018 The compounds differentially antagonised the induction of PXR-regulated genes by rifampicin in primary human hepatocytes. Rifampin 81-91 nuclear receptor subfamily 1 group I member 2 Homo sapiens 58-61 29031874-8 2018 Rifampicin, a potent CYP3A4 inducer, is the first-line treatment of cholestatic pruritus. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-27 29440179-8 2018 Human liver carcinoma cells (HepG2) were transfected with CYP3A4 luciferase and PXR plasmids, followed by treatment with JNK inhibitor (SP600125; SP) and PXR activators rifampicin (RIF) or hyperforin. Rifampin 169-179 nuclear receptor subfamily 1 group I member 2 Homo sapiens 154-157 29343610-5 2018 CYP2J2 has been shown to be resistant to induction by canonical CYP inducers such as phenytoin and rifampin. Rifampin 99-107 peptidylprolyl isomerase G Homo sapiens 0-3 29470228-3 2018 The long half-life of amiodarone and its active metabolite in combination with the late onset and offset of cytochrome P4503A (CYP3A4) induction by rifampicin makes this a challenging drug-drug interaction to cope with in clinical practice. Rifampin 148-158 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 127-133 29635947-9 2018 Rifampicin significantly increases plasma concentrations of both enantiomers through inhibition of OATP1B3, whereas enantioselectivity of fexofenadine uptake by OATP1B3-expressing cells has not been observed. Rifampin 0-10 solute carrier organic anion transporter family member 1B3 Homo sapiens 99-106 29143479-1 2018 Rifampin (RIF) is a bactericidal antibiotic drug and potent inducer of hepatic and intestinal cytochrome P-450 (CYP-450) enzyme systems. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 29143479-1 2018 Rifampin (RIF) is a bactericidal antibiotic drug and potent inducer of hepatic and intestinal cytochrome P-450 (CYP-450) enzyme systems. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-119 29143479-1 2018 Rifampin (RIF) is a bactericidal antibiotic drug and potent inducer of hepatic and intestinal cytochrome P-450 (CYP-450) enzyme systems. Rifampin 10-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 29143479-1 2018 Rifampin (RIF) is a bactericidal antibiotic drug and potent inducer of hepatic and intestinal cytochrome P-450 (CYP-450) enzyme systems. Rifampin 10-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-119 29393813-20 2018 Further consideration should be given to the use of rifampin in the treatment of CSF shunt infection. Rifampin 52-60 colony stimulating factor 2 Homo sapiens 81-84 29435803-0 2018 Rifampicin Prevents SH-SY5Y Cells from Rotenone-Induced Apoptosis via the PI3K/Akt/GSK-3beta/CREB Signaling Pathway. Rifampin 0-10 AKT serine/threonine kinase 1 Homo sapiens 79-82 29435803-0 2018 Rifampicin Prevents SH-SY5Y Cells from Rotenone-Induced Apoptosis via the PI3K/Akt/GSK-3beta/CREB Signaling Pathway. Rifampin 0-10 glycogen synthase kinase 3 beta Homo sapiens 83-92 29435803-0 2018 Rifampicin Prevents SH-SY5Y Cells from Rotenone-Induced Apoptosis via the PI3K/Akt/GSK-3beta/CREB Signaling Pathway. Rifampin 0-10 cAMP responsive element binding protein 1 Homo sapiens 93-97 29435803-3 2018 This study aims to investigate whether rifampicin protects rotenone-lesioned SH-SY5Y cells via regulating PI3K/Akt/GSK-3beta/CREB pathway. Rifampin 39-49 AKT serine/threonine kinase 1 Homo sapiens 111-114 29435803-3 2018 This study aims to investigate whether rifampicin protects rotenone-lesioned SH-SY5Y cells via regulating PI3K/Akt/GSK-3beta/CREB pathway. Rifampin 39-49 glycogen synthase kinase 3 beta Homo sapiens 115-124 29435803-3 2018 This study aims to investigate whether rifampicin protects rotenone-lesioned SH-SY5Y cells via regulating PI3K/Akt/GSK-3beta/CREB pathway. Rifampin 39-49 cAMP responsive element binding protein 1 Homo sapiens 125-129 29098603-5 2018 The model was validated using clinical PK data for co-administration of GXR with ketoconazole (strong CYP3A4 inhibitor) or rifampicin (strong CYP3A4 inducer). Rifampin 123-133 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 142-148 29437631-5 2018 The adjuvant potency of C12-PRP was apparent by its ability to reduce the MIC of minocycline and rifampin below their interpretative susceptibility breakpoints against MDR/XDR P. aeruginosa An attempt to optimize C12-PRP through peptidomimetic modification was performed by replacing all l- to d-amino acids. Rifampin 97-105 prion protein Homo sapiens 28-31 29437631-6 2018 C12-PRP demonstrated that it was amenable to optimization, since synergism with minocycline and rifampin were retained. Rifampin 96-104 prion protein Homo sapiens 4-7 29435803-8 2018 Both the cell viability and the expression of phospho-CREB in cells pretreated with rifampicin were higher than those of cells exposed to rotenone alone. Rifampin 84-94 cAMP responsive element binding protein 1 Homo sapiens 54-58 29435803-9 2018 Moreover, pretreatment of SH-SY5Y cells with rifampicin enhanced phosphorylation of Akt and suppressed activity of GSK-3beta. Rifampin 45-55 AKT serine/threonine kinase 1 Homo sapiens 84-87 29435803-9 2018 Moreover, pretreatment of SH-SY5Y cells with rifampicin enhanced phosphorylation of Akt and suppressed activity of GSK-3beta. Rifampin 45-55 glycogen synthase kinase 3 beta Homo sapiens 115-124 29435803-10 2018 The addition of LY294002, a PI3K inhibitor, could suppress phosphorylation of Akt and CREB and activate GSK-3beta, resulting in abolishment of neuroprotective effects of rifampicin on cells exposed to rotenone. Rifampin 170-180 glycogen synthase kinase 3 beta Homo sapiens 104-113 29435803-11 2018 Rifampicin provides neuroprotection against dopaminergic degeneration, partially via the PI3K/Akt/GSK-3beta/CREB signaling pathway. Rifampin 0-10 AKT serine/threonine kinase 1 Homo sapiens 94-97 29435803-11 2018 Rifampicin provides neuroprotection against dopaminergic degeneration, partially via the PI3K/Akt/GSK-3beta/CREB signaling pathway. Rifampin 0-10 glycogen synthase kinase 3 beta Homo sapiens 98-107 29435803-11 2018 Rifampicin provides neuroprotection against dopaminergic degeneration, partially via the PI3K/Akt/GSK-3beta/CREB signaling pathway. Rifampin 0-10 cAMP responsive element binding protein 1 Homo sapiens 108-112 29437631-3 2018 SPRLPs were further evaluated for their potential to serve as adjuvants in combination with existing antibiotics to enhance antibacterial activity against drug-resistant Pseudomonas aeruginosa Out of 16 prepared SPRLPs, C12-PRP was found to significantly potentiate the antibiotics minocycline and rifampin against multidrug- and extensively drug-resistant (MDR/XDR) P. aeruginosa clinical isolates. Rifampin 298-306 prion protein Homo sapiens 224-227 29610665-8 2018 Physiologically based pharmacokinetic modeling (PBPK) was used to simulate the clinical consequences of rifampin induction of CYP3A4- and UGT1A4-mediated midazolam metabolism. Rifampin 104-112 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 126-132 29610665-8 2018 Physiologically based pharmacokinetic modeling (PBPK) was used to simulate the clinical consequences of rifampin induction of CYP3A4- and UGT1A4-mediated midazolam metabolism. Rifampin 104-112 UDP glucuronosyltransferase family 1 member A4 Homo sapiens 138-144 29285606-9 2018 Regarding phase II metabolism, UGT2B7, 1A8 and 1A3 showed highest activity in glucuronidation of tamoxifen bisphenol and metabolite E. Anti-estrogenic metabolites (Z)-4-hydroxytamoxifen, (Z)-endoxifen and (Z)-norendoxifen inhibited the activity of CYP2C enzymes while tamoxifen bisphenol consistently induced CYPs similar to rifampicin and phenobarbital. Rifampin 325-335 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 31-37 29593688-10 2018 Rifampicin resistance reported by Xpert MTB/RIF slightly deviated from that by phenotypic antibiotic susceptibility testing and requires further study with a larger sample size. Rifampin 0-10 ras homolog family member F, filopodia associated Homo sapiens 44-47 29358328-7 2018 Of note, M. tuberculosis infection and rifampicin exposure synergistically modulated macrophage drug-efflux transporters in vitro We also found that the xenobiotic nuclear receptor pregnane X receptor (PXR) modulates macrophage drug-efflux transporter expression and activity, which compromised the anti-TB efficacy of rifampicin. Rifampin 39-49 nuclear receptor subfamily 1, group I, member 2 Mus musculus 202-205 29358328-7 2018 Of note, M. tuberculosis infection and rifampicin exposure synergistically modulated macrophage drug-efflux transporters in vitro We also found that the xenobiotic nuclear receptor pregnane X receptor (PXR) modulates macrophage drug-efflux transporter expression and activity, which compromised the anti-TB efficacy of rifampicin. Rifampin 319-329 nuclear receptor subfamily 1, group I, member 2 Mus musculus 202-205 29358328-8 2018 We further validated this finding in a TB mouse model in which use of the PXR antagonist ketoconazole rescued rifampicin anti-TB activity. Rifampin 110-120 nuclear receptor subfamily 1, group I, member 2 Mus musculus 74-77 29358328-9 2018 We conclude that PXR activation in macrophages compromises the efficacy of the anti-TB drug rifampicin. Rifampin 92-102 nuclear receptor subfamily 1, group I, member 2 Mus musculus 17-20 28800141-3 2018 This multiarm phase 1 study (Clinicaltrials.gov identifier: NCT01454076) investigated the effect of the strong CYP3A inhibitors ketoconazole and clarithromycin and the strong CYP3A inducer rifampin on the pharmacokinetics of ixazomib. Rifampin 189-197 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 175-180 29315976-2 2018 Here we summarize the underlying mechanisms of the neuroprotective and pro-cognitive effects of rifampicin that are mediated by its anti-inflammatory, anti-tau, anti-amyloid, and cholinergic effects. Rifampin 96-106 microtubule associated protein tau Homo sapiens 156-159 29440136-3 2018 Rifampicin is a common inducer of CYP3A4. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 34-40 28967981-6 2018 In summary, a strong CYP3A4 inducer (rifampin) had a strong effect (>5-fold) on the clinical pharmacokinetics of apatinib, whereas a strong CYP3A inhibitor (itraconazole 100 mg once a day) had a weak effect (1.25- to 2-fold). Rifampin 37-45 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-27 28967981-6 2018 In summary, a strong CYP3A4 inducer (rifampin) had a strong effect (>5-fold) on the clinical pharmacokinetics of apatinib, whereas a strong CYP3A inhibitor (itraconazole 100 mg once a day) had a weak effect (1.25- to 2-fold). Rifampin 37-45 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 21-26 29263072-0 2018 Effect of Genetic Variation of NAT2 on Isoniazid and SLCO1B1 and CES2 on Rifampin Pharmacokinetics in Ghanaian Children with Tuberculosis. Rifampin 73-81 N-acetyltransferase 2 Homo sapiens 31-35 29263072-0 2018 Effect of Genetic Variation of NAT2 on Isoniazid and SLCO1B1 and CES2 on Rifampin Pharmacokinetics in Ghanaian Children with Tuberculosis. Rifampin 73-81 carboxylesterase 2 Homo sapiens 65-69 29636933-2 2018 Reduction in calcineurin inhibitor levels in organ transplant recipients due to enhanced metabolism from interaction with rifampin can predispose these individuals to develop IRIS during the treatment of tuberculosis and mimic sepsis. Rifampin 122-130 calcineurin binding protein 1 Homo sapiens 13-34 30347615-5 2018 METHODS: CYP1A2 and CYP3A4 were induced by omeprazole and rifampicin, respectively. Rifampin 58-68 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 9-15 29191818-5 2018 To analyze the contribution of Cyp3a11 to the protection of atherosclerosis development, Cyp3a11 was induced by Rifampicin (RIF) in ApoE KO and het mice. Rifampin 112-122 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 89-96 29191818-5 2018 To analyze the contribution of Cyp3a11 to the protection of atherosclerosis development, Cyp3a11 was induced by Rifampicin (RIF) in ApoE KO and het mice. Rifampin 112-122 apolipoprotein E Mus musculus 132-136 29191818-5 2018 To analyze the contribution of Cyp3a11 to the protection of atherosclerosis development, Cyp3a11 was induced by Rifampicin (RIF) in ApoE KO and het mice. Rifampin 124-127 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 89-96 29191818-5 2018 To analyze the contribution of Cyp3a11 to the protection of atherosclerosis development, Cyp3a11 was induced by Rifampicin (RIF) in ApoE KO and het mice. Rifampin 124-127 apolipoprotein E Mus musculus 132-136 29357810-5 2018 mRNA expression levels and enzyme activities of CYP1A2, CYP2B6, and CYP3A in HepaRG cells treated with prototypical inducers of each CYP isoform [omeprazole (OME) for CYP1A2, phenobarbital (PB) for CYP2B6, and rifampicin (RIF) for CYP3A] were evaluated. Rifampin 210-220 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 48-54 29357810-5 2018 mRNA expression levels and enzyme activities of CYP1A2, CYP2B6, and CYP3A in HepaRG cells treated with prototypical inducers of each CYP isoform [omeprazole (OME) for CYP1A2, phenobarbital (PB) for CYP2B6, and rifampicin (RIF) for CYP3A] were evaluated. Rifampin 210-220 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-73 29357810-5 2018 mRNA expression levels and enzyme activities of CYP1A2, CYP2B6, and CYP3A in HepaRG cells treated with prototypical inducers of each CYP isoform [omeprazole (OME) for CYP1A2, phenobarbital (PB) for CYP2B6, and rifampicin (RIF) for CYP3A] were evaluated. Rifampin 210-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 29357810-5 2018 mRNA expression levels and enzyme activities of CYP1A2, CYP2B6, and CYP3A in HepaRG cells treated with prototypical inducers of each CYP isoform [omeprazole (OME) for CYP1A2, phenobarbital (PB) for CYP2B6, and rifampicin (RIF) for CYP3A] were evaluated. Rifampin 222-225 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 48-54 29357810-5 2018 mRNA expression levels and enzyme activities of CYP1A2, CYP2B6, and CYP3A in HepaRG cells treated with prototypical inducers of each CYP isoform [omeprazole (OME) for CYP1A2, phenobarbital (PB) for CYP2B6, and rifampicin (RIF) for CYP3A] were evaluated. Rifampin 222-225 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-73 29357810-5 2018 mRNA expression levels and enzyme activities of CYP1A2, CYP2B6, and CYP3A in HepaRG cells treated with prototypical inducers of each CYP isoform [omeprazole (OME) for CYP1A2, phenobarbital (PB) for CYP2B6, and rifampicin (RIF) for CYP3A] were evaluated. Rifampin 222-225 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 28833380-8 2018 Food effects, comedication with itraconazole [a cytochrome P450 (CYP) 3A4 inhibitor], fluconazole (a CYP2C19 inhibitor) and rifampicin (a CYP3A4 inducer) and formulation effects were incorporated into the base model a priori. Rifampin 124-134 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 138-144 30347615-5 2018 METHODS: CYP1A2 and CYP3A4 were induced by omeprazole and rifampicin, respectively. Rifampin 58-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-26 28715853-2 2017 The present study evaluated the possible pharmacokinetic interactions of selexipag with gemfibrozil, a strong CYP2C8 inhibitor, and rifampicin, an inducer of CYP2C8. Rifampin 132-142 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 158-164 29226732-10 2018 Further studies are needed in order to provide definitive recommendations of pharmacogenetic-based individualization: however lower isoniazid doses in NAT2 slow acetylators and higher rifampicin doses in individuals with SLCO1B1 loss of function genes are promising strategies. Rifampin 184-194 solute carrier organic anion transporter family member 1B1 Homo sapiens 221-228 28946447-0 2018 Influence of the ultrasound-assisted synthesis of Cu-BTC metal-organic frameworks nanoparticles on uptake and release properties of rifampicin. Rifampin 132-142 betacellulin Homo sapiens 53-56 28946447-1 2018 In this work, we study uptake and release properties of rifampicin (denoted henceforth as Rif) from ultrasound-assisted synthesis Cu-BTC nanoparticles in comparison with bulk Cu-BTC and activated carbon. Rifampin 56-66 betacellulin Homo sapiens 133-136 28946447-1 2018 In this work, we study uptake and release properties of rifampicin (denoted henceforth as Rif) from ultrasound-assisted synthesis Cu-BTC nanoparticles in comparison with bulk Cu-BTC and activated carbon. Rifampin 90-93 betacellulin Homo sapiens 133-136 29076107-10 2017 CONCLUSION: These findings confirm that amenamevir (as a cytochrome P450 3A4 substrate) can interact with ketoconazole or rifampicin, and (as a cytochrome P450 3A4 inducer) can interact with midazolam; however, no interaction between amenamevir and (S)-warfarin was observed, indicating that amenamevir is not an inducer of cytochrome P450 2C9. Rifampin 122-132 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 57-76 29157826-6 2017 RESULTS: Transient transfection assays showed that UA effectively attenuated CYP3A4 and CYP2B6 promoter activities mediated by rifampin (RIF, human PXR agonist) and CITCO (human CAR agonist). Rifampin 127-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 29157826-6 2017 RESULTS: Transient transfection assays showed that UA effectively attenuated CYP3A4 and CYP2B6 promoter activities mediated by rifampin (RIF, human PXR agonist) and CITCO (human CAR agonist). Rifampin 137-140 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 77-83 29157826-6 2017 RESULTS: Transient transfection assays showed that UA effectively attenuated CYP3A4 and CYP2B6 promoter activities mediated by rifampin (RIF, human PXR agonist) and CITCO (human CAR agonist). Rifampin 137-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 28912253-6 2017 The model was then applied to predict the drug interaction with the strong CYP3A4 inducer rifampin (RIF) and the sensitive CYP3A4 substrate midazolam (MDZ) in lieu of clinical trials. Rifampin 90-98 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 75-81 28912253-6 2017 The model was then applied to predict the drug interaction with the strong CYP3A4 inducer rifampin (RIF) and the sensitive CYP3A4 substrate midazolam (MDZ) in lieu of clinical trials. Rifampin 100-103 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 75-81 28986232-5 2017 Here, we demonstrate that rifampicin pretreatment alleviated rotenone induced release of IL-1beta and IL-6, and its effects were suppressed when autophagy was inhibited by chloroquine. Rifampin 26-36 interleukin 1 beta Mus musculus 89-97 28986232-5 2017 Here, we demonstrate that rifampicin pretreatment alleviated rotenone induced release of IL-1beta and IL-6, and its effects were suppressed when autophagy was inhibited by chloroquine. Rifampin 26-36 interleukin 6 Mus musculus 102-106 29157826-0 2017 Modulation of pregnane X receptor (PXR) and constitutive androstane receptor (CAR) activation by ursolic acid (UA) attenuates rifampin-isoniazid cytotoxicity. Rifampin 126-134 nuclear receptor subfamily 1 group I member 3 Homo sapiens 44-76 29157826-0 2017 Modulation of pregnane X receptor (PXR) and constitutive androstane receptor (CAR) activation by ursolic acid (UA) attenuates rifampin-isoniazid cytotoxicity. Rifampin 126-134 nuclear receptor subfamily 1 group I member 3 Homo sapiens 78-81 29157826-6 2017 RESULTS: Transient transfection assays showed that UA effectively attenuated CYP3A4 and CYP2B6 promoter activities mediated by rifampin (RIF, human PXR agonist) and CITCO (human CAR agonist). Rifampin 127-135 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 77-83 28688954-0 2017 Rifampicin-induced injury in HepG2 cells is alleviated by TUDCA via increasing bile acid transporters expression and enhancing the Nrf2-mediated adaptive response. Rifampin 0-10 NFE2 like bZIP transcription factor 2 Homo sapiens 131-135 29081091-8 2017 Serum immunoglobulin E (IgE) and interleukin-4 (IL-4) levels were also ameliorated in mice treated with rifampicin. Rifampin 104-114 interleukin 4 Mus musculus 33-46 29081091-8 2017 Serum immunoglobulin E (IgE) and interleukin-4 (IL-4) levels were also ameliorated in mice treated with rifampicin. Rifampin 104-114 interleukin 4 Mus musculus 48-52 29081091-10 2017 Rifampicin suppressed the release of beta-hexosaminidase and histamine from human mast cell (HMC)-1 cultures stimulated with compound 48/80. Rifampin 0-10 O-GlcNAcase Homo sapiens 37-56 29081091-11 2017 Treatment with rifampicin also inhibited secretion of inflammatory mediators, such tumor necrosis factor-alpha (TNF-alpha) and prostaglandin D2 (PGD2), in mast cells activated by compound 48/80. Rifampin 15-25 tumor necrosis factor Mus musculus 83-110 29081091-11 2017 Treatment with rifampicin also inhibited secretion of inflammatory mediators, such tumor necrosis factor-alpha (TNF-alpha) and prostaglandin D2 (PGD2), in mast cells activated by compound 48/80. Rifampin 15-25 tumor necrosis factor Mus musculus 112-121 29081091-11 2017 Treatment with rifampicin also inhibited secretion of inflammatory mediators, such tumor necrosis factor-alpha (TNF-alpha) and prostaglandin D2 (PGD2), in mast cells activated by compound 48/80. Rifampin 15-25 prostaglandin D2 synthase (brain) Mus musculus 127-143 29081091-12 2017 The mRNA expression of cyclooxygenase 2 (COX-2) was reduced in the cells treated with rifampicin in a concentration-dependent manner. Rifampin 86-96 prostaglandin-endoperoxide synthase 2 Mus musculus 23-39 29081091-12 2017 The mRNA expression of cyclooxygenase 2 (COX-2) was reduced in the cells treated with rifampicin in a concentration-dependent manner. Rifampin 86-96 prostaglandin-endoperoxide synthase 2 Mus musculus 41-46 29061956-3 2017 Rifampin is a strong inducer of several drug-metabolizing enzymes, including CYP3A. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 77-82 28870519-7 2017 Rifampicin reduced the late increase of alphaSyn oligomers in sigma1R-/- mice. Rifampin 0-10 synuclein, alpha Mus musculus 40-48 28870519-7 2017 Rifampicin reduced the late increase of alphaSyn oligomers in sigma1R-/- mice. Rifampin 0-10 sigma non-opioid intracellular receptor 1 Mus musculus 62-69 28870519-8 2017 Rifampicin or salubrinal could reduce the loss of dopaminergic neurons in sigma1R-/- mice and improved their motor coordination. Rifampin 0-10 sigma non-opioid intracellular receptor 1 Mus musculus 74-81 29093504-1 2017 Xpert MTB/RIF (Xpert) is a widely-used test for tuberculosis (TB) and rifampicin-resistance. Rifampin 70-80 ras homolog family member F, filopodia associated Homo sapiens 10-13 29061956-4 2017 Hence, rifampin-induced CYP3A activity may decrease the effect of oxycodone. Rifampin 7-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-29 28982166-9 2017 Among these patients, only six (54.5%) were found to have rifampin-resistant isolates in period 2, with no significant infectious events. Rifampin 58-66 period circadian regulator 2 Homo sapiens 89-97 28945086-4 2017 OA attenuated the promoter activities, expressions, and enzyme catalytic activities of CYP3A4 and CYP2B6 mediated by rifampin (RIF) and CITCO. Rifampin 117-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 28945086-4 2017 OA attenuated the promoter activities, expressions, and enzyme catalytic activities of CYP3A4 and CYP2B6 mediated by rifampin (RIF) and CITCO. Rifampin 127-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 28407303-2 2017 Rifampicin, an antitubercular agent, is one of the most potent inducers of hepatic and intestinal CYP3A4 thus increasing dihydropyridine metabolism. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 98-104 28856417-6 2017 Although at higher concentration, HBD-1 (x 2 MIC) and Pep-B (x 2 MIC) led to decrease in in vitro dormant mycobacterial load as compared to rifampicin (x 25 MIC) and isoniazid (x 16 MIC). Rifampin 140-150 defensin beta 1 Homo sapiens 34-39 28856417-6 2017 Although at higher concentration, HBD-1 (x 2 MIC) and Pep-B (x 2 MIC) led to decrease in in vitro dormant mycobacterial load as compared to rifampicin (x 25 MIC) and isoniazid (x 16 MIC). Rifampin 140-150 peptidase B Homo sapiens 54-59 29171455-3 2017 Corresponding of CYP2E1 polymorphism in TB patients with the level of isoniazid and rifampicin as well as for the outcome and toxicity development during inpatient TB treatment. Rifampin 84-94 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 17-23 29171455-8 2017 RESULTS: The concentration of rifampicin 6 h after its intake was 17.6% higher in carriers of slow metabolizer (SM) CYP2E1 genotype than in patients with rapid metabolizer (RM) genotype that proved a participation of hepatic enzyme CYP2E1 in metabolism of rifampicin. Rifampin 30-40 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 116-122 29171455-8 2017 RESULTS: The concentration of rifampicin 6 h after its intake was 17.6% higher in carriers of slow metabolizer (SM) CYP2E1 genotype than in patients with rapid metabolizer (RM) genotype that proved a participation of hepatic enzyme CYP2E1 in metabolism of rifampicin. Rifampin 30-40 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 232-238 28942083-4 2017 Treatment with rifampicin, a ligand of PXR, translocated PXR from the cytoplasm to nucleus and increased expression levels of CYP3A4 mRNA in HepG2 cells cultured by the hanging drop method. Rifampin 15-25 nuclear receptor subfamily 1 group I member 2 Homo sapiens 39-42 28942083-4 2017 Treatment with rifampicin, a ligand of PXR, translocated PXR from the cytoplasm to nucleus and increased expression levels of CYP3A4 mRNA in HepG2 cells cultured by the hanging drop method. Rifampin 15-25 nuclear receptor subfamily 1 group I member 2 Homo sapiens 57-60 28942083-4 2017 Treatment with rifampicin, a ligand of PXR, translocated PXR from the cytoplasm to nucleus and increased expression levels of CYP3A4 mRNA in HepG2 cells cultured by the hanging drop method. Rifampin 15-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 126-132 28663285-6 2017 Rifabutin and rifampicin also inhibited several human UGTs including UGT1A4. Rifampin 14-24 UDP glucuronosyltransferase family 1 member A4 Homo sapiens 69-75 29022528-8 2017 GeneXpert MTB/RIF also detected all 4 multidrug-resistant TB cases and an additional 2 rifampicin-resistant cases in culture-negative samples. Rifampin 87-97 ras homolog family member F, filopodia associated Homo sapiens 14-17 28836817-6 2017 The main spa type in this study was spa t030 with frequency as high as 75.5% from intensive care unit (ICU) of the hospitals and high rate of resistance to rifampicin (53%) was found in MRSA isolates. Rifampin 156-166 surfactant protein A2 Homo sapiens 9-12 28414110-0 2017 Rifampicin as a novel tyrosinase inhibitor: Inhibitory activity and mechanism. Rifampin 0-10 tyrosinase Homo sapiens 22-32 28414110-1 2017 In this study, the inhibitory effect and mechanism of rifampicin on the activity of tyrosinase were investigated for developing a novel tyrosinase inhibitor. Rifampin 54-64 tyrosinase Homo sapiens 84-94 28414110-1 2017 In this study, the inhibitory effect and mechanism of rifampicin on the activity of tyrosinase were investigated for developing a novel tyrosinase inhibitor. Rifampin 54-64 tyrosinase Homo sapiens 136-146 28414110-4 2017 The results obtained from intrinsic fluorescence quenching indicated that rifampicin could interact with tyrosinase. Rifampin 74-84 tyrosinase Homo sapiens 105-115 28414110-6 2017 Moreover, the ANS-binding fluorescence analysis suggested that rifampicin binding to tyrosinase changed the polarity of the hydrophobic regions. Rifampin 63-73 tyrosinase Homo sapiens 85-95 28414110-9 2017 This work identified a novel tyrosinase inhibitor and potentially contributed to the usage of rifampicin as a potential hyperpigmentation drug. Rifampin 94-104 tyrosinase Homo sapiens 29-39 28065764-4 2017 Rifampicin and SR12813 significantly upregulated P-gp expression (1.5-fold and 1.9-fold, respectively) compared to control, as assessed by the ICW protocol. Rifampin 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 49-53 27848009-1 2017 This study investigated the effects of rifampicin-modulated P-glycoprotein (P-gp) and cytochrome P450 (CYP450) activity on the development of steroid-induced osteonecrosis of the femoral head. Rifampin 39-49 ATP-dependent translocase ABCB1 Oryctolagus cuniculus 60-74 27848009-1 2017 This study investigated the effects of rifampicin-modulated P-glycoprotein (P-gp) and cytochrome P450 (CYP450) activity on the development of steroid-induced osteonecrosis of the femoral head. Rifampin 39-49 ATP-dependent translocase ABCB1 Oryctolagus cuniculus 76-80 27848009-1 2017 This study investigated the effects of rifampicin-modulated P-glycoprotein (P-gp) and cytochrome P450 (CYP450) activity on the development of steroid-induced osteonecrosis of the femoral head. Rifampin 39-49 cytochrome P-450 Oryctolagus cuniculus 103-109 27848009-8 2017 P-gp activity of mesenchymal stem cells in rifampicin-implanted femoral head was significantly higher than that in the blank control. Rifampin 43-53 ATP-dependent translocase ABCB1 Oryctolagus cuniculus 0-4 27848009-12 2017 P-gp activity locally enhanced by rifampicin decreases the intracellular steroid concentration, but rifampicin does not have significant effects on peripheral P-gp and hepatic CYP450. Rifampin 34-44 ATP-dependent translocase ABCB1 Oryctolagus cuniculus 0-4 28716462-6 2017 Xpert MTB/RIF and Hain line probe assays identified rpoB and inhA mutations consistent with rifampicin and intermediate isoniazid resistance, and a further possible mutation conferring fluoroquinolone resistance. Rifampin 92-102 inhibin subunit alpha Homo sapiens 61-65 28546421-7 2017 Conversely, PXR knockdown abrogated the suppressive effect of rifampicin on NLRP3 inflammasome activation. Rifampin 62-72 nuclear receptor subfamily 1 group I member 2 Homo sapiens 12-15 28495568-4 2017 DDIs with rifampicin and itraconazole were simulated using in vivo Rdif (ratio of diffusional uptake to active uptake) and beta (the fraction of the sum of intrinsic clearances for metabolism and biliary excretion in all possible itineraries of intracellular drugs including basolateral efflux) estimated by static analyses based on the extended clearance concept, in vivo inhibition constant (Ki) for hepatic OATPs reported previously, and in vivo Ki for CYP3A determined from DDI data with midazolam and itraconazole. Rifampin 10-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 456-461 28535976-6 2017 Epigallocatechin gallate, cyclosporin A, rifampicin, and telmisartan inhibited cilostazol uptake in OATP1B1/1B3-HEK293 cells with Ki values close to their clinical plasma concentration, which suggested possible drug-drug interactions in humans via OATP1B1/1B3. Rifampin 41-51 solute carrier organic anion transporter family member 1B1 Homo sapiens 100-107 28535976-6 2017 Epigallocatechin gallate, cyclosporin A, rifampicin, and telmisartan inhibited cilostazol uptake in OATP1B1/1B3-HEK293 cells with Ki values close to their clinical plasma concentration, which suggested possible drug-drug interactions in humans via OATP1B1/1B3. Rifampin 41-51 solute carrier organic anion transporter family member 1B1 Homo sapiens 248-255 28817905-0 2017 Re: CYP3A4 Induction by Rifampin: An Alternative Pathway for Vitamin D Inactivation in Patients with CYP24A1 Mutations. Rifampin 24-32 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 4-10 28817905-0 2017 Re: CYP3A4 Induction by Rifampin: An Alternative Pathway for Vitamin D Inactivation in Patients with CYP24A1 Mutations. Rifampin 24-32 cytochrome P450 family 24 subfamily A member 1 Homo sapiens 101-108 28546421-6 2017 In addition, PXR agonists (rifampicin and SR12813) or overexpression of a constitutively active PXR markedly suppressed the NLRP3 inflammasome activation. Rifampin 27-37 nuclear receptor subfamily 1 group I member 2 Homo sapiens 13-16 28546421-6 2017 In addition, PXR agonists (rifampicin and SR12813) or overexpression of a constitutively active PXR markedly suppressed the NLRP3 inflammasome activation. Rifampin 27-37 NLR family pyrin domain containing 3 Homo sapiens 124-129 28546421-7 2017 Conversely, PXR knockdown abrogated the suppressive effect of rifampicin on NLRP3 inflammasome activation. Rifampin 62-72 NLR family pyrin domain containing 3 Homo sapiens 76-81 28546104-0 2017 Development of a paediatric physiologically based pharmacokinetic model to assess the impact of drug-drug interactions in tuberculosis co-infected malaria subjects: A case study with artemether-lumefantrine and the CYP3A4-inducer rifampicin. Rifampin 230-240 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 215-221 28546104-4 2017 The concomitant treatment of AL with tuberculosis chemotherapy, which includes the CYP3A4 inducer rifampicin, increases the risk of parasite recrudescence and malaria treatment failure. Rifampin 98-108 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-89 28789699-4 2017 We describe a case of severe poisoning with fixed drug combination of isoniazid and rifampicin successfully managed with oral pyridoxine at national hospital of Sri Lanka. Rifampin 84-94 sorcin Homo sapiens 161-164 28819225-5 2017 In contrast, P450/ABC transporter induction with rifampicin markedly impaired ivermectin absorption. Rifampin 49-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-17 28213941-11 2017 An increase in cediranib dose may be needed for cediranib coadministered with strong uridine glucuronosyl transferase/P-glycoprotein inducers such as rifampicin. Rifampin 150-160 ATP binding cassette subfamily B member 1 Homo sapiens 118-132 29187929-9 2017 Finally, GeneXpert MTB/RIF test detected rifampicin resistance in 20.8%. Rifampin 41-51 ras homolog family member F, filopodia associated Homo sapiens 23-26 28571685-0 2017 Activation of the aryl hydrocarbon receptor decreases rifampicin-induced CYP3A4 expression in primary human hepatocytes and HepaRG. Rifampin 54-64 aryl hydrocarbon receptor Homo sapiens 18-43 28571685-0 2017 Activation of the aryl hydrocarbon receptor decreases rifampicin-induced CYP3A4 expression in primary human hepatocytes and HepaRG. Rifampin 54-64 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 73-79 28571685-6 2017 We also investigated whether knockdown of AhR using siRNA affected the basal expression of PXR and CYP3A4 and induction of CYP3A4 by rifampicin, TCDD and 3MI. Rifampin 133-143 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 123-129 28571685-8 2017 Moreover, in both HepaRG and PHHs, AhR activation decreased rifampicin-induced expression of CYP3A4 mRNA. Rifampin 60-70 aryl hydrocarbon receptor Homo sapiens 35-38 28571685-8 2017 Moreover, in both HepaRG and PHHs, AhR activation decreased rifampicin-induced expression of CYP3A4 mRNA. Rifampin 60-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-99 28571685-9 2017 Knock-down of AhR in PHHs increased both basal and rifampicin-induced expression of CYP3A4 mRNA. Rifampin 51-61 aryl hydrocarbon receptor Homo sapiens 14-17 28571685-9 2017 Knock-down of AhR in PHHs increased both basal and rifampicin-induced expression of CYP3A4 mRNA. Rifampin 51-61 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 84-90 27858342-7 2017 Further analysis demonstrated that OATP1B1 inhibition by rifampin or cyclosporine in the existing inhibitor models needs to be approximately tenfold stronger to recapitulate the observed DDI with these two inhibitors. Rifampin 57-65 solute carrier organic anion transporter family member 1B1 Homo sapiens 35-42 28576766-4 2017 All probes were determined in samples from a single study that examined their behavior and their association with rosuvastatin (RSV) pharmacokinetics after administration of an OATP inhibitor rifampin (RIF) in healthy subjects. Rifampin 192-200 solute carrier organic anion transporter family member 1A2 Homo sapiens 177-181 27815731-11 2017 Rosuvastatin, BSP, rifampin, and glycyrrhizic acid all exhibited a certain extent inhibitory effect on the transport of OPD in OATP1B1*1a-HEK293T cells. Rifampin 19-27 solute carrier organic anion transporter family member 1B1 Homo sapiens 127-134 27858342-8 2017 CONCLUSION: Through quantitative assessment of the effect of OATP1B1 genetic polymorphism and inhibitors of transporters and metabolizing enyzmes via PBPK modeling, we confirmed the importance of OATP1B1 in the disposition of these two statins, and explored potential causes for under-prediction of the inhibitory effect of rifampin and cyclosporine. Rifampin 324-332 solute carrier organic anion transporter family member 1B1 Homo sapiens 61-68 27858342-8 2017 CONCLUSION: Through quantitative assessment of the effect of OATP1B1 genetic polymorphism and inhibitors of transporters and metabolizing enyzmes via PBPK modeling, we confirmed the importance of OATP1B1 in the disposition of these two statins, and explored potential causes for under-prediction of the inhibitory effect of rifampin and cyclosporine. Rifampin 324-332 solute carrier organic anion transporter family member 1B1 Homo sapiens 196-203 28371445-4 2017 We found that SG transport by OATP1B1 or murine Oatp1b2 was effectively inhibited by rifampin, and that this agent can significantly increase plasma levels of SG in wildtype mice, but not in Oatp1b2-deficient animals. Rifampin 85-93 solute carrier organic anion transporter family, member 1b2 Mus musculus 48-55 28546420-0 2017 Alterations of Histone Modifications Contribute to Pregnane X Receptor-Mediated Induction of CYP3A4 by Rifampicin. Rifampin 103-113 nuclear receptor subfamily 1 group I member 2 Homo sapiens 51-70 28546420-0 2017 Alterations of Histone Modifications Contribute to Pregnane X Receptor-Mediated Induction of CYP3A4 by Rifampicin. Rifampin 103-113 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-99 28546420-3 2017 This study aims to explore the role of histone modifications in rifampicin-induced expression of CYP3A4 in LS174T cells. Rifampin 64-74 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 97-103 28546420-4 2017 We found that the induction of CYP3A4 mRNA (4- to 15-fold) by rifampicin in LS174T cells was associated with increased levels of histone H3 lysine 4 trimethylation (H3K4me3, above 1.8-fold) and H3 acetylation (above 2-fold) and a decreased level of histone H3 lysine 27 trimethylation (H3K27me3, about 50%) in the CYP3A4 promoter. Rifampin 62-72 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 28546420-4 2017 We found that the induction of CYP3A4 mRNA (4- to 15-fold) by rifampicin in LS174T cells was associated with increased levels of histone H3 lysine 4 trimethylation (H3K4me3, above 1.8-fold) and H3 acetylation (above 2-fold) and a decreased level of histone H3 lysine 27 trimethylation (H3K27me3, about 50%) in the CYP3A4 promoter. Rifampin 62-72 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 314-320 28546420-5 2017 Rifampicin enhanced recruitment to the CYP3A4 promoter of nuclear receptor coactivator 6 (NCOA6, above 3-fold) and histone acetyltransferase p300 (p300, above 1.6-fold). Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 39-45 28546420-5 2017 Rifampicin enhanced recruitment to the CYP3A4 promoter of nuclear receptor coactivator 6 (NCOA6, above 3-fold) and histone acetyltransferase p300 (p300, above 1.6-fold). Rifampin 0-10 nuclear receptor coactivator 6 Homo sapiens 58-88 28546420-5 2017 Rifampicin enhanced recruitment to the CYP3A4 promoter of nuclear receptor coactivator 6 (NCOA6, above 3-fold) and histone acetyltransferase p300 (p300, above 1.6-fold). Rifampin 0-10 nuclear receptor coactivator 6 Homo sapiens 90-95 28546420-5 2017 Rifampicin enhanced recruitment to the CYP3A4 promoter of nuclear receptor coactivator 6 (NCOA6, above 3-fold) and histone acetyltransferase p300 (p300, above 1.6-fold). Rifampin 0-10 E1A binding protein p300 Homo sapiens 115-145 28546420-7 2017 Knockdown of pregnane X receptor (PXR) expression not only suppressed the recruitment of NCOA6 and p300 but also abolished the changes caused by rifampicin in H3K4me3, H3K27me3, and H3 acetylation levels in the CYP3A4 promoter. Rifampin 145-155 nuclear receptor subfamily 1 group I member 2 Homo sapiens 13-32 28546420-7 2017 Knockdown of pregnane X receptor (PXR) expression not only suppressed the recruitment of NCOA6 and p300 but also abolished the changes caused by rifampicin in H3K4me3, H3K27me3, and H3 acetylation levels in the CYP3A4 promoter. Rifampin 145-155 nuclear receptor subfamily 1 group I member 2 Homo sapiens 34-37 28546420-8 2017 Moreover, rifampicin treatment enhanced the nuclear accumulation and interactions between PXR and NCOA6/p300. Rifampin 10-20 nuclear receptor subfamily 1 group I member 2 Homo sapiens 90-93 28546420-8 2017 Moreover, rifampicin treatment enhanced the nuclear accumulation and interactions between PXR and NCOA6/p300. Rifampin 10-20 nuclear receptor coactivator 6 Homo sapiens 98-103 28546420-8 2017 Moreover, rifampicin treatment enhanced the nuclear accumulation and interactions between PXR and NCOA6/p300. Rifampin 10-20 E1A binding protein p300 Homo sapiens 104-108 28546420-9 2017 In conclusion, we show that the alterations of histone modifications contribute to the PXR-mediated induction of CYP3A4 by rifampicin. Rifampin 123-133 nuclear receptor subfamily 1 group I member 2 Homo sapiens 87-90 28546420-9 2017 In conclusion, we show that the alterations of histone modifications contribute to the PXR-mediated induction of CYP3A4 by rifampicin. Rifampin 123-133 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 113-119 28484975-3 2017 Rifampicin (OATP inhibitor; 600 mg, p.o.) Rifampin 0-10 solute carrier organic anion transporter family member 1A2 Homo sapiens 12-16 28403028-4 2017 RECENT FINDINGS: Dose adjustments of standard dose efavirenz are not necessary with rifampicin, because auto-induction of CYP2B6 by efavirenz counteracts the induction of rifampicin. Rifampin 171-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 28403028-5 2017 However, patients with extensive metabolizer CYP2B6 genotypes have lower efavirenz concentrations and may have less auto-induction of CYP2B6; the additive inducing effects of rifampicin on CYP2B6 could result in clinically significant reductions of efavirenz concentrations. Rifampin 175-185 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 28373111-0 2017 Pretreatment With Rifampicin and Tyrosine Kinase Inhibitor Dasatinib Potentiates the Inhibitory Effects Toward OATP1B1- and OATP1B3-Mediated Transport. Rifampin 18-28 solute carrier organic anion transporter family member 1B1 Homo sapiens 111-118 28373111-0 2017 Pretreatment With Rifampicin and Tyrosine Kinase Inhibitor Dasatinib Potentiates the Inhibitory Effects Toward OATP1B1- and OATP1B3-Mediated Transport. Rifampin 18-28 solute carrier organic anion transporter family member 1B3 Homo sapiens 124-131 28373111-1 2017 Present studies determined the effects of pretreatment with rifampicin, an organic anion-transporting polypeptide (OATP) inhibitor, and the tyrosine kinase inhibitor dasatinib on OATP1B1- and OATP1B3-mediated transport, and evaluated the OATP-mediated drug-drug interaction potential of dasatinib using the static R-value and dynamic physiologically based pharmacokinetic models. Rifampin 60-70 solute carrier organic anion transporter family member 1B1 Homo sapiens 179-186 28373111-1 2017 Present studies determined the effects of pretreatment with rifampicin, an organic anion-transporting polypeptide (OATP) inhibitor, and the tyrosine kinase inhibitor dasatinib on OATP1B1- and OATP1B3-mediated transport, and evaluated the OATP-mediated drug-drug interaction potential of dasatinib using the static R-value and dynamic physiologically based pharmacokinetic models. Rifampin 60-70 solute carrier organic anion transporter family member 1B3 Homo sapiens 192-199 28373111-1 2017 Present studies determined the effects of pretreatment with rifampicin, an organic anion-transporting polypeptide (OATP) inhibitor, and the tyrosine kinase inhibitor dasatinib on OATP1B1- and OATP1B3-mediated transport, and evaluated the OATP-mediated drug-drug interaction potential of dasatinib using the static R-value and dynamic physiologically based pharmacokinetic models. Rifampin 60-70 solute carrier organic anion transporter family member 1A2 Homo sapiens 179-183 28373111-2 2017 Rifampicin and dasatinib pretreatment significantly decreased OATP1B1- and OATP1B3-mediated transport. Rifampin 0-10 solute carrier organic anion transporter family member 1B1 Homo sapiens 62-69 28373111-2 2017 Rifampicin and dasatinib pretreatment significantly decreased OATP1B1- and OATP1B3-mediated transport. Rifampin 0-10 solute carrier organic anion transporter family member 1B3 Homo sapiens 75-82 28373111-4 2017 Present studies revealed that estrone-3-sulfate is a less-sensitive OATP1B1 substrate than estradiol-17beta-glucuronide in assessing rifampicin pretreatment effects. Rifampin 133-143 solute carrier organic anion transporter family member 1B1 Homo sapiens 68-75 28373111-5 2017 Pretreatment with rifampicin and dasatinib reduced the inhibition constant (Ki) values against OATP1B1 by 3 and 2.1 fold and against OATP1B3 by 2.4 and 2.1 fold, respectively. Rifampin 18-28 solute carrier organic anion transporter family member 1B1 Homo sapiens 95-102 28373111-5 2017 Pretreatment with rifampicin and dasatinib reduced the inhibition constant (Ki) values against OATP1B1 by 3 and 2.1 fold and against OATP1B3 by 2.4 and 2.1 fold, respectively. Rifampin 18-28 solute carrier organic anion transporter family member 1B3 Homo sapiens 133-140 28373111-9 2017 In summary, we report novel findings that pretreatment with rifampicin and dasatinib potentiates the inhibitory effects toward OATP1B1 and OATP1B3 without affecting plasma membrane levels of the transporters. Rifampin 60-70 solute carrier organic anion transporter family member 1B1 Homo sapiens 127-134 28373111-9 2017 In summary, we report novel findings that pretreatment with rifampicin and dasatinib potentiates the inhibitory effects toward OATP1B1 and OATP1B3 without affecting plasma membrane levels of the transporters. Rifampin 60-70 solute carrier organic anion transporter family member 1B3 Homo sapiens 139-146 28671602-10 2017 In the KEGG pathway enrichment-based clustering analysis, the peroxisome proliferator-activated receptor-gamma (PPARgamma) signaling pathway, cytochrome P450, glutathione metabolism, chemical carcinogenesis, and related proteins increased dose-dependently in rifampicin-treated livers. Rifampin 259-269 peroxisome proliferator activated receptor gamma Mus musculus 62-110 27784136-7 2017 Furthermore the Lnt mutant showed 64 fold and 16 fold increase in susceptibility to rifampicin and ciprofloxacin respectively. Rifampin 84-94 apolipoprotein N-acyltransferase Neisseria meningitidis MC58 16-19 28621455-0 2017 Addition of Rifampicin to Bolton Broth to Inhibit Extended-Spectrum beta-Lactamase-Producing Escherichia coli for the Detection of Campylobacter. Rifampin 12-22 beta-lactamase Escherichia coli 68-82 28199520-2 2017 : Xpert MTB/RIF (Xpert) detects rifampicin-resistant tuberculosis (RR-tuberculosis), enabling physicians to rapidly initiate a World Health Organization-recommended 5-drug regimen while awaiting second-line drug-susceptibility test (DST) results. Rifampin 32-42 ras homolog family member F, filopodia associated Homo sapiens 12-15 28904673-3 2017 The rpoB and katG genes molecular markers are used for detecting rifampicin and isoniazid resistance respectively. Rifampin 65-75 DNA-directed RNA polymerase subunit beta Mycobacterium tuberculosis H37Rv 4-8 28408657-0 2017 Negative Regulation of Human Pregnane X Receptor by MicroRNA-18a-5p: Evidence for Suppression of MicroRNA-18a-5p Expression by Rifampin and Rilpivirine. Rifampin 127-135 nuclear receptor subfamily 1 group I member 2 Homo sapiens 29-48 28408657-8 2017 The decrease in hPXR expression by the miR-18a-5p mimic was associated with a reduction in the extent of hPXR target gene (CYP3A4) induction by rifampin and rilpivirine. Rifampin 144-152 nuclear receptor subfamily 1 group I member 2 Homo sapiens 16-20 28408657-8 2017 The decrease in hPXR expression by the miR-18a-5p mimic was associated with a reduction in the extent of hPXR target gene (CYP3A4) induction by rifampin and rilpivirine. Rifampin 144-152 nuclear receptor subfamily 1 group I member 2 Homo sapiens 105-109 28408657-8 2017 The decrease in hPXR expression by the miR-18a-5p mimic was associated with a reduction in the extent of hPXR target gene (CYP3A4) induction by rifampin and rilpivirine. Rifampin 144-152 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 123-129 28408657-10 2017 In conclusion, miR-18a-5p is a negative regulator of hPXR expression and the hPXR agonists rifampin and rilpivirine are chemical suppressors of miR-18a-5p expression. Rifampin 91-99 nuclear receptor subfamily 1 group I member 2 Homo sapiens 77-81 28173639-0 2017 Bosentan and Rifampin Interactions Modulate Influx Transporter and Cytochrome P450 Expression and Activities in Primary Human Hepatocytes. Rifampin 13-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 28336091-4 2017 Further research showed that the CYP inducers omeprazole, ethanol, and rifampicin inhibited cell viability, in particular, ethanol, a CYP2E1 inducer, induced ROS generation, lipid peroxidation, and apoptosis in HepG2 cells treated with MC-LR. Rifampin 71-81 peptidylprolyl isomerase G Homo sapiens 33-36 28336091-4 2017 Further research showed that the CYP inducers omeprazole, ethanol, and rifampicin inhibited cell viability, in particular, ethanol, a CYP2E1 inducer, induced ROS generation, lipid peroxidation, and apoptosis in HepG2 cells treated with MC-LR. Rifampin 71-81 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 134-140 28594304-1 2017 AIM: We retrospectively evaluate the pharmacogenetic role of single nucleotide polymorphisms involved in rifampicin transport (SLCO1B1, MDR1 and PXR genes) and vitamin D (VDR, CYP24A1 and CYP27B1 genes) metabolism and activity on drug plasma and intracellular concentrations. Rifampin 105-115 solute carrier organic anion transporter family member 1B1 Homo sapiens 127-134 28594304-1 2017 AIM: We retrospectively evaluate the pharmacogenetic role of single nucleotide polymorphisms involved in rifampicin transport (SLCO1B1, MDR1 and PXR genes) and vitamin D (VDR, CYP24A1 and CYP27B1 genes) metabolism and activity on drug plasma and intracellular concentrations. Rifampin 105-115 nuclear receptor subfamily 1 group I member 2 Homo sapiens 145-148 28475796-13 2017 Discordant rifampicin susceptibility results of Xpert MTB/RIF and mycobacteria growth indicator tube (MGIT) were confirmed by using rpoB gene sequencing, which raised the sensitivity of Xpert MTB/RIF in detecting rifampicin resistance to 93.8%. Rifampin 11-21 metallothionein 1J, pseudogene Homo sapiens 54-57 28475796-13 2017 Discordant rifampicin susceptibility results of Xpert MTB/RIF and mycobacteria growth indicator tube (MGIT) were confirmed by using rpoB gene sequencing, which raised the sensitivity of Xpert MTB/RIF in detecting rifampicin resistance to 93.8%. Rifampin 11-21 metallothionein 1J, pseudogene Homo sapiens 192-195 28475796-13 2017 Discordant rifampicin susceptibility results of Xpert MTB/RIF and mycobacteria growth indicator tube (MGIT) were confirmed by using rpoB gene sequencing, which raised the sensitivity of Xpert MTB/RIF in detecting rifampicin resistance to 93.8%. Rifampin 213-223 metallothionein 1J, pseudogene Homo sapiens 192-195 28475796-14 2017 Conclusions: Xpert MTB/RIF is an effective tool in diagnosing PTB but will be more cost-effective for sputum-negative patients and in settings with high prevalence of rifampicin resistance. Rifampin 167-177 metallothionein 1J, pseudogene Homo sapiens 19-22 28575098-7 2017 Pretreatment of rifampin (10 mumol/L), the agonist of PXR alleviated the TBA secretion induced by estradiol (500 nmol/L, 48 h). Rifampin 16-24 nuclear receptor subfamily 1 group I member 2 Homo sapiens 54-57 28575098-9 2017 Both of miR-148a-siRNA and rifampin (10 mumol/L) inhibited the upregulated effect of estradiol on MRP3 expression. Rifampin 27-35 ATP binding cassette subfamily C member 3 Homo sapiens 98-102 28259787-6 2017 Rifampicin activated hPXR as expected, but not cPXR. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 21-25 28173639-3 2017 In this study, the effects of bosentan and rifampin on the expression and activities of organic anion-transporting peptide (OATP) and cytochrome P450 (CYP450) 2C9 and CYP3A4 were investigated in vitro. Rifampin 43-51 solute carrier organic anion transporter family member 1A2 Homo sapiens 88-122 28173639-3 2017 In this study, the effects of bosentan and rifampin on the expression and activities of organic anion-transporting peptide (OATP) and cytochrome P450 (CYP450) 2C9 and CYP3A4 were investigated in vitro. Rifampin 43-51 solute carrier organic anion transporter family member 1A2 Homo sapiens 124-128 28173639-3 2017 In this study, the effects of bosentan and rifampin on the expression and activities of organic anion-transporting peptide (OATP) and cytochrome P450 (CYP450) 2C9 and CYP3A4 were investigated in vitro. Rifampin 43-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-149 28173639-3 2017 In this study, the effects of bosentan and rifampin on the expression and activities of organic anion-transporting peptide (OATP) and cytochrome P450 (CYP450) 2C9 and CYP3A4 were investigated in vitro. Rifampin 43-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-157 28173639-3 2017 In this study, the effects of bosentan and rifampin on the expression and activities of organic anion-transporting peptide (OATP) and cytochrome P450 (CYP450) 2C9 and CYP3A4 were investigated in vitro. Rifampin 43-51 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 167-173 28173639-4 2017 HEK293 cells and primary human hepatocytes overexpressing the target genes were treated with bosentan and various concentrations of rifampin, which decreased the uptake activities of OATP transporters in a dose-dependent manner. Rifampin 132-140 solute carrier organic anion transporter family member 1A2 Homo sapiens 183-187 28173639-5 2017 In primary human hepatocytes, CYP2C9 and CYP3A4 gene expression and activities decreased upon treatment with 20 muM bosentan+200 muM rifampin. Rifampin 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 28173639-5 2017 In primary human hepatocytes, CYP2C9 and CYP3A4 gene expression and activities decreased upon treatment with 20 muM bosentan+200 muM rifampin. Rifampin 133-141 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-47 28173639-7 2017 These results confirm rifampin- and bosentan-induced interactions between OATP transporters and CYP450. Rifampin 22-30 solute carrier organic anion transporter family member 1A2 Homo sapiens 74-78 28173639-7 2017 These results confirm rifampin- and bosentan-induced interactions between OATP transporters and CYP450. Rifampin 22-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-102 28254951-3 2017 At the highest tested concentration of 300 muM, PF-06282999 caused maximal induction in CYP3A4 mRNA and enzyme activity ranging from 56% to 86% and 47% t0 72%, respectively, of rifampicin response across the three hepatocyte donor pools. Rifampin 177-187 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-94 27545757-5 2017 Studies NP28990 and NP29042 enrolled 17 and 24 healthy subjects, respectively, and investigated potent CYP3A inhibition with posaconazole and potent CYP3A induction through rifampin, respectively, on the single oral dose pharmacokinetics (PK) of alectinib. Rifampin 173-181 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 149-154 28157069-0 2017 Delayed de-induction of CYP2C9 compared to CYP3A after discontinuation of rifampicin: Report of two cases : . Rifampin 74-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 28157069-1 2017 OBJECTIVE: Timely dose reduction of concomitant medications is important after withdrawal of rifampicin, a CYP inducer. Rifampin 93-103 peptidylprolyl isomerase G Homo sapiens 107-110 28157069-3 2017 To clarify the time courses of deinduction of CYP2C9 and -CYP3A activities after rifampicin withdrawal, we monitored these enzyme activities in 2 patients over time after discontinuing rifampicin. Rifampin 81-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 28157069-3 2017 To clarify the time courses of deinduction of CYP2C9 and -CYP3A activities after rifampicin withdrawal, we monitored these enzyme activities in 2 patients over time after discontinuing rifampicin. Rifampin 81-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 58-63 28157069-3 2017 To clarify the time courses of deinduction of CYP2C9 and -CYP3A activities after rifampicin withdrawal, we monitored these enzyme activities in 2 patients over time after discontinuing rifampicin. Rifampin 185-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 28157069-5 2017 Warfarin doses were increased due to rifampicin-induced CYP activity. Rifampin 37-47 peptidylprolyl isomerase G Homo sapiens 56-59 28157069-9 2017 CONCLUSION: Our findings suggest that a uniform dose reduction protocol for drugs metabolized by different CYP isoforms may be unsafe after rifampicin withdrawal. Rifampin 140-150 peptidylprolyl isomerase G Homo sapiens 107-110 28329820-5 2017 In HepaRG cells, cytotoxicity started at 20 and 100 microM for posaconazole and ketoconazole, respectively, and was slightly accentuated by cytochrome P450 3A4 induction with rifampicin and 1A2 with 3-methylcholantrene. Rifampin 175-185 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 140-159 28025853-12 2017 CONCLUSIONS/CLINICAL IMPORTANCE: Oral rifampicin combined with topical antimicrobials can be considered an effective therapeutic option for canine superficial and deep pyoderma caused by rifampicin-susceptible multidrug-resistant MRS. Liver enzyme induction might be the most important cause of ALT and ALP increase associated with rifampicin therapy in dogs. Rifampin 38-48 alkaline phosphatase, biomineralization associated Canis lupus familiaris 303-306 28025853-12 2017 CONCLUSIONS/CLINICAL IMPORTANCE: Oral rifampicin combined with topical antimicrobials can be considered an effective therapeutic option for canine superficial and deep pyoderma caused by rifampicin-susceptible multidrug-resistant MRS. Liver enzyme induction might be the most important cause of ALT and ALP increase associated with rifampicin therapy in dogs. Rifampin 187-197 alkaline phosphatase, biomineralization associated Canis lupus familiaris 303-306 28025853-12 2017 CONCLUSIONS/CLINICAL IMPORTANCE: Oral rifampicin combined with topical antimicrobials can be considered an effective therapeutic option for canine superficial and deep pyoderma caused by rifampicin-susceptible multidrug-resistant MRS. Liver enzyme induction might be the most important cause of ALT and ALP increase associated with rifampicin therapy in dogs. Rifampin 187-197 alkaline phosphatase, biomineralization associated Canis lupus familiaris 303-306 28215634-8 2017 We have confirmed that the hepatocyte-like cells prepared by our methods were able to increase gene expression of cytochrome P450 enzymes upon encountering rifampicin, phenobarbital, or omeprazole. Rifampin 156-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-129 28263460-3 2017 Asunaprevir plasma concentrations are markedly increased by single-dose rifampin (an OATP inhibitor) and in subjects with moderate to severe hepatic impairment. Rifampin 72-80 solute carrier organic anion transporter family member 1A2 Homo sapiens 85-89 28253382-0 2017 Kinetic and thermodynamic study of bovine serum albumin interaction with rifampicin using surface plasmon resonance and molecular docking methods. Rifampin 73-83 albumin Homo sapiens 42-55 28077325-0 2017 Role of 14-3-3 sigma in over-expression of P-gp by rifampin and paclitaxel stimulation through interaction with PXR. Rifampin 51-59 stratifin Homo sapiens 8-20 28077325-0 2017 Role of 14-3-3 sigma in over-expression of P-gp by rifampin and paclitaxel stimulation through interaction with PXR. Rifampin 51-59 ATP binding cassette subfamily B member 1 Homo sapiens 43-47 28077325-1 2017 In this study, we presented the role of 14-3-3sigma to activate CK2-Hsp90beta-PXR-MDR1 pathway on rifampin and paclitaxel treated LS174T cells and in vivo LS174T cell-xenografted nude mouse model. Rifampin 98-106 stratifin Homo sapiens 40-51 28077325-1 2017 In this study, we presented the role of 14-3-3sigma to activate CK2-Hsp90beta-PXR-MDR1 pathway on rifampin and paclitaxel treated LS174T cells and in vivo LS174T cell-xenografted nude mouse model. Rifampin 98-106 heat shock protein 90 alpha family class B member 1 Homo sapiens 68-77 28077325-1 2017 In this study, we presented the role of 14-3-3sigma to activate CK2-Hsp90beta-PXR-MDR1 pathway on rifampin and paclitaxel treated LS174T cells and in vivo LS174T cell-xenografted nude mouse model. Rifampin 98-106 nuclear receptor subfamily 1 group I member 2 Homo sapiens 78-81 28077325-1 2017 In this study, we presented the role of 14-3-3sigma to activate CK2-Hsp90beta-PXR-MDR1 pathway on rifampin and paclitaxel treated LS174T cells and in vivo LS174T cell-xenografted nude mouse model. Rifampin 98-106 ATP binding cassette subfamily B member 1 Homo sapiens 82-86 28077325-2 2017 Following several in vitro and in vivo experiments, rifampin and paclitaxel were found to be stimulated the CK2-Hsp90beta-PXR-MDR1 pathway. Rifampin 52-60 heat shock protein 90 alpha family class B member 1 Homo sapiens 112-121 28077325-2 2017 Following several in vitro and in vivo experiments, rifampin and paclitaxel were found to be stimulated the CK2-Hsp90beta-PXR-MDR1 pathway. Rifampin 52-60 nuclear receptor subfamily 1 group I member 2 Homo sapiens 122-125 28077325-2 2017 Following several in vitro and in vivo experiments, rifampin and paclitaxel were found to be stimulated the CK2-Hsp90beta-PXR-MDR1 pathway. Rifampin 52-60 ATP binding cassette subfamily B member 1 Homo sapiens 126-130 28077325-4 2017 We constructed FLAG-PXR-LS174T stable cell lines and discovered 22 proteins that interacted with PXR on rifampin treatment. Rifampin 104-112 nuclear receptor subfamily 1 group I member 2 Homo sapiens 20-23 28077325-4 2017 We constructed FLAG-PXR-LS174T stable cell lines and discovered 22 proteins that interacted with PXR on rifampin treatment. Rifampin 104-112 nuclear receptor subfamily 1 group I member 2 Homo sapiens 97-100 28077325-7 2017 On the other hand, PXR was found to be localized in nucleus after rifampin and paclitaxel treatment by using cell fractionation assay. Rifampin 66-74 nuclear receptor subfamily 1 group I member 2 Homo sapiens 19-22 28077325-10 2017 P-gp overexpression was induced only when 14-3-3sigma transfected LS174T cells were treated with rifampin and paclitaxel, whereas 14-3-3sigma inhibition by nonpeptidic inhibitor, BV02 and 14-3-3sigma siRNA reduced rifampin induced PXR and P-gp expression. Rifampin 97-105 ATP binding cassette subfamily B member 1 Homo sapiens 0-4 28077325-10 2017 P-gp overexpression was induced only when 14-3-3sigma transfected LS174T cells were treated with rifampin and paclitaxel, whereas 14-3-3sigma inhibition by nonpeptidic inhibitor, BV02 and 14-3-3sigma siRNA reduced rifampin induced PXR and P-gp expression. Rifampin 97-105 stratifin Homo sapiens 42-53 29359155-0 2017 The Feasibility of Xpert MTB/RIF Testing to Detect Rifampicin Resistance among Childhood Tuberculosis for Prevalence Surveys in Northern China. Rifampin 51-61 ras homolog family member F, filopodia associated Homo sapiens 29-32 29359155-3 2017 The goal of this work was to generate prevalence data regarding rifampicin- (RIF-) resistant childhood TB in northern China and to test the feasibility of Xpert for surveying pediatric TB drug resistance prevalence. Rifampin 64-74 ras homolog family member F, filopodia associated Homo sapiens 77-80 27273461-0 2017 Pharmacokinetic Evaluation of CYP3A4-Mediated Drug-Drug Interactions of Isavuconazole With Rifampin, Ketoconazole, Midazolam, and Ethinyl Estradiol/Norethindrone in Healthy Adults. Rifampin 91-99 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 30-36 27273461-2 2017 Coadministration of oral isavuconazole (100 mg once daily) with oral rifampin (600 mg once daily; CYP3A4 inducer) decreased isavuconazole area under the concentration-time curve (AUCtau ) during a dosing interval by 90% and maximum concentration (Cmax ) by 75%. Rifampin 69-77 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 98-104 28317812-3 2017 The new polymerase chain reaction-based XpertMTB/Rif (Cepheid Inc., CA, USA), which detects Mycobacterium tuberculosis and rifampicin resistance, was introduced in Cross River State in 2014. Rifampin 123-133 ras homolog family member F, filopodia associated Homo sapiens 49-52 27321774-1 2017 This open-label 2-period study assessed the effect of multiple-dose administration of rifampin, a strong cytochrome P450 3A (CYP3A) and P-glycoprotein inducer, on the pharmacokinetics of odanacatib, a cathepsin K inhibitor. Rifampin 86-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-123 27321774-1 2017 This open-label 2-period study assessed the effect of multiple-dose administration of rifampin, a strong cytochrome P450 3A (CYP3A) and P-glycoprotein inducer, on the pharmacokinetics of odanacatib, a cathepsin K inhibitor. Rifampin 86-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 125-130 27321774-1 2017 This open-label 2-period study assessed the effect of multiple-dose administration of rifampin, a strong cytochrome P450 3A (CYP3A) and P-glycoprotein inducer, on the pharmacokinetics of odanacatib, a cathepsin K inhibitor. Rifampin 86-94 cathepsin K Homo sapiens 201-212 28496040-6 2017 Thalidomide significantly induced P450 3A4/5, 2B6, and pregnane X receptor (PXR) mRNA levels 2 to 3-fold, but rifampicin only enhanced P450 3A5 and PXR mRNA under the modified media conditions. Rifampin 110-120 nuclear receptor subfamily 1 group I member 2 Homo sapiens 148-151 27774592-5 2016 Furthermore, rifampicin dramatically reduced the disruption of blood-brain barrier integrity, down-regulated serum concentration of IL-6 and IL-17A, inhibited pathological Th17 cell differentiation, and modulated the expression of p-STAT3 and p-p65. Rifampin 13-23 interleukin 6 Mus musculus 132-136 27774592-5 2016 Furthermore, rifampicin dramatically reduced the disruption of blood-brain barrier integrity, down-regulated serum concentration of IL-6 and IL-17A, inhibited pathological Th17 cell differentiation, and modulated the expression of p-STAT3 and p-p65. Rifampin 13-23 interleukin 17A Mus musculus 141-147 27774592-5 2016 Furthermore, rifampicin dramatically reduced the disruption of blood-brain barrier integrity, down-regulated serum concentration of IL-6 and IL-17A, inhibited pathological Th17 cell differentiation, and modulated the expression of p-STAT3 and p-p65. Rifampin 13-23 lymphocyte cytosolic protein 1 Mus musculus 243-248 27774592-6 2016 These results suggest that rifampicin is effective for attenuating the clinical severity of EAE mice, which may be related to its inhibitive ability in differentiation of Th17 cell and secretion of its key effector molecule IL-17A via regulation of excessive activation of the key signaling molecules of JAK/STAT pathway. Rifampin 27-37 interleukin 17A Mus musculus 224-230 27965540-0 2016 Rifampicin Attenuated Global Cerebral Ischemia Injury via Activating the Nuclear Factor Erythroid 2-Related Factor Pathway. Rifampin 0-10 nuclear factor, erythroid 2 Rattus norvegicus 73-99 27965540-4 2016 In this study, we examined whether rifampicin exhibits beneficial effects mediated by the Nrf2 pathway after global cerebral ischemia (GCI). Rifampin 35-45 NFE2 like bZIP transcription factor 2 Rattus norvegicus 90-94 27965540-11 2016 In addition, rifampicin significantly elevated the nuclear expression of Nrf2, Nrf2 downstream anti-oxidant protein, HO-1 compared with the control group, and it simultaneously downregulated the expression of COX-2 in the hippocampus on day 3 after ischemia-reperfusion. Rifampin 13-23 NFE2 like bZIP transcription factor 2 Rattus norvegicus 73-77 27965540-11 2016 In addition, rifampicin significantly elevated the nuclear expression of Nrf2, Nrf2 downstream anti-oxidant protein, HO-1 compared with the control group, and it simultaneously downregulated the expression of COX-2 in the hippocampus on day 3 after ischemia-reperfusion. Rifampin 13-23 NFE2 like bZIP transcription factor 2 Rattus norvegicus 79-83 27965540-11 2016 In addition, rifampicin significantly elevated the nuclear expression of Nrf2, Nrf2 downstream anti-oxidant protein, HO-1 compared with the control group, and it simultaneously downregulated the expression of COX-2 in the hippocampus on day 3 after ischemia-reperfusion. Rifampin 13-23 prostaglandin-endoperoxide synthase 2 Rattus norvegicus 209-214 27965540-12 2016 Interestingly, the forenamed effects of rifampicin were abolished by pretreatment with brusatol, a specific inhibitor of Nrf2 activation. Rifampin 40-50 NFE2 like bZIP transcription factor 2 Rattus norvegicus 121-125 27965540-13 2016 Conclusions: Rifampicin exerts neuroprotective effects against global cerebral ischemia, which may be attributed to activation of the Nrf2 pathway. Rifampin 13-23 NFE2 like bZIP transcription factor 2 Rattus norvegicus 134-138 27856484-3 2016 Large-scale clinical trials with single dose rifampicin (SDR) given as post-exposure prophylaxis (PEP) to contacts of newly diagnosed patients with leprosy have shown a 50-60% reduction of the risk of developing leprosy over the following 2 years. Rifampin 45-55 prolyl endopeptidase Homo sapiens 98-101 27603548-3 2016 In the present study, the effects of cyclosporine A and rifampicin on recombinant cynomolgus monkey OATP-mediated pitavastatin uptake were investigated in pre- and simultaneous incubation systems. Rifampin 56-66 solute carrier organic anion transporter family member 1A2 Homo sapiens 100-104 27538915-0 2016 Rifampin-Mediated Induction of Tamoxifen Metabolism in a Humanized PXR-CAR-CYP3A4/3A7-CYP2D6 Mouse Model. Rifampin 0-8 nuclear receptor subfamily 1 group I member 2 Homo sapiens 67-70 27538915-0 2016 Rifampin-Mediated Induction of Tamoxifen Metabolism in a Humanized PXR-CAR-CYP3A4/3A7-CYP2D6 Mouse Model. Rifampin 0-8 nuclear receptor subfamily 1, group I, member 3 Mus musculus 71-74 27538915-0 2016 Rifampin-Mediated Induction of Tamoxifen Metabolism in a Humanized PXR-CAR-CYP3A4/3A7-CYP2D6 Mouse Model. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 75-81 27538915-0 2016 Rifampin-Mediated Induction of Tamoxifen Metabolism in a Humanized PXR-CAR-CYP3A4/3A7-CYP2D6 Mouse Model. Rifampin 0-8 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 86-92 27538915-3 2016 In humans, tamoxifen is metabolized primarily by CYP3A4 and CYP2D6, and multiple-day treatment with rifampin decreased tamoxifen exposure by 6.2-fold. Rifampin 100-108 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 49-55 27538915-3 2016 In humans, tamoxifen is metabolized primarily by CYP3A4 and CYP2D6, and multiple-day treatment with rifampin decreased tamoxifen exposure by 6.2-fold. Rifampin 100-108 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 60-66 27538915-6 2016 In the humanized PXR-CAR (hPXR-CAR) model, rifampin decreased AUC0-8 of tamoxifen and its metabolites by approximately 2-fold. Rifampin 43-51 nuclear receptor subfamily 1 group I member 2 Homo sapiens 17-20 27538915-6 2016 In the humanized PXR-CAR (hPXR-CAR) model, rifampin decreased AUC0-8 of tamoxifen and its metabolites by approximately 2-fold. Rifampin 43-51 nuclear receptor subfamily 1, group I, member 3 Mus musculus 21-24 27538915-6 2016 In the humanized PXR-CAR (hPXR-CAR) model, rifampin decreased AUC0-8 of tamoxifen and its metabolites by approximately 2-fold. Rifampin 43-51 nuclear receptor subfamily 1, group I, member 3 Mus musculus 31-34 27538915-8 2016 In vitro kinetics determined in microsomes prepared from livers of the Tg-composite animals showed that, although Km values were not different between vehicle- and rifampin-treated groups, rifampin increased the Vmax for the CYP3A4-mediated pathways. Rifampin 189-197 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 225-231 27538915-9 2016 These data demonstrate that, although the hPXR-CAR model is responsive to rifampin, the extent of the clinical rifampin-tamoxifen interaction is better represented by the Tg-composite model. Rifampin 74-82 nuclear receptor subfamily 1, group I, member 3 Mus musculus 47-50 27494923-0 2016 Molecular basis of rifampicin resistance in multiresistant porcine livestock-associated MRSA. Rifampin 19-29 solute carrier family 9 member A6 Homo sapiens 88-92 27565568-11 2016 Finally, the validated assay was applied to measure 4-hydroxycholesterol and cholesterol in serum samples of clinical studies in humans and foals that could verify induction of hepatic CYP3A4 (human) and CYP3A89 (foals) after premedication with the known enzyme inducer rifampicin. Rifampin 270-280 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 185-191 27745744-13 2016 IMPLICATIONS: Exposure to olaparib was significantly increased when co-administered with the potent CYP3A4 inhibitor itraconazole, and significantly decreased when co-administered with the potent CYP3A4 inducer rifampin, compared with olaparib alone. Rifampin 211-219 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 196-202 27567379-1 2016 Pregnane X receptor (PXR) is involved in the transactivation of ABCB1 gene by rifampicin (RIF). Rifampin 78-88 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-19 27567379-1 2016 Pregnane X receptor (PXR) is involved in the transactivation of ABCB1 gene by rifampicin (RIF). Rifampin 78-88 nuclear receptor subfamily 1 group I member 2 Homo sapiens 21-24 27567379-1 2016 Pregnane X receptor (PXR) is involved in the transactivation of ABCB1 gene by rifampicin (RIF). Rifampin 78-88 ATP binding cassette subfamily B member 1 Homo sapiens 64-69 27567379-1 2016 Pregnane X receptor (PXR) is involved in the transactivation of ABCB1 gene by rifampicin (RIF). Rifampin 90-93 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-19 27567379-1 2016 Pregnane X receptor (PXR) is involved in the transactivation of ABCB1 gene by rifampicin (RIF). Rifampin 90-93 nuclear receptor subfamily 1 group I member 2 Homo sapiens 21-24 27567379-1 2016 Pregnane X receptor (PXR) is involved in the transactivation of ABCB1 gene by rifampicin (RIF). Rifampin 90-93 ATP binding cassette subfamily B member 1 Homo sapiens 64-69 26320398-11 2016 Compared with vancomycin-susceptible S. aureus, hVISA and VISA isolates were less susceptible to ciprofloxacin, clindamycin, daptomycin, gentamicin, rifampin, and trimethoprim/sulfamethoxazole, and are thus, more likely to have SCCmec II or III element. Rifampin 149-157 mitochondrial antiviral signaling protein Homo sapiens 48-53 27458223-2 2016 Three studies were conducted to evaluate the potential drug-drug interactions between delamanid and antiretroviral drugs, including ritonavir, a strong inhibitor of CYP3A4, and selected anti-TB drugs, including rifampin, a strong inducer of cytochrome P450 (CYP) isozymes. Rifampin 211-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 241-256 27713827-4 2016 The selective PXR agonist rifampicin, a selective CAR activator, 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime (CITCO), and dual activators of CAR and PXR including phenobarbital (PB) were analyzed. Rifampin 26-36 nuclear receptor subfamily 1 group I member 2 Homo sapiens 14-17 27041449-6 2016 Rifampicin and St. John"s wort, the prototypical human PXR agonists, impair glucose tolerance in healthy volunteers. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 55-58 26724677-8 2016 Furthermore, catalytic activities of cytochrome P450 3A4 were modified by omeprazole and rifampicin in the 3D-cultured HepaRG spheroids. Rifampin 89-99 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-56 27795624-2 2016 Rifampicin, a first-line antitubercular therapy (ATT) drug, is a potent inducer of hepatic cytochrome P450 (CYP). Rifampin 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-106 27795624-2 2016 Rifampicin, a first-line antitubercular therapy (ATT) drug, is a potent inducer of hepatic cytochrome P450 (CYP). Rifampin 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 27795624-3 2016 There is potential for pharmacokinetic interaction between rifampicin and anti-hypertensives that are CYP substrates: amlodipine and metoprolol. Rifampin 59-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 27510251-1 2016 OBJECTIVE: To determine the effect of SLCO1B1 gene polymorphisms (rs11045819, rs4149032 and rs4149033) on rifampicin (RMP) concentrations in adult tuberculosis (TB) patients from south India. Rifampin 106-116 solute carrier organic anion transporter family member 1B1 Homo sapiens 38-45 27510251-1 2016 OBJECTIVE: To determine the effect of SLCO1B1 gene polymorphisms (rs11045819, rs4149032 and rs4149033) on rifampicin (RMP) concentrations in adult tuberculosis (TB) patients from south India. Rifampin 118-121 solute carrier organic anion transporter family member 1B1 Homo sapiens 38-45 27631193-2 2016 SLCO1B1 genetic variation is associated, for example, with highly variable rifampicin exposure, thus influencing the cornerstone antituberculosis therapy, especially in sub-Saharan Africa where it is a key therapeutic modality. Rifampin 75-85 solute carrier organic anion transporter family member 1B1 Homo sapiens 0-7 27631194-7 2016 Well-characterized haplotypes, *5 (RS4149056C) and *15 (RS4149056T), have been associated with a strikingly reduced uptake of multiple OATP1B1 substrates, including estrone-3-sulfate, estradiol-17beta-d-glucuronide, atorvastatin, cerivastatin, pravastatin, and rifampicin. Rifampin 261-271 solute carrier organic anion transporter family member 1B1 Homo sapiens 135-142 27557147-3 2016 Rifampin induces gene expression, at least in part, by activating the pregnane X receptor (PXR), which induces gene expression; however, the impact of rifampin on global gene regulation has not been examined under the molecular network frameworks. Rifampin 0-8 nuclear receptor subfamily 1 group I member 2 Homo sapiens 70-89 27557147-3 2016 Rifampin induces gene expression, at least in part, by activating the pregnane X receptor (PXR), which induces gene expression; however, the impact of rifampin on global gene regulation has not been examined under the molecular network frameworks. Rifampin 0-8 nuclear receptor subfamily 1 group I member 2 Homo sapiens 91-94 27197997-6 2016 Activation of CAR and PXR in cells treated with a high dose of CITCO [6-(4-chlorophenyl)-imidazo(2,1-b)thiazole-5-carbaldehyde] or cotreated with rifampicin and tetracycline resulted in synergistic enhancement of CYP3A4, but not CYP2B6, CYP2C9, or UGT1A1, mRNA expression in HepTR/hCAR/hPXR cells. Rifampin 146-156 nuclear receptor subfamily 1 group I member 3 Homo sapiens 14-17 27197997-6 2016 Activation of CAR and PXR in cells treated with a high dose of CITCO [6-(4-chlorophenyl)-imidazo(2,1-b)thiazole-5-carbaldehyde] or cotreated with rifampicin and tetracycline resulted in synergistic enhancement of CYP3A4, but not CYP2B6, CYP2C9, or UGT1A1, mRNA expression in HepTR/hCAR/hPXR cells. Rifampin 146-156 nuclear receptor subfamily 1 group I member 2 Homo sapiens 22-25 27105861-0 2016 Pharmacokinetic drug interactions of the selective androgen receptor modulator GTx-024(Enobosarm) with itraconazole, rifampin, probenecid, celecoxib and rosuvastatin. Rifampin 117-125 androgen receptor Homo sapiens 51-68 27155371-7 2016 Relevant drugs described to up-regulate intestinal MRP2 via PXR are rifampicin, spironolactone and carbamazepine, among others. Rifampin 68-78 ATP binding cassette subfamily C member 2 Homo sapiens 51-55 27155371-7 2016 Relevant drugs described to up-regulate intestinal MRP2 via PXR are rifampicin, spironolactone and carbamazepine, among others. Rifampin 68-78 nuclear receptor subfamily 1 group I member 2 Homo sapiens 60-63 27358806-0 2016 Diagnosis and treatment of TB patients with rifampicin resistance detected using Xpert( ) MTB/RIF in Zimbabwe. Rifampin 44-54 ras homolog family member F, filopodia associated Homo sapiens 94-97 27358806-1 2016 SETTING: In Zimbabwe, there are concerns about the management of tuberculosis (TB) patients with rifampicin (RMP) resistance diagnosed using Xpert( ) MTB/RIF. Rifampin 97-107 ras homolog family member F, filopodia associated Homo sapiens 154-157 27358806-1 2016 SETTING: In Zimbabwe, there are concerns about the management of tuberculosis (TB) patients with rifampicin (RMP) resistance diagnosed using Xpert( ) MTB/RIF. Rifampin 109-112 ras homolog family member F, filopodia associated Homo sapiens 154-157 27297123-8 2016 The competitive inhibitor of NQO1 dicoumarol synergized with rifampin to promote intracellular killing of mycobacteria. Rifampin 61-69 NAD(P)H quinone dehydrogenase 1 Homo sapiens 29-33 27098526-1 2016 Since many drugs are cytochrome P450 (CYP)-3A4 substrates, it has become common practice to assess drug-drug interaction (DDI) potential with a CYP3A4 inhibitor (ketoconazole) or inducer (rifampicin) in early drug development. Rifampin 188-198 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 144-150 27098526-5 2016 We hypothesized that DDIs between anticancer drugs and rifampicin were primarily driven by CYP3A4 induction. Rifampin 55-65 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 91-97 27098526-9 2016 Given the complexity and paradoxical effects arising with DDIs with rifampicin, the continued preference for rifampicin as CYP3A4 inducer needs immediate re-appraisal. Rifampin 109-119 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 123-129 28302396-12 2017 The SAS co-precipitation process using a mixture of ethyl acetate/dimethyl sulfoxide demonstrates that this strategy can be successful for controlling rifampicin delivery. Rifampin 151-161 tetraspanin 31 Homo sapiens 4-7 27918128-4 2017 It was found that Mul A significantly suppressed PXR-mediated P-gp luciferase activity induced by rifampicin (Rif). Rifampin 98-108 nuclear receptor subfamily 1 group I member 2 Homo sapiens 49-52 27918128-4 2017 It was found that Mul A significantly suppressed PXR-mediated P-gp luciferase activity induced by rifampicin (Rif). Rifampin 98-108 ATP binding cassette subfamily B member 1 Homo sapiens 62-66 27918128-4 2017 It was found that Mul A significantly suppressed PXR-mediated P-gp luciferase activity induced by rifampicin (Rif). Rifampin 110-113 nuclear receptor subfamily 1 group I member 2 Homo sapiens 49-52 27918128-4 2017 It was found that Mul A significantly suppressed PXR-mediated P-gp luciferase activity induced by rifampicin (Rif). Rifampin 110-113 ATP binding cassette subfamily B member 1 Homo sapiens 62-66 28324001-0 2017 CYP3A4 Induction by Rifampin: An Alternative Pathway for Vitamin D Inactivation in Patients With CYP24A1 Mutations. Rifampin 20-28 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 28324001-0 2017 CYP3A4 Induction by Rifampin: An Alternative Pathway for Vitamin D Inactivation in Patients With CYP24A1 Mutations. Rifampin 20-28 cytochrome P450 family 24 subfamily A member 1 Homo sapiens 97-104 28324001-5 2017 Objective: Our goal was to determine if rifampin, a potent inducer of CYP3A4, can normalize mineral metabolism in patients with IIH due to mutations in CYP24A1. Rifampin 40-48 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 70-76 28324001-5 2017 Objective: Our goal was to determine if rifampin, a potent inducer of CYP3A4, can normalize mineral metabolism in patients with IIH due to mutations in CYP24A1. Rifampin 40-48 cytochrome P450 family 24 subfamily A member 1 Homo sapiens 152-159 28324001-10 2017 Conclusion: These observations suggest that rifampin-induced overexpression of CYP3A4 provides an alternative pathway for inactivation of vitamin D metabolites in patients who lack CYP24A1 function. Rifampin 44-52 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 79-85 28324001-10 2017 Conclusion: These observations suggest that rifampin-induced overexpression of CYP3A4 provides an alternative pathway for inactivation of vitamin D metabolites in patients who lack CYP24A1 function. Rifampin 44-52 cytochrome P450 family 24 subfamily A member 1 Homo sapiens 181-188 28345929-6 2017 As an assessment of CYP3A4 induction, following rifampin pretreatment to PXR-CAR-3A4/3A7Hep/Int mice, an 8% decrease in vandetanib mean AUC was observed; 39-52% reduction in AUC were observed for dasatinib, ibrutinib, regorafenib, and sorafenib compared to vehicle treated mice. Rifampin 48-56 nuclear receptor subfamily 1, group I, member 2 Mus musculus 73-76 28879160-1 2017 BACKGROUND: Xpert MTB/RIF is a molecular test for the detection of Mycobacterium tuberculosis and rifampicin resistance. Rifampin 99-109 ras homolog family member F, filopodia associated Homo sapiens 23-26 28879160-10 2017 Xpert MTB/RIF correctly identified 185/186 (99.5%) rifampicin-sensitive and 2/2 (100%) rifampicin-resistant M. tuberculosis strains. Rifampin 51-61 ras homolog family member F, filopodia associated Homo sapiens 10-13 28879160-10 2017 Xpert MTB/RIF correctly identified 185/186 (99.5%) rifampicin-sensitive and 2/2 (100%) rifampicin-resistant M. tuberculosis strains. Rifampin 87-97 ras homolog family member F, filopodia associated Homo sapiens 10-13 28879160-11 2017 Mutations were not detected by sequencing in one isolate which was rifampicin resistant on Xpert MTB/RIF but sensitive on MTBDRplus. Rifampin 67-77 ras homolog family member F, filopodia associated Homo sapiens 101-104 28132806-0 2017 Rifampicin pre-treatment inhibits the toxicity of rotenone-induced PC12 cells by enhancing sumoylation modification of alpha-synuclein. Rifampin 0-10 synuclein alpha Rattus norvegicus 119-134 28132806-1 2017 Our previous research revealed that rifampicin could protect PC12 (pheochromocytoma 12) cells from rotenone-induced cytotoxicity by reversing the aggregation of alpha-synuclein. Rifampin 36-46 synuclein alpha Rattus norvegicus 161-176 28132806-3 2017 Here, we investigated whether rifampicin could stabilize alpha-synuclein and prevent rotenone-induced PC12 cells from undergoing apoptosis by enhancing SUMOylation of alpha-synuclein. Rifampin 30-40 synuclein alpha Rattus norvegicus 57-72 28132806-3 2017 Here, we investigated whether rifampicin could stabilize alpha-synuclein and prevent rotenone-induced PC12 cells from undergoing apoptosis by enhancing SUMOylation of alpha-synuclein. Rifampin 30-40 synuclein alpha Rattus norvegicus 167-182 28132806-8 2017 Treatment with 150 mumol/L rifampicin, increased the expressions of SUMO1 and SUMO2/3 in cells by 1.5 times compared with the control group; meanwhile, the cell viability of rotenone-induced cells increased from 20 to 80% (P < 0.05). Rifampin 27-37 small ubiquitin-like modifier 1 Rattus norvegicus 68-73 28132806-8 2017 Treatment with 150 mumol/L rifampicin, increased the expressions of SUMO1 and SUMO2/3 in cells by 1.5 times compared with the control group; meanwhile, the cell viability of rotenone-induced cells increased from 20 to 80% (P < 0.05). Rifampin 27-37 small ubiquitin-like modifier 2 Rattus norvegicus 78-83 28132806-10 2017 SUMOylation of alpha-synuclein was more significant in rifampicin-treated cells and Ubc9 upregulated cells. Rifampin 55-65 synuclein alpha Rattus norvegicus 15-30 28132806-12 2017 In conclusion, rifampicin might reduce the cytotoxicity of rotenone-induced PC12 cells by promoting SUMOylation of alpha-synuclein. Rifampin 15-25 synuclein alpha Rattus norvegicus 115-130 28469522-2 2017 In vivo chemical inhibition of P-gp and Oatp was achieved using elacridar and rifampicin, respectively. Rifampin 78-88 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 31-35 27174018-2 2017 Plasma concentrations of curcumin-O-glucuronide (COG) and curcumin-O-sulfate (COS) significantly increased after Sprague-Dawley rats dealt with the Oatp inhibitor rifampicin, with the Cmax ascending 2.9 and 6.7 times, and the AUC0- ascending 4.4 and 10.8 times, respectively. Rifampin 163-173 solute carrier organic anion transporter family member 1A2 Homo sapiens 148-152 28108285-0 2017 Rifampin suppresses osteoclastogenesis and titanium particle-induced osteolysis via modulating RANKL signaling pathways. Rifampin 0-8 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 95-100 28108285-13 2017 Further investigation revealed that rifampin inhibited osteoclast formation by specifically abrogating RANKL-induced p38 and NF-kappaB signaling. Rifampin 36-44 tumor necrosis factor (ligand) superfamily, member 11 Mus musculus 103-108 28108285-13 2017 Further investigation revealed that rifampin inhibited osteoclast formation by specifically abrogating RANKL-induced p38 and NF-kappaB signaling. Rifampin 36-44 AHA1, activator of heat shock protein ATPase 1 Mus musculus 117-120 27373553-10 2017 Furthermore, we isolated isoniazid/rifampicin-specific T cells from patients, which consisted primarily of CD4+ T cells. Rifampin 35-45 CD4 molecule Homo sapiens 107-110 27373553-11 2017 Drug-specific CD4+ T cells proliferated and secreted IFN-gamma/granzyme B when stimulated with isoniazid or rifampicin, respectively. Rifampin 108-118 CD4 molecule Homo sapiens 14-17 27373553-11 2017 Drug-specific CD4+ T cells proliferated and secreted IFN-gamma/granzyme B when stimulated with isoniazid or rifampicin, respectively. Rifampin 108-118 interferon gamma Homo sapiens 53-62 27373553-11 2017 Drug-specific CD4+ T cells proliferated and secreted IFN-gamma/granzyme B when stimulated with isoniazid or rifampicin, respectively. Rifampin 108-118 granzyme B Homo sapiens 63-73 28077325-10 2017 P-gp overexpression was induced only when 14-3-3sigma transfected LS174T cells were treated with rifampin and paclitaxel, whereas 14-3-3sigma inhibition by nonpeptidic inhibitor, BV02 and 14-3-3sigma siRNA reduced rifampin induced PXR and P-gp expression. Rifampin 214-222 ATP binding cassette subfamily B member 1 Homo sapiens 0-4 28077325-12 2017 This data indicates that 14-3-3sigma contributes to P-gp overexpression through interaction with PXR with rifampin and paclitaxel treatment. Rifampin 106-114 stratifin Homo sapiens 25-36 28077325-12 2017 This data indicates that 14-3-3sigma contributes to P-gp overexpression through interaction with PXR with rifampin and paclitaxel treatment. Rifampin 106-114 ATP binding cassette subfamily B member 1 Homo sapiens 52-56 28077325-12 2017 This data indicates that 14-3-3sigma contributes to P-gp overexpression through interaction with PXR with rifampin and paclitaxel treatment. Rifampin 106-114 nuclear receptor subfamily 1 group I member 2 Homo sapiens 97-100 28952270-4 2017 The results showed that RIF can significantly induce CYP1A2 and CYP2B enzyme activity, and inhibit CYP3A enzyme activity. Rifampin 24-27 cytochrome P450, family 1, subfamily a, polypeptide 2 Rattus norvegicus 53-59 28952270-4 2017 The results showed that RIF can significantly induce CYP1A2 and CYP2B enzyme activity, and inhibit CYP3A enzyme activity. Rifampin 24-27 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 99-104 27693756-10 2017 These results indicate that NQO1 and CPR play an important role in redox cycle of RIF-Q and may thus contribute to RIF-induced adverse reactions. Rifampin 82-86 NAD(P)H quinone dehydrogenase 1 Rattus norvegicus 28-32 27693756-10 2017 These results indicate that NQO1 and CPR play an important role in redox cycle of RIF-Q and may thus contribute to RIF-induced adverse reactions. Rifampin 82-86 cytochrome p450 oxidoreductase Rattus norvegicus 37-40 28674261-5 2017 After 2 d exposure of hepatocyte spheroids to omeprazole, phenobarbital and rifampicin (typical inducers of CYP1A2, 2B6 and 3A4, respectively), CYP1A2, 2B6 and 3A4 mRNA expression levels were significantly increased. Rifampin 76-86 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 108-114 27637290-10 2017 Especially, the down-regulation of hepatic Cyp3a11 suggests that undesirable pharmacokinetic alternations of drugs especially Cyp3a11 substrates like rifampicin might occur in phase with cholecystectomy. Rifampin 150-160 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 43-50 27637290-10 2017 Especially, the down-regulation of hepatic Cyp3a11 suggests that undesirable pharmacokinetic alternations of drugs especially Cyp3a11 substrates like rifampicin might occur in phase with cholecystectomy. Rifampin 150-160 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 126-133 27967330-6 2017 Overexpression of miR-628-3p and miR-641 showed significant decrease of CYP3A4 mRNA level as well as CYP3A4 induction by rifampin. Rifampin 121-129 microRNA 641 Homo sapiens 33-40 27967330-6 2017 Overexpression of miR-628-3p and miR-641 showed significant decrease of CYP3A4 mRNA level as well as CYP3A4 induction by rifampin. Rifampin 121-129 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 101-107 27967330-7 2017 CONCLUSION: miR-628-3p and miR-641 could directly target CYP3A4 and are negatively regulated in CYP3A4 induction by rifampin. Rifampin 116-124 microRNA 641 Homo sapiens 27-34 27967330-7 2017 CONCLUSION: miR-628-3p and miR-641 could directly target CYP3A4 and are negatively regulated in CYP3A4 induction by rifampin. Rifampin 116-124 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 57-63 27967330-7 2017 CONCLUSION: miR-628-3p and miR-641 could directly target CYP3A4 and are negatively regulated in CYP3A4 induction by rifampin. Rifampin 116-124 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 96-102 27898714-3 2016 SAB management was optimized with formal bedside infectious disease specialist consultation in 88%, deep infection foci diagnosed in 77% and adjunctive rifampicin therapy given to 61% of patients. Rifampin 152-162 SH3 domain binding protein 5 Homo sapiens 0-3 27917125-6 2016 After treatment with 20 muM rifampicin, mRNA increased 3- to 50-fold for all CYPs except CYP1A2 and 2D6 for HepaRG and 3D-PHH, 4-fold (CYP2B6) and 17-fold (CYP3A4) for 2D-PHH and four-fold (CYP3A4) for HepG2. Rifampin 28-38 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 89-95 27917125-6 2016 After treatment with 20 muM rifampicin, mRNA increased 3- to 50-fold for all CYPs except CYP1A2 and 2D6 for HepaRG and 3D-PHH, 4-fold (CYP2B6) and 17-fold (CYP3A4) for 2D-PHH and four-fold (CYP3A4) for HepG2. Rifampin 28-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-141 27917125-6 2016 After treatment with 20 muM rifampicin, mRNA increased 3- to 50-fold for all CYPs except CYP1A2 and 2D6 for HepaRG and 3D-PHH, 4-fold (CYP2B6) and 17-fold (CYP3A4) for 2D-PHH and four-fold (CYP3A4) for HepG2. Rifampin 28-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 156-162 27917125-6 2016 After treatment with 20 muM rifampicin, mRNA increased 3- to 50-fold for all CYPs except CYP1A2 and 2D6 for HepaRG and 3D-PHH, 4-fold (CYP2B6) and 17-fold (CYP3A4) for 2D-PHH and four-fold (CYP3A4) for HepG2. Rifampin 28-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 190-196 27806127-0 2016 Rifampicin-Induced Hepatic Lipid Accumulation: Association with Up-Regulation of Peroxisome Proliferator-Activated Receptor gamma in Mouse Liver. Rifampin 0-10 peroxisome proliferator activated receptor gamma Mus musculus 81-129 27806127-7 2016 Moreover, mRNA levels of Fas, Acc and Scd-1, several key genes for fatty acid synthesis, were elevated in rifampicin-treated mice. Rifampin 106-116 anterior capsular cataract Mus musculus 30-33 27806127-7 2016 Moreover, mRNA levels of Fas, Acc and Scd-1, several key genes for fatty acid synthesis, were elevated in rifampicin-treated mice. Rifampin 106-116 stearoyl-Coenzyme A desaturase 1 Mus musculus 38-43 27806127-10 2016 Instead, hepatic PXR was rapidly activated in rifampicin-treated mice. Rifampin 46-56 nuclear receptor subfamily 1, group I, member 2 Mus musculus 17-20 27806127-13 2016 The increased uptake of fatty acids from circulation into liver might be partially attributed to rifampicin-induced up-regulation of PPARgamma and its target genes. Rifampin 97-107 peroxisome proliferator activated receptor gamma Mus musculus 133-142 27550354-2 2016 Ciprofloxacin, linezolid, para-aminosalicylic acid (PAS), and rifampin were observed to have strong inhibitory effects, with the concentrations for a 50% inhibitory effect (IC50s) being 35.1, 31.1, 37.6, and 48.1 muM, respectively, for OAT1 and >100, 21.9, 24.6, and 30.2 muM, respectively, for OAT3. Rifampin 62-70 latexin Homo sapiens 213-216 27550354-2 2016 Ciprofloxacin, linezolid, para-aminosalicylic acid (PAS), and rifampin were observed to have strong inhibitory effects, with the concentrations for a 50% inhibitory effect (IC50s) being 35.1, 31.1, 37.6, and 48.1 muM, respectively, for OAT1 and >100, 21.9, 24.6, and 30.2 muM, respectively, for OAT3. Rifampin 62-70 solute carrier family 22 member 6 Homo sapiens 236-240 27550354-2 2016 Ciprofloxacin, linezolid, para-aminosalicylic acid (PAS), and rifampin were observed to have strong inhibitory effects, with the concentrations for a 50% inhibitory effect (IC50s) being 35.1, 31.1, 37.6, and 48.1 muM, respectively, for OAT1 and >100, 21.9, 24.6, and 30.2 muM, respectively, for OAT3. Rifampin 62-70 latexin Homo sapiens 275-278 27550354-2 2016 Ciprofloxacin, linezolid, para-aminosalicylic acid (PAS), and rifampin were observed to have strong inhibitory effects, with the concentrations for a 50% inhibitory effect (IC50s) being 35.1, 31.1, 37.6, and 48.1 muM, respectively, for OAT1 and >100, 21.9, 24.6, and 30.2 muM, respectively, for OAT3. Rifampin 62-70 solute carrier family 22 member 8 Homo sapiens 298-302 27208659-0 2016 A rifampicin-resistant (rpoB) mutation in Pseudomonas protegens Pf-5 strain leads to improved antifungal activity and elevated production of secondary metabolites. Rifampin 2-12 rpoB Pseudomonas protegens Pf-5 24-28 27470132-0 2016 Rifampicin-induced injury in L02 cells is alleviated by 4-PBA via inhibition of the PERK-ATF4-CHOP pathway. Rifampin 0-10 eukaryotic translation initiation factor 2 alpha kinase 3 Homo sapiens 84-88 27470132-0 2016 Rifampicin-induced injury in L02 cells is alleviated by 4-PBA via inhibition of the PERK-ATF4-CHOP pathway. Rifampin 0-10 activating transcription factor 4 Homo sapiens 89-93 27470132-0 2016 Rifampicin-induced injury in L02 cells is alleviated by 4-PBA via inhibition of the PERK-ATF4-CHOP pathway. Rifampin 0-10 DNA damage inducible transcript 3 Homo sapiens 94-98 27118859-0 2016 Potential pharmacokinetic effect of rifampicin on enrofloxacin in broilers: Roles of P-glycoprotein and BCRP induction by rifampicin. Rifampin 122-132 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 104-108 27118859-2 2016 It has been shown that rifampicin is the typical inducer of P-gp in rodents by activating the nuclear receptor. Rifampin 23-33 phosphoglycolate phosphatase Homo sapiens 60-64 27118859-4 2016 This study explored the effect of rifampicin on mRNA expression of Abcb1, Abcg2, CYP3A37, CXR as well as its effect on the pharmacokinetics of enrofloxacin in broilers. Rifampin 34-44 ATP binding cassette subfamily B member 1 Homo sapiens 67-72 27118859-4 2016 This study explored the effect of rifampicin on mRNA expression of Abcb1, Abcg2, CYP3A37, CXR as well as its effect on the pharmacokinetics of enrofloxacin in broilers. Rifampin 34-44 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 74-79 27118859-5 2016 The mRNA levels of Abcb1, Abcg2, CYP3A37, and CXR were significantly increased in the liver (except Abcg2), kidney, jejunum, and ileum (P < 0.05) but not significantly changed in the duodenum (P > 0.05) after treated with rifampicin. Rifampin 228-238 ATP binding cassette subfamily B member 1 Homo sapiens 19-24 27118859-10 2016 Our study shows that rifampicin up-regulated the small intestinal level of P-gp and BCRP and suggests that P-gp and BCRP are key factors that affected pharmacokinetic behavior of orally administered enrofloxacin by limiting its absorption from the intestine in broilers. Rifampin 21-31 phosphoglycolate phosphatase Homo sapiens 75-79 27118859-10 2016 Our study shows that rifampicin up-regulated the small intestinal level of P-gp and BCRP and suggests that P-gp and BCRP are key factors that affected pharmacokinetic behavior of orally administered enrofloxacin by limiting its absorption from the intestine in broilers. Rifampin 21-31 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 84-88 27118859-10 2016 Our study shows that rifampicin up-regulated the small intestinal level of P-gp and BCRP and suggests that P-gp and BCRP are key factors that affected pharmacokinetic behavior of orally administered enrofloxacin by limiting its absorption from the intestine in broilers. Rifampin 21-31 phosphoglycolate phosphatase Homo sapiens 107-111 27118859-10 2016 Our study shows that rifampicin up-regulated the small intestinal level of P-gp and BCRP and suggests that P-gp and BCRP are key factors that affected pharmacokinetic behavior of orally administered enrofloxacin by limiting its absorption from the intestine in broilers. Rifampin 21-31 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 116-120 27311985-8 2016 Transfection of HepaRG cells with hsa-miR-25-3p mimics inhibited expression of the endogenous CYP2B6 gene and it also decreased rifampicin-dependent induction of CYP2B6 at the mRNA and protein levels. Rifampin 128-138 microRNA 25 Homo sapiens 34-44 27311985-8 2016 Transfection of HepaRG cells with hsa-miR-25-3p mimics inhibited expression of the endogenous CYP2B6 gene and it also decreased rifampicin-dependent induction of CYP2B6 at the mRNA and protein levels. Rifampin 128-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 26678015-6 2016 The exposure of naloxegol was affected substantially by ketoconazole, diltiazem, and rifampin, suggesting that it is a sensitive in vivo substrate of CYP3A4. Rifampin 85-93 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 150-156 27638038-1 2016 Compared with smear microscopy, the Xpert MTB/RIF assay (Xpert), with superior accuracy and capacity to diagnose rifampicin resistance, has advanced TB diagnostic capability. Rifampin 113-123 ras homolog family member F, filopodia associated Homo sapiens 46-49 27062377-8 2016 In subgroup analyses of the RsaI/PstI polymorphism, significant results were found in East Asians, patients who used isoniazid + rifampicin + pyrazinamide + ethambutol and patients with twice the upper limit of normal as the minimum standard for defining ATDH. Rifampin 129-139 serine peptidase inhibitor Kazal type 1 Homo sapiens 33-37 27169677-0 2016 Profound reduction in tamoxifen active metabolite endoxifen in a breast cancer patient treated with rifampin prior to initiation of an anti-TNFalpha biologic for ulcerative colitis: a case report. Rifampin 100-108 tumor necrosis factor Homo sapiens 140-148 26920453-7 2016 KEY RESULTS: OCT1 mRNA in human hepatocytes was down-regulated along with reduced [(3) H]MPP(+) accumulation in differentiated HepaRG cells after treatment with rifampicin. Rifampin 161-171 solute carrier family 22 member 1 Homo sapiens 13-17 27020329-0 2016 Rifampicin is a candidate preventive medicine against amyloid-beta and tau oligomers. Rifampin 0-10 amyloid beta precursor protein Homo sapiens 54-66 27020329-6 2016 Among these compounds, rifampicin, a well-known antibiotic, showed the strongest activities against the accumulation and toxicity (i.e. cytochrome c release from mitochondria) of intracellular amyloid-beta oligomers. Rifampin 23-33 amyloid beta precursor protein Homo sapiens 193-205 27020329-7 2016 Under cell-free conditions, rifampicin inhibited oligomer formation of amyloid-beta, tau, and alpha-synuclein, indicating its broad spectrum. Rifampin 28-38 amyloid beta precursor protein Homo sapiens 71-83 26920453-8 2016 Rifampicin and hyperforin as well as the constitutively active PXR mutant T248D suppressed activity of the 1.8 kb OCT1 promoter construct in gene reporter assays. Rifampin 0-10 solute carrier family 22 member 1 Homo sapiens 114-118 27020329-7 2016 Under cell-free conditions, rifampicin inhibited oligomer formation of amyloid-beta, tau, and alpha-synuclein, indicating its broad spectrum. Rifampin 28-38 synuclein, alpha Mus musculus 94-109 27020329-8 2016 The inhibitory effects of rifampicin against amyloid-beta and tau oligomers were evaluated in APPOSK mice (amyloid-beta oligomer model), Tg2576 mice (Alzheimer"s disease model), and tau609 mice (tauopathy model). Rifampin 26-36 amyloid beta precursor protein Homo sapiens 45-57 26839383-1 2016 BACKGROUND: The Xpert MTB/RIF (Xpert) assay is a rapid nucleic acid amplification test widely used in settings of high tuberculosis prevalence to detect tuberculosis as well asrpoBmutations associated with rifampin resistance. Rifampin 206-214 metallothionein 1J, pseudogene Homo sapiens 22-25 27020329-9 2016 When orally administered to 17-month-old APPOSK mice at 0.5 and 1 mg/day for 1 month, rifampicin reduced the accumulation of amyloid-beta oligomers as well as tau hyperphosphorylation, synapse loss, and microglial activation in a dose-dependent manner. Rifampin 86-96 amyloid beta precursor protein Homo sapiens 125-137 27020329-11 2016 Rifampicin also inhibited cytochrome c release from the mitochondria and caspase 3 activation in the hippocampus. Rifampin 0-10 caspase 3 Mus musculus 73-82 27020329-12 2016 In 13-month-old Tg2576 mice, oral rifampicin at 0.5 mg/day for 1 month decreased amyloid-beta oligomer accumulation, tau hyperphosphorylation, synapse loss, and microglial activation, but not amyloid deposition. Rifampin 34-44 amyloid beta precursor protein Homo sapiens 81-93 27020329-14 2016 In addition, rifampicin decreased the level of p62/sequestosome-1 in the brain without affecting the increased levels of LC3 (microtubule-associated protein light chain 3) conversion, suggesting the restoration of autophagy-lysosomal function. Rifampin 13-23 sequestosome 1 Mus musculus 47-50 27020329-14 2016 In addition, rifampicin decreased the level of p62/sequestosome-1 in the brain without affecting the increased levels of LC3 (microtubule-associated protein light chain 3) conversion, suggesting the restoration of autophagy-lysosomal function. Rifampin 13-23 sequestosome 1 Mus musculus 51-65 26966071-7 2016 Treatment with dexamethasone, phenobarbital, rifampicin, or 1alpha,25-dihydroxyvitamin D3 resulted in 5.8-fold, 13.4-fold, 9.8-fold, or 95.0-fold induction of CYP3A4 expression relative to that in the untreated controls, respectively. Rifampin 45-55 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 159-165 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 15-23 solute carrier organic anion transporter family member 1B3 Homo sapiens 58-65 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 15-23 solute carrier family 47 member 1 Homo sapiens 67-74 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 15-23 solute carrier family 29 member 1 (Augustine blood group) Homo sapiens 76-83 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 15-23 solute carrier family 22 member 9 Homo sapiens 85-92 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 15-23 solute carrier organic anion transporter family member 1B3 Homo sapiens 180-187 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 15-23 solute carrier family 47 member 1 Homo sapiens 197-204 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 15-23 microRNA 95 Homo sapiens 205-211 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 15-23 solute carrier family 29 member 1 (Augustine blood group) Homo sapiens 213-220 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 15-23 microRNA 30d Homo sapiens 221-228 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 15-23 solute carrier family 22 member 9 Homo sapiens 235-242 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 15-23 microRNA 20a Homo sapiens 243-249 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 130-138 solute carrier organic anion transporter family member 1B3 Homo sapiens 58-65 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 130-138 solute carrier family 47 member 1 Homo sapiens 67-74 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 130-138 solute carrier family 29 member 1 (Augustine blood group) Homo sapiens 76-83 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 130-138 solute carrier family 22 member 9 Homo sapiens 85-92 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 130-138 solute carrier organic anion transporter family member 1B3 Homo sapiens 180-187 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 130-138 solute carrier family 47 member 1 Homo sapiens 197-204 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 130-138 microRNA 95 Homo sapiens 205-211 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 130-138 solute carrier family 29 member 1 (Augustine blood group) Homo sapiens 213-220 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 130-138 microRNA 30d Homo sapiens 221-228 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 130-138 solute carrier family 22 member 9 Homo sapiens 235-242 27199754-11 2016 The effects of rifampin on four uptake drug transporters (SLCO1B3, SLC47A1, SLC29A1, SLC22A9) were negatively correlated with the rifampin effects on specific microRNA expression (SLCO1B3/miR-92a, SLC47A1/miR-95, SLC29A1/miR-30d#, and SLC22A9/miR-20; r < -0.79; p < 0.05). Rifampin 130-138 microRNA 20a Homo sapiens 243-249 27045425-0 2016 CYP2B6 genotype-based efavirenz dose recommendations during rifampicin-based antituberculosis cotreatment for a sub-Saharan Africa population. Rifampin 60-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 26749408-2 2016 The effects of rifampin, a strong inducer of CYP3A4 and P-glycoprotein, on the pharmacokinetics of oral and intravenous apixaban were evaluated in an open-label, randomized, sequential crossover study. Rifampin 15-23 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 45-51 26749408-2 2016 The effects of rifampin, a strong inducer of CYP3A4 and P-glycoprotein, on the pharmacokinetics of oral and intravenous apixaban were evaluated in an open-label, randomized, sequential crossover study. Rifampin 15-23 ATP binding cassette subfamily B member 1 Homo sapiens 56-70 27478623-7 2016 Furthermore, these mouse strains showed a significantly different response to rifampicin treatment with regard to Abeta clearance and effect on brain level of its clearance-related proteins. Rifampin 78-88 amyloid beta (A4) precursor protein Mus musculus 114-119 26889608-6 2016 Our finding that in Latvia, time to treatment initiation was decreased for MDR TB cases that were rifampin-resistant TB by XpertMTB/RIF has implications for the use of this test in other settings with a high burden of MDR TB in which rifampin resistance is highly predictive of MDR TB. Rifampin 98-106 ras homolog family member F, filopodia associated Homo sapiens 132-135 26810651-9 2016 After adjusting for age and weight, rifampin exposures for the two formulations used differed inCmax(geometric mean ratio [GMR],2.55; 95% confidence interval [CI], 1.47 to 4.41;P= 0.001) and AUC0-8(GMR, 2.52; 95% CI, 1.34 to 4.73;P= 0.005). Rifampin 36-44 colony stimulating factor 2 receptor subunit alpha Homo sapiens 123-126 26810651-9 2016 After adjusting for age and weight, rifampin exposures for the two formulations used differed inCmax(geometric mean ratio [GMR],2.55; 95% confidence interval [CI], 1.47 to 4.41;P= 0.001) and AUC0-8(GMR, 2.52; 95% CI, 1.34 to 4.73;P= 0.005). Rifampin 36-44 colony stimulating factor 2 receptor subunit alpha Homo sapiens 198-201 27627555-3 2016 MATERIAL AND METHODS: In in vitro experiments, everted parts of rat small intestines were used to evaluate the effects of verapamil (a P-gp inhibitor) and rifampicin (a P-gp inducer) on the intestinal absorption of cyclosporine and tacrolimus. Rifampin 155-165 phosphoglycolate phosphatase Rattus norvegicus 169-173 26808255-13 2016 In conclusion, the major undesired influence of RIF on oral absorption and pulmonary distribution of CLA is associated with induction of intestinal P-gp. Rifampin 48-51 phosphoglycolate phosphatase Equus caballus 148-152 26385839-5 2016 It performed well in recovering the observed effect of the CYP3A4 inhibitors ketoconazole and cyclosporine, and the CYP3A4 inducer rifampicin, as well as in predicting interactions with S-warfarin and sildenafil. Rifampin 131-141 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-122 26739683-0 2016 Effects of CYP3A4 Inhibitors Ketoconazole and Verapamil and the CYP3A4 Inducer Rifampicin on the Pharmacokinetic Parameters of Fostamatinib: Results from In Vitro and Phase I Clinical Studies. Rifampin 79-89 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 64-70 26616219-10 2016 Atorvastatin and rifampicin significantly inhibited glucose uptake and down-regulated GLUT2 and GCK expressions. Rifampin 17-27 solute carrier family 2 member 2 Rattus norvegicus 86-91 26259716-5 2016 The interplay between hepatic uptake and CYP3A4 metabolism was verified by simulations with rifampicin, a potent CYP3A4 inducer and OATP1B1/3 inhibitor, and maintenance doses of cyclosporine. Rifampin 92-102 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-47 26259716-5 2016 The interplay between hepatic uptake and CYP3A4 metabolism was verified by simulations with rifampicin, a potent CYP3A4 inducer and OATP1B1/3 inhibitor, and maintenance doses of cyclosporine. Rifampin 92-102 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 113-119 26782648-0 2016 SUMOylation of pregnane X receptor suppresses rifampicin-induced CYP3A4 and P-gp expression and activity in LS174T cells. Rifampin 46-56 nuclear receptor subfamily 1 group I member 2 Homo sapiens 15-34 26782648-0 2016 SUMOylation of pregnane X receptor suppresses rifampicin-induced CYP3A4 and P-gp expression and activity in LS174T cells. Rifampin 46-56 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-71 26782648-0 2016 SUMOylation of pregnane X receptor suppresses rifampicin-induced CYP3A4 and P-gp expression and activity in LS174T cells. Rifampin 46-56 ATP binding cassette subfamily B member 1 Homo sapiens 76-80 26782648-5 2016 Our results revealed that rifampicin-induced PXR transactivation of the CYP3A4 and P-gp promoter was suppressed by SUMOylation of PXR in reporter gene assay. Rifampin 26-36 nuclear receptor subfamily 1 group I member 2 Homo sapiens 45-48 26782648-5 2016 Our results revealed that rifampicin-induced PXR transactivation of the CYP3A4 and P-gp promoter was suppressed by SUMOylation of PXR in reporter gene assay. Rifampin 26-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 72-78 26782648-5 2016 Our results revealed that rifampicin-induced PXR transactivation of the CYP3A4 and P-gp promoter was suppressed by SUMOylation of PXR in reporter gene assay. Rifampin 26-36 ATP binding cassette subfamily B member 1 Homo sapiens 83-87 26782648-5 2016 Our results revealed that rifampicin-induced PXR transactivation of the CYP3A4 and P-gp promoter was suppressed by SUMOylation of PXR in reporter gene assay. Rifampin 26-36 nuclear receptor subfamily 1 group I member 2 Homo sapiens 130-133 26782648-7 2016 Moreover, CYP3A4 enzymatic assay and Rho123 intracellular assay revealed that rifampicin-induced CYP3A4 and P-gp activity was also suppressed by SUMOylated PXR. Rifampin 78-88 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 10-16 26782648-7 2016 Moreover, CYP3A4 enzymatic assay and Rho123 intracellular assay revealed that rifampicin-induced CYP3A4 and P-gp activity was also suppressed by SUMOylated PXR. Rifampin 78-88 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 97-103 26782648-7 2016 Moreover, CYP3A4 enzymatic assay and Rho123 intracellular assay revealed that rifampicin-induced CYP3A4 and P-gp activity was also suppressed by SUMOylated PXR. Rifampin 78-88 ATP binding cassette subfamily B member 1 Homo sapiens 108-112 26782648-7 2016 Moreover, CYP3A4 enzymatic assay and Rho123 intracellular assay revealed that rifampicin-induced CYP3A4 and P-gp activity was also suppressed by SUMOylated PXR. Rifampin 78-88 nuclear receptor subfamily 1 group I member 2 Homo sapiens 156-159 26782648-8 2016 Our data collectively indicated for the first time that SUMOylation of PXR exerts suppressive effect on rifampicin-induced expression and activity of CYP3A4 and P-gp, which suggest that alteration in the SUMOylation status of PXR will be expected to affect the CYP3A4 mediated drug metabolism and P-gp mediated drug transport. Rifampin 104-114 nuclear receptor subfamily 1 group I member 2 Homo sapiens 71-74 26782648-8 2016 Our data collectively indicated for the first time that SUMOylation of PXR exerts suppressive effect on rifampicin-induced expression and activity of CYP3A4 and P-gp, which suggest that alteration in the SUMOylation status of PXR will be expected to affect the CYP3A4 mediated drug metabolism and P-gp mediated drug transport. Rifampin 104-114 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 150-156 26782648-8 2016 Our data collectively indicated for the first time that SUMOylation of PXR exerts suppressive effect on rifampicin-induced expression and activity of CYP3A4 and P-gp, which suggest that alteration in the SUMOylation status of PXR will be expected to affect the CYP3A4 mediated drug metabolism and P-gp mediated drug transport. Rifampin 104-114 ATP binding cassette subfamily B member 1 Homo sapiens 161-165 26782648-8 2016 Our data collectively indicated for the first time that SUMOylation of PXR exerts suppressive effect on rifampicin-induced expression and activity of CYP3A4 and P-gp, which suggest that alteration in the SUMOylation status of PXR will be expected to affect the CYP3A4 mediated drug metabolism and P-gp mediated drug transport. Rifampin 104-114 nuclear receptor subfamily 1 group I member 2 Homo sapiens 226-229 26782648-8 2016 Our data collectively indicated for the first time that SUMOylation of PXR exerts suppressive effect on rifampicin-induced expression and activity of CYP3A4 and P-gp, which suggest that alteration in the SUMOylation status of PXR will be expected to affect the CYP3A4 mediated drug metabolism and P-gp mediated drug transport. Rifampin 104-114 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 261-267 26616219-3 2016 Rifampicin/PCN served as PXR activator control. Rifampin 0-10 nuclear receptor subfamily 1, group I, member 2 Rattus norvegicus 25-28 26854345-2 2016 The objective of this study was to investigate the patterns of C-reactive protein (CRP), haptoglobin (Hp), ferritin and paraoxonase-1 (PON-1) during treatment of dogs with acute CME with rifampicin. Rifampin 187-197 paraoxonase 1 Canis lupus familiaris 135-140 26658225-5 2016 The EC50 of rifampin for CYP3A4 and CYP2C9 was up to 10-fold lower in HepatoPac than the monocultures. Rifampin 12-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-31 26658225-5 2016 The EC50 of rifampin for CYP3A4 and CYP2C9 was up to 10-fold lower in HepatoPac than the monocultures. Rifampin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 26782648-8 2016 Our data collectively indicated for the first time that SUMOylation of PXR exerts suppressive effect on rifampicin-induced expression and activity of CYP3A4 and P-gp, which suggest that alteration in the SUMOylation status of PXR will be expected to affect the CYP3A4 mediated drug metabolism and P-gp mediated drug transport. Rifampin 104-114 ATP binding cassette subfamily B member 1 Homo sapiens 297-301 26684581-9 2016 Furthermore, uptake of compound 2 was inhibited by two inhibitors of OATP1B1*1a and OATP1B3: rifampicin and ritonavir. Rifampin 93-103 solute carrier organic anion transporter family member 1B1 Homo sapiens 69-76 26684581-9 2016 Furthermore, uptake of compound 2 was inhibited by two inhibitors of OATP1B1*1a and OATP1B3: rifampicin and ritonavir. Rifampin 93-103 solute carrier organic anion transporter family member 1B3 Homo sapiens 84-91 26616219-10 2016 Atorvastatin and rifampicin significantly inhibited glucose uptake and down-regulated GLUT2 and GCK expressions. Rifampin 17-27 glucokinase Rattus norvegicus 96-99 26616219-12 2016 Ketoconazole and resveratrol attenuated the impaired glucose utilization by atorvastatin and rifampicin in both parental and overexpressed PXR cells. Rifampin 93-103 nuclear receptor subfamily 1, group I, member 2 Rattus norvegicus 139-142 26616219-13 2016 PXR knockdown significantly up-regulated GLUT2 and GCK proteins and abolished the decreased glucose consumption and uptake by atorvastatin and rifampicin. Rifampin 143-153 nuclear receptor subfamily 1, group I, member 2 Rattus norvegicus 0-3 26319954-0 2016 Rifampicin Induces Bicarbonate-Rich Choleresis in Rats: Involvement of Anion Exchanger 2. Rifampin 0-10 solute carrier family 4 member 2 Rattus norvegicus 71-88 26549688-6 2016 RT-PCR analysis showed that PXR SUMOylation in presence of rifampicin also enhances the endogenous expression levels of key PXR-regulated genes like CYP3A4, CYP2C9, MDR1 and UGT1A1. Rifampin 59-69 nuclear receptor subfamily 1 group I member 2 Homo sapiens 28-31 26549688-6 2016 RT-PCR analysis showed that PXR SUMOylation in presence of rifampicin also enhances the endogenous expression levels of key PXR-regulated genes like CYP3A4, CYP2C9, MDR1 and UGT1A1. Rifampin 59-69 nuclear receptor subfamily 1 group I member 2 Homo sapiens 124-127 26549688-6 2016 RT-PCR analysis showed that PXR SUMOylation in presence of rifampicin also enhances the endogenous expression levels of key PXR-regulated genes like CYP3A4, CYP2C9, MDR1 and UGT1A1. Rifampin 59-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 26549688-6 2016 RT-PCR analysis showed that PXR SUMOylation in presence of rifampicin also enhances the endogenous expression levels of key PXR-regulated genes like CYP3A4, CYP2C9, MDR1 and UGT1A1. Rifampin 59-69 ATP binding cassette subfamily B member 1 Homo sapiens 165-169 26549688-6 2016 RT-PCR analysis showed that PXR SUMOylation in presence of rifampicin also enhances the endogenous expression levels of key PXR-regulated genes like CYP3A4, CYP2C9, MDR1 and UGT1A1. Rifampin 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 26759747-2 2016 Rifampin is a widely used antimicrobial that has major interactions with several medications including warfarin due to its strong P-glycoprotein and liver enzyme inducer activity especially on CYP2C9, CYP3A4, CYP1A2 and CYP2C19. Rifampin 0-8 ATP binding cassette subfamily B member 1 Homo sapiens 130-144 26759747-2 2016 Rifampin is a widely used antimicrobial that has major interactions with several medications including warfarin due to its strong P-glycoprotein and liver enzyme inducer activity especially on CYP2C9, CYP3A4, CYP1A2 and CYP2C19. Rifampin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 193-199 26759747-2 2016 Rifampin is a widely used antimicrobial that has major interactions with several medications including warfarin due to its strong P-glycoprotein and liver enzyme inducer activity especially on CYP2C9, CYP3A4, CYP1A2 and CYP2C19. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 201-207 26759747-2 2016 Rifampin is a widely used antimicrobial that has major interactions with several medications including warfarin due to its strong P-glycoprotein and liver enzyme inducer activity especially on CYP2C9, CYP3A4, CYP1A2 and CYP2C19. Rifampin 0-8 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 209-215 26759747-2 2016 Rifampin is a widely used antimicrobial that has major interactions with several medications including warfarin due to its strong P-glycoprotein and liver enzyme inducer activity especially on CYP2C9, CYP3A4, CYP1A2 and CYP2C19. Rifampin 0-8 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 220-227 25921553-0 2016 Rifampicin-dependent antibodies target glycoprotein IIb/IIIa and cause clearance of human platelets in NOD/SCID mice. Rifampin 0-10 atrophin 1 Homo sapiens 103-106 26319954-11 2016 After rifampicin treatment, the expression level of anion exchanger 2 (AE2) increased, while the location of hepatic transporters did not change. Rifampin 6-16 solute carrier family 4 member 2 Rattus norvegicus 52-69 26534988-6 2016 Treatment with pifithrin-mu and transfection with anti-CHIP small-interfering RNA reduced the levels of CYP3A4 mRNA induced by rifampicin, suggesting the contribution of Hsp70 and CHIP to the transactivation of hPXR. Rifampin 127-137 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-110 26452722-10 2016 Rifampin-mediated CYP3A4 induction increased midazolam clearance by 73%, while CYP3A4 inhibition with ritonavir decreased midazolam clearance by 79%. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 18-24 26319954-11 2016 After rifampicin treatment, the expression level of anion exchanger 2 (AE2) increased, while the location of hepatic transporters did not change. Rifampin 6-16 solute carrier family 4 member 2 Rattus norvegicus 71-74 26534988-6 2016 Treatment with pifithrin-mu and transfection with anti-CHIP small-interfering RNA reduced the levels of CYP3A4 mRNA induced by rifampicin, suggesting the contribution of Hsp70 and CHIP to the transactivation of hPXR. Rifampin 127-137 heat shock protein family A (Hsp70) member 4 Homo sapiens 170-175 26319954-13 2016 CONCLUSIONS: These results indicate that the increase in bile flow induced by rifampicin is mainly due to increased HCO3 (-) excretion mediated by increased AE2 protein expression and activity. Rifampin 78-88 solute carrier family 4 member 2 Rattus norvegicus 157-160 26534988-6 2016 Treatment with pifithrin-mu and transfection with anti-CHIP small-interfering RNA reduced the levels of CYP3A4 mRNA induced by rifampicin, suggesting the contribution of Hsp70 and CHIP to the transactivation of hPXR. Rifampin 127-137 nuclear receptor subfamily 1 group I member 2 Homo sapiens 211-215 26987268-0 2016 Monoammonium glycyrrhizinate protects rifampicin- and isoniazid-induced hepatotoxicity via regulating the expression of transporter Mrp2, Ntcp, and Oatp1a4 in liver. Rifampin 38-48 ATP binding cassette subfamily C member 2 Rattus norvegicus 132-136 26910068-0 2016 In Vitro Approaches to Study Regulation of Hepatic Cytochrome P450 (CYP) 3A Expression by Paclitaxel and Rifampicin. Rifampin 105-115 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 51-75 26910068-10 2016 The cells were then treated with the pro-inflammatory cytokine, TNFalpha, followed by the prototype CYP3A inducer rifampicin. Rifampin 114-124 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 100-105 26910068-11 2016 It is well established that rifampicin activates PXR, leading to CYP3A4 induction. Rifampin 28-38 nuclear receptor subfamily 1 group I member 2 Homo sapiens 49-52 26910068-11 2016 It is well established that rifampicin activates PXR, leading to CYP3A4 induction. Rifampin 28-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-71 26910068-12 2016 We found that induction of CYP3A4-luciferase activity by rifampicin was significantly attenuated by TNFalpha. Rifampin 57-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 27-33 26910068-12 2016 We found that induction of CYP3A4-luciferase activity by rifampicin was significantly attenuated by TNFalpha. Rifampin 57-67 tumor necrosis factor Homo sapiens 100-108 26636619-2 2015 Covalently linking unacylated tridecaptin A1 (H-TriA1) to rifampicin, vancomycin, and erythromycin enhanced their activity in vitro but not by the same magnitude as coadministration of the peptide and these antibiotics. Rifampin 58-68 T cell receptor induced activation 1 Mus musculus 48-53 26434531-9 2015 Treatments with Dexamethasone (GR ligand), Rifampicin (PXR ligand) and TCDD (AhR ligand) increased the mRNA levels of PON1 at 24 and 48 h. We showed that the activation of GR by Dexamethasone results in PON1 gene induction accompanied by an increase in activity levels. Rifampin 43-53 nuclear receptor subfamily 1 group I member 2 Homo sapiens 55-58 26434531-9 2015 Treatments with Dexamethasone (GR ligand), Rifampicin (PXR ligand) and TCDD (AhR ligand) increased the mRNA levels of PON1 at 24 and 48 h. We showed that the activation of GR by Dexamethasone results in PON1 gene induction accompanied by an increase in activity levels. Rifampin 43-53 paraoxonase 1 Homo sapiens 118-122 26434531-9 2015 Treatments with Dexamethasone (GR ligand), Rifampicin (PXR ligand) and TCDD (AhR ligand) increased the mRNA levels of PON1 at 24 and 48 h. We showed that the activation of GR by Dexamethasone results in PON1 gene induction accompanied by an increase in activity levels. Rifampin 43-53 paraoxonase 1 Homo sapiens 203-207 26559489-6 2015 In addition, CYP3A4 expression in the hiPS-ELCs was induced by treatment with 1, 25-dihydroxyvitamin D3 or rifampicin, which are known to induce CYP3A4 expression, indicating that the hiPS-ELCs have CYP3A4 induction potency. Rifampin 107-117 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 13-19 26474710-6 2015 Using HEK293 cells overexpressing OATP1B1 or OATP1B3, we estimated IC50 values for both rifampicin (0.9 or 0.3 muM) and telmisartan (6.7 or 7.9 muM) in inhibiting OATP-mediated MTX uptake in vitro. Rifampin 88-98 solute carrier organic anion transporter family member 1B1 Homo sapiens 34-41 26474710-6 2015 Using HEK293 cells overexpressing OATP1B1 or OATP1B3, we estimated IC50 values for both rifampicin (0.9 or 0.3 muM) and telmisartan (6.7 or 7.9 muM) in inhibiting OATP-mediated MTX uptake in vitro. Rifampin 88-98 solute carrier organic anion transporter family member 1B3 Homo sapiens 45-52 26474710-6 2015 Using HEK293 cells overexpressing OATP1B1 or OATP1B3, we estimated IC50 values for both rifampicin (0.9 or 0.3 muM) and telmisartan (6.7 or 7.9 muM) in inhibiting OATP-mediated MTX uptake in vitro. Rifampin 88-98 solute carrier organic anion transporter family member 1A2 Homo sapiens 34-38 26474710-7 2015 Using wild-type, Oatp1a/1b-/-, and OATP1B1- or OATP1B3-humanized transgenic mice, we found that rifampicin inhibits hepatic uptake of MTX mediated by the mouse Oatp1a/1b and human OATP1B1 and OATP1B3 transporters at clinically relevant concentrations. Rifampin 96-106 solute carrier organic anion transporter family member 1B1 Homo sapiens 35-42 26474710-7 2015 Using wild-type, Oatp1a/1b-/-, and OATP1B1- or OATP1B3-humanized transgenic mice, we found that rifampicin inhibits hepatic uptake of MTX mediated by the mouse Oatp1a/1b and human OATP1B1 and OATP1B3 transporters at clinically relevant concentrations. Rifampin 96-106 solute carrier organic anion transporter family member 1B3 Homo sapiens 47-54 26474710-7 2015 Using wild-type, Oatp1a/1b-/-, and OATP1B1- or OATP1B3-humanized transgenic mice, we found that rifampicin inhibits hepatic uptake of MTX mediated by the mouse Oatp1a/1b and human OATP1B1 and OATP1B3 transporters at clinically relevant concentrations. Rifampin 96-106 solute carrier organic anion transporter family member 1B1 Homo sapiens 180-187 26474710-7 2015 Using wild-type, Oatp1a/1b-/-, and OATP1B1- or OATP1B3-humanized transgenic mice, we found that rifampicin inhibits hepatic uptake of MTX mediated by the mouse Oatp1a/1b and human OATP1B1 and OATP1B3 transporters at clinically relevant concentrations. Rifampin 96-106 solute carrier organic anion transporter family member 1B3 Homo sapiens 192-199 26381275-6 2015 Co-administration of a single 250-mg crizotinib dose with the strong CYP3A inducer rifampin caused an 82 % decrease in crizotinib AUC0-inf. Rifampin 83-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-74 26614197-2 2015 OBJECTIVE: To document the impact of identifying rifampicin (RMP) resistance using Xpert( ) MTB/RIF on time to diagnosis and time to treatment, and evaluate its performance under programmatic conditions. Rifampin 49-59 ras homolog family member F, filopodia associated Homo sapiens 96-99 26559489-6 2015 In addition, CYP3A4 expression in the hiPS-ELCs was induced by treatment with 1, 25-dihydroxyvitamin D3 or rifampicin, which are known to induce CYP3A4 expression, indicating that the hiPS-ELCs have CYP3A4 induction potency. Rifampin 107-117 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 145-151 26559489-6 2015 In addition, CYP3A4 expression in the hiPS-ELCs was induced by treatment with 1, 25-dihydroxyvitamin D3 or rifampicin, which are known to induce CYP3A4 expression, indicating that the hiPS-ELCs have CYP3A4 induction potency. Rifampin 107-117 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 145-151 26232425-1 2015 Upon treatment with the pregnane X receptor (PXR) activator rifampicin (RIF), human hepatocellular carcinoma HepG2-derived ShP51 cells that stably express PXR showed epithelial-mesenchymal transition (EMT)-like morphological changes and migration. Rifampin 60-70 nuclear receptor subfamily 1 group I member 2 Homo sapiens 24-43 26374116-10 2015 In addition, the levels of IL-10 and IL-6 were significantly elevated in the INH + RIF-induced rats treated with CoQ10. Rifampin 83-86 interleukin 10 Rattus norvegicus 27-32 26374116-10 2015 In addition, the levels of IL-10 and IL-6 were significantly elevated in the INH + RIF-induced rats treated with CoQ10. Rifampin 83-86 interleukin 6 Rattus norvegicus 37-41 26459536-0 2015 Evaluation of Xpert( ) MTB/RIF assay: diagnosis and treatment outcomes in rifampicin-resistant tuberculosis. Rifampin 74-84 ras homolog family member F, filopodia associated Homo sapiens 27-30 26459536-1 2015 SETTING: The Xpert( ) MTB/RIF assay is endorsed by the World Health Organization for the detection of rifampicin (RMP) resistant tuberculosis (TB). Rifampin 102-112 ras homolog family member F, filopodia associated Homo sapiens 26-29 26819743-1 2015 BACKGROUND: Concomitant use of clarithromycin (CAM) and rifampicin (RFP) for the treatment of pulmonary Mycobacterium avium complex (MAC) disease affects the systemic concentrations of both drugs due to CYP3A4-related interactions. Rifampin 56-66 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 203-209 26819743-1 2015 BACKGROUND: Concomitant use of clarithromycin (CAM) and rifampicin (RFP) for the treatment of pulmonary Mycobacterium avium complex (MAC) disease affects the systemic concentrations of both drugs due to CYP3A4-related interactions. Rifampin 68-71 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 203-209 26354948-9 2015 Rifampin treatment induced systemic CYP3A metabolism of (11)C-verapamil; however, it reduced the ER by 6%. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 36-41 26482301-3 2016 Nonlinear mixed-effects modeling found that influential covariates for the pharmacokinetics were weight, sex, and a 30% increased bioavailability among heterozygous carriers of SLCO1B1 rs1104581 (previously associated with low rifampin concentrations). Rifampin 227-235 solute carrier organic anion transporter family member 1B1 Homo sapiens 177-184 26086368-0 2015 Rifampicin attenuates rotenone-induced inflammation via suppressing NLRP3 inflammasome activation in microglia. Rifampin 0-10 NLR family, pyrin domain containing 3 Mus musculus 68-73 26086368-6 2015 Results demonstrated that rifampicin pretreatment significantly reduced rotenone-induced cytotoxicity and gene expression of IL-1beta in BV2 microglia. Rifampin 26-36 interleukin 1 beta Mus musculus 125-133 26086368-7 2015 Moreover, western blot analysis verified that rifampicin pretreatment suppressed NLRP3 inflammasome activation via inhibiting caspase-1 cleavage and protein expression of NLRP3. Rifampin 46-56 NLR family, pyrin domain containing 3 Mus musculus 81-86 26086368-7 2015 Moreover, western blot analysis verified that rifampicin pretreatment suppressed NLRP3 inflammasome activation via inhibiting caspase-1 cleavage and protein expression of NLRP3. Rifampin 46-56 caspase 1 Mus musculus 126-135 26086368-7 2015 Moreover, western blot analysis verified that rifampicin pretreatment suppressed NLRP3 inflammasome activation via inhibiting caspase-1 cleavage and protein expression of NLRP3. Rifampin 46-56 NLR family, pyrin domain containing 3 Mus musculus 171-176 26086368-10 2015 Taken together, our data indicate that rifampicin pretreatment inhibits maturation of IL-1beta and neuroinflammation induced by rotenone via attenuating NLRP3 inflammasome activation. Rifampin 39-49 interleukin 1 beta Mus musculus 86-94 26086368-10 2015 Taken together, our data indicate that rifampicin pretreatment inhibits maturation of IL-1beta and neuroinflammation induced by rotenone via attenuating NLRP3 inflammasome activation. Rifampin 39-49 NLR family, pyrin domain containing 3 Mus musculus 153-158 26182937-0 2015 Role of Specificity Protein 1, Hepatocyte Nuclear Factor 1alpha, and Pregnane X Receptor in the Basal and Rifampicin-Induced Transcriptional Regulation of Porcine Cytochrome P450 3A46. Rifampin 106-116 Sp1 transcription factor Homo sapiens 8-29 26182937-0 2015 Role of Specificity Protein 1, Hepatocyte Nuclear Factor 1alpha, and Pregnane X Receptor in the Basal and Rifampicin-Induced Transcriptional Regulation of Porcine Cytochrome P450 3A46. Rifampin 106-116 HNF1 homeobox A Homo sapiens 31-63 26182937-0 2015 Role of Specificity Protein 1, Hepatocyte Nuclear Factor 1alpha, and Pregnane X Receptor in the Basal and Rifampicin-Induced Transcriptional Regulation of Porcine Cytochrome P450 3A46. Rifampin 106-116 nuclear receptor subfamily 1 group I member 2 Homo sapiens 69-88 26516577-7 2015 In addition, PXR (rifampicin) and CAR-selective (carbamazepine and phenytoin) inducers of CYP3A4 and CYP2B6 induction, respectively, were demonstrated. Rifampin 18-28 nuclear receptor subfamily 1 group I member 2 Homo sapiens 13-16 26516577-7 2015 In addition, PXR (rifampicin) and CAR-selective (carbamazepine and phenytoin) inducers of CYP3A4 and CYP2B6 induction, respectively, were demonstrated. Rifampin 18-28 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-96 26516577-7 2015 In addition, PXR (rifampicin) and CAR-selective (carbamazepine and phenytoin) inducers of CYP3A4 and CYP2B6 induction, respectively, were demonstrated. Rifampin 18-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 26232425-1 2015 Upon treatment with the pregnane X receptor (PXR) activator rifampicin (RIF), human hepatocellular carcinoma HepG2-derived ShP51 cells that stably express PXR showed epithelial-mesenchymal transition (EMT)-like morphological changes and migration. Rifampin 60-70 nuclear receptor subfamily 1 group I member 2 Homo sapiens 45-48 26232425-1 2015 Upon treatment with the pregnane X receptor (PXR) activator rifampicin (RIF), human hepatocellular carcinoma HepG2-derived ShP51 cells that stably express PXR showed epithelial-mesenchymal transition (EMT)-like morphological changes and migration. Rifampin 60-70 nuclear receptor subfamily 1 group I member 2 Homo sapiens 155-158 26232425-1 2015 Upon treatment with the pregnane X receptor (PXR) activator rifampicin (RIF), human hepatocellular carcinoma HepG2-derived ShP51 cells that stably express PXR showed epithelial-mesenchymal transition (EMT)-like morphological changes and migration. Rifampin 72-75 nuclear receptor subfamily 1 group I member 2 Homo sapiens 24-43 26232425-1 2015 Upon treatment with the pregnane X receptor (PXR) activator rifampicin (RIF), human hepatocellular carcinoma HepG2-derived ShP51 cells that stably express PXR showed epithelial-mesenchymal transition (EMT)-like morphological changes and migration. Rifampin 72-75 nuclear receptor subfamily 1 group I member 2 Homo sapiens 45-48 26232425-1 2015 Upon treatment with the pregnane X receptor (PXR) activator rifampicin (RIF), human hepatocellular carcinoma HepG2-derived ShP51 cells that stably express PXR showed epithelial-mesenchymal transition (EMT)-like morphological changes and migration. Rifampin 72-75 nuclear receptor subfamily 1 group I member 2 Homo sapiens 155-158 26232425-7 2015 Subsequently, small interfering RNA knockdown of HNF4alpha connected PXR-mediated gene regulation with the PXR-induced cellular responses, showing that the knockdown resulted in the upregulation of IGFBP1 and EMT-like morphological changes without RIF treatment. Rifampin 248-251 hepatocyte nuclear factor 4 alpha Homo sapiens 49-58 26232425-7 2015 Subsequently, small interfering RNA knockdown of HNF4alpha connected PXR-mediated gene regulation with the PXR-induced cellular responses, showing that the knockdown resulted in the upregulation of IGFBP1 and EMT-like morphological changes without RIF treatment. Rifampin 248-251 nuclear receptor subfamily 1 group I member 2 Homo sapiens 69-72 26232425-7 2015 Subsequently, small interfering RNA knockdown of HNF4alpha connected PXR-mediated gene regulation with the PXR-induced cellular responses, showing that the knockdown resulted in the upregulation of IGFBP1 and EMT-like morphological changes without RIF treatment. Rifampin 248-251 nuclear receptor subfamily 1 group I member 2 Homo sapiens 107-110 27137144-2 2015 This open-label, nonrandomized, fixed-order crossover study evaluated the effect of multiple oral doses of rifampin, a strong CYP3A4 inducer, on the pharmacokinetics of ponatinib (45 mg, single dose). Rifampin 107-115 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 126-132 26757559-4 2015 The results showed that SENP1 could remarkably reduce the rifampicin (RIF)-induced MDR1 reporter activity and mRNA level in hPXR over expressed HepG2 and LS174T cells (P < 0.05), whereas adding SENP1m restored the RIF-induced increases (P < 0.05). Rifampin 58-68 SUMO specific peptidase 1 Homo sapiens 24-29 25776278-8 2015 However, positive controls (50 microM omeprazole and 25 microM rifampin) caused the anticipated CYP induction, but the highest concentration of SIPI5357 (5 microM; 10 times plasma Cmax ) had a minimal effect on CYP1A2 and CYP3A4 mRNA levels in freshly isolated human hepatocytes, suggesting that SIPI5357 is not an inducer of these enzymes. Rifampin 63-71 peptidylprolyl isomerase G Homo sapiens 96-99 26757559-4 2015 The results showed that SENP1 could remarkably reduce the rifampicin (RIF)-induced MDR1 reporter activity and mRNA level in hPXR over expressed HepG2 and LS174T cells (P < 0.05), whereas adding SENP1m restored the RIF-induced increases (P < 0.05). Rifampin 58-68 ATP binding cassette subfamily B member 1 Homo sapiens 83-87 26757559-4 2015 The results showed that SENP1 could remarkably reduce the rifampicin (RIF)-induced MDR1 reporter activity and mRNA level in hPXR over expressed HepG2 and LS174T cells (P < 0.05), whereas adding SENP1m restored the RIF-induced increases (P < 0.05). Rifampin 58-68 nuclear receptor subfamily 1 group I member 2 Homo sapiens 124-128 26757559-4 2015 The results showed that SENP1 could remarkably reduce the rifampicin (RIF)-induced MDR1 reporter activity and mRNA level in hPXR over expressed HepG2 and LS174T cells (P < 0.05), whereas adding SENP1m restored the RIF-induced increases (P < 0.05). Rifampin 58-68 SUMO specific peptidase 1 Homo sapiens 197-202 26757559-4 2015 The results showed that SENP1 could remarkably reduce the rifampicin (RIF)-induced MDR1 reporter activity and mRNA level in hPXR over expressed HepG2 and LS174T cells (P < 0.05), whereas adding SENP1m restored the RIF-induced increases (P < 0.05). Rifampin 70-73 SUMO specific peptidase 1 Homo sapiens 24-29 26757559-4 2015 The results showed that SENP1 could remarkably reduce the rifampicin (RIF)-induced MDR1 reporter activity and mRNA level in hPXR over expressed HepG2 and LS174T cells (P < 0.05), whereas adding SENP1m restored the RIF-induced increases (P < 0.05). Rifampin 70-73 ATP binding cassette subfamily B member 1 Homo sapiens 83-87 26757559-4 2015 The results showed that SENP1 could remarkably reduce the rifampicin (RIF)-induced MDR1 reporter activity and mRNA level in hPXR over expressed HepG2 and LS174T cells (P < 0.05), whereas adding SENP1m restored the RIF-induced increases (P < 0.05). Rifampin 70-73 nuclear receptor subfamily 1 group I member 2 Homo sapiens 124-128 26757559-4 2015 The results showed that SENP1 could remarkably reduce the rifampicin (RIF)-induced MDR1 reporter activity and mRNA level in hPXR over expressed HepG2 and LS174T cells (P < 0.05), whereas adding SENP1m restored the RIF-induced increases (P < 0.05). Rifampin 70-73 SUMO specific peptidase 1 Homo sapiens 197-202 26757559-4 2015 The results showed that SENP1 could remarkably reduce the rifampicin (RIF)-induced MDR1 reporter activity and mRNA level in hPXR over expressed HepG2 and LS174T cells (P < 0.05), whereas adding SENP1m restored the RIF-induced increases (P < 0.05). Rifampin 217-220 SUMO specific peptidase 1 Homo sapiens 24-29 26306393-0 2015 Rifampin use in acute community-acquired meningitis in intensive care units: the French retrospective cohort ACAM-ICU study. Rifampin 0-8 CXADR like membrane protein Homo sapiens 109-113 25862466-5 2015 When 5 mug/ml hBD3-CBD was combined with antibiotics, it decreased the MIC50 and MIC90 of tetracycline, rifampicin, and streptomycin against clinical P. aeruginosa isolates. Rifampin 104-114 defensin beta 103B Homo sapiens 14-18 26067842-2 2015 Rifampin (RMP) and ethambutol (ETB), two anti-tubercular agents, are substrates of P-gp. Rifampin 0-8 ATP binding cassette subfamily B member 1 Homo sapiens 83-87 26067842-2 2015 Rifampin (RMP) and ethambutol (ETB), two anti-tubercular agents, are substrates of P-gp. Rifampin 10-13 ATP binding cassette subfamily B member 1 Homo sapiens 83-87 26067842-4 2015 Genotype/allele frequencies in C1236T, G2677T/A and C3435T SNPs of ABCB1 were obtained from 99 tuberculosis patients susceptible or resistant to RMP and ETB (NoRER or RER). Rifampin 145-148 ATP binding cassette subfamily B member 1 Homo sapiens 67-72 26067842-7 2015 The present study suggests the 2677G>A allele of ABCB1 could be associated with simultaneous resistance to RMP and ETB in pulmonary tuberculosis patients. Rifampin 110-113 ATP binding cassette subfamily B member 1 Homo sapiens 52-57 25995454-0 2015 Casein Kinase 2 (CK2)-mediated Phosphorylation of Hsp90beta as a Novel Mechanism of Rifampin-induced MDR1 Expression. Rifampin 84-92 heat shock protein 90 alpha family class B member 1 Homo sapiens 50-59 26181578-1 2015 BACKGROUND: Xpert MTB/RIF (Xpert) was piloted in Kazakhstan to detect tuberculosis (TB) and rifampicin resistance (RR-)TB among individuals at risk of multidrug-resistant (MDR-) TB. Rifampin 92-102 ras homolog family member F, filopodia associated Homo sapiens 22-25 25760671-9 2015 Rifampin caused a substantial decrease in idelalisib (58% and 75%, Cmax and AUCinf , respectively) and GS-563117 exposures, indicating an enhanced contribution of CYP3A to idelalisib metabolism under a strongly induced state. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 163-168 26214306-4 2015 Intracellular and secreted proteomes from activated THP-1 cells infected with the Mtb H37Rv strain (MOI = 1) and treated with isoniazid and rifampicin for 1 day (infection stage) and 5 days (clearance stage) were analyzed. Rifampin 140-150 GLI family zinc finger 2 Homo sapiens 52-57 25995454-0 2015 Casein Kinase 2 (CK2)-mediated Phosphorylation of Hsp90beta as a Novel Mechanism of Rifampin-induced MDR1 Expression. Rifampin 84-92 ATP binding cassette subfamily B member 1 Homo sapiens 101-105 25720500-0 2015 Rifampicin reduces advanced glycation end products and activates DAF-16 to increase lifespan in Caenorhabditis elegans. Rifampin 0-10 Fork-head domain-containing protein;Forkhead box protein O Caenorhabditis elegans 65-71 26068927-2 2015 The effect of rifampin-induced induction of P-gp and CYP3A4/5 on transport and metabolism of edoxaban through the CYP3A4/5 pathway was investigated in a single-dose edoxaban study. Rifampin 14-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 53-59 26068927-2 2015 The effect of rifampin-induced induction of P-gp and CYP3A4/5 on transport and metabolism of edoxaban through the CYP3A4/5 pathway was investigated in a single-dose edoxaban study. Rifampin 14-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 114-120 26075722-1 2015 BACKGROUND: In March 2012, the Xpert MTB/RIF assay (Xpert) was introduced in three provincial public hospitals in Indonesia as a novel diagnostic to detect tuberculosis and rifampicin resistance among high risk individuals. Rifampin 173-183 ras homolog family member F, filopodia associated Homo sapiens 41-44 25808545-6 2015 Differentiated cells produce albumin and apolipoprotein B100 at levels equivalent to primary human hepatocytes, while demonstrating an 8-fold induction of CYP450 activity in response to aryl hydrocarbon receptor (AhR) agonist omeprazole and a 10-fold induction in response to PXR agonist rifampicin. Rifampin 288-298 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-161 25808545-6 2015 Differentiated cells produce albumin and apolipoprotein B100 at levels equivalent to primary human hepatocytes, while demonstrating an 8-fold induction of CYP450 activity in response to aryl hydrocarbon receptor (AhR) agonist omeprazole and a 10-fold induction in response to PXR agonist rifampicin. Rifampin 288-298 aryl hydrocarbon receptor Homo sapiens 186-211 25808545-6 2015 Differentiated cells produce albumin and apolipoprotein B100 at levels equivalent to primary human hepatocytes, while demonstrating an 8-fold induction of CYP450 activity in response to aryl hydrocarbon receptor (AhR) agonist omeprazole and a 10-fold induction in response to PXR agonist rifampicin. Rifampin 288-298 aryl hydrocarbon receptor Homo sapiens 213-216 25720500-7 2015 For its effect on longevity, rifampicin requires DAF-18 (nematode PTEN) as well as JNK-1 and activates DAF-16, the FOXO homolog. Rifampin 29-39 Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase daf-18 Caenorhabditis elegans 49-55 25720500-7 2015 For its effect on longevity, rifampicin requires DAF-18 (nematode PTEN) as well as JNK-1 and activates DAF-16, the FOXO homolog. Rifampin 29-39 Stress-activated protein kinase jnk-1 Caenorhabditis elegans 83-88 25720500-7 2015 For its effect on longevity, rifampicin requires DAF-18 (nematode PTEN) as well as JNK-1 and activates DAF-16, the FOXO homolog. Rifampin 29-39 Fork-head domain-containing protein;Forkhead box protein O Caenorhabditis elegans 103-109 25720500-10 2015 Together, our data suggest that the dual ability to reduce glycation in vivo and activate prolongevity processes through DAF-16 makes RIF and RSV effective lifespan-extending interventions. Rifampin 134-137 Fork-head domain-containing protein;Forkhead box protein O Caenorhabditis elegans 121-127 25443889-8 2015 Quantifying victim DDI potential was also demonstrated using fmCYP3A estimates from ketoconazole clinical DDI studies to predict the magnitude of interaction on co-administration with the CYP3A inducer, rifampicin (1.6-3.3 fold error). Rifampin 203-213 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 63-68 25835148-0 2015 Expression of hepatic cytochrome P450s and UDP-glucuronosyltransferases in PXR and CAR double humanized mice treated with rifampicin. Rifampin 122-132 nuclear receptor subfamily 1, group I, member 2 Mus musculus 75-78 25715734-1 2015 Arylacetamide deacetylase (AADAC), a microsomal serine esterase, hydrolyzes drugs, such as flutamide, phenacetin and rifampicin. Rifampin 117-127 arylacetamide deacetylase Homo sapiens 0-25 25715734-1 2015 Arylacetamide deacetylase (AADAC), a microsomal serine esterase, hydrolyzes drugs, such as flutamide, phenacetin and rifampicin. Rifampin 117-127 arylacetamide deacetylase Homo sapiens 27-32 25797167-9 2015 Prescribers should be vigilant against combination prescription of NOACs with strong inhibitors (such as ketoconazole) or inducers of P-gp and/or CYP3A4 (such as rifampicin). Rifampin 162-172 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 146-152 26017968-9 2015 CONCLUSIONS: The Xpert MTB/RIF assay was reliable for the detection of rifampicin resistant MTBC strains directly from sputum samples of patients undergoing first-line treatment for two months, being more trustworthy than the simple presence of acid-fast bacilli in the smear. Rifampin 71-81 metallothionein 1J, pseudogene Homo sapiens 23-26 25407545-5 2015 In the biodistribution study, both RFP-Man-NLCs and RFP-NLCs, compared with the commercially available rifampicin solution, displayed superior lung-targeting ability. Rifampin 103-113 tripartite motif containing 27 Homo sapiens 35-38 25996389-8 2015 Compared with the baseline strategy of selective drug-susceptibility testing only for PTB cases at high risk of drug-resistant TB, Xpert MTB/RIF implementation increased rifampicin resistant TB case detection by over fivefold. Rifampin 170-180 ras homolog family member F, filopodia associated Homo sapiens 141-144 25996389-10 2015 Positive predictive value (PPV) of rifampicin resistance detected by Xpert MTB/RIF was 94.7% (CI 91.3-98.1), in comparison to conventional DST. Rifampin 35-45 ras homolog family member F, filopodia associated Homo sapiens 79-82 25996389-11 2015 CONCLUSION: Introduction of Xpert MTB/RIF as initial diagnostic test for TB in public health facilities significantly increased case-notification rates of all bacteriologically confirmed TB by 39% and rifampicin-resistant TB case notification by fivefold. Rifampin 201-211 ras homolog family member F, filopodia associated Homo sapiens 38-41 25865846-5 2015 In in vivo studies, following the oral administration of a P-gp substrate drug, rifampicin, along with compound , the Cmax and AUC0- of rifampicin was enhanced by 31% and 46%, respectively. Rifampin 80-90 ATP binding cassette subfamily B member 1 Homo sapiens 59-63 25865846-5 2015 In in vivo studies, following the oral administration of a P-gp substrate drug, rifampicin, along with compound , the Cmax and AUC0- of rifampicin was enhanced by 31% and 46%, respectively. Rifampin 137-147 ATP binding cassette subfamily B member 1 Homo sapiens 59-63 25407545-5 2015 In the biodistribution study, both RFP-Man-NLCs and RFP-NLCs, compared with the commercially available rifampicin solution, displayed superior lung-targeting ability. Rifampin 103-113 tripartite motif containing 27 Homo sapiens 52-55 25407545-8 2015 CONCLUSIONS: RFP-Man-NLCs provided an alternative strategy for selectively delivering rifampicin to alveolar macrophages. Rifampin 86-96 tripartite motif containing 27 Homo sapiens 13-16 25915745-1 2015 INTRODUCTION: The impact on treatment outcomes of XpertMTB/RIF, a molecular-based test that provides rapid diagnosis of tuberculosis (TB) and rifampicin resistance with high accuracy, has not been reported despite its adoption in a few countries. Rifampin 142-152 ras homolog family member F, filopodia associated Homo sapiens 59-62 25907584-4 2015 We found that both ppk mutants shared most of the phenotypic changes and interestingly many of them related to susceptibility toward numerous and different type of antibiotics such as Ciprofloxacin, Chloramphenicol and Rifampicin. Rifampin 219-229 polyphosphate kinase Pseudomonas aeruginosa PAO1 19-22 25989893-5 2015 Rifampicin (RIF), a typical OATP inhibitor, inhibited this uptake, indicating the involvement of OATP in DHEAS uptake. Rifampin 0-10 sulfotransferase family 2A member 1 Homo sapiens 105-110 25581390-3 2015 In rats administered 20 and 80 mg/kg rifampicin, the plasma levels of bilirubin glucuronides were elevated, gradually decreased, and almost returned to the baseline level at 24 hours after administration without an elevation of alanine aminotransferase (ALT) and aspartate aminotransferase (AST). Rifampin 37-47 glutamic-oxaloacetic transaminase 2 Rattus norvegicus 263-289 25581390-3 2015 In rats administered 20 and 80 mg/kg rifampicin, the plasma levels of bilirubin glucuronides were elevated, gradually decreased, and almost returned to the baseline level at 24 hours after administration without an elevation of alanine aminotransferase (ALT) and aspartate aminotransferase (AST). Rifampin 37-47 glutamic-oxaloacetic transaminase 2 Rattus norvegicus 291-294 25989893-5 2015 Rifampicin (RIF), a typical OATP inhibitor, inhibited this uptake, indicating the involvement of OATP in DHEAS uptake. Rifampin 12-15 sulfotransferase family 2A member 1 Homo sapiens 105-110 25989893-6 2015 In in vivo experiments, the area under the plasma concentration-time curve (AUC) and maximum plasma concentration (Cmax) of DHEAS were significantly increased following administration of RIF 10 mg/kg, although the extent of this increase was lower than that observed with the test-statins used in this study. Rifampin 187-190 sulfotransferase family 2A member 1 Homo sapiens 124-129 24905729-2 2015 We characterized patients with severe persistent hepatocellular secretory failure (PHSF) and treated them with the pregnane X receptor (PXR) agonist, rifampicin. Rifampin 150-160 nuclear receptor subfamily 1 group I member 2 Homo sapiens 115-134 25535219-4 2015 Rifampicin and rifapentine, two anti-TB drugs now being optimized to shorten TB treatment duration, are potent inducers of CYP3A4. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 123-129 26167828-0 2015 [Rifampicin-resistant Mycobacterium bovis BCG strain isolated from an infant with NEMO mutation]. Rifampin 1-11 inhibitor of nuclear factor kappa B kinase regulatory subunit gamma Homo sapiens 82-86 26167828-2 2015 In this report, a rifampicin-resistant M.bovis BCG strain isolated from an infant with NEMO defect was presented. Rifampin 18-28 inhibitor of nuclear factor kappa B kinase regulatory subunit gamma Homo sapiens 87-91 25628225-4 2015 The prototypical PXR activator rifampicin markedly induced the mRNA and protein expression of SLC13A5 in human primary hepatocytes. Rifampin 31-41 nuclear receptor subfamily 1 group I member 2 Homo sapiens 17-20 25628225-4 2015 The prototypical PXR activator rifampicin markedly induced the mRNA and protein expression of SLC13A5 in human primary hepatocytes. Rifampin 31-41 solute carrier family 13 member 5 Homo sapiens 94-101 24905729-2 2015 We characterized patients with severe persistent hepatocellular secretory failure (PHSF) and treated them with the pregnane X receptor (PXR) agonist, rifampicin. Rifampin 150-160 nuclear receptor subfamily 1 group I member 2 Homo sapiens 136-139 24905729-3 2015 We also studied the effect of rifampicin on PXR-dependent expression of genes involved in biotransformation and secretion in vitro. Rifampin 30-40 nuclear receptor subfamily 1 group I member 2 Homo sapiens 44-47 24905729-11 2015 In vitro, rifampicin PXR-dependently upregulated biotransformation phase 1 (CYP3A4), phase 2 (UGT1A1) and phase 3 (MRP2) enzymes/carriers as well as the basolateral bile salt exporter OSTbeta. Rifampin 10-20 nuclear receptor subfamily 1 group I member 2 Homo sapiens 21-24 24905729-11 2015 In vitro, rifampicin PXR-dependently upregulated biotransformation phase 1 (CYP3A4), phase 2 (UGT1A1) and phase 3 (MRP2) enzymes/carriers as well as the basolateral bile salt exporter OSTbeta. Rifampin 10-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 76-82 24905729-11 2015 In vitro, rifampicin PXR-dependently upregulated biotransformation phase 1 (CYP3A4), phase 2 (UGT1A1) and phase 3 (MRP2) enzymes/carriers as well as the basolateral bile salt exporter OSTbeta. Rifampin 10-20 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 24905729-11 2015 In vitro, rifampicin PXR-dependently upregulated biotransformation phase 1 (CYP3A4), phase 2 (UGT1A1) and phase 3 (MRP2) enzymes/carriers as well as the basolateral bile salt exporter OSTbeta. Rifampin 10-20 ATP binding cassette subfamily C member 2 Homo sapiens 115-119 25712654-2 2015 It is considered a strong constitutive androstane receptor activator, however both CBZ and its main metabolite CBZ 10, 11-epoxide have been reported to be pregnane X receptor (PXR) activators whose maximal efficacy and potency are comparable with the human PXR ligand rifampicin. Rifampin 268-278 nuclear receptor subfamily 1 group I member 2 Homo sapiens 176-179 24905729-11 2015 In vitro, rifampicin PXR-dependently upregulated biotransformation phase 1 (CYP3A4), phase 2 (UGT1A1) and phase 3 (MRP2) enzymes/carriers as well as the basolateral bile salt exporter OSTbeta. Rifampin 10-20 solute carrier family 51 subunit beta Homo sapiens 184-191 24905729-14 2015 PXR-dependent induction of CYP3A4, UGT1A1, MRP2 and OSTbeta could contribute to the anticholestatic effect of rifampicin in PHSF. Rifampin 110-120 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-3 24905729-14 2015 PXR-dependent induction of CYP3A4, UGT1A1, MRP2 and OSTbeta could contribute to the anticholestatic effect of rifampicin in PHSF. Rifampin 110-120 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 27-33 24905729-14 2015 PXR-dependent induction of CYP3A4, UGT1A1, MRP2 and OSTbeta could contribute to the anticholestatic effect of rifampicin in PHSF. Rifampin 110-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 24905729-14 2015 PXR-dependent induction of CYP3A4, UGT1A1, MRP2 and OSTbeta could contribute to the anticholestatic effect of rifampicin in PHSF. Rifampin 110-120 ATP binding cassette subfamily C member 2 Homo sapiens 43-47 24905729-14 2015 PXR-dependent induction of CYP3A4, UGT1A1, MRP2 and OSTbeta could contribute to the anticholestatic effect of rifampicin in PHSF. Rifampin 110-120 solute carrier family 51 subunit beta Homo sapiens 52-59 25545714-1 2015 The present study has been designed to investigate the potential of rifampicin [Pregnane X receptors (PXR) agonist] in experimental dementia. Rifampin 68-78 nuclear receptor subfamily 1, group I, member 2 Rattus norvegicus 80-100 25586052-10 2015 Conversely, the CYP3A inducer rifampicin enhanced AR nuclear localization. Rifampin 30-40 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 16-21 25586052-10 2015 Conversely, the CYP3A inducer rifampicin enhanced AR nuclear localization. Rifampin 30-40 androgen receptor Mus musculus 50-52 25545714-9 2015 Rifampicin-treated AlCl3-rats exhibited significant attenuation in memory deficits, biochemical parameters like AChE activity (33+-1.4), TBARS level (4.1+-1.0), nitrite level (64+-2.6), MPO activity (3.6+-1.0) and GSH level (53+-2.4) along with improved histopathological alterations and locomotor activity when compared with AlCl3-treated rats (p<0.05). Rifampin 0-10 acetylcholinesterase Rattus norvegicus 112-116 25545714-1 2015 The present study has been designed to investigate the potential of rifampicin [Pregnane X receptors (PXR) agonist] in experimental dementia. Rifampin 68-78 nuclear receptor subfamily 1, group I, member 2 Rattus norvegicus 102-105 25545714-9 2015 Rifampicin-treated AlCl3-rats exhibited significant attenuation in memory deficits, biochemical parameters like AChE activity (33+-1.4), TBARS level (4.1+-1.0), nitrite level (64+-2.6), MPO activity (3.6+-1.0) and GSH level (53+-2.4) along with improved histopathological alterations and locomotor activity when compared with AlCl3-treated rats (p<0.05). Rifampin 0-10 myeloperoxidase Rattus norvegicus 186-189 25749104-5 2015 We hypothesized that genes induced by SXR in chondrocytes have a protective effect on articular cartilage and identified Fam20a (family with sequence similarity 20a) as an SXR-dependent gene induced by the known SXR ligands, rifampicin and vitamin K2. Rifampin 225-235 FAM20A, golgi associated secretory pathway pseudokinase Mus musculus 121-127 25545714-10 2015 Combined administration of ketoconazole (a PXR antagonist) and rifampicin to AlCl3-treated animals reversed the rifampicin-induced protective effects. Rifampin 112-122 nuclear receptor subfamily 1, group I, member 2 Rattus norvegicus 43-46 25749104-5 2015 We hypothesized that genes induced by SXR in chondrocytes have a protective effect on articular cartilage and identified Fam20a (family with sequence similarity 20a) as an SXR-dependent gene induced by the known SXR ligands, rifampicin and vitamin K2. Rifampin 225-235 nuclear receptor subfamily 1, group I, member 2 Mus musculus 172-175 25749104-5 2015 We hypothesized that genes induced by SXR in chondrocytes have a protective effect on articular cartilage and identified Fam20a (family with sequence similarity 20a) as an SXR-dependent gene induced by the known SXR ligands, rifampicin and vitamin K2. Rifampin 225-235 nuclear receptor subfamily 1, group I, member 2 Mus musculus 38-41 25749104-5 2015 We hypothesized that genes induced by SXR in chondrocytes have a protective effect on articular cartilage and identified Fam20a (family with sequence similarity 20a) as an SXR-dependent gene induced by the known SXR ligands, rifampicin and vitamin K2. Rifampin 225-235 nuclear receptor subfamily 1, group I, member 2 Mus musculus 172-175 25803093-2 2015 This study assessed the safety, tolerability, and pharmacokinetics of bosutinib when coadministered with the CYP3A4 inducer rifampin in 24 healthy men. Rifampin 124-132 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 109-115 27128217-4 2015 The purpose of these studies was to evaluate the effect of ketoconazole, a potent inhibitor of CYP3A4, and rifampin, a potent inducer of CYP3A4, on the pharmacokinetics of tivozanib. Rifampin 107-115 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 137-143 27128217-8 2015 However, coadministration of tivozanib with rifampin caused a significant decrease in the area under the curve from 0 to infinity and half-life and an increase in clearance of tivozanib, which suggest increased clearance via the enhanced CYP3A4-mediated metabolism of tivozanib. Rifampin 44-52 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 238-244 25809761-3 2015 Progress has been made in clinic-based detection tools with the introduction of Xpert MTB/RIF, a nucleic acid amplification test that combines sample processing and analysis in a single instrument to provide a diagnostic result and detection of resistance to rifampicin in under 2h. Rifampin 259-269 ras homolog family member F, filopodia associated Homo sapiens 90-93 25418376-9 2015 By using telmisartan (a non-specific OATP inhibitor) and rifampicin, OATP2B1 was demonstrated to account for <20% of hepatic HMME uptake. Rifampin 57-67 solute carrier organic anion transporter family member 2B1 Homo sapiens 69-76 25658091-8 2015 Upfront Xpert MTB/RIF testing of presumptive PTB and DR-TB cases resulted in diagnosis of 73 (9.5%, CI 7.6-11.8) and 16 (11.2%, CI 6.7-17.1) rifampicin resistance cases, respectively. Rifampin 141-151 ras homolog family member F, filopodia associated Homo sapiens 18-21 25733917-4 2015 As a substrate of CYP 3A4, dolutegravir is affected by rifampin, efavirenz, tipranavir/ritonavir, fosamprenavir/ritonavir, and dose increase is required. Rifampin 55-63 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 18-25 25479155-6 2015 RESULTS: Administration of both rifampicin and BCG-LPS elicited hepatic damage reflected in significantly (p<0.01) increased serum marker enzymes AST, ALT, ALP, GGT, LDH and decreased total proteins, increased MDA and NO formation in liver homogenate. Rifampin 32-42 PDZ and LIM domain 3 Rattus norvegicus 159-162 25670521-4 2015 In vitro reevaluation at therapeutically relevant low-nanomolar concentrations of unbound ASV showed active, saturable human hepatocyte uptake (Km = 0.685 muM) and rifampin-reversible transport by OATP1B1 and OATP2B1, but not OATP1B3. Rifampin 164-172 solute carrier organic anion transporter family member 1B1 Homo sapiens 197-204 25407255-2 2015 Canagliflozin exposures may be affected by coadministration of drugs that induce (e.g., rifampin for UGT) or inhibit (e.g. probenecid for UGT; cyclosporine A for P-gp) these pathways. Rifampin 88-96 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 101-104 25670521-3 2015 Although no significant OATP transport of ASV was observed in vitro at standard micromolar assay concentrations, clinical coadministration of ASV with a single dose of the OATP inhibitor rifampin gave large, variable increases in ASV plasma Cmax (21-fold mean) and AUCinf (15-fold mean), consistent with reduced hepatic uptake. Rifampin 187-195 solute carrier organic anion transporter family member 1A2 Homo sapiens 24-28 25670521-3 2015 Although no significant OATP transport of ASV was observed in vitro at standard micromolar assay concentrations, clinical coadministration of ASV with a single dose of the OATP inhibitor rifampin gave large, variable increases in ASV plasma Cmax (21-fold mean) and AUCinf (15-fold mean), consistent with reduced hepatic uptake. Rifampin 187-195 solute carrier organic anion transporter family member 1A2 Homo sapiens 172-176 25263393-10 2015 Therefore, the interactive mechanism of multidose rifampicin may occur through a combination of OATP and P-gp transporters, thereby altering the pharmacokinetics of fexofenadine enantiomers. Rifampin 50-60 ATP binding cassette subfamily B member 1 Homo sapiens 105-109 25598834-2 2015 RAL should be double-dosed to 800 mg semi-daily in situation of rifampicin co-medication, because RAL is more rapidly metabolized due to rifampicin-induced Uridine-5"-diphosph- gluronosyl-transferase (UGT1A1). Rifampin 64-74 RAS like proto-oncogene A Homo sapiens 0-3 25653502-4 2015 The increased uptake of catalposide via the OAT3, OATP1B1, and OATP1B3 transporters was decreased to basal levels in the presence of representative inhibitors such as probenecid, furosemide, and cimetidine (for OAT3) and cyclosporin A, gemfibrozil, and rifampin (for OATP1B1 and OATP1B3). Rifampin 253-261 solute carrier family 22 member 8 Homo sapiens 44-48 25653502-4 2015 The increased uptake of catalposide via the OAT3, OATP1B1, and OATP1B3 transporters was decreased to basal levels in the presence of representative inhibitors such as probenecid, furosemide, and cimetidine (for OAT3) and cyclosporin A, gemfibrozil, and rifampin (for OATP1B1 and OATP1B3). Rifampin 253-261 solute carrier organic anion transporter family member 1B3 Homo sapiens 63-70 25263393-1 2015 WHAT IS KNOWN AND OBJECTIVE: Rifampicin is a potent inducer of P-glycoprotein (P-gp) and inhibitor of organic anion-transporting polypeptides (OATPs), with fexofenadine acting as a substrate for both mechanisms. Rifampin 29-39 ATP binding cassette subfamily B member 1 Homo sapiens 63-77 25263393-1 2015 WHAT IS KNOWN AND OBJECTIVE: Rifampicin is a potent inducer of P-glycoprotein (P-gp) and inhibitor of organic anion-transporting polypeptides (OATPs), with fexofenadine acting as a substrate for both mechanisms. Rifampin 29-39 ATP binding cassette subfamily B member 1 Homo sapiens 79-83 25263393-2 2015 Simultaneous administration of single- or multiple-dose rifampicin 600 mg significantly increases the concentrations of fexofenadine enantiomers by inhibiting OATP transporters. Rifampin 56-66 solute carrier organic anion transporter family member 1A2 Homo sapiens 159-163 25263393-8 2015 These results indicated that multiple doses of rifampicin 450 mg may be sufficient to inhibit the renal influx transporter and OATP-mediated hepatic uptake of both enantiomers. Rifampin 47-57 solute carrier organic anion transporter family member 1A2 Homo sapiens 127-131 25263393-9 2015 Moreover, these effects may be greater than the P-gp-inductive effects of rifampicin. Rifampin 74-84 ATP binding cassette subfamily B member 1 Homo sapiens 48-52 25263393-10 2015 Therefore, the interactive mechanism of multidose rifampicin may occur through a combination of OATP and P-gp transporters, thereby altering the pharmacokinetics of fexofenadine enantiomers. Rifampin 50-60 solute carrier organic anion transporter family member 1A2 Homo sapiens 96-100 25648393-14 2015 Imipenem/meropenem/rifampicin in combination with polymyxin B demonstrated consistent bactericidal effect against 49 bla OXA-23-harbouring XDR A. baumannii clinical isolates, suggesting a role of combination therapy in the treatment of these infections. Rifampin 19-29 class D beta-lactamase OXA-23 Acinetobacter baumannii 121-127 25598834-2 2015 RAL should be double-dosed to 800 mg semi-daily in situation of rifampicin co-medication, because RAL is more rapidly metabolized due to rifampicin-induced Uridine-5"-diphosph- gluronosyl-transferase (UGT1A1). Rifampin 137-147 RAS like proto-oncogene A Homo sapiens 0-3 25598834-2 2015 RAL should be double-dosed to 800 mg semi-daily in situation of rifampicin co-medication, because RAL is more rapidly metabolized due to rifampicin-induced Uridine-5"-diphosph- gluronosyl-transferase (UGT1A1). Rifampin 137-147 RAS like proto-oncogene A Homo sapiens 98-101 25598834-2 2015 RAL should be double-dosed to 800 mg semi-daily in situation of rifampicin co-medication, because RAL is more rapidly metabolized due to rifampicin-induced Uridine-5"-diphosph- gluronosyl-transferase (UGT1A1). Rifampin 137-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 201-207 25598834-3 2015 Recently, it was speculated that chewed RAL might lead to increased absorption, which might compensate the inductive effect of rifampicin-rapid metabolized RAL, as part of cost-saving effects in countries with high-tuberculosis prevalence and less economic power. Rifampin 127-137 RAS like proto-oncogene A Homo sapiens 156-159 25069801-5 2015 Protective effects of AITC on rifampin-induced cytotoxicity were observed, and transient transfection assays showed that AITC was able to effectively attenuate the agonist effects of rifampin and CITCO on human PXR and CAR activity, respectively. Rifampin 183-191 nuclear receptor subfamily 1 group I member 2 Homo sapiens 211-214 26168679-4 2015 The highest levels of the MRSA resistance were stated against ciprofloxacin--92%(MIC50 32 mcg/ml), gentamicin--85% (MIC50 128 mcg/ml), erythromycin--54% (MIC50 32-mcg/ml) and clindainycin - 45% (MIC50 0.03 mcg/ml), as well as against rifampicin--38% (MIC50 0.06 mcg/ml). Rifampin 234-244 solute carrier family 9 member A6 Homo sapiens 26-30 26094899-2 2015 Rifampicin, rifabutin, and thioacetazone inhibited one CYP reaction. Rifampin 0-10 peptidylprolyl isomerase G Homo sapiens 55-58 26574146-12 2015 Unlike CYP1 and 3 and other CYP2 members, CYP2D6 is resistant to typical inducers such as rifampin, phenobarbital and dexamethasone. Rifampin 90-98 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 42-48 27128001-2 2015 Hence, the potential for drug-drug interactions when alisporivir is co-administered with CYP3A4 and/or P-gp inhibitors such as ketoconazole, azithromycin and CYP3A4 inducers such as rifampin were evaluated in three separate clinical studies. Rifampin 182-190 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 158-164 27128001-5 2015 Rifampin (a CYP3A4 inducer) caused an approximate 90% reduction in alisporivir Cmax and AUC and a fourfold reduction in alisporivir terminal elimination half-life. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 12-18 26405974-2 2015 Previous studies have shown that rifampin can indirectly influence drug deposition through the regulation of molecular interactions of miRNA, PXR and other genes. Rifampin 33-41 nuclear receptor subfamily 1 group I member 2 Homo sapiens 142-145 24464437-10 2015 A short course treatment with isoniazid and rifampin provided efficacy, good tolerability and good completion rate in patients with rheumatic conditions proposed for anti-TNF therapy. Rifampin 44-52 tumor necrosis factor Homo sapiens 171-174 25351763-3 2015 The predominant role of OATPs was further supported by a dramatic inhibition of flavopiridol uptake in the presence of the OATP substrate rifampicin. Rifampin 138-148 solute carrier organic anion transporter family member 1A2 Homo sapiens 24-28 25600204-2 2015 To evaluate time-dependent cytochrome P450 induction precisely, induction of CYP1A2, CYP2B6, and CYP3A4 mRNA was confirmed (>2-fold) by the treatment with omeprazole, phenobarbital, and rifampicin, respectively, for 24 or 48 h on day 3 from the start of culture. Rifampin 189-199 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 77-83 25600204-2 2015 To evaluate time-dependent cytochrome P450 induction precisely, induction of CYP1A2, CYP2B6, and CYP3A4 mRNA was confirmed (>2-fold) by the treatment with omeprazole, phenobarbital, and rifampicin, respectively, for 24 or 48 h on day 3 from the start of culture. Rifampin 189-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 25209610-2 2015 The aim of this study was to assess the non-inferiority of a combination of trimethoprim/sulfamethoxazole plus rifampicin versus linezolid alone for the treatment of MRSA infection. Rifampin 111-121 solute carrier family 9 member A6 Homo sapiens 166-170 25209610-13 2015 CONCLUSIONS: Compared with linezolid, trimethoprim/sulfamethoxazole and rifampicin seems to be non-inferior in the treatment of MRSA infection. Rifampin 72-82 solute carrier family 9 member A6 Homo sapiens 128-132 24934663-8 2015 A combination of rifampin and clarithromycin released from the polymer film provided >99.9% kill of an MRSA inoculate for up to 72 h. CONCLUSION: Results showed that combining multiple drugs in copolymer films with unique surface properties, initial hydrophilicity and increase in roughness, can be an effective way to prevent biofilm formation. Rifampin 17-25 solute carrier family 9 member A6 Homo sapiens 106-110 26506671-4 2015 MATERIAL AND METHODS: The specific zone mutations in rpoB, katG and inhA gene for rifampicin and isoniazid were investigated with molecular methods in 198 recently isolated unique strains from patients diagnosed with pulmonary tuberculosis. Rifampin 82-92 inhibin subunit alpha Homo sapiens 68-72 25761590-8 2015 Inhibitors of MRP2, (MK571 and rifampicin) and co-incubation with kaempferol (10 muM), increased transfer from the apical to the basolateral side by three to five fold. Rifampin 31-41 ATP binding cassette subfamily C member 2 Homo sapiens 14-18 25886055-0 2015 The inhibition of hepatic bile acids transporters Ntcp and Bsep is involved in the pathogenesis of isoniazid/rifampicin-induced hepatotoxicity. Rifampin 109-119 solute carrier family 10 (sodium/bile acid cotransporter family), member 1 Mus musculus 50-54 25886055-0 2015 The inhibition of hepatic bile acids transporters Ntcp and Bsep is involved in the pathogenesis of isoniazid/rifampicin-induced hepatotoxicity. Rifampin 109-119 ATP-binding cassette, sub-family B (MDR/TAP), member 11 Mus musculus 59-63 25625052-0 2014 Verapamil and rifampin effect on p-glycoprotein expression in hepatocellular carcinoma. Rifampin 14-22 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 33-47 26068524-5 2015 Our studies indicated that berberine uptake was significantly suppressed by rifampicin, cyclosporine A and glycyrrhizic acid, which act as specific inhibitors of different Oatp isoforms (Oatp1a1, Oatp1a4 and Oatp1b2) in rat hepatocytes. Rifampin 76-86 solute carrier organic anion transporter family member 1A2 Homo sapiens 172-176 25096076-1 2014 OBJECTIVES: To assess the effect of the major efavirenz metabolizing enzyme (CYP2B6) genotype and the effects of rifampicin co-treatment on induction of CYP3A by efavirenz. Rifampin 113-123 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 153-158 25047139-12 2014 WHAT IS NEW AND CONCLUSION: Co-administration navitoclax with rifampin moderately decreased navitoclax AUC, which could be partly due to the induction effect of rifampin on CYP3A4. Rifampin 62-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 173-179 25047139-12 2014 WHAT IS NEW AND CONCLUSION: Co-administration navitoclax with rifampin moderately decreased navitoclax AUC, which could be partly due to the induction effect of rifampin on CYP3A4. Rifampin 161-169 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 173-179 25302422-0 2014 Protective effects of resveratrol on hepatotoxicity induced by isoniazid and rifampicin via SIRT1 modulation. Rifampin 77-87 sirtuin 1 Mus musculus 92-97 25302422-8 2014 The results presented herein show that 1 was able to largely prevent the hepatotoxicity induced by isoniazid and rifampicin in mice, mainly by modulating SIRT1 mRNA expression. Rifampin 113-123 sirtuin 1 Mus musculus 154-159 25008118-6 2014 The model predicted an alisporivir AUC increase by 9.4-fold and a decrease by 86% when alisporivir was co-administrated with ketoconazole (CYP3A4 inhibitor) or rifampin (CYP3A4 inducer), respectively. Rifampin 160-168 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 170-176 26521872-4 2015 We found that flutamide, phenacetin, rifamycins (rifampicin, rifabutin, and rifapentine), and indiplon are hydrolyzed by arylacetamide deacetylase (AADAC), which is highly expressed in human liver and gastrointestinal tissues. Rifampin 49-59 arylacetamide deacetylase Homo sapiens 148-153 25395048-1 2014 BACKGROUND: The Xpert MTB/RIF assay (Xpert assay; Cepheid, Sunnyvale, CA) is becoming the test of choice for the rapid diagnosis of tuberculosis and rifampin (RIF) resistance. Rifampin 149-157 ras homolog family member F, filopodia associated Homo sapiens 16-29 24962564-9 2014 However, the effect of CYP3A4 induction by rifampicin on apremilast clearance is much more pronounced than that of CYP3A4 inhibition by ketoconazole. Rifampin 43-53 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 23-29 25625052-3 2014 OBJECTIVES: The aim of this study was to elucidate the effect of potent carcinogen nitrosamine with and without verapamil and rifampin drugs on P-gp expression at the mRNA level in HCC. Rifampin 126-134 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 144-148 25625052-8 2014 Rifampin administrated group had a decreased level of the mdr1a mRNA compared to the control group (P <= 0.006). Rifampin 0-8 ATP binding cassette subfamily B member 1A Rattus norvegicus 58-63 25625052-11 2014 CONCLUSIONS: Verapamil and rifampin were found specific and effective against P-gp expression in HCC. Rifampin 27-35 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 78-82 25022720-8 2014 RESULTS: Single-dose rifampicin (P-gp inhibition) increased area under the plasma concentration-time curve from time zero to infinity (AUC0- ) of free and total lenvatinib by 32 and 31 %, respectively. Rifampin 21-31 ATP binding cassette subfamily B member 1 Homo sapiens 33-37 25145495-7 2014 Additionally, using a chequerboard assay, we have shown that hepcidin has an antagonistic effect in combination with the antibiotics rifampicin and tetracycline against Staphylococcus aureus, Pseudomonas aeruginosa and Streptococcus pyogenes, evidenced by a fractional inhibitory concentration index (FICI) > 4. Rifampin 133-143 hepcidin antimicrobial peptide Homo sapiens 61-69 24617605-8 2014 In vitro studies showed that rifampicin was an inhibitor of OATP1B1 and OATP1B3, breast cancer resistance protein and multidrug resistance protein 2, but not of organic anion transporter 3. Rifampin 29-39 solute carrier organic anion transporter family member 1B1 Homo sapiens 60-67 24617605-8 2014 In vitro studies showed that rifampicin was an inhibitor of OATP1B1 and OATP1B3, breast cancer resistance protein and multidrug resistance protein 2, but not of organic anion transporter 3. Rifampin 29-39 solute carrier organic anion transporter family member 1B3 Homo sapiens 72-79 25070100-9 2014 In human hepatocytes, ketoconazole dose dependently inhibited the formation of N(+)-glucuronides (50% inhibitory concentration [IC50], 1.5 muM), while rifampin induced the mRNA level of UGT1A9 by 28% and the activity of UGT1A9 by 53%. Rifampin 151-159 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 186-192 25070100-9 2014 In human hepatocytes, ketoconazole dose dependently inhibited the formation of N(+)-glucuronides (50% inhibitory concentration [IC50], 1.5 muM), while rifampin induced the mRNA level of UGT1A9 by 28% and the activity of UGT1A9 by 53%. Rifampin 151-159 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 220-226 25074870-4 2014 The amount of the unphosphorylated wild-type yellow fluorescent protein-hPXR fusion protein but not the T290A mutant increased on Phos-tag gels in response to stimulations with rifampicin and cyclin-dependent kinase 2 inhibitor roscovitine, and a marked increase was observed in the unphosphorylated levels of the T290A mutant in nontreated cells. Rifampin 177-187 nuclear receptor subfamily 1 group I member 2 Homo sapiens 72-76 25074870-8 2014 After the rifampicin and roscovitine stimulations, PP1 was recruited to the wild-type hPXR but not the T290A mutant. Rifampin 10-20 neuropeptide Y receptor Y4 Homo sapiens 51-54 25074870-8 2014 After the rifampicin and roscovitine stimulations, PP1 was recruited to the wild-type hPXR but not the T290A mutant. Rifampin 10-20 nuclear receptor subfamily 1 group I member 2 Homo sapiens 86-90 25119251-0 2014 Rifampicin improves neuronal apoptosis in LPS-stimulated co-cultured BV2 cells through inhibition of the TLR-4 pathway. Rifampin 0-10 toll-like receptor 4 Mus musculus 42-45 25119251-0 2014 Rifampicin improves neuronal apoptosis in LPS-stimulated co-cultured BV2 cells through inhibition of the TLR-4 pathway. Rifampin 0-10 toll-like receptor 4 Mus musculus 105-110 25119251-3 2014 Our previous study showed that rifampicin inhibits the production of lipopolysaccharide (LPS)-induced pro-inflammatory mediators and improves neuron survival in inflammation; however, the mechanism through which rifampicin inhibits microglial inflammation and its neuroprotective effects are not completely understood. Rifampin 31-41 toll-like receptor 4 Mus musculus 89-92 25119251-3 2014 Our previous study showed that rifampicin inhibits the production of lipopolysaccharide (LPS)-induced pro-inflammatory mediators and improves neuron survival in inflammation; however, the mechanism through which rifampicin inhibits microglial inflammation and its neuroprotective effects are not completely understood. Rifampin 212-222 toll-like receptor 4 Mus musculus 89-92 25119251-4 2014 In this study, we examined the effects of rifampicin on morphological changes induced by LPS in murine microglial BV2 cells. Rifampin 42-52 toll-like receptor 4 Mus musculus 89-92 25119251-5 2014 Then we investigated, in BV2 microglia, the effects of rifampicin on two signaling pathway componentss stimulated by LPS, the Toll-like receptor-4 (TLR-4) and the nuclear factor-kappaB (NF-kappaB). Rifampin 55-65 toll-like receptor 4 Mus musculus 117-120 25119251-5 2014 Then we investigated, in BV2 microglia, the effects of rifampicin on two signaling pathway componentss stimulated by LPS, the Toll-like receptor-4 (TLR-4) and the nuclear factor-kappaB (NF-kappaB). Rifampin 55-65 toll-like receptor 4 Mus musculus 126-146 25119251-7 2014 Rifampicin inhibited LPS-stimulated expression of the TLR-4 gene. Rifampin 0-10 toll-like receptor 4 Mus musculus 21-24 25119251-7 2014 Rifampicin inhibited LPS-stimulated expression of the TLR-4 gene. Rifampin 0-10 toll-like receptor 4 Mus musculus 54-59 25119251-8 2014 When neurons were co-cultured with LPS-stimulated BV2 microglia, pre-treatment with rifampicin increased neuronal viability and reduced the number of apoptotic cells. Rifampin 84-94 toll-like receptor 4 Mus musculus 35-38 25022720-9 2014 Multiple-dose rifampicin (simultaneous P-gp and CYP3A4 induction) decreased lenvatinib AUC0- (total: 18 %; free: 9 %). Rifampin 14-24 ATP binding cassette subfamily B member 1 Homo sapiens 39-43 25022720-9 2014 Multiple-dose rifampicin (simultaneous P-gp and CYP3A4 induction) decreased lenvatinib AUC0- (total: 18 %; free: 9 %). Rifampin 14-24 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-54 24890593-6 2014 Respiratory epithelial cell-derived MMP-3 was suppressed by moxifloxacin, rifampicin, and azithromycin in a dose-dependent manner. Rifampin 74-84 matrix metallopeptidase 3 Homo sapiens 36-41 24996840-1 2014 We aimed to analyze the efficacy and safety of high doses of daptomycin (10 mg/kg/d) plus rifampin (D10 + R) for prosthetic joint infection (PJI). Rifampin 90-98 CHRNA7 (exons 5-10) and FAM7A (exons A-E) fusion Homo sapiens 100-103 25361020-3 2014 Xpert MTB/RIF allows for the rapid diagnosis of Mycobacterium tuberculosis and identification of rifampicin susceptibility. Rifampin 97-107 ras homolog family member F, filopodia associated Homo sapiens 10-13 25141173-7 2014 Lansoprazole dose-dependently activated pregnane X receptor PXR in gene reporter assays, and slightly modulated rifampicin-inducible PXR activity, with similar potency for each enantiomer. Rifampin 112-122 nuclear receptor subfamily 1 group I member 2 Homo sapiens 133-136 25141173-8 2014 Omeprazole dose-dependently activated PXR and inhibited rifampicin-inducible PXR activity. Rifampin 56-66 nuclear receptor subfamily 1 group I member 2 Homo sapiens 77-80 25158258-6 2014 In addition, increased rifampicin concentrations inhibited the formation of calcium nodules, OPN/BGP, and COL1A1 in osteoblasts after 28 days of induction. Rifampin 23-33 secreted phosphoprotein 1 Homo sapiens 93-96 25158258-6 2014 In addition, increased rifampicin concentrations inhibited the formation of calcium nodules, OPN/BGP, and COL1A1 in osteoblasts after 28 days of induction. Rifampin 23-33 bone gamma-carboxyglutamate protein Homo sapiens 97-100 25158258-6 2014 In addition, increased rifampicin concentrations inhibited the formation of calcium nodules, OPN/BGP, and COL1A1 in osteoblasts after 28 days of induction. Rifampin 23-33 collagen type I alpha 1 chain Homo sapiens 106-112 25140877-10 2014 Upfront Xpert MTB/RIF testing of presumptive PTB and presumptive DR-TB cases resulted in diagnosis of 79 and 12 rifampicin resistance cases, respectively. Rifampin 112-122 ras homolog family member F, filopodia associated Homo sapiens 18-21 25140877-12 2014 CONCLUSION: Upfront access to Xpert MTB/RIF testing in pediatric presumptive PTB cases was associated with a two-fold increase in bacteriologically-confirmed PTB, and increased detection of rifampicin-resistant TB cases under routine operational conditions across India. Rifampin 190-200 ras homolog family member F, filopodia associated Homo sapiens 40-43 25129689-1 2014 BACKGROUND: An algorithm instituted following Xpert MTB/RIF (Xpert) introduction in South Africa advocates for treating all Xpert rifampicin resistant patients as MDR-TB cases while awaiting confirmation by phenotypic or genotypic drug susceptibility testing. Rifampin 130-140 ras homolog family member F, filopodia associated Homo sapiens 56-59 25116663-9 2014 Polymorphism frequency in the SLCO1B1 (rs4149032) drug transporter gene was high (0.76) and was associated with low median rifampicin C2.5 hr, 3.7 (2.8-5.0) microg/mL in the heterozygous and 3.4 (2.7-4.7) microg/mL in the homozygous variant carriers. Rifampin 123-133 solute carrier organic anion transporter family member 1B1 Homo sapiens 30-37 25116663-13 2014 CONCLUSION: Approximated peak rifampicin concentrations were well below the recommended target range of 8 to 24 microg/mL in this patient population with its high frequency of the SLCO1B1 (rs4149032) polymorphism. Rifampin 30-40 solute carrier organic anion transporter family member 1B1 Homo sapiens 180-187 24839948-0 2014 Comparison of two endogenous biomarkers of CYP3A4 activity in a drug-drug interaction study between midostaurin and rifampicin. Rifampin 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 43-49 24839948-11 2014 CONCLUSIONS: Both 6betaCR and 4betaHC levels showed a good dynamic response range upon strong CYP3A4 induction with rifampicin. Rifampin 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 94-100 24752503-4 2014 Here, we present a simple suspension culture of aggregates of ES cell-derived hepatocytes that compared to conventional monolayer adherent culture significantly increases induction of CYP 1A2 by omeprazole and 3A4 by rifampicin. Rifampin 217-227 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 184-191 24820076-4 2014 In vitro studies were conducted to determine whether one or more of the antituberculosis drugs (rifampin, isoniazid, pyrazinamide, or ethambutol) potently inhibit efavirenz 8-hydroxylation by CYP2B6 or efavirenz 7-hydroxylation by CYP2A6, the main mechanisms of efavirenz clearance. Rifampin 96-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-198 24820076-4 2014 In vitro studies were conducted to determine whether one or more of the antituberculosis drugs (rifampin, isoniazid, pyrazinamide, or ethambutol) potently inhibit efavirenz 8-hydroxylation by CYP2B6 or efavirenz 7-hydroxylation by CYP2A6, the main mechanisms of efavirenz clearance. Rifampin 96-104 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 231-237 24627312-6 2014 RESULTS: The anthracycline Antibiotic 301A(1) was isolated from the fermentation broth of a strain of Streptomyces (WAC450); the molecule was shown to be highly synergistic with rifampicin (fractional inhibitory concentration index = 0.156) and moderately synergistic with linezolid (FIC index = 0.25) in both E. coli and Acinetobacter baumannii. Rifampin 178-188 Fic Escherichia coli 284-287 24446527-7 2014 Finally, adjunctive transferrin reduced the emergence of rifampin-resistant mutants of S. aureus in infected mice treated with rifampin. Rifampin 57-65 transferrin Homo sapiens 20-31 24446527-7 2014 Finally, adjunctive transferrin reduced the emergence of rifampin-resistant mutants of S. aureus in infected mice treated with rifampin. Rifampin 127-135 transferrin Homo sapiens 20-31 24725118-5 2014 A2 increased the mRNA expression of OATP1B3 (an organic anion-transporting polypeptide) and uptake of A2 was inhibited by rifampin (inhibitor of OATP1B3), which indicated that the transporter-mediated transport of A2 was mediated by OATP1B3. Rifampin 122-130 solute carrier organic anion transporter family member 1B3 Homo sapiens 36-43 24725118-5 2014 A2 increased the mRNA expression of OATP1B3 (an organic anion-transporting polypeptide) and uptake of A2 was inhibited by rifampin (inhibitor of OATP1B3), which indicated that the transporter-mediated transport of A2 was mediated by OATP1B3. Rifampin 122-130 solute carrier organic anion transporter family member 1B3 Homo sapiens 145-152 24725118-5 2014 A2 increased the mRNA expression of OATP1B3 (an organic anion-transporting polypeptide) and uptake of A2 was inhibited by rifampin (inhibitor of OATP1B3), which indicated that the transporter-mediated transport of A2 was mediated by OATP1B3. Rifampin 122-130 solute carrier organic anion transporter family member 1B3 Homo sapiens 145-152 24742608-3 2014 Hepatic damage induced in Wistar rats by isoniazid and rifampicin resulted in significant alterations in biomarkers of liver function, namely, bilirubin, aspartate transaminase, alanine transaminase, alkaline phosphatase, and oxidative stress markers such as superoxide dismutase, catalase, glutathione, and thiobarbituric acid reactive substances. Rifampin 55-65 catalase Rattus norvegicus 281-289 24964282-1 2014 A Bayesian mechanism-based pharmacokinetic/pharmacodynamic model of cytochrome P450 3A4 (CYP3A4) activity was developed based on a clinical study of the effects of ketoconazole and rifampin on midazolam exposure and plasma 4beta-hydroxycholesterol (4betaHC) concentrations. Rifampin 181-189 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-87 24880706-9 2014 The up-regulation of Th1 immunity by piperine can be synergistically combined with rifampicin to improve its therapeutic efficacy in immune-compromised TB patients. Rifampin 83-93 negative elongation factor complex member C/D Homo sapiens 21-24 24964282-1 2014 A Bayesian mechanism-based pharmacokinetic/pharmacodynamic model of cytochrome P450 3A4 (CYP3A4) activity was developed based on a clinical study of the effects of ketoconazole and rifampin on midazolam exposure and plasma 4beta-hydroxycholesterol (4betaHC) concentrations. Rifampin 181-189 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 89-95 24641623-9 2014 Induction of UGT1A4 and UGT1A3 gene expression was also determined in human hepatocytes treated with pregnane X receptor/constitutive androstane receptor agonists, such as rifampin, carbamazepine, and phenobarbital. Rifampin 172-180 UDP glucuronosyltransferase family 1 member A4 Homo sapiens 13-19 25420292-4 2014 The mutations that appear for Rifampicin are in region rpoB, for Isoniazid in region katG and inhA, for Ethambutol-embB, Quinolone-gyrA, Aminoglicozid and Cyclical Peptides-rrs. Rifampin 30-40 inhibin subunit alpha Homo sapiens 94-98 24530864-0 2014 Physiologically based pharmacokinetic modeling of CYP3A4 induction by rifampicin in human: influence of time between substrate and inducer administration. Rifampin 70-80 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-56 24530864-2 2014 Rifampicin (RIF) is a potent inducer of CYP3A4 and also acts as a competitive inhibitor which can partially mask the induction. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-46 24530864-2 2014 Rifampicin (RIF) is a potent inducer of CYP3A4 and also acts as a competitive inhibitor which can partially mask the induction. Rifampin 12-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-46 24777631-6 2014 Rifampicin is also an inducer of the uridine diphosphate glucuronosyltransferase (UGT) 1A1 enzymes and interferes with drugs, such as integrase inhibitors, that are metabolized by this metabolic pathway. Rifampin 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-90 24641623-9 2014 Induction of UGT1A4 and UGT1A3 gene expression was also determined in human hepatocytes treated with pregnane X receptor/constitutive androstane receptor agonists, such as rifampin, carbamazepine, and phenobarbital. Rifampin 172-180 UDP glucuronosyltransferase family 1 member A3 Homo sapiens 24-30 24486740-4 2014 Functionally, activation of human PXR by rifampicin or ectopic expression of constitutively-activated human VP-PXR inhibited cervical cell proliferation. Rifampin 41-51 nuclear receptor subfamily 1 group I member 2 Homo sapiens 34-37 24553381-6 2014 In contrast, of the 8 hepatocyte lots treated with rifampicin, CYP2C8 and CYP2C9 mRNA were not induced in 5 and 2 hepatocyte lots, respectively, and CYP2C19 mRNA was not induced in any of the 8 hepatocyte lots tested. Rifampin 51-61 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 63-69 24486481-9 2014 Stimulation of Caco-2 intestinal epithelial cells with rifaximin, rifampicin and SR12813, all potent agonists of the PXR, significantly increased wound closure. Rifampin 66-76 nuclear receptor subfamily 1 group I member 2 Homo sapiens 117-120 24553381-5 2014 The results revealed that CYP1A2, CYP2B6, and CYP3A4 were induced (>2.0-fold) by omeprazole, phenobarbital, and rifampicin, respectively, in all the hepatocyte lots tested at enzyme activity level (23 lots) and mRNA level (8 lots). Rifampin 115-125 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 26-32 24553381-5 2014 The results revealed that CYP1A2, CYP2B6, and CYP3A4 were induced (>2.0-fold) by omeprazole, phenobarbital, and rifampicin, respectively, in all the hepatocyte lots tested at enzyme activity level (23 lots) and mRNA level (8 lots). Rifampin 115-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 24553381-5 2014 The results revealed that CYP1A2, CYP2B6, and CYP3A4 were induced (>2.0-fold) by omeprazole, phenobarbital, and rifampicin, respectively, in all the hepatocyte lots tested at enzyme activity level (23 lots) and mRNA level (8 lots). Rifampin 115-125 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 46-52 24595680-4 2014 Following multiple oral administration of rifampicin (a known CYP3A inducer) at 15 mg/kg/d in cynomolgus monkeys, the mean serum 4beta-hydroxycholesterol levels increased 4-fold from the baseline of 55.3 +- 21.7 to 221 +- 53.4 ng/ml. Rifampin 42-52 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-67 24595680-8 2014 The serum exposure (area under the curve) of midazolam was markedly decreased by ~95%, confirming the induction of CYP3A catalytic activity by rifampicin treatment in monkeys. Rifampin 143-153 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 115-120 24788541-10 2014 In addition, all the above expression levels in the transfected cell models could be further induced with additional treatment of Rifampicin, a specific inducer for CYP3A4. Rifampin 130-140 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 165-171 24733486-7 2014 Furthermore, the CYP3A4 activity in the HNF1alpha-transfected cells could be induced by CYP3A4-specific inducer, rifampicin, and metabolized nifedipine in a dose-dependent manner. Rifampin 113-123 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 17-23 24733486-7 2014 Furthermore, the CYP3A4 activity in the HNF1alpha-transfected cells could be induced by CYP3A4-specific inducer, rifampicin, and metabolized nifedipine in a dose-dependent manner. Rifampin 113-123 HNF1 homeobox A Homo sapiens 40-49 24733486-7 2014 Furthermore, the CYP3A4 activity in the HNF1alpha-transfected cells could be induced by CYP3A4-specific inducer, rifampicin, and metabolized nifedipine in a dose-dependent manner. Rifampin 113-123 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-94 24420910-9 2014 However, drugs that could specifically interfere with the metabolic pathways of SGLT2 inhibitors [rifampicin, inhibitors or inducers of uridine diphosphate-glucuronosyltransferase (UGT)] may result in significant changes in the exposure of SGLT2 inhibitors, as shown for dapagliflozin and canagliflozin. Rifampin 98-108 solute carrier family 5 member 2 Homo sapiens 80-85 24464802-7 2014 In a panel of 24 individual HLM samples, the indiplon hydrolase activities were significantly correlated with the hydrolase activities of flutamide, phenacetin, and rifampicin, which are known AADAC substrates. Rifampin 165-175 oxysterol binding protein 2 Homo sapiens 28-31 24464802-7 2014 In a panel of 24 individual HLM samples, the indiplon hydrolase activities were significantly correlated with the hydrolase activities of flutamide, phenacetin, and rifampicin, which are known AADAC substrates. Rifampin 165-175 arylacetamide deacetylase Homo sapiens 193-198 24420910-9 2014 However, drugs that could specifically interfere with the metabolic pathways of SGLT2 inhibitors [rifampicin, inhibitors or inducers of uridine diphosphate-glucuronosyltransferase (UGT)] may result in significant changes in the exposure of SGLT2 inhibitors, as shown for dapagliflozin and canagliflozin. Rifampin 98-108 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 136-179 24420910-9 2014 However, drugs that could specifically interfere with the metabolic pathways of SGLT2 inhibitors [rifampicin, inhibitors or inducers of uridine diphosphate-glucuronosyltransferase (UGT)] may result in significant changes in the exposure of SGLT2 inhibitors, as shown for dapagliflozin and canagliflozin. Rifampin 98-108 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 181-184 24420910-9 2014 However, drugs that could specifically interfere with the metabolic pathways of SGLT2 inhibitors [rifampicin, inhibitors or inducers of uridine diphosphate-glucuronosyltransferase (UGT)] may result in significant changes in the exposure of SGLT2 inhibitors, as shown for dapagliflozin and canagliflozin. Rifampin 98-108 solute carrier family 5 member 2 Homo sapiens 240-245 24638036-0 2014 Rifampicin protects PC12 cells from rotenone-induced cytotoxicity by activating GRP78 via PERK-eIF2alpha-ATF4 pathway. Rifampin 0-10 heat shock protein family A (Hsp70) member 5 Rattus norvegicus 80-85 24447252-0 2014 In vitro activity of rifampicin alone and in combination with imipenem against multidrug-resistant Acinetobacter baumannii harboring the blaOXA-72 resistance gene. Rifampin 21-31 carbapenem-hydrolyzing oxacillinase Acinetobacter baumannii 137-146 24638036-0 2014 Rifampicin protects PC12 cells from rotenone-induced cytotoxicity by activating GRP78 via PERK-eIF2alpha-ATF4 pathway. Rifampin 0-10 eukaryotic translation initiation factor 2A Rattus norvegicus 95-104 24638036-0 2014 Rifampicin protects PC12 cells from rotenone-induced cytotoxicity by activating GRP78 via PERK-eIF2alpha-ATF4 pathway. Rifampin 0-10 activating transcription factor 4 Rattus norvegicus 105-109 24638036-4 2014 In this study, using the comprehensive proteomic analysis, we identified that the 78 kDa glucose-regulated protein (GRP78), a hallmark of the unfolded protein response (UPR), was upregulated in rifampicin-treated PC12 cells. Rifampin 194-204 heat shock protein family A (Hsp70) member 5 Rattus norvegicus 82-114 24638036-4 2014 In this study, using the comprehensive proteomic analysis, we identified that the 78 kDa glucose-regulated protein (GRP78), a hallmark of the unfolded protein response (UPR), was upregulated in rifampicin-treated PC12 cells. Rifampin 194-204 heat shock protein family A (Hsp70) member 5 Rattus norvegicus 116-121 24638036-6 2014 GRP78 functions cytoprotectively in stressed cells, therefore, we hypothesized that GRP78 mediated rifampicin-induced neuroprotection. Rifampin 99-109 heat shock protein family A (Hsp70) member 5 Rattus norvegicus 0-5 24638036-6 2014 GRP78 functions cytoprotectively in stressed cells, therefore, we hypothesized that GRP78 mediated rifampicin-induced neuroprotection. Rifampin 99-109 heat shock protein family A (Hsp70) member 5 Rattus norvegicus 84-89 24638036-7 2014 Using RNA interference, we found that GRP78 gene knockdown significantly attenuated the neuroprotective effects of rifampicin. Rifampin 115-125 heat shock protein family A (Hsp70) member 5 Rattus norvegicus 38-43 24638036-9 2014 Our results showed that PERK, eukaryotic initiation factor 2alpha (eIF2alpha), and activating transcription factor 4 (ATF4) were activated in rifampicin-treated PC12 cells. Rifampin 142-152 eukaryotic translation initiation factor 2A Rattus norvegicus 67-76 24638036-9 2014 Our results showed that PERK, eukaryotic initiation factor 2alpha (eIF2alpha), and activating transcription factor 4 (ATF4) were activated in rifampicin-treated PC12 cells. Rifampin 142-152 activating transcription factor 4 Rattus norvegicus 83-116 24638036-9 2014 Our results showed that PERK, eukaryotic initiation factor 2alpha (eIF2alpha), and activating transcription factor 4 (ATF4) were activated in rifampicin-treated PC12 cells. Rifampin 142-152 activating transcription factor 4 Rattus norvegicus 118-122 24638036-12 2014 Taken together, our data suggested that rifampicin induced GRP78 via the PERK-eIF2alpha-ATF4 pathway to protect neurons against rotenone-induced cell damage. Rifampin 40-50 heat shock protein family A (Hsp70) member 5 Rattus norvegicus 59-64 24638036-12 2014 Taken together, our data suggested that rifampicin induced GRP78 via the PERK-eIF2alpha-ATF4 pathway to protect neurons against rotenone-induced cell damage. Rifampin 40-50 activating transcription factor 4 Rattus norvegicus 73-92 24474302-0 2014 The effect of rifampicin, a prototypical CYP3A4 inducer, on erlotinib pharmacokinetics in healthy subjects. Rifampin 14-24 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-47 24411494-2 2014 High expression of ABCB1 induced by forskolin (FSK) and rifampicin (RIF) in the bovine blastocysts reportedly improves the cellular quality. Rifampin 56-66 LOW QUALITY PROTEIN: multidrug resistance protein 1 Bos taurus 19-24 24411494-2 2014 High expression of ABCB1 induced by forskolin (FSK) and rifampicin (RIF) in the bovine blastocysts reportedly improves the cellular quality. Rifampin 68-71 LOW QUALITY PROTEIN: multidrug resistance protein 1 Bos taurus 19-24 24411494-3 2014 In the present study, interferon-alpha, similar to FSK and RIF, was highly potent in inducing the expression of ABCB1 in the bovine blastocysts but did not exhibit an additive effect with FSK and RIF. Rifampin 59-62 LOW QUALITY PROTEIN: multidrug resistance protein 1 Bos taurus 112-117 24474302-3 2014 Two phase I studies were conducted in healthy male subjects to evaluate the effect of pre- or co-administered rifampicin, a CYP3A4 inducer, on the pharmacokinetics of erlotinib. Rifampin 110-120 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 124-130 24335393-6 2014 SULT1C2 protein content was strongly increased by 1,25(OH)2D3 treatment and moderately increased by GW3965, GW4064, and rifampicin. Rifampin 120-130 sulfotransferase family 1C member 4 Homo sapiens 0-7 24399452-2 2014 As afatinib is a substrate for the P-glycoprotein (P-gp) pump transporter the three studies presented here investigated the pharmacokinetics of afatinib in the presence of a potent inhibitor (ritonavir) or inducer [rifampicin (rifampin)] of P-gp. Rifampin 215-225 ATP binding cassette subfamily B member 1 Homo sapiens 51-55 24399452-2 2014 As afatinib is a substrate for the P-glycoprotein (P-gp) pump transporter the three studies presented here investigated the pharmacokinetics of afatinib in the presence of a potent inhibitor (ritonavir) or inducer [rifampicin (rifampin)] of P-gp. Rifampin 227-235 ATP binding cassette subfamily B member 1 Homo sapiens 51-55 24259679-5 2014 Here we investigated five synthetic TRPC6-activating phloroglucinol derivatives and four TRPC6-nonactivating compounds compared with hyperforin and rifampicin for their potential to activate PXR in silico and in vitro. Rifampin 148-158 nuclear receptor subfamily 1 group I member 2 Homo sapiens 191-194 24259679-8 2014 Hyperforin and rifampicin treatment of primary human hepatocytes resulted in highly correlated induction of PXR target genes, whereas treatment with the phloroglucinol derivatives elicited moderate gene expression changes that were only weakly correlated with those of rifampicin and hyperforin treatment. Rifampin 15-25 nuclear receptor subfamily 1 group I member 2 Homo sapiens 108-111 24831405-0 2014 Lymphadenitis caused by infection with an isoniazid- and rifampin-resistant strain of Mycobacterium bovis BCG in an infant with IFN-gamma/IL-12 pathway defect. Rifampin 57-65 interferon gamma Homo sapiens 128-137 24259679-7 2014 Hyperforin and rifampicin treatment of HepG2 cells cotransfected with human PXR expression vector and a CYP3A4 promoter-reporter construct resulted in potent PXR-dependent induction, whereas all TRPC6-activating compounds failed to show any PXR activation or to antagonize rifampicin-mediated CYP3A4 promoter induction. Rifampin 15-25 nuclear receptor subfamily 1 group I member 2 Homo sapiens 76-79 24259679-7 2014 Hyperforin and rifampicin treatment of HepG2 cells cotransfected with human PXR expression vector and a CYP3A4 promoter-reporter construct resulted in potent PXR-dependent induction, whereas all TRPC6-activating compounds failed to show any PXR activation or to antagonize rifampicin-mediated CYP3A4 promoter induction. Rifampin 15-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-110 24259679-7 2014 Hyperforin and rifampicin treatment of HepG2 cells cotransfected with human PXR expression vector and a CYP3A4 promoter-reporter construct resulted in potent PXR-dependent induction, whereas all TRPC6-activating compounds failed to show any PXR activation or to antagonize rifampicin-mediated CYP3A4 promoter induction. Rifampin 15-25 nuclear receptor subfamily 1 group I member 2 Homo sapiens 158-161 24259679-7 2014 Hyperforin and rifampicin treatment of HepG2 cells cotransfected with human PXR expression vector and a CYP3A4 promoter-reporter construct resulted in potent PXR-dependent induction, whereas all TRPC6-activating compounds failed to show any PXR activation or to antagonize rifampicin-mediated CYP3A4 promoter induction. Rifampin 15-25 transient receptor potential cation channel subfamily C member 6 Homo sapiens 195-200 24259679-7 2014 Hyperforin and rifampicin treatment of HepG2 cells cotransfected with human PXR expression vector and a CYP3A4 promoter-reporter construct resulted in potent PXR-dependent induction, whereas all TRPC6-activating compounds failed to show any PXR activation or to antagonize rifampicin-mediated CYP3A4 promoter induction. Rifampin 15-25 nuclear receptor subfamily 1 group I member 2 Homo sapiens 158-161 24259679-7 2014 Hyperforin and rifampicin treatment of HepG2 cells cotransfected with human PXR expression vector and a CYP3A4 promoter-reporter construct resulted in potent PXR-dependent induction, whereas all TRPC6-activating compounds failed to show any PXR activation or to antagonize rifampicin-mediated CYP3A4 promoter induction. Rifampin 15-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 293-299 24259679-7 2014 Hyperforin and rifampicin treatment of HepG2 cells cotransfected with human PXR expression vector and a CYP3A4 promoter-reporter construct resulted in potent PXR-dependent induction, whereas all TRPC6-activating compounds failed to show any PXR activation or to antagonize rifampicin-mediated CYP3A4 promoter induction. Rifampin 273-283 nuclear receptor subfamily 1 group I member 2 Homo sapiens 76-79 24259679-7 2014 Hyperforin and rifampicin treatment of HepG2 cells cotransfected with human PXR expression vector and a CYP3A4 promoter-reporter construct resulted in potent PXR-dependent induction, whereas all TRPC6-activating compounds failed to show any PXR activation or to antagonize rifampicin-mediated CYP3A4 promoter induction. Rifampin 273-283 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-110 24586675-1 2014 BACKGROUND: Xpert MTB/RIF is an automated cartridge-based nucleic acid amplification test that has demonstrated its potential to detect tuberculosis and rifampicin resistance with high accuracy. Rifampin 153-163 ras homolog family member F, filopodia associated Homo sapiens 22-25 24420746-10 2014 Solute carrier organic anion transporter family member 1B1 (SLCO1B1) c.388A G and SLCO1B1 c.463C A polymorphisms jointly had significant effect on rifampin Cmax (R(2) = 0.75). Rifampin 147-155 solute carrier organic anion transporter family member 1B1 Homo sapiens 60-67 24420746-10 2014 Solute carrier organic anion transporter family member 1B1 (SLCO1B1) c.388A G and SLCO1B1 c.463C A polymorphisms jointly had significant effect on rifampin Cmax (R(2) = 0.75). Rifampin 147-155 solute carrier organic anion transporter family member 1B1 Homo sapiens 82-89 24710854-13 2014 The expression of Cyp2b10 was increased 35-fold by phenobarbital, and Cyp3a11 was increased 4.5-fold by rifampin. Rifampin 104-112 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 70-77 24710854-15 2014 Both phenobarbital and rifampin increased Ugt1a1, Ugt1a6, Ugt1a9, and icariin but did not show any suppressive effects on the Ugt1 family genes. Rifampin 23-31 UDP glucuronosyltransferase 1 family, polypeptide A1 Mus musculus 42-48 24710854-15 2014 Both phenobarbital and rifampin increased Ugt1a1, Ugt1a6, Ugt1a9, and icariin but did not show any suppressive effects on the Ugt1 family genes. Rifampin 23-31 UDP glucuronosyltransferase 1 family, polypeptide A6A Mus musculus 50-56 24710854-15 2014 Both phenobarbital and rifampin increased Ugt1a1, Ugt1a6, Ugt1a9, and icariin but did not show any suppressive effects on the Ugt1 family genes. Rifampin 23-31 UDP glucuronosyltransferase 1 family, polypeptide A9 Mus musculus 58-64 24710854-15 2014 Both phenobarbital and rifampin increased Ugt1a1, Ugt1a6, Ugt1a9, and icariin but did not show any suppressive effects on the Ugt1 family genes. Rifampin 23-31 UDP glucuronosyltransferase 1 family, polypeptide A2 Mus musculus 42-46 24507091-2 2014 In a mouse model of multiple system atrophy, rifampicin inhibited formation of alpha-synuclein fibrils, the neuropathological hallmark of the disease. Rifampin 45-55 synuclein, alpha Mus musculus 79-94 23990572-0 2014 Dependence of efavirenz- and rifampicin-isoniazid-based antituberculosis treatment drug-drug interaction on CYP2B6 and NAT2 genetic polymorphisms: ANRS 12154 study in Cambodia. Rifampin 29-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 24316028-4 2014 There was a significant difference between CYP1A2 -739T/G and T/T genotypes when patients were treated with EFV-containing therapy combined with rifampicin-based TB treatment, but not when EFV-containing therapy was alone. Rifampin 145-155 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 43-49 24316028-5 2014 CYP2B6 516T/T genotype was associated with high EFV levels compared to other CYP2B6 516G>T genotypes in the presence and in the absence of rifampicin-based TB treatment. Rifampin 142-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 24316028-6 2014 Predictive factors of EFV plasma levels in the presence of rifampicin-based TB treatment were CYP2A6 1093G>A, CYP2B6 516G>T, and CYP2B6 983T>C accounting for 27%, 43%, and 29% of the total variance in EFV levels, respectively. Rifampin 59-69 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 94-100 23990572-0 2014 Dependence of efavirenz- and rifampicin-isoniazid-based antituberculosis treatment drug-drug interaction on CYP2B6 and NAT2 genetic polymorphisms: ANRS 12154 study in Cambodia. Rifampin 29-39 N-acetyltransferase 2 Homo sapiens 119-123 24448973-2 2014 Xpert MTB/RIF assay is an automated test that can detect both TB and rifampicin resistance, generally within two hours after starting the test, with minimal hands-on technical time. Rifampin 70-80 ras homolog family member F, filopodia associated Homo sapiens 11-14 24092658-9 2014 Mutations of rpoB and fusA following the use of rifampicin and fusidic acid were identified in the second step of evolution, which corresponded to the development of dual resistance to rifampicin and fusidic acid and the conversion of hVISA to VISA. Rifampin 48-58 mitochondrial antiviral signaling protein Homo sapiens 235-240 24092658-9 2014 Mutations of rpoB and fusA following the use of rifampicin and fusidic acid were identified in the second step of evolution, which corresponded to the development of dual resistance to rifampicin and fusidic acid and the conversion of hVISA to VISA. Rifampin 48-58 mitochondrial antiviral signaling protein Homo sapiens 236-240 24092658-9 2014 Mutations of rpoB and fusA following the use of rifampicin and fusidic acid were identified in the second step of evolution, which corresponded to the development of dual resistance to rifampicin and fusidic acid and the conversion of hVISA to VISA. Rifampin 185-195 mitochondrial antiviral signaling protein Homo sapiens 235-240 24092658-9 2014 Mutations of rpoB and fusA following the use of rifampicin and fusidic acid were identified in the second step of evolution, which corresponded to the development of dual resistance to rifampicin and fusidic acid and the conversion of hVISA to VISA. Rifampin 185-195 mitochondrial antiviral signaling protein Homo sapiens 236-240 24448973-23 2014 Rifampicin resistance For rifampicin resistance detection, Xpert MTB/RIF pooled sensitivity was 95% (95% CrI 90% to 97%; 17 studies, 555 rifampicin resistance positives) and pooled specificity was 98% (95% CrI 97% to 99%; 24 studies, 2411 rifampicin resistance negatives). Rifampin 0-10 ras homolog family member F, filopodia associated Homo sapiens 70-73 24448973-25 2014 Where 5% of those with symptoms are rifampicin resistant, Xpert MTB/RIF would correctly identify 48 individuals as rifampicin resistant and miss three cases and correctly identify 931 individuals as rifampicin susceptible and misclassify 19 individuals as resistant. Rifampin 36-46 ras homolog family member F, filopodia associated Homo sapiens 69-72 24448973-25 2014 Where 5% of those with symptoms are rifampicin resistant, Xpert MTB/RIF would correctly identify 48 individuals as rifampicin resistant and miss three cases and correctly identify 931 individuals as rifampicin susceptible and misclassify 19 individuals as resistant. Rifampin 116-126 ras homolog family member F, filopodia associated Homo sapiens 69-72 24448973-25 2014 Where 5% of those with symptoms are rifampicin resistant, Xpert MTB/RIF would correctly identify 48 individuals as rifampicin resistant and miss three cases and correctly identify 931 individuals as rifampicin susceptible and misclassify 19 individuals as resistant. Rifampin 116-126 ras homolog family member F, filopodia associated Homo sapiens 69-72 24448973-30 2014 For rifampicin resistance detection, Xpert MTB/RIF provides accurate results and can allow rapid initiation of MDR-TB treatment, pending results from conventional culture and DST. Rifampin 4-14 ras homolog family member F, filopodia associated Homo sapiens 48-51 24191259-5 2014 Differentiated HepaRG cells were pretreated with dexamethasone (100 muM) or the prototypical CYP3A4 inducer rifampicin (4 muM) for 72 hours, followed by incubation with lapatinib (0-100 muM) for 24 hours. Rifampin 108-118 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-99 24204015-7 2014 Rifampicin treatment of two different samples of human primary hepatocytes revealed that PXR induces G6Pase in the presence of high levels of SGK2, whereas PXR represses G6Pase in its absence. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 156-159 24558228-6 2014 The modulations of ABCB1 and ABCC1 expressions were analyzed after ex vivo treatment of human skin with rifampicin and dexamethasone. Rifampin 104-114 ATP binding cassette subfamily B member 1 Homo sapiens 19-24 24674673-8 2014 Multiple factors may affect CYP3A expression and activity, such as inducers like rifampicin, phenobarbital, 3-methylcholantrene, beta-naphtoflavone, and dexamethasone. Rifampin 81-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 28-33 27128230-3 2014 This open-label, nonrandomized study investigated effects of CYP3A4 induction, by rifampin, on sirolimus pharmacokinetics. Rifampin 82-90 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 61-67 24204015-7 2014 Rifampicin treatment of two different samples of human primary hepatocytes revealed that PXR induces G6Pase in the presence of high levels of SGK2, whereas PXR represses G6Pase in its absence. Rifampin 0-10 glucose-6-phosphatase catalytic subunit 1 Homo sapiens 170-176 24204015-3 2014 Both SGK2 and G6Pase mRNAs were increased in rifampicin-treated HepG2 cells stably expressing human PXR. Rifampin 45-55 serum/glucocorticoid regulated kinase 2 Homo sapiens 5-9 25335408-1 2014 The goal of this work was to study cytochrome-450 (CYP) 2C9 (CYP2C9) gene polymorphism in patients with tuberculosis (TB) and its meaning for development, progress, and outcome of TB, for the pharmacokinetics of the antituberculosis antibiotic rifampicin on the basis of the southern region of Ukraine. Rifampin 244-254 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-59 24204015-3 2014 Both SGK2 and G6Pase mRNAs were increased in rifampicin-treated HepG2 cells stably expressing human PXR. Rifampin 45-55 glucose-6-phosphatase catalytic subunit 1 Homo sapiens 14-20 24204015-3 2014 Both SGK2 and G6Pase mRNAs were increased in rifampicin-treated HepG2 cells stably expressing human PXR. Rifampin 45-55 nuclear receptor subfamily 1 group I member 2 Homo sapiens 100-103 24204015-7 2014 Rifampicin treatment of two different samples of human primary hepatocytes revealed that PXR induces G6Pase in the presence of high levels of SGK2, whereas PXR represses G6Pase in its absence. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 89-92 24204015-7 2014 Rifampicin treatment of two different samples of human primary hepatocytes revealed that PXR induces G6Pase in the presence of high levels of SGK2, whereas PXR represses G6Pase in its absence. Rifampin 0-10 glucose-6-phosphatase catalytic subunit 1 Homo sapiens 101-107 24204015-7 2014 Rifampicin treatment of two different samples of human primary hepatocytes revealed that PXR induces G6Pase in the presence of high levels of SGK2, whereas PXR represses G6Pase in its absence. Rifampin 0-10 serum/glucocorticoid regulated kinase 2 Homo sapiens 142-146 25341566-4 2014 Here, we showed that resveratrol (RES), a natural polyphenolic compound could significantly suppress the rifampicin-induced PXR transactivation of the CYP3A4 promoter. Rifampin 105-115 nuclear receptor subfamily 1, group I, member 2 Mus musculus 124-127 25335408-1 2014 The goal of this work was to study cytochrome-450 (CYP) 2C9 (CYP2C9) gene polymorphism in patients with tuberculosis (TB) and its meaning for development, progress, and outcome of TB, for the pharmacokinetics of the antituberculosis antibiotic rifampicin on the basis of the southern region of Ukraine. Rifampin 244-254 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 24362577-4 2013 The CYP1A1 and 1A2, and CYP3A4 mRNA expression levels increased significantly in the PXB-mouse livers with 20 mg/kg of 3-MC (Cmax, 12.2 ng/mL), and 10 mg/kg rifampicin (Cmax, 6.9 microg/mL), respectively. Rifampin 157-167 cytochrome P450, family 1, subfamily a, polypeptide 1 Mus musculus 4-18 24362577-6 2013 The CYP1A2 and CYP3A4 mRNA expression levels increased significantly with 50 ng/mL of 3-MC, and 5 mug/mL of rifampicin, respectively, which indicated that the sensitivities were similar between in vivo and in vitro studies. Rifampin 108-118 cytochrome P450, family 1, subfamily a, polypeptide 2 Mus musculus 4-10 24085261-6 2013 RESULTS: Inhibition of CYP3A4 by ketoconazole increased midostaurin exposure more than tenfold, and induction of CYP3A4 by rifampicin decreased midostaurin exposure by more than tenfold. Rifampin 123-133 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 113-119 24060875-0 2013 Induction of influx and efflux transporters and cytochrome P450 3A4 in primary human hepatocytes by rifampin, rifabutin, and rifapentine. Rifampin 100-108 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-67 24060875-1 2013 Rifampin is a potent inducer of cytochrome P450 (CYP) enzymes and transporters. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 24060875-1 2013 Rifampin is a potent inducer of cytochrome P450 (CYP) enzymes and transporters. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 24060875-4 2013 Rifampin and rifabutin significantly induced CYP3A4 (80-fold and 20-fold, respectively) in primary human hepatocytes. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 45-51 24085261-8 2013 CONCLUSION: The pharmacokinetics of midostaurin and its metabolites was affected substantially by ketoconazole and rifampicin, suggesting that midostaurin is a sensitive CYP3A4 substrate. Rifampin 115-125 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 170-176 23852652-8 2013 Strong CYP3A or CYP2C9 inducers (e.g. rifampicin) would be expected to decrease the exposure to ospemifene. Rifampin 38-48 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 7-12 23770199-5 2013 In primary human hepatocytes, clopidogrel (10 and 100 muM) was cytotoxic only after cytochrome P450 (CYP) induction by rifampicin. Rifampin 119-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 23770199-5 2013 In primary human hepatocytes, clopidogrel (10 and 100 muM) was cytotoxic only after cytochrome P450 (CYP) induction by rifampicin. Rifampin 119-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 23916555-0 2013 PXR-ALAS1: a key regulatory pathway in liver toxicity induced by isoniazid-rifampicin antituberculosis treatment. Rifampin 75-85 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-3 23916555-0 2013 PXR-ALAS1: a key regulatory pathway in liver toxicity induced by isoniazid-rifampicin antituberculosis treatment. Rifampin 75-85 5'-aminolevulinate synthase 1 Homo sapiens 4-9 23974699-0 2013 Chronological effects of rifampicin discontinuation on cytochrome P450 activity in healthy Japanese volunteers, using the cocktail method. Rifampin 25-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 23974699-5 2013 Seven-day rifampicin administration increased CYP2C19 and CYP3A enzyme activities. Rifampin 10-20 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 46-53 23974699-5 2013 Seven-day rifampicin administration increased CYP2C19 and CYP3A enzyme activities. Rifampin 10-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 58-63 23974699-6 2013 The induced CYP2C19 and CYP3A activities remained elevated at 4 days after rifampicin discontinuation and returned to baseline levels 8 days after rifampicin discontinuation. Rifampin 75-85 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 12-19 23974699-6 2013 The induced CYP2C19 and CYP3A activities remained elevated at 4 days after rifampicin discontinuation and returned to baseline levels 8 days after rifampicin discontinuation. Rifampin 75-85 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-29 23974699-6 2013 The induced CYP2C19 and CYP3A activities remained elevated at 4 days after rifampicin discontinuation and returned to baseline levels 8 days after rifampicin discontinuation. Rifampin 147-157 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 12-19 23974699-6 2013 The induced CYP2C19 and CYP3A activities remained elevated at 4 days after rifampicin discontinuation and returned to baseline levels 8 days after rifampicin discontinuation. Rifampin 147-157 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-29 23974699-7 2013 CYP1A2 and CYP2D6 enzyme activities showed no significant changes, and CYP2C9 enzyme activity was increased with rifampicin administration, with a tendency toward statistical significance. Rifampin 113-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 23852652-8 2013 Strong CYP3A or CYP2C9 inducers (e.g. rifampicin) would be expected to decrease the exposure to ospemifene. Rifampin 38-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 23852717-8 2013 LRP1- and P-gp-mediated 125I-Abeta40 biliary excretion was inducible and increased BEI% by 26% after rifampicin pretreatment. Rifampin 101-111 LDL receptor related protein 1 Rattus norvegicus 0-4 23852717-8 2013 LRP1- and P-gp-mediated 125I-Abeta40 biliary excretion was inducible and increased BEI% by 26% after rifampicin pretreatment. Rifampin 101-111 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 10-14 23258774-1 2013 Xpert MTB/RIF is a rapid test to diagnose tuberculosis (TB) and rifampicin-resistant TB. Rifampin 64-74 ras homolog family member F, filopodia associated Homo sapiens 10-13 23935064-4 2013 Rifampin induced the expression of 10 clinically important and 13 additional P450 genes and repressed the expression of 9 other P450 genes (P < 0.05). Rifampin 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-81 23935064-4 2013 Rifampin induced the expression of 10 clinically important and 13 additional P450 genes and repressed the expression of 9 other P450 genes (P < 0.05). Rifampin 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-132 23935064-6 2013 Several of these changes were highly negatively correlated with the rifampin-induced changes in the expression of their predicted target P450 mRNAs, supporting the possibility of miRNA-induced regulation of P450 mRNA expression. Rifampin 68-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-141 23935064-6 2013 Several of these changes were highly negatively correlated with the rifampin-induced changes in the expression of their predicted target P450 mRNAs, supporting the possibility of miRNA-induced regulation of P450 mRNA expression. Rifampin 68-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 207-211 23935064-9 2013 In conclusion, rifampin treatment alters miRNA expression patterns in human hepatocytes, and some of the changes were correlated with the rifampin-induced changes in expression of the P450 mRNAs they are predicted to target. Rifampin 15-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 184-188 23935064-9 2013 In conclusion, rifampin treatment alters miRNA expression patterns in human hepatocytes, and some of the changes were correlated with the rifampin-induced changes in expression of the P450 mRNAs they are predicted to target. Rifampin 138-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 184-188 23928187-5 2013 Here we report the virtual screening of ~25,000 natural product derivatives from the ZINC database using the Molecular Operating Environment docking software tool against the PXR-rifampicin complex X-ray crystal structure. Rifampin 179-189 nuclear receptor subfamily 1 group I member 2 Homo sapiens 175-178 23073537-7 2013 Basal expression of CYP3A4 protein in HepG2-PGC-1alpha cells was increased but rifampicin-inducible expression of CYP3A4 protein was lowered in comparison with parent HepG2 cells. Rifampin 79-89 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 114-120 23929936-3 2013 Cyclosporine A (CsA) and rifampicin (RIF), typical OATP inhibitors, inhibited active uptake of pitavastatin into monkey hepatocytes with half-maximal inhibitory concentration values comparable with those in human hepatocytes. Rifampin 25-35 solute carrier organic anion transporter family member 1A2 Homo sapiens 51-55 23929936-3 2013 Cyclosporine A (CsA) and rifampicin (RIF), typical OATP inhibitors, inhibited active uptake of pitavastatin into monkey hepatocytes with half-maximal inhibitory concentration values comparable with those in human hepatocytes. Rifampin 37-40 solute carrier organic anion transporter family member 1A2 Homo sapiens 51-55 24086247-0 2013 Modeling of rifampicin-induced CYP3A4 activation dynamics for the prediction of clinical drug-drug interactions from in vitro data. Rifampin 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 24086247-3 2013 Rifampicin is a well-known CYP3A4 inducer, and is commonly used as a positive control for evaluating the CYP3A4 induction potential of test compounds. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 27-33 24086247-3 2013 Rifampicin is a well-known CYP3A4 inducer, and is commonly used as a positive control for evaluating the CYP3A4 induction potential of test compounds. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-111 24086247-4 2013 Herein, we report the compilation of in vitro induction data for CYP3A4 by rifampicin in human hepatocytes, and the transcription/translation model developed for this enzyme using an extended least squares method that can account for inherent inter-individual variability. Rifampin 75-85 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-71 23764483-0 2013 Hepatocyte growth factor protects against isoniazid/rifampicin-induced oxidative liver damage. Rifampin 52-62 hepatocyte growth factor Mus musculus 0-24 23712732-7 2013 In human cryopreserved hepatocytes, JPH203 uptake was saturable and inhibited by rifampicin, a prototypical OATP inhibitor. Rifampin 81-91 solute carrier organic anion transporter family member 1A2 Homo sapiens 108-112 23581556-9 2013 CYP3A12 and CYP2B11 induction after phenobarbital or rifampin treatment was robust in both cell types but stronger in S cells. Rifampin 53-61 cytochrome P450 3A12 Canis lupus familiaris 0-7 23581556-9 2013 CYP3A12 and CYP2B11 induction after phenobarbital or rifampin treatment was robust in both cell types but stronger in S cells. Rifampin 53-61 cytochrome P450 2B11 Canis lupus familiaris 12-19 23677771-14 2013 In the presence of ketoconazole, a strong CYP3A4/5 inhibitor, axitinib C max and AUC increased by 1.5- and 2-fold, respectively, whereas co-administration of rifampin, a strong CYP3A4/5 inducer, resulted in a 71 and 79 % decrease in the C max and AUC , respectively. Rifampin 159-167 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 178-184 23686764-6 2013 The predicted decrease in AUC and Cmax values of digoxin following administration of rifampicin because of P-gp induction was 1.57- (range: 1.42-1.77) and 1.62-fold (range: 1.53-1.70), which were reasonably consistent with observed values of 1.4- and 2.2-fold, respectively. Rifampin 85-95 ATP binding cassette subfamily B member 1 Homo sapiens 107-111 23703021-1 2013 A prerequisite for the prediction of the magnitude of P-glycoprotein (P-gp)-mediated drug-drug interactions between digoxin and P-gp inhibitors (e.g. verapamil and its metabolite norverapamil) or P-gp inducers (e.g. rifampicin) is a predictive pharmacokinetic model for digoxin itself. Rifampin 216-226 ATP binding cassette subfamily B member 1 Homo sapiens 54-68 23703021-1 2013 A prerequisite for the prediction of the magnitude of P-glycoprotein (P-gp)-mediated drug-drug interactions between digoxin and P-gp inhibitors (e.g. verapamil and its metabolite norverapamil) or P-gp inducers (e.g. rifampicin) is a predictive pharmacokinetic model for digoxin itself. Rifampin 216-226 ATP binding cassette subfamily B member 1 Homo sapiens 70-74 23991219-0 2013 Development of novel rifampicin-derived P-glycoprotein activators/inducers. Rifampin 21-31 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 40-54 23764483-4 2013 The present work was focused to address the antioxidant and antiapoptotic effects of HGF on isoniazid- and rifampicin-induced hepatotoxicity. Rifampin 107-117 hepatocyte growth factor Mus musculus 85-88 23764483-8 2013 Furthermore, inhibition of endogenous HGF with anti-HGF antibody (iv) enhanced the isoniazid- and rifampicin-induced oxidative stress damage and decreased the GSH content, aggravating liver damage. Rifampin 98-108 hepatocyte growth factor Mus musculus 38-41 23764483-8 2013 Furthermore, inhibition of endogenous HGF with anti-HGF antibody (iv) enhanced the isoniazid- and rifampicin-induced oxidative stress damage and decreased the GSH content, aggravating liver damage. Rifampin 98-108 hepatocyte growth factor Mus musculus 52-55 24101390-8 2013 In the present study, findings of the P-gp expression were inconclusive but regarding previous studies, it can be suggested that the beneficial effects of rifampicin and spironolactone may be partly due to their action as a P-gp ligand. Rifampin 155-165 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 224-228 23674608-0 2013 Comparison of endogenous 4beta-hydroxycholesterol with midazolam as markers for CYP3A4 induction by rifampicin. Rifampin 100-110 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 80-86 23674608-5 2013 The CYP3A4 induction could be detected even at the lowest dose of rifampicin (10 mg) via the estimated midazolam clearance, the 4beta-hydroxycholesterol ratio (both P < 0.01), and the 6beta-hydroxycortisol ratio (P < 0.05). Rifampin 66-76 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 4-10 23851904-0 2013 GC-MS-based quantitative signatures of cytochrome P450-mediated steroid oxidation induced by rifampicin. Rifampin 93-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 23851904-4 2013 Twelve healthy subjects were orally administered 600 mg of rifampicin a day for 7 days, and their CYP enzyme activity was evaluated. Rifampin 59-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 23851904-7 2013 Administration of the CYP3A4 inducer rifampicin produced distinct differences in CYP3A4 and CYP11B1, CYP19A1, HSD11B, and HSD17B, which were indicated by their heat map-visualized steroid signatures. Rifampin 37-47 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 22-28 23851904-7 2013 Administration of the CYP3A4 inducer rifampicin produced distinct differences in CYP3A4 and CYP11B1, CYP19A1, HSD11B, and HSD17B, which were indicated by their heat map-visualized steroid signatures. Rifampin 37-47 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-87 23851904-7 2013 Administration of the CYP3A4 inducer rifampicin produced distinct differences in CYP3A4 and CYP11B1, CYP19A1, HSD11B, and HSD17B, which were indicated by their heat map-visualized steroid signatures. Rifampin 37-47 cytochrome P450 family 11 subfamily B member 1 Homo sapiens 92-99 23851904-7 2013 Administration of the CYP3A4 inducer rifampicin produced distinct differences in CYP3A4 and CYP11B1, CYP19A1, HSD11B, and HSD17B, which were indicated by their heat map-visualized steroid signatures. Rifampin 37-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-108 23851904-7 2013 Administration of the CYP3A4 inducer rifampicin produced distinct differences in CYP3A4 and CYP11B1, CYP19A1, HSD11B, and HSD17B, which were indicated by their heat map-visualized steroid signatures. Rifampin 37-47 hydroxysteroid 11-beta dehydrogenase 1 Homo sapiens 110-116 24180002-0 2013 Effects of rifampin-based antituberculosis therapy on plasma efavirenz concentrations in children vary by CYP2B6 genotype. Rifampin 11-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 24180002-2 2013 However, rifampin, a key component of antituberculosis therapy, induces CYP2B6. Rifampin 9-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 23670789-5 2013 The GMRs and 90% CIs for anacetrapib AUC0- and Cmax were 1.25 (1.04, 1.51) and 1.43 (1.13, 1.82) for SD rifampin (Period 2/Period 1) and 0.35 (0.29, 0.42) and 0.26 (0.21, 0.32) for MD rifampin (Period 3/Period 1), respectively. Rifampin 105-113 period circadian regulator 2 Homo sapiens 115-123 23685082-0 2013 Mechanisms of P-gp inhibition and effects on membrane fluidity of a new rifampicin derivative, 1,8-dibenzoyl-rifampicin. Rifampin 72-82 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 14-18 23685082-1 2013 PURPOSE: To assess P-glycoprotein (P-gp)-modulation ability and the mechanisms of P-gp inhibition mediated by a new synthetic rifampicin derivative, 1,8-dibenzoyl-rifampicin (DiBenzRif), in an in vitro model of the blood-brain barrier (BBB), RBE4 cells, and in membrane mimetic models (liposomes). Rifampin 126-136 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 82-86 23624173-3 2013 We previously identified zebrafish CYP3A65 as a CYP3A ortholog that is constitutively expressed in gastrointestinal tissues, and is upregulated by treatment with dexamethasone, rifampicin or tetrachlorodibenzo-p-dioxin (TCDD). Rifampin 177-187 cytochrome P450, family 3, subfamily A, polypeptide 65 Danio rerio 35-42 23591749-1 2013 Isoniazid (INH) and rifampicin (RIF) are the first-line drugs for antituberculosis (anti-TB) chemotherapy. Rifampin 20-30 alpha fetoprotein regulation 2, inducibility Mus musculus 32-35 23180585-3 2013 Xpert MTB/RIF, a new assay employing automated nucleic acid amplification to detect Mycobacterium tuberculosis, as well as mutations that confer rifampicin resistance, holds the promise to largely overcome these operational challenges. Rifampin 145-155 ras homolog family member F, filopodia associated Homo sapiens 10-13 23377313-8 2013 In the subgroup analyses, the pooled results showed that GSTM1 null allele carriers had a significant association with ADIH risk in East Asians and the patients who used isoniazid (INH) + rifampicin (RMP) + pyrazinamide (PZA) + ethambutol (EMB), or + streptomycin (SM) (HRZES), but the opposite result was observed for patients using HR. Rifampin 188-198 glutathione S-transferase mu 1 Homo sapiens 57-62 23377313-8 2013 In the subgroup analyses, the pooled results showed that GSTM1 null allele carriers had a significant association with ADIH risk in East Asians and the patients who used isoniazid (INH) + rifampicin (RMP) + pyrazinamide (PZA) + ethambutol (EMB), or + streptomycin (SM) (HRZES), but the opposite result was observed for patients using HR. Rifampin 200-203 glutathione S-transferase mu 1 Homo sapiens 57-62 23874477-3 2013 In this study, we found that rifampicin, an agonist of human PXR, induced lipid accumulation in HepG2 cells. Rifampin 29-39 nuclear receptor subfamily 1 group I member 2 Homo sapiens 61-64 23874477-6 2013 Treatment of HepG2 cells with rifampicin induced the expression of the free fatty acid transporter CD36 and ABCG1, as well as several lipogenic enzymes, including stearoyl-CoA desaturase-1 (SCD1), long chain free fatty acid elongase (FAE), and lecithin-cholesterol acyltransferase (LCAT), while the expression of acyl:cholesterol acetyltransferase(ACAT1) was not affected. Rifampin 30-40 stearoyl-CoA desaturase Homo sapiens 190-194 23874477-7 2013 Moreover, in PXR over-expressing HepG2 cells (HepG2-PXR), the SCD1 expression was significantly higher than in HepG2-Vector cells, even in the absence of rifampicin. Rifampin 154-164 nuclear receptor subfamily 1 group I member 2 Homo sapiens 13-16 23874477-8 2013 Down-regulation of PXR by shRNA abolished the rifampicin-induced SCD1 gene expression in HepG2 cells. Rifampin 46-56 nuclear receptor subfamily 1 group I member 2 Homo sapiens 19-22 23874477-8 2013 Down-regulation of PXR by shRNA abolished the rifampicin-induced SCD1 gene expression in HepG2 cells. Rifampin 46-56 stearoyl-CoA desaturase Homo sapiens 65-69 23701163-2 2013 Cytochrome P450 2E1 (CYP2E1) is thought to contribute to the synergistic effects of RIF and INH. Rifampin 84-87 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 0-19 23701163-2 2013 Cytochrome P450 2E1 (CYP2E1) is thought to contribute to the synergistic effects of RIF and INH. Rifampin 84-87 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 21-27 23701163-9 2013 RESULTS: Mannitol inhibited CYP2E1 activity by 54% in mice with INH/RIF-induced hepatotoxicity (p < 0.005). Rifampin 68-71 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 28-34 23670789-5 2013 The GMRs and 90% CIs for anacetrapib AUC0- and Cmax were 1.25 (1.04, 1.51) and 1.43 (1.13, 1.82) for SD rifampin (Period 2/Period 1) and 0.35 (0.29, 0.42) and 0.26 (0.21, 0.32) for MD rifampin (Period 3/Period 1), respectively. Rifampin 185-193 period circadian regulator 2 Homo sapiens 115-123 23558944-8 2013 In in situ liver perfusion model, the hepatic extraction ratio of MTC-220 was reduced to 40 % of control in the presence of rifampicin, an Oatps inhibitor, and the cumulative biliary excretion rates of MTC-220 were reduced to 52.9, 71.5 and 62.9 % of control when concomitant perfusion with probenecid, novobiocin and verapamil, the inhibitors of Mrp2, Bcrp and P-gp, respectively. Rifampin 124-134 ATP binding cassette subfamily C member 2 Rattus norvegicus 347-351 23558944-8 2013 In in situ liver perfusion model, the hepatic extraction ratio of MTC-220 was reduced to 40 % of control in the presence of rifampicin, an Oatps inhibitor, and the cumulative biliary excretion rates of MTC-220 were reduced to 52.9, 71.5 and 62.9 % of control when concomitant perfusion with probenecid, novobiocin and verapamil, the inhibitors of Mrp2, Bcrp and P-gp, respectively. Rifampin 124-134 phosphoglycolate phosphatase Rattus norvegicus 362-366 23471941-0 2013 Relationship between CES2 genetic variations and rifampicin metabolism. Rifampin 49-59 carboxylesterase 2 Homo sapiens 21-25 24089608-1 2013 Xpert MTB/RIF is an automated real-time polymerase chain reaction test for simultaneous detection of tuberculosis and rifampicin resistance. Rifampin 118-128 ras homolog family member F, filopodia associated Homo sapiens 10-13 24252529-5 2013 Coadministration of RPV with drugs that increase gastric pH, such as omeprazole, or those inducing CYP3A4, such as rifampicin, can significantly reduce RPV concentrations and is contraindicated. Rifampin 115-125 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-105 23471941-11 2013 CONCLUSIONS: Variations in CES2, especially c.-2263A > G in the promoter region, may alter rifampicin metabolism by affecting expression of the gene. Rifampin 94-104 carboxylesterase 2 Homo sapiens 27-31 23479137-10 2013 Surprisingly, only in two cell lines, PXR activity was increased by the well-known inductor rifampicin. Rifampin 92-102 nuclear receptor subfamily 1 group I member 2 Homo sapiens 38-41 23393219-0 2013 Quantitative prediction of repaglinide-rifampicin complex drug interactions using dynamic and static mechanistic models: delineating differential CYP3A4 induction and OATP1B1 inhibition potential of rifampicin. Rifampin 199-209 solute carrier organic anion transporter family member 1B1 Homo sapiens 167-174 23396419-7 2013 (-)-Epigallocatechin gallate, cyclosporin A, rifampicin, and telmisartan inhibited transport of dioscin in OATP1B3-HEK293 cells. Rifampin 45-55 solute carrier organic anion transporter family member 1B3 Homo sapiens 107-114 23562850-2 2013 We have previously shown that silencing mediator for retinoid and thyroid receptors (SMRT) interacts with SXR even in the presence of rifampicin on cytochrome P450 monooxygenase 3A4 (CYP3A4) promoter in HepG2 cells. Rifampin 134-144 nuclear receptor corepressor 2 Homo sapiens 30-83 23562850-2 2013 We have previously shown that silencing mediator for retinoid and thyroid receptors (SMRT) interacts with SXR even in the presence of rifampicin on cytochrome P450 monooxygenase 3A4 (CYP3A4) promoter in HepG2 cells. Rifampin 134-144 nuclear receptor corepressor 2 Homo sapiens 85-89 23562850-2 2013 We have previously shown that silencing mediator for retinoid and thyroid receptors (SMRT) interacts with SXR even in the presence of rifampicin on cytochrome P450 monooxygenase 3A4 (CYP3A4) promoter in HepG2 cells. Rifampin 134-144 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 148-181 23562850-7 2013 KEY FINDINGS: SMRT, but not NCoR suppressed rifampicin-induced SXR-mediated transcription. Rifampin 44-54 nuclear receptor corepressor 2 Homo sapiens 14-18 23562850-7 2013 KEY FINDINGS: SMRT, but not NCoR suppressed rifampicin-induced SXR-mediated transcription. Rifampin 44-54 nuclear receptor subfamily 1 group I member 2 Homo sapiens 63-66 23562850-8 2013 The SXR-mediated MDR1 mRNA expression was augmented in the presence of rifampicin, whereas it suppressed the expression following the overexpression of SMRT. Rifampin 71-81 nuclear receptor subfamily 1 group I member 2 Homo sapiens 4-7 23562850-8 2013 The SXR-mediated MDR1 mRNA expression was augmented in the presence of rifampicin, whereas it suppressed the expression following the overexpression of SMRT. Rifampin 71-81 ATP binding cassette subfamily B member 1 Homo sapiens 17-21 23562850-9 2013 In mammalian one-hybrid assay, only SMRT but not NCoR interacted with SXR on MDR1 promoter in the presence of rifampicin. Rifampin 110-120 nuclear receptor corepressor 2 Homo sapiens 36-40 23562850-9 2013 In mammalian one-hybrid assay, only SMRT but not NCoR interacted with SXR on MDR1 promoter in the presence of rifampicin. Rifampin 110-120 nuclear receptor subfamily 1 group I member 2 Homo sapiens 70-73 23562850-9 2013 In mammalian one-hybrid assay, only SMRT but not NCoR interacted with SXR on MDR1 promoter in the presence of rifampicin. Rifampin 110-120 ATP binding cassette subfamily B member 1 Homo sapiens 77-81 23475203-0 2013 Human PXR modulates hepatotoxicity associated with rifampicin and isoniazid co-therapy. Rifampin 51-61 nuclear receptor subfamily 1 group I member 2 Homo sapiens 6-9 23212742-4 2013 With human hepatocytes, treatment with phenobarbital, hyperforin, carbamazepine, and rifampin significantly increased the levels of CYP3A4, but not CYP24A1 or CYP27B1 mRNA. Rifampin 85-93 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 132-138 23212742-4 2013 With human hepatocytes, treatment with phenobarbital, hyperforin, carbamazepine, and rifampin significantly increased the levels of CYP3A4, but not CYP24A1 or CYP27B1 mRNA. Rifampin 85-93 cytochrome P450 family 24 subfamily A member 1 Homo sapiens 148-155 23212742-4 2013 With human hepatocytes, treatment with phenobarbital, hyperforin, carbamazepine, and rifampin significantly increased the levels of CYP3A4, but not CYP24A1 or CYP27B1 mRNA. Rifampin 85-93 cytochrome P450 family 27 subfamily B member 1 Homo sapiens 159-166 23395915-5 2013 To further confirm Pgp induction in rifampin treated mice, digoxin, a known substrate of Pgp, was administered to the rifampin pre-treated mice, and a significant drop in the digoxin exposure was observed compared to the control group. Rifampin 36-44 phosphoglycolate phosphatase Mus musculus 19-22 23395915-5 2013 To further confirm Pgp induction in rifampin treated mice, digoxin, a known substrate of Pgp, was administered to the rifampin pre-treated mice, and a significant drop in the digoxin exposure was observed compared to the control group. Rifampin 118-126 phosphoglycolate phosphatase Mus musculus 89-92 23395915-6 2013 Collectively, our results show that repeated administration of rifampin in mice leads to a reduction in oral exposure due to induction of Pgp-mediated efflux of rifampin, and not via induction of CYP3A isoforms. Rifampin 63-71 phosphoglycolate phosphatase Mus musculus 138-141 23395915-6 2013 Collectively, our results show that repeated administration of rifampin in mice leads to a reduction in oral exposure due to induction of Pgp-mediated efflux of rifampin, and not via induction of CYP3A isoforms. Rifampin 161-169 phosphoglycolate phosphatase Mus musculus 138-141 23238784-7 2013 The CYP3A4 activity could be induced 2-fold by rifampicin, whereas CYP2C9 activity remained equally high. Rifampin 47-57 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 4-10 23493125-2 2013 The data show that electroporation of HaCaT cells in combination with rifampicin induces cytoskeleton disruption and increases permeability of cell monolayer due to cell-cell junctions" interruption, visualized by fluorescent imaging of E-cadherin and actin integrity. Rifampin 70-80 cadherin 1 Homo sapiens 237-247 23143285-5 2013 Furthermore, correlation analysis revealed that the over-expression of both Rv1217c and Rv1218c resulted in the higher minimum inhibition concentrations (MICs) of rifampicin (RIF) (OR = 1.01, P < 0.05 of Rv1217c; OR = 1.23, P < 0.05 of Rv1218c), while the over-expression of Rv1218c only led to the higher MICs of isoniazid (INH) (OR = 1.17, P < 0.05). Rifampin 163-173 tetronasin ABC transporter integral membrane protein Mycobacterium tuberculosis H37Rv 76-83 23143285-5 2013 Furthermore, correlation analysis revealed that the over-expression of both Rv1217c and Rv1218c resulted in the higher minimum inhibition concentrations (MICs) of rifampicin (RIF) (OR = 1.01, P < 0.05 of Rv1217c; OR = 1.23, P < 0.05 of Rv1218c), while the over-expression of Rv1218c only led to the higher MICs of isoniazid (INH) (OR = 1.17, P < 0.05). Rifampin 163-173 tetronasin ABC transporter ATP-binding protein Mycobacterium tuberculosis H37Rv 88-95 23143285-5 2013 Furthermore, correlation analysis revealed that the over-expression of both Rv1217c and Rv1218c resulted in the higher minimum inhibition concentrations (MICs) of rifampicin (RIF) (OR = 1.01, P < 0.05 of Rv1217c; OR = 1.23, P < 0.05 of Rv1218c), while the over-expression of Rv1218c only led to the higher MICs of isoniazid (INH) (OR = 1.17, P < 0.05). Rifampin 163-173 tetronasin ABC transporter integral membrane protein Mycobacterium tuberculosis H37Rv 207-214 23143285-5 2013 Furthermore, correlation analysis revealed that the over-expression of both Rv1217c and Rv1218c resulted in the higher minimum inhibition concentrations (MICs) of rifampicin (RIF) (OR = 1.01, P < 0.05 of Rv1217c; OR = 1.23, P < 0.05 of Rv1218c), while the over-expression of Rv1218c only led to the higher MICs of isoniazid (INH) (OR = 1.17, P < 0.05). Rifampin 163-173 tetronasin ABC transporter ATP-binding protein Mycobacterium tuberculosis H37Rv 242-249 23143285-5 2013 Furthermore, correlation analysis revealed that the over-expression of both Rv1217c and Rv1218c resulted in the higher minimum inhibition concentrations (MICs) of rifampicin (RIF) (OR = 1.01, P < 0.05 of Rv1217c; OR = 1.23, P < 0.05 of Rv1218c), while the over-expression of Rv1218c only led to the higher MICs of isoniazid (INH) (OR = 1.17, P < 0.05). Rifampin 163-173 tetronasin ABC transporter ATP-binding protein Mycobacterium tuberculosis H37Rv 242-249 23360470-6 2013 The P-gp inhibitor GF120918 increased the permeability for darunavir by 400% in rifampicin-treated mice, whereas this was only 56% in mice that were not treated, thus indicating P-gp induction by rifampicin. Rifampin 80-90 phosphoglycolate phosphatase Mus musculus 4-8 23360470-6 2013 The P-gp inhibitor GF120918 increased the permeability for darunavir by 400% in rifampicin-treated mice, whereas this was only 56% in mice that were not treated, thus indicating P-gp induction by rifampicin. Rifampin 80-90 phosphoglycolate phosphatase Mus musculus 178-182 23360470-6 2013 The P-gp inhibitor GF120918 increased the permeability for darunavir by 400% in rifampicin-treated mice, whereas this was only 56% in mice that were not treated, thus indicating P-gp induction by rifampicin. Rifampin 196-206 phosphoglycolate phosphatase Mus musculus 4-8 23360470-6 2013 The P-gp inhibitor GF120918 increased the permeability for darunavir by 400% in rifampicin-treated mice, whereas this was only 56% in mice that were not treated, thus indicating P-gp induction by rifampicin. Rifampin 196-206 phosphoglycolate phosphatase Mus musculus 178-182 23360470-8 2013 Quantitative Western blot analysis of the intestinal tissue showed that rifampicin treatment induced intestinal P-gp levels 4-fold, while CYP3A4 levels remained unchanged. Rifampin 72-82 phosphoglycolate phosphatase Mus musculus 112-116 23320515-0 2013 Global metabolomics and targeted steroid profiling reveal that rifampin, a strong human PXR activator, alters endogenous urinary steroid markers. Rifampin 63-71 nuclear receptor subfamily 1 group I member 2 Homo sapiens 88-91 23255551-6 2013 Further study on other Oatp1a2 substrates showed that the high affinity component for E-3-S is responsible for the interaction with taurocholate, bromsulphthalein, and rifampicin and is sensitive to proton concentration change, whereas the low affinity binding site is only involved in the binding of the antitumor drug methotrexate and had no response to change of pH. Rifampin 168-178 solute carrier organic anion transporter family member 1A2 Homo sapiens 23-30 23320515-2 2013 To identify endogenous biomarkers of PXR activation in humans, rifampin, a strong PXR activator, was administered to 12 healthy male subjects, and their urine was analyzed before and after rifampin administration. Rifampin 63-71 nuclear receptor subfamily 1 group I member 2 Homo sapiens 37-40 23320515-2 2013 To identify endogenous biomarkers of PXR activation in humans, rifampin, a strong PXR activator, was administered to 12 healthy male subjects, and their urine was analyzed before and after rifampin administration. Rifampin 63-71 nuclear receptor subfamily 1 group I member 2 Homo sapiens 82-85 23264496-5 2013 The PXR ligand rifampicin further increased the expression of CYP3A4, and siRNA-mediated knock-down of PXR in confluent cells resulted in decreased CYP3A4 expression. Rifampin 15-25 nuclear receptor subfamily 1 group I member 2 Homo sapiens 4-7 23264496-5 2013 The PXR ligand rifampicin further increased the expression of CYP3A4, and siRNA-mediated knock-down of PXR in confluent cells resulted in decreased CYP3A4 expression. Rifampin 15-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 22642697-9 2013 CONCLUSION: CYP3A inducers, rifampicin and efavirenz, can reduce telaprevir exposure to varying degrees based on their potency. Rifampin 28-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 12-17 23071008-1 2013 Induction of the cytochrome P450 (CYP) family of enzymes by coadministered compounds can result in drug-drug interactions, as in the case of the coadministration of rifampicin with many CYP3A substrates, including midazolam. Rifampin 165-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-32 22803519-2 2013 The aim of this study was to determine the effect of rifampicin, a CYP3A4 inducer, on the plasma pharmacokinetics of eribulin in patients with solid tumours. Rifampin 53-63 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 67-73 23071008-1 2013 Induction of the cytochrome P450 (CYP) family of enzymes by coadministered compounds can result in drug-drug interactions, as in the case of the coadministration of rifampicin with many CYP3A substrates, including midazolam. Rifampin 165-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 23071008-1 2013 Induction of the cytochrome P450 (CYP) family of enzymes by coadministered compounds can result in drug-drug interactions, as in the case of the coadministration of rifampicin with many CYP3A substrates, including midazolam. Rifampin 165-175 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 186-191 23071008-7 2013 The E(max) and EC(50) values in two separate lots of hepatocytes for CYP3A induction by rifampicin in a 96-well format were similar when discrete probe was compared with the probe cocktail. Rifampin 88-98 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-74 23440123-2 2013 We detected the C526T and C531T mutations in the rifampicin resistance-determining region (RRDR) of the rpoB gene with qPCR-HRM using plasmid-based controls. Rifampin 49-59 DNA-directed RNA polymerase subunit beta Mycobacterium tuberculosis H37Rv 104-108 22889553-0 2013 Expression and inducibility of CYP1A1, 1A2, 1B1 by beta-naphthoflavone and CYP2B22, CYP3As by rifampicin in heart regions and coronary arteries of pig. Rifampin 94-104 cytochrome P450 family 1 subfamily A member 1 Sus scrofa 31-37 22889553-0 2013 Expression and inducibility of CYP1A1, 1A2, 1B1 by beta-naphthoflavone and CYP2B22, CYP3As by rifampicin in heart regions and coronary arteries of pig. Rifampin 94-104 cytochrome P450 family 2 subfamily B member 6B Sus scrofa 75-82 22889553-3 2013 The rifampicin treatment resulted in an induction of CYP2B22 and CYP3As, at the transcriptional, activity and protein level in liver but not in heart nor in the coronary arteries, despite the expression of CAR and PXR in the cardiac tissues. Rifampin 4-14 cytochrome P450 family 2 subfamily B member 6B Sus scrofa 53-60 22889553-3 2013 The rifampicin treatment resulted in an induction of CYP2B22 and CYP3As, at the transcriptional, activity and protein level in liver but not in heart nor in the coronary arteries, despite the expression of CAR and PXR in the cardiac tissues. Rifampin 4-14 CXADR Ig-like cell adhesion molecule Sus scrofa 206-209 22889553-3 2013 The rifampicin treatment resulted in an induction of CYP2B22 and CYP3As, at the transcriptional, activity and protein level in liver but not in heart nor in the coronary arteries, despite the expression of CAR and PXR in the cardiac tissues. Rifampin 4-14 nuclear receptor subfamily 1 group I member 2 Sus scrofa 214-217 23160820-8 2013 Of importance, ectopic overexpression and silencing of endogenous RBCK1 in primary human hepatocytes resulted in a decrease and increase, respectively, in endogenous PXR protein levels and in the induction of PXR target genes by rifampicin. Rifampin 229-239 RANBP2-type and C3HC4-type zinc finger containing 1 Homo sapiens 66-71 23374512-10 2013 RESULTS: The vancomycin-resistant isolate CP2 was found to be resistant to oxacillin, chloramphenicol, erythromycin, rifampicin, gentamicin, tetracycline and ciprofloxacin, as well. Rifampin 117-127 ceruloplasmin Homo sapiens 42-45 23339213-36 2013 Thirteen days later, the CSF culture became positive for Mycobacterium tuberculosis sensitive to streptomycin, isoniazid, ethambutol, rifampin, and pyrazinamide. Rifampin 134-142 colony stimulating factor 2 Homo sapiens 25-28 23400746-4 2013 In vitro studies indicated that ketoconazole inhibited the uptake of fimasartan into cells expressing OATP1B1 with a K(i) of 107.7 microM, and rifampicin inhibited OAT1- and OATP1B1-mediated fimasartan transport with a K(i) of 212 microM and 12.2 microM, respectively. Rifampin 143-153 solute carrier family 22 member 6 Homo sapiens 164-168 23273219-3 2013 beta-Amyloid aggregation results showed that all compounds exhibited remarkable Abeta fibril aggregation inhibition activity with a nearly similar potential as the reference compound rifampicin, which makes them promising anti-Alzheimer drug candidates. Rifampin 183-193 amyloid beta precursor protein Homo sapiens 80-85 23095030-13 2013 Isolates with a DNA profile compatible with the BI/NAP1/027 ribotype had, on the average, higher MICs of rifampin. Rifampin 105-113 nucleosome assembly protein 1 like 1 Homo sapiens 51-55 23123212-5 2013 Xenobiotics like rifampicin and hyperforin activate PXR and induce Abcb1 expression. Rifampin 17-27 nuclear receptor subfamily 1 group I member 2 Homo sapiens 52-55 23123212-5 2013 Xenobiotics like rifampicin and hyperforin activate PXR and induce Abcb1 expression. Rifampin 17-27 ATP binding cassette subfamily B member 1 Homo sapiens 67-72 23123212-8 2013 For activation of the pregnane X receptor the ligands hyperforin and rifampicin were used. Rifampin 69-79 nuclear receptor subfamily 1 group I member 2 Homo sapiens 22-41 23123212-12 2013 In conclusion our data prove PXR activation by rifampicin and hyperforin lead to an increased ABC-transporter expression and transport activity. Rifampin 47-57 nuclear receptor subfamily 1 group I member 2 Homo sapiens 29-32 23123212-12 2013 In conclusion our data prove PXR activation by rifampicin and hyperforin lead to an increased ABC-transporter expression and transport activity. Rifampin 47-57 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 94-109 23137910-8 2013 Cotreatment with the prototypical pregnane X receptor activator, rifampicin, significantly reversed decitabine-induced ABCC3 and SLCO2A1 expression. Rifampin 65-75 nuclear receptor subfamily 1 group I member 2 Homo sapiens 34-53 23137910-8 2013 Cotreatment with the prototypical pregnane X receptor activator, rifampicin, significantly reversed decitabine-induced ABCC3 and SLCO2A1 expression. Rifampin 65-75 ATP binding cassette subfamily C member 3 Homo sapiens 119-124 23137910-8 2013 Cotreatment with the prototypical pregnane X receptor activator, rifampicin, significantly reversed decitabine-induced ABCC3 and SLCO2A1 expression. Rifampin 65-75 solute carrier organic anion transporter family member 2A1 Homo sapiens 129-136 24852442-6 2013 Rifampicin significantly inhibited the overexpression of TLR2, TLR4, CD14 and IL8Rs. Rifampin 0-10 toll like receptor 2 Homo sapiens 57-61 24852442-6 2013 Rifampicin significantly inhibited the overexpression of TLR2, TLR4, CD14 and IL8Rs. Rifampin 0-10 toll like receptor 4 Homo sapiens 63-67 24852442-6 2013 Rifampicin significantly inhibited the overexpression of TLR2, TLR4, CD14 and IL8Rs. Rifampin 0-10 CD14 molecule Homo sapiens 69-73 23115085-5 2013 Rifampicin inhibited the uptake of fexofenadine enantiomers by human hepatocytes via organic anion transporter (OAT) OATP1B3 and its basal-to-apical transport in Caco-2 cells, but not OAT3-mediated or multidrug and toxic compound extrusion 1 (MATE1)-mediated transport. Rifampin 0-10 solute carrier organic anion transporter family member 1B3 Homo sapiens 117-124 23115085-5 2013 Rifampicin inhibited the uptake of fexofenadine enantiomers by human hepatocytes via organic anion transporter (OAT) OATP1B3 and its basal-to-apical transport in Caco-2 cells, but not OAT3-mediated or multidrug and toxic compound extrusion 1 (MATE1)-mediated transport. Rifampin 0-10 solute carrier family 22 member 8 Homo sapiens 184-188 23115085-5 2013 Rifampicin inhibited the uptake of fexofenadine enantiomers by human hepatocytes via organic anion transporter (OAT) OATP1B3 and its basal-to-apical transport in Caco-2 cells, but not OAT3-mediated or multidrug and toxic compound extrusion 1 (MATE1)-mediated transport. Rifampin 0-10 solute carrier family 47 member 1 Homo sapiens 243-248 23115085-7 2013 The rifampicin-sensitive uptake by hepatocytes was 1.6 times higher for R-fexofenadine, whereas the transport activities by OATP1B3, OAT3, MATE1, or P-glycoprotein were identical for both enantiomers. Rifampin 4-14 solute carrier family 22 member 8 Homo sapiens 133-137 23115085-9 2013 In conclusion, rifampicin has multiple interaction sites with fexofenadine, all of which contribute to increasing the area under the curve of fexofenadine when they are given simultaneously, to surpass the effect of the induction of P-glycoprotein elicited by multiple doses. Rifampin 15-25 ATP binding cassette subfamily B member 1 Homo sapiens 233-247 23400746-4 2013 In vitro studies indicated that ketoconazole inhibited the uptake of fimasartan into cells expressing OATP1B1 with a K(i) of 107.7 microM, and rifampicin inhibited OAT1- and OATP1B1-mediated fimasartan transport with a K(i) of 212 microM and 12.2 microM, respectively. Rifampin 143-153 solute carrier organic anion transporter family member 1B1 Homo sapiens 174-181 23841924-3 2013 The metabolism and bioavailability of cyclosporin can be significantly and persistently influenced through induction of CYP3A4 caused by the concomitant use of rifampicin. Rifampin 160-170 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 120-126 23600152-5 2013 The positive hPXR/hCAR activators rifampicin and CITCO were applied to make sure that the reporter gene model was successfully established. Rifampin 34-44 CXADR Ig-like cell adhesion molecule Homo sapiens 18-22 23164983-4 2013 When bovine embryos were cultured with forskolin and/or rifampicin, the ABCB1 level was significantly increased in blastocysts but embryo development was not significantly improved. Rifampin 56-66 LOW QUALITY PROTEIN: multidrug resistance protein 1 Bos taurus 72-77 23402082-4 2012 Rifampicin, the most effective antipruritic agent in cholestatic itch, has been shown to reduce autotaxin transcription in vitro. Rifampin 0-10 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 96-105 23153560-5 2013 The potent PXR ligand - rifampicin (300mg/d) was given alone for 7days in arm 1, or in daily combination with 450mumol SFN (Broccoli Sprout extract) in arm 2; SFN was given alone in arm 3. Rifampin 24-34 nuclear receptor subfamily 1 group I member 2 Homo sapiens 11-14 23153560-7 2013 Rifampicin alone decreased midazolam AUC by 70%, indicative of the expected increase in CYP3A4 activity. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-94 22676424-3 2012 We examined the effect rifampicin, a well-known inducer of many cytochrome P450 (CYP) enzymes and transporters, on the pharmacokinetics of intravenous and oral S-ketamine in healthy volunteers. Rifampin 23-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 22834478-6 2012 Whereas positive controls (100 microM omeprazole and 10 microM rifampin) caused the anticipated CYP induction, the highest concentration of mirabegron (10 microM; 37 times plasma C(max)) had minimal effect on CYP1A2 and CYP3A4/5 activity, and CYP1A2 and CYP3A4 mRNA levels in freshly isolated human hepatocytes, suggesting that mirabegron is not an inducer of these enzymes. Rifampin 63-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 22676424-3 2012 We examined the effect rifampicin, a well-known inducer of many cytochrome P450 (CYP) enzymes and transporters, on the pharmacokinetics of intravenous and oral S-ketamine in healthy volunteers. Rifampin 23-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 22250979-6 2012 When rifampicin was stopped, IDV/r was reduced to 400/100 mg BID. Rifampin 5-15 BH3 interacting domain death agonist Homo sapiens 61-64 22809809-7 2012 The compounds 3-methylcholanthrene (a CYP1A2 inducer), phenobarbital (a CYP2B6 inducer), and rifampicin (a CYP3A4/5 inducer) served as positive controls. Rifampin 93-103 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 107-113 22473838-5 2012 Rifampicin (RMP) significantly reduced itch intensity and ATX activity in patients with pruritus not responding to bile salt sequestrants. Rifampin 0-10 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 58-61 22445438-14 2012 Coadministration of CYP3A4 modulators should be avoided where possible, and another type of contraception should be used when coadministration of CYP3A4 inducers like rifampicin is unavoidable. Rifampin 167-177 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 146-152 22829544-5 2012 Although phosphomimetic mutations at Thr290 and Thr408 retained the PXR protein in cytoplasm and attenuated the induction of UGT1A1 expression by both roscovitine and rifampicin, a mutation at Ser350 specifically reduced the activity of PXR induced by roscovitine. Rifampin 167-177 nuclear receptor subfamily 1 group I member 2 Homo sapiens 68-71 22829544-5 2012 Although phosphomimetic mutations at Thr290 and Thr408 retained the PXR protein in cytoplasm and attenuated the induction of UGT1A1 expression by both roscovitine and rifampicin, a mutation at Ser350 specifically reduced the activity of PXR induced by roscovitine. Rifampin 167-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 22829544-5 2012 Although phosphomimetic mutations at Thr290 and Thr408 retained the PXR protein in cytoplasm and attenuated the induction of UGT1A1 expression by both roscovitine and rifampicin, a mutation at Ser350 specifically reduced the activity of PXR induced by roscovitine. Rifampin 167-177 nuclear receptor subfamily 1 group I member 2 Homo sapiens 237-240 22473838-5 2012 Rifampicin (RMP) significantly reduced itch intensity and ATX activity in patients with pruritus not responding to bile salt sequestrants. Rifampin 12-15 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 58-61 22875972-8 2012 In contrast, when virion envelope formation was blocked at a later step by repression of A14 expression or by rifampin treatment, A11 colocalized with virion membrane proteins in the factories. Rifampin 110-118 DXS435E Homo sapiens 130-133 22733800-7 2012 Differentiating cells showed significant induction in the expression of CYP1A1, 2B6, 2E1, 3A4, AHR, CAR, PXR, and GSTP1-1 when exposed to rifampin, a known universal inducer of CYPs. Rifampin 138-146 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 72-78 22638872-4 2012 DDI with OATP1B1 inhibitors, cyclosporine, gemfibrozil and rifampin, was also simulated using inhibition constant (Ki) values. Rifampin 59-67 solute carrier organic anion transporter family member 1B1 Homo sapiens 9-16 22733800-7 2012 Differentiating cells showed significant induction in the expression of CYP1A1, 2B6, 2E1, 3A4, AHR, CAR, PXR, and GSTP1-1 when exposed to rifampin, a known universal inducer of CYPs. Rifampin 138-146 aryl hydrocarbon receptor Homo sapiens 95-98 22733800-7 2012 Differentiating cells showed significant induction in the expression of CYP1A1, 2B6, 2E1, 3A4, AHR, CAR, PXR, and GSTP1-1 when exposed to rifampin, a known universal inducer of CYPs. Rifampin 138-146 nuclear receptor subfamily 1 group I member 3 Homo sapiens 100-103 22825545-6 2012 We demonstrated that higher concentrations of rifampicin (>50 muM) change the expression of reference genes and thus may complicate and adulterate normalization of qRT-PCR data. Rifampin 46-56 latexin Homo sapiens 65-68 22733800-7 2012 Differentiating cells showed significant induction in the expression of CYP1A1, 2B6, 2E1, 3A4, AHR, CAR, PXR, and GSTP1-1 when exposed to rifampin, a known universal inducer of CYPs. Rifampin 138-146 nuclear receptor subfamily 1 group I member 2 Homo sapiens 105-108 22733800-7 2012 Differentiating cells showed significant induction in the expression of CYP1A1, 2B6, 2E1, 3A4, AHR, CAR, PXR, and GSTP1-1 when exposed to rifampin, a known universal inducer of CYPs. Rifampin 138-146 glutathione S-transferase pi 1 Homo sapiens 114-121 22727860-3 2012 Pretreatment with rifampin significantly decreased serum alanine transaminase (ALT) activities, aspartate transaminase (AST) activities and prevented severe liver necrosis following acetaminophen overdose. Rifampin 18-26 glutamic pyruvic transaminase, soluble Mus musculus 57-77 22847250-0 2012 Effect of pH, mucin and bovine serum on rifampicin permeability through Caco-2 cells. Rifampin 40-50 LOC100508689 Homo sapiens 14-19 22727860-3 2012 Pretreatment with rifampin significantly decreased serum alanine transaminase (ALT) activities, aspartate transaminase (AST) activities and prevented severe liver necrosis following acetaminophen overdose. Rifampin 18-26 glutamic pyruvic transaminase, soluble Mus musculus 79-82 22727860-3 2012 Pretreatment with rifampin significantly decreased serum alanine transaminase (ALT) activities, aspartate transaminase (AST) activities and prevented severe liver necrosis following acetaminophen overdose. Rifampin 18-26 solute carrier family 17 (anion/sugar transporter), member 5 Mus musculus 96-118 22727860-3 2012 Pretreatment with rifampin significantly decreased serum alanine transaminase (ALT) activities, aspartate transaminase (AST) activities and prevented severe liver necrosis following acetaminophen overdose. Rifampin 18-26 solute carrier family 17 (anion/sugar transporter), member 5 Mus musculus 120-123 22664347-4 2012 Cells were exposed to a range of non-coplanar PCBs (99, 138, 153, 180 and 194), or the coplanar PCB77: Direct activation of PXR and CAR was measured using a mammalian receptor activation assay in human liver cells, with rifampicin and CITCO used as positive controls ligands for PXR and CAR, respectively; activation of target gene expression was examined using reporter gene plasmids for CYP3A4 and MDR1 transfected into liver, intestine and lung cell lines. Rifampin 220-230 nuclear receptor subfamily 1 group I member 2 Homo sapiens 124-127 23015129-1 2012 This study investigated the therapeutic effects of interleukin (IL)-2 and granulocyte-macrophage colony-stimulating factor (GM-CSF) co-administrated with antibacterial agents isoniazid (INH) and rifampin (RIF) to treat a mouse model of tuberculosis (TB) infection. Rifampin 195-203 colony stimulating factor 2 (granulocyte-macrophage) Mus musculus 124-130 23015129-1 2012 This study investigated the therapeutic effects of interleukin (IL)-2 and granulocyte-macrophage colony-stimulating factor (GM-CSF) co-administrated with antibacterial agents isoniazid (INH) and rifampin (RIF) to treat a mouse model of tuberculosis (TB) infection. Rifampin 205-208 colony stimulating factor 2 (granulocyte-macrophage) Mus musculus 124-130 22525741-2 2012 Rifampicin has been reported to be an effective treatment of jaundice and pruritus in patients with BRIC. Rifampin 0-10 ATPase phospholipid transporting 8B1 Homo sapiens 100-104 22525741-3 2012 Proposed mechanisms of effect for rifampicin include enhancement of multidrug-resistance protein 2 expression, activation of the enzymes of uridine diphosphate glucuronosyltransferase 1A1 and cytochrome P450 3A4, and stimulation of 6alpha-hydroxylation of bile acids. Rifampin 34-44 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 192-211 22525741-4 2012 METHODS: To confirm the diagnosis of BRIC-1 and demonstrate the effect of rifampicin treatment on bile acid metabolism, we analyzed the patient"s ATP8B1 gene and bile acids in urine. Rifampin 74-84 ATPase phospholipid transporting 8B1 Homo sapiens 146-152 22525741-5 2012 RESULTS: We detected 2 heterozygous mutations in the ATP8B1 gene, and increasing amounts of unusual bile acids such as 1beta-hydroxylated cholic acid, 2beta-hydroxylated cholic acid, 4beta-hydroxylated cholic acid, 6alpha-hydroxylated cholic acid, and hyocholic acid in urine during rifampicin treatment. Rifampin 283-293 ATPase phospholipid transporting 8B1 Homo sapiens 53-59 22339773-3 2012 Thus, the aim of this study is to investigate maternal, placental and foetal Abcb1 expression and function in pregnant rats after induction with rifampicin, dexamethasone, St. John"s wort (SJW) or thyroxine. Rifampin 145-155 ATP binding cassette subfamily B member 1A Rattus norvegicus 77-82 22644021-11 2012 While rifampin plus ceftriaxone versus ceftriaxone also led to lower CSF inflammation (P < 0.02 for IL-6 at 6 h), it had no significant effect on apoptosis and hearing capacity. Rifampin 6-14 interleukin 6 Rattus norvegicus 103-107 22644026-10 2012 Lersivirine should not be coadministered with rifampin, which is a potent inducer of CYP3A4, UGT2B7, and P-glycoprotein activity and thus substantially lowers lersivirine exposure. Rifampin 46-54 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 85-91 22644026-10 2012 Lersivirine should not be coadministered with rifampin, which is a potent inducer of CYP3A4, UGT2B7, and P-glycoprotein activity and thus substantially lowers lersivirine exposure. Rifampin 46-54 UDP glucuronosyltransferase family 2 member B7 Homo sapiens 93-99 22339773-9 2012 Rifampicin induced Abcb1 in all maternal and foetal organs. Rifampin 0-10 ATP binding cassette subfamily B member 1A Rattus norvegicus 19-24 22339773-12 2012 Although rifampicin regulates maternal and foetal Abcb1 expression, organ distribution of talinolol remains unchanged most likely caused by the known inhibitory effect of rifampicin on Abcb1 function. Rifampin 9-19 ATP binding cassette subfamily B member 1A Rattus norvegicus 50-55 22339773-12 2012 Although rifampicin regulates maternal and foetal Abcb1 expression, organ distribution of talinolol remains unchanged most likely caused by the known inhibitory effect of rifampicin on Abcb1 function. Rifampin 171-181 ATP binding cassette subfamily B member 1A Rattus norvegicus 185-190 22636321-7 2012 Oral rifampicin was effective in clearing infection and halting further progression of focal inflammation from infected IFN-gamma(-/-) mice, although some symptoms and swelling remained. Rifampin 5-15 interferon gamma Mus musculus 120-129 22562045-3 2012 Treatment with the hPXR agonist rifampin significantly increased CYP3A4 mRNA content and catalytic activity, but had no effect on CYP24A1 or TRPV6 mRNA content. Rifampin 32-40 nuclear receptor subfamily 1 group I member 2 Homo sapiens 19-23 22562045-3 2012 Treatment with the hPXR agonist rifampin significantly increased CYP3A4 mRNA content and catalytic activity, but had no effect on CYP24A1 or TRPV6 mRNA content. Rifampin 32-40 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 65-71 22562045-4 2012 Pre-treating cells with rifampin for 48h, prior to a 24h 1alpha,25(OH)(2)D(3) treatment phase, was associated with a subsequent 48% increase in the elimination of 1alpha,25(OH)(2)D(3) and a 35% reduction of peak TRPV6 mRNA. Rifampin 24-32 transient receptor potential cation channel subfamily V member 6 Homo sapiens 212-217 22562045-5 2012 Introduction of the CYP3A4 inhibitor, 6",7"-dihydroxybergamottin, an active inhibitor in grapefruit juice, reversed the effects of rifampin on 1alpha,25(OH)(2)D(3) clearance and TRPV6 expression. Rifampin 131-139 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-26 22562045-5 2012 Introduction of the CYP3A4 inhibitor, 6",7"-dihydroxybergamottin, an active inhibitor in grapefruit juice, reversed the effects of rifampin on 1alpha,25(OH)(2)D(3) clearance and TRPV6 expression. Rifampin 131-139 transient receptor potential cation channel subfamily V member 6 Homo sapiens 178-183 22562045-6 2012 Over-expression of hPXR in LS180 cells greatly enhanced the CYP3A4 responsiveness to rifampin pretreatment, and elicited a greater relative suppression of TRPV6 expression and an increase in 1alpha,25(OH)(2)D(3) disappearance rate, compared to vector expressed cells, following hormone administration. Rifampin 85-93 nuclear receptor subfamily 1 group I member 2 Homo sapiens 19-23 22562045-6 2012 Over-expression of hPXR in LS180 cells greatly enhanced the CYP3A4 responsiveness to rifampin pretreatment, and elicited a greater relative suppression of TRPV6 expression and an increase in 1alpha,25(OH)(2)D(3) disappearance rate, compared to vector expressed cells, following hormone administration. Rifampin 85-93 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 60-66 22585454-10 2012 CONCLUSION: Chemoprophylaxis with isoniazid and rifampicin generally decreased IFN-gamma levels among tuberculosis contacts. Rifampin 48-58 interferon gamma Homo sapiens 79-88 23087510-2 2012 Rifampicin is a strong CYP enzyme inducer while isoniazid is a potent CYP inhibitor. Rifampin 0-10 peptidylprolyl isomerase G Homo sapiens 23-26 22617226-0 2012 Effects of CYP induction by rifampicin on tamoxifen exposure. Rifampin 28-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-14 22617226-3 2012 We evaluated the effects of cytochrome P450 (CYP) induction by rifampicin on the exposure levels of tamoxifen and its metabolites and found that coadministration of rifampicin resulted in markedly reduced (up to 86%, P <= 0.040) concentrations of tamoxifen and its metabolites. Rifampin 63-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 22617226-3 2012 We evaluated the effects of cytochrome P450 (CYP) induction by rifampicin on the exposure levels of tamoxifen and its metabolites and found that coadministration of rifampicin resulted in markedly reduced (up to 86%, P <= 0.040) concentrations of tamoxifen and its metabolites. Rifampin 63-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 22617226-3 2012 We evaluated the effects of cytochrome P450 (CYP) induction by rifampicin on the exposure levels of tamoxifen and its metabolites and found that coadministration of rifampicin resulted in markedly reduced (up to 86%, P <= 0.040) concentrations of tamoxifen and its metabolites. Rifampin 165-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 22617226-3 2012 We evaluated the effects of cytochrome P450 (CYP) induction by rifampicin on the exposure levels of tamoxifen and its metabolites and found that coadministration of rifampicin resulted in markedly reduced (up to 86%, P <= 0.040) concentrations of tamoxifen and its metabolites. Rifampin 165-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 21602517-4 2012 Pretreatment with rifampin, a potent CYP3A4 inducer, decreased ruxolitinib AUC(0- ) by 71% while resulting in only a 10% decrease in the overall PD activity. Rifampin 18-26 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-43 22620706-9 2012 RESULTS: Rifampicin treatment upregulated duodenal P-gp in dogs and significantly reduced the area under the plasma concentration-time curve of prednisolone. Rifampin 9-19 PGP Canis lupus familiaris 51-55 22562052-0 2012 SLCO1B1 *15 haplotype is associated with rifampin-induced liver injury. Rifampin 41-49 solute carrier organic anion transporter family member 1B1 Homo sapiens 0-7 22562052-1 2012 The organic anion transporting polypeptide 1B1 (OATP1B1, encoded by SLCO1B1) plays an important role in the transport of endogenous and xenobiotic compounds, such as bile acids and rifampin. Rifampin 181-189 solute carrier organic anion transporter family member 1B1 Homo sapiens 4-46 22562052-1 2012 The organic anion transporting polypeptide 1B1 (OATP1B1, encoded by SLCO1B1) plays an important role in the transport of endogenous and xenobiotic compounds, such as bile acids and rifampin. Rifampin 181-189 solute carrier organic anion transporter family member 1B1 Homo sapiens 48-55 22562052-1 2012 The organic anion transporting polypeptide 1B1 (OATP1B1, encoded by SLCO1B1) plays an important role in the transport of endogenous and xenobiotic compounds, such as bile acids and rifampin. Rifampin 181-189 solute carrier organic anion transporter family member 1B1 Homo sapiens 68-75 22562052-13 2012 These results suggest that the OATP1B1 *15 haplotype is an important predisposing factor for rifampin-induced liver injury. Rifampin 93-101 solute carrier organic anion transporter family member 1B1 Homo sapiens 31-38 22559005-3 2012 Here we investigated the effects of exposure of carp (Cyprinus carpio) primary hepatocytes to the human PXR agonist rifampicin (RIF) on expression of target genes involved in phase I (cyp2k, cyp3a) and phase II (gstalpha, gstpi) drug metabolism and drug transporters mdr1 and mrp2. Rifampin 116-126 nuclear receptor subfamily 1 group I member 2 Homo sapiens 104-107 22559005-3 2012 Here we investigated the effects of exposure of carp (Cyprinus carpio) primary hepatocytes to the human PXR agonist rifampicin (RIF) on expression of target genes involved in phase I (cyp2k, cyp3a) and phase II (gstalpha, gstpi) drug metabolism and drug transporters mdr1 and mrp2. Rifampin 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 191-196 22559005-3 2012 Here we investigated the effects of exposure of carp (Cyprinus carpio) primary hepatocytes to the human PXR agonist rifampicin (RIF) on expression of target genes involved in phase I (cyp2k, cyp3a) and phase II (gstalpha, gstpi) drug metabolism and drug transporters mdr1 and mrp2. Rifampin 116-126 ATP binding cassette subfamily B member 1 Homo sapiens 267-271 22559005-3 2012 Here we investigated the effects of exposure of carp (Cyprinus carpio) primary hepatocytes to the human PXR agonist rifampicin (RIF) on expression of target genes involved in phase I (cyp2k, cyp3a) and phase II (gstalpha, gstpi) drug metabolism and drug transporters mdr1 and mrp2. Rifampin 116-126 ATP binding cassette subfamily C member 2 Homo sapiens 276-280 22559005-3 2012 Here we investigated the effects of exposure of carp (Cyprinus carpio) primary hepatocytes to the human PXR agonist rifampicin (RIF) on expression of target genes involved in phase I (cyp2k, cyp3a) and phase II (gstalpha, gstpi) drug metabolism and drug transporters mdr1 and mrp2. Rifampin 128-131 nuclear receptor subfamily 1 group I member 2 Homo sapiens 104-107 22559005-3 2012 Here we investigated the effects of exposure of carp (Cyprinus carpio) primary hepatocytes to the human PXR agonist rifampicin (RIF) on expression of target genes involved in phase I (cyp2k, cyp3a) and phase II (gstalpha, gstpi) drug metabolism and drug transporters mdr1 and mrp2. Rifampin 128-131 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 191-196 22559005-3 2012 Here we investigated the effects of exposure of carp (Cyprinus carpio) primary hepatocytes to the human PXR agonist rifampicin (RIF) on expression of target genes involved in phase I (cyp2k, cyp3a) and phase II (gstalpha, gstpi) drug metabolism and drug transporters mdr1 and mrp2. Rifampin 128-131 ATP binding cassette subfamily B member 1 Homo sapiens 267-271 22559005-3 2012 Here we investigated the effects of exposure of carp (Cyprinus carpio) primary hepatocytes to the human PXR agonist rifampicin (RIF) on expression of target genes involved in phase I (cyp2k, cyp3a) and phase II (gstalpha, gstpi) drug metabolism and drug transporters mdr1 and mrp2. Rifampin 128-131 ATP binding cassette subfamily C member 2 Homo sapiens 276-280 22534255-3 2012 OBJECTIVE: The effects of multiple oral doses of ketoconazole (a potent CYP3A4 inhibitor) and multiple oral doses of rifampicin (a potent CYP3A4 inducer) on the pharmacokinetic properties of a single oral dose of gemigliptin were evaluated in fasting healthy male Korean volunteers. Rifampin 117-127 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 138-144 22499612-8 2012 Rifampicin (600 mg by mouth), a potent OATP inhibitor, significantly reduced the radioactivity excreted into the bile (by 44%) by inhibiting both uptake (by 45%) and subsequent canalicular efflux (by 62%). Rifampin 0-10 solute carrier organic anion transporter family member 1A2 Homo sapiens 39-43 22314385-5 2012 Walrycin A was as efficient as the reference PXR agonist rifampicin to activate PXR in a transactivation assay at noncytotoxic concentrations. Rifampin 57-67 nuclear receptor subfamily 1 group I member 2 Homo sapiens 45-48 22314385-5 2012 Walrycin A was as efficient as the reference PXR agonist rifampicin to activate PXR in a transactivation assay at noncytotoxic concentrations. Rifampin 57-67 nuclear receptor subfamily 1 group I member 2 Homo sapiens 80-83 22207054-1 2012 Human arylacetamide deacetylase (AADAC) is a major esterase responsible for the hydrolysis of clinical drugs such as flutamide, phenacetin, and rifampicin. Rifampin 144-154 arylacetamide deacetylase Homo sapiens 6-31 22207054-1 2012 Human arylacetamide deacetylase (AADAC) is a major esterase responsible for the hydrolysis of clinical drugs such as flutamide, phenacetin, and rifampicin. Rifampin 144-154 arylacetamide deacetylase Homo sapiens 33-38 22664508-0 2012 Need to confirm isoniazid susceptibility in Xpert MTB/RIF rifampin susceptible cases. Rifampin 58-66 ras homolog family member F, filopodia associated Homo sapiens 54-57 22213739-6 2012 Between January 2006 and December 2008, LiPA saved 25.3 and 32.2 days for TB diagnosis and rifampicin resistance of smear-positive samples, respectively. Rifampin 91-101 lipase A, lysosomal acid type Homo sapiens 40-44 22126990-5 2012 We evaluated the inductive effect of rifampicin (RIF), a typical human PXR ligand, on the plasma exposure to the four P450 substrate drugs (triazolam/CYP3A4, pioglitazone/CYP2C8, (S)-warfarin/CYP2C9, and (S)-(-)-mephenytoin/CYP2C19) by cassette dosing in PXB mice. Rifampin 37-47 nuclear receptor subfamily 1 group I member 2 Homo sapiens 71-74 22126990-5 2012 We evaluated the inductive effect of rifampicin (RIF), a typical human PXR ligand, on the plasma exposure to the four P450 substrate drugs (triazolam/CYP3A4, pioglitazone/CYP2C8, (S)-warfarin/CYP2C9, and (S)-(-)-mephenytoin/CYP2C19) by cassette dosing in PXB mice. Rifampin 49-52 nuclear receptor subfamily 1 group I member 2 Homo sapiens 71-74 22253426-8 2012 Glucocorticoids, including dexamethasone, cortisol, corticosterone, and cortisone all induced the expression of CYP3A7 mRNA, whereas rifampicin, an activator of PXR and an inducer of CYP3A4 in adult liver, had no effect on CYP3A7 expression. Rifampin 133-143 nuclear receptor subfamily 1, group I, member 2 Mus musculus 161-164 22253426-8 2012 Glucocorticoids, including dexamethasone, cortisol, corticosterone, and cortisone all induced the expression of CYP3A7 mRNA, whereas rifampicin, an activator of PXR and an inducer of CYP3A4 in adult liver, had no effect on CYP3A7 expression. Rifampin 133-143 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 183-189 22253426-8 2012 Glucocorticoids, including dexamethasone, cortisol, corticosterone, and cortisone all induced the expression of CYP3A7 mRNA, whereas rifampicin, an activator of PXR and an inducer of CYP3A4 in adult liver, had no effect on CYP3A7 expression. Rifampin 133-143 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 223-229 22281341-7 2012 In the rifampicin group, increases in the expression of mRNA for TNF-alpha, IL-1beta, IL-6, IL-8, and lactoferrin were observed at 6 h postinfection and in the expression of mRNA for TLR2 but not for TLR4 at 12 h postinfection. Rifampin 7-17 tumor necrosis factor Bos taurus 65-74 22281341-7 2012 In the rifampicin group, increases in the expression of mRNA for TNF-alpha, IL-1beta, IL-6, IL-8, and lactoferrin were observed at 6 h postinfection and in the expression of mRNA for TLR2 but not for TLR4 at 12 h postinfection. Rifampin 7-17 interleukin 1 beta Bos taurus 76-84 22281341-7 2012 In the rifampicin group, increases in the expression of mRNA for TNF-alpha, IL-1beta, IL-6, IL-8, and lactoferrin were observed at 6 h postinfection and in the expression of mRNA for TLR2 but not for TLR4 at 12 h postinfection. Rifampin 7-17 interferon beta-2 Bos taurus 86-90 22281341-7 2012 In the rifampicin group, increases in the expression of mRNA for TNF-alpha, IL-1beta, IL-6, IL-8, and lactoferrin were observed at 6 h postinfection and in the expression of mRNA for TLR2 but not for TLR4 at 12 h postinfection. Rifampin 7-17 C-X-C motif chemokine ligand 8 Bos taurus 92-96 22281341-7 2012 In the rifampicin group, increases in the expression of mRNA for TNF-alpha, IL-1beta, IL-6, IL-8, and lactoferrin were observed at 6 h postinfection and in the expression of mRNA for TLR2 but not for TLR4 at 12 h postinfection. Rifampin 7-17 lactotransferrin Bos taurus 102-113 22281341-7 2012 In the rifampicin group, increases in the expression of mRNA for TNF-alpha, IL-1beta, IL-6, IL-8, and lactoferrin were observed at 6 h postinfection and in the expression of mRNA for TLR2 but not for TLR4 at 12 h postinfection. Rifampin 7-17 toll like receptor 2 Bos taurus 183-187 22281341-7 2012 In the rifampicin group, increases in the expression of mRNA for TNF-alpha, IL-1beta, IL-6, IL-8, and lactoferrin were observed at 6 h postinfection and in the expression of mRNA for TLR2 but not for TLR4 at 12 h postinfection. Rifampin 7-17 toll like receptor 4 Bos taurus 200-204 22178124-9 2012 Among them, bufalin and cinobufagin significantly promoted the CYP3A4 enzyme activity, mRNA and protein levels, with the maximal induction challenging or exceeding that of the induction by rifampicin, indicating that they might play a critical role in CYP3A4 enzyme-inducing effects of SBP. Rifampin 189-199 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 63-69 22687401-3 2012 The induction of CALUX in HPL-A3 by hPXR activators, including rifampicin, occurred in time- and concentration-dependent fashions, whereas no such induction was observed using rat/mouse PXR activators, such as pregnenolone-16alpha-carbonitrile and dexamethasone. Rifampin 63-73 galectin 1 Homo sapiens 26-29 22687401-3 2012 The induction of CALUX in HPL-A3 by hPXR activators, including rifampicin, occurred in time- and concentration-dependent fashions, whereas no such induction was observed using rat/mouse PXR activators, such as pregnenolone-16alpha-carbonitrile and dexamethasone. Rifampin 63-73 nuclear receptor subfamily 1 group I member 2 Homo sapiens 36-40 22687401-3 2012 The induction of CALUX in HPL-A3 by hPXR activators, including rifampicin, occurred in time- and concentration-dependent fashions, whereas no such induction was observed using rat/mouse PXR activators, such as pregnenolone-16alpha-carbonitrile and dexamethasone. Rifampin 63-73 nuclear receptor subfamily 1, group I, member 2 Mus musculus 37-40 22076448-6 2012 The CYP3A4 reporter activity enhanced by overexpression of human PXR was further increased by treatment with PAHs and TCDD as well as by treatment with rifampicin. Rifampin 152-162 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 4-10 22076448-6 2012 The CYP3A4 reporter activity enhanced by overexpression of human PXR was further increased by treatment with PAHs and TCDD as well as by treatment with rifampicin. Rifampin 152-162 nuclear receptor subfamily 1 group I member 2 Homo sapiens 65-68 22504320-3 2012 In this study, we have hypothesized that enhanced amyloid-beta (Abeta) clearance from the brain across the blood-brain barrier (BBB) of wild-type mice treated with rifampicin or caffeine is caused by both drugs potential to upregulate low-density lipoprotein receptor related protein-1 (LRP1) and/or P-glycoprotein (P-gp) at the BBB. Rifampin 164-174 low density lipoprotein receptor-related protein 1 Mus musculus 235-285 22504320-3 2012 In this study, we have hypothesized that enhanced amyloid-beta (Abeta) clearance from the brain across the blood-brain barrier (BBB) of wild-type mice treated with rifampicin or caffeine is caused by both drugs potential to upregulate low-density lipoprotein receptor related protein-1 (LRP1) and/or P-glycoprotein (P-gp) at the BBB. Rifampin 164-174 low density lipoprotein receptor-related protein 1 Mus musculus 287-291 22504320-3 2012 In this study, we have hypothesized that enhanced amyloid-beta (Abeta) clearance from the brain across the blood-brain barrier (BBB) of wild-type mice treated with rifampicin or caffeine is caused by both drugs potential to upregulate low-density lipoprotein receptor related protein-1 (LRP1) and/or P-glycoprotein (P-gp) at the BBB. Rifampin 164-174 phosphoglycolate phosphatase Mus musculus 300-314 22504320-3 2012 In this study, we have hypothesized that enhanced amyloid-beta (Abeta) clearance from the brain across the blood-brain barrier (BBB) of wild-type mice treated with rifampicin or caffeine is caused by both drugs potential to upregulate low-density lipoprotein receptor related protein-1 (LRP1) and/or P-glycoprotein (P-gp) at the BBB. Rifampin 164-174 phosphoglycolate phosphatase Mus musculus 316-320 22504320-4 2012 Expression studies of LRP1 and P-gp in brain endothelial cells and isolated mice brain microvessels following treatment with rifampicin or caffeine demonstrated both drugs as P-gp inducers, and only rifampicin as an LRP1 inducer. Rifampin 125-135 low density lipoprotein receptor-related protein 1 Mus musculus 22-26 22504320-4 2012 Expression studies of LRP1 and P-gp in brain endothelial cells and isolated mice brain microvessels following treatment with rifampicin or caffeine demonstrated both drugs as P-gp inducers, and only rifampicin as an LRP1 inducer. Rifampin 125-135 phosphoglycolate phosphatase Mus musculus 31-35 22467028-8 2012 Quantitative reverse transcription-polymerase chain reaction (RT-PCR) analysis showed that the human-selective ligand, rifampicin induced Cyp3a11 and Carboxylesterase 6 (Ces6) mRNA expression in liver and intestine, whereas the murine-selective ligand, pregnenolone-16-carbonitrile did not. Rifampin 119-129 carboxylesterase 4A Homo sapiens 150-168 22467028-8 2012 Quantitative reverse transcription-polymerase chain reaction (RT-PCR) analysis showed that the human-selective ligand, rifampicin induced Cyp3a11 and Carboxylesterase 6 (Ces6) mRNA expression in liver and intestine, whereas the murine-selective ligand, pregnenolone-16-carbonitrile did not. Rifampin 119-129 carboxylesterase 4A Homo sapiens 170-174 22363234-0 2012 Fucoxanthin attenuates rifampin-induced cytochrome P450 3A4 (CYP3A4) and multiple drug resistance 1 (MDR1) gene expression through pregnane X receptor (PXR)-mediated pathways in human hepatoma HepG2 and colon adenocarcinoma LS174T cells. Rifampin 23-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-59 22363234-0 2012 Fucoxanthin attenuates rifampin-induced cytochrome P450 3A4 (CYP3A4) and multiple drug resistance 1 (MDR1) gene expression through pregnane X receptor (PXR)-mediated pathways in human hepatoma HepG2 and colon adenocarcinoma LS174T cells. Rifampin 23-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 61-67 22363234-0 2012 Fucoxanthin attenuates rifampin-induced cytochrome P450 3A4 (CYP3A4) and multiple drug resistance 1 (MDR1) gene expression through pregnane X receptor (PXR)-mediated pathways in human hepatoma HepG2 and colon adenocarcinoma LS174T cells. Rifampin 23-31 ATP binding cassette subfamily B member 1 Homo sapiens 73-99 22363234-0 2012 Fucoxanthin attenuates rifampin-induced cytochrome P450 3A4 (CYP3A4) and multiple drug resistance 1 (MDR1) gene expression through pregnane X receptor (PXR)-mediated pathways in human hepatoma HepG2 and colon adenocarcinoma LS174T cells. Rifampin 23-31 ATP binding cassette subfamily B member 1 Homo sapiens 101-105 22363234-0 2012 Fucoxanthin attenuates rifampin-induced cytochrome P450 3A4 (CYP3A4) and multiple drug resistance 1 (MDR1) gene expression through pregnane X receptor (PXR)-mediated pathways in human hepatoma HepG2 and colon adenocarcinoma LS174T cells. Rifampin 23-31 nuclear receptor subfamily 1 group I member 2 Homo sapiens 131-150 22363234-0 2012 Fucoxanthin attenuates rifampin-induced cytochrome P450 3A4 (CYP3A4) and multiple drug resistance 1 (MDR1) gene expression through pregnane X receptor (PXR)-mediated pathways in human hepatoma HepG2 and colon adenocarcinoma LS174T cells. Rifampin 23-31 nuclear receptor subfamily 1 group I member 2 Homo sapiens 152-155 21567510-1 2012 This article studied the possible effect of rifampicin (RIF), an inhibitor of organic anion transporting polypeptide (Oatp), on the pharmacokinetics of salvianolic acid B (SAB) in rats. Rifampin 44-54 SH3-domain binding protein 5 Rattus norvegicus 172-175 21567510-1 2012 This article studied the possible effect of rifampicin (RIF), an inhibitor of organic anion transporting polypeptide (Oatp), on the pharmacokinetics of salvianolic acid B (SAB) in rats. Rifampin 56-59 SH3-domain binding protein 5 Rattus norvegicus 172-175 21567510-5 2012 These results suggested that pretreatment with rifampicin prior to SAB administration could decrease significantly the total and bile elimination of SAB and alter its pharmacokinetic profiles. Rifampin 47-57 SH3-domain binding protein 5 Rattus norvegicus 67-70 21567510-5 2012 These results suggested that pretreatment with rifampicin prior to SAB administration could decrease significantly the total and bile elimination of SAB and alter its pharmacokinetic profiles. Rifampin 47-57 SH3-domain binding protein 5 Rattus norvegicus 149-152 21567510-6 2012 The influence of rifampicin on the pharmacokinetics of SAB may be attributed to the inhibition of Oatp-mediated influx. Rifampin 17-27 SH3-domain binding protein 5 Rattus norvegicus 55-58 22970271-10 2012 For rifampicin resistance detection, sensitivity of MTB/RIF was 100% (95% CI 61.0-100.0) (6/6); specificity was 91.0% (95% CI 76.4-96.9) (30/33); the positive predictive value was 66.7% (95% CI 35.4-87.9) (6/9); and the negative predictive value was 100% (95% CI 88.7 -100.0) (30/30). Rifampin 4-14 ras homolog family member F, filopodia associated Homo sapiens 56-59 22629310-3 2012 In this study, using 2-dimensional gel electrophoresis (2-DE) and mass spectrometry (MS), we discovered that 26S protease regulatory subunit 7 (MSS1) was decreased in rifampicin-treated microglia. Rifampin 167-177 proteasome 26S subunit, ATPase 2 Homo sapiens 109-142 22629310-3 2012 In this study, using 2-dimensional gel electrophoresis (2-DE) and mass spectrometry (MS), we discovered that 26S protease regulatory subunit 7 (MSS1) was decreased in rifampicin-treated microglia. Rifampin 167-177 proteasome 26S subunit, ATPase 2 Homo sapiens 144-148 22629310-4 2012 Western blot analysis verified the downregulation of MSS1 expression by rifampicin. Rifampin 72-82 proteasome 26S subunit, ATPase 2 Homo sapiens 53-57 22864345-4 2012 Phenyl analogue menaquinone-3 was as active as the known SXR ligand rifampicin. Rifampin 68-78 nuclear receptor subfamily 1 group I member 2 Homo sapiens 57-60 21930886-5 2011 Rifampin significantly increased the secretion of interleukin 8 (IL-8) in both untreated cells (P < 0.001) and cytokine-treated cells (P < 0.006). Rifampin 0-8 C-X-C motif chemokine ligand 8 Homo sapiens 50-63 21930886-5 2011 Rifampin significantly increased the secretion of interleukin 8 (IL-8) in both untreated cells (P < 0.001) and cytokine-treated cells (P < 0.006). Rifampin 0-8 C-X-C motif chemokine ligand 8 Homo sapiens 65-69 21930886-6 2011 An array screening assay revealed that rifampin stimulated the production of IL-1beta and gamma interferon-induced protein-10 (IP-10) in untreated cells and increased the secretion of RANTES in cytokine-treated cells. Rifampin 39-47 interleukin 1 beta Homo sapiens 77-85 21930886-6 2011 An array screening assay revealed that rifampin stimulated the production of IL-1beta and gamma interferon-induced protein-10 (IP-10) in untreated cells and increased the secretion of RANTES in cytokine-treated cells. Rifampin 39-47 C-X-C motif chemokine ligand 10 Homo sapiens 127-132 21930886-6 2011 An array screening assay revealed that rifampin stimulated the production of IL-1beta and gamma interferon-induced protein-10 (IP-10) in untreated cells and increased the secretion of RANTES in cytokine-treated cells. Rifampin 39-47 C-C motif chemokine ligand 5 Homo sapiens 184-190 22005110-1 2011 Xpert MTB/RIF offers new and important possibilities for the diagnosis of sputum smear-negative tuberculosis (TB) and/or rifampicin (RMP) resistance, and many are encouraging rapid and widespread implementation. Rifampin 123-133 ras homolog family member F, filopodia associated Homo sapiens 12-15 22005110-1 2011 Xpert MTB/RIF offers new and important possibilities for the diagnosis of sputum smear-negative tuberculosis (TB) and/or rifampicin (RMP) resistance, and many are encouraging rapid and widespread implementation. Rifampin 135-138 ras homolog family member F, filopodia associated Homo sapiens 12-15 22115371-10 2011 In addition, rifampicin, a P-gp inducer, resulted in some limited reduction in Salmonella and Klebsiella attachment only. Rifampin 13-23 ATP binding cassette subfamily B member 1 Homo sapiens 27-31 21871880-8 2011 Induction of CYP3A4 mRNA by rifampicin was about 1.5-2.5 fold (clones 2, 4, 6, 7) and it was not significantly different from CYP3A4 mRNA induction in parent HepG2. Rifampin 28-38 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 13-19 21871880-9 2011 The basal expression of CYP3A4 protein was increased in all clones, but rifampicin-induced expression of CYP3A4 protein was in all clones lower than in parent HepG2. Rifampin 72-82 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-111 21764778-2 2011 Given our finding that PXR activation by rifampicin (RIF) represses the estrogen sulfotransferase (SULT1E1) gene in human primary hepatocytes and hepatocellular carcinoma Huh7 cells, here we have investigated the molecular mechanism of this repression. Rifampin 41-51 nuclear receptor subfamily 1 group I member 2 Homo sapiens 23-26 21764778-2 2011 Given our finding that PXR activation by rifampicin (RIF) represses the estrogen sulfotransferase (SULT1E1) gene in human primary hepatocytes and hepatocellular carcinoma Huh7 cells, here we have investigated the molecular mechanism of this repression. Rifampin 41-51 sulfotransferase family 1E member 1 Homo sapiens 99-106 21764778-2 2011 Given our finding that PXR activation by rifampicin (RIF) represses the estrogen sulfotransferase (SULT1E1) gene in human primary hepatocytes and hepatocellular carcinoma Huh7 cells, here we have investigated the molecular mechanism of this repression. Rifampin 53-56 nuclear receptor subfamily 1 group I member 2 Homo sapiens 23-26 21764778-2 2011 Given our finding that PXR activation by rifampicin (RIF) represses the estrogen sulfotransferase (SULT1E1) gene in human primary hepatocytes and hepatocellular carcinoma Huh7 cells, here we have investigated the molecular mechanism of this repression. Rifampin 53-56 sulfotransferase family 1E member 1 Homo sapiens 99-106 22310326-10 2012 When rifampicin-treated primary human hepatocyte cultures were exposed to arsenite concentrations as low as 50 nM, CYP3A mRNA was decreased. Rifampin 5-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 115-120 22205755-7 2012 Formation of 4beta,25(OH)(2)D(3) in primary human hepatocytes was induced by rifampin and inhibited by CYP3A4-specific inhibitors. Rifampin 77-85 iodothyronine deiodinase 3 Homo sapiens 28-32 22205755-8 2012 Short-term treatment of healthy volunteers (n = 6) with rifampin selectively induced CYP3A4-dependent 4beta,25(OH)(2)D(3), but not CYP24A1-dependent 24R,25-dihydroxyvitamin D(3) formation, and altered systemic mineral homeostasis. Rifampin 56-64 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 85-91 22205755-8 2012 Short-term treatment of healthy volunteers (n = 6) with rifampin selectively induced CYP3A4-dependent 4beta,25(OH)(2)D(3), but not CYP24A1-dependent 24R,25-dihydroxyvitamin D(3) formation, and altered systemic mineral homeostasis. Rifampin 56-64 iodothyronine deiodinase 3 Homo sapiens 117-121 22205755-8 2012 Short-term treatment of healthy volunteers (n = 6) with rifampin selectively induced CYP3A4-dependent 4beta,25(OH)(2)D(3), but not CYP24A1-dependent 24R,25-dihydroxyvitamin D(3) formation, and altered systemic mineral homeostasis. Rifampin 56-64 iodothyronine deiodinase 3 Homo sapiens 173-177 22229830-7 2012 CONCLUSIONS: Both rifampicin and plasmapheresis represent important therapeutic options of acute cholestatic attacks in patients with BRIC. Rifampin 18-28 ATPase phospholipid transporting 8B1 Homo sapiens 134-138 22159698-6 2012 In the presence of 2,3,7,8-tetrachlorodibenzo-p-dioxin as an inducer for CYP1A and rifampin as an inducer for CYP3A, As(III) decreased the enzymatic activity of the four P450s more than it decreased their mRNA or protein expression levels. Rifampin 83-91 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 110-115 22236925-1 2012 A study was carried out to establish the relative contribution of human cytochrome P450 (CYP450) enzymes in the metabolism of rifampicin (RMP), rifapentine (RPT) and rifabutin (RFB). Rifampin 126-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 22236925-1 2012 A study was carried out to establish the relative contribution of human cytochrome P450 (CYP450) enzymes in the metabolism of rifampicin (RMP), rifapentine (RPT) and rifabutin (RFB). Rifampin 126-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-95 22236925-1 2012 A study was carried out to establish the relative contribution of human cytochrome P450 (CYP450) enzymes in the metabolism of rifampicin (RMP), rifapentine (RPT) and rifabutin (RFB). Rifampin 138-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 22236925-1 2012 A study was carried out to establish the relative contribution of human cytochrome P450 (CYP450) enzymes in the metabolism of rifampicin (RMP), rifapentine (RPT) and rifabutin (RFB). Rifampin 138-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-95 22291651-6 2012 In this study, we studied the effect of rifampicin, a prototype PXR ligand, on SHP mRNA, and protein expression in three primary human hepatocyte cultures. Rifampin 40-50 nuclear receptor subfamily 0 group B member 2 Homo sapiens 79-82 22185585-0 2012 Rifampicin-independent interactions between the pregnane X receptor ligand binding domain and peptide fragments of coactivator and corepressor proteins. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 48-67 22185585-4 2012 In this work, steady-state total internal reflection fluorescence microscopy (TIRFM) and total internal reflection with fluorescence recovery after photobleaching were used to measure the thermodynamics and kinetics of the interaction between the PXR ligand binding domain and a peptide fragment of the steroid receptor coactivator-1 (SRC-1) in the presence and absence of the established PXR agonist, rifampicin. Rifampin 402-412 nuclear receptor subfamily 1 group I member 2 Homo sapiens 247-250 22185585-7 2012 An equilibrium dissociation constant of ~70 muM was obtained for SMRT in the presence and absence of rifampicin. Rifampin 101-111 nuclear receptor corepressor 2 Homo sapiens 65-69 22333269-0 2012 Lower expression of HNF4alpha and PGC1alpha might impair rifampicin-mediated CYP3A4 induction under conditions where PXR is overexpressed in human fetal liver cells. Rifampin 57-67 hepatocyte nuclear factor 4 alpha Homo sapiens 20-29 22333269-0 2012 Lower expression of HNF4alpha and PGC1alpha might impair rifampicin-mediated CYP3A4 induction under conditions where PXR is overexpressed in human fetal liver cells. Rifampin 57-67 PPARG coactivator 1 alpha Homo sapiens 34-43 22333269-0 2012 Lower expression of HNF4alpha and PGC1alpha might impair rifampicin-mediated CYP3A4 induction under conditions where PXR is overexpressed in human fetal liver cells. Rifampin 57-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 77-83 22333269-0 2012 Lower expression of HNF4alpha and PGC1alpha might impair rifampicin-mediated CYP3A4 induction under conditions where PXR is overexpressed in human fetal liver cells. Rifampin 57-67 nuclear receptor subfamily 1 group I member 2 Homo sapiens 117-120 22333269-2 2012 CYP3A4 was induced by rifampicin (RIF), mifepristone (RU486), clotrimazole (CTZ), and dexamethasone (DEX) in HepG2 cells, while these PXR ligands with the exception of DEX did not induce CYP3A4 mRNA expression in HFL cells. Rifampin 22-32 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 22333269-2 2012 CYP3A4 was induced by rifampicin (RIF), mifepristone (RU486), clotrimazole (CTZ), and dexamethasone (DEX) in HepG2 cells, while these PXR ligands with the exception of DEX did not induce CYP3A4 mRNA expression in HFL cells. Rifampin 34-37 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 22333269-10 2012 These results suggested that lower expression of HNF4alpha and PGC1alpha may impair RIF-mediated CYP3A4 induction under conditions of PXR overexpression in HFL cells. Rifampin 84-87 hepatocyte nuclear factor 4 alpha Homo sapiens 49-58 22333269-10 2012 These results suggested that lower expression of HNF4alpha and PGC1alpha may impair RIF-mediated CYP3A4 induction under conditions of PXR overexpression in HFL cells. Rifampin 84-87 PPARG coactivator 1 alpha Homo sapiens 63-72 22333269-10 2012 These results suggested that lower expression of HNF4alpha and PGC1alpha may impair RIF-mediated CYP3A4 induction under conditions of PXR overexpression in HFL cells. Rifampin 84-87 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 97-103 22333269-10 2012 These results suggested that lower expression of HNF4alpha and PGC1alpha may impair RIF-mediated CYP3A4 induction under conditions of PXR overexpression in HFL cells. Rifampin 84-87 nuclear receptor subfamily 1 group I member 2 Homo sapiens 134-137 22504320-8 2012 In conclusion, our results demonstrated the upregulation of LRP1 and P-gp at the BBB by rifampicin and caffeine enhanced brain Abeta clearance, and this effect could explain, at least in part, the protective effect of rifampicin and caffeine against AD. Rifampin 88-98 low density lipoprotein receptor-related protein 1 Mus musculus 60-64 22504320-8 2012 In conclusion, our results demonstrated the upregulation of LRP1 and P-gp at the BBB by rifampicin and caffeine enhanced brain Abeta clearance, and this effect could explain, at least in part, the protective effect of rifampicin and caffeine against AD. Rifampin 88-98 phosphoglycolate phosphatase Mus musculus 69-73 22504320-8 2012 In conclusion, our results demonstrated the upregulation of LRP1 and P-gp at the BBB by rifampicin and caffeine enhanced brain Abeta clearance, and this effect could explain, at least in part, the protective effect of rifampicin and caffeine against AD. Rifampin 218-228 low density lipoprotein receptor-related protein 1 Mus musculus 60-64 22504320-8 2012 In conclusion, our results demonstrated the upregulation of LRP1 and P-gp at the BBB by rifampicin and caffeine enhanced brain Abeta clearance, and this effect could explain, at least in part, the protective effect of rifampicin and caffeine against AD. Rifampin 218-228 phosphoglycolate phosphatase Mus musculus 69-73 22007302-3 2012 Recently the WHO endorsed Xpert MTB/RIF, a highly sensitive, real-time PCR assay for Mycobacterium tuberculosis that simultaneously detects rifampicin resistance directly from sputum and provides results within 100 minutes. Rifampin 140-150 ras homolog family member F, filopodia associated Homo sapiens 36-39 22363234-3 2012 In this study, we determined whether fucoxanthin could overcome drug resistance through attenuation of rifampin-induced CYP3A4 and MDR1 gene expression by PXR-mediated pathways in HepG2 hepatoma cells. Rifampin 103-111 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 120-126 22363234-3 2012 In this study, we determined whether fucoxanthin could overcome drug resistance through attenuation of rifampin-induced CYP3A4 and MDR1 gene expression by PXR-mediated pathways in HepG2 hepatoma cells. Rifampin 103-111 ATP binding cassette subfamily B member 1 Homo sapiens 131-135 22363234-3 2012 In this study, we determined whether fucoxanthin could overcome drug resistance through attenuation of rifampin-induced CYP3A4 and MDR1 gene expression by PXR-mediated pathways in HepG2 hepatoma cells. Rifampin 103-111 nuclear receptor subfamily 1 group I member 2 Homo sapiens 155-158 22363234-4 2012 We found that fucoxanthin (1-10 muM) significantly attenuated rifampin (20 muM)-induced CYP3A4, MDR1 mRNA and CYP3A4 protein expression at 24 h of incubation. Rifampin 62-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-94 22363234-4 2012 We found that fucoxanthin (1-10 muM) significantly attenuated rifampin (20 muM)-induced CYP3A4, MDR1 mRNA and CYP3A4 protein expression at 24 h of incubation. Rifampin 62-70 ATP binding cassette subfamily B member 1 Homo sapiens 96-100 22363234-4 2012 We found that fucoxanthin (1-10 muM) significantly attenuated rifampin (20 muM)-induced CYP3A4, MDR1 mRNA and CYP3A4 protein expression at 24 h of incubation. Rifampin 62-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 110-116 22363234-8 2012 Thus, fucoxanthin can decrease rifampin-induced CYP3A4 and MDR1 expression through attenuation of PXR-mediated CYP3A4 promoter activation and interaction between PXR and co-activator. Rifampin 31-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-54 22363234-8 2012 Thus, fucoxanthin can decrease rifampin-induced CYP3A4 and MDR1 expression through attenuation of PXR-mediated CYP3A4 promoter activation and interaction between PXR and co-activator. Rifampin 31-39 ATP binding cassette subfamily B member 1 Homo sapiens 59-63 22363234-8 2012 Thus, fucoxanthin can decrease rifampin-induced CYP3A4 and MDR1 expression through attenuation of PXR-mediated CYP3A4 promoter activation and interaction between PXR and co-activator. Rifampin 31-39 nuclear receptor subfamily 1 group I member 2 Homo sapiens 98-101 22363234-8 2012 Thus, fucoxanthin can decrease rifampin-induced CYP3A4 and MDR1 expression through attenuation of PXR-mediated CYP3A4 promoter activation and interaction between PXR and co-activator. Rifampin 31-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 111-117 22363234-8 2012 Thus, fucoxanthin can decrease rifampin-induced CYP3A4 and MDR1 expression through attenuation of PXR-mediated CYP3A4 promoter activation and interaction between PXR and co-activator. Rifampin 31-39 nuclear receptor subfamily 1 group I member 2 Homo sapiens 162-165 21998292-9 2012 Quantification of the transcripts by RNA-Seq and real time quantitative PCR revealed that the CYP3A4 transcript with shorter 3"-UTR was preferentially expressed in developed livers, differentiated hepatocytes, and in rifampicin- and phenobarbital-induced hepatocytes. Rifampin 217-227 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 94-100 23185139-1 2012 BACKGROUND: The Xpert MTB/RIF test enables rapid detection of tuberculosis (TB) and rifampicin resistance. Rifampin 84-94 metallothionein 1J, pseudogene Homo sapiens 22-25 23185139-1 2012 BACKGROUND: The Xpert MTB/RIF test enables rapid detection of tuberculosis (TB) and rifampicin resistance. Rifampin 84-94 ras homolog family member F, filopodia associated Homo sapiens 26-29 23236531-5 2012 This study examined the effect of MDT components on host lipid metabolism in vitro, and the outcome of rifampicin, dapsone and clofazimine treatment on ADRP and HSL expression in THP-1 cells. Rifampin 103-113 perilipin 2 Homo sapiens 152-156 23236531-5 2012 This study examined the effect of MDT components on host lipid metabolism in vitro, and the outcome of rifampicin, dapsone and clofazimine treatment on ADRP and HSL expression in THP-1 cells. Rifampin 103-113 lipase E, hormone sensitive type Homo sapiens 161-164 22629310-8 2012 Taken together, our findings suggested that rifampicin inhibited microglial inflammation by suppressing MSS1 protein production. Rifampin 44-54 proteasome 26S subunit, ATPase 2 Homo sapiens 104-108 21856291-0 2011 Human arylacetamide deacetylase is responsible for deacetylation of rifamycins: rifampicin, rifabutin, and rifapentine. Rifampin 80-90 arylacetamide deacetylase Homo sapiens 6-31 22087865-13 2011 Concomitant administration of bortezomib with strong CYP3A4 inducers such as rifampicin is not recommended, as it may result in a reduction of the clinical effect, whereas concomitant administration of weak CYP3A4 inducers such as dexamethasone does not affect the pharmacological profile of bortezomib. Rifampin 77-87 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 53-59 21930825-3 2011 Statistically significant induction of CYP1A2 (2.1-, 2.9-, and 2.2-fold), CYP2B6 (1.8-, 2.4-, and 4-fold), and CYP2C9 (1.3-, 1.8-, and 2.6-fold) was observed after NFV, RTV, or RIF treatment, respectively (as expected, CYP2D6 was not induced). Rifampin 177-180 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 39-45 21930825-3 2011 Statistically significant induction of CYP1A2 (2.1-, 2.9-, and 2.2-fold), CYP2B6 (1.8-, 2.4-, and 4-fold), and CYP2C9 (1.3-, 1.8-, and 2.6-fold) was observed after NFV, RTV, or RIF treatment, respectively (as expected, CYP2D6 was not induced). Rifampin 177-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 21930825-3 2011 Statistically significant induction of CYP1A2 (2.1-, 2.9-, and 2.2-fold), CYP2B6 (1.8-, 2.4-, and 4-fold), and CYP2C9 (1.3-, 1.8-, and 2.6-fold) was observed after NFV, RTV, or RIF treatment, respectively (as expected, CYP2D6 was not induced). Rifampin 177-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 20623161-4 2011 The patient received anti-tuberculosis therapy including rifampicin, a potent CYP3A4/5 inducer. Rifampin 57-67 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 78-84 20623161-5 2011 After introduction of rifampicin-based treatment, the patient experienced tumour progression, leaving questionable the potential pharmacokinetic interaction between rifampicin and temsirolimus, a substrate for CYP3A4. Rifampin 22-32 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 210-216 20623161-5 2011 After introduction of rifampicin-based treatment, the patient experienced tumour progression, leaving questionable the potential pharmacokinetic interaction between rifampicin and temsirolimus, a substrate for CYP3A4. Rifampin 165-175 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 210-216 22100163-3 2011 In mice, the ABL family tyrosine kinase inhibitor, imatinib (Gleevec), when administered prophylactically or therapeutically, reduced both the number of granulomatous lesions and bacterial load in infected organs and was also effective against a rifampicin-resistant strain. Rifampin 246-256 c-abl oncogene 1, non-receptor tyrosine kinase Mus musculus 13-16 22139693-8 2011 These results showed that Labrasol and Pluronic F68 might inhibit the function of P-gp in the intestine, thereby increasing intestinal absorption and changing the pharmacokinetic parameters of rifampicin. Rifampin 193-203 phosphoglycolate phosphatase Rattus norvegicus 82-86 22000295-3 2011 METHODS: PXR-knockdown experiments using RNAi showed that the cell cycle phase arrest mediated by rifampicin based on activation of PXR. Rifampin 98-108 nuclear receptor subfamily 1 group I member 2 Homo sapiens 9-12 22000295-3 2011 METHODS: PXR-knockdown experiments using RNAi showed that the cell cycle phase arrest mediated by rifampicin based on activation of PXR. Rifampin 98-108 nuclear receptor subfamily 1 group I member 2 Homo sapiens 132-135 22000295-5 2011 Retinoid X receptor alpha (RXRalpha) expression level in cells is another key factor for cell cycle phase arrest mediated by rifampicin. Rifampin 125-135 retinoid X receptor alpha Homo sapiens 0-25 22000295-5 2011 Retinoid X receptor alpha (RXRalpha) expression level in cells is another key factor for cell cycle phase arrest mediated by rifampicin. Rifampin 125-135 retinoid X receptor alpha Homo sapiens 27-35 22000295-6 2011 Both over expression and lacking expression of RXRalpha in cell reduced the cell arrest efficiency mediated by rifampicin. Rifampin 111-121 retinoid X receptor alpha Homo sapiens 47-55 22000295-9 2011 Competitive bind of rifampicin-activated PXR with RXRalpha is one main reason to arrest cell cycle phase through inhibiting combination of RXRalpha with other partners. Rifampin 20-30 nuclear receptor subfamily 1 group I member 2 Homo sapiens 41-44 22000295-9 2011 Competitive bind of rifampicin-activated PXR with RXRalpha is one main reason to arrest cell cycle phase through inhibiting combination of RXRalpha with other partners. Rifampin 20-30 retinoid X receptor alpha Homo sapiens 50-58 22000295-9 2011 Competitive bind of rifampicin-activated PXR with RXRalpha is one main reason to arrest cell cycle phase through inhibiting combination of RXRalpha with other partners. Rifampin 20-30 retinoid X receptor alpha Homo sapiens 139-147 22000295-10 2011 Rifampicin could promote cell growth rate when RXRalpha expressed more excessively than PXR in cells. Rifampin 0-10 retinoid X receptor alpha Homo sapiens 47-55 22000295-10 2011 Rifampicin could promote cell growth rate when RXRalpha expressed more excessively than PXR in cells. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 88-91 21771933-1 2011 Rifampin is a potent inducer of CYP3A4 in vitro and precipitates numerous drug-drug interactions (DDIs) when coadministered with CYP3A4 substrates. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-38 21771933-1 2011 Rifampin is a potent inducer of CYP3A4 in vitro and precipitates numerous drug-drug interactions (DDIs) when coadministered with CYP3A4 substrates. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 129-135 22010421-6 2011 PXR-ligands include a wide variety of pharmaceutical agents, such as antiepileptic drugs, taxol, rifampicin, and human immunodeficiency virus protease inhibitors such as ritonavir and saquinavir. Rifampin 97-107 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-3 22499612-9 2012 (15R)-(11)C-TIC is an in vitro substrate of OATP1B1 and OATP1B3, and clinically relevant concentrations of rifampicin inhibited uptake by OATP1B1 and OATP1B3. Rifampin 107-117 solute carrier organic anion transporter family member 1B1 Homo sapiens 138-145 22499612-9 2012 (15R)-(11)C-TIC is an in vitro substrate of OATP1B1 and OATP1B3, and clinically relevant concentrations of rifampicin inhibited uptake by OATP1B1 and OATP1B3. Rifampin 107-117 solute carrier organic anion transporter family member 1B3 Homo sapiens 150-157 21555117-7 2011 Moreover, rifampicin suppressed LPS-induced nuclear factor-kappa B activation by blocking the degradation of the inhibitor of the nuclear transcription factor NF-kappa B. Rifampin 10-20 nuclear factor kappa B subunit 1 Homo sapiens 159-169 21709081-0 2011 The SLCO1B1 rs4149032 polymorphism is highly prevalent in South Africans and is associated with reduced rifampin concentrations: dosing implications. Rifampin 104-112 solute carrier organic anion transporter family member 1B1 Homo sapiens 4-11 21709081-3 2011 The objectives were to identify genetic polymorphisms of drug transporters and the transcriptional regulators pregnane X receptor (PXR) and constitutive androstane receptor (CAR) with an impact on rifampin pharmacokinetics in South Africans. Rifampin 197-205 nuclear receptor subfamily 1 group I member 2 Homo sapiens 110-129 21709081-3 2011 The objectives were to identify genetic polymorphisms of drug transporters and the transcriptional regulators pregnane X receptor (PXR) and constitutive androstane receptor (CAR) with an impact on rifampin pharmacokinetics in South Africans. Rifampin 197-205 nuclear receptor subfamily 1 group I member 2 Homo sapiens 131-134 21709081-3 2011 The objectives were to identify genetic polymorphisms of drug transporters and the transcriptional regulators pregnane X receptor (PXR) and constitutive androstane receptor (CAR) with an impact on rifampin pharmacokinetics in South Africans. Rifampin 197-205 nuclear receptor subfamily 1 group I member 3 Homo sapiens 140-172 21709081-3 2011 The objectives were to identify genetic polymorphisms of drug transporters and the transcriptional regulators pregnane X receptor (PXR) and constitutive androstane receptor (CAR) with an impact on rifampin pharmacokinetics in South Africans. Rifampin 197-205 nuclear receptor subfamily 1 group I member 3 Homo sapiens 174-177 21709081-11 2011 SLCO1B1 rs4149032 was present in most patients and was associated with substantially reduced rifampin exposure. Rifampin 93-101 solute carrier organic anion transporter family member 1B1 Homo sapiens 0-7 21605667-6 2011 Presence of the known OATP inhibitor rifampicin (25 muM) significantly (p<0.01) decreased the accumulation of estrone-3-sulfate and E(2)-17beta-G to 48% and 70% of the control value, respectively, while no significant effect on digoxin accumulation was observed. Rifampin 37-47 solute carrier organic anion transporter family member 1A2 Homo sapiens 22-26 21651615-6 2011 Similar maximum % inhibition and area under the % inhibition-time effect curve over 24 h for DPP-4 activity were observed when saxagliptin was administered alone or with rifampicin. Rifampin 170-180 dipeptidyl peptidase 4 Homo sapiens 93-98 21561094-4 2011 Phenyl analogue menaquinone-3 was as active as the known SXR ligand rifampicin. Rifampin 68-78 nuclear receptor subfamily 1 group I member 2 Homo sapiens 57-60 21746940-9 2011 Furthermore, 3 (33%) of 10 independently isolated VISA strains established from the heterogeneous subpopulations of Mu3graR were found to possess rpoB mutations with or without an accompanying rifampin-resistance phenotype. Rifampin 193-201 mitochondrial antiviral signaling protein Homo sapiens 50-54 21958145-0 2011 Xpert MTB/RIF assay: development, evaluation and implementation of a new rapid molecular diagnostic for tuberculosis and rifampicin resistance. Rifampin 122-132 ras homolog family member F, filopodia associated Homo sapiens 11-14 21958145-3 2011 The Xpert( ) MTB/RIF assay is a rapid molecular assay that can be used close to the point of care by operators with minimal technical expertise, enabling diagnosis of TB and simultaneous assessment of rifampicin resistance to be completed within 2 h. Moreover, this can be accomplished using unprocessed sputum samples as well as clinical specimens from extrapulmonary sites. Rifampin 201-211 ras homolog family member F, filopodia associated Homo sapiens 17-20 21832258-5 2011 In the 96-well plate induction assay, the CYP3A4 inducers rifampin, phenobarbital, carbamazepine, phenytoin, troglitazone, rosiglitazone, and pioglitazone yielded dose-dependent induction of LIPA metabolism, whereas the CYP1A2 inducers omeprazole and 3-methylcholanthrene did not display any induction in the CYP3A4 activity. Rifampin 58-66 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 21526375-6 2011 The beneficial properties of rifampicin were associated with an increase in DME and export bile acid systems (multidrug resistance-associated protein 4, MRP4, and bile acid export pump to bile duct, BSEP) expression, as well as a reduction in NTCP expression. Rifampin 29-39 ATP binding cassette subfamily C member 4 Homo sapiens 110-151 21526375-6 2011 The beneficial properties of rifampicin were associated with an increase in DME and export bile acid systems (multidrug resistance-associated protein 4, MRP4, and bile acid export pump to bile duct, BSEP) expression, as well as a reduction in NTCP expression. Rifampin 29-39 ATP binding cassette subfamily C member 4 Homo sapiens 153-157 21526375-6 2011 The beneficial properties of rifampicin were associated with an increase in DME and export bile acid systems (multidrug resistance-associated protein 4, MRP4, and bile acid export pump to bile duct, BSEP) expression, as well as a reduction in NTCP expression. Rifampin 29-39 ATP binding cassette subfamily B member 11 Homo sapiens 199-203 21526375-6 2011 The beneficial properties of rifampicin were associated with an increase in DME and export bile acid systems (multidrug resistance-associated protein 4, MRP4, and bile acid export pump to bile duct, BSEP) expression, as well as a reduction in NTCP expression. Rifampin 29-39 solute carrier family 10 member 1 Homo sapiens 243-247 21555507-6 2011 IL-6 suppressed induction of CYP1A2 enzyme activity by omeprazole and CYP3A4 enzyme activity by rifampicin but only at supraphysiological concentrations of IL-6. Rifampin 96-106 interleukin 6 Homo sapiens 0-4 21555507-6 2011 IL-6 suppressed induction of CYP1A2 enzyme activity by omeprazole and CYP3A4 enzyme activity by rifampicin but only at supraphysiological concentrations of IL-6. Rifampin 96-106 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 29-35 21555507-6 2011 IL-6 suppressed induction of CYP1A2 enzyme activity by omeprazole and CYP3A4 enzyme activity by rifampicin but only at supraphysiological concentrations of IL-6. Rifampin 96-106 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 70-76 21540293-3 2011 We screened the expression of 377 human miRNAs in HepG2 cells after 48 h of treatment with 5 muM rifampicin [a pregnane X receptor (PXR) ligand] or vehicle using reverse transcription-polymerase chain reaction-based low-density arrays. Rifampin 97-107 nuclear receptor subfamily 1 group I member 2 Homo sapiens 111-130 21540293-3 2011 We screened the expression of 377 human miRNAs in HepG2 cells after 48 h of treatment with 5 muM rifampicin [a pregnane X receptor (PXR) ligand] or vehicle using reverse transcription-polymerase chain reaction-based low-density arrays. Rifampin 97-107 nuclear receptor subfamily 1 group I member 2 Homo sapiens 132-135 21466844-8 2011 At concentration of 1 ng/ml, trantinterol was about 3.6 +- 3.1% as effective as rifampin (CYP3A4/5 inducer). Rifampin 80-88 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-96 21436404-6 2011 Ketoconazole treatment decreased the CYP3A4 activity by 69%, and rifampicin induced the CYP3A4- and CYP2B6-dependent activity 6-fold, which predicts well the magnitude of changes observed in vivo. Rifampin 65-75 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-94 21436404-6 2011 Ketoconazole treatment decreased the CYP3A4 activity by 69%, and rifampicin induced the CYP3A4- and CYP2B6-dependent activity 6-fold, which predicts well the magnitude of changes observed in vivo. Rifampin 65-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 21441468-1 2011 Rifampin and carbamazepine have been recommended in the U.S. Food and Drug Administration draft drug interaction guidance as CYP3A4 inducers for clinical drug-drug interaction (DDI) studies. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 125-131 21441468-8 2011 Comparison of the maximal CYP3A4 induction potential among the three inducers indicated that rifampin is the most potent inducer and is the best choice for clinical CYP3A4 induction DDI studies. Rifampin 93-101 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 26-32 21441468-8 2011 Comparison of the maximal CYP3A4 induction potential among the three inducers indicated that rifampin is the most potent inducer and is the best choice for clinical CYP3A4 induction DDI studies. Rifampin 93-101 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 165-171 21441468-9 2011 Moreover, a near-maximal CYP3A4 DDI was predicted to result from administration of rifampin for approximately 7 days at 450 to 600 mg q.d. Rifampin 83-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-31 21714930-3 2011 FINDINGS: Oxidative stress-acclimatized bacteria thrive in rifampicin by generating antibiotic-detoxifying nitric oxide (NO), which can be repressed by artesunate or an inhibitor of nitric oxide synthase (NOS). Rifampin 59-69 nitric oxide synthase 2 Homo sapiens 182-203 21555117-6 2011 Our results showed that rifampicin inhibited the LPS-stimulated expression of inducible nitric oxide synthase, cyclooxygenase-2, tumor necrosis factor-alpha, and interleukin-1beta, as well as the production of nitric oxide and prostaglandin E2. Rifampin 24-34 prostaglandin-endoperoxide synthase 2 Homo sapiens 111-127 21555117-6 2011 Our results showed that rifampicin inhibited the LPS-stimulated expression of inducible nitric oxide synthase, cyclooxygenase-2, tumor necrosis factor-alpha, and interleukin-1beta, as well as the production of nitric oxide and prostaglandin E2. Rifampin 24-34 tumor necrosis factor Homo sapiens 129-156 21555117-6 2011 Our results showed that rifampicin inhibited the LPS-stimulated expression of inducible nitric oxide synthase, cyclooxygenase-2, tumor necrosis factor-alpha, and interleukin-1beta, as well as the production of nitric oxide and prostaglandin E2. Rifampin 24-34 interleukin 1 beta Homo sapiens 162-179 21508826-10 2011 CONCLUSIONS: : A novel observation is the large effect of rifampicin on S-norketamine concentrations and indicates that rifampicin induces the elimination of S-ketamine"s metabolite, S-norketamine, probably via induction of the CYP3A4 and/or CYP2B6 enzymes. Rifampin 120-130 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 228-234 21508826-10 2011 CONCLUSIONS: : A novel observation is the large effect of rifampicin on S-norketamine concentrations and indicates that rifampicin induces the elimination of S-ketamine"s metabolite, S-norketamine, probably via induction of the CYP3A4 and/or CYP2B6 enzymes. Rifampin 120-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 242-248 21508826-4 2011 The aim of this study was to investigate the effect of CYP enzyme induction by rifampicin on the pharmacokinetics of S-ketamine and its major metabolite, S-norketamine, in healthy volunteers. Rifampin 79-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 21430235-5 2011 Finally, budesonide, cyclosporine, and rifampin were identified as inhibitors of OATP1B1-, OATP1B3-, and OATP2B1-meditated mesalazine uptake. Rifampin 39-47 solute carrier organic anion transporter family member 1B1 Homo sapiens 81-88 21297316-7 2011 Rifampicin and some HIV protease inhibitors are also OATP1B1 inhibitors. Rifampin 0-10 solute carrier organic anion transporter family member 1B1 Homo sapiens 53-60 21297316-8 2011 Rifampicin is also an inducer of metabolic enzymes, and although its single coadministration produces an increase in the plasma concentration of the affected drugs, multiple coadministrations may result in reductions in the plasma concentrations of OATP1B1 and CYP3A4 bisubstrates. Rifampin 0-10 solute carrier organic anion transporter family member 1B1 Homo sapiens 249-256 21297316-8 2011 Rifampicin is also an inducer of metabolic enzymes, and although its single coadministration produces an increase in the plasma concentration of the affected drugs, multiple coadministrations may result in reductions in the plasma concentrations of OATP1B1 and CYP3A4 bisubstrates. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 261-267 21415249-6 2011 The ratios of IC50 values obtained from the biliary excretion index over the IC(50, QTLI) were 1.34, 1.94, and 1.94, but ratios over IC50 values in CDF uptake by Mrp2-expressing membrane vesicles varied more: 6.69, 3.07, and 2.43 for rifampicin, cyclosporine, and MK-571, respectively. Rifampin 234-244 ATP binding cassette subfamily C member 2 Rattus norvegicus 162-166 21430235-5 2011 Finally, budesonide, cyclosporine, and rifampin were identified as inhibitors of OATP1B1-, OATP1B3-, and OATP2B1-meditated mesalazine uptake. Rifampin 39-47 solute carrier organic anion transporter family member 1B3 Homo sapiens 91-98 21430235-5 2011 Finally, budesonide, cyclosporine, and rifampin were identified as inhibitors of OATP1B1-, OATP1B3-, and OATP2B1-meditated mesalazine uptake. Rifampin 39-47 solute carrier organic anion transporter family member 2B1 Homo sapiens 105-112 20852006-2 2011 A prior investigation of raltegravir coadministered with rifampin demonstrated a decrease in plasma concentrations of raltegravir likely secondary to induction of UGT1A1, the enzyme primarily responsible for the metabolism of raltegravir. Rifampin 57-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 163-169 21419764-5 2011 We show that serum alanine aminotransferase (ALT) and alkaline phosphatase (ALP) were significantly increased in mice treated with isoniazid plus rifampicin. Rifampin 146-156 glutamic pyruvic transaminase, soluble Mus musculus 19-43 21402137-3 2011 MDMA significantly inhibited hPXR-mediated CYP3A4-reporter gene expression induced by the human PXR activator rifampicin (IC50 1.26 +- 0.36 mM) or the therapeutic drugs paroxetine, fluoxetine, clozapine, diazepam and risperidone. Rifampin 110-120 nuclear receptor subfamily 1 group I member 2 Homo sapiens 29-33 21402137-3 2011 MDMA significantly inhibited hPXR-mediated CYP3A4-reporter gene expression induced by the human PXR activator rifampicin (IC50 1.26 +- 0.36 mM) or the therapeutic drugs paroxetine, fluoxetine, clozapine, diazepam and risperidone. Rifampin 110-120 nuclear receptor subfamily 1 group I member 2 Homo sapiens 30-33 21419764-5 2011 We show that serum alanine aminotransferase (ALT) and alkaline phosphatase (ALP) were significantly increased in mice treated with isoniazid plus rifampicin. Rifampin 146-156 glutamic pyruvic transaminase, soluble Mus musculus 45-48 21419764-7 2011 In addition, isoniazid plus rifampicin resulted in hepatic apoptosis, as determined by terminal dUTP nick-end labeling (TUNEL) staining and caspase-3 activation. Rifampin 28-38 caspase 3 Mus musculus 140-149 21507477-10 2011 MTB/RIF test sensitivity for rifampicin resistance was 94 4% (236 of 250) and specificity was 98 3% (796 of 810). Rifampin 29-39 ras homolog family member F, filopodia associated Homo sapiens 4-7 21292004-2 2011 Constitutive and induced expressions of these enzymes are under the control of HNF4alpha and rifampicin activated PXR. Rifampin 93-103 nuclear receptor subfamily 1 group I member 2 Homo sapiens 114-117 21292004-6 2011 NCOA6 enhances the synergistic activation of CYP2C9 and CYP3A4 promoter activity by PXR and HNF4alpha in the presence of rifampicin. Rifampin 121-131 nuclear receptor coactivator 6 Homo sapiens 0-5 21292004-6 2011 NCOA6 enhances the synergistic activation of CYP2C9 and CYP3A4 promoter activity by PXR and HNF4alpha in the presence of rifampicin. Rifampin 121-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 21292004-6 2011 NCOA6 enhances the synergistic activation of CYP2C9 and CYP3A4 promoter activity by PXR and HNF4alpha in the presence of rifampicin. Rifampin 121-131 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 56-62 21210720-11 2011 Drug-induced cytochrome P450 gene expression was increased with rifampicin induction. Rifampin 64-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 21111054-4 2011 METHODS: To evaluate the xenobiotic-mediated induction of hepatocellular gene expression, the prototypical inducers rifampicin (10 muM) and phenobarbital (1 mM) were used for CYP3A4, CITCO (1 muM) and artemisinin (50 muM) were used for CYP2B6, and 3-methylcholanthrene (1 muM) and omeprazole (50 muM) were utilized for induction of CYP1A2. Rifampin 116-126 latexin Homo sapiens 131-134 21111054-4 2011 METHODS: To evaluate the xenobiotic-mediated induction of hepatocellular gene expression, the prototypical inducers rifampicin (10 muM) and phenobarbital (1 mM) were used for CYP3A4, CITCO (1 muM) and artemisinin (50 muM) were used for CYP2B6, and 3-methylcholanthrene (1 muM) and omeprazole (50 muM) were utilized for induction of CYP1A2. Rifampin 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 175-181 21111054-4 2011 METHODS: To evaluate the xenobiotic-mediated induction of hepatocellular gene expression, the prototypical inducers rifampicin (10 muM) and phenobarbital (1 mM) were used for CYP3A4, CITCO (1 muM) and artemisinin (50 muM) were used for CYP2B6, and 3-methylcholanthrene (1 muM) and omeprazole (50 muM) were utilized for induction of CYP1A2. Rifampin 116-126 latexin Homo sapiens 192-195 21111054-4 2011 METHODS: To evaluate the xenobiotic-mediated induction of hepatocellular gene expression, the prototypical inducers rifampicin (10 muM) and phenobarbital (1 mM) were used for CYP3A4, CITCO (1 muM) and artemisinin (50 muM) were used for CYP2B6, and 3-methylcholanthrene (1 muM) and omeprazole (50 muM) were utilized for induction of CYP1A2. Rifampin 116-126 latexin Homo sapiens 192-195 21111054-4 2011 METHODS: To evaluate the xenobiotic-mediated induction of hepatocellular gene expression, the prototypical inducers rifampicin (10 muM) and phenobarbital (1 mM) were used for CYP3A4, CITCO (1 muM) and artemisinin (50 muM) were used for CYP2B6, and 3-methylcholanthrene (1 muM) and omeprazole (50 muM) were utilized for induction of CYP1A2. Rifampin 116-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 236-242 21111054-4 2011 METHODS: To evaluate the xenobiotic-mediated induction of hepatocellular gene expression, the prototypical inducers rifampicin (10 muM) and phenobarbital (1 mM) were used for CYP3A4, CITCO (1 muM) and artemisinin (50 muM) were used for CYP2B6, and 3-methylcholanthrene (1 muM) and omeprazole (50 muM) were utilized for induction of CYP1A2. Rifampin 116-126 latexin Homo sapiens 192-195 21111054-4 2011 METHODS: To evaluate the xenobiotic-mediated induction of hepatocellular gene expression, the prototypical inducers rifampicin (10 muM) and phenobarbital (1 mM) were used for CYP3A4, CITCO (1 muM) and artemisinin (50 muM) were used for CYP2B6, and 3-methylcholanthrene (1 muM) and omeprazole (50 muM) were utilized for induction of CYP1A2. Rifampin 116-126 latexin Homo sapiens 192-195 21111054-4 2011 METHODS: To evaluate the xenobiotic-mediated induction of hepatocellular gene expression, the prototypical inducers rifampicin (10 muM) and phenobarbital (1 mM) were used for CYP3A4, CITCO (1 muM) and artemisinin (50 muM) were used for CYP2B6, and 3-methylcholanthrene (1 muM) and omeprazole (50 muM) were utilized for induction of CYP1A2. Rifampin 116-126 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 332-338 21482342-6 2011 Anti-TNF therapy was stopped even though the patient had received 3 months of prophylactic treatment with rifampicin and isoniazid before starting etanercept. Rifampin 106-116 tumor necrosis factor Homo sapiens 5-8 21456052-0 2011 Inhibitory effects of ketoconazole and rifampin on OAT1 and OATP1B1 transport activities: considerations on drug-drug interactions. Rifampin 39-47 solute carrier family 22 member 6 Homo sapiens 51-55 21456052-0 2011 Inhibitory effects of ketoconazole and rifampin on OAT1 and OATP1B1 transport activities: considerations on drug-drug interactions. Rifampin 39-47 solute carrier organic anion transporter family member 1B1 Homo sapiens 60-67 21456052-2 2011 To determine the underlying mechanisms of DDI, this study was performed to investigate the inhibitory effects of ketoconazole and rifampin on the functions of OAT1 and OATP1B1, and to evaluate the potential of ketoconazole and rifampin for DDI with substrate drugs for these transporters in a clinical setting. Rifampin 130-138 solute carrier family 22 member 6 Homo sapiens 159-163 21456052-2 2011 To determine the underlying mechanisms of DDI, this study was performed to investigate the inhibitory effects of ketoconazole and rifampin on the functions of OAT1 and OATP1B1, and to evaluate the potential of ketoconazole and rifampin for DDI with substrate drugs for these transporters in a clinical setting. Rifampin 130-138 solute carrier organic anion transporter family member 1B1 Homo sapiens 168-175 21456052-3 2011 Ketoconazole inhibited OATP1B1-mediated transport activity, while rifampin inhibited OAT1 and OATP1B1. Rifampin 66-74 solute carrier family 22 member 6 Homo sapiens 85-89 21456052-3 2011 Ketoconazole inhibited OATP1B1-mediated transport activity, while rifampin inhibited OAT1 and OATP1B1. Rifampin 66-74 solute carrier organic anion transporter family member 1B1 Homo sapiens 94-101 21395646-0 2011 In silico prediction of efavirenz and rifampicin drug-drug interaction considering weight and CYP2B6 phenotype. Rifampin 38-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 21395646-4 2011 The model was used to simulate the effects of rifampicin on efavirenz pharmacokinetics in 400 virtual patients, taking into account bodyweight and CYP2B6 phenotype. Rifampin 46-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-153 19780977-0 2011 In vitro interactions between amphotericin B and other antifungal agents and rifampin against Fusarium spp. Rifampin 77-85 histocompatibility minor 13 Homo sapiens 103-106 21205914-0 2011 Pharmacokinetic-pharmacodynamic modeling of rifampicin-mediated Cyp3a11 induction in steroid and xenobiotic X receptor humanized mice. Rifampin 44-54 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 64-71 21205914-0 2011 Pharmacokinetic-pharmacodynamic modeling of rifampicin-mediated Cyp3a11 induction in steroid and xenobiotic X receptor humanized mice. Rifampin 44-54 nuclear receptor subfamily 1, group I, member 2 Mus musculus 85-118 21205914-1 2011 The purpose of this study was to develop a mechanistic pharmacokinetic-pharmacodynamic (PK-PD) model to describe the effects of rifampicin on hepatic Cyp3a11 RNA, enzymatic activity, and triazolam pharmacokinetics. Rifampin 128-138 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 150-157 21205914-2 2011 Rifampicin was administered to steroid and xenobiotic X receptor (SXR) humanized mice at 10 mg/kg p.o. Rifampin 0-10 nuclear receptor subfamily 1, group I, member 2 Mus musculus 31-64 21205914-2 2011 Rifampicin was administered to steroid and xenobiotic X receptor (SXR) humanized mice at 10 mg/kg p.o. Rifampin 0-10 nuclear receptor subfamily 1, group I, member 2 Mus musculus 66-69 21205914-6 2011 Elevations in Cyp3a11 RNA expression were observed 24 h after the first dose of rifampicin, reaching a maximum (~10 times baseline) after the third dose and were sustained until day 4 and began declining 48 h after the last rifampicin dose. Rifampin 80-90 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 14-21 21205914-6 2011 Elevations in Cyp3a11 RNA expression were observed 24 h after the first dose of rifampicin, reaching a maximum (~10 times baseline) after the third dose and were sustained until day 4 and began declining 48 h after the last rifampicin dose. Rifampin 224-234 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 14-21 21205914-9 2011 The final PK-PD model incorporated rifampicin liver concentration as the driving force for the time-delayed Cyp3a11 induction governed by in vitro potency estimates, which in turn regulated the turnover of enzyme activity. Rifampin 35-45 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 108-115 21205914-10 2011 The PK-PD model was able to recapitulate the delayed induction of Cyp3a11 mRNA and enzymatic activity by rifampicin. Rifampin 105-115 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 66-73 21270793-0 2011 Rifampin enhances the glucose-lowering effect of metformin and increases OCT1 mRNA levels in healthy participants. Rifampin 0-8 solute carrier family 22 member 1 Homo sapiens 73-77 20876786-0 2011 Effects of pregnane X receptor (NR1I2) and CYP2B6 genetic polymorphisms on the induction of bupropion hydroxylation by rifampin. Rifampin 119-127 nuclear receptor subfamily 1 group I member 2 Homo sapiens 11-30 21150552-0 2011 Paradoxically elevated efavirenz concentrations in HIV/tuberculosis-coinfected patients with CYP2B6 516TT genotype on rifampin-containing antituberculous therapy. Rifampin 118-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-99 21150552-3 2011 Two-way analysis of variance revealed a significant interaction between CYP2B6 516G T polymorphism and rifampin-containing therapy, suggesting that efavirenz dose adjustment may need to be individualized on the basis of the patient"s genotype. Rifampin 103-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 21191377-1 2011 We studied the time course for the reversal of rifampin"s effect on the pharmacokinetics of oral midazolam (a cytochrome P450 (CYP) 3A4 substrate) and digoxin (a P-glycoprotein (P-gp) substrate). Rifampin 47-55 DNA segment, 3a4 Mus musculus 132-135 21191377-1 2011 We studied the time course for the reversal of rifampin"s effect on the pharmacokinetics of oral midazolam (a cytochrome P450 (CYP) 3A4 substrate) and digoxin (a P-glycoprotein (P-gp) substrate). Rifampin 47-55 phosphoglycolate phosphatase Mus musculus 162-176 21191377-1 2011 We studied the time course for the reversal of rifampin"s effect on the pharmacokinetics of oral midazolam (a cytochrome P450 (CYP) 3A4 substrate) and digoxin (a P-glycoprotein (P-gp) substrate). Rifampin 47-55 phosphoglycolate phosphatase Mus musculus 178-182 21191377-7 2011 However, rifampin did inhibit the P-gp-mediated transport of digoxin with a half-maximal inhibitory concentration (IC(50)) below anticipated gut lumen concentrations, suggesting that rifampin inhibits digoxin efflux from the enterocyte to the intestinal lumen. Rifampin 9-17 phosphoglycolate phosphatase Mus musculus 34-38 21191377-7 2011 However, rifampin did inhibit the P-gp-mediated transport of digoxin with a half-maximal inhibitory concentration (IC(50)) below anticipated gut lumen concentrations, suggesting that rifampin inhibits digoxin efflux from the enterocyte to the intestinal lumen. Rifampin 183-191 phosphoglycolate phosphatase Mus musculus 34-38 21372390-7 2011 Rifampicin and penicillin were transported by both OATP1B1 and OATP1B3. Rifampin 0-10 solute carrier organic anion transporter family member 1B1 Homo sapiens 51-58 21372390-7 2011 Rifampicin and penicillin were transported by both OATP1B1 and OATP1B3. Rifampin 0-10 solute carrier organic anion transporter family member 1B3 Homo sapiens 63-70 21410294-16 2011 CONCLUSIONS: Exposure to vandetanib, as assessed by AUC(504) in healthy subjects, was reduced by around 40% when a single dose was given in combination with the potent CYP3A4 inducer rifampicin. Rifampin 183-193 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 168-174 20876786-0 2011 Effects of pregnane X receptor (NR1I2) and CYP2B6 genetic polymorphisms on the induction of bupropion hydroxylation by rifampin. Rifampin 119-127 nuclear receptor subfamily 1 group I member 2 Homo sapiens 32-37 20876786-0 2011 Effects of pregnane X receptor (NR1I2) and CYP2B6 genetic polymorphisms on the induction of bupropion hydroxylation by rifampin. Rifampin 119-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 20876786-1 2011 We investigated genetic polymorphisms in the pregnane X receptor (NR1I2) in Korean individuals (n = 83) and the effects of NR1I2 genotypes on rifampin-mediated induction of bupropion hydroxylation. Rifampin 142-150 nuclear receptor subfamily 1 group I member 2 Homo sapiens 123-128 20876786-3 2011 The area under the time-concentration curve (AUC) ratio of hydroxybupropion to bupropion in CYP2B6*6 carriers was significantly lower than that in CYP2B6*6 noncarriers in both the basal and rifampin-induced states (p = 0.012). Rifampin 190-198 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 20876786-7 2011 In conclusion, it is suggested the NR1I2 TGT genotype decreases the bupropion hydroxylation induced by treatment with rifampin, particularly in CYP2B6*6 carriers. Rifampin 118-126 nuclear receptor subfamily 1 group I member 2 Homo sapiens 35-40 20876786-7 2011 In conclusion, it is suggested the NR1I2 TGT genotype decreases the bupropion hydroxylation induced by treatment with rifampin, particularly in CYP2B6*6 carriers. Rifampin 118-126 queuine tRNA-ribosyltransferase catalytic subunit 1 Homo sapiens 41-44 20876786-7 2011 In conclusion, it is suggested the NR1I2 TGT genotype decreases the bupropion hydroxylation induced by treatment with rifampin, particularly in CYP2B6*6 carriers. Rifampin 118-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-150 21047828-1 2011 BACKGROUND: rifampicin lowers nevirapine plasma concentrations by inducing cytochrome P450. Rifampin 12-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 20702754-4 2011 In the induction study, administration of rifampin 600 mg once daily for 8 days significantly increased urinary 6beta-hydroxycortisol/ cortisol ratio, from a preinduction baseline of 5.8 +- 2.7 to 18.0 +- 10.2 after 8 days of rifampin treatment, confirming an inductive effect on CYP3A4. Rifampin 42-50 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 280-286 21436605-5 2011 Furthermore, CYP3A4 protein levels were significantly increased by the addition of rifampicin and dexamethasone to the culture media, indicating that the induction potential was maintained. Rifampin 83-93 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 13-19 21733412-10 2011 The rank order of these drugs potencies to up-regulate P-gp activity was as following: hyperforin >>> dexamethasone ~ beta-estradiol > caffeine > rifampicin ~ pentylenetetrazole > verapamil. Rifampin 161-171 ATP binding cassette subfamily B member 1 Homo sapiens 55-59 21789183-8 2011 Our findings establish an endogenous cellular function for a mono-ADP-ribosyltransferase apart from its role in mediating Rifampin resistance. Rifampin 122-130 ADP-ribosyltransferase 3 (inactive) Homo sapiens 61-88 20583967-0 2010 A multi-endpoint evaluation of cytochrome P450 1A2, 2B6 and 3A4 induction response in human hepatocyte cultures after treatment with beta-naphthoflavone, phenobarbital and rifampicin. Rifampin 172-182 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 31-50 20583967-3 2010 The concentration-dependent response and time-course for the induction of CYP1A2, CYP2B6 and CYP3A4 by inducing agents beta-naphthoflavone, phenobarbital and rifampicin, respectively were examined in two or more donors using multiple end-points (mRNA, enzyme activity and Western blot analysis). Rifampin 158-168 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 74-80 20583967-3 2010 The concentration-dependent response and time-course for the induction of CYP1A2, CYP2B6 and CYP3A4 by inducing agents beta-naphthoflavone, phenobarbital and rifampicin, respectively were examined in two or more donors using multiple end-points (mRNA, enzyme activity and Western blot analysis). Rifampin 158-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 20583967-3 2010 The concentration-dependent response and time-course for the induction of CYP1A2, CYP2B6 and CYP3A4 by inducing agents beta-naphthoflavone, phenobarbital and rifampicin, respectively were examined in two or more donors using multiple end-points (mRNA, enzyme activity and Western blot analysis). Rifampin 158-168 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-99 20962047-6 2010 In human prostate cancer cells, treatment with the PXR agonist rifampicin (RIF) inhibited androgen-dependent proliferation of LAPC-4 cells but had little effect on the growth of the androgen-independent isogenic LA99 cells. Rifampin 63-73 nuclear receptor subfamily 1 group I member 2 Homo sapiens 51-54 20837660-4 2010 The efficacy of MG132 was increased by inclusion of ketoconazole in the hepatocyte incubations and decreased by prior treatment of the cultures with the CYP3A inducers phenobarbital or rifampicin. Rifampin 185-195 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 153-158 20861156-5 2010 In DPX2 cells, the PXR-driven 5.5- and 6.5-fold induction responses to omeprazole and 10 muM rifampicin were attenuated to 4- and 3.5-fold, respectively. Rifampin 93-103 nuclear receptor subfamily 1 group I member 2 Homo sapiens 19-22 20962047-6 2010 In human prostate cancer cells, treatment with the PXR agonist rifampicin (RIF) inhibited androgen-dependent proliferation of LAPC-4 cells but had little effect on the growth of the androgen-independent isogenic LA99 cells. Rifampin 75-78 nuclear receptor subfamily 1 group I member 2 Homo sapiens 51-54 20832446-7 2010 A number of genes were regulated in a cell type and species-specific manner after incubation with the prototypical PXR agonists rifampicin or dexamethasone, respectively. Rifampin 128-138 nuclear receptor subfamily 1 group I member 2 Homo sapiens 115-118 21131707-5 2010 The patient was treated with 3 months therapy of Ethambutol and Rifampicin with good clinical response.The clinical presentation of the case is discussed with a review of the literature about current guidelines for prophylaxis and other preventive strategy for infection among patients receiving TNF antagonists. Rifampin 64-74 tumor necrosis factor Homo sapiens 296-299 20887388-9 2010 Steroid and xenobiotic receptor (SXR), which was recently identified as a receptor of VK2, was expressed on myeloid progenitors, and the SXR agonist rifampicin (RIF) also upregulated CD11b and CD14 expressions on myeloid progenitors. Rifampin 149-159 nuclear receptor subfamily 1 group I member 2 Homo sapiens 33-36 20887388-9 2010 Steroid and xenobiotic receptor (SXR), which was recently identified as a receptor of VK2, was expressed on myeloid progenitors, and the SXR agonist rifampicin (RIF) also upregulated CD11b and CD14 expressions on myeloid progenitors. Rifampin 149-159 nuclear receptor subfamily 1 group I member 2 Homo sapiens 137-140 20887388-9 2010 Steroid and xenobiotic receptor (SXR), which was recently identified as a receptor of VK2, was expressed on myeloid progenitors, and the SXR agonist rifampicin (RIF) also upregulated CD11b and CD14 expressions on myeloid progenitors. Rifampin 149-159 integrin subunit alpha M Homo sapiens 183-188 20887388-9 2010 Steroid and xenobiotic receptor (SXR), which was recently identified as a receptor of VK2, was expressed on myeloid progenitors, and the SXR agonist rifampicin (RIF) also upregulated CD11b and CD14 expressions on myeloid progenitors. Rifampin 149-159 CD14 molecule Homo sapiens 193-197 20887388-9 2010 Steroid and xenobiotic receptor (SXR), which was recently identified as a receptor of VK2, was expressed on myeloid progenitors, and the SXR agonist rifampicin (RIF) also upregulated CD11b and CD14 expressions on myeloid progenitors. Rifampin 161-164 nuclear receptor subfamily 1 group I member 2 Homo sapiens 33-36 20887388-9 2010 Steroid and xenobiotic receptor (SXR), which was recently identified as a receptor of VK2, was expressed on myeloid progenitors, and the SXR agonist rifampicin (RIF) also upregulated CD11b and CD14 expressions on myeloid progenitors. Rifampin 161-164 nuclear receptor subfamily 1 group I member 2 Homo sapiens 137-140 20887388-9 2010 Steroid and xenobiotic receptor (SXR), which was recently identified as a receptor of VK2, was expressed on myeloid progenitors, and the SXR agonist rifampicin (RIF) also upregulated CD11b and CD14 expressions on myeloid progenitors. Rifampin 161-164 integrin subunit alpha M Homo sapiens 183-188 20887388-9 2010 Steroid and xenobiotic receptor (SXR), which was recently identified as a receptor of VK2, was expressed on myeloid progenitors, and the SXR agonist rifampicin (RIF) also upregulated CD11b and CD14 expressions on myeloid progenitors. Rifampin 161-164 CD14 molecule Homo sapiens 193-197 20871212-3 2010 When human estrogen receptor (hERalpha) and SXR were exogenously expressed, treatment with either rifampicin or corticosterone promoted significantly the SXR-mediated transactivation of LXRE reporter gene in HepG2. Rifampin 98-108 estrogen receptor 1 Homo sapiens 11-39 20871212-3 2010 When human estrogen receptor (hERalpha) and SXR were exogenously expressed, treatment with either rifampicin or corticosterone promoted significantly the SXR-mediated transactivation of LXRE reporter gene in HepG2. Rifampin 98-108 nuclear receptor subfamily 1 group I member 2 Homo sapiens 44-47 20871212-3 2010 When human estrogen receptor (hERalpha) and SXR were exogenously expressed, treatment with either rifampicin or corticosterone promoted significantly the SXR-mediated transactivation of LXRE reporter gene in HepG2. Rifampin 98-108 nuclear receptor subfamily 1 group I member 2 Homo sapiens 154-157 20699409-4 2010 Concentration-dependent CYP3A induction of rifampicin was reproducible with the EC(50) values of 0.36 +- 0.28 muM from four batches of human hepatocytes using the 96-well plate with TL Matrigel. Rifampin 43-53 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-29 20699409-6 2010 Cotreatment of avasimibe or efavirenz with 10 muM rifampicin was found to reduce CYP3A activities induced by rifampicin at a lower rate than treatment with rifampicin alone, whereas treatment with phenobarbital and carbamazepine had no effect. Rifampin 50-60 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-86 20699409-6 2010 Cotreatment of avasimibe or efavirenz with 10 muM rifampicin was found to reduce CYP3A activities induced by rifampicin at a lower rate than treatment with rifampicin alone, whereas treatment with phenobarbital and carbamazepine had no effect. Rifampin 109-119 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-86 20699409-6 2010 Cotreatment of avasimibe or efavirenz with 10 muM rifampicin was found to reduce CYP3A activities induced by rifampicin at a lower rate than treatment with rifampicin alone, whereas treatment with phenobarbital and carbamazepine had no effect. Rifampin 109-119 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-86 20699409-8 2010 The cotreatment assay of test compound with rifampicin allows us to exclude the false-negative results caused by the cytotoxicity and/or the mechanism-based inactivation, when the drug candidate"s ability for CYP3A induction is evaluating the enzyme activity. Rifampin 44-54 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 209-214 20624464-4 2010 In this study we aimed to further characterize this cell line by focusing on the time dependence of P-gp expression, localization and function in the presence of rifampicin, a P-gp inducer. Rifampin 162-172 ATP binding cassette subfamily B member 1 Homo sapiens 100-104 20719936-1 2010 Long-term treatment of patients with the macrolide antibiotic and prototypical activator of pregnane X receptor (PXR) rifampicin (Rif) inhibits the inflammatory response in liver. Rifampin 118-128 nuclear receptor subfamily 1 group I member 2 Homo sapiens 92-111 20719936-1 2010 Long-term treatment of patients with the macrolide antibiotic and prototypical activator of pregnane X receptor (PXR) rifampicin (Rif) inhibits the inflammatory response in liver. Rifampin 118-128 nuclear receptor subfamily 1 group I member 2 Homo sapiens 113-116 20719936-1 2010 Long-term treatment of patients with the macrolide antibiotic and prototypical activator of pregnane X receptor (PXR) rifampicin (Rif) inhibits the inflammatory response in liver. Rifampin 130-133 nuclear receptor subfamily 1 group I member 2 Homo sapiens 92-111 20719936-1 2010 Long-term treatment of patients with the macrolide antibiotic and prototypical activator of pregnane X receptor (PXR) rifampicin (Rif) inhibits the inflammatory response in liver. Rifampin 130-133 nuclear receptor subfamily 1 group I member 2 Homo sapiens 113-116 20624464-4 2010 In this study we aimed to further characterize this cell line by focusing on the time dependence of P-gp expression, localization and function in the presence of rifampicin, a P-gp inducer. Rifampin 162-172 ATP binding cassette subfamily B member 1 Homo sapiens 176-180 20624464-5 2010 P-gp protein expression was increased in a time and dose dependent manner following exposure of cells to rifampicin (5-50 muM). Rifampin 105-115 ATP binding cassette subfamily B member 1 Homo sapiens 0-4 20624464-5 2010 P-gp protein expression was increased in a time and dose dependent manner following exposure of cells to rifampicin (5-50 muM). Rifampin 105-115 latexin Homo sapiens 122-125 20624464-6 2010 The induction of P-gp by rifampicin and its inhibition by ketoconazole (an inhibitor of PXR mediated P-gp induction) confirms the suitability of these cells for PXR induction studies. Rifampin 25-35 ATP binding cassette subfamily B member 1 Homo sapiens 17-21 20624464-6 2010 The induction of P-gp by rifampicin and its inhibition by ketoconazole (an inhibitor of PXR mediated P-gp induction) confirms the suitability of these cells for PXR induction studies. Rifampin 25-35 nuclear receptor subfamily 1 group I member 2 Homo sapiens 161-164 20945562-17 2004 The current treatment regime for drug-sensitive TB involves the use of rifampicin (RIF), INH, pyrazinamide (PZA), and ethambutol or streptomycin for two months, followed by four months of continued dosing with INH and RIF (11, 12). Rifampin 71-81 ras homolog family member F, filopodia associated Homo sapiens 83-86 20939914-0 2010 Protective effect of rifampicin and clindamycin impregnated devices against Staphylococcus spp. Rifampin 21-31 histocompatibility minor 13 Homo sapiens 91-94 20599501-7 2010 Rifampicin, a model CYP3A4 inducer, significantly induced CYP3A4 mRNA in both types of cells. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-26 20599501-7 2010 Rifampicin, a model CYP3A4 inducer, significantly induced CYP3A4 mRNA in both types of cells. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 58-64 20599501-8 2010 The level of CYP3A4 protein in human hepatocytes was increased by Orlistat after 48h, while rifampicin strongly induced CYP3A4 protein level. Rifampin 92-102 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 120-126 20540932-10 2010 Molecules known to interact with OATPs, including cyclosporin A, rifampicin, and glibenclamide, each demonstrated concentration-dependent inhibition of 8-FcA transport by OATP1B1 and OATP1B3. Rifampin 65-75 solute carrier organic anion transporter family member 1B1 Homo sapiens 171-178 20540932-10 2010 Molecules known to interact with OATPs, including cyclosporin A, rifampicin, and glibenclamide, each demonstrated concentration-dependent inhibition of 8-FcA transport by OATP1B1 and OATP1B3. Rifampin 65-75 solute carrier organic anion transporter family member 1B3 Homo sapiens 183-190 20660695-3 2010 In this study, we evaluated rifampin pharmacokinetics in relation to tuberculosis, geographic region, race, and single nucleotide polymorphisms of the human transporter genes SLCO1B1, SLCO1B3, and MDR1. Rifampin 28-36 ATP binding cassette subfamily B member 1 Homo sapiens 197-201 20660695-3 2010 In this study, we evaluated rifampin pharmacokinetics in relation to tuberculosis, geographic region, race, and single nucleotide polymorphisms of the human transporter genes SLCO1B1, SLCO1B3, and MDR1. Rifampin 28-36 solute carrier organic anion transporter family member 1B1 Homo sapiens 175-182 20660695-6 2010 However, in multivariable analyses, the rifampin AUC(0-24) was significantly affected by rifampin dosage (in mg/kg of body weight), polymorphisms in the SLCO1B1 gene, and the presence of tuberculosis by geographic region. Rifampin 40-48 solute carrier organic anion transporter family member 1B1 Homo sapiens 153-160 20660695-11 2010 Lower rifampin exposure was associated with the polymorphism of the SLCO1B1 c.463C>A gene. Rifampin 6-14 solute carrier organic anion transporter family member 1B1 Homo sapiens 68-75 20827821-0 2004 (11)C-Labeled rifampicin Rifampicin (RIF) (or rifampin) is a rifamycin derivative with a clinically effective group of 4-methyl-1-piperazinaminyl. Rifampin 14-24 ras homolog family member F, filopodia associated Homo sapiens 37-40 20624854-5 2010 Cell-based promoter reporter assays demonstrated that CYP2B6 luciferase activity was synergistically enhanced in the presence of both -82T C mutation and rifampicin (RIF)-activated PXR. Rifampin 154-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 20624854-5 2010 Cell-based promoter reporter assays demonstrated that CYP2B6 luciferase activity was synergistically enhanced in the presence of both -82T C mutation and rifampicin (RIF)-activated PXR. Rifampin 154-164 nuclear receptor subfamily 1 group I member 2 Homo sapiens 181-184 20624854-5 2010 Cell-based promoter reporter assays demonstrated that CYP2B6 luciferase activity was synergistically enhanced in the presence of both -82T C mutation and rifampicin (RIF)-activated PXR. Rifampin 166-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 20624854-5 2010 Cell-based promoter reporter assays demonstrated that CYP2B6 luciferase activity was synergistically enhanced in the presence of both -82T C mutation and rifampicin (RIF)-activated PXR. Rifampin 166-169 nuclear receptor subfamily 1 group I member 2 Homo sapiens 181-184 20703222-1 2010 Based on in vitro rat and human hepatocyte uptake studies showing inhibition of warfarin uptake in the presence of the nonspecific organic anion-transporting polypeptide (OATP) inhibitor rifampin, a clinical study was conducted in 10 healthy volunteers to examine the in vivo relevance of OATP hepatic uptake on the pharmacokinetics of warfarin. Rifampin 187-195 solute carrier organic anion transporter family member 1A2 Homo sapiens 131-169 20703222-1 2010 Based on in vitro rat and human hepatocyte uptake studies showing inhibition of warfarin uptake in the presence of the nonspecific organic anion-transporting polypeptide (OATP) inhibitor rifampin, a clinical study was conducted in 10 healthy volunteers to examine the in vivo relevance of OATP hepatic uptake on the pharmacokinetics of warfarin. Rifampin 187-195 solute carrier organic anion transporter family member 1A2 Homo sapiens 171-175 20703222-1 2010 Based on in vitro rat and human hepatocyte uptake studies showing inhibition of warfarin uptake in the presence of the nonspecific organic anion-transporting polypeptide (OATP) inhibitor rifampin, a clinical study was conducted in 10 healthy volunteers to examine the in vivo relevance of OATP hepatic uptake on the pharmacokinetics of warfarin. Rifampin 187-195 solute carrier organic anion transporter family member 1A2 Homo sapiens 289-293 20827821-0 2004 (11)C-Labeled rifampicin Rifampicin (RIF) (or rifampin) is a rifamycin derivative with a clinically effective group of 4-methyl-1-piperazinaminyl. Rifampin 25-35 ras homolog family member F, filopodia associated Homo sapiens 37-40 20827821-0 2004 (11)C-Labeled rifampicin Rifampicin (RIF) (or rifampin) is a rifamycin derivative with a clinically effective group of 4-methyl-1-piperazinaminyl. Rifampin 46-54 ras homolog family member F, filopodia associated Homo sapiens 37-40 20818746-7 2010 Upon activation of PXR by rifampicin, the messenger RNA levels of CYP3A4, CYP3A5, and CYP3A7 increased 49- to 213-fold versus HepG2 cells. Rifampin 26-36 nuclear receptor subfamily 1 group I member 2 Homo sapiens 19-22 20690781-12 2010 Therefore, exposure to saxagliptin and its primary metabolite may be significantly modified when saxagliptin is coadministered with specific strong inhibitors (ketoconazole, diltiazem) or inducers (rifampicin) of CYP3A4/5 isoforms. Rifampin 198-208 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 213-219 20504912-6 2010 CPT treatment resulted in delayed metabolism of nifedipine in human hepatocytes treated with rifampicin, suggesting a potential prevention of drug-drug interactions between CYP3A4 inducers and CYP3A4-metabolized drugs. Rifampin 93-103 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 173-179 20504912-6 2010 CPT treatment resulted in delayed metabolism of nifedipine in human hepatocytes treated with rifampicin, suggesting a potential prevention of drug-drug interactions between CYP3A4 inducers and CYP3A4-metabolized drugs. Rifampin 93-103 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 193-199 20818746-7 2010 Upon activation of PXR by rifampicin, the messenger RNA levels of CYP3A4, CYP3A5, and CYP3A7 increased 49- to 213-fold versus HepG2 cells. Rifampin 26-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 66-72 20818746-7 2010 Upon activation of PXR by rifampicin, the messenger RNA levels of CYP3A4, CYP3A5, and CYP3A7 increased 49- to 213-fold versus HepG2 cells. Rifampin 26-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 74-80 20818746-7 2010 Upon activation of PXR by rifampicin, the messenger RNA levels of CYP3A4, CYP3A5, and CYP3A7 increased 49- to 213-fold versus HepG2 cells. Rifampin 26-36 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 86-92 20818746-8 2010 According to reporter gene assays with different inducers, the highest increase in CYP3A4 promoter activity (131-fold) was observed upon induction with rifampicin. Rifampin 152-162 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-89 20642445-5 2010 ABT also modulated CYP-inducing actions of CITCO and rifampin, the known inducers of CYP2B6 and CYP3A4. Rifampin 53-61 peptidylprolyl isomerase G Homo sapiens 19-22 20488228-0 2010 Lack of P-glycoprotein induction by rifampicin and phenobarbital in human lymphocytes. Rifampin 36-46 ATP binding cassette subfamily B member 1 Homo sapiens 8-22 20488228-3 2010 Rifampicin and phenobarbital have been shown to induce P-gp in hepatic and intestinal cells through the activation of the nuclear receptors PXR and CAR. Rifampin 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 55-59 20488228-3 2010 Rifampicin and phenobarbital have been shown to induce P-gp in hepatic and intestinal cells through the activation of the nuclear receptors PXR and CAR. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 140-143 20488228-3 2010 Rifampicin and phenobarbital have been shown to induce P-gp in hepatic and intestinal cells through the activation of the nuclear receptors PXR and CAR. Rifampin 0-10 CXADR pseudogene 1 Homo sapiens 148-151 20488228-12 2010 Therefore, P-gp induction by rifampicin and phenobarbital may play a negligible role in drug interactions occurring within lymphocytes. Rifampin 29-39 ATP binding cassette subfamily B member 1 Homo sapiens 11-15 20642445-5 2010 ABT also modulated CYP-inducing actions of CITCO and rifampin, the known inducers of CYP2B6 and CYP3A4. Rifampin 53-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 20642445-5 2010 ABT also modulated CYP-inducing actions of CITCO and rifampin, the known inducers of CYP2B6 and CYP3A4. Rifampin 53-61 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 96-102 20492465-0 2010 Potentiation of clopidogrel active metabolite formation by rifampicin leads to greater P2Y12 receptor blockade and inhibition of platelet aggregation after clopidogrel. Rifampin 59-69 purinergic receptor P2Y12 Homo sapiens 87-92 20584229-4 2010 Regarding drug-drug interactions, dabigatran requires caution when used in combination with strong inhibitors or inducers of P-glycoprotein, such as amiodarone or rifampicin. Rifampin 163-173 ATP binding cassette subfamily B member 1 Homo sapiens 125-139 20233841-6 2010 The CYP3A4 activity in confluent cells was also inducible by rifampicin. Rifampin 61-71 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 4-10 20014428-2 2010 Extracellular sodium depletion or coincubation with the OATP/Oatp inhibitors rifampicin and digoxin revealed that about 35% of active CGamF accumulation was mediated by Ntcp/NTCP in rat and human hepatocytes, while the contribution of this sodium-dependent transporter reached 50-60% in dog and pig hepatocytes. Rifampin 77-87 solute carrier organic anion transporter family member 1A2 Homo sapiens 56-60 20014428-2 2010 Extracellular sodium depletion or coincubation with the OATP/Oatp inhibitors rifampicin and digoxin revealed that about 35% of active CGamF accumulation was mediated by Ntcp/NTCP in rat and human hepatocytes, while the contribution of this sodium-dependent transporter reached 50-60% in dog and pig hepatocytes. Rifampin 77-87 solute carrier organic anion transporter family member 1A2 Homo sapiens 61-65 20014428-2 2010 Extracellular sodium depletion or coincubation with the OATP/Oatp inhibitors rifampicin and digoxin revealed that about 35% of active CGamF accumulation was mediated by Ntcp/NTCP in rat and human hepatocytes, while the contribution of this sodium-dependent transporter reached 50-60% in dog and pig hepatocytes. Rifampin 77-87 solute carrier family 10 member 1 Rattus norvegicus 169-173 20803980-7 2010 Anti-NTM chemotherapy consisting of rifampicin, ethambutol and clarithromycin was started in addition to anticancer chemotherapy, without severe side effects. Rifampin 36-46 neurotrimin Homo sapiens 5-8 20406852-5 2010 In comparison, the IC(50) values for rifampicin were 11.9 (OATP1B1), 1.4 (OATP1B3), and 80.5 muM (OATP2B1), respectively. Rifampin 37-47 solute carrier organic anion transporter family member 1B1 Homo sapiens 59-66 20406852-5 2010 In comparison, the IC(50) values for rifampicin were 11.9 (OATP1B1), 1.4 (OATP1B3), and 80.5 muM (OATP2B1), respectively. Rifampin 37-47 solute carrier organic anion transporter family member 1B3 Homo sapiens 74-81 20406852-5 2010 In comparison, the IC(50) values for rifampicin were 11.9 (OATP1B1), 1.4 (OATP1B3), and 80.5 muM (OATP2B1), respectively. Rifampin 37-47 latexin Homo sapiens 93-96 20406852-5 2010 In comparison, the IC(50) values for rifampicin were 11.9 (OATP1B1), 1.4 (OATP1B3), and 80.5 muM (OATP2B1), respectively. Rifampin 37-47 solute carrier organic anion transporter family member 2B1 Homo sapiens 98-105 20179914-1 2010 PURPOSE: This study aimed to investigate the effect of rifampicin, an inducer of CYP3A4 and P-glycoprotein, on the pharmacokinetics and pharmacodynamics of aliskiren, a renin inhibitor used in the treatment of hypertension. Rifampin 55-65 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-87 20179914-1 2010 PURPOSE: This study aimed to investigate the effect of rifampicin, an inducer of CYP3A4 and P-glycoprotein, on the pharmacokinetics and pharmacodynamics of aliskiren, a renin inhibitor used in the treatment of hypertension. Rifampin 55-65 ATP binding cassette subfamily B member 1 Homo sapiens 92-106 20179914-1 2010 PURPOSE: This study aimed to investigate the effect of rifampicin, an inducer of CYP3A4 and P-glycoprotein, on the pharmacokinetics and pharmacodynamics of aliskiren, a renin inhibitor used in the treatment of hypertension. Rifampin 55-65 renin Homo sapiens 169-174 20179914-6 2010 Plasma renin activity 24 h after aliskiren intake was 61% higher during the rifampicin phase than during the placebo phase (P = 0.008). Rifampin 76-86 renin Homo sapiens 7-12 20179914-7 2010 CONCLUSIONS: Rifampicin considerably reduces the plasma concentrations and the renin-inhibiting effect of aliskiren by decreasing its oral bioavailability. Rifampin 13-23 renin Homo sapiens 79-84 20222094-9 2010 Most genes showing changes greater than 3-fold were known to be rifampicin-responsive, and this suggested a PXR-dependent mode of regulation. Rifampin 64-74 nuclear receptor subfamily 1 group I member 2 Homo sapiens 108-111 22009927-14 2010 CONCLUSION: Rifampicin may decrease the risk of steroid-induced ONFH by enhancing P-gp activity, thus preventing steroid-induced BMSC adipogenesis. Rifampin 12-22 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 82-86 20447852-0 2010 Pregnancy during TNFalpha antagonist therapy: beware the rifampin-oral contraceptive interaction. Rifampin 57-65 tumor necrosis factor Homo sapiens 17-25 20086032-7 2010 Mutational studies showed that the HNF4alpha sites are needed for up-regulation of the CYP2C8 and CYP2C9 promoters by rifampicin. Rifampin 118-128 hepatocyte nuclear factor 4 alpha Homo sapiens 35-44 20086032-7 2010 Mutational studies showed that the HNF4alpha sites are needed for up-regulation of the CYP2C8 and CYP2C9 promoters by rifampicin. Rifampin 118-128 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 87-93 20086032-7 2010 Mutational studies showed that the HNF4alpha sites are needed for up-regulation of the CYP2C8 and CYP2C9 promoters by rifampicin. Rifampin 118-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 20086032-8 2010 Furthermore, silencing of HNF4alpha abolished transactivation of the CYP2C8 and CYP2C9 promoters by rifampicin. Rifampin 100-110 hepatocyte nuclear factor 4 alpha Homo sapiens 26-35 20086032-8 2010 Furthermore, silencing of HNF4alpha abolished transactivation of the CYP2C8 and CYP2C9 promoters by rifampicin. Rifampin 100-110 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 69-75 20086032-8 2010 Furthermore, silencing of HNF4alpha abolished transactivation of the CYP2C8 and CYP2C9 promoters by rifampicin. Rifampin 100-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 20086032-10 2010 Quantitative polymerase chain reaction analysis demonstrated that silencing HNF4alpha reduced the constitutive expression of CYP2C8 (53%), CYP2C9 (55%), and CYP2C19 (43%) mRNAs and significantly decreased the magnitude of the rifampicin-mediated induction of CYP2C8 (6.6- versus 2.7-fold), CYP2C9 (3- versus 1.5-fold), and CYP2C19 (1.8- versus 1.1-fold). Rifampin 226-236 hepatocyte nuclear factor 4 alpha Homo sapiens 76-85 20086032-10 2010 Quantitative polymerase chain reaction analysis demonstrated that silencing HNF4alpha reduced the constitutive expression of CYP2C8 (53%), CYP2C9 (55%), and CYP2C19 (43%) mRNAs and significantly decreased the magnitude of the rifampicin-mediated induction of CYP2C8 (6.6- versus 2.7-fold), CYP2C9 (3- versus 1.5-fold), and CYP2C19 (1.8- versus 1.1-fold). Rifampin 226-236 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 125-131 20086032-11 2010 These results provide clear evidence that HNF4alpha contributes to the constitutive expression of the human CYP2C genes and is also important for up-regulation by the PXR agonist rifampicin. Rifampin 179-189 hepatocyte nuclear factor 4 alpha Homo sapiens 42-51 20086032-11 2010 These results provide clear evidence that HNF4alpha contributes to the constitutive expression of the human CYP2C genes and is also important for up-regulation by the PXR agonist rifampicin. Rifampin 179-189 nuclear receptor subfamily 1 group I member 2 Homo sapiens 167-170 20446914-4 2010 Rifampicin, CITCO and T1317 significantly enhanced interactions for human PXR, human CAR and human LXR, respectively. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 74-77 20179914-0 2010 Rifampicin reduces the plasma concentrations and the renin-inhibiting effect of aliskiren. Rifampin 0-10 renin Homo sapiens 53-58 20446914-4 2010 Rifampicin, CITCO and T1317 significantly enhanced interactions for human PXR, human CAR and human LXR, respectively. Rifampin 0-10 nuclear receptor subfamily 1 group I member 3 Homo sapiens 85-88 19844775-5 2010 After pig treatment with rifampicin, the mRNA expression of CYPs and transporters from brain regions did not appear to change, except CYP3A22 and 3A29 in cortex and hippocampus, CYP2B22 in meninges. Rifampin 25-35 cytochrome P450 family 2 subfamily B member 6B Sus scrofa 178-185 19844775-1 2010 The in vivo effect of rifampicin, a potent ligand of PXR, on gene expression of CYP2B22, 3A22, 3A29, 3A46, CAR, PXR and MDR1, MRP1, MRP2, LRP transporters in liver and cortex, cerebellum, midbrain, hippocampus, meninges and brain capillaries of pig was investigated. Rifampin 22-32 nuclear receptor subfamily 1 group I member 2 Sus scrofa 53-56 19844775-1 2010 The in vivo effect of rifampicin, a potent ligand of PXR, on gene expression of CYP2B22, 3A22, 3A29, 3A46, CAR, PXR and MDR1, MRP1, MRP2, LRP transporters in liver and cortex, cerebellum, midbrain, hippocampus, meninges and brain capillaries of pig was investigated. Rifampin 22-32 cytochrome P450 family 2 subfamily B member 6B Sus scrofa 80-87 19844775-1 2010 The in vivo effect of rifampicin, a potent ligand of PXR, on gene expression of CYP2B22, 3A22, 3A29, 3A46, CAR, PXR and MDR1, MRP1, MRP2, LRP transporters in liver and cortex, cerebellum, midbrain, hippocampus, meninges and brain capillaries of pig was investigated. Rifampin 22-32 CXADR Ig-like cell adhesion molecule Sus scrofa 107-110 19844775-1 2010 The in vivo effect of rifampicin, a potent ligand of PXR, on gene expression of CYP2B22, 3A22, 3A29, 3A46, CAR, PXR and MDR1, MRP1, MRP2, LRP transporters in liver and cortex, cerebellum, midbrain, hippocampus, meninges and brain capillaries of pig was investigated. Rifampin 22-32 nuclear receptor subfamily 1 group I member 2 Sus scrofa 112-115 19844775-1 2010 The in vivo effect of rifampicin, a potent ligand of PXR, on gene expression of CYP2B22, 3A22, 3A29, 3A46, CAR, PXR and MDR1, MRP1, MRP2, LRP transporters in liver and cortex, cerebellum, midbrain, hippocampus, meninges and brain capillaries of pig was investigated. Rifampin 22-32 ATP-binding cassette, sub-family B (MDR/TAP), member 1 Sus scrofa 120-124 19844775-1 2010 The in vivo effect of rifampicin, a potent ligand of PXR, on gene expression of CYP2B22, 3A22, 3A29, 3A46, CAR, PXR and MDR1, MRP1, MRP2, LRP transporters in liver and cortex, cerebellum, midbrain, hippocampus, meninges and brain capillaries of pig was investigated. Rifampin 22-32 LOW QUALITY PROTEIN: multidrug resistance-associated protein 6 Sus scrofa 126-130 19844775-1 2010 The in vivo effect of rifampicin, a potent ligand of PXR, on gene expression of CYP2B22, 3A22, 3A29, 3A46, CAR, PXR and MDR1, MRP1, MRP2, LRP transporters in liver and cortex, cerebellum, midbrain, hippocampus, meninges and brain capillaries of pig was investigated. Rifampin 22-32 ATP binding cassette subfamily C member 2 Sus scrofa 132-136 19844775-5 2010 After pig treatment with rifampicin, the mRNA expression of CYPs and transporters from brain regions did not appear to change, except CYP3A22 and 3A29 in cortex and hippocampus, CYP2B22 in meninges. Rifampin 25-35 cytochrome P450 family 3 subfamily A member 22 Sus scrofa 134-141 20079722-10 2010 Indeed, over-expression of PXR in HepG2 cells enhanced the steatogenic effect of hyperforin and rifampicin. Rifampin 96-106 nuclear receptor subfamily 1 group I member 2 Homo sapiens 27-30 20338069-11 2010 This is the first report to demonstrate the effects of CYP2B6 G516T polymorphisms on plasma efavirenz and nevirapine concentrations when co-administered with rifampicin in HIV/TB co-infected Thai adults. Rifampin 158-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 20086032-0 2010 Hepatocyte nuclear factor 4{alpha} regulates rifampicin-mediated induction of CYP2C genes in primary cultures of human hepatocytes. Rifampin 45-55 hepatocyte nuclear factor 4 alpha Homo sapiens 0-33 20086032-0 2010 Hepatocyte nuclear factor 4{alpha} regulates rifampicin-mediated induction of CYP2C genes in primary cultures of human hepatocytes. Rifampin 45-55 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 78-83 20086032-3 2010 In the present study, we used primary cultures of human hepatocytes to investigate the role of HNF4alpha in the pregnane X receptor (PXR)/rifampicin-mediated up-regulation of CYP2C8, CYP2C9, and CYP2C19 gene expression. Rifampin 138-148 hepatocyte nuclear factor 4 alpha Homo sapiens 95-104 20086032-3 2010 In the present study, we used primary cultures of human hepatocytes to investigate the role of HNF4alpha in the pregnane X receptor (PXR)/rifampicin-mediated up-regulation of CYP2C8, CYP2C9, and CYP2C19 gene expression. Rifampin 138-148 nuclear receptor subfamily 1 group I member 2 Homo sapiens 112-131 20086032-3 2010 In the present study, we used primary cultures of human hepatocytes to investigate the role of HNF4alpha in the pregnane X receptor (PXR)/rifampicin-mediated up-regulation of CYP2C8, CYP2C9, and CYP2C19 gene expression. Rifampin 138-148 nuclear receptor subfamily 1 group I member 2 Homo sapiens 133-136 20086032-3 2010 In the present study, we used primary cultures of human hepatocytes to investigate the role of HNF4alpha in the pregnane X receptor (PXR)/rifampicin-mediated up-regulation of CYP2C8, CYP2C9, and CYP2C19 gene expression. Rifampin 138-148 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 175-181 20086032-3 2010 In the present study, we used primary cultures of human hepatocytes to investigate the role of HNF4alpha in the pregnane X receptor (PXR)/rifampicin-mediated up-regulation of CYP2C8, CYP2C9, and CYP2C19 gene expression. Rifampin 138-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 183-189 20171174-6 2010 TRalpha1, TRbeta1, COUP-TFI, and COUP-TFII further demonstrated dose-dependent repression of PXR-mediated CYP3A4 enhancer/promoter reporter activity in transient transfection in the presence and absence of the PXR inducer rifampin, while VDR showed this effect only in the absence of treatment. Rifampin 222-230 nuclear receptor subfamily 2 group F member 1 Homo sapiens 19-27 20171174-6 2010 TRalpha1, TRbeta1, COUP-TFI, and COUP-TFII further demonstrated dose-dependent repression of PXR-mediated CYP3A4 enhancer/promoter reporter activity in transient transfection in the presence and absence of the PXR inducer rifampin, while VDR showed this effect only in the absence of treatment. Rifampin 222-230 nuclear receptor subfamily 2 group F member 2 Homo sapiens 33-42 20171174-6 2010 TRalpha1, TRbeta1, COUP-TFI, and COUP-TFII further demonstrated dose-dependent repression of PXR-mediated CYP3A4 enhancer/promoter reporter activity in transient transfection in the presence and absence of the PXR inducer rifampin, while VDR showed this effect only in the absence of treatment. Rifampin 222-230 nuclear receptor subfamily 1 group I member 2 Homo sapiens 93-96 20171174-6 2010 TRalpha1, TRbeta1, COUP-TFI, and COUP-TFII further demonstrated dose-dependent repression of PXR-mediated CYP3A4 enhancer/promoter reporter activity in transient transfection in the presence and absence of the PXR inducer rifampin, while VDR showed this effect only in the absence of treatment. Rifampin 222-230 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 106-112 20171174-6 2010 TRalpha1, TRbeta1, COUP-TFI, and COUP-TFII further demonstrated dose-dependent repression of PXR-mediated CYP3A4 enhancer/promoter reporter activity in transient transfection in the presence and absence of the PXR inducer rifampin, while VDR showed this effect only in the absence of treatment. Rifampin 222-230 nuclear receptor subfamily 1 group I member 2 Homo sapiens 210-213 20171174-6 2010 TRalpha1, TRbeta1, COUP-TFI, and COUP-TFII further demonstrated dose-dependent repression of PXR-mediated CYP3A4 enhancer/promoter reporter activity in transient transfection in the presence and absence of the PXR inducer rifampin, while VDR showed this effect only in the absence of treatment. Rifampin 222-230 vitamin D receptor Homo sapiens 238-241 19879123-0 2010 Serial interferon-gamma release assays after rifampicin prophylaxis in a tuberculosis outbreak. Rifampin 45-55 interferon gamma Homo sapiens 7-23 19854261-5 2010 On the other hand, the model inducers, i.e. 2,3,7,8- tetrachlorodibenzo-p-dioxin (TCDD) and rifampicin, significantly increased the levels of CYP1A2 and CYP3A4 mRNAs in all cultures. Rifampin 92-102 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 142-148 19854261-5 2010 On the other hand, the model inducers, i.e. 2,3,7,8- tetrachlorodibenzo-p-dioxin (TCDD) and rifampicin, significantly increased the levels of CYP1A2 and CYP3A4 mRNAs in all cultures. Rifampin 92-102 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 153-159 20035846-5 2010 CYP3A was induced by EMD in human HepG2 cells exceeding the level induced by rifampicin, but was not induced in rat H4IIE cells. Rifampin 77-87 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-5 20460758-9 2010 However, rifampicin the nuclear receptor steroid and xenobiotic receptor (SXR) ligand had no effect of IL-beta, suggesting that IL-1beta is involved in VK(2) dependent gamma-calboxylation but not SXR-activation. Rifampin 9-19 nuclear receptor subfamily 1 group I member 2 Homo sapiens 74-77 20019244-2 2010 In the present study, we carefully analyzed mRNA expression and activity of the major P450s and their responsiveness to three prototypical inducers, phenobarbital, rifampicin, and omeprazole, in differentiated HepaRG cell cultures over a 4-week period after low and high seeding. Rifampin 164-174 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-91 20019244-7 2010 Thus, CYP1A2, CYP2B6, and CYP3A4 were found to accurately respond to their respective prototypical inducers, i.e., omeprazole, phenobarbital, and rifampicin. Rifampin 146-156 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 6-12 20019244-7 2010 Thus, CYP1A2, CYP2B6, and CYP3A4 were found to accurately respond to their respective prototypical inducers, i.e., omeprazole, phenobarbital, and rifampicin. Rifampin 146-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 20019244-7 2010 Thus, CYP1A2, CYP2B6, and CYP3A4 were found to accurately respond to their respective prototypical inducers, i.e., omeprazole, phenobarbital, and rifampicin. Rifampin 146-156 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 26-32 20034695-4 2010 Current evidence indicates that ursodeoxycholic acid (UDCA) can be effective in selected patients with PFIC3 (ABCB4 deficiency), while rifampicin reduces pruritus in patients with PFIC1 (ATP8B1 deficiency) and PFIC2 (ABCB11 deficiency), and might abort cholestatic episodes in BRIC (mild ATP8B1 or ABCB11 deficiency). Rifampin 135-145 ATPase phospholipid transporting 8B1 Homo sapiens 187-193 20012942-2 2010 Since they interact with ABCB1, ABCC2, and organic anion transporting proteins (OATPs), which are localized to lung tissue, pulmonary concentration may be influenced by rifampicin (RIF), an inducer and modulator of efflux and uptake transporters. Rifampin 169-179 ATP binding cassette subfamily B member 1 Canis lupus familiaris 25-30 20012942-2 2010 Since they interact with ABCB1, ABCC2, and organic anion transporting proteins (OATPs), which are localized to lung tissue, pulmonary concentration may be influenced by rifampicin (RIF), an inducer and modulator of efflux and uptake transporters. Rifampin 169-179 ATP binding cassette subfamily C member 2 Canis lupus familiaris 32-37 20012942-2 2010 Since they interact with ABCB1, ABCC2, and organic anion transporting proteins (OATPs), which are localized to lung tissue, pulmonary concentration may be influenced by rifampicin (RIF), an inducer and modulator of efflux and uptake transporters. Rifampin 181-184 ATP binding cassette subfamily B member 1 Canis lupus familiaris 25-30 20012942-2 2010 Since they interact with ABCB1, ABCC2, and organic anion transporting proteins (OATPs), which are localized to lung tissue, pulmonary concentration may be influenced by rifampicin (RIF), an inducer and modulator of efflux and uptake transporters. Rifampin 181-184 ATP binding cassette subfamily C member 2 Canis lupus familiaris 32-37 19712670-7 2010 In contrast, eNR3A4 is involved only in the basal transactivation in intestinal cells and in the PXR-mediated rifampicin-induced transactivation of CYP3A4 in LS174T intestinal cells. Rifampin 110-120 nuclear receptor subfamily 1 group I member 2 Homo sapiens 97-100 19712670-7 2010 In contrast, eNR3A4 is involved only in the basal transactivation in intestinal cells and in the PXR-mediated rifampicin-induced transactivation of CYP3A4 in LS174T intestinal cells. Rifampin 110-120 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 148-154 19898817-10 2010 Exposure of fathead minnow (FHM) cells with the CYP3A inducer rifampicin leads to dose-dependent increase in putative CYP3A enzyme activity. Rifampin 62-72 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-53 19898817-10 2010 Exposure of fathead minnow (FHM) cells with the CYP3A inducer rifampicin leads to dose-dependent increase in putative CYP3A enzyme activity. Rifampin 62-72 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 118-123 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. Rifampin 142-150 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. Rifampin 142-150 ATP binding cassette subfamily B member 1 Homo sapiens 94-108 21265468-5 2010 Rifampicin and minocycline at 10-100 microg/ml significantly inhibited interleukin-2 production from mitogen- or superantigen-stimulated peripheral-blood mononuclear cells (p < 0.025). Rifampin 0-10 interleukin 2 Homo sapiens 71-84 20923245-3 2010 OBJECTIVE: To assess the effects of rifampicin (rifampin), a potent inducer of CYP3A4 and inhibitor of organic anion transporter polypeptides (OATPs), on the steady-state pharmacokinetics, safety and tolerability of ambrisentan. Rifampin 36-46 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 79-85 20016271-2 2010 The in vitro effects of rifampicin to rapidly downregulate angiogenesis and mitogenesis-related genes in cultured endothelial cells are reminiscent of endostatin, one of the most well-studied angiogenesis inhibitors. Rifampin 24-34 collagen type XVIII alpha 1 chain Homo sapiens 151-161 20923245-15 2010 No dose adjustment of ambrisentan should be required when it is co-administered with rifampicin, a strong inducer of CYP3A4 activity and inhibitor of OATPs. Rifampin 85-95 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 117-123 20923245-3 2010 OBJECTIVE: To assess the effects of rifampicin (rifampin), a potent inducer of CYP3A4 and inhibitor of organic anion transporter polypeptides (OATPs), on the steady-state pharmacokinetics, safety and tolerability of ambrisentan. Rifampin 48-56 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 79-85 19833845-7 2010 Second, two known human CYP3A4 inducers, rifampicin and hyperforin, were tested in monkey and human primary hepatocytes for induction of CYP3A enzymes. Rifampin 41-51 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-30 19833845-7 2010 Second, two known human CYP3A4 inducers, rifampicin and hyperforin, were tested in monkey and human primary hepatocytes for induction of CYP3A enzymes. Rifampin 41-51 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-29 20212331-0 2010 Discovery of rifampicin as a new anti-glycating compound by matrix-assisted laser desorption/ionization mass spectrometry-based insulin glycation assay. Rifampin 13-23 insulin Homo sapiens 128-135 19915937-4 2010 Following exposure of PICM-19H cells to either 3-methylcholanthrene, rifampicin or phenobarbital, the induced activities of cytochrome P450 (CYP450) isozymes CYP-1A, -2, and-3A were assessed. Rifampin 69-79 cytochrome P450 family 2 subfamily D member 6 Sus scrofa 124-139 20201776-4 2010 We determined the induction of CYP3A4 by EFV, RTV, ATV, EFV+RTV, EFV+ATV, EFV+RTV+ATV and rifampicin (RIF) employing primary human hepatocytes from 3 donors. Rifampin 90-100 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 20201776-4 2010 We determined the induction of CYP3A4 by EFV, RTV, ATV, EFV+RTV, EFV+ATV, EFV+RTV+ATV and rifampicin (RIF) employing primary human hepatocytes from 3 donors. Rifampin 102-105 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 20201776-8 2010 hPXR activation data in LS180 cells were consistent with the induction of transcripts and the estimated EC(50) values were 0.87 microM, 0.44 microM and 3.7 microM for RIF, RTV and EFV, respectively. Rifampin 167-170 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-4 19915937-4 2010 Following exposure of PICM-19H cells to either 3-methylcholanthrene, rifampicin or phenobarbital, the induced activities of cytochrome P450 (CYP450) isozymes CYP-1A, -2, and-3A were assessed. Rifampin 69-79 cytochrome P450 family 2 subfamily D member 6 Sus scrofa 141-147 19915937-7 2010 Rifampicin induction of CYP450 isozyme activities was confirmed by conversion of testosterone to 6beta-OH-, 2alpha-OH- and 2beta-OH-testosterone, as determined by mass spectrometry. Rifampin 0-10 cytochrome P450 family 2 subfamily D member 6 Sus scrofa 24-30 19836313-8 2009 In addition, the ability of MUTYH and the variants to suppress mutations and complement for the absence of MutY in Escherichia coli was assessed using rifampicin resistance assays. Rifampin 151-161 mutY DNA glycosylase Homo sapiens 28-33 20392161-1 2010 Intranasal administration of lipid microspheres (LM) containing rifampicin (LM-RFP) exhibited the bacteriostatic effect against Mycobacterium tuberculosis H37Rv. Rifampin 64-74 tripartite motif-containing 27 Mus musculus 79-82 20392161-2 2010 The efficacies of intranasal LM-RFP in lung were markedly higher in immunodeficient BALB/c nude mice (p < 0.001), but not different in immunocompetent BALB/c mice (p = 1.000) compared to the results of oral rifampicin groups. Rifampin 210-220 tripartite motif-containing 27 Mus musculus 32-35 20392161-4 2010 These results suggested that intranasal LM-RFP could improve the anti-tuberculosis activity in immunodeficient host and minimize drug interaction between rifampicin and nevirapine. Rifampin 154-164 tripartite motif-containing 27 Mus musculus 43-46 20305954-10 2010 Results showed rifampicin alone to raise (p < 0.05) liver function enzymes (Aspartate amino transferase [AST], Serum alanine amino transferase [ALT] and Total Bilirubin) when compared with controls. Rifampin 15-25 solute carrier family 17 member 5 Homo sapiens 79-106 20305954-10 2010 Results showed rifampicin alone to raise (p < 0.05) liver function enzymes (Aspartate amino transferase [AST], Serum alanine amino transferase [ALT] and Total Bilirubin) when compared with controls. Rifampin 15-25 solute carrier family 17 member 5 Homo sapiens 108-111 20305954-10 2010 Results showed rifampicin alone to raise (p < 0.05) liver function enzymes (Aspartate amino transferase [AST], Serum alanine amino transferase [ALT] and Total Bilirubin) when compared with controls. Rifampin 15-25 glutamic--pyruvic transaminase Homo sapiens 120-145 20305954-10 2010 Results showed rifampicin alone to raise (p < 0.05) liver function enzymes (Aspartate amino transferase [AST], Serum alanine amino transferase [ALT] and Total Bilirubin) when compared with controls. Rifampin 15-25 glutamic--pyruvic transaminase Homo sapiens 147-150 20110038-3 2009 OBJECTIVE: The aim of this study was to investigate the effects of a potent inhibitor (ketoconazole) and inducer (rifampicin) of the CYP3A4 isozyme on the pharmacokinetics of mirodenafil to meet the regulatory requirements for the marketing of mirodenafil in Korea. Rifampin 114-124 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 133-139 19451401-7 2009 LIPA metabolism in human hepatocytes was found to be induced by the treatment of human hepatocytes with the prototypical CYP3A4 inducers rifampin, carbamazepine, omeprazole, phenobarbital, and phenytoin but not by the CYP1A2 inducer 3-methylcholanthrene. Rifampin 137-145 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 121-127 20054495-0 2009 Rifampicin Inhibits the LPS-induced Expression of Toll-like Receptor 2 via the Suppression of NF-kappaB DNA-binding Activity in RAW 264.7 Cells. Rifampin 0-10 toll-like receptor 2 Mus musculus 50-70 20054495-3 2009 Therefore, we hypothesized that rifampicin may influence the TLR2 expression in LPS-activated RAW 264.7 cells. Rifampin 32-42 toll-like receptor 2 Mus musculus 61-65 20054495-4 2009 In this study, we determined that rifampicin suppresses LPS-induced TLR2 mRNA expression. Rifampin 34-44 toll-like receptor 2 Mus musculus 68-72 20054495-8 2009 However, rifampicin slightly inhibited the nuclear translocation of NF-kappaB p65. Rifampin 9-19 v-rel reticuloendotheliosis viral oncogene homolog A (avian) Mus musculus 78-81 20054495-9 2009 In addition, rifampicin increased physical interaction between pregnane X receptor, a receptor for rifampicin, and NF-kappaB p65, suggesting pregnane X receptor interferes with NF-kappaB binding to DNA. Rifampin 13-23 nuclear receptor subfamily 1, group I, member 2 Mus musculus 63-82 20054495-9 2009 In addition, rifampicin increased physical interaction between pregnane X receptor, a receptor for rifampicin, and NF-kappaB p65, suggesting pregnane X receptor interferes with NF-kappaB binding to DNA. Rifampin 13-23 v-rel reticuloendotheliosis viral oncogene homolog A (avian) Mus musculus 125-128 20054495-9 2009 In addition, rifampicin increased physical interaction between pregnane X receptor, a receptor for rifampicin, and NF-kappaB p65, suggesting pregnane X receptor interferes with NF-kappaB binding to DNA. Rifampin 13-23 nuclear receptor subfamily 1, group I, member 2 Mus musculus 141-160 20054495-9 2009 In addition, rifampicin increased physical interaction between pregnane X receptor, a receptor for rifampicin, and NF-kappaB p65, suggesting pregnane X receptor interferes with NF-kappaB binding to DNA. Rifampin 99-109 nuclear receptor subfamily 1, group I, member 2 Mus musculus 63-82 20054495-9 2009 In addition, rifampicin increased physical interaction between pregnane X receptor, a receptor for rifampicin, and NF-kappaB p65, suggesting pregnane X receptor interferes with NF-kappaB binding to DNA. Rifampin 99-109 v-rel reticuloendotheliosis viral oncogene homolog A (avian) Mus musculus 125-128 20054495-9 2009 In addition, rifampicin increased physical interaction between pregnane X receptor, a receptor for rifampicin, and NF-kappaB p65, suggesting pregnane X receptor interferes with NF-kappaB binding to DNA. Rifampin 99-109 nuclear receptor subfamily 1, group I, member 2 Mus musculus 141-160 20054495-10 2009 Taken together, our results demonstrate that rifampicin inhibits LPS-induced TLR2 expression, at least in part, via the suppression of NF-kappaB DNA-binding activity in RAW 264.7 cells. Rifampin 45-55 toll-like receptor 2 Mus musculus 77-81 20054495-11 2009 Thus, the present results suggest that the rifampicin-mediated inhibition of TLR2 via the suppression of NF-kappaB DNA-binding activity may be a novel mechanism of the immune-suppressive effects of rifampicin. Rifampin 43-53 toll-like receptor 2 Mus musculus 77-81 20054495-11 2009 Thus, the present results suggest that the rifampicin-mediated inhibition of TLR2 via the suppression of NF-kappaB DNA-binding activity may be a novel mechanism of the immune-suppressive effects of rifampicin. Rifampin 198-208 toll-like receptor 2 Mus musculus 77-81 19686824-6 2009 The levels of CYP1A1, CYP1A2, CY2C9 and CYP3A4 mRNAs were not altered by Zolpidem, whereas model inducers dioxin and rifampicin significantly induced CYP1A and CYP2/3 gene expression, respectively. Rifampin 117-127 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 14-20 19686824-6 2009 The levels of CYP1A1, CYP1A2, CY2C9 and CYP3A4 mRNAs were not altered by Zolpidem, whereas model inducers dioxin and rifampicin significantly induced CYP1A and CYP2/3 gene expression, respectively. Rifampin 117-127 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 22-28 19686824-6 2009 The levels of CYP1A1, CYP1A2, CY2C9 and CYP3A4 mRNAs were not altered by Zolpidem, whereas model inducers dioxin and rifampicin significantly induced CYP1A and CYP2/3 gene expression, respectively. Rifampin 117-127 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-46 19686824-6 2009 The levels of CYP1A1, CYP1A2, CY2C9 and CYP3A4 mRNAs were not altered by Zolpidem, whereas model inducers dioxin and rifampicin significantly induced CYP1A and CYP2/3 gene expression, respectively. Rifampin 117-127 peptidylprolyl isomerase F Homo sapiens 160-166 19577586-8 2009 The integrity of hepatocyte ZO-1 and occludin was altered by a seven-day administration of rifampicin. Rifampin 91-101 tight junction protein 1 Mus musculus 28-32 19577586-8 2009 The integrity of hepatocyte ZO-1 and occludin was altered by a seven-day administration of rifampicin. Rifampin 91-101 occludin Mus musculus 37-45 19577586-10 2009 The expression of hepatic ZO-1 and ZO-2 mRNA was significantly decreased, beginning as early as 30 min and remaining a lower level 12 h after a single dose of rifampicin. Rifampin 159-169 tight junction protein 1 Mus musculus 26-30 19577586-10 2009 The expression of hepatic ZO-1 and ZO-2 mRNA was significantly decreased, beginning as early as 30 min and remaining a lower level 12 h after a single dose of rifampicin. Rifampin 159-169 tight junction protein 2 Mus musculus 35-39 19520773-5 2009 Real-time polymerase chain reaction analysis of human hepatocyte cultures revealed that MD induces the mRNA expression of CYP2B6, CYP3A4, UGT1A1, and multidrug resistance 1 in a concentration-related manner, with the maximal induction of CYP2B6 challenging that of the induction by rifampicin. Rifampin 282-292 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 19520773-5 2009 Real-time polymerase chain reaction analysis of human hepatocyte cultures revealed that MD induces the mRNA expression of CYP2B6, CYP3A4, UGT1A1, and multidrug resistance 1 in a concentration-related manner, with the maximal induction of CYP2B6 challenging that of the induction by rifampicin. Rifampin 282-292 ATP binding cassette subfamily B member 1 Homo sapiens 150-172 19460945-0 2009 Rifampicin-activated human pregnane X receptor and CYP3A4 induction enhance acetaminophen-induced toxicity. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 27-46 19460945-3 2009 Human PXR activation and CYP3A4 induction enhanced APAP-induced hepatotoxicity as revealed by hepatic alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities elevated in serum, and hepatic necrosis after coadministration of rifampicin and APAP, compared with APAP administration alone. Rifampin 247-257 nuclear receptor subfamily 1 group I member 2 Homo sapiens 6-9 19460945-3 2009 Human PXR activation and CYP3A4 induction enhanced APAP-induced hepatotoxicity as revealed by hepatic alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities elevated in serum, and hepatic necrosis after coadministration of rifampicin and APAP, compared with APAP administration alone. Rifampin 247-257 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-31 19460945-6 2009 Moreover, mRNA analysis demonstrated that CYP3A4 and other PXR target genes were significantly induced by rifampicin treatment. Rifampin 106-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 19460945-6 2009 Moreover, mRNA analysis demonstrated that CYP3A4 and other PXR target genes were significantly induced by rifampicin treatment. Rifampin 106-116 nuclear receptor subfamily 1 group I member 2 Homo sapiens 59-62 19845431-7 2009 Using rifampicin as a reference standard, compounds with a C(ss,u)/RF value greater than 7.31 nmol l(-1) may induce CYP3A in vivo in humans. Rifampin 6-16 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-121 19843061-9 2009 CONCLUSIONS: Co-administration of rifampicin and roflumilast led to a reduction in total PDE4 inhibitory activity of roflumilast by about 58%. Rifampin 34-44 phosphodiesterase 4A Homo sapiens 89-93 19593667-6 2009 The expressions of PXR, SP1, and MRP3 were markedly enhanced after rifampicin treatment. Rifampin 67-77 nuclear receptor subfamily 1 group I member 2 Homo sapiens 19-22 19593667-6 2009 The expressions of PXR, SP1, and MRP3 were markedly enhanced after rifampicin treatment. Rifampin 67-77 ATP binding cassette subfamily C member 3 Homo sapiens 33-37 19593667-8 2009 It also observed that PXR, activated by rifampicin or knocked down via short hairpin RNAs, could enhance or reduce cells resistance to the chemotherapeutic agents through 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Rifampin 40-50 nuclear receptor subfamily 1 group I member 2 Homo sapiens 22-25 19703346-8 2009 Rifampicin (a potent inhibitor of organic anion transporting polypeptide (Oatp) 2), but not indometacin (a representative inhibitor of organic anion transporter (Oat) 2 and Oatp1) treatment, significantly inhibited the uptake of paclitaxel and TAX-2"-Et. Rifampin 0-10 solute carrier organic anion transporter family, member 1a4 Rattus norvegicus 34-81 19703346-9 2009 We characterized the rifampicin-sensitive uptake of paclitaxel and TAX-2"-Et using rat hepatocytes treated with PCN, which dramatically enhances hepatic Oatp2 protein levels. Rifampin 21-31 solute carrier organic anion transporter family, member 1a4 Rattus norvegicus 153-158 19364845-9 2009 In addition, daptomycin at a high dose (30 mg/kg) completely prevented the emergence of rifampin resistance in planktonic and adherent MRSA cells. Rifampin 88-96 solute carrier family 9 member A6 Homo sapiens 135-139 19364845-10 2009 Daptomycin at a high dose, corresponding to 6 mg/kg in humans, in combination with rifampin showed the highest activity against planktonic and adherent MRSA. Rifampin 83-91 solute carrier family 9 member A6 Homo sapiens 152-156 19364845-11 2009 Daptomycin plus rifampin is a promising treatment option for implant-associated MRSA infections. Rifampin 16-24 solute carrier family 9 member A6 Homo sapiens 80-84 19432554-7 2009 Median rifampin and pyrazinamide levels differed significantly by HIV status and CD4 cell count category. Rifampin 7-15 CD4 molecule Homo sapiens 81-84 19374892-0 2009 Rifampicin alters atorvastatin plasma concentration on the basis of SLCO1B1 521T>C polymorphism. Rifampin 0-10 solute carrier organic anion transporter family member 1B1 Homo sapiens 68-75 19374892-1 2009 BACKGROUND: Both atorvastatin and rifampicin are substrates of OATP1B1 (organic anion transporting polypeptide 1B1) encoded by SLCO1B1 gene. Rifampin 34-44 solute carrier organic anion transporter family member 1B1 Homo sapiens 63-70 19374892-1 2009 BACKGROUND: Both atorvastatin and rifampicin are substrates of OATP1B1 (organic anion transporting polypeptide 1B1) encoded by SLCO1B1 gene. Rifampin 34-44 solute carrier organic anion transporter family member 1B1 Homo sapiens 72-114 19374892-1 2009 BACKGROUND: Both atorvastatin and rifampicin are substrates of OATP1B1 (organic anion transporting polypeptide 1B1) encoded by SLCO1B1 gene. Rifampin 34-44 solute carrier organic anion transporter family member 1B1 Homo sapiens 127-134 19374892-2 2009 Rifampicin is a potent inhibitor of SLCO1B1 (IC50 1.5 umol/l) and SLCO1B1 521T>C functional genetic polymorphism alters the kinetics of atorvastatin in vivo. Rifampin 0-10 solute carrier organic anion transporter family member 1B1 Homo sapiens 36-43 19374892-3 2009 We hypothesize that rifampicin might influence atorvastatin kinetics in a SLCO1B1 polymorphism dependent manner. Rifampin 20-30 solute carrier organic anion transporter family member 1B1 Homo sapiens 74-81 19374892-7 2009 RESULTS: Rifampicin increased atorvastatin plasma concentration in accordance with SLCO1B1 521T>C genotype while the increasing percentage of AUC((0-48)) among c.521TT, c.521TC and c.521CC individuals were 833+/-245% vs 468+/-233% vs 330+/-223% (P=0.007). Rifampin 9-19 solute carrier organic anion transporter family member 1B1 Homo sapiens 83-90 19374892-9 2009 CONCLUSIONS: These results suggested that rifampicin elevated the plasma concentration of atorvastatin depending on SLCO1B1 genotype and rifampicin pharmacokinetics were not altered by SLCO1B1 genotype. Rifampin 42-52 solute carrier organic anion transporter family member 1B1 Homo sapiens 116-123 19369937-9 2009 But the difference in activity levels between the CYP2C9*1/*1 and *3/*3 genotypes before the administration of rifampin was sixfold. Rifampin 111-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 19702536-3 2009 A variety of xenobiotics such as phenobarbital, rifampicin, and hyperforin have been shown to induce the transcriptional expression of CYP2C genes in primary human hepatocytes and to increase the metabolism of CYP2C substrates in vivo in man. Rifampin 48-58 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 135-140 19702536-3 2009 A variety of xenobiotics such as phenobarbital, rifampicin, and hyperforin have been shown to induce the transcriptional expression of CYP2C genes in primary human hepatocytes and to increase the metabolism of CYP2C substrates in vivo in man. Rifampin 48-58 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 210-215 19584503-0 2009 Characterization of RPO B gene for detection of rifampicin drug resistance by SSCP and sequence analysis. Rifampin 48-58 DNA-directed RNA polymerase subunit beta Mycobacterium tuberculosis H37Rv 20-25 19429255-7 2009 In agreement, pretreatment with selective CYP inducers, beta-naphthoflavone and rifampin, could increase the cytotoxicity of ginkgolic acid in HepG2 cells. Rifampin 80-88 peptidylprolyl isomerase G Homo sapiens 42-45 19464568-0 2009 Expression and inducibility of CYP1A1, 1A2, 1B1 by beta-naphthoflavone and CYP2B22, 3A22, 3A29, 3A46 by rifampicin in the respiratory and olfactory mucosa of pig. Rifampin 104-114 cytochrome P450 family 1 subfamily A member 1 Sus scrofa 31-37 19457725-2 2009 Rifampicin is a CYP3A4 inducer while clarithromycin is known to inhibit CYP3A4. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 16-22 19464568-0 2009 Expression and inducibility of CYP1A1, 1A2, 1B1 by beta-naphthoflavone and CYP2B22, 3A22, 3A29, 3A46 by rifampicin in the respiratory and olfactory mucosa of pig. Rifampin 104-114 cytochrome P450 family 2 subfamily B member 6B Sus scrofa 75-82 19464568-4 2009 The rifampicin treatment resulted in a transcriptional activation of CYP2B22 and CYP3As genes in liver but not in respiratory and olfactory mucosa. Rifampin 4-14 cytochrome P450 family 2 subfamily B member 6B Sus scrofa 69-76 19417618-4 2009 Their hypothesis was that rifampin enhances the CYP3A-mediated metabolism of oxycodone and attenuates its pharmacologic effect. Rifampin 26-34 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-53 19371336-0 2009 Does CYP2E1 play a major role in the aggravation of isoniazid toxicity by rifampicin in human hepatocytes? Rifampin 74-84 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 5-11 19371336-4 2009 The main conclusion was that the difference in cytochrome P450 2E1 (CYP2E1) induction by rifampicin between rat and human hepatocytes accounted for the difference in exacerbation of isoniazid hepatotoxicity by rifampicin. Rifampin 89-99 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 47-66 19371336-4 2009 The main conclusion was that the difference in cytochrome P450 2E1 (CYP2E1) induction by rifampicin between rat and human hepatocytes accounted for the difference in exacerbation of isoniazid hepatotoxicity by rifampicin. Rifampin 89-99 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 68-74 19371336-4 2009 The main conclusion was that the difference in cytochrome P450 2E1 (CYP2E1) induction by rifampicin between rat and human hepatocytes accounted for the difference in exacerbation of isoniazid hepatotoxicity by rifampicin. Rifampin 210-220 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 47-66 19371336-4 2009 The main conclusion was that the difference in cytochrome P450 2E1 (CYP2E1) induction by rifampicin between rat and human hepatocytes accounted for the difference in exacerbation of isoniazid hepatotoxicity by rifampicin. Rifampin 210-220 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 68-74 19371336-6 2009 The authors presented data showing that rifampicin enhanced 4-NP activity and CYP2E1 mRNA expression in human hepatocytes, but not in rat hepatocytes. Rifampin 40-50 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 78-84 19371336-8 2009 Rifampicin is a strong inducer of human CYP3A; thus, the increase in 4-NP activity in human hepatocytes could be due to the induction of CYP3A. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-45 19371336-8 2009 Rifampicin is a strong inducer of human CYP3A; thus, the increase in 4-NP activity in human hepatocytes could be due to the induction of CYP3A. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 137-142 19371336-10 2009 Additionally, more experiments are needed to support the conclusion that rifampicin increased CYP2E1 mRNA expression in human hepatocytes because of the small sample size and the limitations of semi-quantitative RT-PCR. Rifampin 73-83 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 94-100 19251824-5 2009 In HepG2 and LS180 cells stably transduced with PXR.1, PXR target genes (CYP3A4, MDR1, CYP2B6, and UGT1A1) were higher than mock-transduced cells in the absence of ligand and were further induced in the presence of rifampin. Rifampin 215-223 peroxisomal biogenesis factor 5 Homo sapiens 48-53 19251824-5 2009 In HepG2 and LS180 cells stably transduced with PXR.1, PXR target genes (CYP3A4, MDR1, CYP2B6, and UGT1A1) were higher than mock-transduced cells in the absence of ligand and were further induced in the presence of rifampin. Rifampin 215-223 nuclear receptor subfamily 1 group I member 2 Homo sapiens 48-51 19251824-5 2009 In HepG2 and LS180 cells stably transduced with PXR.1, PXR target genes (CYP3A4, MDR1, CYP2B6, and UGT1A1) were higher than mock-transduced cells in the absence of ligand and were further induced in the presence of rifampin. Rifampin 215-223 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 73-79 19251824-5 2009 In HepG2 and LS180 cells stably transduced with PXR.1, PXR target genes (CYP3A4, MDR1, CYP2B6, and UGT1A1) were higher than mock-transduced cells in the absence of ligand and were further induced in the presence of rifampin. Rifampin 215-223 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 19480548-7 2009 The effective CYP3A29 induction in liver was also detected by rifampin oral intubation. Rifampin 62-70 cytochrome P450 family 3 subfamily A member 29 Sus scrofa 14-21 19074998-5 2009 Introduction of a mutation into either an alpha or beta half-site of CYP3A4 reporter genes almost completely diminished the rifampicin-induced transcription. Rifampin 124-134 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-75 19223781-1 2009 SETTING: Rifampicin may reduce plasma efavirenz concentrations by inducing the expression of the cytochrome P450 2B6, which metabolizes efavirenz. Rifampin 9-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-116 19144646-6 2009 The chromatin immunoprecipitation (ChIP) assay showed that PRMT1 was recruited to the regulatory region of the PXR target gene cytochrome P450 3A4 (CYP3A4), with a concomitant methylation of arginine 3 of histone H4, in response to the PXR agonist rifampicin. Rifampin 248-258 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 148-154 19188390-8 2009 Moreover, rifampin, a known inducer of efflux transport proteins, upregulated the expression of P-glycoprotein in Calu-3 cells and enhanced MXF active transport. Rifampin 10-18 ATP binding cassette subfamily B member 1 Homo sapiens 96-110 19381336-1 2009 OBJECTIVES: Rifampin is a potent inducer of the cytochrome P450 3A4 isoenzyme (CYP3A4) that metabolizes most protease inhibitor (PI) antiretrovirals. Rifampin 12-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-77 19381336-1 2009 OBJECTIVES: Rifampin is a potent inducer of the cytochrome P450 3A4 isoenzyme (CYP3A4) that metabolizes most protease inhibitor (PI) antiretrovirals. Rifampin 12-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 79-85 19199552-7 2009 Rifampin resistance emerged more frequently among hVISA isolates than among MRSA isolates (44% vs. 5.9%; P < .001). Rifampin 0-8 mitochondrial antiviral signaling protein Homo sapiens 50-55 19199552-9 2009 CONCLUSIONS: hVISA bacteremia was significantly associated with prolonged bacteremia duration, greater rates of complications, and emergence of rifampin resistance, compared with MRSA bacteremia. Rifampin 144-152 mitochondrial antiviral signaling protein Homo sapiens 13-18 18855943-7 2009 Treatment of hCMEC/D3 cells for 72 hr with rifampin or SR12813 (two well-established hPXR ligands) or PIs (atazanavir or ritonavir) resulted in an increase in P-gp expression by 1.8-, 6-, and 2-fold, respectively, with no effect observed for MRP1 expression. Rifampin 43-51 nuclear receptor subfamily 1 group I member 2 Homo sapiens 85-89 18855943-7 2009 Treatment of hCMEC/D3 cells for 72 hr with rifampin or SR12813 (two well-established hPXR ligands) or PIs (atazanavir or ritonavir) resulted in an increase in P-gp expression by 1.8-, 6-, and 2-fold, respectively, with no effect observed for MRP1 expression. Rifampin 43-51 phosphoglycolate phosphatase Homo sapiens 159-163 18855943-7 2009 Treatment of hCMEC/D3 cells for 72 hr with rifampin or SR12813 (two well-established hPXR ligands) or PIs (atazanavir or ritonavir) resulted in an increase in P-gp expression by 1.8-, 6-, and 2-fold, respectively, with no effect observed for MRP1 expression. Rifampin 43-51 ATP binding cassette subfamily B member 1 Homo sapiens 242-246 19144646-6 2009 The chromatin immunoprecipitation (ChIP) assay showed that PRMT1 was recruited to the regulatory region of the PXR target gene cytochrome P450 3A4 (CYP3A4), with a concomitant methylation of arginine 3 of histone H4, in response to the PXR agonist rifampicin. Rifampin 248-258 protein arginine methyltransferase 1 Homo sapiens 59-64 19144646-6 2009 The chromatin immunoprecipitation (ChIP) assay showed that PRMT1 was recruited to the regulatory region of the PXR target gene cytochrome P450 3A4 (CYP3A4), with a concomitant methylation of arginine 3 of histone H4, in response to the PXR agonist rifampicin. Rifampin 248-258 nuclear receptor subfamily 1 group I member 2 Homo sapiens 111-114 19144646-6 2009 The chromatin immunoprecipitation (ChIP) assay showed that PRMT1 was recruited to the regulatory region of the PXR target gene cytochrome P450 3A4 (CYP3A4), with a concomitant methylation of arginine 3 of histone H4, in response to the PXR agonist rifampicin. Rifampin 248-258 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 127-146 19114679-0 2009 Roles of NF-kappaB activation and peroxisome proliferator-activated receptor gamma inhibition in the effect of rifampin on inducible nitric oxide synthase transcription in human lung epithelial cells. Rifampin 111-119 peroxisome proliferator activated receptor gamma Homo sapiens 34-82 19114679-2 2009 Previous studies have found that rifampin increases inducible nitric oxide (NO) synthase (iNOS) expression and NO production. Rifampin 33-41 nitric oxide synthase 2 Homo sapiens 90-94 19114679-5 2009 The addition of rifampin increased the iNOS level by 1.9 +/- 0.3-fold at a dose of 10 microg/ml (P < 0.01) and by 4.0 +/- 0.3-fold at a dose of 50 microg/ml (P < 0.001). Rifampin 16-24 nitric oxide synthase 2 Homo sapiens 39-43 19114679-6 2009 Rifampin treatment also affected the transcription factors that regulate iNOS mRNA: there was an increased and prolonged degradation of the inhibitory subunit of NF-kappaB, a corresponding increase in the level of cytokine-induced DNA binding of NF-kappaB (2.1 +/- 0.2-fold), and a decrease in the level of expression of peroxisome proliferator-activated receptor gamma (PPARgamma). Rifampin 0-8 nitric oxide synthase 2 Homo sapiens 73-77 19114679-6 2009 Rifampin treatment also affected the transcription factors that regulate iNOS mRNA: there was an increased and prolonged degradation of the inhibitory subunit of NF-kappaB, a corresponding increase in the level of cytokine-induced DNA binding of NF-kappaB (2.1 +/- 0.2-fold), and a decrease in the level of expression of peroxisome proliferator-activated receptor gamma (PPARgamma). Rifampin 0-8 nuclear factor kappa B subunit 1 Homo sapiens 162-171 19114679-6 2009 Rifampin treatment also affected the transcription factors that regulate iNOS mRNA: there was an increased and prolonged degradation of the inhibitory subunit of NF-kappaB, a corresponding increase in the level of cytokine-induced DNA binding of NF-kappaB (2.1 +/- 0.2-fold), and a decrease in the level of expression of peroxisome proliferator-activated receptor gamma (PPARgamma). Rifampin 0-8 nuclear factor kappa B subunit 1 Homo sapiens 246-255 19114679-6 2009 Rifampin treatment also affected the transcription factors that regulate iNOS mRNA: there was an increased and prolonged degradation of the inhibitory subunit of NF-kappaB, a corresponding increase in the level of cytokine-induced DNA binding of NF-kappaB (2.1 +/- 0.2-fold), and a decrease in the level of expression of peroxisome proliferator-activated receptor gamma (PPARgamma). Rifampin 0-8 peroxisome proliferator activated receptor gamma Homo sapiens 321-369 19114679-6 2009 Rifampin treatment also affected the transcription factors that regulate iNOS mRNA: there was an increased and prolonged degradation of the inhibitory subunit of NF-kappaB, a corresponding increase in the level of cytokine-induced DNA binding of NF-kappaB (2.1 +/- 0.2-fold), and a decrease in the level of expression of peroxisome proliferator-activated receptor gamma (PPARgamma). Rifampin 0-8 peroxisome proliferator activated receptor gamma Homo sapiens 371-380 19114679-11 2009 The increases in the levels of NF-kappaB activation and NO production probably contribute to the therapeutic effects of rifampin. Rifampin 120-128 nuclear factor kappa B subunit 1 Homo sapiens 31-40 19114679-12 2009 However, given the role of NF-kappaB in upregulating many inflammatory genes and the roles of PPARgamma in downregulating inflammatory genes and in lipid and glucose metabolism, these findings have implications for potential adverse effects of rifampin in patients with chronic inflammatory diseases and glucose or lipid disorders. Rifampin 244-252 peroxisome proliferator activated receptor gamma Homo sapiens 94-103 19336894-3 2009 CYP1A1 and CYP3A8 mRNAs were significantly induced by 3-methylcholanthrene and rifampicin, respectively. Rifampin 79-89 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 0-6 19074998-7 2009 HepG2-based transactivation assays with the reporter gene constructs with or without mutations in the PXR binding element(s) demonstrated that this DR4 motif is essential for the transcriptional activation not only by rifampicin but also by various human PXR activators. Rifampin 218-228 nuclear receptor subfamily 1 group I member 2 Homo sapiens 102-105 19074998-7 2009 HepG2-based transactivation assays with the reporter gene constructs with or without mutations in the PXR binding element(s) demonstrated that this DR4 motif is essential for the transcriptional activation not only by rifampicin but also by various human PXR activators. Rifampin 218-228 major histocompatibility complex, class II, DR beta 4 Homo sapiens 148-151 19206053-6 2009 Inducers of metabolism (rifampicin, carbamazepine, St. John"s Wort) also induce the expression of drug transporters like ABCB1. Rifampin 24-34 ATP binding cassette subfamily B member 1 Homo sapiens 121-126 19147857-8 2009 To activate PXR, we used the PXR ligands, rifampicin, hyperforin, and pregnenolone-16alpha-carbonitrile (PCN), and measured abcb1 mRNA with quantitative polymerase chain reaction, P-glycoprotein expression with Western blotting, and P-glycoprotein transport activity with a calcein assay. Rifampin 42-52 nuclear receptor subfamily 1 group I member 2 Homo sapiens 12-15 19147857-9 2009 We provide first proof of principle that the human PXR ligands, rifampicin and hyperforin, but not the rodent PXR ligand, PCN, activate pig PXR at the blood-brain barrier and induce mRNA, protein expression, and transport activity of P-glycoprotein. Rifampin 64-74 nuclear receptor subfamily 1 group I member 2 Homo sapiens 51-54 19129222-1 2009 Chromatin immunoprecipitation (ChIP) studies were conducted in human hepatocytes treated with rifampicin in order to identify new pregnane-X receptor (PXR) target genes. Rifampin 94-104 nuclear receptor subfamily 1 group I member 2 Homo sapiens 130-149 19129222-1 2009 Chromatin immunoprecipitation (ChIP) studies were conducted in human hepatocytes treated with rifampicin in order to identify new pregnane-X receptor (PXR) target genes. Rifampin 94-104 nuclear receptor subfamily 1 group I member 2 Homo sapiens 151-154 19129222-4 2009 Of these, only CYP4F12 demonstrated increased binding in the presence of rifampicin. Rifampin 73-83 cytochrome P450 family 4 subfamily F member 12 Homo sapiens 15-22 19129222-7 2009 Both PXR and the steroid receptor coactivator (SRC-1) were found to bind to PXREs in the absence of rifampicin, although binding was stronger after rifampicin treatment. Rifampin 100-110 steroid receptor RNA activator 1 Homo sapiens 17-45 19129222-7 2009 Both PXR and the steroid receptor coactivator (SRC-1) were found to bind to PXREs in the absence of rifampicin, although binding was stronger after rifampicin treatment. Rifampin 100-110 nuclear receptor coactivator 1 Homo sapiens 47-52 19129222-7 2009 Both PXR and the steroid receptor coactivator (SRC-1) were found to bind to PXREs in the absence of rifampicin, although binding was stronger after rifampicin treatment. Rifampin 148-158 nuclear receptor subfamily 1 group I member 2 Homo sapiens 5-8 19129222-7 2009 Both PXR and the steroid receptor coactivator (SRC-1) were found to bind to PXREs in the absence of rifampicin, although binding was stronger after rifampicin treatment. Rifampin 148-158 steroid receptor RNA activator 1 Homo sapiens 17-45 19129222-7 2009 Both PXR and the steroid receptor coactivator (SRC-1) were found to bind to PXREs in the absence of rifampicin, although binding was stronger after rifampicin treatment. Rifampin 148-158 nuclear receptor coactivator 1 Homo sapiens 47-52 19129222-9 2009 In conclusion, our findings indicate that PXR is involved in the regulation of CYP4F12 and that PXR along with SRC1 binds to a broad range of promoters but that many of these are not inducible by rifampicin. Rifampin 196-206 nuclear receptor coactivator 1 Homo sapiens 111-115 18606396-2 2009 Two major groups of ligand-activated nuclear receptors/xenosensors evolved: the Ah receptor (activated by aryl hydrocarbons and drugs such as omeprazole) and type 2 steroid receptors such as PXR and CAR, activated by drugs such as rifampicin, carbamazepin and phenytoin. Rifampin 231-241 nuclear receptor subfamily 1 group I member 2 Homo sapiens 191-194 18606396-2 2009 Two major groups of ligand-activated nuclear receptors/xenosensors evolved: the Ah receptor (activated by aryl hydrocarbons and drugs such as omeprazole) and type 2 steroid receptors such as PXR and CAR, activated by drugs such as rifampicin, carbamazepin and phenytoin. Rifampin 231-241 CXADR pseudogene 1 Homo sapiens 199-202 19013243-0 2009 Effects of rifampin on CYP2E1-dependent hepatotoxicity of isoniazid in rats. Rifampin 11-19 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 23-29 18981260-5 2009 The degree of the PXR-mediated locking of SMRT depends on the relative concentration of vitamin D(3) to the human PXR activator rifampicin; SMRT increased its dissociation as this ratio increased. Rifampin 128-138 nuclear receptor subfamily 1 group I member 2 Homo sapiens 18-21 18981260-5 2009 The degree of the PXR-mediated locking of SMRT depends on the relative concentration of vitamin D(3) to the human PXR activator rifampicin; SMRT increased its dissociation as this ratio increased. Rifampin 128-138 nuclear receptor corepressor 2 Homo sapiens 42-46 18981260-5 2009 The degree of the PXR-mediated locking of SMRT depends on the relative concentration of vitamin D(3) to the human PXR activator rifampicin; SMRT increased its dissociation as this ratio increased. Rifampin 128-138 nuclear receptor corepressor 2 Homo sapiens 140-144 19013243-11 2009 Rifampin co-administration significantly attenuated isoniazid-induced CYP2E1 levels (p<0.01) and inhibition of mu GST (p<0.01). Rifampin 0-8 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 70-76 19228077-12 2009 For example, sorafenib exposure was reduced by an average 37% with concomitant administration of the CYP3A4 inducer rifampicin (rifampin); sorafenib concentrations may also be decreased by other CYP3A4 inducers. Rifampin 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 101-107 19006545-4 2009 In the present study we aimed to determine the time course of the decrease in plasma 4beta-hydroxycholesterol after termination of induction of CYP3A4/5 by rifampicin. Rifampin 156-166 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 144-150 19515014-10 2009 CYP2C9 is subject to induction by rifampin, phenobarbital, and dexamethasone, indicating the involvement of pregnane X receptor, constitutive androstane receptor and glucocorticoid receptor in the regulation of CYP2C9. Rifampin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-10 2009 CYP2C9 is subject to induction by rifampin, phenobarbital, and dexamethasone, indicating the involvement of pregnane X receptor, constitutive androstane receptor and glucocorticoid receptor in the regulation of CYP2C9. Rifampin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 211-217 19881256-3 2009 Positive controls for CYP1A2 and CYP3A4 used beta-naphthoflavone (beta-NF) and rifampicin (Rif), respectively. Rifampin 79-89 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 22-28 19881256-3 2009 Positive controls for CYP1A2 and CYP3A4 used beta-naphthoflavone (beta-NF) and rifampicin (Rif), respectively. Rifampin 91-94 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 22-28 19697859-1 2009 The effects of isoniazid and rifampicin on the peripheral mononuclear production of IL-2, IL-12, IL-10, IFN-gamma, and TGF-beta were evaluated in patients with infiltrative pulmonary tuberculosis. Rifampin 29-39 interleukin 2 Homo sapiens 84-88 19697859-2 2009 Irrespective of the susceptibility of the causative agent to the essential antituberculous drugs, rifampicin was ascertained to initiate increased IL-2 secretion and to reduce the generation of IL-12, IFN-gamma and TGF-beta. Rifampin 98-108 interleukin 2 Homo sapiens 147-151 19697859-2 2009 Irrespective of the susceptibility of the causative agent to the essential antituberculous drugs, rifampicin was ascertained to initiate increased IL-2 secretion and to reduce the generation of IL-12, IFN-gamma and TGF-beta. Rifampin 98-108 interferon gamma Homo sapiens 201-210 19697859-2 2009 Irrespective of the susceptibility of the causative agent to the essential antituberculous drugs, rifampicin was ascertained to initiate increased IL-2 secretion and to reduce the generation of IL-12, IFN-gamma and TGF-beta. Rifampin 98-108 transforming growth factor beta 1 Homo sapiens 215-223 19704172-1 2009 BACKGROUND: Rifampicin induces expression of the cytochrome P450 isoenzyme 2B6 (CYP2B6), which metabolizes efavirenz. Rifampin 12-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-78 19704172-1 2009 BACKGROUND: Rifampicin induces expression of the cytochrome P450 isoenzyme 2B6 (CYP2B6), which metabolizes efavirenz. Rifampin 12-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 19601803-7 2009 CYP2D6 is largely uninducible by prototypical CYP inducers such as phenobarbital, rifampin and dexamethasone, but it is regulated by hepatocyte nuclear factor-4alpha, a nuclear receptor. Rifampin 82-90 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-6 19601803-7 2009 CYP2D6 is largely uninducible by prototypical CYP inducers such as phenobarbital, rifampin and dexamethasone, but it is regulated by hepatocyte nuclear factor-4alpha, a nuclear receptor. Rifampin 82-90 peptidylprolyl isomerase G Homo sapiens 0-3 19601803-7 2009 CYP2D6 is largely uninducible by prototypical CYP inducers such as phenobarbital, rifampin and dexamethasone, but it is regulated by hepatocyte nuclear factor-4alpha, a nuclear receptor. Rifampin 82-90 hepatocyte nuclear factor 4 alpha Homo sapiens 133-165 19202563-3 2009 Clotrimazole, rifampin, ritonavir, phenytoin, and phenobarbital induced CYP2C9 consistent with previous findings for CYP3A4. Rifampin 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 19202563-3 2009 Clotrimazole, rifampin, ritonavir, phenytoin, and phenobarbital induced CYP2C9 consistent with previous findings for CYP3A4. Rifampin 14-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 117-123 19202563-6 2009 In 17 donors, rifampin increased mean basal CYP2C9 activity from 59 +/- 43 to 143 +/- 68 pmol/mg protein/min; fold induction ranged from 1.4- to 6.4-fold. Rifampin 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 19077665-3 2009 It was recently reported that hepatocyte nuclear receptor 4alpha (HNF4alpha), a master regulator of several nuclear receptors, associates with PXR thus regulates the expression of CYP3A4 under rifampin treatment. Rifampin 193-201 hepatocyte nuclear factor 4 alpha Homo sapiens 41-75 19077665-3 2009 It was recently reported that hepatocyte nuclear receptor 4alpha (HNF4alpha), a master regulator of several nuclear receptors, associates with PXR thus regulates the expression of CYP3A4 under rifampin treatment. Rifampin 193-201 nuclear receptor subfamily 1 group I member 2 Homo sapiens 143-146 19077665-3 2009 It was recently reported that hepatocyte nuclear receptor 4alpha (HNF4alpha), a master regulator of several nuclear receptors, associates with PXR thus regulates the expression of CYP3A4 under rifampin treatment. Rifampin 193-201 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 180-186 19077665-4 2009 We therefore focused on the role of PXR-HNF4alpha interaction in the transcriptional regulation of CYP3A4 under rifampin-mediated ketoconazole inhibition. Rifampin 112-120 nuclear receptor subfamily 1 group I member 2 Homo sapiens 36-39 19077665-4 2009 We therefore focused on the role of PXR-HNF4alpha interaction in the transcriptional regulation of CYP3A4 under rifampin-mediated ketoconazole inhibition. Rifampin 112-120 hepatocyte nuclear factor 4 alpha Homo sapiens 40-49 19077665-4 2009 We therefore focused on the role of PXR-HNF4alpha interaction in the transcriptional regulation of CYP3A4 under rifampin-mediated ketoconazole inhibition. Rifampin 112-120 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-105 18799805-6 2008 In TgCYP3A4/hPXR mice, CYP3A4 was strongly induced by rifampicin, a human-specific PXR ligand, but not by pregnenolone 16alpha-carbonitrile, a rodent-specific PXR ligand. Rifampin 54-64 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 5-11 18799805-6 2008 In TgCYP3A4/hPXR mice, CYP3A4 was strongly induced by rifampicin, a human-specific PXR ligand, but not by pregnenolone 16alpha-carbonitrile, a rodent-specific PXR ligand. Rifampin 54-64 nuclear receptor subfamily 1 group I member 2 Homo sapiens 13-16 18799805-6 2008 In TgCYP3A4/hPXR mice, CYP3A4 was strongly induced by rifampicin, a human-specific PXR ligand, but not by pregnenolone 16alpha-carbonitrile, a rodent-specific PXR ligand. Rifampin 54-64 nuclear receptor subfamily 1 group I member 2 Homo sapiens 83-86 19228077-12 2009 For example, sorafenib exposure was reduced by an average 37% with concomitant administration of the CYP3A4 inducer rifampicin (rifampin); sorafenib concentrations may also be decreased by other CYP3A4 inducers. Rifampin 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 195-201 19228077-12 2009 For example, sorafenib exposure was reduced by an average 37% with concomitant administration of the CYP3A4 inducer rifampicin (rifampin); sorafenib concentrations may also be decreased by other CYP3A4 inducers. Rifampin 128-136 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 101-107 19228077-12 2009 For example, sorafenib exposure was reduced by an average 37% with concomitant administration of the CYP3A4 inducer rifampicin (rifampin); sorafenib concentrations may also be decreased by other CYP3A4 inducers. Rifampin 128-136 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 195-201 18989830-4 2008 CYP3A4 mRNA and activity were induced by the prototypical pregnane X receptor (PXR) ligands, rifampicin (E(max) = 36- and 6-fold, respectively, and EC(50) = 4 microM) and phenobarbital (E(max) = 12- and 4-fold, respectively, and EC(50) = 205 microM). Rifampin 93-103 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 18989830-4 2008 CYP3A4 mRNA and activity were induced by the prototypical pregnane X receptor (PXR) ligands, rifampicin (E(max) = 36- and 6-fold, respectively, and EC(50) = 4 microM) and phenobarbital (E(max) = 12- and 4-fold, respectively, and EC(50) = 205 microM). Rifampin 93-103 nuclear receptor subfamily 1 group I member 2 Homo sapiens 58-77 18989830-4 2008 CYP3A4 mRNA and activity were induced by the prototypical pregnane X receptor (PXR) ligands, rifampicin (E(max) = 36- and 6-fold, respectively, and EC(50) = 4 microM) and phenobarbital (E(max) = 12- and 4-fold, respectively, and EC(50) = 205 microM). Rifampin 93-103 nuclear receptor subfamily 1 group I member 2 Homo sapiens 79-82 19010908-6 2008 Interestingly, treatment of breast cancer cells in vitro with the PXR agonist rifampin induced OATP1A2 expression in a time-dependent and concentration-dependent manner. Rifampin 78-86 nuclear receptor subfamily 1 group I member 2 Homo sapiens 66-69 19010908-6 2008 Interestingly, treatment of breast cancer cells in vitro with the PXR agonist rifampin induced OATP1A2 expression in a time-dependent and concentration-dependent manner. Rifampin 78-86 solute carrier organic anion transporter family member 1A2 Homo sapiens 95-102 19010908-8 2008 The rifampin response was abrogated after small interfering RNA targeting of PXR. Rifampin 4-12 nuclear receptor subfamily 1 group I member 2 Homo sapiens 77-80 18981575-1 2008 This study investigated the changes in the mRNA levels of the ATP binding cassette (ABC) transporters multidrug resistance 1 (MDR1), multidrug resistance-associated protein 1 (MRP1), and multidrug resistance-associated protein 2 (MRP2) following exposure to the prototypical microsomal enzyme inducers rifampicin (Rif), dexamethasone (Dex), and omeprazole (Ome) in primary cultures of cryopreserved human and cynomolgus monkey hepatocytes. Rifampin 302-312 ATP binding cassette subfamily B member 1 Homo sapiens 102-124 18981575-1 2008 This study investigated the changes in the mRNA levels of the ATP binding cassette (ABC) transporters multidrug resistance 1 (MDR1), multidrug resistance-associated protein 1 (MRP1), and multidrug resistance-associated protein 2 (MRP2) following exposure to the prototypical microsomal enzyme inducers rifampicin (Rif), dexamethasone (Dex), and omeprazole (Ome) in primary cultures of cryopreserved human and cynomolgus monkey hepatocytes. Rifampin 302-312 ATP binding cassette subfamily C member 2 Homo sapiens 230-234 18981575-1 2008 This study investigated the changes in the mRNA levels of the ATP binding cassette (ABC) transporters multidrug resistance 1 (MDR1), multidrug resistance-associated protein 1 (MRP1), and multidrug resistance-associated protein 2 (MRP2) following exposure to the prototypical microsomal enzyme inducers rifampicin (Rif), dexamethasone (Dex), and omeprazole (Ome) in primary cultures of cryopreserved human and cynomolgus monkey hepatocytes. Rifampin 314-317 ATP binding cassette subfamily B member 1 Homo sapiens 102-124 18981575-1 2008 This study investigated the changes in the mRNA levels of the ATP binding cassette (ABC) transporters multidrug resistance 1 (MDR1), multidrug resistance-associated protein 1 (MRP1), and multidrug resistance-associated protein 2 (MRP2) following exposure to the prototypical microsomal enzyme inducers rifampicin (Rif), dexamethasone (Dex), and omeprazole (Ome) in primary cultures of cryopreserved human and cynomolgus monkey hepatocytes. Rifampin 314-317 ATP binding cassette subfamily C member 2 Homo sapiens 230-234 18658192-8 2008 In contrast, rifampicin concentrations above the MIC were required to exert sustained killing in CSF(CO(2)). Rifampin 13-23 complement C2 Homo sapiens 97-107 18786598-0 2008 Expression and induction by rifampicin of CAR- and PXR-regulated CYP2B and CYP3A in liver, kidney and airways of pig. Rifampin 28-38 CXADR Ig-like cell adhesion molecule Sus scrofa 42-46 18786598-0 2008 Expression and induction by rifampicin of CAR- and PXR-regulated CYP2B and CYP3A in liver, kidney and airways of pig. Rifampin 28-38 nuclear receptor subfamily 1 group I member 2 Sus scrofa 51-54 18786598-9 2008 Thus, it is likely that rifampicin induced CYP2B22 both in liver and kidney of pig, not via activation of CAR, but via PXR, through a cross-talk mechanism, as previously observed in human liver. Rifampin 24-34 cytochrome P450 family 2 subfamily B member 6B Sus scrofa 43-50 18786598-9 2008 Thus, it is likely that rifampicin induced CYP2B22 both in liver and kidney of pig, not via activation of CAR, but via PXR, through a cross-talk mechanism, as previously observed in human liver. Rifampin 24-34 nuclear receptor subfamily 1 group I member 2 Sus scrofa 119-122 18959805-9 2008 She remains on her original cART regimen with an undetectable viral load and normal CD4 count (34%; 1398 x 106/l).The combination of rifampicin and azithromycin was well tolerated, simple to administer and effective. Rifampin 133-143 CD4 molecule Homo sapiens 84-87 18800312-5 2008 Reporter assays were used to evaluate the function of all known hPXR variants in response to the secondary bile acid lithocholic acid and therapeutic agents rifampicin, ursodeoxycholic acid and dexamethasone. Rifampin 157-167 nuclear receptor subfamily 1 group I member 2 Homo sapiens 64-68 18647599-2 2008 The extent of CYP3A4 induction (measured as RIF ratio in comparison to rifampicin) and EC50 was obtained from the dose-response curve. Rifampin 71-81 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-20 18712882-7 2008 However, the BDO-hAGT complex at Cys(150) exhibited stability for more than 1 h. The effect of hAGT and mutants on BDO-induced genotoxicity was studied in E. coli using the forward assay to rifampicin resistance. Rifampin 190-200 angiotensinogen Homo sapiens 17-21 18799805-9 2008 By using TgCYP3A4/hPXR mice, human PXR-CYP3A4-mediated rifampicin-protease inhibitor interactions were recapitulated, as the metabolic stability of amprenavir, nelfinavir, and saquinavir decreased 52, 53, and 99%, respectively, in the liver microsomes of TgCYP3A4/hPXR mice pretreated with rifampicin. Rifampin 55-65 nuclear receptor subfamily 1, group I, member 2 Mus musculus 19-22 18799805-9 2008 By using TgCYP3A4/hPXR mice, human PXR-CYP3A4-mediated rifampicin-protease inhibitor interactions were recapitulated, as the metabolic stability of amprenavir, nelfinavir, and saquinavir decreased 52, 53, and 99%, respectively, in the liver microsomes of TgCYP3A4/hPXR mice pretreated with rifampicin. Rifampin 55-65 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 11-17 18765524-10 2008 In mice harboring SKOV-3 xenografts, rifampicin (PXR agonist) induces cell proliferation and tumor growth. Rifampin 37-47 nuclear receptor subfamily 1, group I, member 2 Mus musculus 49-52 18544536-4 2008 In agreement with previous reports, we found that rexinoids are weak activators of SXR, but we also found that they can antagonize SXR activation by the potent SXR agonist rifampicin. Rifampin 172-182 nuclear receptor subfamily 1 group I member 2 Homo sapiens 131-134 18573861-9 2008 Metabolism of 17-OHPC was significantly greater in FHH treated with the CYP3A inducers, rifampin and phenobarbital. Rifampin 88-96 calcium sensing receptor Homo sapiens 51-54 18534776-1 2008 The protective effects of Ginkgoselect Phytosome (GBP) on Rifampicin (RMP) induced hepatotoxicity and the probable mechanism(s) involved in this protection were investigated in rats. Rifampin 58-68 transmembrane protein 132A Rattus norvegicus 50-53 18523138-10 2008 In LS180 human intestinal cells, rifampicin increased UGT2A3 mRNA by more than 4.5-fold compared with vehicle, whereas levels were not significantly affected by the arylhydrocarbon receptor ligand beta-naphthoflavone. Rifampin 33-43 UDP glucuronosyltransferase family 2 member A3 Homo sapiens 54-60 18695506-0 2008 Rifampicin reduces alpha-synuclein in a transgenic mouse model of multiple system atrophy. Rifampin 0-10 synuclein, alpha Mus musculus 19-34 18695506-4 2008 This study investigates the ability of the antibiotic rifampicin to reduce alpha-synuclein aggregation and the associated neurodegeneration in a transgenic mouse model of MSA. Rifampin 54-64 synuclein, alpha Mus musculus 75-90 18695506-5 2008 We report a reduction in monomeric and oligomeric alpha-synuclein and a reduction in phosphorylated alpha-synuclein (S129) upon rifampicin treatment. Rifampin 128-138 synuclein, alpha Mus musculus 100-115 18556442-8 2008 The in vivo results showed a differential effect of Oatp1b2 on hepatic uptake of the model compounds, indicating that Oatp1b2 plays a more significant role in the hepatobiliary disposition of rifampicin and lovastatin than the other compounds tested. Rifampin 192-202 solute carrier organic anion transporter family, member 1b2 Mus musculus 52-59 18556442-8 2008 The in vivo results showed a differential effect of Oatp1b2 on hepatic uptake of the model compounds, indicating that Oatp1b2 plays a more significant role in the hepatobiliary disposition of rifampicin and lovastatin than the other compounds tested. Rifampin 192-202 solute carrier organic anion transporter family, member 1b2 Mus musculus 118-125 18559486-9 2008 Stimulation with 3-methylcholanthrene and rifampicin markedly increased CYP1A1/2 or CYP3A4 activities, which could be selectively inhibited by nifedipine, verapamil, ketoconazole, and quercetin. Rifampin 42-52 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 72-78 18559486-9 2008 Stimulation with 3-methylcholanthrene and rifampicin markedly increased CYP1A1/2 or CYP3A4 activities, which could be selectively inhibited by nifedipine, verapamil, ketoconazole, and quercetin. Rifampin 42-52 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 84-90 18728241-6 2008 Our results indicate that CYP2B6 c.516TT genotype can be used to identify efavirenz poor metabolizers in patients co-treated with rifampin. Rifampin 130-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-32 18544536-4 2008 In agreement with previous reports, we found that rexinoids are weak activators of SXR, but we also found that they can antagonize SXR activation by the potent SXR agonist rifampicin. Rifampin 172-182 nuclear receptor subfamily 1 group I member 2 Homo sapiens 131-134 18544536-5 2008 This antagonism included suppression of rifampicin-induced expression of SXR target genes, as well as reduced binding of SXR/RXR to SXR response elements both in vivo and in vitro. Rifampin 40-50 nuclear receptor subfamily 1 group I member 2 Homo sapiens 73-76 18544536-9 2008 We suggest that the ability of rexinoids to induce degradation of both SXR and RXR, in combination with competition for binding to SXR, can also explain why rexinoids antagonize the activation of SXR by drugs like rifampicin. Rifampin 214-224 nuclear receptor subfamily 1 group I member 2 Homo sapiens 71-74 18544536-9 2008 We suggest that the ability of rexinoids to induce degradation of both SXR and RXR, in combination with competition for binding to SXR, can also explain why rexinoids antagonize the activation of SXR by drugs like rifampicin. Rifampin 214-224 retinoid X receptor alpha Homo sapiens 79-82 18544536-9 2008 We suggest that the ability of rexinoids to induce degradation of both SXR and RXR, in combination with competition for binding to SXR, can also explain why rexinoids antagonize the activation of SXR by drugs like rifampicin. Rifampin 214-224 nuclear receptor subfamily 1 group I member 2 Homo sapiens 131-134 18544536-9 2008 We suggest that the ability of rexinoids to induce degradation of both SXR and RXR, in combination with competition for binding to SXR, can also explain why rexinoids antagonize the activation of SXR by drugs like rifampicin. Rifampin 214-224 nuclear receptor subfamily 1 group I member 2 Homo sapiens 131-134 18490434-6 2008 Rifampin increased ERBTs in CYP3A5(*)1 carriers (4.68 versus 2.60%, P = 0.0008) and noncarriers (3.55 versus 2.11%, P = 0.0017), whereas dexamethasone increased ERBTs only in CYP3A5(*)1 noncarriers (3.03 versus 2.14%, P = 0.031). Rifampin 0-8 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 175-181 18727801-5 2008 Extracellular (broth) and intracellular (THP-1 macrophages) activities of rifampicin, linezolid and fusidic acid at C(max), and of vancomycin, daptomycin, quinupristin-dalfopristin and oritavancin over a wide range of extracellular concentrations (with pharmacological modelling to determine E(max)), were measured at 24 h. Increases in vancomycin MICs correlated with increased drug binding, and decreased cell wall turnover and detergent-induced autolysis. Rifampin 74-84 GLI family zinc finger 2 Homo sapiens 41-46 18505790-6 2008 CYP3A4 activity was significantly increased by rifampin (9.7-fold), nafcillin and dicloxacillin (5.9-fold), and weakly induced (2-fold) by tetracycline, sufisoxazole, troleandomycin, and clindamycin. Rifampin 47-55 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 18413659-0 2008 Targeted disruption of murine organic anion-transporting polypeptide 1b2 (Oatp1b2/Slco1b2) significantly alters disposition of prototypical drug substrates pravastatin and rifampin. Rifampin 172-180 solute carrier organic anion transporter family, member 1b2 Mus musculus 74-81 19356089-0 2008 Quantitative relationship between rifampicin exposure and induction of Cyp3a11 in SXR humanized mice: extrapolation to human CYP3A4 induction potential. Rifampin 34-44 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 71-78 19356089-0 2008 Quantitative relationship between rifampicin exposure and induction of Cyp3a11 in SXR humanized mice: extrapolation to human CYP3A4 induction potential. Rifampin 34-44 transposition, Chr Y, Cattanach 1 Mus musculus 82-85 19356089-1 2008 The SXR humanized mouse model was used to quantitatively assess an in vivo induction response of the human PXR agonist, rifampicin. Rifampin 120-130 transposition, Chr Y, Cattanach 1 Mus musculus 4-7 19356089-1 2008 The SXR humanized mouse model was used to quantitatively assess an in vivo induction response of the human PXR agonist, rifampicin. Rifampin 120-130 nuclear receptor subfamily 1 group I member 2 Homo sapiens 107-110 19356089-2 2008 Three days of rifampicin treatment increased RNA expression and microsomal enzyme activity of CYP3A11, as well as significantly reduced triazolam plasma exposure. Rifampin 14-24 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 94-101 18413659-8 2008 After a single intravenous dose of rifampin (1 mg/kg), a 1.7-fold increase in plasma area under the concentration-time curve (AUC) was observed, whereas the liver-to-plasma ratio was reduced by 5-fold, and nearly 8-fold when assessed at steady-state conditions after 24 h of continuous subcutaneous infusion in Slco1b2(-/-) mice. Rifampin 35-43 solute carrier organic anion transporter family, member 1b2 Mus musculus 311-318 18509604-4 2008 RESULTS: MEB (80 pM) transport by OATP1B1 and OATP1B3 was inhibited by rifampicin (50 microM) to 10% and 4% of control, respectively. Rifampin 71-81 solute carrier organic anion transporter family member 1B1 Homo sapiens 34-41 18509604-4 2008 RESULTS: MEB (80 pM) transport by OATP1B1 and OATP1B3 was inhibited by rifampicin (50 microM) to 10% and 4% of control, respectively. Rifampin 71-81 solute carrier organic anion transporter family member 1B3 Homo sapiens 46-53 18601745-9 2008 RESULTS: Treatment of rats with INH+RIF or INH+RIF+PZA induced hepatotoxicity as evidenced by biochemical measurements: serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP) activities and the levels of total bilirubin were elevated, and the levels of albumin and total protein were decreased in drugs-treated animals. Rifampin 36-39 glutamic-oxaloacetic transaminase 2 Rattus norvegicus 158-184 18691987-1 2008 BACKGROUND: Rifampicin (rifampin) has been reported to have drug-drug interaction with several 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors due to its ability to influence the function of cytochrome P450 enzymes or transporters. Rifampin 12-22 3-hydroxy-3-methylglutaryl-CoA reductase Homo sapiens 95-142 18691987-1 2008 BACKGROUND: Rifampicin (rifampin) has been reported to have drug-drug interaction with several 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors due to its ability to influence the function of cytochrome P450 enzymes or transporters. Rifampin 24-32 3-hydroxy-3-methylglutaryl-CoA reductase Homo sapiens 95-142 18457428-3 2008 Recent studies have highlighted the potential use of rifampicin as an inhibitor of amyloid formation by a variety of polypeptides; however, there are conflicting reports on its ability to inhibit amyloid formation by islet amyloid polypeptide (IAPP). Rifampin 53-63 islet amyloid polypeptide Homo sapiens 217-242 18457428-3 2008 Recent studies have highlighted the potential use of rifampicin as an inhibitor of amyloid formation by a variety of polypeptides; however, there are conflicting reports on its ability to inhibit amyloid formation by islet amyloid polypeptide (IAPP). Rifampin 53-63 islet amyloid polypeptide Homo sapiens 244-248 18457428-7 2008 However, the effects of oxidized and reduced rifampicin are similar, in that neither prevents amyloid formation by IAPP. Rifampin 45-55 islet amyloid polypeptide Homo sapiens 115-119 18332078-5 2008 By using an optimized induction assay for Fa2N-4 cells, CYP3A4 induction by rifampicin, the prototypical PXR activator, increased from 1.5- to 7-fold at the level of functional activity. Rifampin 76-86 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 56-62 18332078-5 2008 By using an optimized induction assay for Fa2N-4 cells, CYP3A4 induction by rifampicin, the prototypical PXR activator, increased from 1.5- to 7-fold at the level of functional activity. Rifampin 76-86 nuclear receptor subfamily 1 group I member 2 Homo sapiens 105-108 18332078-10 2008 The EC(50) value for rifampicin-mediated CYP3A4 induction was 10-fold higher than that in human hepatocytes. Rifampin 21-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-47 18473749-9 2008 A small number of drugs such as rifampin, phenytoin and ritonavir are identified as inducers of CYP3A4. Rifampin 32-40 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 96-102 18560033-15 2008 The biochip-based analysis of RIF and INH susceptibility provides fast (less than 24 hours) and accurate identification of the mutations in gene rpoB, katG, inhA, ahpC responsible for resistance to rifampin and isoniazid. Rifampin 198-206 inhibin subunit alpha Homo sapiens 157-161 18490434-6 2008 Rifampin increased ERBTs in CYP3A5(*)1 carriers (4.68 versus 2.60%, P = 0.0008) and noncarriers (3.55 versus 2.11%, P = 0.0017), whereas dexamethasone increased ERBTs only in CYP3A5(*)1 noncarriers (3.03 versus 2.14%, P = 0.031). Rifampin 0-8 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 28-34 18492796-10 2008 performed a pharmacokinetic analysis with the human OATP1B1 and -1B3 substrates rifampicin and pravastatin and demonstrated a reduced liver-to-plasma ratio for these drugs in knockout compared with control mice, providing strong evidence that Oatp1b2 played an important role in the disposition of these drugs. Rifampin 80-90 solute carrier organic anion transporter family member 1B1 Homo sapiens 52-68 18492796-10 2008 performed a pharmacokinetic analysis with the human OATP1B1 and -1B3 substrates rifampicin and pravastatin and demonstrated a reduced liver-to-plasma ratio for these drugs in knockout compared with control mice, providing strong evidence that Oatp1b2 played an important role in the disposition of these drugs. Rifampin 80-90 solute carrier organic anion transporter family, member 1b2 Mus musculus 243-250 19468476-11 2008 Rifampicin has to be avoided in allograft recipients as it activates cytochrome-P450 enzymes and thereby decreases the therapeutic levels of cyclosporine and prednisolone. Rifampin 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-84 18332086-5 2008 CYP3A4 activity (1"-hydroxymidazolam formation) was increased (2-fold) by rifampin (10 microM) but was reduced by the PIs (1.5- to 7-fold). Rifampin 74-82 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 18332086-8 2008 Furthermore, in a PXR-knockdown stable LS180 cell line, induction of CYP3A4 and MDR1 mRNA after treatment with PIs and rifampin was significantly reduced (1.4- to 5-fold) compared with that in PXR nonsilenced cells. Rifampin 119-127 nuclear receptor subfamily 1 group I member 2 Homo sapiens 18-21 18332086-8 2008 Furthermore, in a PXR-knockdown stable LS180 cell line, induction of CYP3A4 and MDR1 mRNA after treatment with PIs and rifampin was significantly reduced (1.4- to 5-fold) compared with that in PXR nonsilenced cells. Rifampin 119-127 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-75 18332086-8 2008 Furthermore, in a PXR-knockdown stable LS180 cell line, induction of CYP3A4 and MDR1 mRNA after treatment with PIs and rifampin was significantly reduced (1.4- to 5-fold) compared with that in PXR nonsilenced cells. Rifampin 119-127 ATP binding cassette subfamily B member 1 Homo sapiens 80-84 18332086-8 2008 Furthermore, in a PXR-knockdown stable LS180 cell line, induction of CYP3A4 and MDR1 mRNA after treatment with PIs and rifampin was significantly reduced (1.4- to 5-fold) compared with that in PXR nonsilenced cells. Rifampin 119-127 nuclear receptor subfamily 1 group I member 2 Homo sapiens 193-196 18535361-6 2008 For example,the antibiotic rifampicin, one of the strongest inducers of the human gene CYP3A4, was the strongest inducer of the worm ortholog CYP13A7. Rifampin 27-37 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 87-93 18332862-3 2008 The ability of rifampin, a known PXR agonist and digoxin, a model MDR1 substrate, to regulate MDR1 expression and transport activity has been tested, in these T84 cells. Rifampin 15-23 ATP binding cassette subfamily B member 1 Homo sapiens 94-98 18332862-6 2008 KEY RESULTS: Rifampin exposure (10 microM, 72 hours) increased MDR1 transcript levels (3.4 fold), MDR1 total protein levels (4.4 fold), apical MDR1 protein (2.7 fold) and functional activity of MDR1 (1.2 fold). Rifampin 13-21 ATP binding cassette subfamily B member 1 Homo sapiens 63-67 18332862-6 2008 KEY RESULTS: Rifampin exposure (10 microM, 72 hours) increased MDR1 transcript levels (3.4 fold), MDR1 total protein levels (4.4 fold), apical MDR1 protein (2.7 fold) and functional activity of MDR1 (1.2 fold). Rifampin 13-21 ATP binding cassette subfamily B member 1 Homo sapiens 98-102 18332862-6 2008 KEY RESULTS: Rifampin exposure (10 microM, 72 hours) increased MDR1 transcript levels (3.4 fold), MDR1 total protein levels (4.4 fold), apical MDR1 protein (2.7 fold) and functional activity of MDR1 (1.2 fold). Rifampin 13-21 ATP binding cassette subfamily B member 1 Homo sapiens 98-102 18332862-6 2008 KEY RESULTS: Rifampin exposure (10 microM, 72 hours) increased MDR1 transcript levels (3.4 fold), MDR1 total protein levels (4.4 fold), apical MDR1 protein (2.7 fold) and functional activity of MDR1 (1.2 fold). Rifampin 13-21 ATP binding cassette subfamily B member 1 Homo sapiens 98-102 18332862-8 2008 Whereas PXR expression was increased by rifampin incubation (2 fold), digoxin reduced PXR expression (0.3 fold). Rifampin 40-48 nuclear receptor subfamily 1 group I member 2 Homo sapiens 8-11 18413659-0 2008 Targeted disruption of murine organic anion-transporting polypeptide 1b2 (Oatp1b2/Slco1b2) significantly alters disposition of prototypical drug substrates pravastatin and rifampin. Rifampin 172-180 solute carrier organic anion transporter family, member 1b2 Mus musculus 82-89 18413659-7 2008 Pharmacological characterization was carried out using two prototypical substrates of human OATP1B1 and -1B3, rifampin and pravastatin. Rifampin 110-118 solute carrier organic anion transporter family member 1B1 Homo sapiens 92-108 18321482-5 2008 They mediate transport of a variety of drugs including the pregnane X receptor ligands rifampicin and dexamethasone. Rifampin 87-97 nuclear receptor subfamily 1 group I member 2 Homo sapiens 59-78 18328680-8 2008 Enzyme activity of CYP3A4, measured by testosterone metabolism, was increased after 24h by RIF. Rifampin 91-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-25 18180276-8 2008 As a result, cyclosporin A and rifampicin were found to have the potential to interact with OATP1B3-mediated uptake at clinical concentrations. Rifampin 31-41 solute carrier organic anion transporter family member 1B3 Homo sapiens 92-99 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 21-31 nuclear receptor subfamily 1 group I member 2 Homo sapiens 39-58 18334920-0 2008 Influence of genetic polymorphisms on intestinal expression and rifampicin-type induction of ABCC2 and on bioavailability of talinolol. Rifampin 64-74 ATP binding cassette subfamily C member 2 Homo sapiens 93-98 18334920-4 2008 Moreover, the effects of rifampicin-type induction (600 mg, 8 days) of duodenal ABCC2 were quantified in 22 participants with regard to genetic polymorphisms. Rifampin 25-35 ATP binding cassette subfamily C member 2 Homo sapiens 80-85 18334920-9 2008 Intestinal ABCC2 mRNA and protein expression were upregulated by rifampicin treatment, a genetic influence could be detected in only four cases heterozygote for 3563T>A or 4544G>A. Rifampin 65-75 ATP binding cassette subfamily C member 2 Homo sapiens 11-16 18096673-5 2008 A-792611 also attenuated the rifampin and ritonavir-mediated activation of the human pregnane X receptor (PXR) in luciferase reporter assays. Rifampin 29-37 nuclear receptor subfamily 1 group I member 2 Homo sapiens 85-104 18096673-5 2008 A-792611 also attenuated the rifampin and ritonavir-mediated activation of the human pregnane X receptor (PXR) in luciferase reporter assays. Rifampin 29-37 nuclear receptor subfamily 1 group I member 2 Homo sapiens 106-109 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 21-31 nuclear receptor subfamily 1 group I member 2 Homo sapiens 60-63 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 21-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-87 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 21-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 21-31 UDP glucuronosyltransferase family 1 member A6 Homo sapiens 117-123 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 21-31 ATP binding cassette subfamily B member 1 Homo sapiens 140-162 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 21-31 ATP binding cassette subfamily B member 1 Homo sapiens 164-168 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 21-31 ATP binding cassette subfamily B member 1 Homo sapiens 170-175 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 21-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 238-244 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 33-36 nuclear receptor subfamily 1 group I member 2 Homo sapiens 39-58 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 33-36 nuclear receptor subfamily 1 group I member 2 Homo sapiens 60-63 18379929-1 2008 The study demonstrated that lipid microspheres (LM) containing rifampicin (LM-RFP) could deliver the drug to alveolar macrophages in vitro and in vivo, and that intranasal administration to animals could achieve preferential accumulation in the lungs with less effect on the liver. Rifampin 63-73 tripartite motif containing 27 Homo sapiens 78-81 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 33-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-87 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 33-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 33-36 UDP glucuronosyltransferase family 1 member A6 Homo sapiens 117-123 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 33-36 ATP binding cassette subfamily B member 1 Homo sapiens 140-162 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 33-36 ATP binding cassette subfamily B member 1 Homo sapiens 164-168 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 33-36 ATP binding cassette subfamily B member 1 Homo sapiens 170-175 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 33-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 238-244 17760873-8 2008 Rifampicin, indomethacin and benzbromarone decreased 5,(6)-carboxy-2",7"-dichlorofluorescein transport by multidrug resistance-associated protein (Mrp)2 as visualized by confocal laser microscopy and in vesicular transport experiments. Rifampin 0-10 ATP binding cassette subfamily C member 2 Rattus norvegicus 106-152 17760873-9 2008 Interestingly, rifampicin decreased the MRP3 activity in vesicular transport experiments using 17-beta-estradiol-17-beta-D-glucuronide as substrate, in contrast to that observed in bilirubin-glucuronide transport experiments. Rifampin 15-25 ATP binding cassette subfamily C member 3 Rattus norvegicus 40-44 17998298-0 2008 Azole antimycotics differentially affect rifampicin-induced pregnane X receptor-mediated CYP3A4 gene expression. Rifampin 41-51 nuclear receptor subfamily 1 group I member 2 Homo sapiens 60-79 21279172-3 2008 The concomitant use of either CYP3A4 inducer rifampicin or CYP3A4 inhibitor erythromycin may influence the results of MEGX test. Rifampin 45-55 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 30-36 17998298-0 2008 Azole antimycotics differentially affect rifampicin-induced pregnane X receptor-mediated CYP3A4 gene expression. Rifampin 41-51 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 89-95 17962516-8 2008 In addition to pregnenolone 16alpha-carbonitrile, several clinically relevant PXR ligands (rifampicin and dexamethasone) all increased ATRA metabolism both in vitro and in vivo, which was PXR-dependent, and up-regulation of Cyp3a was the major contributor. Rifampin 91-101 nuclear receptor subfamily 1, group I, member 2 Mus musculus 78-81 18071298-5 2008 For demonstration of the differential effects of rifampicin on human and rat hepatocytes, induction by rifampicin of cytochrome P450 (CYP) 2E1, a major enzyme associated with isoniazid hepatotoxicity, was detected by 4-nitrocatechol formation and RT-PCR analysis. Rifampin 49-59 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 117-142 18071298-5 2008 For demonstration of the differential effects of rifampicin on human and rat hepatocytes, induction by rifampicin of cytochrome P450 (CYP) 2E1, a major enzyme associated with isoniazid hepatotoxicity, was detected by 4-nitrocatechol formation and RT-PCR analysis. Rifampin 103-113 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 117-142 18071298-9 2008 CONCLUSIONS AND IMPLICATIONS: The difference in induction of CYP 2E1 by rifampicin between rat and human hepatocytes accounted for the difference in exacerbation of isoniazid hepatocyte toxicity by rifampicin, with more significant toxicity in gel entrapment than in monolayer cultures. Rifampin 72-82 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 61-68 18071298-9 2008 CONCLUSIONS AND IMPLICATIONS: The difference in induction of CYP 2E1 by rifampicin between rat and human hepatocytes accounted for the difference in exacerbation of isoniazid hepatocyte toxicity by rifampicin, with more significant toxicity in gel entrapment than in monolayer cultures. Rifampin 198-208 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 61-68 17962516-8 2008 In addition to pregnenolone 16alpha-carbonitrile, several clinically relevant PXR ligands (rifampicin and dexamethasone) all increased ATRA metabolism both in vitro and in vivo, which was PXR-dependent, and up-regulation of Cyp3a was the major contributor. Rifampin 91-101 nuclear receptor subfamily 1, group I, member 2 Mus musculus 188-191 17962516-8 2008 In addition to pregnenolone 16alpha-carbonitrile, several clinically relevant PXR ligands (rifampicin and dexamethasone) all increased ATRA metabolism both in vitro and in vivo, which was PXR-dependent, and up-regulation of Cyp3a was the major contributor. Rifampin 91-101 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 224-229 18783297-9 2008 Using this equation, the ICCYP3A4 was calculated for seven inducers (bosentan, carbamazepine, efavirenz, phenytoin, pioglitazone, rifampicin [rifampin], and St John"s wort [hypericum]) on the basis of the reduction in the AUC of a coadministered standard substrate of CYP3A4, such as simvastatin, in ten DDI studies. Rifampin 130-140 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 27-33 17764444-7 2008 Consistent with the hypothesis, fusion of the far module to the proximal promoter of CYP3A4 markedly increased rifampicin-induced reporter activity. Rifampin 111-121 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 85-91 18783297-9 2008 Using this equation, the ICCYP3A4 was calculated for seven inducers (bosentan, carbamazepine, efavirenz, phenytoin, pioglitazone, rifampicin [rifampin], and St John"s wort [hypericum]) on the basis of the reduction in the AUC of a coadministered standard substrate of CYP3A4, such as simvastatin, in ten DDI studies. Rifampin 142-150 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 27-33 18783297-17 2008 It was indicated that coadministration of rifampicin, phenytoin and carbamazepine may reduce plasma AUCs to less than half for a broad range of CYP3A4 substrate drugs, with CRCYP3A4 values greater than 0.13, 0.21 and 0.33, respectively. Rifampin 42-52 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 144-150 17826359-3 2008 In the present study, we determined the in vivo and in vitro effects of typical mammalian CYP3A inducers (rifampicin, phenobarbital and dexamethasone) on CYP3A-related enzyme activities in a freshwater teleost, the grass carp (Ctenopharyngodon idellus). Rifampin 106-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-95 17925385-3 2008 PXR single nucleotide polymorphisms (SNP) were then genotyped in donor human livers phenotyped for CYP3A4 and multidrug resistance protein 1 mRNA and primary human hepatocytes phenotyped for basal and rifampin-inducible CYP3A4 activity. Rifampin 201-209 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-3 17826359-3 2008 In the present study, we determined the in vivo and in vitro effects of typical mammalian CYP3A inducers (rifampicin, phenobarbital and dexamethasone) on CYP3A-related enzyme activities in a freshwater teleost, the grass carp (Ctenopharyngodon idellus). Rifampin 106-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 154-159 17826359-6 2008 Using erythromycin and testosterone as substrates, we demonstrated that CYP3A catalysis exhibited non-Michaelis-Menten kinetics in GCL cells, and that V(max)/K(m) values were significantly increased due to rifampicin-treatment. Rifampin 206-216 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 72-77 18608530-4 2008 These results show that rifampicin inhibits rapamycin-induced autophagy, at least in part, via the suppression of PP2A activity. Rifampin 24-34 protein phosphatase 2, regulatory subunit A, alpha Mus musculus 114-118 18305373-3 2008 Rif, Dex, and Ome increased SULT2A1 mRNA level in both human and cynomolgus monkey hepatocytes in dose-dependent manner, but not SULT1A1 mRNA level. Rifampin 0-3 sulfotransferase family 2A member 1 Homo sapiens 28-35 18305373-4 2008 Rif, Dex, and Ome increased the mRNA level of UGT1A1 in both human and cynomolgus monkey hepatocytes, Ome more potently in humans and Rif and Ome more potently in monkeys. Rifampin 0-3 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 18305373-4 2008 Rif, Dex, and Ome increased the mRNA level of UGT1A1 in both human and cynomolgus monkey hepatocytes, Ome more potently in humans and Rif and Ome more potently in monkeys. Rifampin 134-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 18031740-5 2008 Rifampicin significantly suppressed HUVEC tube formation and proliferation, and its effects appeared to be mediated at least in part through inhibition of extracellular signal-regulated kinase (ERK) 1/2 phosphorylation. Rifampin 0-10 mitogen-activated protein kinase 3 Mus musculus 155-202 18031740-7 2008 Rifampicin was given subcutaneously at 20mg/kg once a day from immediately after hyperoxia (P12) to P16. Rifampin 0-10 CDK2 (cyclin-dependent kinase 2)-associated protein 1 Mus musculus 92-95 18031740-7 2008 Rifampicin was given subcutaneously at 20mg/kg once a day from immediately after hyperoxia (P12) to P16. Rifampin 0-10 cyclin dependent kinase inhibitor 2A Mus musculus 100-103 18618311-4 2008 Mice treated with LPS alone showed markedly increased plasma levels of TNF, IL-1beta, IL-6, and IL-10, while mice pretreated with rifampicin showed significantly lower plasma levels of these cytokines compared to the LPS alone. Rifampin 130-140 interleukin 1 beta Mus musculus 76-84 18618311-4 2008 Mice treated with LPS alone showed markedly increased plasma levels of TNF, IL-1beta, IL-6, and IL-10, while mice pretreated with rifampicin showed significantly lower plasma levels of these cytokines compared to the LPS alone. Rifampin 130-140 interleukin 6 Mus musculus 86-90 18618311-4 2008 Mice treated with LPS alone showed markedly increased plasma levels of TNF, IL-1beta, IL-6, and IL-10, while mice pretreated with rifampicin showed significantly lower plasma levels of these cytokines compared to the LPS alone. Rifampin 130-140 interleukin 10 Mus musculus 96-101 18094221-0 2008 Effect of rifampin, an inducer of CYP3A and P-glycoprotein, on the pharmacokinetics of risperidone. Rifampin 10-18 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 34-39 17998298-4 2008 We investigated individual effects of the tested azoles as well as their action on rifampicin-induced PXR-mediated transactivation and expression of CYP3A4 in LS174T cell line and primary human hepatocytes, their interactions with PXR ligand-binding domain, and azole-mediated recruitment of SRC-1 to PXR. Rifampin 83-93 nuclear receptor subfamily 1 group I member 2 Homo sapiens 102-105 17998298-4 2008 We investigated individual effects of the tested azoles as well as their action on rifampicin-induced PXR-mediated transactivation and expression of CYP3A4 in LS174T cell line and primary human hepatocytes, their interactions with PXR ligand-binding domain, and azole-mediated recruitment of SRC-1 to PXR. Rifampin 83-93 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 149-155 17998298-4 2008 We investigated individual effects of the tested azoles as well as their action on rifampicin-induced PXR-mediated transactivation and expression of CYP3A4 in LS174T cell line and primary human hepatocytes, their interactions with PXR ligand-binding domain, and azole-mediated recruitment of SRC-1 to PXR. Rifampin 83-93 nuclear receptor subfamily 1 group I member 2 Homo sapiens 231-234 17998298-4 2008 We investigated individual effects of the tested azoles as well as their action on rifampicin-induced PXR-mediated transactivation and expression of CYP3A4 in LS174T cell line and primary human hepatocytes, their interactions with PXR ligand-binding domain, and azole-mediated recruitment of SRC-1 to PXR. Rifampin 83-93 nuclear receptor coactivator 1 Homo sapiens 292-297 17998298-4 2008 We investigated individual effects of the tested azoles as well as their action on rifampicin-induced PXR-mediated transactivation and expression of CYP3A4 in LS174T cell line and primary human hepatocytes, their interactions with PXR ligand-binding domain, and azole-mediated recruitment of SRC-1 to PXR. Rifampin 83-93 nuclear receptor subfamily 1 group I member 2 Homo sapiens 231-234 17998298-5 2008 In addition, applying the pharmacodynamic approach and dose-response analysis, we aimed to describe the nature of potential interactions of tested azole antimycotics coadministered with a prototypical PXR ligand rifampicin in transactivation of CYP3A4 gene. Rifampin 212-222 nuclear receptor subfamily 1 group I member 2 Homo sapiens 201-204 17998298-5 2008 In addition, applying the pharmacodynamic approach and dose-response analysis, we aimed to describe the nature of potential interactions of tested azole antimycotics coadministered with a prototypical PXR ligand rifampicin in transactivation of CYP3A4 gene. Rifampin 212-222 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 245-251 17998298-6 2008 We describe additive and antagonistic interactions of partial and full agonists of PXR nuclear receptor in the therapeutic group of azole antimycotics in rifampicin-mediated transactivation of CYP3A4. Rifampin 154-164 nuclear receptor subfamily 1 group I member 2 Homo sapiens 83-86 17998298-6 2008 We describe additive and antagonistic interactions of partial and full agonists of PXR nuclear receptor in the therapeutic group of azole antimycotics in rifampicin-mediated transactivation of CYP3A4. Rifampin 154-164 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 193-199 18094221-0 2008 Effect of rifampin, an inducer of CYP3A and P-glycoprotein, on the pharmacokinetics of risperidone. Rifampin 10-18 ATP binding cassette subfamily B member 1 Homo sapiens 44-58 18094221-1 2008 The authors studied the effect of rifampin, a dual inducer of CYP3A and P-glycoprotein, on the pharmacokinetics and pharmacodynamics of risperidone in humans. Rifampin 34-42 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-67 18094221-1 2008 The authors studied the effect of rifampin, a dual inducer of CYP3A and P-glycoprotein, on the pharmacokinetics and pharmacodynamics of risperidone in humans. Rifampin 34-42 ATP binding cassette subfamily B member 1 Homo sapiens 72-86 18094221-8 2008 Thus, rifampin reduced the exposure to risperidone, probably because of a decrease in its bioavailability through the induction of CYP3A and probably P-glycoprotein. Rifampin 6-14 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 131-136 18094221-8 2008 Thus, rifampin reduced the exposure to risperidone, probably because of a decrease in its bioavailability through the induction of CYP3A and probably P-glycoprotein. Rifampin 6-14 ATP binding cassette subfamily B member 1 Homo sapiens 150-164 17978482-6 2007 In HFL cells, CYP3A4 and CYP3A7 mRNA expression levels were markedly up-regulated by dexamethasone (DEX), but not by rifampicin (RIF). Rifampin 117-127 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 25-31 17998298-7 2008 We show that oxiconazole is a highly efficacious activator of CYP3A4 transactivation, which could be antagonized by rifampicin in a competitive manner. Rifampin 116-126 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 17998298-8 2008 In addition, we show that activation of the CYP3A4 promoter is a complex process, which is not exclusively determined by azole-PXR interactions, and we suggest that the ability of some azoles to affect recruitment of SRC-1 to PXR modulates their net effects in transactivation of CYP3A4 both in the absence or presence of rifampicin. Rifampin 322-332 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 44-50 17998298-8 2008 In addition, we show that activation of the CYP3A4 promoter is a complex process, which is not exclusively determined by azole-PXR interactions, and we suggest that the ability of some azoles to affect recruitment of SRC-1 to PXR modulates their net effects in transactivation of CYP3A4 both in the absence or presence of rifampicin. Rifampin 322-332 nuclear receptor coactivator 1 Homo sapiens 217-222 17936189-9 2007 Induction (rifampicin) or inhibition (ketoconazole) of alpha-tocopherol metabolism and overexpression of PXR showed that the increase in PXR-related CYP3A4 expression caused by alpha-tocopherol enhanced cell death in hepatocytes. Rifampin 11-21 nuclear receptor subfamily 1 group I member 2 Homo sapiens 137-140 17936189-9 2007 Induction (rifampicin) or inhibition (ketoconazole) of alpha-tocopherol metabolism and overexpression of PXR showed that the increase in PXR-related CYP3A4 expression caused by alpha-tocopherol enhanced cell death in hepatocytes. Rifampin 11-21 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 149-155 18314648-1 2007 INTRODUCTION: Steroid and Xenobiotic Receptor (SXR) has demonstrated its activation by numerous drugs, including cytochrome P450 potent inducers like rifampicina or cotrimazol. Rifampin 150-161 nuclear receptor subfamily 1 group I member 2 Homo sapiens 14-45 18314648-1 2007 INTRODUCTION: Steroid and Xenobiotic Receptor (SXR) has demonstrated its activation by numerous drugs, including cytochrome P450 potent inducers like rifampicina or cotrimazol. Rifampin 150-161 nuclear receptor subfamily 1 group I member 2 Homo sapiens 47-50 17635106-3 2007 Adenoviral-mediated expression of human PXR (hPXR) and its activation by rifampicin strongly repressed cAMP-dependent induction of the endogenous G6Pase gene in Huh7 cells. Rifampin 73-83 nuclear receptor subfamily 1 group I member 2 Homo sapiens 40-43 17635106-3 2007 Adenoviral-mediated expression of human PXR (hPXR) and its activation by rifampicin strongly repressed cAMP-dependent induction of the endogenous G6Pase gene in Huh7 cells. Rifampin 73-83 glucose-6-phosphatase catalytic subunit 1 Homo sapiens 146-152 17978482-8 2007 On the other hand, CYP3A4, CYP3A5, and CYP3A7 mRNA expression levels in HepG2 cells were increased from 2- to 3-fold by treatment with DEX and RIF. Rifampin 143-146 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-25 17978482-8 2007 On the other hand, CYP3A4, CYP3A5, and CYP3A7 mRNA expression levels in HepG2 cells were increased from 2- to 3-fold by treatment with DEX and RIF. Rifampin 143-146 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 27-33 17978482-8 2007 On the other hand, CYP3A4, CYP3A5, and CYP3A7 mRNA expression levels in HepG2 cells were increased from 2- to 3-fold by treatment with DEX and RIF. Rifampin 143-146 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 39-45 17686907-4 2007 Considering that cyclosporin A and rifampicin inhibit multiple uptake/efflux transporters, the interactions of CP-671,305 with major human hepatic drug transporters, multidrug resistance protein 1 (MDR1), multidrug resistance-associated protein 2 (MRP2), breast cancer resistant protein (BCRP), and organic anion-transporting polypeptide (OATPs) were evaluated in vitro. Rifampin 35-45 ATP binding cassette subfamily B member 1 Homo sapiens 198-202 17686907-4 2007 Considering that cyclosporin A and rifampicin inhibit multiple uptake/efflux transporters, the interactions of CP-671,305 with major human hepatic drug transporters, multidrug resistance protein 1 (MDR1), multidrug resistance-associated protein 2 (MRP2), breast cancer resistant protein (BCRP), and organic anion-transporting polypeptide (OATPs) were evaluated in vitro. Rifampin 35-45 ATP binding cassette subfamily C member 2 Homo sapiens 205-246 17686907-4 2007 Considering that cyclosporin A and rifampicin inhibit multiple uptake/efflux transporters, the interactions of CP-671,305 with major human hepatic drug transporters, multidrug resistance protein 1 (MDR1), multidrug resistance-associated protein 2 (MRP2), breast cancer resistant protein (BCRP), and organic anion-transporting polypeptide (OATPs) were evaluated in vitro. Rifampin 35-45 ATP binding cassette subfamily C member 2 Homo sapiens 248-252 17687072-8 2007 The suppressive effect of rifampicin was diminished, whereas the activating effect was augmented, in HepG2 cells with RNA interference-mediated PXR knockdown. Rifampin 26-36 nuclear receptor subfamily 1 group I member 2 Homo sapiens 144-147 17687072-9 2007 These results suggest that HNF4alpha plays a central role in the control of SULT2A1 transcription and that rifampicin-liganded PXR suppresses SULT2A1 expression by interfering with HNF4alpha activity. Rifampin 107-117 nuclear receptor subfamily 1 group I member 2 Homo sapiens 127-130 17687072-0 2007 Positive and negative regulation of human hepatic hydroxysteroid sulfotransferase (SULT2A1) gene transcription by rifampicin: roles of hepatocyte nuclear factor 4alpha and pregnane X receptor. Rifampin 114-124 sulfotransferase family 2A member 1 Homo sapiens 83-90 17687072-1 2007 The effects of rifampicin treatment on SULT2A1 mRNA expression were evaluated in 23 preparations of primary cultured human hepatocytes. Rifampin 15-25 sulfotransferase family 2A member 1 Homo sapiens 39-46 17687072-9 2007 These results suggest that HNF4alpha plays a central role in the control of SULT2A1 transcription and that rifampicin-liganded PXR suppresses SULT2A1 expression by interfering with HNF4alpha activity. Rifampin 107-117 sulfotransferase family 2A member 1 Homo sapiens 142-149 17687072-2 2007 In contrast to the consistently occurring induction of CYP3A4, a prototypical pregnane X receptor (PXR) target gene, rifampicin treatment increased SULT2A1 mRNA levels in 12 of the hepatocyte preparations, but it produced little change or even suppression in the others. Rifampin 117-127 nuclear receptor subfamily 1 group I member 2 Homo sapiens 78-97 17687072-9 2007 These results suggest that HNF4alpha plays a central role in the control of SULT2A1 transcription and that rifampicin-liganded PXR suppresses SULT2A1 expression by interfering with HNF4alpha activity. Rifampin 107-117 hepatocyte nuclear factor 4 alpha Homo sapiens 181-190 17687072-2 2007 In contrast to the consistently occurring induction of CYP3A4, a prototypical pregnane X receptor (PXR) target gene, rifampicin treatment increased SULT2A1 mRNA levels in 12 of the hepatocyte preparations, but it produced little change or even suppression in the others. Rifampin 117-127 nuclear receptor subfamily 1 group I member 2 Homo sapiens 99-102 17687072-2 2007 In contrast to the consistently occurring induction of CYP3A4, a prototypical pregnane X receptor (PXR) target gene, rifampicin treatment increased SULT2A1 mRNA levels in 12 of the hepatocyte preparations, but it produced little change or even suppression in the others. Rifampin 117-127 sulfotransferase family 2A member 1 Homo sapiens 148-155 17687072-3 2007 Transient transfection of HepG2 cells with a series of reporter constructs implicated two SULT2A1 5"-flanking regions as containing rifampicin-responsive information. Rifampin 132-142 sulfotransferase family 2A member 1 Homo sapiens 90-97 17687072-10 2007 By contrast, the rifampicin-inducible SULT2A1 expression that occurs in many human hepatocyte preparations seems to be mediated through a PXR-independent mechanism. Rifampin 17-27 sulfotransferase family 2A member 1 Homo sapiens 38-45 17687072-6 2007 Cotransfection of HepG2 cells with pregnane X receptor (PXR) dose-dependently suppressed reporter expression from SULT2A1 constructs containing the HNF4 sites, and rifampicin treatment augmented the suppression. Rifampin 164-174 nuclear receptor subfamily 1 group I member 2 Homo sapiens 35-54 17687072-10 2007 By contrast, the rifampicin-inducible SULT2A1 expression that occurs in many human hepatocyte preparations seems to be mediated through a PXR-independent mechanism. Rifampin 17-27 nuclear receptor subfamily 1 group I member 2 Homo sapiens 138-141 17687072-7 2007 Rifampicin treatment concentration-dependently suppressed SULT2A1 reporter expression at the same concentrations that progressively induced expression from a PXR-responsive CYP3A4 reporter, whereas higher rifampicin concentrations reversed the SULT2A1 suppression. Rifampin 0-10 sulfotransferase family 2A member 1 Homo sapiens 58-65 17687072-7 2007 Rifampicin treatment concentration-dependently suppressed SULT2A1 reporter expression at the same concentrations that progressively induced expression from a PXR-responsive CYP3A4 reporter, whereas higher rifampicin concentrations reversed the SULT2A1 suppression. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 158-161 17687072-7 2007 Rifampicin treatment concentration-dependently suppressed SULT2A1 reporter expression at the same concentrations that progressively induced expression from a PXR-responsive CYP3A4 reporter, whereas higher rifampicin concentrations reversed the SULT2A1 suppression. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 173-179 17687072-7 2007 Rifampicin treatment concentration-dependently suppressed SULT2A1 reporter expression at the same concentrations that progressively induced expression from a PXR-responsive CYP3A4 reporter, whereas higher rifampicin concentrations reversed the SULT2A1 suppression. Rifampin 0-10 sulfotransferase family 2A member 1 Homo sapiens 244-251 17687072-7 2007 Rifampicin treatment concentration-dependently suppressed SULT2A1 reporter expression at the same concentrations that progressively induced expression from a PXR-responsive CYP3A4 reporter, whereas higher rifampicin concentrations reversed the SULT2A1 suppression. Rifampin 205-215 sulfotransferase family 2A member 1 Homo sapiens 244-251 17696253-8 2007 Overexpression of PXR and its dimerization partner retinoid X receptor (RXR) and stimulation with LCA or the potent PXR ligand rifampicin leads to a significant induction of FGF19 promoter activity in intestinal cells. Rifampin 127-137 nuclear receptor subfamily 1 group I member 2 Homo sapiens 18-21 18183335-7 2007 Targeting for rapid drug resistance detection, in hospitalized patients at "Servico de Pneumologia 2 do Hospital Pulido Valente", the test INNO-LIPA Rif.TB, to identify the rifampicin resistance as a marker of multi-drug resistance, was evaluated. Rifampin 173-183 lipase A, lysosomal acid type Homo sapiens 144-148 17639026-5 2007 The induction by rifampin of CYP3A activity was significantly correlated with its induction of CYP3A4 transcripts (r = 0.96, p < 0.05) and CYP3A protein (r = 0.89, p < 0.05). Rifampin 17-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 95-100 17768708-1 2007 In neutral medium, rifamycin antibiotics such as rifapentin (RFPT), rifampicin (RFP), rifandin (RFD) and rifamycin SV (RFSV) can bind with human serum albumin (HSA) and bovine serum albumin (BSA) to form complexes, resulting in the quenching of the intrinsic fluorescence (lambda(ex)/lambda(em) = 285/355 nm) of the BSA and HSA. Rifampin 68-78 albumin Homo sapiens 145-158 17768708-1 2007 In neutral medium, rifamycin antibiotics such as rifapentin (RFPT), rifampicin (RFP), rifandin (RFD) and rifamycin SV (RFSV) can bind with human serum albumin (HSA) and bovine serum albumin (BSA) to form complexes, resulting in the quenching of the intrinsic fluorescence (lambda(ex)/lambda(em) = 285/355 nm) of the BSA and HSA. Rifampin 68-78 albumin Homo sapiens 176-189 17696253-8 2007 Overexpression of PXR and its dimerization partner retinoid X receptor (RXR) and stimulation with LCA or the potent PXR ligand rifampicin leads to a significant induction of FGF19 promoter activity in intestinal cells. Rifampin 127-137 retinoid X receptor alpha Homo sapiens 72-75 17696253-8 2007 Overexpression of PXR and its dimerization partner retinoid X receptor (RXR) and stimulation with LCA or the potent PXR ligand rifampicin leads to a significant induction of FGF19 promoter activity in intestinal cells. Rifampin 127-137 nuclear receptor subfamily 1 group I member 2 Homo sapiens 116-119 17696253-8 2007 Overexpression of PXR and its dimerization partner retinoid X receptor (RXR) and stimulation with LCA or the potent PXR ligand rifampicin leads to a significant induction of FGF19 promoter activity in intestinal cells. Rifampin 127-137 fibroblast growth factor 19 Homo sapiens 174-179 17496208-0 2007 Bosentan is a substrate of human OATP1B1 and OATP1B3: inhibition of hepatic uptake as the common mechanism of its interactions with cyclosporin A, rifampicin, and sildenafil. Rifampin 147-157 solute carrier organic anion transporter family member 1B1 Homo sapiens 33-40 17624828-3 2007 First, induction of cytochrome P-450 enzymes and P-glycoprotein by rifampin results in reduced concentrations of nonnucleoside reverse-transcriptase inhibitors and, particularly, protease inhibitors. Rifampin 67-75 ATP binding cassette subfamily B member 1 Homo sapiens 49-63 17496208-5 2007 Bosentan uptake into Chinese hamster ovary cells expressing these OATP transporters was efficiently inhibited by cyclosporin A and rifampicin with IC(50) values significantly below their effective plasma concentrations in humans. Rifampin 131-141 solute carrier organic anion transporter family member 1A2 Homo sapiens 66-70 17635174-13 2007 GSE occur frequently in patients receiving rifampicin for osteoarticular infection but without an association with rifampicin plasma concentrations. Rifampin 43-53 CELIAC2 Homo sapiens 0-3 17553741-0 2007 Rifampicin-induced CYP3A4 activation in CTX patients cannot replace chenodeoxycholic acid treatment. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-25 17553741-6 2007 Rifampicin (600 mg/day x 7 days), known to induce the PXR, and thereby to increase CYP3A activity, was used. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 54-57 17553741-6 2007 Rifampicin (600 mg/day x 7 days), known to induce the PXR, and thereby to increase CYP3A activity, was used. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-88 17553741-7 2007 The degree of CYP3A4 induction was assessed by comparing midazolam pharmacokinetics before and after rifampicin treatment. Rifampin 101-111 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-20 17553741-9 2007 The results show that despite 60% increase in CYP3A4 activity following rifampicin treatment, there is no significant change in cholestanol levels. Rifampin 72-82 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 46-52 17392393-11 2007 Furthermore, we suggest that VPA synergistically augments the effect of rifampicin in transactivation of CYP3A4 in primary human hepatocytes. Rifampin 72-82 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-111 17510066-2 2007 Exposure to some xenobiotics, such as the antibiotic rifampicin, increases P-gp expression. Rifampin 53-63 ATP binding cassette subfamily B member 1 Homo sapiens 75-79 17434476-7 2007 A significant augmentation in the expression and activity of CYP1A2 and CYP2E1 was observed following pyrogallol and rifampicin+pyrogallol treatment; however, rifampicin exhibited a significant induction of CYP2E1 only. Rifampin 117-127 cytochrome P450, family 1, subfamily a, polypeptide 2 Mus musculus 61-67 17434476-7 2007 A significant augmentation in the expression and activity of CYP1A2 and CYP2E1 was observed following pyrogallol and rifampicin+pyrogallol treatment; however, rifampicin exhibited a significant induction of CYP2E1 only. Rifampin 117-127 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 72-78 17434476-7 2007 A significant augmentation in the expression and activity of CYP1A2 and CYP2E1 was observed following pyrogallol and rifampicin+pyrogallol treatment; however, rifampicin exhibited a significant induction of CYP2E1 only. Rifampin 159-169 cytochrome P450, family 1, subfamily a, polypeptide 2 Mus musculus 61-67 17434476-7 2007 A significant augmentation in the expression and activity of CYP1A2 and CYP2E1 was observed following pyrogallol and rifampicin+pyrogallol treatment; however, rifampicin exhibited a significant induction of CYP2E1 only. Rifampin 159-169 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 72-78 17434476-7 2007 A significant augmentation in the expression and activity of CYP1A2 and CYP2E1 was observed following pyrogallol and rifampicin+pyrogallol treatment; however, rifampicin exhibited a significant induction of CYP2E1 only. Rifampin 159-169 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 207-213 17434476-8 2007 Attenuation of glutathione-S-transferase, glutathione reductase and glutathione peroxidase activities and augmentation of lipid peroxidation were observed following rifampicin and/or pyrogallol treatment and a cumulative effect was seen when the two drugs were administered in combination. Rifampin 165-175 hematopoietic prostaglandin D synthase Mus musculus 15-40 17434476-8 2007 Attenuation of glutathione-S-transferase, glutathione reductase and glutathione peroxidase activities and augmentation of lipid peroxidation were observed following rifampicin and/or pyrogallol treatment and a cumulative effect was seen when the two drugs were administered in combination. Rifampin 165-175 glutathione reductase Mus musculus 42-63 17434476-9 2007 Silymarin restored the rifampicin- and/or pyrogallol-induced alterations in the expression and activity of CYP1A2 and CYP2E1, the activity of glutathione-S-transferase, glutathione reductase, and glutathione peroxidase, and lipid peroxidation. Rifampin 23-33 cytochrome P450, family 1, subfamily a, polypeptide 2 Mus musculus 107-113 17434476-9 2007 Silymarin restored the rifampicin- and/or pyrogallol-induced alterations in the expression and activity of CYP1A2 and CYP2E1, the activity of glutathione-S-transferase, glutathione reductase, and glutathione peroxidase, and lipid peroxidation. Rifampin 23-33 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 118-124 17434476-9 2007 Silymarin restored the rifampicin- and/or pyrogallol-induced alterations in the expression and activity of CYP1A2 and CYP2E1, the activity of glutathione-S-transferase, glutathione reductase, and glutathione peroxidase, and lipid peroxidation. Rifampin 23-33 hematopoietic prostaglandin D synthase Mus musculus 142-167 17434476-9 2007 Silymarin restored the rifampicin- and/or pyrogallol-induced alterations in the expression and activity of CYP1A2 and CYP2E1, the activity of glutathione-S-transferase, glutathione reductase, and glutathione peroxidase, and lipid peroxidation. Rifampin 23-33 glutathione reductase Mus musculus 169-190 17434476-10 2007 The results demonstrate the role of CYP1A2, CYP2E1, glutathione-S-transferase, glutathione reductase and glutathione peroxidase in rifampicin- and pyrogallol-induced hepatotoxicity and provide evidence for the involvement of silymarin in attenuation of drug-induced hepatotoxicity. Rifampin 131-141 cytochrome P450, family 1, subfamily a, polypeptide 2 Mus musculus 36-42 17434476-10 2007 The results demonstrate the role of CYP1A2, CYP2E1, glutathione-S-transferase, glutathione reductase and glutathione peroxidase in rifampicin- and pyrogallol-induced hepatotoxicity and provide evidence for the involvement of silymarin in attenuation of drug-induced hepatotoxicity. Rifampin 131-141 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 44-50 17434476-10 2007 The results demonstrate the role of CYP1A2, CYP2E1, glutathione-S-transferase, glutathione reductase and glutathione peroxidase in rifampicin- and pyrogallol-induced hepatotoxicity and provide evidence for the involvement of silymarin in attenuation of drug-induced hepatotoxicity. Rifampin 131-141 hematopoietic prostaglandin D synthase Mus musculus 52-77 17434476-10 2007 The results demonstrate the role of CYP1A2, CYP2E1, glutathione-S-transferase, glutathione reductase and glutathione peroxidase in rifampicin- and pyrogallol-induced hepatotoxicity and provide evidence for the involvement of silymarin in attenuation of drug-induced hepatotoxicity. Rifampin 131-141 glutathione reductase Mus musculus 79-100 17198732-11 2007 However, 11/14 (79%) MRSA patients who received rifampin combinations, other than vancomycin and rifampin simultaneously, were apparently cured. Rifampin 48-56 solute carrier family 9 member A6 Homo sapiens 21-25 17639026-4 2007 At 10 microM, rifampin induced CYP3A4 transcripts, CYP3A protein, and activity by 23-, 12-, and 13-fold, respectively. Rifampin 14-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 17639026-4 2007 At 10 microM, rifampin induced CYP3A4 transcripts, CYP3A protein, and activity by 23-, 12-, and 13-fold, respectively. Rifampin 14-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-36 17639026-5 2007 The induction by rifampin of CYP3A activity was significantly correlated with its induction of CYP3A4 transcripts (r = 0.96, p < 0.05) and CYP3A protein (r = 0.89, p < 0.05). Rifampin 17-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-34 17639026-5 2007 The induction by rifampin of CYP3A activity was significantly correlated with its induction of CYP3A4 transcripts (r = 0.96, p < 0.05) and CYP3A protein (r = 0.89, p < 0.05). Rifampin 17-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 95-101 17361125-2 2007 This study examines whether presumed changes in CYP3A4 activity are in fact related to inhibition of an uptake organic anion transporter using rifampin and inhibition of the efflux hepatic P-glycoprotein transporter using lansoprazole. Rifampin 143-151 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-54 17369605-7 2007 Using HEK293-OATP1B3 cells, the cytotoxicity was attenuated by substrates and inhibitors of OATP1B3, including bromosulfophthalein, rifampicin, and cyclosporin A. Rifampin 132-142 solute carrier organic anion transporter family member 1B3 Homo sapiens 13-20 17369605-7 2007 Using HEK293-OATP1B3 cells, the cytotoxicity was attenuated by substrates and inhibitors of OATP1B3, including bromosulfophthalein, rifampicin, and cyclosporin A. Rifampin 132-142 solute carrier organic anion transporter family member 1B3 Homo sapiens 92-99 17293382-5 2007 Surprisingly, pyributicarb was found to increase the CYP3A4 reporter activity at 0.1 to 1 microM more strongly than typical CYP3A4 inducer rifampicin. Rifampin 139-149 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 124-130 17207564-8 2007 HepG2 CYP3A4 activity was unaffected by 48 h DEX pretreatment; therefore, studies were done in DPX-2 cells, a HepG2 derivative engineered to overexpress pregnane-X receptor (PXR) that exhibits rifampicin (RIF)-inducible endogenous CYP3A4. Rifampin 193-203 nuclear receptor subfamily 1 group I member 2 Homo sapiens 153-172 17207564-10 2007 CYP3A4 activity of DPX-2 cells treated with RIF (10 microM, 48 h) was twice that of untreated cells, but RIF did not increase alachlor toxicity. Rifampin 44-47 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 17429319-4 2007 Instead, as shown using a mammalian two-hybrid assay, it decreased the interaction of pregnane X receptor with steroid receptor coactivator-1 in the presence of rifampin, clotrimazole, paclitaxel, or nifedipine but not in their absence. Rifampin 161-169 nuclear receptor subfamily 1 group I member 2 Homo sapiens 86-105 17429319-4 2007 Instead, as shown using a mammalian two-hybrid assay, it decreased the interaction of pregnane X receptor with steroid receptor coactivator-1 in the presence of rifampin, clotrimazole, paclitaxel, or nifedipine but not in their absence. Rifampin 161-169 nuclear receptor coactivator 1 Homo sapiens 111-141 17429319-5 2007 Rifampin treatment markedly increased pregnane X receptor protein in the wild-type pregnane X receptor-transfected cells as shown by coimmunoprecipitation but not in Q158K pregnane X receptor-transfected cells. Rifampin 0-8 nuclear receptor subfamily 1 group I member 2 Homo sapiens 38-57 17429319-5 2007 Rifampin treatment markedly increased pregnane X receptor protein in the wild-type pregnane X receptor-transfected cells as shown by coimmunoprecipitation but not in Q158K pregnane X receptor-transfected cells. Rifampin 0-8 nuclear receptor subfamily 1 group I member 2 Homo sapiens 83-102 17429319-5 2007 Rifampin treatment markedly increased pregnane X receptor protein in the wild-type pregnane X receptor-transfected cells as shown by coimmunoprecipitation but not in Q158K pregnane X receptor-transfected cells. Rifampin 0-8 nuclear receptor subfamily 1 group I member 2 Homo sapiens 83-102 17381666-0 2007 Rifampin, a cytochrome P450 3A inducer, decreases plasma concentrations of antipsychotic risperidone in healthy volunteers. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 17381666-2 2007 Rifampin, a potent CYP3A inducer, has been known to markedly decrease plasma concentrations of various drugs, which are concomitantly administered during treatment. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-24 17389551-1 2007 The effect of rifampin, a cytochrome P450 3A4 inducer, on the pharmacokinetics of atrasentan was assessed in 12 healthy male subjects in an open-label study. Rifampin 14-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 26-45 17470658-11 2007 However; the Cox regression analysis showed that rifampin was the only independent predictor associated with a lower risk of thrombocytopenia (hazard ratio, 0.37; 95% confidence interval, 0.14 to 0.98; P = 0.045). Rifampin 49-57 cytochrome c oxidase subunit 8A Homo sapiens 13-16 17259447-13 2007 Changes in plasma R/S methadone ratios observed after rifampin or troleandomycin pretreatment in humans in vivo were successfully predicted by CYP2B6- but not CYP3A4-catalyzed methadone N-demethylation. Rifampin 54-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 143-149 17093002-5 2007 Treatment of PXR-humanized mice with the PXR ligands mimicked the human response, since both hepatic and intestinal CYP3As were strongly induced by rifampicin, a human-specific PXR ligand, but not by pregnenolone 16alpha-carbonitrile, a rodent-specific PXR ligand. Rifampin 148-158 nuclear receptor subfamily 1, group I, member 2 Mus musculus 13-16 17251982-7 2007 Rifampin potently inhibited the uptake of bosentan into cells expressing human OATP1B1 and -1B3. Rifampin 0-8 solute carrier organic anion transporter family member 1B1 Homo sapiens 79-95 17280646-0 2007 Rifampicin protects PC12 cells against MPP+-induced apoptosis and inhibits the expression of an alpha-Synuclein multimer. Rifampin 0-10 synuclein alpha Rattus norvegicus 96-111 17280646-5 2007 The expression of the higher molecular mass alpha-Synuclein was increased by MPP(+) exposure, and its expression was inversely related to cell survival in the rifampicin-treated cells. Rifampin 159-169 synuclein alpha Rattus norvegicus 44-59 17280646-6 2007 Importantly, rifampicin suppressed apoptosis almost completely, without shifting the death cascade to necrosis, which is a problem that has been reported with caspase inhibitors of apoptosis (Hartmann, A., Troadec, J.D., Hunot, S., Kikly, K., Faucheux, B.A., Mouatt-Prigent, A., Ruberg, M. Agid, Y., Hirsch, E.C., 2001. Rifampin 13-23 caspase 8 Rattus norvegicus 159-166 17280646-10 2007 These results suggest that rifampicin improves survival of catecholamine- and alpha-Synuclein-containing cells, which degenerate in Parkinson"s disease (PD), and thus may be therapeutic in this disease. Rifampin 27-37 synuclein alpha Rattus norvegicus 78-93 17088262-6 2007 A human PXR ligand, rifampicin, induced CYP27A1 mRNA expression in intestine cells but not in liver cells. Rifampin 20-30 nuclear receptor subfamily 1 group I member 2 Homo sapiens 8-11 17088262-6 2007 A human PXR ligand, rifampicin, induced CYP27A1 mRNA expression in intestine cells but not in liver cells. Rifampin 20-30 cytochrome P450 family 27 subfamily A member 1 Homo sapiens 40-47 17093002-5 2007 Treatment of PXR-humanized mice with the PXR ligands mimicked the human response, since both hepatic and intestinal CYP3As were strongly induced by rifampicin, a human-specific PXR ligand, but not by pregnenolone 16alpha-carbonitrile, a rodent-specific PXR ligand. Rifampin 148-158 nuclear receptor subfamily 1, group I, member 2 Mus musculus 41-44 17088262-7 2007 Rifampicin induced CYP27A1 gene transcription, increased intracellular 27-HOC levels, and induced ABCA1 and ABCG1 mRNA expression only in intestine cells. Rifampin 0-10 cytochrome P450 family 27 subfamily A member 1 Homo sapiens 19-26 17093002-5 2007 Treatment of PXR-humanized mice with the PXR ligands mimicked the human response, since both hepatic and intestinal CYP3As were strongly induced by rifampicin, a human-specific PXR ligand, but not by pregnenolone 16alpha-carbonitrile, a rodent-specific PXR ligand. Rifampin 148-158 peptidylprolyl isomerase F Homo sapiens 116-120 17088262-7 2007 Rifampicin induced CYP27A1 gene transcription, increased intracellular 27-HOC levels, and induced ABCA1 and ABCG1 mRNA expression only in intestine cells. Rifampin 0-10 ATP binding cassette subfamily A member 1 Homo sapiens 98-103 17093002-5 2007 Treatment of PXR-humanized mice with the PXR ligands mimicked the human response, since both hepatic and intestinal CYP3As were strongly induced by rifampicin, a human-specific PXR ligand, but not by pregnenolone 16alpha-carbonitrile, a rodent-specific PXR ligand. Rifampin 148-158 nuclear receptor subfamily 1 group I member 2 Homo sapiens 41-44 17088262-7 2007 Rifampicin induced CYP27A1 gene transcription, increased intracellular 27-HOC levels, and induced ABCA1 and ABCG1 mRNA expression only in intestine cells. Rifampin 0-10 ATP binding cassette subfamily G member 1 Homo sapiens 108-113 17088262-9 2007 Chromatin immunoprecipitation assays revealed that rifampicin induced the PXR recruitment of steroid receptor coactivator 1 to CYP27A1 chromatin. Rifampin 51-61 nuclear receptor subfamily 1 group I member 2 Homo sapiens 74-77 17093002-5 2007 Treatment of PXR-humanized mice with the PXR ligands mimicked the human response, since both hepatic and intestinal CYP3As were strongly induced by rifampicin, a human-specific PXR ligand, but not by pregnenolone 16alpha-carbonitrile, a rodent-specific PXR ligand. Rifampin 148-158 nuclear receptor subfamily 1 group I member 2 Homo sapiens 41-44 17088262-9 2007 Chromatin immunoprecipitation assays revealed that rifampicin induced the PXR recruitment of steroid receptor coactivator 1 to CYP27A1 chromatin. Rifampin 51-61 nuclear receptor coactivator 1 Homo sapiens 93-123 17088262-9 2007 Chromatin immunoprecipitation assays revealed that rifampicin induced the PXR recruitment of steroid receptor coactivator 1 to CYP27A1 chromatin. Rifampin 51-61 cytochrome P450 family 27 subfamily A member 1 Homo sapiens 127-134 17093002-6 2007 In rifampicin-pretreated PXR-humanized mice, an approximately 60% decrease was observed for both the maximal midazolam serum concentration (C(max)) and the area under the concentration-time curve, as a result of a 3-fold increase in midazolam 1"-hydroxylation. Rifampin 3-13 nuclear receptor subfamily 1, group I, member 2 Mus musculus 25-28 17088262-11 2007 Rifampicin, 27-HOC, and a potent LXRalpha agonist, T0901317, induced ABCA1 and ABCG1 protein expression and stimulated cholesterol efflux from intestine cells to apolipoprotein A-I and HDL. Rifampin 0-10 ATP binding cassette subfamily A member 1 Homo sapiens 69-74 17069749-6 2007 NDGA and RIF inhibited the binding of Abeta to GM1 liposomes by competitively binding to the membranes and/or direct interaction with Abeta in solution, thus at least partly preventing fibrils from forming. Rifampin 9-12 amyloid beta precursor protein Rattus norvegicus 38-43 17088262-11 2007 Rifampicin, 27-HOC, and a potent LXRalpha agonist, T0901317, induced ABCA1 and ABCG1 protein expression and stimulated cholesterol efflux from intestine cells to apolipoprotein A-I and HDL. Rifampin 0-10 ATP binding cassette subfamily G member 1 Homo sapiens 79-84 17088262-11 2007 Rifampicin, 27-HOC, and a potent LXRalpha agonist, T0901317, induced ABCA1 and ABCG1 protein expression and stimulated cholesterol efflux from intestine cells to apolipoprotein A-I and HDL. Rifampin 0-10 apolipoprotein A1 Homo sapiens 162-180 17108049-7 2007 In addition, BKRF3 was able to complement an E. coli ung mutant in rifampin and nalidixic acid resistance mutator assays. Rifampin 67-75 uracil-DNA glycosylase Human gammaherpesvirus 4 13-18 17108049-7 2007 In addition, BKRF3 was able to complement an E. coli ung mutant in rifampin and nalidixic acid resistance mutator assays. Rifampin 67-75 uracil DNA glycosylase Homo sapiens 53-56 17069749-6 2007 NDGA and RIF inhibited the binding of Abeta to GM1 liposomes by competitively binding to the membranes and/or direct interaction with Abeta in solution, thus at least partly preventing fibrils from forming. Rifampin 9-12 amyloid beta precursor protein Rattus norvegicus 134-139 17936928-10 2007 Since species differences exist in ligand specificity between human and mice, a PXR-humanized mouse (hPXR) was produced that responds to human PXR activators such as rifampicin but does not respond to the rodent activator pregnenalone 16alpha-carbonitrile. Rifampin 166-176 nuclear receptor subfamily 1, group I, member 2 Mus musculus 80-83 17253885-4 2007 For some time it has been known that inducers of drug-metabolising enzymes (such as the CYP3A4 inducer rifampicin [rifampin]) can lead to breakthrough bleeding and contraceptive failure. Rifampin 103-113 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-94 17253885-4 2007 For some time it has been known that inducers of drug-metabolising enzymes (such as the CYP3A4 inducer rifampicin [rifampin]) can lead to breakthrough bleeding and contraceptive failure. Rifampin 115-123 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-94 17936928-10 2007 Since species differences exist in ligand specificity between human and mice, a PXR-humanized mouse (hPXR) was produced that responds to human PXR activators such as rifampicin but does not respond to the rodent activator pregnenalone 16alpha-carbonitrile. Rifampin 166-176 nuclear receptor subfamily 1 group I member 2 Homo sapiens 102-105 17365992-2 2007 The aim of this study was to examine Pgp activity in relation to age, gender, medical treatment (rifampicin or ketoconazole) and the multidrug resistance (MDR1) gene single nucleotide polymorphisms (SNPs) G2677T and C3435T using the model drug digoxin. Rifampin 97-107 ATP binding cassette subfamily B member 1 Homo sapiens 37-40 16973756-3 2007 Activation of PXR by genetic (transgene) or pharmacological (ligand, such as rifampicin) markedly increased plasma concentrations of corticosterone and aldosterone, the respective primary glucocorticoid and mineralocorticoid in rodents. Rifampin 77-87 nuclear receptor subfamily 1, group I, member 2 Mus musculus 14-17 17365992-7 2007 RESULTS: Rifampicin treatment resulted in increased Pgp activity, duodenal MDR1 mRNA expression and Pgp detection compared with that in the control group (p<0.05 for all), Pgp activity being associated with duodenal MDR1 mRNA level (p<0.05). Rifampin 9-19 ATP binding cassette subfamily B member 1 Homo sapiens 52-55 17365992-7 2007 RESULTS: Rifampicin treatment resulted in increased Pgp activity, duodenal MDR1 mRNA expression and Pgp detection compared with that in the control group (p<0.05 for all), Pgp activity being associated with duodenal MDR1 mRNA level (p<0.05). Rifampin 9-19 ATP binding cassette subfamily B member 1 Homo sapiens 75-79 17365992-7 2007 RESULTS: Rifampicin treatment resulted in increased Pgp activity, duodenal MDR1 mRNA expression and Pgp detection compared with that in the control group (p<0.05 for all), Pgp activity being associated with duodenal MDR1 mRNA level (p<0.05). Rifampin 9-19 ATP binding cassette subfamily B member 1 Homo sapiens 100-103 17365992-7 2007 RESULTS: Rifampicin treatment resulted in increased Pgp activity, duodenal MDR1 mRNA expression and Pgp detection compared with that in the control group (p<0.05 for all), Pgp activity being associated with duodenal MDR1 mRNA level (p<0.05). Rifampin 9-19 ATP binding cassette subfamily B member 1 Homo sapiens 100-103 17365992-7 2007 RESULTS: Rifampicin treatment resulted in increased Pgp activity, duodenal MDR1 mRNA expression and Pgp detection compared with that in the control group (p<0.05 for all), Pgp activity being associated with duodenal MDR1 mRNA level (p<0.05). Rifampin 9-19 ATP binding cassette subfamily B member 1 Homo sapiens 219-223 17365992-10 2007 CONCLUSIONS: Our data confirm that rifampicin increases Pgp activity, by increasing MDR1 mRNA and Pgp levels. Rifampin 35-45 ATP binding cassette subfamily B member 1 Homo sapiens 56-59 17365992-10 2007 CONCLUSIONS: Our data confirm that rifampicin increases Pgp activity, by increasing MDR1 mRNA and Pgp levels. Rifampin 35-45 ATP binding cassette subfamily B member 1 Homo sapiens 84-88 17365992-10 2007 CONCLUSIONS: Our data confirm that rifampicin increases Pgp activity, by increasing MDR1 mRNA and Pgp levels. Rifampin 35-45 ATP binding cassette subfamily B member 1 Homo sapiens 98-101 17112804-11 2006 CONCLUSIONS: Coadministration of rifampin increases the maximum serum concentrations of ezetimibe but reduces its enterosystemic recycling, most likely by inhibition of the secretion of ezetimibe and its glucuronide via P-gp and MRP2. Rifampin 33-41 ATP binding cassette subfamily B member 1 Homo sapiens 220-224 16849627-5 2006 Rifampicin prevents hIAPP amyloid fibril formation and has been proposed as a potential target for prevention of T2DM. Rifampin 0-10 islet amyloid polypeptide Homo sapiens 20-25 16849627-7 2006 We report that rifampicin (Acocella G. Clin Pharmacokinet 3: 108-127, 1978) prevents hIAPP fibril formation, but not formation of toxic hIAPP oligomers (Bates G. Lancet 361: 1642-1644, 2003), and does not protect beta-cells from apoptosis induced by either overexpression or application of hIAPP. Rifampin 15-25 islet amyloid polypeptide Homo sapiens 85-90 17220562-9 2006 CYP3A4 mRNA expression was increased significantly by rifampicin (Rif) exposure in human hepatocytes, whereas Rif-induced increases in CYP3A4 mRNA expression in chimeric mouse hepatocytes was seen for two of the three donors. Rifampin 54-64 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 17220562-9 2006 CYP3A4 mRNA expression was increased significantly by rifampicin (Rif) exposure in human hepatocytes, whereas Rif-induced increases in CYP3A4 mRNA expression in chimeric mouse hepatocytes was seen for two of the three donors. Rifampin 66-69 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 17220562-9 2006 CYP3A4 mRNA expression was increased significantly by rifampicin (Rif) exposure in human hepatocytes, whereas Rif-induced increases in CYP3A4 mRNA expression in chimeric mouse hepatocytes was seen for two of the three donors. Rifampin 110-113 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 135-141 17112807-12 2006 CONCLUSION: This study demonstrates an interaction between mycophenolate mofetil and rifampin, which is a result of induction of MPA glucuronidation and possibly also rifampin-associated alterations in MRP2-mediated transport of MPAG and AcMPAG. Rifampin 85-93 ATP binding cassette subfamily C member 2 Homo sapiens 202-206 17112807-12 2006 CONCLUSION: This study demonstrates an interaction between mycophenolate mofetil and rifampin, which is a result of induction of MPA glucuronidation and possibly also rifampin-associated alterations in MRP2-mediated transport of MPAG and AcMPAG. Rifampin 167-175 ATP binding cassette subfamily C member 2 Homo sapiens 202-206 17112804-11 2006 CONCLUSIONS: Coadministration of rifampin increases the maximum serum concentrations of ezetimibe but reduces its enterosystemic recycling, most likely by inhibition of the secretion of ezetimibe and its glucuronide via P-gp and MRP2. Rifampin 33-41 ATP binding cassette subfamily C member 2 Homo sapiens 229-233 16632523-2 2006 Rifampin, the classical activator of CYP3A4, binds to SXR directly. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-43 16952291-0 2006 Identification of a novel 974C-->G nonsense mutation of the MRP2/ABCC2 gene in a patient with Dubin-Johnson syndrome and analysis of the effects of rifampicin and ursodeoxycholic acid on serum bilirubin and bile acids. Rifampin 148-158 ATP binding cassette subfamily C member 2 Homo sapiens 60-64 16837569-4 2006 With a threshold corresponding to a 1.5-fold factor change in mRNA levels, observed in at least three of seven independent human hepatocyte cultures, efflux transporters such as MDR1, MRP2 and BCRP were up-regulated by PB, RIF, and OPZ, whereas MRP3 was induced by OPZ and RIF. Rifampin 223-226 ATP binding cassette subfamily B member 1 Homo sapiens 178-182 16837569-4 2006 With a threshold corresponding to a 1.5-fold factor change in mRNA levels, observed in at least three of seven independent human hepatocyte cultures, efflux transporters such as MDR1, MRP2 and BCRP were up-regulated by PB, RIF, and OPZ, whereas MRP3 was induced by OPZ and RIF. Rifampin 223-226 ATP binding cassette subfamily C member 2 Homo sapiens 184-188 16837569-4 2006 With a threshold corresponding to a 1.5-fold factor change in mRNA levels, observed in at least three of seven independent human hepatocyte cultures, efflux transporters such as MDR1, MRP2 and BCRP were up-regulated by PB, RIF, and OPZ, whereas MRP3 was induced by OPZ and RIF. Rifampin 223-226 BCR pseudogene 1 Homo sapiens 193-197 16837569-4 2006 With a threshold corresponding to a 1.5-fold factor change in mRNA levels, observed in at least three of seven independent human hepatocyte cultures, efflux transporters such as MDR1, MRP2 and BCRP were up-regulated by PB, RIF, and OPZ, whereas MRP3 was induced by OPZ and RIF. Rifampin 223-226 ATP binding cassette subfamily C member 3 Homo sapiens 245-249 16837569-4 2006 With a threshold corresponding to a 1.5-fold factor change in mRNA levels, observed in at least three of seven independent human hepatocyte cultures, efflux transporters such as MDR1, MRP2 and BCRP were up-regulated by PB, RIF, and OPZ, whereas MRP3 was induced by OPZ and RIF. Rifampin 273-276 ATP binding cassette subfamily B member 1 Homo sapiens 178-182 16837569-4 2006 With a threshold corresponding to a 1.5-fold factor change in mRNA levels, observed in at least three of seven independent human hepatocyte cultures, efflux transporters such as MDR1, MRP2 and BCRP were up-regulated by PB, RIF, and OPZ, whereas MRP3 was induced by OPZ and RIF. Rifampin 273-276 ATP binding cassette subfamily C member 2 Homo sapiens 184-188 16837569-4 2006 With a threshold corresponding to a 1.5-fold factor change in mRNA levels, observed in at least three of seven independent human hepatocyte cultures, efflux transporters such as MDR1, MRP2 and BCRP were up-regulated by PB, RIF, and OPZ, whereas MRP3 was induced by OPZ and RIF. Rifampin 273-276 BCR pseudogene 1 Homo sapiens 193-197 16837569-4 2006 With a threshold corresponding to a 1.5-fold factor change in mRNA levels, observed in at least three of seven independent human hepatocyte cultures, efflux transporters such as MDR1, MRP2 and BCRP were up-regulated by PB, RIF, and OPZ, whereas MRP3 was induced by OPZ and RIF. Rifampin 273-276 ATP binding cassette subfamily C member 3 Homo sapiens 245-249 16837625-5 2006 P-glycoprotein expression and transport activity in brain capillaries from transgenic mice was significantly increased when capillaries were exposed to the hPXR ligands, rifampin and hyperforin, in vitro and when the mice were dosed with rifampin in vivo. Rifampin 170-178 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 16837625-5 2006 P-glycoprotein expression and transport activity in brain capillaries from transgenic mice was significantly increased when capillaries were exposed to the hPXR ligands, rifampin and hyperforin, in vitro and when the mice were dosed with rifampin in vivo. Rifampin 170-178 nuclear receptor subfamily 1 group I member 2 Homo sapiens 156-160 16837625-5 2006 P-glycoprotein expression and transport activity in brain capillaries from transgenic mice was significantly increased when capillaries were exposed to the hPXR ligands, rifampin and hyperforin, in vitro and when the mice were dosed with rifampin in vivo. Rifampin 238-246 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 16837625-5 2006 P-glycoprotein expression and transport activity in brain capillaries from transgenic mice was significantly increased when capillaries were exposed to the hPXR ligands, rifampin and hyperforin, in vitro and when the mice were dosed with rifampin in vivo. Rifampin 238-246 nuclear receptor subfamily 1 group I member 2 Homo sapiens 156-160 16782766-2 2006 In monkey hepatocytes, rifampin markedly induced CYP3A64 mRNA (EC50 = 0.5 microM; Emax = 6-fold) and midazolam (MDZ) 1"-hydroxylase activity (EC50 = 0.2 microM; Emax = 2-fold). Rifampin 23-31 cytochrome P450 3A64 Macaca mulatta 49-56 16782766-4 2006 Similar corresponding results also were obtained with human CYP3A4 in the presence of rifampin or compound A. Rifampin 86-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 60-66 16842400-0 2006 Induction of P-glycoprotein in lymphocytes by carbamazepine and rifampicin: the role of nuclear hormone response elements. Rifampin 64-74 ATP binding cassette subfamily B member 1 Homo sapiens 13-27 16842400-4 2006 METHODS: Pgp expression was assessed by flow cytometry and real-time reverse transcriptase-polymerase chain reaction using lymphocytes from four healthy subjects after incubation with therapeutic concentrations of CBZ, using rifampicin as a positive control. Rifampin 225-235 ATP binding cassette subfamily B member 1 Homo sapiens 9-12 16946557-3 2006 In primary cultures of human hepatocytes, mRNA levels of MDR and MRP1 were increased by about 1.5 fold and 1.3 fold, respectively, by exposure to Rif at 2 to 50 microM as compared with 0.1% DMSO-treated controls. Rifampin 146-149 mutS homolog 3 Homo sapiens 65-69 16946557-4 2006 MRP2 mRNA levels in the same human hepatocytes were significantly increased by 1.2 to 1.8 fold by exposure to Rif at 50 microM as compared with controls. Rifampin 110-113 ATP binding cassette subfamily C member 2 Homo sapiens 0-4 16822884-8 2006 Furthermore, the addition of rifampicin increased the gene expression of CYP3A4, which is similar with the activities of human liver cells. Rifampin 29-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 73-79 16713641-5 2006 We used real time PCR to examine whether known and suspected CYP3A inducers (dexamethasone, metyrapone, rifampicin, and toxaphene) up-regulate steady state levels of hepatic CYP3A77 transcript to determine if induction patterns in female juvenile alligators are similar to those reported in other vertebrates and whether toxaphene, an organochlorine compound found in high concentrations in Lake Apopka alligators, induces this gene. Rifampin 104-114 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 61-66 16606623-4 2006 To explore the SXR-mediated vitamin K2 signaling network in bone homeostasis, we identified genes up-regulated by both vitamin K2 and the prototypical SXR ligand, rifampicin, in osteoblastic cells using oligonucleotide microarray analysis and quantitative reverse transcription-PCR. Rifampin 163-173 nuclear receptor subfamily 1 group I member 2 Homo sapiens 15-18 16606623-4 2006 To explore the SXR-mediated vitamin K2 signaling network in bone homeostasis, we identified genes up-regulated by both vitamin K2 and the prototypical SXR ligand, rifampicin, in osteoblastic cells using oligonucleotide microarray analysis and quantitative reverse transcription-PCR. Rifampin 163-173 nuclear receptor subfamily 1 group I member 2 Homo sapiens 151-154 16620787-6 2006 Rifampin (10 microM) caused significant induction of ABCB1 (595+/-263%, p<0.05) in LS174T cells but induction was absent in Caco-2 or A549 cells. Rifampin 0-8 ATP binding cassette subfamily B member 1 Homo sapiens 53-58 16691293-7 2006 Moreover, 1,25(OH)2D3-induced CYP24 expression was enhanced in mice lacking the SXR ortholog pregnane X receptor, and treatment of humans with the SXR agonist rifampicin had no effect on intestinal CYP24 expression, despite demonstration of marked CYP3A4 induction. Rifampin 159-169 nuclear receptor subfamily 1 group I member 2 Homo sapiens 147-150 16857725-3 2006 Rifampicin, phenobarbital, and 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime, which are activators of pregnane X receptor (PXR) and constitutive androstane receptor (CAR), induced CYP2A6 mRNA in human hepatocytes. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 145-164 16857725-3 2006 Rifampicin, phenobarbital, and 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime, which are activators of pregnane X receptor (PXR) and constitutive androstane receptor (CAR), induced CYP2A6 mRNA in human hepatocytes. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 166-169 16857725-3 2006 Rifampicin, phenobarbital, and 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime, which are activators of pregnane X receptor (PXR) and constitutive androstane receptor (CAR), induced CYP2A6 mRNA in human hepatocytes. Rifampin 0-10 nuclear receptor subfamily 1 group I member 3 Homo sapiens 175-207 16857725-3 2006 Rifampicin, phenobarbital, and 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime, which are activators of pregnane X receptor (PXR) and constitutive androstane receptor (CAR), induced CYP2A6 mRNA in human hepatocytes. Rifampin 0-10 nuclear receptor subfamily 1 group I member 3 Homo sapiens 209-212 16857725-3 2006 Rifampicin, phenobarbital, and 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime, which are activators of pregnane X receptor (PXR) and constitutive androstane receptor (CAR), induced CYP2A6 mRNA in human hepatocytes. Rifampin 0-10 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 223-229 16760228-7 2006 The well known cholestatic drugs, rifampicin, rifamycin SV, glibenclamide, and cyclosporin A, reduced the basal-to-apical transport and the apical efflux clearance of taurocholate across NTCP- and BSEP-coexpressing cell monolayers. Rifampin 34-44 solute carrier family 10 member 1 Homo sapiens 187-191 16760228-7 2006 The well known cholestatic drugs, rifampicin, rifamycin SV, glibenclamide, and cyclosporin A, reduced the basal-to-apical transport and the apical efflux clearance of taurocholate across NTCP- and BSEP-coexpressing cell monolayers. Rifampin 34-44 ATP binding cassette subfamily B member 11 Homo sapiens 197-201 16849379-8 2006 In contrast, the combinations doxycycline/gentamicin and doxycycline/rifampicin were effective in the clearance of Rev 1 infection, but only the former improved significantly the therapeutic efficacy as compared with that of the antibiotics given alone. Rifampin 69-79 REV1, DNA directed polymerase Mus musculus 115-120 16831602-4 2006 RESULTS: The pregnane X receptor was expressed in primary cultures at the level of messenger RNA and protein and was activated by the ligand rifampicin as judged by increases in binding of proteins to the pregnane X receptor ER6 DNA response element and by increases in ER6-dependent reporter gene expression. Rifampin 141-151 nuclear receptor subfamily 1 group I member 2 Homo sapiens 13-32 16831602-4 2006 RESULTS: The pregnane X receptor was expressed in primary cultures at the level of messenger RNA and protein and was activated by the ligand rifampicin as judged by increases in binding of proteins to the pregnane X receptor ER6 DNA response element and by increases in ER6-dependent reporter gene expression. Rifampin 141-151 nuclear receptor subfamily 1 group I member 2 Homo sapiens 205-224 16831602-5 2006 Short-term treatment of hepatic stellate cells with rifampicin inhibited the expression of selected fibrosis-related genes (transforming growth factor beta1, alpha-smooth muscle actin), proliferation-related genes, and WNT signaling-associated genes. Rifampin 52-62 transforming growth factor beta 1 Homo sapiens 124-156 16479312-7 2006 Rifampin and taurocholic acid (a substrate of OATP-C) reversed the increased toxicity of arsenate and arsenite seen in OATP-C-transfected cells. Rifampin 0-8 solute carrier organic anion transporter family member 1B1 Homo sapiens 46-52 16479312-7 2006 Rifampin and taurocholic acid (a substrate of OATP-C) reversed the increased toxicity of arsenate and arsenite seen in OATP-C-transfected cells. Rifampin 0-8 solute carrier organic anion transporter family member 1B1 Homo sapiens 119-125 16632523-2 2006 Rifampin, the classical activator of CYP3A4, binds to SXR directly. Rifampin 0-8 nuclear receptor subfamily 1 group I member 2 Homo sapiens 54-57 16109480-6 2006 However, in highly differentiated primary human hepatocytes treated with rifampicin an agonist of the pregnane X receptor (PXR), omeprazole-sulphide behaves as an agonist of AhR. Rifampin 73-83 nuclear receptor subfamily 1 group I member 2 Homo sapiens 102-121 16109480-6 2006 However, in highly differentiated primary human hepatocytes treated with rifampicin an agonist of the pregnane X receptor (PXR), omeprazole-sulphide behaves as an agonist of AhR. Rifampin 73-83 nuclear receptor subfamily 1 group I member 2 Homo sapiens 123-126 16501008-6 2006 Induction response for CYP3A4 to 100 microM clevidipine was approximately 20% of that of the positive control inducer rifampicin. Rifampin 118-128 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 23-29 16455805-0 2006 Rifampicin induction of CYP3A4 requires pregnane X receptor cross talk with hepatocyte nuclear factor 4alpha and coactivators, and suppression of small heterodimer partner gene expression. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-30 16455805-0 2006 Rifampicin induction of CYP3A4 requires pregnane X receptor cross talk with hepatocyte nuclear factor 4alpha and coactivators, and suppression of small heterodimer partner gene expression. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 40-59 16455805-0 2006 Rifampicin induction of CYP3A4 requires pregnane X receptor cross talk with hepatocyte nuclear factor 4alpha and coactivators, and suppression of small heterodimer partner gene expression. Rifampin 0-10 hepatocyte nuclear factor 4 alpha Homo sapiens 76-108 16455805-0 2006 Rifampicin induction of CYP3A4 requires pregnane X receptor cross talk with hepatocyte nuclear factor 4alpha and coactivators, and suppression of small heterodimer partner gene expression. Rifampin 0-10 nuclear receptor subfamily 0 group B member 2 Homo sapiens 146-171 16455805-3 2006 Rifampicin dose dependently induced the CYP3A4 but inhibited small heterodimer partner (SHP) mRNA expression levels in primary human hepatocytes. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-46 16455805-3 2006 Rifampicin dose dependently induced the CYP3A4 but inhibited small heterodimer partner (SHP) mRNA expression levels in primary human hepatocytes. Rifampin 0-10 nuclear receptor subfamily 0 group B member 2 Homo sapiens 61-86 16455805-3 2006 Rifampicin dose dependently induced the CYP3A4 but inhibited small heterodimer partner (SHP) mRNA expression levels in primary human hepatocytes. Rifampin 0-10 nuclear receptor subfamily 0 group B member 2 Homo sapiens 88-91 16455805-4 2006 Rifampicin strongly stimulated PXR and hepatocyte nuclear factor 4alpha (HNF4alpha) interaction, and CYP3A4 reporter activity, which was further stimulated by peroxisome proliferators-activated receptorgamma co-activator 1alpha (PGC-1alpha) and steroid receptor coactivator-1 (SRC-1) but inhibited by SHP. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 31-34 16455805-4 2006 Rifampicin strongly stimulated PXR and hepatocyte nuclear factor 4alpha (HNF4alpha) interaction, and CYP3A4 reporter activity, which was further stimulated by peroxisome proliferators-activated receptorgamma co-activator 1alpha (PGC-1alpha) and steroid receptor coactivator-1 (SRC-1) but inhibited by SHP. Rifampin 0-10 hepatocyte nuclear factor 4 alpha Homo sapiens 39-71 16455805-4 2006 Rifampicin strongly stimulated PXR and hepatocyte nuclear factor 4alpha (HNF4alpha) interaction, and CYP3A4 reporter activity, which was further stimulated by peroxisome proliferators-activated receptorgamma co-activator 1alpha (PGC-1alpha) and steroid receptor coactivator-1 (SRC-1) but inhibited by SHP. Rifampin 0-10 hepatocyte nuclear factor 4 alpha Homo sapiens 73-82 16455805-4 2006 Rifampicin strongly stimulated PXR and hepatocyte nuclear factor 4alpha (HNF4alpha) interaction, and CYP3A4 reporter activity, which was further stimulated by peroxisome proliferators-activated receptorgamma co-activator 1alpha (PGC-1alpha) and steroid receptor coactivator-1 (SRC-1) but inhibited by SHP. Rifampin 0-10 PPARG coactivator 1 alpha Homo sapiens 229-239 16455805-4 2006 Rifampicin strongly stimulated PXR and hepatocyte nuclear factor 4alpha (HNF4alpha) interaction, and CYP3A4 reporter activity, which was further stimulated by peroxisome proliferators-activated receptorgamma co-activator 1alpha (PGC-1alpha) and steroid receptor coactivator-1 (SRC-1) but inhibited by SHP. Rifampin 0-10 nuclear receptor coactivator 1 Homo sapiens 245-275 16455805-4 2006 Rifampicin strongly stimulated PXR and hepatocyte nuclear factor 4alpha (HNF4alpha) interaction, and CYP3A4 reporter activity, which was further stimulated by peroxisome proliferators-activated receptorgamma co-activator 1alpha (PGC-1alpha) and steroid receptor coactivator-1 (SRC-1) but inhibited by SHP. Rifampin 0-10 nuclear receptor coactivator 1 Homo sapiens 277-282 16455805-4 2006 Rifampicin strongly stimulated PXR and hepatocyte nuclear factor 4alpha (HNF4alpha) interaction, and CYP3A4 reporter activity, which was further stimulated by peroxisome proliferators-activated receptorgamma co-activator 1alpha (PGC-1alpha) and steroid receptor coactivator-1 (SRC-1) but inhibited by SHP. Rifampin 0-10 nuclear receptor subfamily 0 group B member 2 Homo sapiens 301-304 16455805-6 2006 Chromatin immunoprecipitation assays revealed that rifampicin-activated PXR recruited HNF4alpha and SRC-1 to the CYP3A4 chromatin. Rifampin 51-61 nuclear receptor subfamily 1 group I member 2 Homo sapiens 72-75 16455805-6 2006 Chromatin immunoprecipitation assays revealed that rifampicin-activated PXR recruited HNF4alpha and SRC-1 to the CYP3A4 chromatin. Rifampin 51-61 hepatocyte nuclear factor 4 alpha Homo sapiens 86-95 16455805-6 2006 Chromatin immunoprecipitation assays revealed that rifampicin-activated PXR recruited HNF4alpha and SRC-1 to the CYP3A4 chromatin. Rifampin 51-61 nuclear receptor coactivator 1 Homo sapiens 100-105 16455805-6 2006 Chromatin immunoprecipitation assays revealed that rifampicin-activated PXR recruited HNF4alpha and SRC-1 to the CYP3A4 chromatin. Rifampin 51-61 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 113-119 16455805-9 2006 Upon activation by rifampicin, PXR inhibited SHP promoter activity. Rifampin 19-29 nuclear receptor subfamily 1 group I member 2 Homo sapiens 31-34 16455805-9 2006 Upon activation by rifampicin, PXR inhibited SHP promoter activity. Rifampin 19-29 nuclear receptor subfamily 0 group B member 2 Homo sapiens 45-48 16488578-5 2006 In addition, expression of canalicular transporters in HepaRG cells was found to be up-regulated by known inducers of transporters such as rifampicin, phenobarbital and chenodeoxycholate acting on P-glycoprotein, MRP2 and BSEP, respectively. Rifampin 139-149 ATP binding cassette subfamily B member 1 Homo sapiens 197-211 16488578-5 2006 In addition, expression of canalicular transporters in HepaRG cells was found to be up-regulated by known inducers of transporters such as rifampicin, phenobarbital and chenodeoxycholate acting on P-glycoprotein, MRP2 and BSEP, respectively. Rifampin 139-149 ATP binding cassette subfamily C member 2 Homo sapiens 213-217 16488578-5 2006 In addition, expression of canalicular transporters in HepaRG cells was found to be up-regulated by known inducers of transporters such as rifampicin, phenobarbital and chenodeoxycholate acting on P-glycoprotein, MRP2 and BSEP, respectively. Rifampin 139-149 ATP binding cassette subfamily B member 11 Homo sapiens 222-226 16836137-0 2006 [Fluorescence spectroscopy study of human serum albumin quenched by rifampicin capsules]. Rifampin 68-78 albumin Homo sapiens 42-55 16495352-9 2006 In conclusion, we have identified OATP1B3 as the human hepatic uptake transporter for amatoxins; moreover, substrates and inhibitors of OATP1B3, among others rifampicin, may be useful for the treatment of human amatoxin poisoning. Rifampin 158-168 solute carrier organic anion transporter family member 1B3 Homo sapiens 34-41 16525793-2 2006 Substrates for members of the OATP family include bile salts, hormones, and steroid conjugates as well as drugs like the HMG-CoA-reductase inhibitors (statins), cardiac glycosides, anticancer agents like methotrexate, and antibiotics like rifampicin. Rifampin 239-249 solute carrier organic anion transporter family member 1A2 Homo sapiens 30-34 16944591-6 2006 The results of sequence analysis were shown as: amomg 18 rifampin-resistant strains, only one has deletion in codons 513 and 514 of rpoB gene, which is a new report, the others have point mutation; among 12 rifampin- susceptible strains, 3 positive strains in SSCP occur gene mutations directly related to rifampin resistance. Rifampin 57-65 DNA-directed RNA polymerase subunit beta Mycobacterium tuberculosis H37Rv 132-136 16498570-6 2006 Losartan appears to be primarily a P-glycoprotein (P-gp) substrate, as previously reported, but MRP inhibitors such as MK-571 and rifampicin also affect the difference between apical to basolateral and basolateral to apical transport. Rifampin 130-140 ATP binding cassette subfamily C member 1 Homo sapiens 96-99 16473453-11 2006 In conclusion, human hepatocytes cryopreserved on collagen gels show a clear induction of CYP3A4 by rifampicin, although the basal activities are reduced compared to non-frozen cells. Rifampin 100-110 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-96 16513445-3 2006 This study evaluates the contribution of these factors to the disposition and cholesterol-lowering effect of ezetimibe before and after induction of UGT1A1, P-gp, and MRP2 with rifampin (INN, rifampicin). Rifampin 177-185 ATP binding cassette subfamily C member 2 Homo sapiens 167-171 16513445-7 2006 RESULTS: Up-regulation of intestinal P-gp, MRP2, and UGT1A1 (but not of NPC1L1) by rifampin was associated with markedly decreased areas under the curve of ezetimibe and its glucuronide (116 +/- 78.1 ng.h/mL versus 49.9 +/- 31.0 ng.h/mL and 635 +/- 302 ng.h/mL versus 225 +/- 86.4 ng.h/mL, respectively; both P = .002) and increased intestinal clearances (2400 +/- 1560 mL/min versus 5500 +/- 4610 mL/min [P = .003] and 76.6 +/- 113 mL/min versus 316 +/- 457 mL/min [P = .010], respectively) and nearly abolished sterol-lowering effects. Rifampin 83-91 ATP binding cassette subfamily B member 1 Homo sapiens 37-41 16513445-7 2006 RESULTS: Up-regulation of intestinal P-gp, MRP2, and UGT1A1 (but not of NPC1L1) by rifampin was associated with markedly decreased areas under the curve of ezetimibe and its glucuronide (116 +/- 78.1 ng.h/mL versus 49.9 +/- 31.0 ng.h/mL and 635 +/- 302 ng.h/mL versus 225 +/- 86.4 ng.h/mL, respectively; both P = .002) and increased intestinal clearances (2400 +/- 1560 mL/min versus 5500 +/- 4610 mL/min [P = .003] and 76.6 +/- 113 mL/min versus 316 +/- 457 mL/min [P = .010], respectively) and nearly abolished sterol-lowering effects. Rifampin 83-91 ATP binding cassette subfamily C member 2 Homo sapiens 43-47 16513445-7 2006 RESULTS: Up-regulation of intestinal P-gp, MRP2, and UGT1A1 (but not of NPC1L1) by rifampin was associated with markedly decreased areas under the curve of ezetimibe and its glucuronide (116 +/- 78.1 ng.h/mL versus 49.9 +/- 31.0 ng.h/mL and 635 +/- 302 ng.h/mL versus 225 +/- 86.4 ng.h/mL, respectively; both P = .002) and increased intestinal clearances (2400 +/- 1560 mL/min versus 5500 +/- 4610 mL/min [P = .003] and 76.6 +/- 113 mL/min versus 316 +/- 457 mL/min [P = .010], respectively) and nearly abolished sterol-lowering effects. Rifampin 83-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 16433874-0 2006 Pharmacokinetics of ruboxistaurin are significantly altered by rifampicin-mediated CYP3A4 induction. Rifampin 63-73 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-89 16580901-13 2006 Conversely, the magnitude of CYP3A4 induction by rifampin is greater in persons with the MDR1 2677G allele and is inversely related to baseline CYP3A4 expression. Rifampin 49-57 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-35 16580901-13 2006 Conversely, the magnitude of CYP3A4 induction by rifampin is greater in persons with the MDR1 2677G allele and is inversely related to baseline CYP3A4 expression. Rifampin 49-57 ATP binding cassette subfamily B member 1 Homo sapiens 89-93 16580901-13 2006 Conversely, the magnitude of CYP3A4 induction by rifampin is greater in persons with the MDR1 2677G allele and is inversely related to baseline CYP3A4 expression. Rifampin 49-57 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 144-150 16414346-8 2006 TCDD, dexamethasone, and rifampicin, which up-regulated CYP3A65 mRNA in zebrafish larvae, did not alter the CYP3C1 transcript levels suggesting regulatory differences between CYP3A and CYP3C enzymes in this species. Rifampin 25-35 cytochrome P450, family 3, subfamily A, polypeptide 65 Danio rerio 56-63 16414346-8 2006 TCDD, dexamethasone, and rifampicin, which up-regulated CYP3A65 mRNA in zebrafish larvae, did not alter the CYP3C1 transcript levels suggesting regulatory differences between CYP3A and CYP3C enzymes in this species. Rifampin 25-35 cytochrome P450, family 3, subfamily c, polypeptide 1 Danio rerio 56-61 16480505-0 2006 Roles of rifampicin in drug-drug interactions: underlying molecular mechanisms involving the nuclear pregnane X receptor. Rifampin 9-19 nuclear receptor subfamily 1 group I member 2 Homo sapiens 101-120 16480505-3 2006 The concomitantly administered effects of rifampicin on other drugs can result in their altered metabolism or transportation that are metabolised by cytochromes P450 or transported by p-glycoprotein in the gastrointestinal tract and liver. Rifampin 42-52 ATP binding cassette subfamily B member 1 Homo sapiens 184-198 16480505-5 2006 In general, rifampicin can act on a pattern: rifampicin activates the nuclear pregnane X receptor that in turn affects cytochromes P450, glucuronosyltransferases and p-glycoprotein activities. Rifampin 12-22 nuclear receptor subfamily 1 group I member 2 Homo sapiens 78-97 16480505-5 2006 In general, rifampicin can act on a pattern: rifampicin activates the nuclear pregnane X receptor that in turn affects cytochromes P450, glucuronosyltransferases and p-glycoprotein activities. Rifampin 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 166-180 16480505-5 2006 In general, rifampicin can act on a pattern: rifampicin activates the nuclear pregnane X receptor that in turn affects cytochromes P450, glucuronosyltransferases and p-glycoprotein activities. Rifampin 45-55 nuclear receptor subfamily 1 group I member 2 Homo sapiens 78-97 16480505-5 2006 In general, rifampicin can act on a pattern: rifampicin activates the nuclear pregnane X receptor that in turn affects cytochromes P450, glucuronosyltransferases and p-glycoprotein activities. Rifampin 45-55 ATP binding cassette subfamily B member 1 Homo sapiens 166-180 16433874-13 2006 CONCLUSIONS: The effect of rifampicin co-administration on the exposure of ruboxistaurin is consistent with ruboxistaurin being a substrate of CYP3A4. Rifampin 27-37 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 143-149 16489682-6 2006 RESULTS: The treatment of rats with INH+RIF (50 mg/kg per day each) induced hepatotoxicity in all the treated animals as judged by elevated serum ALT, AST, and bilirubin levels, presence of focal hepatocytic necrosis (6/8) and portal triaditis (8/8). Rifampin 40-43 glutamic-oxaloacetic transaminase 2 Rattus norvegicus 151-154 16547390-1 2006 The mRNA levels of human cytochrome P450 (CYP)2Cs and CYP3As in primary cultures of freshly isolated human hepatocytes were assessed after exposure to NO-1886 and rifampicin, a typical inducer of CYP3As. Rifampin 163-173 peptidylprolyl isomerase F Homo sapiens 54-58 16482631-7 2006 Partial protection was observed with NAC against RMP-induced changes in liver, which was evidenced from the prevention of increase in lipid peroxidation and the reduction in SOD and catalase enzyme levels. Rifampin 49-52 catalase Rattus norvegicus 182-190 16390353-11 2006 CONCLUSIONS: Rifampicin caused a substantial decrease in the plasma concentration of pioglitazone, probably by induction of CYP2C8. Rifampin 13-23 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 124-130 17181373-5 2006 For example, protease inhibitors often must be avoided if the potent CYP inducer rifampicin is co-administered. Rifampin 81-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 16288819-0 2006 Successful treatment of severe unconjugated hyperbilirubinemia via induction of UGT1A1 by rifampicin. Rifampin 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 16219912-3 2006 PXR activates cytochrome P450 3A4 gene expression upon binding to rifampicin (Rif) and clotrimazole (CTZ) by recruiting transcriptional coactivators. Rifampin 66-76 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-3 16219912-3 2006 PXR activates cytochrome P450 3A4 gene expression upon binding to rifampicin (Rif) and clotrimazole (CTZ) by recruiting transcriptional coactivators. Rifampin 78-81 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-3 16129589-10 2006 Uptake can be inhibited completely by the addition of the known OATP1B3-inhibitor rifampicin proving that Fluo-3 is transported by OATP1B3. Rifampin 82-92 solute carrier organic anion transporter family member 1B3 Homo sapiens 64-71 16129589-10 2006 Uptake can be inhibited completely by the addition of the known OATP1B3-inhibitor rifampicin proving that Fluo-3 is transported by OATP1B3. Rifampin 82-92 solute carrier organic anion transporter family member 1B3 Homo sapiens 131-138 16615094-3 2006 The purpose of this study was (1) to determine the temporal kinetics of drug-induced changes in canine CYP1A, CYP2B, and CYP3A mRNA and enzymatic activity, and (2) to characterize concentration-response relationships for CYP1A2, CYP2B11, and CYP3A12 using primary cultures of canine hepatocytes treated with beta-naphthoflavone (BNF), phenobarbital (PB), and rifampin (RIF), respectively. Rifampin 359-367 cytochrome P450 family 1 subfamily A member 2 Canis lupus familiaris 221-227 16615094-3 2006 The purpose of this study was (1) to determine the temporal kinetics of drug-induced changes in canine CYP1A, CYP2B, and CYP3A mRNA and enzymatic activity, and (2) to characterize concentration-response relationships for CYP1A2, CYP2B11, and CYP3A12 using primary cultures of canine hepatocytes treated with beta-naphthoflavone (BNF), phenobarbital (PB), and rifampin (RIF), respectively. Rifampin 369-372 cytochrome P450 family 1 subfamily A member 2 Canis lupus familiaris 221-227 16615094-6 2006 CYP3A12 and CYP3A26 mRNA levels were increased maximally after 72 h of treatment with PB and RIF (CYP3A12, 35-fold and 18-fold, and CYP3A26, 72-fold and 22-fold with PB and RIF treatment, respectively). Rifampin 93-96 cytochrome P450 3A12 Canis lupus familiaris 0-7 16615094-6 2006 CYP3A12 and CYP3A26 mRNA levels were increased maximally after 72 h of treatment with PB and RIF (CYP3A12, 35-fold and 18-fold, and CYP3A26, 72-fold and 22-fold with PB and RIF treatment, respectively). Rifampin 93-96 cytochrome P450 3A26 Canis lupus familiaris 12-19 16615094-6 2006 CYP3A12 and CYP3A26 mRNA levels were increased maximally after 72 h of treatment with PB and RIF (CYP3A12, 35-fold and 18-fold, and CYP3A26, 72-fold and 22-fold with PB and RIF treatment, respectively). Rifampin 93-96 cytochrome P450 3A12 Canis lupus familiaris 98-105 16615094-6 2006 CYP3A12 and CYP3A26 mRNA levels were increased maximally after 72 h of treatment with PB and RIF (CYP3A12, 35-fold and 18-fold, and CYP3A26, 72-fold and 22-fold with PB and RIF treatment, respectively). Rifampin 173-176 cytochrome P450 3A12 Canis lupus familiaris 0-7 16615094-6 2006 CYP3A12 and CYP3A26 mRNA levels were increased maximally after 72 h of treatment with PB and RIF (CYP3A12, 35-fold and 18-fold, and CYP3A26, 72-fold and 22-fold with PB and RIF treatment, respectively). Rifampin 173-176 cytochrome P450 3A26 Canis lupus familiaris 12-19 17065468-6 2006 In hok/sok-carrying cells, anti Sok PNAs were more bactericidal than rifampicin. Rifampin 69-79 modulator of post-segregation killing protein Escherichia coli 3-6 16135659-7 2005 All 23 single assays were validated by assessing the effects (induction or repression) of known inducers (ethanol, 3-methylcholanthrene, rifampicin, dexamethasone, phenobarbital) on P450 expression in human primary hepatocytes. Rifampin 137-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 182-186 16304167-7 2005 In agreement with the published fMLP-like activity of rifampin, the inhibitory effect of rifampin was significantly greater for fMLP (IC50 of 5.6 mg/liter) than for PMA (IC50 of 58 mg/liter) stimulation. Rifampin 54-62 formyl peptide receptor 1 Homo sapiens 32-36 16304167-7 2005 In agreement with the published fMLP-like activity of rifampin, the inhibitory effect of rifampin was significantly greater for fMLP (IC50 of 5.6 mg/liter) than for PMA (IC50 of 58 mg/liter) stimulation. Rifampin 89-97 formyl peptide receptor 1 Homo sapiens 32-36 16304167-7 2005 In agreement with the published fMLP-like activity of rifampin, the inhibitory effect of rifampin was significantly greater for fMLP (IC50 of 5.6 mg/liter) than for PMA (IC50 of 58 mg/liter) stimulation. Rifampin 89-97 formyl peptide receptor 1 Homo sapiens 128-132 16120810-3 2005 We have now detected a low basal level of UGT1A5 expression in cultured human hepatocytes, and treatment with rifampicin or 3-methylcholanthrene increased the level of UGT1A5 mRNA. Rifampin 110-120 UDP glucuronosyltransferase family 1 member A5 Homo sapiens 42-48 16120810-3 2005 We have now detected a low basal level of UGT1A5 expression in cultured human hepatocytes, and treatment with rifampicin or 3-methylcholanthrene increased the level of UGT1A5 mRNA. Rifampin 110-120 UDP glucuronosyltransferase family 1 member A5 Homo sapiens 168-174 16120810-6 2005 Full-length UGT1A5 cDNA was isolated from Caco-2 cells that had been transfected with the pregnane X receptor and treated with rifampicin. Rifampin 127-137 UDP glucuronosyltransferase 1 family, polypeptide A5 Mus musculus 12-18 16176562-8 2005 Rifampicin inhibited CYP2C8 (Ki 30.2 microM), CYP3A4 (Ki 18.5 microM) and repaglinide metabolism (IC50 13.7 microM). Rifampin 0-10 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 21-27 15885729-5 2005 The hPXR activation at the top concentrations tested (10 microM) relative to the positive control 10 microM rifampicin ranged from 1.3% (trans-resveratrol) to 152% (ICI 182780). Rifampin 108-118 nuclear receptor subfamily 1 group I member 2 Homo sapiens 4-8 16324277-1 2005 OBJECTIVE: To study the combined effect of isoniazid and rifampicin on the activities of CYP1A2 and 3A4 in primary hepatocytes from healthy human adults. Rifampin 57-67 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 89-95 16324277-10 2005 The activity of CYP1A2 of groups with two kinds of different concentrations of isoniazid and rifampicin combined groups was (3.27 +/- 0.96), (3.97 +/- 0.25) mAU.min respectively, which had significant difference compared with that in the negative control group (P < 0.05). Rifampin 93-103 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 16-22 16324277-12 2005 The activity of CYP3A4 in rifampicin groups with concentrations of 12.5 micromol/L and 25 micromol/L was (165.17 +/- 11.47), (120.20 +/- 15.73) mAU.min respectively, which had significant difference compared with that in the negative control group (P < 0.01). Rifampin 26-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 16-22 16324277-13 2005 The activity of CYP3A4 in the three isoniazid and rifampicin combined groups with three kinds of different concentrations was (118.37 +/- 8.90), (77.53 +/- 6.91), (68.73 +/- 4.72) mAU.min respectively, which had significant difference compared with that in the negative control group (P < 0.01), but they were lower than those in rifampicin groups with corresponding concentrations (P < 0.05). Rifampin 50-60 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 16-22 16324277-13 2005 The activity of CYP3A4 in the three isoniazid and rifampicin combined groups with three kinds of different concentrations was (118.37 +/- 8.90), (77.53 +/- 6.91), (68.73 +/- 4.72) mAU.min respectively, which had significant difference compared with that in the negative control group (P < 0.01), but they were lower than those in rifampicin groups with corresponding concentrations (P < 0.05). Rifampin 333-343 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 16-22 16324277-15 2005 Isoniazid in the range of maximum clinical blood concentration can inhibit the activity of CYP3A4, while rifampicin can induce the activity of CYP3A4; the combined effect of isoniazid and rifampicin being the induction of CYP3A4 activity, but the inducing effect was less than that of rifampicin alone with the same concentration. Rifampin 105-115 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 143-149 16324277-15 2005 Isoniazid in the range of maximum clinical blood concentration can inhibit the activity of CYP3A4, while rifampicin can induce the activity of CYP3A4; the combined effect of isoniazid and rifampicin being the induction of CYP3A4 activity, but the inducing effect was less than that of rifampicin alone with the same concentration. Rifampin 105-115 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 143-149 16324277-15 2005 Isoniazid in the range of maximum clinical blood concentration can inhibit the activity of CYP3A4, while rifampicin can induce the activity of CYP3A4; the combined effect of isoniazid and rifampicin being the induction of CYP3A4 activity, but the inducing effect was less than that of rifampicin alone with the same concentration. Rifampin 188-198 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 91-97 16324277-15 2005 Isoniazid in the range of maximum clinical blood concentration can inhibit the activity of CYP3A4, while rifampicin can induce the activity of CYP3A4; the combined effect of isoniazid and rifampicin being the induction of CYP3A4 activity, but the inducing effect was less than that of rifampicin alone with the same concentration. Rifampin 188-198 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 143-149 16324277-15 2005 Isoniazid in the range of maximum clinical blood concentration can inhibit the activity of CYP3A4, while rifampicin can induce the activity of CYP3A4; the combined effect of isoniazid and rifampicin being the induction of CYP3A4 activity, but the inducing effect was less than that of rifampicin alone with the same concentration. Rifampin 188-198 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 143-149 16324277-15 2005 Isoniazid in the range of maximum clinical blood concentration can inhibit the activity of CYP3A4, while rifampicin can induce the activity of CYP3A4; the combined effect of isoniazid and rifampicin being the induction of CYP3A4 activity, but the inducing effect was less than that of rifampicin alone with the same concentration. Rifampin 188-198 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 91-97 16324277-15 2005 Isoniazid in the range of maximum clinical blood concentration can inhibit the activity of CYP3A4, while rifampicin can induce the activity of CYP3A4; the combined effect of isoniazid and rifampicin being the induction of CYP3A4 activity, but the inducing effect was less than that of rifampicin alone with the same concentration. Rifampin 188-198 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 143-149 16324277-15 2005 Isoniazid in the range of maximum clinical blood concentration can inhibit the activity of CYP3A4, while rifampicin can induce the activity of CYP3A4; the combined effect of isoniazid and rifampicin being the induction of CYP3A4 activity, but the inducing effect was less than that of rifampicin alone with the same concentration. Rifampin 188-198 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 143-149 16176562-8 2005 Rifampicin inhibited CYP2C8 (Ki 30.2 microM), CYP3A4 (Ki 18.5 microM) and repaglinide metabolism (IC50 13.7 microM). Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 46-52 16176562-11 2005 Rifampicin competitively inhibits both CYP2C8 and CYP3A4, which can counteract its inducing effect in humans. Rifampin 0-10 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 39-45 16176562-11 2005 Rifampicin competitively inhibits both CYP2C8 and CYP3A4, which can counteract its inducing effect in humans. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-56 16041547-2 2005 We investigated whether changes in mRNA expression for CYP1A2,CYP2C19, CYP2D6 and CYP3A4 in peripheral blood lymphocytes (PBLs) may serve as surrogate markers for changes in CYP enzyme activity following the administration of rifampin. Rifampin 226-234 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 71-77 16143978-7 2005 However, this study shows contrasting actions with the antibiotic rifampicin and magnesium addition leading to a decrease of the alpha-synuclein-herbicide interaction even if other metals are present in the bulk solvent. Rifampin 66-76 synuclein alpha Homo sapiens 129-144 16041547-2 2005 We investigated whether changes in mRNA expression for CYP1A2,CYP2C19, CYP2D6 and CYP3A4 in peripheral blood lymphocytes (PBLs) may serve as surrogate markers for changes in CYP enzyme activity following the administration of rifampin. Rifampin 226-234 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 82-88 15933212-6 2005 We initially identified a distal PXR/CAR-binding site in the CYP2C8 promoter that confers inducibility of CYP2C8 via the PXR agonist/ligand rifampicin and the CAR agonist/ligand CITCO [6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime]. Rifampin 140-150 nuclear receptor subfamily 1 group I member 2 Homo sapiens 33-36 15919766-2 2005 Recent studies in our laboratory have shown that the nuclear receptor pregnane X receptor (PXR) is important in the transcriptional activation of the CYP2C9 promoter by drugs such as rifampicin and that the essential element is a constitutive androstane receptor (CAR)/PXR site -1839 bp upstream of the translation start site. Rifampin 183-193 nuclear receptor subfamily 1 group I member 2 Homo sapiens 70-89 15919766-2 2005 Recent studies in our laboratory have shown that the nuclear receptor pregnane X receptor (PXR) is important in the transcriptional activation of the CYP2C9 promoter by drugs such as rifampicin and that the essential element is a constitutive androstane receptor (CAR)/PXR site -1839 bp upstream of the translation start site. Rifampin 183-193 nuclear receptor subfamily 1 group I member 2 Homo sapiens 91-94 15919766-2 2005 Recent studies in our laboratory have shown that the nuclear receptor pregnane X receptor (PXR) is important in the transcriptional activation of the CYP2C9 promoter by drugs such as rifampicin and that the essential element is a constitutive androstane receptor (CAR)/PXR site -1839 bp upstream of the translation start site. Rifampin 183-193 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 15919766-2 2005 Recent studies in our laboratory have shown that the nuclear receptor pregnane X receptor (PXR) is important in the transcriptional activation of the CYP2C9 promoter by drugs such as rifampicin and that the essential element is a constitutive androstane receptor (CAR)/PXR site -1839 bp upstream of the translation start site. Rifampin 183-193 nuclear receptor subfamily 1 group I member 3 Homo sapiens 264-267 15919766-2 2005 Recent studies in our laboratory have shown that the nuclear receptor pregnane X receptor (PXR) is important in the transcriptional activation of the CYP2C9 promoter by drugs such as rifampicin and that the essential element is a constitutive androstane receptor (CAR)/PXR site -1839 bp upstream of the translation start site. Rifampin 183-193 nuclear receptor subfamily 1 group I member 2 Homo sapiens 269-272 15919766-6 2005 HNF4alpha synergizes with CAR and with PXR in HepG2 cells treated with rifampicin. Rifampin 71-81 hepatocyte nuclear factor 4 alpha Homo sapiens 0-9 15919766-6 2005 HNF4alpha synergizes with CAR and with PXR in HepG2 cells treated with rifampicin. Rifampin 71-81 nuclear receptor subfamily 1 group I member 2 Homo sapiens 39-42 15919766-8 2005 Mutation of the two HNF4alpha binding sites differentially prevented up-regulation of CYP2C9 promoter by both CAR as well as HNF4alpha, synergy between the two receptors, and essentially abolished induction by rifampicin in HepG2 cells transfected with PXR. Rifampin 210-220 hepatocyte nuclear factor 4 alpha Homo sapiens 20-29 15919766-8 2005 Mutation of the two HNF4alpha binding sites differentially prevented up-regulation of CYP2C9 promoter by both CAR as well as HNF4alpha, synergy between the two receptors, and essentially abolished induction by rifampicin in HepG2 cells transfected with PXR. Rifampin 210-220 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 15919766-8 2005 Mutation of the two HNF4alpha binding sites differentially prevented up-regulation of CYP2C9 promoter by both CAR as well as HNF4alpha, synergy between the two receptors, and essentially abolished induction by rifampicin in HepG2 cells transfected with PXR. Rifampin 210-220 nuclear receptor subfamily 1 group I member 3 Homo sapiens 110-113 15919766-8 2005 Mutation of the two HNF4alpha binding sites differentially prevented up-regulation of CYP2C9 promoter by both CAR as well as HNF4alpha, synergy between the two receptors, and essentially abolished induction by rifampicin in HepG2 cells transfected with PXR. Rifampin 210-220 nuclear receptor subfamily 1 group I member 2 Homo sapiens 253-256 16099622-0 2005 The occurrence of rare rpoB mutations in rifampicin-resistant clinical Mycobacterium tuberculosis isolates from Kuwait. Rifampin 41-51 DNA-directed RNA polymerase subunit beta Mycobacterium tuberculosis H37Rv 23-27 16099622-1 2005 Mutations associated with rifampicin (RIF) resistance in two regions of the rpoB gene were studied by line probe (INNO-LiPA Rif. Rifampin 26-36 DNA-directed RNA polymerase subunit beta Mycobacterium tuberculosis H37Rv 76-80 16099622-1 2005 Mutations associated with rifampicin (RIF) resistance in two regions of the rpoB gene were studied by line probe (INNO-LiPA Rif. Rifampin 38-41 DNA-directed RNA polymerase subunit beta Mycobacterium tuberculosis H37Rv 76-80 15933212-6 2005 We initially identified a distal PXR/CAR-binding site in the CYP2C8 promoter that confers inducibility of CYP2C8 via the PXR agonist/ligand rifampicin and the CAR agonist/ligand CITCO [6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime]. Rifampin 140-150 nuclear receptor subfamily 1 group I member 3 Homo sapiens 37-40 15933212-6 2005 We initially identified a distal PXR/CAR-binding site in the CYP2C8 promoter that confers inducibility of CYP2C8 via the PXR agonist/ligand rifampicin and the CAR agonist/ligand CITCO [6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime]. Rifampin 140-150 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 61-67 15933212-6 2005 We initially identified a distal PXR/CAR-binding site in the CYP2C8 promoter that confers inducibility of CYP2C8 via the PXR agonist/ligand rifampicin and the CAR agonist/ligand CITCO [6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime]. Rifampin 140-150 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 106-112 15933212-6 2005 We initially identified a distal PXR/CAR-binding site in the CYP2C8 promoter that confers inducibility of CYP2C8 via the PXR agonist/ligand rifampicin and the CAR agonist/ligand CITCO [6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime]. Rifampin 140-150 nuclear receptor subfamily 1 group I member 2 Homo sapiens 121-124 16193213-8 2005 At 3 days, explanted rifampin-soaked grafts exhibited no MRSA growth (4 of 4 grafts) and a > or =97% mean log reduction of MRSA CFUs from the adjacent aorta and perigraft fluid (PGF). Rifampin 21-29 placental growth factor Canis lupus familiaris 181-184 16193213-10 2005 The 7-day rifampin group had an average log reduction in MRSA CFU of 72% (graft), 58% (PGF), 75% (aorta). Rifampin 10-18 placental growth factor Canis lupus familiaris 87-90 16193213-11 2005 Quantitative cultures of 14-day rifampin grafted demonstrated continued bacterial growth suppression with mean MRSA CFU log reductions of 82%, graft; 72%, PGF; 89%, aorta. Rifampin 32-40 placental growth factor Canis lupus familiaris 155-158 16084850-15 2005 It is advisable to increase the dosage of atorvastatin and preferable to administer it in the evening to guarantee adequate concentrations during the nighttime rapid cholesterol synthesis when rifampin or other potent inducers of cytochrome P450 3A4 are coadministered. Rifampin 193-201 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 230-249 16308281-3 2005 The present study examined the in vivo induction potency of human CYP3A in chimeric mice with humanized liver, recently established in Japan, by a specific inducer of human CYP3A enzyme activity in this experimental condition, rifabutin, which is an analogue of rifampicin. Rifampin 262-272 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 66-71 16028152-8 2005 Low serum concentrations of rifampin were found in both HIV-infected and non-HIV-infected patients, and such patients were less likely to have >4 weeks of symptoms, more likely to have lymphadenopathy, and more likely to have low serum albumin levels (P<.05 for all). Rifampin 28-36 albumin Homo sapiens 239-246 16003296-1 2005 The tuberculostatic compound rifampin (INN, rifampicin) induces the expression of a number of drug metabolism-related genes involved in multidrug resistance (P-glycoprotein and multidrug resistance proteins 1 and 2), cytochromes (cytochrome P450 [CYP] 3A4), uridine diphosphate-glucuronosyltransferases, monoamine oxidases, and glutathione S -transferases. Rifampin 29-37 ATP binding cassette subfamily B member 1 Homo sapiens 158-214 16083704-7 2005 RESULTS: RIFA enhanced bile acid detoxification as well as bilirubin conjugation and excretion as reflected by enhanced expression of CYP3A4, UGT1A1, and MRP2. Rifampin 9-13 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 134-140 16083704-7 2005 RESULTS: RIFA enhanced bile acid detoxification as well as bilirubin conjugation and excretion as reflected by enhanced expression of CYP3A4, UGT1A1, and MRP2. Rifampin 9-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 16083704-7 2005 RESULTS: RIFA enhanced bile acid detoxification as well as bilirubin conjugation and excretion as reflected by enhanced expression of CYP3A4, UGT1A1, and MRP2. Rifampin 9-13 ATP binding cassette subfamily C member 2 Homo sapiens 154-158 15833926-8 2005 Treatment with arsenite in the presence of CYP3A4 inducers, rifampicin (Rif) or phenobarbital, caused major decreases in CYP3A4 mRNA, protein, and activity. Rifampin 60-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 43-49 15833926-8 2005 Treatment with arsenite in the presence of CYP3A4 inducers, rifampicin (Rif) or phenobarbital, caused major decreases in CYP3A4 mRNA, protein, and activity. Rifampin 60-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 121-127 15833926-8 2005 Treatment with arsenite in the presence of CYP3A4 inducers, rifampicin (Rif) or phenobarbital, caused major decreases in CYP3A4 mRNA, protein, and activity. Rifampin 72-75 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 43-49 15833926-8 2005 Treatment with arsenite in the presence of CYP3A4 inducers, rifampicin (Rif) or phenobarbital, caused major decreases in CYP3A4 mRNA, protein, and activity. Rifampin 72-75 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 121-127 15833926-11 2005 We found that arsenite failed to affect expression of PXR or the transcription factor Sp1, yet caused a significant decrease in PXR responsiveness to Rif. Rifampin 150-153 nuclear receptor subfamily 1 group I member 2 Homo sapiens 128-131 16003296-1 2005 The tuberculostatic compound rifampin (INN, rifampicin) induces the expression of a number of drug metabolism-related genes involved in multidrug resistance (P-glycoprotein and multidrug resistance proteins 1 and 2), cytochromes (cytochrome P450 [CYP] 3A4), uridine diphosphate-glucuronosyltransferases, monoamine oxidases, and glutathione S -transferases. Rifampin 29-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-255 16003296-1 2005 The tuberculostatic compound rifampin (INN, rifampicin) induces the expression of a number of drug metabolism-related genes involved in multidrug resistance (P-glycoprotein and multidrug resistance proteins 1 and 2), cytochromes (cytochrome P450 [CYP] 3A4), uridine diphosphate-glucuronosyltransferases, monoamine oxidases, and glutathione S -transferases. Rifampin 44-54 ATP binding cassette subfamily B member 1 Homo sapiens 158-214 16003296-1 2005 The tuberculostatic compound rifampin (INN, rifampicin) induces the expression of a number of drug metabolism-related genes involved in multidrug resistance (P-glycoprotein and multidrug resistance proteins 1 and 2), cytochromes (cytochrome P450 [CYP] 3A4), uridine diphosphate-glucuronosyltransferases, monoamine oxidases, and glutathione S -transferases. Rifampin 44-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-255 15907539-5 2005 When harboring traA mutants, rifampin-sensitive hosts exhibited constitutive clumping, whereas rifampin-resistant hosts did not-despite the fact that the latter host exhibited a normal pheromone-inducible clumping response when harboring a wild-type plasmid. Rifampin 29-37 pheromone-binding protein Enterococcus faecalis 15-19 15883047-8 2005 The expression of ABCA1 and SR-BI was inhibited by the PXR activators rifampicin and lithocholic acid (LCA) in HepG2 cells and pregnenolone 16alpha-carbonitrile (PCN) in primary rat hepatocytes. Rifampin 70-80 ATP binding cassette subfamily A member 1 Homo sapiens 18-23 15883047-8 2005 The expression of ABCA1 and SR-BI was inhibited by the PXR activators rifampicin and lithocholic acid (LCA) in HepG2 cells and pregnenolone 16alpha-carbonitrile (PCN) in primary rat hepatocytes. Rifampin 70-80 scavenger receptor class B member 1 Homo sapiens 28-33 15883047-8 2005 The expression of ABCA1 and SR-BI was inhibited by the PXR activators rifampicin and lithocholic acid (LCA) in HepG2 cells and pregnenolone 16alpha-carbonitrile (PCN) in primary rat hepatocytes. Rifampin 70-80 nuclear receptor subfamily 1 group I member 2 Homo sapiens 55-58 15769886-5 2005 The expression levels of human CYP3A4 mRNA and CYP3A4 protein and dexamethasone 6-hydroxylase activity, specific for human CYP3A4, were increased 8- to 22-, 3- to 10-, and 5- to 12-fold, respectively, by treatment with rifampicin. Rifampin 219-229 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 15922010-5 2005 Like the human CYP3A genes, CYP3A65 transcription in the foregut region was enhanced by treatment of the zebrafish larvae with the steroid dexamethasone and the macrocyclic antibiotic rifampicin. Rifampin 184-194 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 15-20 15922010-5 2005 Like the human CYP3A genes, CYP3A65 transcription in the foregut region was enhanced by treatment of the zebrafish larvae with the steroid dexamethasone and the macrocyclic antibiotic rifampicin. Rifampin 184-194 cytochrome P450, family 3, subfamily A, polypeptide 65 Danio rerio 28-35 15769886-5 2005 The expression levels of human CYP3A4 mRNA and CYP3A4 protein and dexamethasone 6-hydroxylase activity, specific for human CYP3A4, were increased 8- to 22-, 3- to 10-, and 5- to 12-fold, respectively, by treatment with rifampicin. Rifampin 219-229 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-53 15769886-5 2005 The expression levels of human CYP3A4 mRNA and CYP3A4 protein and dexamethasone 6-hydroxylase activity, specific for human CYP3A4, were increased 8- to 22-, 3- to 10-, and 5- to 12-fold, respectively, by treatment with rifampicin. Rifampin 219-229 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-53 15883178-10 2005 In particular, rifampicin and the fluoroquinolones were the most active compounds, both in extracellular medium and in THP-1 cells. Rifampin 15-25 GLI family zinc finger 2 Homo sapiens 119-124 15705662-0 2005 Structural disorder in the complex of human pregnane X receptor and the macrolide antibiotic rifampicin. Rifampin 93-103 nuclear receptor subfamily 1 group I member 2 Homo sapiens 44-63 15705662-2 2005 The macrolide antibiotic rifampicin, a front-line treatment for tuberculosis, is an established PXR agonist and, at 823 Da, is one of the largest known ligands for the receptor. Rifampin 25-35 nuclear receptor subfamily 1 group I member 2 Homo sapiens 96-99 15705662-3 2005 We present the 2.8 A crystal structure of the ligand-binding domain of human PXR in complex with rifampicin. Rifampin 97-107 nuclear receptor subfamily 1 group I member 2 Homo sapiens 77-80 15864135-6 2005 CYP3A4-luciferase reporter assays revealed that the Q158K variant gave rise to much lower levels of CYP3A4 promoter activity in LS174T and HepG2 cells exposed to the PXR ligands, rifampin and paclitaxel, than did wild-type PXR. Rifampin 179-187 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 15864135-6 2005 CYP3A4-luciferase reporter assays revealed that the Q158K variant gave rise to much lower levels of CYP3A4 promoter activity in LS174T and HepG2 cells exposed to the PXR ligands, rifampin and paclitaxel, than did wild-type PXR. Rifampin 179-187 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 100-106 15864135-6 2005 CYP3A4-luciferase reporter assays revealed that the Q158K variant gave rise to much lower levels of CYP3A4 promoter activity in LS174T and HepG2 cells exposed to the PXR ligands, rifampin and paclitaxel, than did wild-type PXR. Rifampin 179-187 nuclear receptor subfamily 1 group I member 2 Homo sapiens 223-226 15855724-3 2005 beta-naphthoflavone (beta-NF) and rifampicin (Rif) were used as typical cytochrome P450 (CYP) inducers for CYP1A2 and CYP3A4, respectively. Rifampin 34-44 cytochrome P450, family 1, subfamily a, polypeptide 2 Mus musculus 107-113 15855724-3 2005 beta-naphthoflavone (beta-NF) and rifampicin (Rif) were used as typical cytochrome P450 (CYP) inducers for CYP1A2 and CYP3A4, respectively. Rifampin 34-44 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 118-124 15855724-3 2005 beta-naphthoflavone (beta-NF) and rifampicin (Rif) were used as typical cytochrome P450 (CYP) inducers for CYP1A2 and CYP3A4, respectively. Rifampin 46-49 cytochrome P450, family 1, subfamily a, polypeptide 2 Mus musculus 107-113 15778422-1 2005 This study investigated 2 hypotheses about genotype-phenotype relationships for the efflux transporter, P-glycoprotein: (1) the presence of a synonymous C3435T variant in exon 26 of the MDR1 gene correlates to higher plasma concentrations of a P-glycoprotein substrate, dicloxacillin, and (2) the effects of genotypic differences decrease under conditions of P-glycoprotein induction by rifampin. Rifampin 387-395 ATP binding cassette subfamily B member 1 Homo sapiens 186-190 16146350-5 2005 CYP3A4 enzyme activity in noninduced and rifampicin-induced DPX-2 cells was also assessed using Vivid fluorogenic substrates. Rifampin 41-51 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 16146350-6 2005 Significantly elevated CYP3A4 activity levels (2.8-fold +/- 0.2-fold above DMSO-treated cells) were found in DPX-2 cells after 48 hours of exposure to rifampicin, but were undetectable in parental HepG2 cells. Rifampin 151-161 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 23-29 16146350-7 2005 Rifampicin-induced activity levels were found to be suitable for assessing the inhibitory potential of new chemical entities in downstream CYP3A4 inhibition assays. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 139-145 15832810-6 2005 Similarly, the steroid family of orphan nuclear receptors, the constitutive androstane receptor (CAR) and pregnane X receptor (PXR), both heterodimerize with the retinoid X receptor (RXR), are shown to transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 358-366 nuclear receptor subfamily 1 group I member 3 Homo sapiens 63-95 15832810-6 2005 Similarly, the steroid family of orphan nuclear receptors, the constitutive androstane receptor (CAR) and pregnane X receptor (PXR), both heterodimerize with the retinoid X receptor (RXR), are shown to transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 358-366 nuclear receptor subfamily 1 group I member 3 Homo sapiens 97-100 15832810-6 2005 Similarly, the steroid family of orphan nuclear receptors, the constitutive androstane receptor (CAR) and pregnane X receptor (PXR), both heterodimerize with the retinoid X receptor (RXR), are shown to transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 358-366 nuclear receptor subfamily 1 group I member 2 Homo sapiens 106-125 15832810-6 2005 Similarly, the steroid family of orphan nuclear receptors, the constitutive androstane receptor (CAR) and pregnane X receptor (PXR), both heterodimerize with the retinoid X receptor (RXR), are shown to transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 358-366 nuclear receptor subfamily 1 group I member 2 Homo sapiens 127-130 15832810-6 2005 Similarly, the steroid family of orphan nuclear receptors, the constitutive androstane receptor (CAR) and pregnane X receptor (PXR), both heterodimerize with the retinoid X receptor (RXR), are shown to transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 358-366 retinoid X receptor alpha Homo sapiens 162-181 15832810-6 2005 Similarly, the steroid family of orphan nuclear receptors, the constitutive androstane receptor (CAR) and pregnane X receptor (PXR), both heterodimerize with the retinoid X receptor (RXR), are shown to transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 358-366 retinoid X receptor alpha Homo sapiens 183-186 15832810-6 2005 Similarly, the steroid family of orphan nuclear receptors, the constitutive androstane receptor (CAR) and pregnane X receptor (PXR), both heterodimerize with the retinoid X receptor (RXR), are shown to transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 358-366 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 256-261 15832810-6 2005 Similarly, the steroid family of orphan nuclear receptors, the constitutive androstane receptor (CAR) and pregnane X receptor (PXR), both heterodimerize with the retinoid X receptor (RXR), are shown to transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 358-366 nuclear receptor subfamily 1 group I member 3 Homo sapiens 331-334 15832810-6 2005 Similarly, the steroid family of orphan nuclear receptors, the constitutive androstane receptor (CAR) and pregnane X receptor (PXR), both heterodimerize with the retinoid X receptor (RXR), are shown to transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 358-366 nuclear receptor subfamily 1 group I member 2 Homo sapiens 383-386 15635047-4 2005 We found that GBS exposed to rifampin or clindamycin (versus beta-lactam antibiotics) stimulated less TNF secretion and inducible nitric oxide synthase (iNOS) protein accumulation in RAW 264.7 cells. Rifampin 29-37 tumor necrosis factor Mus musculus 102-105 15855724-3 2005 beta-naphthoflavone (beta-NF) and rifampicin (Rif) were used as typical cytochrome P450 (CYP) inducers for CYP1A2 and CYP3A4, respectively. Rifampin 46-49 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 118-124 15726657-9 2005 In contrast, administration of rifampicin markedly induced CYP3A metabolism, resulting in abolished budesonide plasma levels and high urinary excretion of 6beta-hydroxycortisol. Rifampin 31-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 59-64 15635047-4 2005 We found that GBS exposed to rifampin or clindamycin (versus beta-lactam antibiotics) stimulated less TNF secretion and inducible nitric oxide synthase (iNOS) protein accumulation in RAW 264.7 cells. Rifampin 29-37 nitric oxide synthase 2, inducible Mus musculus 120-151 15843291-4 2005 Rifampin is known to induce the metabolism of cytochrome P450 3A4 substrates and thus plays a role in several drug-drug interactions. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 46-65 15635047-4 2005 We found that GBS exposed to rifampin or clindamycin (versus beta-lactam antibiotics) stimulated less TNF secretion and inducible nitric oxide synthase (iNOS) protein accumulation in RAW 264.7 cells. Rifampin 29-37 nitric oxide synthase 2, inducible Mus musculus 153-157 15670600-1 2005 The nuclear receptor pregnane X receptor (PXR) acts as a sensor for a broad variety of natural and synthetic lipophilic compounds, such as bile acids and rifampicin, and regulates the expression of proteins that are involved in the metabolism and transport of these compounds. Rifampin 154-164 nuclear receptor subfamily 1 group I member 2 Homo sapiens 21-40 15670600-1 2005 The nuclear receptor pregnane X receptor (PXR) acts as a sensor for a broad variety of natural and synthetic lipophilic compounds, such as bile acids and rifampicin, and regulates the expression of proteins that are involved in the metabolism and transport of these compounds. Rifampin 154-164 nuclear receptor subfamily 1 group I member 2 Homo sapiens 42-45 15670600-8 2005 Induction of OATP2 and SHP1 mRNA expression by rifampicin confirmed that both genes are primary human PXR responding genes. Rifampin 47-57 solute carrier organic anion transporter family member 1B1 Homo sapiens 13-18 15670600-8 2005 Induction of OATP2 and SHP1 mRNA expression by rifampicin confirmed that both genes are primary human PXR responding genes. Rifampin 47-57 nuclear receptor subfamily 0 group B member 2 Homo sapiens 23-27 15670600-8 2005 Induction of OATP2 and SHP1 mRNA expression by rifampicin confirmed that both genes are primary human PXR responding genes. Rifampin 47-57 nuclear receptor subfamily 1 group I member 2 Homo sapiens 102-105 15710403-4 2005 Rifampicin, which inhibits hIAPP-induced membrane permeabilisation in functional studies, prevents membrane insertion. Rifampin 0-10 islet amyloid polypeptide Homo sapiens 27-32 15652233-9 2005 By employing this cellular model we were able to demonstrate for the first time that OATP1B1 together with MRP2 mediates the trans-cellular transport of rifampicin. Rifampin 153-163 solute carrier organic anion transporter family member 1B1 Homo sapiens 85-92 15652233-9 2005 By employing this cellular model we were able to demonstrate for the first time that OATP1B1 together with MRP2 mediates the trans-cellular transport of rifampicin. Rifampin 153-163 ATP binding cassette subfamily C member 2 Sus scrofa 107-111 15489503-5 2005 This unusual structural feature explains the known promiscuity of Tlc for various ligands, with chemical structures ranging from lipids and retinoids to the macrocyclic antibiotic rifampin and even to microbial siderophores. Rifampin 180-188 lipocalin 1 Homo sapiens 66-69 15676042-1 2005 AIMS: To investigate the potential induction by rifampicin of intestinal CYP2C8, CYP2C9, CYP2D6 and CYP3A4 using preparations of human enterocytes. Rifampin 48-58 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 73-79 15676042-1 2005 AIMS: To investigate the potential induction by rifampicin of intestinal CYP2C8, CYP2C9, CYP2D6 and CYP3A4 using preparations of human enterocytes. Rifampin 48-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 15676042-1 2005 AIMS: To investigate the potential induction by rifampicin of intestinal CYP2C8, CYP2C9, CYP2D6 and CYP3A4 using preparations of human enterocytes. Rifampin 48-58 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 89-95 15676042-1 2005 AIMS: To investigate the potential induction by rifampicin of intestinal CYP2C8, CYP2C9, CYP2D6 and CYP3A4 using preparations of human enterocytes. Rifampin 48-58 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 100-106 15676042-8 2005 Rifampicin administration also resulted in a significant induction of CYP3A4 protein (34.1 +/- 10.7 vs. 113.9 +/- 31.1 pmol mg(-1) protein (P < 0.001, 95% CI: 51.8-107.6)). Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 70-76 15676042-11 2005 Induction of intestinal CYP2C8 and CYP2C9 might contribute in part to rifampicin - mediated drug interactions, in addition to their hepatic counterparts and intestinal and hepatic CYP3A4. Rifampin 70-80 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 24-30 15676042-11 2005 Induction of intestinal CYP2C8 and CYP2C9 might contribute in part to rifampicin - mediated drug interactions, in addition to their hepatic counterparts and intestinal and hepatic CYP3A4. Rifampin 70-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 15523048-5 2005 Transcriptome profiles before and after exposure to the CYP3A transcriptional activators rifampicin, dexamethasone, pregnane-16alpha-carbonitrile, and phenobarbital were subsequently examined. Rifampin 89-99 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 56-61 15331348-0 2005 Mechanism of rifampicin and pregnane X receptor inhibition of human cholesterol 7 alpha-hydroxylase gene transcription. Rifampin 13-23 cytochrome P450 family 7 subfamily A member 1 Homo sapiens 68-99 15331348-2 2005 Rifampicin, a human PXR agonist, inhibits bile acid synthesis and has been used to treat cholestatic diseases. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 20-23 15331348-5 2005 Rifampicin reduced CYP7A1 and small heterodimer partner (SHP; NR02B) mRNA expression suggesting that SHP was not involved in PXR inhibition of CYP7A1. Rifampin 0-10 cytochrome P450 family 7 subfamily A member 1 Homo sapiens 19-25 15331348-5 2005 Rifampicin reduced CYP7A1 and small heterodimer partner (SHP; NR02B) mRNA expression suggesting that SHP was not involved in PXR inhibition of CYP7A1. Rifampin 0-10 nuclear receptor subfamily 0 group B member 2 Homo sapiens 30-55 15331348-5 2005 Rifampicin reduced CYP7A1 and small heterodimer partner (SHP; NR02B) mRNA expression suggesting that SHP was not involved in PXR inhibition of CYP7A1. Rifampin 0-10 nuclear receptor subfamily 0 group B member 2 Homo sapiens 57-60 15331348-6 2005 Rifampicin inhibited CYP7A1 reporter activity and a PXR binding site was localized to the bile acid response element-I. Rifampin 0-10 cytochrome P450 family 7 subfamily A member 1 Homo sapiens 21-27 15331348-7 2005 Mammalian two-hybrid assays revealed that PXR interacted with hepatic nuclear factor 4 alpha (HNF4 alpha, NR2A1) and rifampicin was required. Rifampin 117-127 nuclear receptor subfamily 1 group I member 2 Homo sapiens 42-45 15331348-10 2005 Rifampicin enhanced PXR interaction with HNF4 alpha and reduced PGC-1 alpha interaction with HNF4 alpha. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 20-23 15331348-10 2005 Rifampicin enhanced PXR interaction with HNF4 alpha and reduced PGC-1 alpha interaction with HNF4 alpha. Rifampin 0-10 hepatocyte nuclear factor 4 alpha Homo sapiens 41-51 15331348-10 2005 Rifampicin enhanced PXR interaction with HNF4 alpha and reduced PGC-1 alpha interaction with HNF4 alpha. Rifampin 0-10 PPARG coactivator 1 alpha Homo sapiens 64-75 15331348-10 2005 Rifampicin enhanced PXR interaction with HNF4 alpha and reduced PGC-1 alpha interaction with HNF4 alpha. Rifampin 0-10 hepatocyte nuclear factor 4 alpha Homo sapiens 93-103 15331348-11 2005 Chromatin immunoprecipitation assay showed that PXR, HNF4 alpha, and PGC-1 alpha bound to CYP7A1 chromatin, and rifampicin dissociated PGC-1 alpha from chromatin. Rifampin 112-122 PPARG coactivator 1 alpha Homo sapiens 135-146 15331348-12 2005 These results suggest that activation of PXR by rifampicin promotes PXR interaction with HNF4 alpha and blocks PGC-1 alpha activation with HNF4 alpha and results in inhibition of CYP7A1 gene transcription. Rifampin 48-58 nuclear receptor subfamily 1 group I member 2 Homo sapiens 41-44 15331348-12 2005 These results suggest that activation of PXR by rifampicin promotes PXR interaction with HNF4 alpha and blocks PGC-1 alpha activation with HNF4 alpha and results in inhibition of CYP7A1 gene transcription. Rifampin 48-58 nuclear receptor subfamily 1 group I member 2 Homo sapiens 68-71 15331348-12 2005 These results suggest that activation of PXR by rifampicin promotes PXR interaction with HNF4 alpha and blocks PGC-1 alpha activation with HNF4 alpha and results in inhibition of CYP7A1 gene transcription. Rifampin 48-58 hepatocyte nuclear factor 4 alpha Homo sapiens 89-99 15331348-12 2005 These results suggest that activation of PXR by rifampicin promotes PXR interaction with HNF4 alpha and blocks PGC-1 alpha activation with HNF4 alpha and results in inhibition of CYP7A1 gene transcription. Rifampin 48-58 PPARG coactivator 1 alpha Homo sapiens 111-122 15331348-12 2005 These results suggest that activation of PXR by rifampicin promotes PXR interaction with HNF4 alpha and blocks PGC-1 alpha activation with HNF4 alpha and results in inhibition of CYP7A1 gene transcription. Rifampin 48-58 hepatocyte nuclear factor 4 alpha Homo sapiens 139-149 15331348-12 2005 These results suggest that activation of PXR by rifampicin promotes PXR interaction with HNF4 alpha and blocks PGC-1 alpha activation with HNF4 alpha and results in inhibition of CYP7A1 gene transcription. Rifampin 48-58 cytochrome P450 family 7 subfamily A member 1 Homo sapiens 179-185 16292757-6 2005 Through promoter-reporter gene assays, we found that rifampicin showed multiple-folds activation of a 1.23-kb UGDH promoter construct, the region likely to respond to rifampicin treatment is located within the range -632 to -1,050. Rifampin 53-63 UDP-glucose 6-dehydrogenase Homo sapiens 110-114 15966756-11 2005 RESULTS: Compared with controls, CYP3A induction (or inhibition) decreased (or increased) plasma LAAM concentrations and mean area under the plasma concentration-time curve from time zero to infinity (AUC(infinity) 199 +/- 91 [control] versus 11.3 +/- 4.0 [rifampicin] and 731 +/- 229 ng . Rifampin 257-267 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 33-38 15849716-3 2005 Concentration-dependent changes in UGT1A1 response were evaluated in hepatocyte cultures after treatment with 3-methylchloranthrene, beta-napthoflavone, rifampicin, or phenobarbital. Rifampin 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 15849716-7 2005 UGT1A1 was most responsive to the pregnane-X-receptor-agonists rifampicin, ritonavir, and clotrimazole at the mRNA level and, to a lesser extent, the constitutive androstane receptor-activators, phenobarbital and phenytoin. Rifampin 63-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 15849716-7 2005 UGT1A1 was most responsive to the pregnane-X-receptor-agonists rifampicin, ritonavir, and clotrimazole at the mRNA level and, to a lesser extent, the constitutive androstane receptor-activators, phenobarbital and phenytoin. Rifampin 63-73 nuclear receptor subfamily 1 group I member 2 Homo sapiens 34-53 16292757-9 2005 Cotransfection with the PPARalpha and retinoid X receptor-alpha expression vectors and subsequent treatment with the PPARalpha agonist led to the suppression of the UGDH promoter activity either in the presence or absence of rifampicin. Rifampin 225-235 UDP-glucose 6-dehydrogenase Homo sapiens 165-169 16292757-10 2005 Our study, for the first time, shows the UGDH gene to be under xenobiotic regulation and delineates a motif responsible for rifampicin response and transcriptional repression of the UGDH gene. Rifampin 124-134 UDP-glucose 6-dehydrogenase Homo sapiens 41-45 16292757-6 2005 Through promoter-reporter gene assays, we found that rifampicin showed multiple-folds activation of a 1.23-kb UGDH promoter construct, the region likely to respond to rifampicin treatment is located within the range -632 to -1,050. Rifampin 167-177 UDP-glucose 6-dehydrogenase Homo sapiens 110-114 16292757-8 2005 A mutation at the predicted PPAR recognizing motif eliminated normal suppression as well as the rifampicin activation effect on the UGDH promoter activity. Rifampin 96-106 UDP-glucose 6-dehydrogenase Homo sapiens 132-136 15630458-6 2005 CYP24 mRNA is upregulated in vivo in mice by pregnenolone 16alpha-carbonitrile and dexamethasone, 2 murine PXR agonists, and in vitro in human hepatocytes by rifampicin and hyperforin, 2 human PXR agonists. Rifampin 158-168 cytochrome P450, family 24, subfamily a, polypeptide 1 Mus musculus 0-5 15630458-8 2005 Transfection of PXR in human embryonic kidney cells resulted in rifampicin-mediated induction of CYP24 mRNA. Rifampin 64-74 nuclear receptor subfamily 1 group I member 2 Homo sapiens 16-19 15630458-8 2005 Transfection of PXR in human embryonic kidney cells resulted in rifampicin-mediated induction of CYP24 mRNA. Rifampin 64-74 cytochrome P450 family 24 subfamily A member 1 Homo sapiens 97-102 15448116-10 2004 The inducibility of CYP2C19 by agents (e.g., rifampicin) often taken concurrently with nelfinavir, together with this P450"s known polymorphic nature, may thus be important determinants of nelfinavir"s antiviral potency. Rifampin 45-55 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 20-27 15771232-7 2005 Significant increases (3- to 6-fold) in amodiaquine N-deethylase (a functional probe for CYP2C8 activity) also were observed with clofibric acid, fenofibric acid, and rifampin, in agreement with the mRNA finding. Rifampin 167-175 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 89-95 15771234-6 2005 RESULTS: Rifampin inhibited the P-gp-mediated efflux of [3H]-verapamil, regardless of delivery route (Imax = 62 +/- 6%). Rifampin 9-17 phosphoglycolate phosphatase Mus musculus 32-36 15548381-2 2004 After selection for neomycin resistance, one clone, displaying high luciferase activity in response to rifampicin (RIF), was isolated and the stable expression of hPXR was confirmed by reverse transcription polymerase chain reaction (RT-PCR). Rifampin 103-113 nuclear receptor subfamily 1 group I member 2 Homo sapiens 163-167 15548381-2 2004 After selection for neomycin resistance, one clone, displaying high luciferase activity in response to rifampicin (RIF), was isolated and the stable expression of hPXR was confirmed by reverse transcription polymerase chain reaction (RT-PCR). Rifampin 115-118 nuclear receptor subfamily 1 group I member 2 Homo sapiens 163-167 15322103-3 2004 Consistent with these studies, the antibiotic rifampicin, a ligand for human PXR, reduces hepatic bile acid levels in cholestasis patients. Rifampin 46-56 nuclear receptor subfamily 1 group I member 2 Homo sapiens 77-80 15681896-5 2004 CYP3A4 and CYP3A5 mRNA levels after exposure to 50 microM rifampicin (positive control for CYP3As) were significantly increased by 5.8 (p<0.01) and 2.0 times (p<0.01), respectively, compared with untreated controls. Rifampin 58-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 15681896-5 2004 CYP3A4 and CYP3A5 mRNA levels after exposure to 50 microM rifampicin (positive control for CYP3As) were significantly increased by 5.8 (p<0.01) and 2.0 times (p<0.01), respectively, compared with untreated controls. Rifampin 58-68 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 11-17 15555995-3 2004 provide strong evidence that rifampicin interacts with alpha-synuclein and inhibits its fibrillization. Rifampin 29-39 synuclein alpha Homo sapiens 55-70 15556002-0 2004 Rifampicin inhibits alpha-synuclein fibrillation and disaggregates fibrils. Rifampin 0-10 synuclein alpha Homo sapiens 20-35 15556002-2 2004 We show that the antibiotic rifampicin inhibited alpha-synuclein fibrillation and disaggregated existing fibrils in a concentration-dependent manner. Rifampin 28-38 synuclein alpha Homo sapiens 49-64 15556002-3 2004 Size-exclusion chromatography data indicated that rifampicin stabilized alpha-synuclein as both a monomer and soluble oligomers comprised of partially folded alpha-synuclein. Rifampin 50-60 synuclein alpha Homo sapiens 72-87 15556002-3 2004 Size-exclusion chromatography data indicated that rifampicin stabilized alpha-synuclein as both a monomer and soluble oligomers comprised of partially folded alpha-synuclein. Rifampin 50-60 synuclein alpha Homo sapiens 158-173 15556002-5 2004 These results indicate that rifampicin-mediated inhibition of alpha-synuclein fibrillation and disaggregation of fibrils involves preferential stabilization of monomeric and soluble oligomeric forms, and that rifampicin potentially may have therapeutic application for Parkinson"s disease. Rifampin 28-38 synuclein alpha Homo sapiens 62-77 15531725-7 2004 We identified defective steroid receptor co-activator 1 (SRC1) recruitment to the GR AF-2 in COR L103 cells, although SRC1 was successfully recruited to the steroid X receptor with rifampicin, suggesting a defect in the GR. Rifampin 181-191 nuclear receptor coactivator 1 Homo sapiens 118-122 15322103-6 2004 The coactivator PGC-1 enhanced transcriptional activity of HNF-4, and this enhancement was suppressed by rifampicin-activated PXR. Rifampin 105-115 hepatocyte nuclear factor 4 alpha Homo sapiens 59-64 15322103-6 2004 The coactivator PGC-1 enhanced transcriptional activity of HNF-4, and this enhancement was suppressed by rifampicin-activated PXR. Rifampin 105-115 nuclear receptor subfamily 1 group I member 2 Homo sapiens 126-129 15322103-8 2004 Rifampicin-dependent interaction of PXR with PGC-1 was shown in cells by coimmunoprecipitation, and intranuclear localization studies using confocal microscopy provided further evidence for this interaction. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Sus scrofa 36-39 15322103-8 2004 Rifampicin-dependent interaction of PXR with PGC-1 was shown in cells by coimmunoprecipitation, and intranuclear localization studies using confocal microscopy provided further evidence for this interaction. Rifampin 0-10 PPARG coactivator 1 alpha Sus scrofa 45-50 15309450-4 2004 Carotenoids and retinol revealed a 5-6 fold reporter gene activity in HepG2 cells in comparison to a 7-fold induction by the well-known PXR agonist rifampicin, whereas apo-carotenals and lycopene exerted less or no activation potential. Rifampin 148-158 nuclear receptor subfamily 1 group I member 2 Homo sapiens 136-139 15309450-6 2004 In addition, carotenoid- or retinol-mediated gene expression of PXR-responsive genes like CYP3A4/CYP3A7, CYP3A5, MDR-1 and MRP-2 has been determined in HepG2 cells by RT-PCR with up-regulative properties of beta-carotene or retinol being comparable to or even higher than that of rifampicin. Rifampin 280-290 nuclear receptor subfamily 1 group I member 2 Homo sapiens 64-67 15309450-6 2004 In addition, carotenoid- or retinol-mediated gene expression of PXR-responsive genes like CYP3A4/CYP3A7, CYP3A5, MDR-1 and MRP-2 has been determined in HepG2 cells by RT-PCR with up-regulative properties of beta-carotene or retinol being comparable to or even higher than that of rifampicin. Rifampin 280-290 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-96 15309450-6 2004 In addition, carotenoid- or retinol-mediated gene expression of PXR-responsive genes like CYP3A4/CYP3A7, CYP3A5, MDR-1 and MRP-2 has been determined in HepG2 cells by RT-PCR with up-regulative properties of beta-carotene or retinol being comparable to or even higher than that of rifampicin. Rifampin 280-290 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 105-111 15309450-6 2004 In addition, carotenoid- or retinol-mediated gene expression of PXR-responsive genes like CYP3A4/CYP3A7, CYP3A5, MDR-1 and MRP-2 has been determined in HepG2 cells by RT-PCR with up-regulative properties of beta-carotene or retinol being comparable to or even higher than that of rifampicin. Rifampin 280-290 ATP binding cassette subfamily B member 1 Homo sapiens 113-118 15322103-9 2004 In chromatin immunoprecipitation studies, rifampicin treatment did not inhibit HNF-4 binding to the native promoters of CYP7A1 and CYP8B1 but resulted in dissociation of PGC-1 and concomitant gene repression. Rifampin 42-52 PPARG coactivator 1 alpha Homo sapiens 170-175 15322103-10 2004 Most interestingly, these rifampicin effects were also observed in the phosphoenolpyruvate carboxykinase gene that contains a functional HNF-4-binding site and is central to hepatic gluconeogenesis. Rifampin 26-36 hepatocyte nuclear factor 4 alpha Homo sapiens 137-142 15322103-5 2004 Treatment with rifampicin resulted in repression of endogenous human CYP7A1 expression in HepG2 cells that was reversed by PXR small interfering RNA. Rifampin 15-25 cytochrome P450 family 7 subfamily A member 1 Homo sapiens 69-75 15322103-5 2004 Treatment with rifampicin resulted in repression of endogenous human CYP7A1 expression in HepG2 cells that was reversed by PXR small interfering RNA. Rifampin 15-25 nuclear receptor subfamily 1 group I member 2 Homo sapiens 123-126 15322103-6 2004 The coactivator PGC-1 enhanced transcriptional activity of HNF-4, and this enhancement was suppressed by rifampicin-activated PXR. Rifampin 105-115 PPARG coactivator 1 alpha Homo sapiens 16-21 15342613-3 2004 In part II, 8 different subjects were administered a single dose of 60 mg etoricoxib alone and following daily doses of 600 mg rifampin, a known strong inducer of CYP3A. Rifampin 127-135 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 163-168 15364537-8 2004 Primary cultures of hepatocytes were treated with beta-naphthoflavone, an inducer of CYP1A2, phenobarbital or rifampin, both of which induce CYP2B6, CYP2C9, CYP2C19, and CYP3A4, albeit it to different extents. Rifampin 110-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-147 15364537-8 2004 Primary cultures of hepatocytes were treated with beta-naphthoflavone, an inducer of CYP1A2, phenobarbital or rifampin, both of which induce CYP2B6, CYP2C9, CYP2C19, and CYP3A4, albeit it to different extents. Rifampin 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 15364537-8 2004 Primary cultures of hepatocytes were treated with beta-naphthoflavone, an inducer of CYP1A2, phenobarbital or rifampin, both of which induce CYP2B6, CYP2C9, CYP2C19, and CYP3A4, albeit it to different extents. Rifampin 110-118 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 157-164 15364537-8 2004 Primary cultures of hepatocytes were treated with beta-naphthoflavone, an inducer of CYP1A2, phenobarbital or rifampin, both of which induce CYP2B6, CYP2C9, CYP2C19, and CYP3A4, albeit it to different extents. Rifampin 110-118 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 170-176 15364542-9 2004 Further studies revealed that hPXR-D163G did respond to rifampicin, but required approximately 40-fold higher concentrations than wild-type receptor, suggesting that the ligand-binding domain D163G variant has impaired ligand-binding activity. Rifampin 56-66 nuclear receptor subfamily 1 group I member 2 Homo sapiens 30-34 15252010-3 2004 We report the induction of CYP3A5 mRNA in 13 of 16 hepatocyte preparations exposed to rifampin. Rifampin 86-94 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 27-33 15554232-2 2004 Upon application of this method to patients receiving different kinds of drug therapy, Herbert Remmer was the first to describe that total human hepatic cytochrome P450 was markedly elevated by the new anti-tuberculosis drug rifampicin. Rifampin 225-235 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-168 15554232-4 2004 In 1975, Herbert Remmer"s group described the unique species difference that induction of cytochrome P450 by rifampicin did not occur in the rat. Rifampin 109-119 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 90-105 15554232-9 2004 The inducibility of CYP3A4 by barbiturates and rifampicin explains the effects of inducers to enhance the clearance of ethynylestradiol and thereby to reduce the effectiveness of oral contraceptives, rifampicin being one of the most potent inducers of human CYP3A4 gene expression. Rifampin 47-57 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-26 15554232-9 2004 The inducibility of CYP3A4 by barbiturates and rifampicin explains the effects of inducers to enhance the clearance of ethynylestradiol and thereby to reduce the effectiveness of oral contraceptives, rifampicin being one of the most potent inducers of human CYP3A4 gene expression. Rifampin 47-57 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 258-264 15554232-9 2004 The inducibility of CYP3A4 by barbiturates and rifampicin explains the effects of inducers to enhance the clearance of ethynylestradiol and thereby to reduce the effectiveness of oral contraceptives, rifampicin being one of the most potent inducers of human CYP3A4 gene expression. Rifampin 200-210 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-26 15554232-9 2004 The inducibility of CYP3A4 by barbiturates and rifampicin explains the effects of inducers to enhance the clearance of ethynylestradiol and thereby to reduce the effectiveness of oral contraceptives, rifampicin being one of the most potent inducers of human CYP3A4 gene expression. Rifampin 200-210 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 258-264 15554232-12 2004 It now appears that PXR is a key mediator of complex induction processes of xenobiotic processing enzymes, which are triggered by rifampicin and other inducers. Rifampin 130-140 nuclear receptor subfamily 1 group I member 2 Homo sapiens 20-23 15554232-13 2004 Studies of the structure and substrate affinities of PXR have provided the rational explanation of the unique species difference of rifampicin induction between humans and rats that was first described by Herbert Remmer. Rifampin 132-142 nuclear receptor subfamily 1 group I member 2 Homo sapiens 53-56 15371985-0 2004 Effects of trimethoprim and rifampin on the pharmacokinetics of the cytochrome P450 2C8 substrate rosiglitazone. Rifampin 28-36 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 68-87 15331414-9 2004 The mRNAs for hCYP3A4 and hCYP1A1/2 were induced in the liver in a CYP type-specific manner when the mice were treated with rifampicin and 3-methylcholanthrene, respectively. Rifampin 124-134 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 14-21 15331414-9 2004 The mRNAs for hCYP3A4 and hCYP1A1/2 were induced in the liver in a CYP type-specific manner when the mice were treated with rifampicin and 3-methylcholanthrene, respectively. Rifampin 124-134 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 26-35 15371985-2 2004 Rifampin (INN, rifampicin) is a potent inducer of several CYP enzymes, and in vitro studies have suggested that it also induces CYP2C8. Rifampin 0-8 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 128-134 15371985-3 2004 OBJECTIVE: Our aims were to investigate possible effects of trimethoprim and rifampin on CYP2C8 activity by use of rosiglitazone, a thiazolidinedione antidiabetic drug metabolized primarily by CYP2C8, as an in vivo probe. Rifampin 77-85 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 89-95 15371985-3 2004 OBJECTIVE: Our aims were to investigate possible effects of trimethoprim and rifampin on CYP2C8 activity by use of rosiglitazone, a thiazolidinedione antidiabetic drug metabolized primarily by CYP2C8, as an in vivo probe. Rifampin 77-85 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 193-199 15371985-16 2004 CONCLUSIONS: Trimethoprim raises and rifampin reduces the plasma concentrations of rosiglitazone by inhibiting and inducing, respectively, the CYP2C8-catalyzed biotransformation of rosiglitazone. Rifampin 37-45 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 143-149 16026006-8 2004 Addition of verapamil, an inhibitor for P-glycoprotein (Pgp), multidrug resistance associated protein-2 (MRP-2), and other related transporters, increased absorption of rifampicin in jejunum and ileum by 2-3-fold and decreased secretion by almost 4-fold. Rifampin 169-179 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 56-59 15319341-8 2004 Real-time polymerase chain reaction and Western blot analysis revealed significantly increased CYP3A4 mRNA and protein levels in hepatocytes treated with 10 microM rifampicin compared with untreated cells, but only modest effects of rifampicin on CYP3A5 induction. Rifampin 164-174 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 95-101 15319341-8 2004 Real-time polymerase chain reaction and Western blot analysis revealed significantly increased CYP3A4 mRNA and protein levels in hepatocytes treated with 10 microM rifampicin compared with untreated cells, but only modest effects of rifampicin on CYP3A5 induction. Rifampin 164-174 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 247-253 15319343-5 2004 In intestine, rifampin and SJW induced P-gp expression 3.7- and 1.6-fold and CYP3A 3.5- and 2.4-fold, respectively, whereas dexamethasone and PCN induced CYP3A only. Rifampin 14-22 phosphoglycolate phosphatase Mus musculus 39-43 15319343-5 2004 In intestine, rifampin and SJW induced P-gp expression 3.7- and 1.6-fold and CYP3A 3.5- and 2.4-fold, respectively, whereas dexamethasone and PCN induced CYP3A only. Rifampin 14-22 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 77-82 15319343-5 2004 In intestine, rifampin and SJW induced P-gp expression 3.7- and 1.6-fold and CYP3A 3.5- and 2.4-fold, respectively, whereas dexamethasone and PCN induced CYP3A only. Rifampin 14-22 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 154-159 16026006-8 2004 Addition of verapamil, an inhibitor for P-glycoprotein (Pgp), multidrug resistance associated protein-2 (MRP-2), and other related transporters, increased absorption of rifampicin in jejunum and ileum by 2-3-fold and decreased secretion by almost 4-fold. Rifampin 169-179 ATP binding cassette subfamily C member 2 Rattus norvegicus 105-110 16026006-13 2004 As Pgp varies from person to person to an extent of 2-8-fold, it can be one direct reason for the interindividual variable bioavailability shown by rifampicin. Rifampin 148-158 phosphoglycolate phosphatase Homo sapiens 3-6 15332711-3 2004 Secretion of IL-1beta and TNF-a were significantly inhibited (P<0.002) whereas secretion of IL-6 and IL-10 were significantly increased (P<0.003) by rifampicin treated mononuclear cells. Rifampin 155-165 interleukin 6 Homo sapiens 95-99 15276086-8 2004 Rifampicin, shown to cause clinical drug interactions via a PXR-mediated increase in PGP expression, exhibited a very similar time-course and extent of induction. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 60-63 15276086-8 2004 Rifampicin, shown to cause clinical drug interactions via a PXR-mediated increase in PGP expression, exhibited a very similar time-course and extent of induction. Rifampin 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 85-88 15332711-3 2004 Secretion of IL-1beta and TNF-a were significantly inhibited (P<0.002) whereas secretion of IL-6 and IL-10 were significantly increased (P<0.003) by rifampicin treated mononuclear cells. Rifampin 155-165 interleukin 10 Homo sapiens 104-109 15183191-6 2004 LPS also represses the upregulation of CYP3A11 mRNA levels and erythromycin N-demethylase (ERND) catalytic activity in mice pretreated with PXR ligands dexamethasone, rifampicin, mifepristone, and phenobarbital. Rifampin 167-177 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 39-46 15183191-6 2004 LPS also represses the upregulation of CYP3A11 mRNA levels and erythromycin N-demethylase (ERND) catalytic activity in mice pretreated with PXR ligands dexamethasone, rifampicin, mifepristone, and phenobarbital. Rifampin 167-177 nuclear receptor subfamily 1, group I, member 2 Mus musculus 140-143 15128731-1 2004 Poly (DL-lactide-co-glycolide) (PLG) nanoparticles encapsulating three front-line antitubercular drugs, i.e. rifampicin, isoniazid and pyrazinamide, were prepared by the multiple emulsion technique and administered subcutaneously to mice for pharmacokinetic/chemotherapeutic study. Rifampin 109-119 plasminogen Mus musculus 32-35 15273801-6 2004 After treatment with rifampicina, isoniazida, and pirazinamida, only the levels of IFN-gamma increased significantly (p < 0.01). Rifampin 21-32 interferon gamma Homo sapiens 83-92 15222722-1 2004 OBJECTIVE: It has been demonstrated in preliminary studies that rifampin, a semisynthetic antibiotic and known inducer of hepatic cytochrome P450 3A4, reduces serum concentrations of total bile acids only in individuals with liver disease and elevated serum bile acid levels. Rifampin 64-72 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 130-149 15196345-11 2004 90% (63/70) rifampicin-resistant strains had rpoB gene mutation. Rifampin 12-22 DNA-directed RNA polymerase subunit beta Mycobacterium tuberculosis H37Rv 45-49 15196345-16 2004 CONCLUSION: Gene array might become a rapid, simple, and accurate method for detecting rpoB mutations in most of the rifampicin-resistant Mycobacterium tuberculosis. Rifampin 117-127 DNA-directed RNA polymerase subunit beta Mycobacterium tuberculosis H37Rv 87-91 15180306-1 2004 The expression of CYP3A4 gene is induced by a variety of structurally unrelated xenobiotics including the antibiotic rifampicin, pregnenolone 16-carbonitrile (PCN), and endogenous hormones, that might mediate through steroid and xenobiotic receptor (SXR) system. Rifampin 117-127 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 18-24 15180306-1 2004 The expression of CYP3A4 gene is induced by a variety of structurally unrelated xenobiotics including the antibiotic rifampicin, pregnenolone 16-carbonitrile (PCN), and endogenous hormones, that might mediate through steroid and xenobiotic receptor (SXR) system. Rifampin 117-127 nuclear receptor subfamily 1 group I member 2 Homo sapiens 217-248 15180306-1 2004 The expression of CYP3A4 gene is induced by a variety of structurally unrelated xenobiotics including the antibiotic rifampicin, pregnenolone 16-carbonitrile (PCN), and endogenous hormones, that might mediate through steroid and xenobiotic receptor (SXR) system. Rifampin 117-127 nuclear receptor subfamily 1 group I member 2 Homo sapiens 250-253 15180306-6 2004 In HepG2 cells, CYP3A4 inducers, such as rifampicin, PCN and RU486 showed minimal stimulation of CYP3A4 proximal promoter activity in the absence of SXR and histone deacetylase (HDAC) inhibitors. Rifampin 41-51 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 16-22 15180306-6 2004 In HepG2 cells, CYP3A4 inducers, such as rifampicin, PCN and RU486 showed minimal stimulation of CYP3A4 proximal promoter activity in the absence of SXR and histone deacetylase (HDAC) inhibitors. Rifampin 41-51 histone deacetylase 9 Homo sapiens 178-182 15180306-8 2004 The results of this study demonstrated that both HDAC inhibitors and SXR are essential to increase of CYP3A4 proximal promoter activity by CYP3A4 inducers such as PCN, rifampicin, and RU486. Rifampin 168-178 histone deacetylase 9 Homo sapiens 49-53 15180306-8 2004 The results of this study demonstrated that both HDAC inhibitors and SXR are essential to increase of CYP3A4 proximal promoter activity by CYP3A4 inducers such as PCN, rifampicin, and RU486. Rifampin 168-178 nuclear receptor subfamily 1 group I member 2 Homo sapiens 69-72 15180306-8 2004 The results of this study demonstrated that both HDAC inhibitors and SXR are essential to increase of CYP3A4 proximal promoter activity by CYP3A4 inducers such as PCN, rifampicin, and RU486. Rifampin 168-178 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 102-108 15180306-8 2004 The results of this study demonstrated that both HDAC inhibitors and SXR are essential to increase of CYP3A4 proximal promoter activity by CYP3A4 inducers such as PCN, rifampicin, and RU486. Rifampin 168-178 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 139-145 15180307-8 2004 Taken together, these results indicated that the inhibition of histone deacetylation was required to SXR-mediated increase in CYP3A4 proximal promoter region when rifampicin, or PCN was treated. Rifampin 163-173 nuclear receptor subfamily 1 group I member 2 Homo sapiens 101-104 15180307-8 2004 Taken together, these results indicated that the inhibition of histone deacetylation was required to SXR-mediated increase in CYP3A4 proximal promoter region when rifampicin, or PCN was treated. Rifampin 163-173 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 126-132 14989258-4 2004 In an in vitro system, both vitamin E and rifampicin, a known stimulator of xenobiotic metabolism, activated the pregnane X receptor (PXR), an orphan nuclear receptor. Rifampin 42-52 nuclear receptor subfamily 1 group I member 2 Homo sapiens 113-132 15100175-8 2004 Finally, the role of these sites was examined in activation of CYP3A4 expression by rifampicin, metyrapone, clotrimazole, and phenobarbital. Rifampin 84-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 63-69 15499176-4 2004 Although overexpression of retinoid X receptor alpha (RXRalpha) had no clear effect for both CYP3A reporters, co-expression of apolipoprotein AI regulatory protein-1 (ARP-1) with hPXR resulted in the rifampicin-induced transactivation of the CYP3A1 reporter. Rifampin 200-210 nuclear receptor subfamily 2 group F member 2 Homo sapiens 127-165 15499176-4 2004 Although overexpression of retinoid X receptor alpha (RXRalpha) had no clear effect for both CYP3A reporters, co-expression of apolipoprotein AI regulatory protein-1 (ARP-1) with hPXR resulted in the rifampicin-induced transactivation of the CYP3A1 reporter. Rifampin 200-210 nuclear receptor subfamily 2 group F member 2 Homo sapiens 167-172 15499176-4 2004 Although overexpression of retinoid X receptor alpha (RXRalpha) had no clear effect for both CYP3A reporters, co-expression of apolipoprotein AI regulatory protein-1 (ARP-1) with hPXR resulted in the rifampicin-induced transactivation of the CYP3A1 reporter. Rifampin 200-210 nuclear receptor subfamily 1 group I member 2 Homo sapiens 179-183 15499176-5 2004 A truncated CYP3A4 reporter retaining the both motifs showed the rifampicin-induced transactivation by overexpression of hPXR and ARP-1, while the transactivation in hPXR-overexpressed cells was not observed. Rifampin 65-75 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 12-18 15499176-5 2004 A truncated CYP3A4 reporter retaining the both motifs showed the rifampicin-induced transactivation by overexpression of hPXR and ARP-1, while the transactivation in hPXR-overexpressed cells was not observed. Rifampin 65-75 nuclear receptor subfamily 1 group I member 2 Homo sapiens 121-125 15499176-5 2004 A truncated CYP3A4 reporter retaining the both motifs showed the rifampicin-induced transactivation by overexpression of hPXR and ARP-1, while the transactivation in hPXR-overexpressed cells was not observed. Rifampin 65-75 nuclear receptor subfamily 2 group F member 2 Homo sapiens 130-135 15499176-5 2004 A truncated CYP3A4 reporter retaining the both motifs showed the rifampicin-induced transactivation by overexpression of hPXR and ARP-1, while the transactivation in hPXR-overexpressed cells was not observed. Rifampin 65-75 nuclear receptor subfamily 1 group I member 2 Homo sapiens 166-170 14722322-9 2004 Treatment with 10 microM rifampin resulted in increases in CYP3A4 mRNA (17-fold) and activity (6-beta-hydroxytestoterone formation, 9-fold); and in CYP2C9 mRNA (4-fold) and activity (4"-hydroxydiclofenac formation, 2-fold). Rifampin 25-33 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 59-65 14722322-9 2004 Treatment with 10 microM rifampin resulted in increases in CYP3A4 mRNA (17-fold) and activity (6-beta-hydroxytestoterone formation, 9-fold); and in CYP2C9 mRNA (4-fold) and activity (4"-hydroxydiclofenac formation, 2-fold). Rifampin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 14722322-11 2004 UGT1A mRNA was induced by beta-naphthoflavone (2-fold), and MDR1 (P-glycoprotein) mRNA was induced by rifampin (3-fold). Rifampin 102-110 ATP binding cassette subfamily B member 1 Homo sapiens 60-64 14722322-11 2004 UGT1A mRNA was induced by beta-naphthoflavone (2-fold), and MDR1 (P-glycoprotein) mRNA was induced by rifampin (3-fold). Rifampin 102-110 ATP binding cassette subfamily B member 1 Homo sapiens 66-80 15211874-1 2004 OBJECTIVES: To examine the incidence rate by age and gender of leukopenia caused by chemotherapy including rifampicin (RFP) and isoniazid (INH), and to study the relationships between the leukopenia and the hepatic side effect or other haematological disorders such as thrombocytopenia. Rifampin 107-117 tripartite motif containing 27 Homo sapiens 119-122 14989258-4 2004 In an in vitro system, both vitamin E and rifampicin, a known stimulator of xenobiotic metabolism, activated the pregnane X receptor (PXR), an orphan nuclear receptor. Rifampin 42-52 nuclear receptor subfamily 1 group I member 2 Homo sapiens 134-137 15104253-2 2004 Chrysin- and rifampicin-response activities were traced to the same element as a 290-bp distal enhancer module (-3483/-3194), in which the reporter activities were enhanced by activators of nuclear receptors [constitutive androstane receptor (CAR) and pregnane X receptor (PXR)] and transcription factor [aryl hydrocarbon receptor (AhR)]. Rifampin 13-23 nuclear receptor subfamily 1 group I member 3 Homo sapiens 209-241 15104253-2 2004 Chrysin- and rifampicin-response activities were traced to the same element as a 290-bp distal enhancer module (-3483/-3194), in which the reporter activities were enhanced by activators of nuclear receptors [constitutive androstane receptor (CAR) and pregnane X receptor (PXR)] and transcription factor [aryl hydrocarbon receptor (AhR)]. Rifampin 13-23 nuclear receptor subfamily 1 group I member 3 Homo sapiens 243-246 15104253-2 2004 Chrysin- and rifampicin-response activities were traced to the same element as a 290-bp distal enhancer module (-3483/-3194), in which the reporter activities were enhanced by activators of nuclear receptors [constitutive androstane receptor (CAR) and pregnane X receptor (PXR)] and transcription factor [aryl hydrocarbon receptor (AhR)]. Rifampin 13-23 nuclear receptor subfamily 1 group I member 2 Homo sapiens 252-271 15104253-2 2004 Chrysin- and rifampicin-response activities were traced to the same element as a 290-bp distal enhancer module (-3483/-3194), in which the reporter activities were enhanced by activators of nuclear receptors [constitutive androstane receptor (CAR) and pregnane X receptor (PXR)] and transcription factor [aryl hydrocarbon receptor (AhR)]. Rifampin 13-23 nuclear receptor subfamily 1 group I member 2 Homo sapiens 273-276 15104253-2 2004 Chrysin- and rifampicin-response activities were traced to the same element as a 290-bp distal enhancer module (-3483/-3194), in which the reporter activities were enhanced by activators of nuclear receptors [constitutive androstane receptor (CAR) and pregnane X receptor (PXR)] and transcription factor [aryl hydrocarbon receptor (AhR)]. Rifampin 13-23 aryl hydrocarbon receptor Homo sapiens 305-330 15104253-2 2004 Chrysin- and rifampicin-response activities were traced to the same element as a 290-bp distal enhancer module (-3483/-3194), in which the reporter activities were enhanced by activators of nuclear receptors [constitutive androstane receptor (CAR) and pregnane X receptor (PXR)] and transcription factor [aryl hydrocarbon receptor (AhR)]. Rifampin 13-23 aryl hydrocarbon receptor Homo sapiens 332-335 15104253-6 2004 Chrysin and rifampicin induced the activation of the wild-type reporter gene and T-3263G-mutated gene to a similar extent in HepG2 cells cotransfected with expression vectors of CAR and PXR. Rifampin 12-22 nuclear receptor subfamily 1 group I member 3 Homo sapiens 178-181 15104253-6 2004 Chrysin and rifampicin induced the activation of the wild-type reporter gene and T-3263G-mutated gene to a similar extent in HepG2 cells cotransfected with expression vectors of CAR and PXR. Rifampin 12-22 nuclear receptor subfamily 1 group I member 2 Homo sapiens 186-189 15001973-11 2004 Furthermore, we measured the disposition of long-term oral carvedilol after comedication of the pregnane X receptor ligand rifampin (INN, rifampicin) (600 mg, 9 days), which up-regulates both P-glycoprotein and MRP2 but not CYP2D6. Rifampin 123-131 ATP binding cassette subfamily B member 1 Homo sapiens 192-206 14977870-7 2004 EC50 values for CYP2B6 and CYP3A4 induction by clotrimazole, phenobarbital, phenytoin, and rifampin were strongly correlated (r2 = 0.99) and were statistically indistinguishable for clotrimazole, phenytoin, and rifampin. Rifampin 91-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 14977870-7 2004 EC50 values for CYP2B6 and CYP3A4 induction by clotrimazole, phenobarbital, phenytoin, and rifampin were strongly correlated (r2 = 0.99) and were statistically indistinguishable for clotrimazole, phenytoin, and rifampin. Rifampin 91-99 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 27-33 14977870-7 2004 EC50 values for CYP2B6 and CYP3A4 induction by clotrimazole, phenobarbital, phenytoin, and rifampin were strongly correlated (r2 = 0.99) and were statistically indistinguishable for clotrimazole, phenytoin, and rifampin. Rifampin 211-219 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 14977870-7 2004 EC50 values for CYP2B6 and CYP3A4 induction by clotrimazole, phenobarbital, phenytoin, and rifampin were strongly correlated (r2 = 0.99) and were statistically indistinguishable for clotrimazole, phenytoin, and rifampin. Rifampin 211-219 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 27-33 14977870-8 2004 Kinetic constants governing time-dependent induction by phenobarbital and rifampin were also similar between CYP2B6 and CYP3A4. Rifampin 74-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 15001973-11 2004 Furthermore, we measured the disposition of long-term oral carvedilol after comedication of the pregnane X receptor ligand rifampin (INN, rifampicin) (600 mg, 9 days), which up-regulates both P-glycoprotein and MRP2 but not CYP2D6. Rifampin 123-131 ATP binding cassette subfamily C member 2 Homo sapiens 211-215 15001973-11 2004 Furthermore, we measured the disposition of long-term oral carvedilol after comedication of the pregnane X receptor ligand rifampin (INN, rifampicin) (600 mg, 9 days), which up-regulates both P-glycoprotein and MRP2 but not CYP2D6. Rifampin 123-131 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 224-230 15001973-11 2004 Furthermore, we measured the disposition of long-term oral carvedilol after comedication of the pregnane X receptor ligand rifampin (INN, rifampicin) (600 mg, 9 days), which up-regulates both P-glycoprotein and MRP2 but not CYP2D6. Rifampin 138-148 ATP binding cassette subfamily B member 1 Homo sapiens 192-206 14634033-9 2004 It is concluded that rifampicin limits the hepatic entrance of digoxin and reduced the hepatic exposure of digoxin to CYP3A by inhibiting the basolateral Oatp2 uptake transport, whereas quinidine increased the hepatic exposure of digoxin to CYP3A by inhibiting the canalicular P-gp transport. Rifampin 21-31 solute carrier organic anion transporter family, member 1a4 Rattus norvegicus 154-159 14634033-1 2004 The disposition of digoxin and the influence of the organic anion transporting polypeptide (Oatp)2 inhibitor rifampicin and the P-glycoprotein (P-gp) inhibitor quinidine on its hepatic disposition were examined in the isolated perfused rat liver. Rifampin 109-119 solute carrier organic anion transporter family, member 1a4 Rattus norvegicus 92-98 14634033-9 2004 It is concluded that rifampicin limits the hepatic entrance of digoxin and reduced the hepatic exposure of digoxin to CYP3A by inhibiting the basolateral Oatp2 uptake transport, whereas quinidine increased the hepatic exposure of digoxin to CYP3A by inhibiting the canalicular P-gp transport. Rifampin 21-31 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 277-281 14748817-1 2004 AIMS: Previous work has shown that rifampicin, a potent inducer of several cytochrome P450 (CYP) enzymes and transporters, decreased the plasma concentrations of simvastatin acid by more than 90%. Rifampin 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 14605865-1 2004 OBJECTIVE: This study was carried out to investigate the influence of CYP3A induction with rifampicin on imatinib (Gleevec) exposure. Rifampin 91-101 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 70-75 14748817-1 2004 AIMS: Previous work has shown that rifampicin, a potent inducer of several cytochrome P450 (CYP) enzymes and transporters, decreased the plasma concentrations of simvastatin acid by more than 90%. Rifampin 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 14709631-7 2004 UGT1A9-mediated propofol glucuronidation was induced by phenobarbital (up to 2.2-fold) and rifampicin (up to 1.7-fold). Rifampin 91-101 UDP glucuronosyltransferase family 1 member A9 Homo sapiens 0-6 14600250-0 2004 Induction of human CYP2C9 by rifampicin, hyperforin, and phenobarbital is mediated by the pregnane X receptor. Rifampin 29-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 14600250-0 2004 Induction of human CYP2C9 by rifampicin, hyperforin, and phenobarbital is mediated by the pregnane X receptor. Rifampin 29-39 nuclear receptor subfamily 1 group I member 2 Homo sapiens 90-109 14600250-2 2004 Several studies have reported that certain drugs such as rifampicin and phenobarbital induce CYP2C9, but the molecular basis for this induction remains unknown. Rifampin 57-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 14600250-3 2004 In the present study, we demonstrate that the human pregnane X receptor (hPXR) mediates induction of CYP2C9 by the prototype drugs rifampicin, hyperforin (found in St. John"s Wart), and phenobarbital. Rifampin 131-141 nuclear receptor subfamily 1 group I member 2 Homo sapiens 52-71 14600250-3 2004 In the present study, we demonstrate that the human pregnane X receptor (hPXR) mediates induction of CYP2C9 by the prototype drugs rifampicin, hyperforin (found in St. John"s Wart), and phenobarbital. Rifampin 131-141 nuclear receptor subfamily 1 group I member 2 Homo sapiens 73-77 14600250-3 2004 In the present study, we demonstrate that the human pregnane X receptor (hPXR) mediates induction of CYP2C9 by the prototype drugs rifampicin, hyperforin (found in St. John"s Wart), and phenobarbital. Rifampin 131-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 14600250-4 2004 Deletion and mutagenesis studies with luciferase reporter constructs showed that a functional PXR-responsive element located -1839/-1824 base pairs upstream from the translation start site was the primary binding site mediating the rifampicin induction of CYP2C9. Rifampin 232-242 nuclear receptor subfamily 1 group I member 2 Homo sapiens 94-97 14600250-4 2004 Deletion and mutagenesis studies with luciferase reporter constructs showed that a functional PXR-responsive element located -1839/-1824 base pairs upstream from the translation start site was the primary binding site mediating the rifampicin induction of CYP2C9. Rifampin 232-242 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 256-262 14600250-6 2004 Mutational analysis of 3- and 12-kilobase CYP2C9 promoter fragments indicated that this proximal binding site was essential for rifampicin inducibility, although a cooperative effect could be attributed to a second CAR-RE located at -2899/-2883. Rifampin 128-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 14600250-7 2004 In summary, we have demonstrated rifampicin induction of CYP2C9 promoter constructs that is consistent with the magnitude of induction of CYP2C9 protein and mRNA reported in vivo and in primary human hepatocytes, and we have identified the cis-element essential for this response. Rifampin 33-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 14600250-7 2004 In summary, we have demonstrated rifampicin induction of CYP2C9 promoter constructs that is consistent with the magnitude of induction of CYP2C9 protein and mRNA reported in vivo and in primary human hepatocytes, and we have identified the cis-element essential for this response. Rifampin 33-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 14600250-8 2004 This is the first report to demonstrate that the nuclear receptor PXR mediates induction of CYP2C9 with rifampicin, phenobarbital, and hyperforin. Rifampin 104-114 nuclear receptor subfamily 1 group I member 2 Homo sapiens 66-69 14600250-8 2004 This is the first report to demonstrate that the nuclear receptor PXR mediates induction of CYP2C9 with rifampicin, phenobarbital, and hyperforin. Rifampin 104-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 14707025-6 2004 Individual platelet aggregation was studied in 10 healthy volunteers after the coadministration of clopidogrel and rifampin (a CYP3A4 inducer). Rifampin 115-123 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 127-133 14709618-0 2004 The pregnane X receptor binds to response elements in a genomic context-dependent manner, and PXR activator rifampicin selectively alters the binding among target genes. Rifampin 108-118 nuclear receptor subfamily 1 group I member 2 Homo sapiens 94-97 14709618-10 2004 However, such abundant interactions were markedly diminished when cells were treated with PXR activator rifampicin. Rifampin 104-114 nuclear receptor subfamily 1 group I member 2 Homo sapiens 90-93 14614579-14 2004 CONCLUSION: Rifampicin pretreatment reduces plasma celiprolol concentrations, possibly by induction of the efflux transporter P-glycoprotein, particularly in the intestinal wall, which leads to decreased absorption of celiprolol. Rifampin 12-22 ATP binding cassette subfamily B member 1 Homo sapiens 126-140 15001966-12 2004 CONCLUSION: This study showed that rifampin affected the disposition of rosiglitazone in humans, probably by the induction of cytochrome P450 (CYP) 2C8 and, to a lesser extent, CYP2C9. Rifampin 35-43 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 126-151 15001966-12 2004 CONCLUSION: This study showed that rifampin affected the disposition of rosiglitazone in humans, probably by the induction of cytochrome P450 (CYP) 2C8 and, to a lesser extent, CYP2C9. Rifampin 35-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 177-183 14722235-2 2004 Interestingly, certain PXR ligands such as rifampin have been shown to readily induce human and rabbit but not rodent members of the cytochrome P450 3A. Rifampin 43-51 nuclear receptor subfamily 1 group I member 2 Homo sapiens 23-26 14722235-8 2004 Rifampin, paclitaxel, and hyperforin sensitivity was conferred to rat PXR when Phe305 was converted to leucine, whereas attenuation of sensitivity was observed when Leu308 of human PXR was replaced with phenylalanine. Rifampin 0-8 nuclear receptor subfamily 1, group I, member 2 Rattus norvegicus 70-73 14636322-10 2003 Comparatively, rifampicin (10 microM) effected 14.5-, 25.3-, and a 20.3-fold increase in CYP3A4 activity, immunoreactive protein levels, and mRNA levels, respectively. Rifampin 15-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 89-95 14586385-0 2003 Cytochrome p450 3A4 messenger ribonucleic acid induction by rifampin in human peripheral blood mononuclear cells: correlation with alprazolam pharmacokinetics. Rifampin 60-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-19 12920130-7 2003 The known SXR activators rifampicin and hyperforin induced this panel of bone markers to an extent similar to vitamin K2. Rifampin 25-35 nuclear receptor subfamily 1, group I, member 2 Mus musculus 10-13 14709322-0 2004 Induction of proteins involved in multidrug resistance (P-glycoprotein, MRP1, MRP2, LRP) and of CYP 3A4 by rifampicin in LLC-PK1 cells. Rifampin 107-117 LOW QUALITY PROTEIN: multidrug resistance-associated protein 6 Sus scrofa 72-76 14709322-2 2004 In the LLC-PK(1) tubular renal cell line, a 15-day treatment with 25 microM rifampicin significantly increased the mRNA levels of P-glycoprotein, MRP1, MRP2, LRP and cytochrome P450 3A4 (CYP 3A4). Rifampin 76-86 LOW QUALITY PROTEIN: multidrug resistance-associated protein 6 Sus scrofa 146-150 14709322-2 2004 In the LLC-PK(1) tubular renal cell line, a 15-day treatment with 25 microM rifampicin significantly increased the mRNA levels of P-glycoprotein, MRP1, MRP2, LRP and cytochrome P450 3A4 (CYP 3A4). Rifampin 76-86 ATP binding cassette subfamily C member 2 Sus scrofa 152-156 14705863-6 2003 This explains the risk for drug interactions that is inherent to pharmacotherapy with PXR ligands such as rifampin, phenobarbital, statins, and St. John"s wort. Rifampin 106-114 nuclear receptor subfamily 1 group I member 2 Homo sapiens 86-89 14586385-8 2003 Leukocyte CYP3A4 mRNA was detectable in all samples with a median of 28 molecules per 1 ng total ribonucleic acid before (range, 10-128 molecules per 1 ng total ribonucleic acid) and 50 molecules per 1 ng total ribonucleic acid after (range, 9-484 molecules per 1 ng total ribonucleic acid) rifampin treatment (P <.001). Rifampin 291-299 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 10-16 12814912-4 2003 The PrP(82-146) WT- and PrP(82-146) (127-146) SC-formed cation channels with fast kinetics are Cu2+ sensitive and rifampicin (RIF) insensitive, whereas the time-dependent inactivating channels formed by these same peptides are both Cu2+ and RIF insensitive. Rifampin 114-124 prion protein Homo sapiens 4-7 14534520-8 2003 CONCLUSION: The effect of rifampin on the pharmacokinetics and pharmacodynamics of gliclazide suggests that rifampin affects the disposition of gliclazide in humans, possibly by the induction of cytochrome P450 2C9. Rifampin 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-214 12966371-1 2003 BACKGROUND: The relative susceptibility of intestinal and hepatic cytochrome P450 (CYP) 3A to induction by rifampin (INN, rifampicin), as a function of age and sex, was investigated with the CYP3A substrate midazolam. Rifampin 107-115 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 66-90 12966371-1 2003 BACKGROUND: The relative susceptibility of intestinal and hepatic cytochrome P450 (CYP) 3A to induction by rifampin (INN, rifampicin), as a function of age and sex, was investigated with the CYP3A substrate midazolam. Rifampin 107-115 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 191-196 12966371-1 2003 BACKGROUND: The relative susceptibility of intestinal and hepatic cytochrome P450 (CYP) 3A to induction by rifampin (INN, rifampicin), as a function of age and sex, was investigated with the CYP3A substrate midazolam. Rifampin 122-132 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 66-90 12966371-16 2003 CONCLUSION: Sex-related differences exist in the extent of intestinal and hepatic CYP3A induction by rifampin. Rifampin 101-109 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 82-87 12948020-4 2003 Additional experiments were performed to assess the potential for enhancing brain uptake by inhibiting P-gp with intranasal rifampin. Rifampin 124-132 phosphoglycolate phosphatase Mus musculus 103-107 12948020-7 2003 Co-administration of rifampin enhanced brain uptake of relevant substrates, and resulted in complete elimination of P-gp-mediated transport for 3H- verapamil. Rifampin 21-29 phosphoglycolate phosphatase Mus musculus 116-120 12843640-10 2003 Exposure to rifampicin reduced CYP3A4 mRNA levels for all donors at 0.5% and 2.5% DMSO. Rifampin 12-22 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-37 12843640-11 2003 Exposure to rifampicin also reduced CYP3A5 and CYP3A7 mRNA levels for donors 1 and 3 at 0.5% and 2.5% DMSO. Rifampin 12-22 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 36-42 12843640-11 2003 Exposure to rifampicin also reduced CYP3A5 and CYP3A7 mRNA levels for donors 1 and 3 at 0.5% and 2.5% DMSO. Rifampin 12-22 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 47-53 12814963-4 2003 A range of PXR activation was observed, 0 to 150% of the activation of the known human CYP3A inducer rifampicin. Rifampin 101-111 nuclear receptor subfamily 1 group I member 2 Homo sapiens 11-14 12814963-4 2003 A range of PXR activation was observed, 0 to 150% of the activation of the known human CYP3A inducer rifampicin. Rifampin 101-111 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 87-92 14599235-10 2003 Similarly, treatment of cryopreserved human hepatocytes with rifampicin (25 microM) for 3 d increased testosterone 6 beta-hydroxylase activity (CYP3A4) by five- to eightfold over untreated cultures and increased CYP3A4 mRNA expression by four- to eightfold. Rifampin 61-71 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 144-150 14599235-10 2003 Similarly, treatment of cryopreserved human hepatocytes with rifampicin (25 microM) for 3 d increased testosterone 6 beta-hydroxylase activity (CYP3A4) by five- to eightfold over untreated cultures and increased CYP3A4 mRNA expression by four- to eightfold. Rifampin 61-71 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 212-218 14604466-2 2003 The possibility of cytochrome p450 (CYP) induction by bupropion (10 microM) was evaluated in-vitro by comparing catalytic activity, immunoreactive protein and CYP mRNA levels from human hepatocytes in primary culture versus cells treated with vehicle (0.5% methanol) and with rifampicin (rifampin) as a positive control. Rifampin 276-286 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 14604466-2 2003 The possibility of cytochrome p450 (CYP) induction by bupropion (10 microM) was evaluated in-vitro by comparing catalytic activity, immunoreactive protein and CYP mRNA levels from human hepatocytes in primary culture versus cells treated with vehicle (0.5% methanol) and with rifampicin (rifampin) as a positive control. Rifampin 276-286 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 14604466-2 2003 The possibility of cytochrome p450 (CYP) induction by bupropion (10 microM) was evaluated in-vitro by comparing catalytic activity, immunoreactive protein and CYP mRNA levels from human hepatocytes in primary culture versus cells treated with vehicle (0.5% methanol) and with rifampicin (rifampin) as a positive control. Rifampin 288-296 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 14604466-2 2003 The possibility of cytochrome p450 (CYP) induction by bupropion (10 microM) was evaluated in-vitro by comparing catalytic activity, immunoreactive protein and CYP mRNA levels from human hepatocytes in primary culture versus cells treated with vehicle (0.5% methanol) and with rifampicin (rifampin) as a positive control. Rifampin 288-296 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 14604466-4 2003 CYP2B6 activity, protein and mRNA levels were increased by 2.5, 1.5 and 2.1 fold, respectively, by 20 microM rifampicin. Rifampin 109-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 14604466-7 2003 Rifampicin (20 microM) increased CYP2E1 protein by 2.1 fold, while 10 microM bupropion minimally altered CYP2E1 protein (1.2 fold). Rifampin 0-10 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 33-39 12721332-2 2003 Potential P-gp inducers (rifampin and morphine) were administered subchorionically to P-gp-competent [mdr1a(+/+)] mice to induce P-gp expression in brain; the impact of rifampin pretreatment on brain penetration of verapamil also was evaluated with an in situ brain perfusion technique. Rifampin 25-33 phosphoglycolate phosphatase Mus musculus 10-14 12721332-3 2003 In addition, the effect of single-dose rifampin on P-gp BBB transport activity was assessed with brain perfusion using verapamil and quinidine as model P-gp substrates. Rifampin 39-47 phosphoglycolate phosphatase Mus musculus 51-55 12721332-4 2003 Chronic exposure to rifampin or morphine induced P-gp expression in mouse brain to a modest extent. Rifampin 20-28 phosphoglycolate phosphatase Mus musculus 49-53 12721332-5 2003 However, single-dose rifampin treatment increased the brain uptake of verapamil and quinidine in mdr1a(+/+) mice in a dose- and concentration-dependent manner, consistent with P-gp inhibition. Rifampin 21-29 phosphoglycolate phosphatase Mus musculus 176-180 12721332-6 2003 Maximum inhibition of P-gp-mediated efflux of verapamil by rifampin pretreatment in vivo (150 mg/kg) was approximately 55%, whereas there was only approximately 12% inhibition of P-gp-mediated efflux of quinidine at that rifampin dose. Rifampin 59-67 phosphoglycolate phosphatase Mus musculus 22-26 12721332-6 2003 Maximum inhibition of P-gp-mediated efflux of verapamil by rifampin pretreatment in vivo (150 mg/kg) was approximately 55%, whereas there was only approximately 12% inhibition of P-gp-mediated efflux of quinidine at that rifampin dose. Rifampin 221-229 phosphoglycolate phosphatase Mus musculus 22-26 12721332-7 2003 Coperfusion of rifampin at a concentration of 500 microM abolished P-gp-mediated efflux of verapamil at the BBB. Rifampin 15-23 phosphoglycolate phosphatase Mus musculus 67-71 12721332-8 2003 However, only approximately 40% inhibition of P-gp-mediated efflux of quinidine was observed with coperfusion of rifampin, even at a 2-fold higher rifampin concentration (1000 microM). Rifampin 113-121 phosphoglycolate phosphatase Mus musculus 46-50 12721332-8 2003 However, only approximately 40% inhibition of P-gp-mediated efflux of quinidine was observed with coperfusion of rifampin, even at a 2-fold higher rifampin concentration (1000 microM). Rifampin 147-155 phosphoglycolate phosphatase Mus musculus 46-50 12721332-9 2003 The present studies demonstrate that P-gp function at the BBB can be modulated by rifampin in a dose- and concentration-dependent manner. Rifampin 82-90 phosphoglycolate phosphatase Mus musculus 37-41 12721332-10 2003 The degree to which rifampin inhibits P-gp-mediated transport is dependent on the substrate molecule. Rifampin 20-28 phosphoglycolate phosphatase Mus musculus 38-42 12814912-4 2003 The PrP(82-146) WT- and PrP(82-146) (127-146) SC-formed cation channels with fast kinetics are Cu2+ sensitive and rifampicin (RIF) insensitive, whereas the time-dependent inactivating channels formed by these same peptides are both Cu2+ and RIF insensitive. Rifampin 114-124 prion protein Homo sapiens 24-27 12814912-4 2003 The PrP(82-146) WT- and PrP(82-146) (127-146) SC-formed cation channels with fast kinetics are Cu2+ sensitive and rifampicin (RIF) insensitive, whereas the time-dependent inactivating channels formed by these same peptides are both Cu2+ and RIF insensitive. Rifampin 126-129 prion protein Homo sapiens 4-7 12814912-4 2003 The PrP(82-146) WT- and PrP(82-146) (127-146) SC-formed cation channels with fast kinetics are Cu2+ sensitive and rifampicin (RIF) insensitive, whereas the time-dependent inactivating channels formed by these same peptides are both Cu2+ and RIF insensitive. Rifampin 126-129 prion protein Homo sapiens 24-27 12781341-4 2003 Since this heterodimer then binds to xenobiotic response elements to activate transcription of CYP3A4, we examined a 13kb promoter region of CYP3A4 for responsiveness to dexamethasone and rifampicin. Rifampin 188-198 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 141-147 12869636-2 2003 Previous studies show that compounds such as rifampicin and dexamethasone induce CYP2C19 both in vivo in humans and in vitro in human hepatocytes. Rifampin 45-55 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 81-88 12869636-9 2003 Rifampicin produced a modest increase in promoter activity in cells cotransfected with hPXR. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Homo sapiens 87-91 12815172-4 2003 Colchicine (COL) decreased both basal and rifampicin- and phenobarbital-inducible expression of CYP2B6, CYP2C8/9, and CYP3A4. Rifampin 42-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 12815172-4 2003 Colchicine (COL) decreased both basal and rifampicin- and phenobarbital-inducible expression of CYP2B6, CYP2C8/9, and CYP3A4. Rifampin 42-52 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 104-110 12815172-4 2003 Colchicine (COL) decreased both basal and rifampicin- and phenobarbital-inducible expression of CYP2B6, CYP2C8/9, and CYP3A4. Rifampin 42-52 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 118-124 12781341-8 2003 Co-injection with a human PXR expression vector resulted in a dramatic increase in rifampicin-induced activity and a smaller increase of dexamethasone-induced activity. Rifampin 83-93 nuclear receptor subfamily 1 group I member 2 Homo sapiens 26-29 12781341-9 2003 Co-injection of an antisense murine PXR construct with the CYP3A4-luc reduced both the dexamethasone- and rifampicin-induced responses, thus demonstrating that the murine PXR receptor can participate in the regulation of the human CYP3A4 promoter in mice. Rifampin 106-116 nuclear receptor subfamily 1, group I, member 2 Mus musculus 36-39 12781341-9 2003 Co-injection of an antisense murine PXR construct with the CYP3A4-luc reduced both the dexamethasone- and rifampicin-induced responses, thus demonstrating that the murine PXR receptor can participate in the regulation of the human CYP3A4 promoter in mice. Rifampin 106-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 59-65 12781341-9 2003 Co-injection of an antisense murine PXR construct with the CYP3A4-luc reduced both the dexamethasone- and rifampicin-induced responses, thus demonstrating that the murine PXR receptor can participate in the regulation of the human CYP3A4 promoter in mice. Rifampin 106-116 nuclear receptor subfamily 1, group I, member 2 Mus musculus 171-174 12781341-9 2003 Co-injection of an antisense murine PXR construct with the CYP3A4-luc reduced both the dexamethasone- and rifampicin-induced responses, thus demonstrating that the murine PXR receptor can participate in the regulation of the human CYP3A4 promoter in mice. Rifampin 106-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 231-237 12695351-4 2003 In contrast, treatment with 0.1 microM DEX enhanced CYP2B6 induction by different pregnane X receptor (PXR) activators, including rifampin, phenytoin, clotrimazole, and phenobarbital. Rifampin 130-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 12695351-4 2003 In contrast, treatment with 0.1 microM DEX enhanced CYP2B6 induction by different pregnane X receptor (PXR) activators, including rifampin, phenytoin, clotrimazole, and phenobarbital. Rifampin 130-138 nuclear receptor subfamily 1 group I member 2 Homo sapiens 82-101 12695351-4 2003 In contrast, treatment with 0.1 microM DEX enhanced CYP2B6 induction by different pregnane X receptor (PXR) activators, including rifampin, phenytoin, clotrimazole, and phenobarbital. Rifampin 130-138 nuclear receptor subfamily 1 group I member 2 Homo sapiens 103-106 12673034-3 2003 In this study, we demonstrated the inductive effects of phenobarbital, rifampicin, carbamazepine, phenytoin, prednisolone, ciclosporin and clotrimazole on CYP3A4, CYP3A5 and CYP3A7 mRNA expression, and established the relationship between the expression of human glucocorticoid receptor alpha (hGR) mRNA and the induction of CYP3A4 mRNA in cultured HepG2 cells by reverse transcription polymerase chain reaction (RT-PCR). Rifampin 71-81 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 155-161 12673034-3 2003 In this study, we demonstrated the inductive effects of phenobarbital, rifampicin, carbamazepine, phenytoin, prednisolone, ciclosporin and clotrimazole on CYP3A4, CYP3A5 and CYP3A7 mRNA expression, and established the relationship between the expression of human glucocorticoid receptor alpha (hGR) mRNA and the induction of CYP3A4 mRNA in cultured HepG2 cells by reverse transcription polymerase chain reaction (RT-PCR). Rifampin 71-81 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 163-169 12673034-4 2003 Treatment with prednisolone, rifampicin and carbamazepine rapidly induced the level of CYP3A4 mRNA expression by 3- to 6-fold. Rifampin 29-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 87-93 12673034-3 2003 In this study, we demonstrated the inductive effects of phenobarbital, rifampicin, carbamazepine, phenytoin, prednisolone, ciclosporin and clotrimazole on CYP3A4, CYP3A5 and CYP3A7 mRNA expression, and established the relationship between the expression of human glucocorticoid receptor alpha (hGR) mRNA and the induction of CYP3A4 mRNA in cultured HepG2 cells by reverse transcription polymerase chain reaction (RT-PCR). Rifampin 71-81 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 174-180 12673034-7 2003 Treatment with phenytoin, rifampicin, carbamazepine and ciclosporin induced approximately 2-fold increases in the expression of CYP3A5 mRNA, although prednisolone, phenytoin and clotrimazole had no effect. Rifampin 26-36 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 128-134 12673034-3 2003 In this study, we demonstrated the inductive effects of phenobarbital, rifampicin, carbamazepine, phenytoin, prednisolone, ciclosporin and clotrimazole on CYP3A4, CYP3A5 and CYP3A7 mRNA expression, and established the relationship between the expression of human glucocorticoid receptor alpha (hGR) mRNA and the induction of CYP3A4 mRNA in cultured HepG2 cells by reverse transcription polymerase chain reaction (RT-PCR). Rifampin 71-81 nuclear receptor subfamily 3 group C member 1 Homo sapiens 263-286 12673034-3 2003 In this study, we demonstrated the inductive effects of phenobarbital, rifampicin, carbamazepine, phenytoin, prednisolone, ciclosporin and clotrimazole on CYP3A4, CYP3A5 and CYP3A7 mRNA expression, and established the relationship between the expression of human glucocorticoid receptor alpha (hGR) mRNA and the induction of CYP3A4 mRNA in cultured HepG2 cells by reverse transcription polymerase chain reaction (RT-PCR). Rifampin 71-81 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 325-331 12673034-8 2003 Treatment with rifampicin, phenytoin, clotrimazole and ciclosporin resulted in approximately a 2-fold induction of the CYP3A7 mRNA level. Rifampin 15-25 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 119-125 12721102-9 2003 promazine metabolism in a primary culture of human hepatocytes treated with specific inducers (rifampicin-CYP3A4, CYP2B6 and CYP2C inducer, 2,3,7,8-tetrachlordibenzeno-p-dioxin (TCDD)-CYP1A1/1A2 inducer). Rifampin 95-105 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 106-112 12689656-0 2003 Rifampicin and dexamethasone have similar effects on macrophage phagocytosis of zymosan, but differ in their effects on nitrite and TNF-alpha production. Rifampin 0-10 tumor necrosis factor Homo sapiens 132-141 12689656-6 2003 At high doses, rifampicin moderately suppressed TNF-alpha while dexamethasone inhibited it in a dose-dependent manner, which was ameliorated by the presence of RU486. Rifampin 15-25 tumor necrosis factor Homo sapiens 48-57 12682816-0 2003 Rifampin-impregnated silicone catheters: a potential tool for prevention and treatment of CSF shunt infections. Rifampin 0-8 colony stimulating factor 2 Homo sapiens 90-93 12721102-19 2003 In a primary culture of human hepatocytes, TCDD (a CYP1A subfamily inducer), as well as rifampicin (mainly a CYP3A4 inducer) induced the formation of promazine 5-sulphoxide and N-desmethylpromazine. Rifampin 88-98 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 109-115 12908606-0 2003 Modulation of taurine on CYP3A4 induction by rifampicin in a HepG2 cell line. Rifampin 45-55 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-31 12601364-4 2003 The CYP3A4 messenger RNA (mRNA) expression level 48 hours after rifampicin treatment in the RBF was approximately 100 times higher than that in a monolayer culture. Rifampin 64-74 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 4-10 12601364-6 2003 When testosterone, a substrate for CYP3A4, was added to the rifampicin-treated cell culture, 6 beta-hydroxy testosterone as a metabolite was formed. Rifampin 60-70 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-41 12601364-7 2003 Electrophoretic mobility shift assay (EMSA) with a CYP3A4 ER6 probe demonstrated that relatively high molecular weight complex containing pregnane X receptor (PXR)/retinoid X receptor alpha(RXR alpha), compared with that in the monolayer culture, is possibly generated in the RFB culture of FLC-5 treated with rifampicin. Rifampin 310-320 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 51-57 12601364-7 2003 Electrophoretic mobility shift assay (EMSA) with a CYP3A4 ER6 probe demonstrated that relatively high molecular weight complex containing pregnane X receptor (PXR)/retinoid X receptor alpha(RXR alpha), compared with that in the monolayer culture, is possibly generated in the RFB culture of FLC-5 treated with rifampicin. Rifampin 310-320 nuclear receptor subfamily 1 group I member 2 Homo sapiens 138-157 12601364-7 2003 Electrophoretic mobility shift assay (EMSA) with a CYP3A4 ER6 probe demonstrated that relatively high molecular weight complex containing pregnane X receptor (PXR)/retinoid X receptor alpha(RXR alpha), compared with that in the monolayer culture, is possibly generated in the RFB culture of FLC-5 treated with rifampicin. Rifampin 310-320 nuclear receptor subfamily 1 group I member 2 Homo sapiens 159-162 12601364-7 2003 Electrophoretic mobility shift assay (EMSA) with a CYP3A4 ER6 probe demonstrated that relatively high molecular weight complex containing pregnane X receptor (PXR)/retinoid X receptor alpha(RXR alpha), compared with that in the monolayer culture, is possibly generated in the RFB culture of FLC-5 treated with rifampicin. Rifampin 310-320 retinoid X receptor alpha Homo sapiens 164-189 12601364-7 2003 Electrophoretic mobility shift assay (EMSA) with a CYP3A4 ER6 probe demonstrated that relatively high molecular weight complex containing pregnane X receptor (PXR)/retinoid X receptor alpha(RXR alpha), compared with that in the monolayer culture, is possibly generated in the RFB culture of FLC-5 treated with rifampicin. Rifampin 310-320 retinoid X receptor alpha Homo sapiens 190-199 12504802-4 2003 In HepG2 cells transfected with the human PXR and the chloramphenicol acetyl transferase (CAT) gene linked to two PXR responsive elements, CAT activity was most strongly induced by alpha- and gamma-tocotrienol followed by rifampicin, delta-, alpha- and gamma-tocopherol. Rifampin 222-232 nuclear receptor subfamily 1 group I member 2 Homo sapiens 42-45 12504802-4 2003 In HepG2 cells transfected with the human PXR and the chloramphenicol acetyl transferase (CAT) gene linked to two PXR responsive elements, CAT activity was most strongly induced by alpha- and gamma-tocotrienol followed by rifampicin, delta-, alpha- and gamma-tocopherol. Rifampin 222-232 nuclear receptor subfamily 1 group I member 2 Homo sapiens 114-117 12504802-6 2003 Up-regulation of endogenous CYP3A4 and CYP3A5 mRNA was obtained by gamma-tocotrienol, the most potent activator of PXR, with the same efficacy as with rifampicin. Rifampin 151-161 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 28-34 12504802-6 2003 Up-regulation of endogenous CYP3A4 and CYP3A5 mRNA was obtained by gamma-tocotrienol, the most potent activator of PXR, with the same efficacy as with rifampicin. Rifampin 151-161 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 39-45 12504802-6 2003 Up-regulation of endogenous CYP3A4 and CYP3A5 mRNA was obtained by gamma-tocotrienol, the most potent activator of PXR, with the same efficacy as with rifampicin. Rifampin 151-161 nuclear receptor subfamily 1 group I member 2 Homo sapiens 115-118 12882588-1 2003 The antituberculosis drug rifampicin (rifampin) induces a number of drug-metabolising enzymes, having the greatest effects on the expression of cytochrome P450 (CYP) 3A4 in the liver and in the small intestine. Rifampin 26-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 144-169 13677133-0 2003 [Formation of virulent antigen-modified mutants (Fra-, Fra-Tox-) of plague bacteria resistant to rifampicin and quinolones]. Rifampin 97-107 rabaptin, RAB GTPase binding effector protein 2 Mus musculus 49-52 13677133-0 2003 [Formation of virulent antigen-modified mutants (Fra-, Fra-Tox-) of plague bacteria resistant to rifampicin and quinolones]. Rifampin 97-107 rabaptin, RAB GTPase binding effector protein 2 Mus musculus 55-58 13677133-0 2003 [Formation of virulent antigen-modified mutants (Fra-, Fra-Tox-) of plague bacteria resistant to rifampicin and quinolones]. Rifampin 97-107 thymocyte selection-associated high mobility group box Mus musculus 59-62 12882588-1 2003 The antituberculosis drug rifampicin (rifampin) induces a number of drug-metabolising enzymes, having the greatest effects on the expression of cytochrome P450 (CYP) 3A4 in the liver and in the small intestine. Rifampin 38-46 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 144-169 12882588-2 2003 In addition, rifampicin induces some drug transporter proteins, such as intestinal and hepatic P-glycoprotein. Rifampin 13-23 ATP binding cassette subfamily B member 1 Homo sapiens 95-109 12882588-4 2003 Rifampicin has its greatest effects on the pharmacokinetics of orally administered drugs that are metabolised by CYP3A4 and/or are transported by P-glycoprotein. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 113-119 12882588-4 2003 Rifampicin has its greatest effects on the pharmacokinetics of orally administered drugs that are metabolised by CYP3A4 and/or are transported by P-glycoprotein. Rifampin 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 146-160 12882588-10 2003 Rifampicin also induces CYP2C-mediated metabolism and thus reduces the plasma concentrations of, for example, the CYP2C9 substrate (S)-warfarin and the sulfonylurea antidiabetic drugs. Rifampin 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 12882588-11 2003 In addition, rifampicin can reduce the plasma concentrations of drugs that are not metabolised (e.g. digoxin) by inducing drug transporters such as P-glycoprotein. Rifampin 13-23 ATP binding cassette subfamily B member 1 Homo sapiens 148-162 12645856-0 2003 Molecular basis of rifampicin-induced inhibition of anti-CD95-induced apoptosis of peripheral blood T lymphocytes: the role of CD95 ligand and FLIPs. Rifampin 19-29 Fas cell surface death receptor Homo sapiens 57-61 12486675-0 2003 Unexpected lack of effect of the rifampin-induced P-glycoprotein on the oral bioavailability of its substrate, talinolol, in humans: implication in phenotyping. Rifampin 33-41 ATP binding cassette subfamily B member 1 Homo sapiens 50-64 12490595-1 2003 Rifampin, a member of the rifamycin class of antibiotics, is well known for its ability to induce drug-metabolizing enzymes and transporters, through activation of the pregnane X receptor. Rifampin 0-8 nuclear receptor subfamily 1 group I member 2 Homo sapiens 168-187 12490595-5 2003 Indeed, both OATP-C and OATP8 seemed capable of mediating rifampin uptake into HeLa cells. Rifampin 58-66 solute carrier organic anion transporter family member 1B1 Homo sapiens 13-19 12490595-5 2003 Indeed, both OATP-C and OATP8 seemed capable of mediating rifampin uptake into HeLa cells. Rifampin 58-66 solute carrier organic anion transporter family member 1B3 Homo sapiens 24-29 12490595-8 2003 In cell-based, transactivation assays, OATP-C expression was associated with increased cellular rifampin retention as well as potentiation of PXR reporter gene activity. Rifampin 96-104 solute carrier organic anion transporter family member 1B1 Homo sapiens 39-45 12431788-0 2002 Taurine modulates induction of cytochrome P450 3A4 mRNA by rifampicin in the HepG2 cell line. Rifampin 59-69 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 31-50 12492608-1 2002 AIMS: A noncoding single nucleotide polymorphism (SNP) in exon 26 3435C > T of the highly polymorphic MDR1 gene has been demonstrated to alter digoxin absorption after induction of the MDR1 gene product P-glycoprotein by rifampicin or after multiple oral dosing. Rifampin 224-234 ATP binding cassette subfamily B member 1 Homo sapiens 105-109 12492608-1 2002 AIMS: A noncoding single nucleotide polymorphism (SNP) in exon 26 3435C > T of the highly polymorphic MDR1 gene has been demonstrated to alter digoxin absorption after induction of the MDR1 gene product P-glycoprotein by rifampicin or after multiple oral dosing. Rifampin 224-234 ATP binding cassette subfamily B member 1 Homo sapiens 188-192 12492608-1 2002 AIMS: A noncoding single nucleotide polymorphism (SNP) in exon 26 3435C > T of the highly polymorphic MDR1 gene has been demonstrated to alter digoxin absorption after induction of the MDR1 gene product P-glycoprotein by rifampicin or after multiple oral dosing. Rifampin 224-234 ATP binding cassette subfamily B member 1 Homo sapiens 206-220 12645856-0 2003 Molecular basis of rifampicin-induced inhibition of anti-CD95-induced apoptosis of peripheral blood T lymphocytes: the role of CD95 ligand and FLIPs. Rifampin 19-29 Fas cell surface death receptor Homo sapiens 127-131 12645856-4 2003 Therefore, we examined the effect of rifampicin on signaling pathway of anti-CD95-induced apoptosis in peripheral blood lymphocytes. Rifampin 37-47 Fas cell surface death receptor Homo sapiens 77-81 12645856-5 2003 Rifampicin, in a concentration-dependent manner, inhibited anti-CD95-induced apoptosis in both CD4+ and CD8+ T cells, which was associated with the inhibition of activation of both caspase-3 and caspase-8. Rifampin 0-10 Fas cell surface death receptor Homo sapiens 64-68 12645856-5 2003 Rifampicin, in a concentration-dependent manner, inhibited anti-CD95-induced apoptosis in both CD4+ and CD8+ T cells, which was associated with the inhibition of activation of both caspase-3 and caspase-8. Rifampin 0-10 caspase 3 Homo sapiens 181-190 12645856-5 2003 Rifampicin, in a concentration-dependent manner, inhibited anti-CD95-induced apoptosis in both CD4+ and CD8+ T cells, which was associated with the inhibition of activation of both caspase-3 and caspase-8. Rifampin 0-10 caspase 8 Homo sapiens 195-204 12645856-6 2003 In addition, rifampicin down-regulated the expression of CD95L and Bax. Rifampin 13-23 Fas ligand Homo sapiens 57-62 12645856-6 2003 In addition, rifampicin down-regulated the expression of CD95L and Bax. Rifampin 13-23 BCL2 associated X, apoptosis regulator Homo sapiens 67-70 17535057-9 2003 CONCLUSIONS: The increase in rifampicin levels on administration of a PGP/CYP3A4 blocker suggests a pivotal role for PGP/CYP3A4 in the absorption of rifampicin in patients with TB, which may be responsible for lower or variable levels of rifampicin. Rifampin 29-39 ATP binding cassette subfamily B member 1 Homo sapiens 70-73 12645856-7 2003 The inhibitory effects of rifampicin on apoptosis and caspase activation as well as its effect on the expression of CD95L and FLIPs were reversed by RU486, an antagonist of glucocorticoid receptor. Rifampin 26-36 Fas ligand Homo sapiens 116-121 12645856-7 2003 The inhibitory effects of rifampicin on apoptosis and caspase activation as well as its effect on the expression of CD95L and FLIPs were reversed by RU486, an antagonist of glucocorticoid receptor. Rifampin 26-36 nuclear receptor subfamily 3 group C member 1 Homo sapiens 173-196 17535057-9 2003 CONCLUSIONS: The increase in rifampicin levels on administration of a PGP/CYP3A4 blocker suggests a pivotal role for PGP/CYP3A4 in the absorption of rifampicin in patients with TB, which may be responsible for lower or variable levels of rifampicin. Rifampin 29-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 74-80 17535057-9 2003 CONCLUSIONS: The increase in rifampicin levels on administration of a PGP/CYP3A4 blocker suggests a pivotal role for PGP/CYP3A4 in the absorption of rifampicin in patients with TB, which may be responsible for lower or variable levels of rifampicin. Rifampin 29-39 ATP binding cassette subfamily B member 1 Homo sapiens 117-120 12645856-8 2003 These data suggest that rifampicin inhibits anti-CD95-mediated apoptosis in lymphocytes by modulating the expression of certain proteins that regulate apoptosis, at least in part, via glucocorticoid receptors. Rifampin 24-34 Fas cell surface death receptor Homo sapiens 49-53 17535057-9 2003 CONCLUSIONS: The increase in rifampicin levels on administration of a PGP/CYP3A4 blocker suggests a pivotal role for PGP/CYP3A4 in the absorption of rifampicin in patients with TB, which may be responsible for lower or variable levels of rifampicin. Rifampin 29-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 121-127 12426514-3 2002 Rifampin (INN, rifampicin), a potent enzyme inducer of CYP-mediated metabolism (especially CYP2C9, CYP2C19, and CYP3A4), is known to markedly decrease plasma concentrations and effects of a number coadministered drugs. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 17535057-9 2003 CONCLUSIONS: The increase in rifampicin levels on administration of a PGP/CYP3A4 blocker suggests a pivotal role for PGP/CYP3A4 in the absorption of rifampicin in patients with TB, which may be responsible for lower or variable levels of rifampicin. Rifampin 149-159 ATP binding cassette subfamily B member 1 Homo sapiens 70-73 17535057-9 2003 CONCLUSIONS: The increase in rifampicin levels on administration of a PGP/CYP3A4 blocker suggests a pivotal role for PGP/CYP3A4 in the absorption of rifampicin in patients with TB, which may be responsible for lower or variable levels of rifampicin. Rifampin 149-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 74-80 17535057-9 2003 CONCLUSIONS: The increase in rifampicin levels on administration of a PGP/CYP3A4 blocker suggests a pivotal role for PGP/CYP3A4 in the absorption of rifampicin in patients with TB, which may be responsible for lower or variable levels of rifampicin. Rifampin 149-159 ATP binding cassette subfamily B member 1 Homo sapiens 117-120 17535057-9 2003 CONCLUSIONS: The increase in rifampicin levels on administration of a PGP/CYP3A4 blocker suggests a pivotal role for PGP/CYP3A4 in the absorption of rifampicin in patients with TB, which may be responsible for lower or variable levels of rifampicin. Rifampin 149-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 121-127 17535057-9 2003 CONCLUSIONS: The increase in rifampicin levels on administration of a PGP/CYP3A4 blocker suggests a pivotal role for PGP/CYP3A4 in the absorption of rifampicin in patients with TB, which may be responsible for lower or variable levels of rifampicin. Rifampin 149-159 ATP binding cassette subfamily B member 1 Homo sapiens 70-73 17535057-9 2003 CONCLUSIONS: The increase in rifampicin levels on administration of a PGP/CYP3A4 blocker suggests a pivotal role for PGP/CYP3A4 in the absorption of rifampicin in patients with TB, which may be responsible for lower or variable levels of rifampicin. Rifampin 149-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 74-80 17535057-9 2003 CONCLUSIONS: The increase in rifampicin levels on administration of a PGP/CYP3A4 blocker suggests a pivotal role for PGP/CYP3A4 in the absorption of rifampicin in patients with TB, which may be responsible for lower or variable levels of rifampicin. Rifampin 149-159 ATP binding cassette subfamily B member 1 Homo sapiens 117-120 17535057-9 2003 CONCLUSIONS: The increase in rifampicin levels on administration of a PGP/CYP3A4 blocker suggests a pivotal role for PGP/CYP3A4 in the absorption of rifampicin in patients with TB, which may be responsible for lower or variable levels of rifampicin. Rifampin 149-159 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 121-127 12426516-13 2002 The erythromycin-rifampin interaction cannot be attributed to CYP3A4 induction alone and probably also reflected intestinal P-glycoprotein induction. Rifampin 17-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 62-68 12426516-13 2002 The erythromycin-rifampin interaction cannot be attributed to CYP3A4 induction alone and probably also reflected intestinal P-glycoprotein induction. Rifampin 17-25 ATP binding cassette subfamily B member 1 Homo sapiens 124-138 12426514-3 2002 Rifampin (INN, rifampicin), a potent enzyme inducer of CYP-mediated metabolism (especially CYP2C9, CYP2C19, and CYP3A4), is known to markedly decrease plasma concentrations and effects of a number coadministered drugs. Rifampin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 12426514-3 2002 Rifampin (INN, rifampicin), a potent enzyme inducer of CYP-mediated metabolism (especially CYP2C9, CYP2C19, and CYP3A4), is known to markedly decrease plasma concentrations and effects of a number coadministered drugs. Rifampin 0-8 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 99-106 12426514-3 2002 Rifampin (INN, rifampicin), a potent enzyme inducer of CYP-mediated metabolism (especially CYP2C9, CYP2C19, and CYP3A4), is known to markedly decrease plasma concentrations and effects of a number coadministered drugs. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 112-118 12426514-3 2002 Rifampin (INN, rifampicin), a potent enzyme inducer of CYP-mediated metabolism (especially CYP2C9, CYP2C19, and CYP3A4), is known to markedly decrease plasma concentrations and effects of a number coadministered drugs. Rifampin 15-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 12426514-3 2002 Rifampin (INN, rifampicin), a potent enzyme inducer of CYP-mediated metabolism (especially CYP2C9, CYP2C19, and CYP3A4), is known to markedly decrease plasma concentrations and effects of a number coadministered drugs. Rifampin 15-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 12426514-3 2002 Rifampin (INN, rifampicin), a potent enzyme inducer of CYP-mediated metabolism (especially CYP2C9, CYP2C19, and CYP3A4), is known to markedly decrease plasma concentrations and effects of a number coadministered drugs. Rifampin 15-25 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 99-106 12426514-3 2002 Rifampin (INN, rifampicin), a potent enzyme inducer of CYP-mediated metabolism (especially CYP2C9, CYP2C19, and CYP3A4), is known to markedly decrease plasma concentrations and effects of a number coadministered drugs. Rifampin 15-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 112-118 12426516-0 2002 Cytochrome P450 3A4 and P-glycoprotein mediate the interaction between an oral erythromycin breath test and rifampin. Rifampin 108-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-19 12426516-0 2002 Cytochrome P450 3A4 and P-glycoprotein mediate the interaction between an oral erythromycin breath test and rifampin. Rifampin 108-116 ATP binding cassette subfamily B member 1 Homo sapiens 24-38 12386126-7 2002 Rifampin caused a 13-fold induction of testosterone 6beta-hydroxylase (CYP3A12) activity in vitro and up to a 4.5-fold increase in vivo. Rifampin 0-8 cytochrome P450 3A12 Canis lupus familiaris 71-78 12404239-10 2002 Cyclosporin A, rifampicin, and glibenclamide were proved to be competitive inhibitors of BSEP taurocholate transport, with inhibition constant values of 9.5 micromol/L, 31 micromol/L, and 27.5 micromol/L, respectively. Rifampin 15-25 ATP binding cassette subfamily B member 11 Homo sapiens 89-93 12369894-10 2002 Similarly, the steroid family of orphan receptors, the constitutive androstane receptor (CAR) and pregnane X receptors (PXR), heterodimerize with the retinoid X receptor (RXR), transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 333-341 nuclear receptor subfamily 1 group I member 3 Homo sapiens 55-87 12369894-10 2002 Similarly, the steroid family of orphan receptors, the constitutive androstane receptor (CAR) and pregnane X receptors (PXR), heterodimerize with the retinoid X receptor (RXR), transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 333-341 nuclear receptor subfamily 1 group I member 3 Homo sapiens 89-92 12369894-10 2002 Similarly, the steroid family of orphan receptors, the constitutive androstane receptor (CAR) and pregnane X receptors (PXR), heterodimerize with the retinoid X receptor (RXR), transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 333-341 nuclear receptor subfamily 1 group I member 2 Homo sapiens 98-118 12369894-10 2002 Similarly, the steroid family of orphan receptors, the constitutive androstane receptor (CAR) and pregnane X receptors (PXR), heterodimerize with the retinoid X receptor (RXR), transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 333-341 nuclear receptor subfamily 1 group I member 2 Homo sapiens 120-123 12369894-10 2002 Similarly, the steroid family of orphan receptors, the constitutive androstane receptor (CAR) and pregnane X receptors (PXR), heterodimerize with the retinoid X receptor (RXR), transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 333-341 retinoid X receptor alpha Homo sapiens 150-169 12369894-10 2002 Similarly, the steroid family of orphan receptors, the constitutive androstane receptor (CAR) and pregnane X receptors (PXR), heterodimerize with the retinoid X receptor (RXR), transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 333-341 retinoid X receptor alpha Homo sapiens 171-174 12369894-10 2002 Similarly, the steroid family of orphan receptors, the constitutive androstane receptor (CAR) and pregnane X receptors (PXR), heterodimerize with the retinoid X receptor (RXR), transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 333-341 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 231-236 12369894-10 2002 Similarly, the steroid family of orphan receptors, the constitutive androstane receptor (CAR) and pregnane X receptors (PXR), heterodimerize with the retinoid X receptor (RXR), transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 333-341 nuclear receptor subfamily 1 group I member 3 Homo sapiens 306-309 12369894-10 2002 Similarly, the steroid family of orphan receptors, the constitutive androstane receptor (CAR) and pregnane X receptors (PXR), heterodimerize with the retinoid X receptor (RXR), transcriptionally activate the promoters of CYP2B and CYP3A gene expression by xenobiotics such as phenobarbital-like compounds (CAR) and dexamethasone and rifampin-type of agents (PXR). Rifampin 333-341 nuclear receptor subfamily 1 group I member 2 Homo sapiens 358-361 12169622-5 2002 The S487L change is the predominant change found in rpoB mutations sequenced from rifampin-resistant clinical isolates of Mycobacterium tuberculosis. Rifampin 82-90 RNA polymerase (beta subunit) Bacillus subtilis subsp. subtilis str. 168 52-56 12395981-3 2002 However, an important issue associated with a rifampicin-containing FDC is its quality. Rifampin 46-56 CMD1B Homo sapiens 68-71 12235278-8 2002 Treatment of transformants with various concentrations of rifampicin produced a dose-response curve with maximal induction at 10 microM (5.6 +/- 0.18- and 2.1 +/- 0.3-fold above dimethyl sulfoxide (DMSO)-treated cells in transformants with and without PXR, respectively). Rifampin 58-68 nuclear receptor subfamily 1 group I member 2 Homo sapiens 252-255 12395192-2 2002 We have found that certain rifampicin-resistant ( rif) mutants isolated from either relA or relC strains regain the ability to produce actinorhodin at the same level as the wild-type strain, although their capacity to synthesize ppGpp is unchanged. Rifampin 27-37 RELA proto-oncogene, NF-kB subunit Homo sapiens 84-88 12167558-6 2002 hPXR"s capability of binding to extremely large ligands, such as rifampicin, implies that its binding cavity may be able to expand further through the flexibility of a surface loop. Rifampin 65-75 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-4 12072427-2 2002 Many inducers of human CYP3A4, such as rifampicin, bind to the orphan nuclear receptor SXR (steroid and xenobiotic receptor) as ligands and stimulate transcription on xenobiotic response elements located in the CYP3A4 promoter. Rifampin 39-49 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 23-29 12072427-2 2002 Many inducers of human CYP3A4, such as rifampicin, bind to the orphan nuclear receptor SXR (steroid and xenobiotic receptor) as ligands and stimulate transcription on xenobiotic response elements located in the CYP3A4 promoter. Rifampin 39-49 nuclear receptor subfamily 1 group I member 2 Homo sapiens 87-90 12072427-2 2002 Many inducers of human CYP3A4, such as rifampicin, bind to the orphan nuclear receptor SXR (steroid and xenobiotic receptor) as ligands and stimulate transcription on xenobiotic response elements located in the CYP3A4 promoter. Rifampin 39-49 nuclear receptor subfamily 1 group I member 2 Homo sapiens 92-123 12072427-2 2002 Many inducers of human CYP3A4, such as rifampicin, bind to the orphan nuclear receptor SXR (steroid and xenobiotic receptor) as ligands and stimulate transcription on xenobiotic response elements located in the CYP3A4 promoter. Rifampin 39-49 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 211-217 12072427-5 2002 Cotransfection of SMRT, but not NCoR, inhibited not only basal but also rifampicin-induced transcriptional activity of SXR on the CYP3A4 promoter through specific SMRT-SXR interaction in HepG2 cells. Rifampin 72-82 nuclear receptor corepressor 2 Homo sapiens 18-22 12072427-5 2002 Cotransfection of SMRT, but not NCoR, inhibited not only basal but also rifampicin-induced transcriptional activity of SXR on the CYP3A4 promoter through specific SMRT-SXR interaction in HepG2 cells. Rifampin 72-82 nuclear receptor subfamily 1 group I member 2 Homo sapiens 119-122 12072427-5 2002 Cotransfection of SMRT, but not NCoR, inhibited not only basal but also rifampicin-induced transcriptional activity of SXR on the CYP3A4 promoter through specific SMRT-SXR interaction in HepG2 cells. Rifampin 72-82 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 130-136 12072427-5 2002 Cotransfection of SMRT, but not NCoR, inhibited not only basal but also rifampicin-induced transcriptional activity of SXR on the CYP3A4 promoter through specific SMRT-SXR interaction in HepG2 cells. Rifampin 72-82 nuclear receptor corepressor 2 Homo sapiens 163-167 12072427-5 2002 Cotransfection of SMRT, but not NCoR, inhibited not only basal but also rifampicin-induced transcriptional activity of SXR on the CYP3A4 promoter through specific SMRT-SXR interaction in HepG2 cells. Rifampin 72-82 nuclear receptor subfamily 1 group I member 2 Homo sapiens 168-171 12072427-6 2002 Interestingly, rifampicin also increased the interaction of SXR with SMRT as well as with coactivator SRC-1. Rifampin 15-25 nuclear receptor subfamily 1 group I member 2 Homo sapiens 60-63 12072427-6 2002 Interestingly, rifampicin also increased the interaction of SXR with SMRT as well as with coactivator SRC-1. Rifampin 15-25 nuclear receptor corepressor 2 Homo sapiens 69-73 12072427-6 2002 Interestingly, rifampicin also increased the interaction of SXR with SMRT as well as with coactivator SRC-1. Rifampin 15-25 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 102-107 11991950-3 2002 Full VDR response of various CYP3A4 heterologous/homologous promoter-reporter constructs requires both the proximal ER6 and the distal DR3 motifs, as observed previously with rifampicin-activated PXR. Rifampin 175-185 vitamin D receptor Homo sapiens 5-8 12567899-4 2002 RESULTS: The concentration of rifampicin was linear with ln(F0/F) when concentration was in the range of 10-170 micrograms/mL-1. Rifampin 30-40 L1 cell adhesion molecule Mus musculus 123-127 12130704-2 2002 We used primary cultures of human hepatocytes from 15 subjects to assess the inducibility of CYP2C enzyme expression by prototypical inducer agents, including rifampicin, dexamethasone, and phenobarbital. Rifampin 159-169 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 93-98 12130704-5 2002 Parallel increases in CYP2C mRNAs, measured by Northern blotting and/or RNase protection, were found in rifampicin-treated hepatocytes, with CYP2C8, CYP2C9, and CYP2C19 transcripts exhibiting increases of 688 +/- 635, 207 +/- 49, and 230 +/- 60%, respectively, versus an 8.8-fold enhancement of CYP3A4 mRNA levels. Rifampin 104-114 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 22-27 12130704-5 2002 Parallel increases in CYP2C mRNAs, measured by Northern blotting and/or RNase protection, were found in rifampicin-treated hepatocytes, with CYP2C8, CYP2C9, and CYP2C19 transcripts exhibiting increases of 688 +/- 635, 207 +/- 49, and 230 +/- 60%, respectively, versus an 8.8-fold enhancement of CYP3A4 mRNA levels. Rifampin 104-114 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 141-147 12130704-5 2002 Parallel increases in CYP2C mRNAs, measured by Northern blotting and/or RNase protection, were found in rifampicin-treated hepatocytes, with CYP2C8, CYP2C9, and CYP2C19 transcripts exhibiting increases of 688 +/- 635, 207 +/- 49, and 230 +/- 60%, respectively, versus an 8.8-fold enhancement of CYP3A4 mRNA levels. Rifampin 104-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 12130704-5 2002 Parallel increases in CYP2C mRNAs, measured by Northern blotting and/or RNase protection, were found in rifampicin-treated hepatocytes, with CYP2C8, CYP2C9, and CYP2C19 transcripts exhibiting increases of 688 +/- 635, 207 +/- 49, and 230 +/- 60%, respectively, versus an 8.8-fold enhancement of CYP3A4 mRNA levels. Rifampin 104-114 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 161-168 12130704-5 2002 Parallel increases in CYP2C mRNAs, measured by Northern blotting and/or RNase protection, were found in rifampicin-treated hepatocytes, with CYP2C8, CYP2C9, and CYP2C19 transcripts exhibiting increases of 688 +/- 635, 207 +/- 49, and 230 +/- 60%, respectively, versus an 8.8-fold enhancement of CYP3A4 mRNA levels. Rifampin 104-114 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 295-301 12130704-8 2002 Our results show that CYP2C enzyme expression in human hepatocytes is highly inducible by rifampicin, dexamethasone, and phenobarbital. Rifampin 90-100 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 22-27 12167569-8 2002 This mutation disrupts induction of a reporter gene construct by the glucocorticoids dexamethasone and hydrocortisone; yet induction by the potent PXR ligand rifampicin is unaffected. Rifampin 158-168 nuclear receptor subfamily 1 group I member 2 Homo sapiens 147-150 11991950-3 2002 Full VDR response of various CYP3A4 heterologous/homologous promoter-reporter constructs requires both the proximal ER6 and the distal DR3 motifs, as observed previously with rifampicin-activated PXR. Rifampin 175-185 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-35 11991950-3 2002 Full VDR response of various CYP3A4 heterologous/homologous promoter-reporter constructs requires both the proximal ER6 and the distal DR3 motifs, as observed previously with rifampicin-activated PXR. Rifampin 175-185 TNF receptor superfamily member 25 Homo sapiens 135-138 11991950-3 2002 Full VDR response of various CYP3A4 heterologous/homologous promoter-reporter constructs requires both the proximal ER6 and the distal DR3 motifs, as observed previously with rifampicin-activated PXR. Rifampin 175-185 nuclear receptor subfamily 1 group I member 2 Homo sapiens 196-199 12135489-7 2002 Bsep promoter activity was reduced by rifampin and beta-estradiol. Rifampin 38-46 ATP binding cassette subfamily B member 11 Rattus norvegicus 0-4 12065438-7 2002 Clotrimazole, phenobarbital, rifampin, and sulfinpyrazone highly activated PXR and increased CYP3A4 activity; carbamazepine, dexamethasone, dexamethasone-t-butylacetate, phenytoin, sulfadimidine, and taxol weakly activated PXR and induced CYP3A4 activity, and methotrexate and probenecid showed no marked activation in either system. Rifampin 29-37 nuclear receptor subfamily 1 group I member 2 Homo sapiens 75-78 12065438-7 2002 Clotrimazole, phenobarbital, rifampin, and sulfinpyrazone highly activated PXR and increased CYP3A4 activity; carbamazepine, dexamethasone, dexamethasone-t-butylacetate, phenytoin, sulfadimidine, and taxol weakly activated PXR and induced CYP3A4 activity, and methotrexate and probenecid showed no marked activation in either system. Rifampin 29-37 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-99 12065438-7 2002 Clotrimazole, phenobarbital, rifampin, and sulfinpyrazone highly activated PXR and increased CYP3A4 activity; carbamazepine, dexamethasone, dexamethasone-t-butylacetate, phenytoin, sulfadimidine, and taxol weakly activated PXR and induced CYP3A4 activity, and methotrexate and probenecid showed no marked activation in either system. Rifampin 29-37 nuclear receptor subfamily 1 group I member 2 Homo sapiens 223-226 12065438-7 2002 Clotrimazole, phenobarbital, rifampin, and sulfinpyrazone highly activated PXR and increased CYP3A4 activity; carbamazepine, dexamethasone, dexamethasone-t-butylacetate, phenytoin, sulfadimidine, and taxol weakly activated PXR and induced CYP3A4 activity, and methotrexate and probenecid showed no marked activation in either system. Rifampin 29-37 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 239-245 12085361-2 2002 In rats, rifamycin SV and rifampicin were shown to interfere with hepatic organic anion uptake by inhibition of the organic anion transporting polypeptides Oatp1 and Oatp2. Rifampin 26-36 solute carrier organic anion transporter family, member 1a1 Rattus norvegicus 156-161 12085361-2 2002 In rats, rifamycin SV and rifampicin were shown to interfere with hepatic organic anion uptake by inhibition of the organic anion transporting polypeptides Oatp1 and Oatp2. Rifampin 26-36 solute carrier organic anion transporter family, member 1a4 Rattus norvegicus 166-171 12085361-7 2002 Rifampicin (10 micromol/L) inhibited OATP8-mediated BSP uptake by 50%, whereas inhibition of OATP-C-, OATP-B-, and OATP-A-mediated BSP transport was below 15%. Rifampin 0-10 solute carrier organic anion transporter family member 1B3 Homo sapiens 37-42 12085361-8 2002 100 micromol/L rifampicin inhibited OATP-C- and OATP8-, OATP-B- and OATP-A-mediated BSP uptake by 66%, 96%, 25%, and 49%, respectively. Rifampin 15-25 solute carrier organic anion transporter family member 1B1 Homo sapiens 36-42 12085361-8 2002 100 micromol/L rifampicin inhibited OATP-C- and OATP8-, OATP-B- and OATP-A-mediated BSP uptake by 66%, 96%, 25%, and 49%, respectively. Rifampin 15-25 solute carrier organic anion transporter family member 1B3 Homo sapiens 48-53 12085361-8 2002 100 micromol/L rifampicin inhibited OATP-C- and OATP8-, OATP-B- and OATP-A-mediated BSP uptake by 66%, 96%, 25%, and 49%, respectively. Rifampin 15-25 solute carrier organic anion transporter family member 2B1 Homo sapiens 56-62 12085361-8 2002 100 micromol/L rifampicin inhibited OATP-C- and OATP8-, OATP-B- and OATP-A-mediated BSP uptake by 66%, 96%, 25%, and 49%, respectively. Rifampin 15-25 solute carrier organic anion transporter family member 1A2 Homo sapiens 68-74 12085361-10 2002 Direct transport of rifampicin could be shown for OATP-C (apparent K(m) value 13 micromol/L) and OATP8 (2.3 micromol/L). Rifampin 20-30 solute carrier organic anion transporter family member 1B1 Homo sapiens 50-56 12085361-10 2002 Direct transport of rifampicin could be shown for OATP-C (apparent K(m) value 13 micromol/L) and OATP8 (2.3 micromol/L). Rifampin 20-30 solute carrier organic anion transporter family member 1B3 Homo sapiens 97-102 12051692-4 2002 Phenobarbital induced CYP2B and rifampicin induced CYP3A, respectively, in addition to NADPH-cytochrome P450 reductase. Rifampin 32-42 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 51-56 11978200-11 2002 CYP2A6 activity was markedly inhibited (> 50 %) by pilocarpine, diethyldithio carbamic (DDC), and rifampicin, the IC50 was 5.31 micromol/L, 156.35 micromol/L, and 38.81 micromol/L, respectively. Rifampin 101-111 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 0-6 11978200-15 2002 Not only pilocarpine, CYP2A6 specific inhibitor, but also rifampicin and DDC inhibited CYP2A6 activity selectively. Rifampin 58-68 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 87-93 11950795-12 2002 The efficacy of tamoxifen and 4-hydroxytamoxifen relative to rifampicin for hPXR activation was approximately 30 and 60%, respectively. Rifampin 61-71 nuclear receptor subfamily 1 group I member 2 Homo sapiens 76-80 12040753-6 2002 The levels of CYP1A1, CYP2B6, CYP2C8, CYP3A4, CYP3A5, ADH3, and ABCG1 mRNA in human hepatocytes increased after exposure to rifampicin. Rifampin 124-134 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 14-20 12040753-6 2002 The levels of CYP1A1, CYP2B6, CYP2C8, CYP3A4, CYP3A5, ADH3, and ABCG1 mRNA in human hepatocytes increased after exposure to rifampicin. Rifampin 124-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 12040753-6 2002 The levels of CYP1A1, CYP2B6, CYP2C8, CYP3A4, CYP3A5, ADH3, and ABCG1 mRNA in human hepatocytes increased after exposure to rifampicin. Rifampin 124-134 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 30-36 12040753-6 2002 The levels of CYP1A1, CYP2B6, CYP2C8, CYP3A4, CYP3A5, ADH3, and ABCG1 mRNA in human hepatocytes increased after exposure to rifampicin. Rifampin 124-134 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 38-44 12040753-6 2002 The levels of CYP1A1, CYP2B6, CYP2C8, CYP3A4, CYP3A5, ADH3, and ABCG1 mRNA in human hepatocytes increased after exposure to rifampicin. Rifampin 124-134 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 46-52 12040753-6 2002 The levels of CYP1A1, CYP2B6, CYP2C8, CYP3A4, CYP3A5, ADH3, and ABCG1 mRNA in human hepatocytes increased after exposure to rifampicin. Rifampin 124-134 alcohol dehydrogenase 1C (class I), gamma polypeptide Homo sapiens 54-58 12040753-6 2002 The levels of CYP1A1, CYP2B6, CYP2C8, CYP3A4, CYP3A5, ADH3, and ABCG1 mRNA in human hepatocytes increased after exposure to rifampicin. Rifampin 124-134 ATP binding cassette subfamily G member 1 Homo sapiens 64-69 12040753-7 2002 The levels of CYP1A1, CYP2B6, CYP3A4, CYP3A5, and ABCG1 mRNA recovered after a change to media without rifampicin. Rifampin 103-113 ATP binding cassette subfamily G member 1 Homo sapiens 50-55 11744607-0 2002 Induction of multidrug resistance-1 and cytochrome P450 mRNAs in human mononuclear cells by rifampin. Rifampin 92-100 ATP binding cassette subfamily B member 1 Homo sapiens 13-35 11893771-4 2002 Interestingly, both the antibiotic rifampicin and the herbal antidepressant St. John"s wort activate PXR and have anticholestatic properties, which suggests that more potent, selective PXR agonists may be useful in the treatment of biliary cholestasis or other diseases characterized by the accumulation of bile acids or other toxins in the liver. Rifampin 35-45 nuclear receptor subfamily 1 group I member 2 Homo sapiens 101-104 11893771-4 2002 Interestingly, both the antibiotic rifampicin and the herbal antidepressant St. John"s wort activate PXR and have anticholestatic properties, which suggests that more potent, selective PXR agonists may be useful in the treatment of biliary cholestasis or other diseases characterized by the accumulation of bile acids or other toxins in the liver. Rifampin 35-45 nuclear receptor subfamily 1 group I member 2 Homo sapiens 185-188 11883641-5 2002 whereas digoxin, quinine, and rifampicin preferentially inhibited Oatp2 at low concentrations. Rifampin 30-40 solute carrier organic anion transporter family, member 1a4 Rattus norvegicus 66-71 11679585-3 2002 In previous work, we have shown that CYP2C9 is inducible in primary human hepatocytes by xenobiotics including dexamethasone, rifampicin, and phenobarbital. Rifampin 126-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 11679585-8 2002 Identification of these functional elements provides rational mechanistic basis for CYP2C9 induction by dexamethasone (submicromolar concentrations), and by phenobarbital and rifampicin, respectively. Rifampin 175-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 11836020-1 2002 In the present study, we investigated the inducibility of the drug conjugate transporter genes MRP1 and MRP2 by redox-active compounds such as tertiary butylated hydroquinone (tBHQ) and quercetin and by chemicals known to activate the pregnane X receptor (PXR) such as rifampicin and clotrimazol and by the metalloid compound arsenite. Rifampin 269-279 ATP binding cassette subfamily C member 1 Homo sapiens 95-99 11836020-1 2002 In the present study, we investigated the inducibility of the drug conjugate transporter genes MRP1 and MRP2 by redox-active compounds such as tertiary butylated hydroquinone (tBHQ) and quercetin and by chemicals known to activate the pregnane X receptor (PXR) such as rifampicin and clotrimazol and by the metalloid compound arsenite. Rifampin 269-279 synaptonemal complex central element protein 1 like Homo sapiens 104-108 11836020-2 2002 The human MRP2 gene was found to be inducible in HepG2 cells by rifampicin, clotrimazol, arsenite and tBHQ. Rifampin 64-74 synaptonemal complex central element protein 1 like Homo sapiens 10-14 11829457-4 2002 Marked induction was detected in the cases of dexamethasone, clofibrate, and rifampicin treatments for 96 h both in enzyme activity (178, 176, and 168%) and in UGT1A1 protein level (362, 328, and 250%). Rifampin 77-87 UDP glucuronosyltransferase family 1 member A1 Rattus norvegicus 160-166 12206135-3 2002 In addition, MRP2 levels are altered in hepatocytes in response to hormones such as glucocorticoids and to structurally unrelated drugs such as rifampicin, phenobarbital, ritonavir, and cisplatin. Rifampin 144-154 ATP binding cassette subfamily C member 2 Homo sapiens 13-17 11744607-0 2002 Induction of multidrug resistance-1 and cytochrome P450 mRNAs in human mononuclear cells by rifampin. Rifampin 92-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 11744607-5 2002 A significant (P < 0.05; n = 37) increase in the expression of MDR1 mRNA from 176,900 +/- 122,000 to 248,500 +/- 162,300 molecules/microg of RNA was observed following rifampin administration in the human lymphocytes. Rifampin 171-179 ATP binding cassette subfamily B member 1 Homo sapiens 66-70 11744607-11 2002 The mRNA of CYP2E1, CYP3A5, CYP3A7, CYP4A11, and CYP4B1 genes were variably expressed at baseline and following rifampin treatment. Rifampin 112-120 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 12-18 11744607-11 2002 The mRNA of CYP2E1, CYP3A5, CYP3A7, CYP4A11, and CYP4B1 genes were variably expressed at baseline and following rifampin treatment. Rifampin 112-120 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 20-26 11744607-11 2002 The mRNA of CYP2E1, CYP3A5, CYP3A7, CYP4A11, and CYP4B1 genes were variably expressed at baseline and following rifampin treatment. Rifampin 112-120 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 28-34 11744607-11 2002 The mRNA of CYP2E1, CYP3A5, CYP3A7, CYP4A11, and CYP4B1 genes were variably expressed at baseline and following rifampin treatment. Rifampin 112-120 cytochrome P450 family 4 subfamily A member 11 Homo sapiens 36-43 11744607-11 2002 The mRNA of CYP2E1, CYP3A5, CYP3A7, CYP4A11, and CYP4B1 genes were variably expressed at baseline and following rifampin treatment. Rifampin 112-120 cytochrome P450 family 4 subfamily B member 1 Homo sapiens 49-55 11744607-12 2002 In contrast, CYP2C9 and CYP3A4 mRNAs were undetectable in lymphocytes both before and after rifampin dosing. Rifampin 92-100 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-30 11389115-2 2001 In this study, Poly (DL-lactide-co-glycolide) (PLG) microparticles containing a combination of isoniazid and rifampicin were developed as sustained release carrier systems. Rifampin 109-119 plasminogen Mus musculus 47-50 11958592-0 2002 Rifampicin inhibits CD95-mediated apoptosis of Jurkat T cells via glucocorticoid receptors by modifying the expression of molecules regulating apoptosis. Rifampin 0-10 Fas cell surface death receptor Homo sapiens 20-24 11958592-5 2002 Rifampicin, in a concentration-dependent manner, inhibited anti-CD95-induced apoptosis. Rifampin 0-10 Fas cell surface death receptor Homo sapiens 64-68 11958592-6 2002 Furthermore, rifampicin down-regulated the expression of Bax and CD95L and up-regulated the expression of Bcl-2, Bcl-xL, and Flice-inhibitory protein-L (FLIPL); however, rifampicin had no effect on CD95 or XIAP expression. Rifampin 13-23 BCL2 associated X, apoptosis regulator Homo sapiens 57-60 11958592-6 2002 Furthermore, rifampicin down-regulated the expression of Bax and CD95L and up-regulated the expression of Bcl-2, Bcl-xL, and Flice-inhibitory protein-L (FLIPL); however, rifampicin had no effect on CD95 or XIAP expression. Rifampin 13-23 Fas ligand Homo sapiens 65-70 11958592-6 2002 Furthermore, rifampicin down-regulated the expression of Bax and CD95L and up-regulated the expression of Bcl-2, Bcl-xL, and Flice-inhibitory protein-L (FLIPL); however, rifampicin had no effect on CD95 or XIAP expression. Rifampin 13-23 BCL2 apoptosis regulator Homo sapiens 106-111 11958592-6 2002 Furthermore, rifampicin down-regulated the expression of Bax and CD95L and up-regulated the expression of Bcl-2, Bcl-xL, and Flice-inhibitory protein-L (FLIPL); however, rifampicin had no effect on CD95 or XIAP expression. Rifampin 13-23 BCL2 like 1 Homo sapiens 113-119 11958592-6 2002 Furthermore, rifampicin down-regulated the expression of Bax and CD95L and up-regulated the expression of Bcl-2, Bcl-xL, and Flice-inhibitory protein-L (FLIPL); however, rifampicin had no effect on CD95 or XIAP expression. Rifampin 13-23 Fas cell surface death receptor Homo sapiens 65-69 11958592-6 2002 Furthermore, rifampicin down-regulated the expression of Bax and CD95L and up-regulated the expression of Bcl-2, Bcl-xL, and Flice-inhibitory protein-L (FLIPL); however, rifampicin had no effect on CD95 or XIAP expression. Rifampin 13-23 X-linked inhibitor of apoptosis Homo sapiens 206-210 11958592-10 2002 Taken together, these findings suggest that rifampicin inhibits anti-CD95-induced apoptosis in Jurkat T cells by modulating the expression of various molecules regulating apoptosis and its effect appears to be mediated via GR and at least in part through NF-kappaB signaling pathway. Rifampin 44-54 Fas cell surface death receptor Homo sapiens 69-73 11714868-5 2001 The oligonucleotide-based array made it possible to detect different levels of induction within the CYP2C family, with rifampin causing a 6.5-fold increase in expression of CYP2C8 and a 3.7-fold increase in CYP2C9 while having no effect on the level of CYP2C18 mRNA. Rifampin 119-127 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 173-179 11714868-5 2001 The oligonucleotide-based array made it possible to detect different levels of induction within the CYP2C family, with rifampin causing a 6.5-fold increase in expression of CYP2C8 and a 3.7-fold increase in CYP2C9 while having no effect on the level of CYP2C18 mRNA. Rifampin 119-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 207-213 11714868-5 2001 The oligonucleotide-based array made it possible to detect different levels of induction within the CYP2C family, with rifampin causing a 6.5-fold increase in expression of CYP2C8 and a 3.7-fold increase in CYP2C9 while having no effect on the level of CYP2C18 mRNA. Rifampin 119-127 cytochrome P450 family 2 subfamily C member 18 Homo sapiens 253-260 11714868-6 2001 Rifampin also induced other CYP enzymes including CYP2B6 and all three members of the CYP3A family, with CYP3A4 showing the highest level of induction at 55.1-fold. Rifampin 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 11714868-6 2001 Rifampin also induced other CYP enzymes including CYP2B6 and all three members of the CYP3A family, with CYP3A4 showing the highest level of induction at 55.1-fold. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-111 11668216-12 2001 The relevance of each of the 38 PXR SNPs identified in DNA of individuals whose CYP3A basal and rifampin-inducible CYP3A4 expression was determined in vivo and/or in vitro was demonstrated by univariate statistical analysis. Rifampin 96-104 nuclear receptor subfamily 1 group I member 2 Homo sapiens 32-35 11668216-12 2001 The relevance of each of the 38 PXR SNPs identified in DNA of individuals whose CYP3A basal and rifampin-inducible CYP3A4 expression was determined in vivo and/or in vitro was demonstrated by univariate statistical analysis. Rifampin 96-104 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 115-121 11502872-4 2001 In primary cultures of human hepatocytes, CYP2B6 was highly inducible by a number of compounds known to be human PXR ligands, including rifampicin and hyperforin. Rifampin 136-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 11502872-4 2001 In primary cultures of human hepatocytes, CYP2B6 was highly inducible by a number of compounds known to be human PXR ligands, including rifampicin and hyperforin. Rifampin 136-146 nuclear receptor subfamily 1 group I member 2 Homo sapiens 113-116 11503007-3 2001 The aim of this study was to investigate modulation of the CYP-mediated metabolism of ifosfamide with ketoconazole, a potent inhibitor of CYP3A4, and rifampin (INN, rifampicin), an inducer of CYP3A4/CYP2B6. Rifampin 150-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 199-205 11471773-5 2001 METHODS: Pharmacokinetic parameters of midazolam and 6beta-OH cortisol urinary ratio were evaluated in 8 volunteers before and after 6 days treatment with rifampin, a potent inducer of CYP3A, and after cessation of rifampin treatment. Rifampin 155-163 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 185-190 12405865-13 2002 Rifampicin (rifampin), phenobarbital, phenytoin, carbamazepine, nevirapine, and efavirenz decrease methadone blood concentrations, probably by induction of CYP3A4 activity, which can result in severe withdrawal symptoms. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 156-162 12405865-13 2002 Rifampicin (rifampin), phenobarbital, phenytoin, carbamazepine, nevirapine, and efavirenz decrease methadone blood concentrations, probably by induction of CYP3A4 activity, which can result in severe withdrawal symptoms. Rifampin 12-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 156-162 11965845-2 2002 In view of the worldwide re-emergency of tuberculosis (TB) and the rise of pulmonary TB (TBP) resistant to the two main first line drugs, namely isoniazid (H) and rifampin (R), so called multidrug resistant TBP (MDR-TBP), it was considered necessary to carry out a national, non randomized, multicenter, prospective pilot survey to determine the rates of resistant Mycobacterium tuberculosis (MTB) in patients with no previous antituberculous treatment (primary resistance). Rifampin 163-171 TATA-box binding protein Homo sapiens 89-92 11965845-2 2002 In view of the worldwide re-emergency of tuberculosis (TB) and the rise of pulmonary TB (TBP) resistant to the two main first line drugs, namely isoniazid (H) and rifampin (R), so called multidrug resistant TBP (MDR-TBP), it was considered necessary to carry out a national, non randomized, multicenter, prospective pilot survey to determine the rates of resistant Mycobacterium tuberculosis (MTB) in patients with no previous antituberculous treatment (primary resistance). Rifampin 163-171 TATA-box binding protein Homo sapiens 207-210 11965845-2 2002 In view of the worldwide re-emergency of tuberculosis (TB) and the rise of pulmonary TB (TBP) resistant to the two main first line drugs, namely isoniazid (H) and rifampin (R), so called multidrug resistant TBP (MDR-TBP), it was considered necessary to carry out a national, non randomized, multicenter, prospective pilot survey to determine the rates of resistant Mycobacterium tuberculosis (MTB) in patients with no previous antituberculous treatment (primary resistance). Rifampin 163-171 TATA-box binding protein Homo sapiens 207-210 11868391-6 2002 For human CYP3A4 genes, pregnane X receptor (PXR) binds to the xenobiotic-responsive enhancer module (XREM) and upon induction by rifampicin, a PXR:RXR heterodimer could transactivate XREM. Rifampin 130-140 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 10-16 11868391-6 2002 For human CYP3A4 genes, pregnane X receptor (PXR) binds to the xenobiotic-responsive enhancer module (XREM) and upon induction by rifampicin, a PXR:RXR heterodimer could transactivate XREM. Rifampin 130-140 nuclear receptor subfamily 1 group I member 2 Homo sapiens 24-43 11868391-6 2002 For human CYP3A4 genes, pregnane X receptor (PXR) binds to the xenobiotic-responsive enhancer module (XREM) and upon induction by rifampicin, a PXR:RXR heterodimer could transactivate XREM. Rifampin 130-140 nuclear receptor subfamily 1 group I member 2 Homo sapiens 45-48 11868391-6 2002 For human CYP3A4 genes, pregnane X receptor (PXR) binds to the xenobiotic-responsive enhancer module (XREM) and upon induction by rifampicin, a PXR:RXR heterodimer could transactivate XREM. Rifampin 130-140 nuclear receptor subfamily 1 group I member 2 Homo sapiens 144-147 11868391-6 2002 For human CYP3A4 genes, pregnane X receptor (PXR) binds to the xenobiotic-responsive enhancer module (XREM) and upon induction by rifampicin, a PXR:RXR heterodimer could transactivate XREM. Rifampin 130-140 retinoid X receptor alpha Homo sapiens 148-151 11602520-6 2001 Known ligands for pregnane X receptor (PXR) (rifampin, dexamethasone, and dexamethasone t-butyl acetate) markedly induced CYP3A4 expression in human hepatocytes. Rifampin 45-53 nuclear receptor subfamily 1 group I member 2 Homo sapiens 18-37 11602520-6 2001 Known ligands for pregnane X receptor (PXR) (rifampin, dexamethasone, and dexamethasone t-butyl acetate) markedly induced CYP3A4 expression in human hepatocytes. Rifampin 45-53 nuclear receptor subfamily 1 group I member 2 Homo sapiens 39-42 11602520-6 2001 Known ligands for pregnane X receptor (PXR) (rifampin, dexamethasone, and dexamethasone t-butyl acetate) markedly induced CYP3A4 expression in human hepatocytes. Rifampin 45-53 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 122-128 11601668-5 2001 However, potent inducers of CYP, such as rifampin, may decrease rofecoxib concentrations because of induction of general hepatic metabolic activity. Rifampin 41-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 11503007-3 2001 The aim of this study was to investigate modulation of the CYP-mediated metabolism of ifosfamide with ketoconazole, a potent inhibitor of CYP3A4, and rifampin (INN, rifampicin), an inducer of CYP3A4/CYP2B6. Rifampin 150-158 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 192-198 11389115-3 2001 A single dose of PLG microparticles exhibited a sustained release of isoniazid and rifampicin in vivo up to 7 and 6 weeks, respectively. Rifampin 83-93 plasminogen Mus musculus 17-20 11336975-1 2001 Induction of P450 isoforms 1A (CYP1A) and 3A (CYP3A) by model inducers dexamethasone, omeprazole and rifampin was evaluated in primary cultured hepatocytes from man and laboratory animals. Rifampin 101-109 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 46-51 11336975-3 2001 Results with human hepatocytes from six human donors consistently show that both rifampin and dexamethasone were inducers of CYP3A activity (measured as testosterone 6beta-hydroxylase activity), with rifampin being more potent. Rifampin 81-89 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 125-130 11336976-5 2001 Pregnenolone 16alpha-carbonitrile, a known CYP3A inducer in rodents, is a very efficacious activator of mouse and rat PXR, whereas rifampicin, a known inducer of CYP3A in humans and rabbits, is a very efficacious activator of human and rabbit PXR. Rifampin 131-141 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 162-167 11336977-5 2001 The activity of intestinal 6beta-OHT in both control and Rif-treated dogs was specifically inhibited by anti-CYP3A12 antiserum. Rifampin 57-60 cytochrome P450 3A12 Canis lupus familiaris 109-116 11336977-10 2001 These results demonstrate that Rif induces not only hepatic CYP3A12 but also intestinal CYP3A in dogs. Rifampin 31-34 cytochrome P450 3A12 Canis lupus familiaris 60-67 11745918-8 2001 Rifampin increased CYP3A6 activity by 2- to 3-fold in hepatic microsomes compared to vehicle. Rifampin 0-8 cytochrome P450 3A6 Oryctolagus cuniculus 19-25 11297522-4 2001 So far, the mechanisms by which rifampin induces MDR1 expression are poorly understood. Rifampin 32-40 ATP binding cassette subfamily B member 1 Homo sapiens 49-53 11297522-7 2001 The human colon carcinoma cell line LS174T was used as an intestinal model to study induction because in these cells the endogenous MDR1 gene is highly inducible by rifampin. Rifampin 165-173 ATP binding cassette subfamily B member 1 Homo sapiens 132-136 11491386-9 2001 Since CYP3A6 is mainly induced by rifampicin in rabbit-cultured hepatocytes, the data suggest that this isoform is involved in the biotransformation of MDEA to 3OH-MDEA. Rifampin 34-44 cytochrome P450 3A6 Oryctolagus cuniculus 6-12 11745918-3 2001 The rabbit isoform CYP3A6 and the human isoform CYP3A4 have similar P-450 predominance and substrate specificity and are both induced by rifampin. Rifampin 137-145 cytochrome P450 3A6 Oryctolagus cuniculus 19-25 11745918-3 2001 The rabbit isoform CYP3A6 and the human isoform CYP3A4 have similar P-450 predominance and substrate specificity and are both induced by rifampin. Rifampin 137-145 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-54 11745918-5 2001 We determined the in vivo and ex vivo CYP3A6 induction by 4 days of treatment with rifabutin (100 mg/kg), rifampin (100 mg/kg), or vehicle (DMSO) in the rabbit. Rifampin 106-114 cytochrome P450 3A6 Oryctolagus cuniculus 38-44 11322768-7 2001 Of the MRP inhibitors including probenecid, ofloxacin, erythromycin, and rifampicin used in this study, only probenecid showed a marked chemosensitizing effect in AML-2/DX100 but not in HL-60/Adr, suggesting that the chemosensitizing effects of the MRP inhibitors vary according to the type of resistant cells. Rifampin 73-83 RUNX family transcription factor 3 Homo sapiens 163-168 11323161-4 2001 Furthermore, the antibiotic rifampicin was identified as MRP2 inducer in HepG2 cells. Rifampin 28-38 ATP binding cassette subfamily C member 2 Homo sapiens 57-61 11296180-9 2001 Treatment with clarithromycin combined with two or three other antibiotics, including ethambutol, rifampicin, ofloxacin, or ciprofloxacin, for at least 6 months resulted in a significant fall in plasma HBD-2 concentrations in responders, but not in nonresponders. Rifampin 98-108 defensin beta 4A Homo sapiens 202-207 11323161-6 2001 However, MRP3 and 5 mRNAs were detected in addition to MRP2 and their expression was found to be increased by 2-AAF, cisplatin and rifampicin. Rifampin 131-141 ATP binding cassette subfamily C member 3 Homo sapiens 9-19 11323161-6 2001 However, MRP3 and 5 mRNAs were detected in addition to MRP2 and their expression was found to be increased by 2-AAF, cisplatin and rifampicin. Rifampin 131-141 ATP binding cassette subfamily C member 2 Homo sapiens 55-59 11181490-4 2001 Compounds known to activate the pregnane X receptor (PXR) such as rifampicin, or the constitutively activated receptor (CAR) such as phenobarbital, induced CYP2C8, CYP2C9, and to a lesser extent CYP2C19 mRNAs and proteins. Rifampin 66-76 nuclear receptor subfamily 1 group I member 2 Homo sapiens 32-51 11257517-5 2001 Treatment with phenobarbitone caused CYP2B, CYP2C-2 and CYP3A21 levels to increase, rifampicin caused increases in CYP2B and CYP2C-1 and a decrease in CYP3A21 levels, whereas dioxin caused CYP1A1 and CYP1A2 levels to increase and CYP2E1 levels to decrease. Rifampin 84-94 cytochrome P450 3A21 Callithrix jacchus 151-158 11257517-5 2001 Treatment with phenobarbitone caused CYP2B, CYP2C-2 and CYP3A21 levels to increase, rifampicin caused increases in CYP2B and CYP2C-1 and a decrease in CYP3A21 levels, whereas dioxin caused CYP1A1 and CYP1A2 levels to increase and CYP2E1 levels to decrease. Rifampin 84-94 cytochrome P450 family 1 subfamily A member 1 Callithrix jacchus 189-195 11257517-5 2001 Treatment with phenobarbitone caused CYP2B, CYP2C-2 and CYP3A21 levels to increase, rifampicin caused increases in CYP2B and CYP2C-1 and a decrease in CYP3A21 levels, whereas dioxin caused CYP1A1 and CYP1A2 levels to increase and CYP2E1 levels to decrease. Rifampin 84-94 cytochrome P450 1A2 Callithrix jacchus 200-206 11257517-5 2001 Treatment with phenobarbitone caused CYP2B, CYP2C-2 and CYP3A21 levels to increase, rifampicin caused increases in CYP2B and CYP2C-1 and a decrease in CYP3A21 levels, whereas dioxin caused CYP1A1 and CYP1A2 levels to increase and CYP2E1 levels to decrease. Rifampin 84-94 cytochrome P450 2E1 Callithrix jacchus 230-236 11257517-7 2001 P450 enzyme activities were assessed using 8 different substrates and increases were found after treatment with phenobarbitone, rifampicin, and dioxin. Rifampin 128-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 11181490-4 2001 Compounds known to activate the pregnane X receptor (PXR) such as rifampicin, or the constitutively activated receptor (CAR) such as phenobarbital, induced CYP2C8, CYP2C9, and to a lesser extent CYP2C19 mRNAs and proteins. Rifampin 66-76 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 156-162 11181490-4 2001 Compounds known to activate the pregnane X receptor (PXR) such as rifampicin, or the constitutively activated receptor (CAR) such as phenobarbital, induced CYP2C8, CYP2C9, and to a lesser extent CYP2C19 mRNAs and proteins. Rifampin 66-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 164-170 11181490-4 2001 Compounds known to activate the pregnane X receptor (PXR) such as rifampicin, or the constitutively activated receptor (CAR) such as phenobarbital, induced CYP2C8, CYP2C9, and to a lesser extent CYP2C19 mRNAs and proteins. Rifampin 66-76 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 195-202 11181490-6 2001 The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. Rifampin 71-81 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 32-38 11181490-6 2001 The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. Rifampin 71-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 11181490-6 2001 The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. Rifampin 71-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 11181490-6 2001 The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. Rifampin 192-202 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 32-38 11181490-6 2001 The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. Rifampin 192-202 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 11181490-6 2001 The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. Rifampin 192-202 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 11181490-9 2001 Furthermore, dexamethasone, which has been recently shown to up-regulate PXR and CAR expression through the glucocorticoid receptor, potentiated CYP2C8 and CYP2C9 mRNA induction in response to rifampicin and phenobarbital. Rifampin 193-203 nuclear receptor subfamily 1 group I member 2 Homo sapiens 73-76 11181490-9 2001 Furthermore, dexamethasone, which has been recently shown to up-regulate PXR and CAR expression through the glucocorticoid receptor, potentiated CYP2C8 and CYP2C9 mRNA induction in response to rifampicin and phenobarbital. Rifampin 193-203 nuclear receptor subfamily 1 group I member 3 Homo sapiens 81-84 11181490-9 2001 Furthermore, dexamethasone, which has been recently shown to up-regulate PXR and CAR expression through the glucocorticoid receptor, potentiated CYP2C8 and CYP2C9 mRNA induction in response to rifampicin and phenobarbital. Rifampin 193-203 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 145-151 11181490-9 2001 Furthermore, dexamethasone, which has been recently shown to up-regulate PXR and CAR expression through the glucocorticoid receptor, potentiated CYP2C8 and CYP2C9 mRNA induction in response to rifampicin and phenobarbital. Rifampin 193-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 11266076-8 2001 CYP3A43 is also expressed in several other tissues including liver, where it can be induced by rifampicin. Rifampin 95-105 cytochrome P450 family 3 subfamily A member 43 Homo sapiens 0-7 11327199-6 2001 Concomitant use of CYP inducers, such as rifampicin (rifampin), rifabutin, phenytoin, phenobarbital or carbamazepine, should be avoided since delavirdine plasma concentrations are significantly lowered. Rifampin 41-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 11327199-6 2001 Concomitant use of CYP inducers, such as rifampicin (rifampin), rifabutin, phenytoin, phenobarbital or carbamazepine, should be avoided since delavirdine plasma concentrations are significantly lowered. Rifampin 53-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 11432536-4 2001 The relative potency of the rifamycins as CYP3A inducers is rifampin > rifapentine > rifabutin; rifabutin is also a CYP3A substrate. Rifampin 60-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-47 11432536-4 2001 The relative potency of the rifamycins as CYP3A inducers is rifampin > rifapentine > rifabutin; rifabutin is also a CYP3A substrate. Rifampin 60-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 122-127 11406737-12 2001 The mechanism underlying the interaction between rifampin and glyburide is probably induction of either CYP2C9 or P-glycoprotein or both. Rifampin 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 11406737-12 2001 The mechanism underlying the interaction between rifampin and glyburide is probably induction of either CYP2C9 or P-glycoprotein or both. Rifampin 49-57 ATP binding cassette subfamily B member 1 Homo sapiens 114-128 11735605-5 2001 On the other hand, rifampicin is a nonspecific inducer of CYP-mediated metabolism (especially of CYP2C9, CYP2C19 and CYP3A4) and may therefore cause therapeutic failure of other coadministered drugs by increasing their clearance. Rifampin 19-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 11735605-5 2001 On the other hand, rifampicin is a nonspecific inducer of CYP-mediated metabolism (especially of CYP2C9, CYP2C19 and CYP3A4) and may therefore cause therapeutic failure of other coadministered drugs by increasing their clearance. Rifampin 19-29 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 105-112 11735605-5 2001 On the other hand, rifampicin is a nonspecific inducer of CYP-mediated metabolism (especially of CYP2C9, CYP2C19 and CYP3A4) and may therefore cause therapeutic failure of other coadministered drugs by increasing their clearance. Rifampin 19-29 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 117-123 11460875-8 2001 However, such an interaction effect was masked following administration at 22.00 h. The time-dependent influence of rifampicin on the pharmacokinetics of zidovudine may be due to time-dependent changes in absorption and elimination of rifampicin, thus modifying its induction effect on the levels of UDP glucuronyl transferase and cytochrome P-450 content in liver which are responsible for metabolism of zidovudine. Rifampin 116-126 cytochrome P-450 Oryctolagus cuniculus 331-347 11181490-4 2001 Compounds known to activate the pregnane X receptor (PXR) such as rifampicin, or the constitutively activated receptor (CAR) such as phenobarbital, induced CYP2C8, CYP2C9, and to a lesser extent CYP2C19 mRNAs and proteins. Rifampin 66-76 nuclear receptor subfamily 1 group I member 2 Homo sapiens 53-56 11136298-7 2000 CONCLUSIONS: The effects of rifampicin on the pharmacokinetics of glimepiride suggest that rifampicin induced the CYP2C9-mediated metabolism of glimepiride and thereby slightly increased its systemic clearance. Rifampin 28-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 11136298-7 2000 CONCLUSIONS: The effects of rifampicin on the pharmacokinetics of glimepiride suggest that rifampicin induced the CYP2C9-mediated metabolism of glimepiride and thereby slightly increased its systemic clearance. Rifampin 91-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 11180018-1 2000 BACKGROUND: Rifampin (rifampicin) is a potent inducer of several cytochrome P450 (CYP) enzymes, including CYP3A4. Rifampin 12-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 11147822-12 2000 Simultaneous expression of the H and L ferritin genes in Escherichia coli grown in a deficient medium with minimal iron and with the addition of rifampicin resulted in the production of a range of recombinant human apoferritin heteromers that could be separated based on their subunit composition. Rifampin 145-155 ferritin heavy chain 1 Homo sapiens 215-226 11180018-1 2000 BACKGROUND: Rifampin (rifampicin) is a potent inducer of several cytochrome P450 (CYP) enzymes, including CYP3A4. Rifampin 12-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 11180018-1 2000 BACKGROUND: Rifampin (rifampicin) is a potent inducer of several cytochrome P450 (CYP) enzymes, including CYP3A4. Rifampin 12-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 106-112 11180018-1 2000 BACKGROUND: Rifampin (rifampicin) is a potent inducer of several cytochrome P450 (CYP) enzymes, including CYP3A4. Rifampin 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 11180018-1 2000 BACKGROUND: Rifampin (rifampicin) is a potent inducer of several cytochrome P450 (CYP) enzymes, including CYP3A4. Rifampin 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 11180018-1 2000 BACKGROUND: Rifampin (rifampicin) is a potent inducer of several cytochrome P450 (CYP) enzymes, including CYP3A4. Rifampin 22-32 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 106-112 11180018-11 2000 Because the elimination half-life of simvastatin was not affected by rifampin, induction of the CYP3A4-mediated first-pass metabolism of simvastatin in the intestine and the liver probably explains this interaction. Rifampin 69-77 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 96-102 11082432-8 2000 The induction of CYP3A by rifampicin enhanced the metabolism, and the induction of all-trans-RA metabolism was also observed after preincubation of the cells with all-trans-RA. Rifampin 26-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 17-22 11061580-0 2000 In vivo modulation of CYP enzymes by quinidine and rifampin. Rifampin 51-59 peptidylprolyl isomerase G Homo sapiens 22-25 11090943-8 2000 And finally, PXRs from different species are differentially activated by certain compounds such as rifampicin and pregnenolone 16alpha-carbonitrile (PCN) in a manner that correlates with species-specific induction of CYP3A gene expression. Rifampin 99-109 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 217-222 11073816-0 2000 The effect of rifampin treatment on intestinal expression of human MRP transporters. Rifampin 14-22 ATP binding cassette subfamily C member 1 Homo sapiens 67-70 11073816-2 2000 For example, induction of intestinal P-glycoprotein by rifampin appears to be the underlying mechanism of decreased plasma concentrations of P-glycoprotein substrates such as digoxin with concomitant rifampin therapy. Rifampin 55-63 ATP binding cassette subfamily B member 1 Homo sapiens 37-51 11073816-2 2000 For example, induction of intestinal P-glycoprotein by rifampin appears to be the underlying mechanism of decreased plasma concentrations of P-glycoprotein substrates such as digoxin with concomitant rifampin therapy. Rifampin 55-63 ATP binding cassette subfamily B member 1 Homo sapiens 141-155 11073816-4 2000 Therefore, we tested in this study the hypothesis whether the conjugate export pump MRP2 (cMOAT), which is another member of the ABC transporter family, is inducible by rifampin in humans. Rifampin 169-177 ATP binding cassette subfamily C member 2 Homo sapiens 84-88 11073816-4 2000 Therefore, we tested in this study the hypothesis whether the conjugate export pump MRP2 (cMOAT), which is another member of the ABC transporter family, is inducible by rifampin in humans. Rifampin 169-177 ATP binding cassette subfamily C member 2 Homo sapiens 90-95 11073816-7 2000 Rifampin induced duodenal MRP2 mRNA in 14 out of 16 individuals. Rifampin 0-8 ATP binding cassette subfamily C member 2 Homo sapiens 26-30 11073816-8 2000 Moreover, MRP2 protein, which was expressed in the apical membrane of enterocytes, was significantly induced by rifampin in 10 out of 16 subjects. Rifampin 112-120 ATP binding cassette subfamily C member 2 Homo sapiens 10-14 11073816-9 2000 In summary, rifampin induces MRP2 mRNA and protein in human duodenum. Rifampin 12-20 ATP binding cassette subfamily C member 2 Homo sapiens 29-33 11073816-10 2000 Increased elimination of MRP2 substrates (eg, drug conjugates) into the lumen of the gastrointestinal tract during treatment with rifampin could be a new mechanism of drug interactions. Rifampin 130-138 ATP binding cassette subfamily C member 2 Homo sapiens 25-29 11061574-2 2000 It has been demonstrated that rifampin (INN, rifampicin) induces P-glycoprotein, particularly in the gut wall. Rifampin 30-38 ATP binding cassette subfamily B member 1 Homo sapiens 65-79 11061574-2 2000 It has been demonstrated that rifampin (INN, rifampicin) induces P-glycoprotein, particularly in the gut wall. Rifampin 45-55 ATP binding cassette subfamily B member 1 Homo sapiens 65-79 11061574-3 2000 We therefore hypothesized that rifampin affects pharmacokinetics of the P-glycoprotein substrate talinolol, a beta1-blocker without appreciable metabolic disposition but intense intestinal secretion in human beings. Rifampin 31-39 ATP binding cassette subfamily B member 1 Homo sapiens 72-86 11061574-6 2000 Treatment with rifampin resulted in a significantly increased expression of duodenal P-glycoprotein content 4.2-fold (2.9, 6.51) (Western blot) and messenger RNA was increased in six of the eight volunteers. Rifampin 15-23 ATP binding cassette subfamily B member 1 Homo sapiens 85-99 11144420-5 2000 The overall frequency of drug resistance traits among the 1,749 MRSA strains was high (over 70% and up to and over 90% of the strains) to ciprofloxacin, erythromycin, clindamycin, gentamicin, and tetracycline, and was somewhat less frequent to sulfamethoxazole-trimethoprim (45%), chloramphenicol (30%), and rifampin (38%). Rifampin 308-316 solute carrier family 9 member A6 Homo sapiens 64-68 11129093-1 2000 It has recently been shown that P-glycoprotein (P-gp) is inducible by rifampicin in the human gut as shown in intestinal biopsies. Rifampin 70-80 ATP binding cassette subfamily B member 1 Homo sapiens 32-46 11129093-1 2000 It has recently been shown that P-glycoprotein (P-gp) is inducible by rifampicin in the human gut as shown in intestinal biopsies. Rifampin 70-80 ATP binding cassette subfamily B member 1 Homo sapiens 48-52 11129093-8 2000 Cytochrome P4503A4 (CYP3A4) inducibility was measured by comparing the urinary metabolic ratio of 6beta-hydroxycortisol/cortisol on day 14 and 19, Lymphocyte P-gp expression and activity was not induced by rifampicin, while it increased CYP3A4 activity from 5.0 +/- 4.0 to 22.9 +/- 16.6 (P < 0.001). Rifampin 206-216 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-18 11129093-8 2000 Cytochrome P4503A4 (CYP3A4) inducibility was measured by comparing the urinary metabolic ratio of 6beta-hydroxycortisol/cortisol on day 14 and 19, Lymphocyte P-gp expression and activity was not induced by rifampicin, while it increased CYP3A4 activity from 5.0 +/- 4.0 to 22.9 +/- 16.6 (P < 0.001). Rifampin 206-216 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 20-26 10975612-0 2000 Quantitative RT-PCR for CYP3A4 mRNA in human peripheral lymphocytes: induction of CYP3A4 in lymphocytes and in liver by rifampicin. Rifampin 120-130 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 82-88 10859152-9 2000 Under identical experimental conditions, rifampicin and lansoprazole significantly elevated CYP3A4 mRNA expression (P <.05), whereas 3-methylcholanthrene, omeprazole, and dimethyl sulfoxide were without significant effect. Rifampin 41-51 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 92-98 10924340-4 2000 In parallel, IL-6 decreases both rifampicin- and phenobarbital-mediated induction of CYP2B6, CYP2C8, CYP2C9, and CYP3A4. Rifampin 33-43 interleukin 6 Homo sapiens 13-17 10924340-4 2000 In parallel, IL-6 decreases both rifampicin- and phenobarbital-mediated induction of CYP2B6, CYP2C8, CYP2C9, and CYP3A4. Rifampin 33-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 10924340-4 2000 In parallel, IL-6 decreases both rifampicin- and phenobarbital-mediated induction of CYP2B6, CYP2C8, CYP2C9, and CYP3A4. Rifampin 33-43 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 93-99 10924340-4 2000 In parallel, IL-6 decreases both rifampicin- and phenobarbital-mediated induction of CYP2B6, CYP2C8, CYP2C9, and CYP3A4. Rifampin 33-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 10924340-4 2000 In parallel, IL-6 decreases both rifampicin- and phenobarbital-mediated induction of CYP2B6, CYP2C8, CYP2C9, and CYP3A4. Rifampin 33-43 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 113-119 10908304-3 2000 We report that dexamethasone increases both retinoid X receptor-alpha (RXRalpha) and PXR mRNA expression in cultured human hepatocytes, whereas PXR activators such as rifampicin and clotrimazole do not. Rifampin 167-177 nuclear receptor subfamily 1 group I member 2 Homo sapiens 144-147 10869292-0 2000 Rifamycin SV and rifampicin exhibit differential inhibition of the hepatic rat organic anion transporting polypeptides, Oatp1 and Oatp2. Rifampin 17-27 solute carrier organic anion transporter family, member 1a1 Rattus norvegicus 120-125 10869292-0 2000 Rifamycin SV and rifampicin exhibit differential inhibition of the hepatic rat organic anion transporting polypeptides, Oatp1 and Oatp2. Rifampin 17-27 solute carrier organic anion transporter family, member 1a4 Rattus norvegicus 130-135 10869292-6 2000 Rifampicin potently inhibited Oatp2-mediated taurocholate uptake, but did not interfere with Oatp1-mediated taurocholate uptake. Rifampin 0-10 solute carrier organic anion transporter family, member 1a4 Rattus norvegicus 30-35 10869292-7 2000 Similar effects of rifamycin SV and rifampicin were found for Oatp1- and Oatp2-mediated estradiol-17beta-glucuronide transport. Rifampin 36-46 solute carrier organic anion transporter family, member 1a1 Rattus norvegicus 62-67 10869292-7 2000 Similar effects of rifamycin SV and rifampicin were found for Oatp1- and Oatp2-mediated estradiol-17beta-glucuronide transport. Rifampin 36-46 solute carrier organic anion transporter family, member 1a4 Rattus norvegicus 73-78 11061580-8 2000 The quinidine/rifampin combination resulted in selective CYP2D6 inhibition and induction of all other enzymes evaluated over this time period, suggesting that predictable complex interactions occur with the drug combination. Rifampin 14-22 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 57-63 10873607-4 2000 Model inducers like rifampicin, phenobarbital, or 3-methylcholanthrene and beta-naphtoflavone were able to induce CYP1A or CYP3A4 as well as EROD or T6H activities for up to 30 days. Rifampin 20-30 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 123-129 10869292-9 2000 These results demonstrate that rifamycin SV and rifampicin exhibit differential inhibition on Oatp1 and Oatp2, and identify rifampicin as a selective Oatp2 inhibitor. Rifampin 48-58 solute carrier organic anion transporter family, member 1a1 Rattus norvegicus 94-99 10869292-9 2000 These results demonstrate that rifamycin SV and rifampicin exhibit differential inhibition on Oatp1 and Oatp2, and identify rifampicin as a selective Oatp2 inhibitor. Rifampin 48-58 solute carrier organic anion transporter family, member 1a4 Rattus norvegicus 104-109 10869292-9 2000 These results demonstrate that rifamycin SV and rifampicin exhibit differential inhibition on Oatp1 and Oatp2, and identify rifampicin as a selective Oatp2 inhibitor. Rifampin 124-134 solute carrier organic anion transporter family, member 1a4 Rattus norvegicus 150-155 10903421-4 2000 The goal of these investigations was to evaluate the effect of altering culture conditions on rifampin-mediated induction of CYP3A isoforms in cultured porcine hepatocytes. Rifampin 94-102 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 125-130 10824630-3 2000 METHODS: Six extensive metabolizers of CYP2D6 (age, 70.5 +/- 3.5 years) ingested 600 mg rifampin once daily for 9 consecutive days. Rifampin 88-96 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 39-45 10620362-12 2000 Finally, in both chicken embryo liver and chicken hepatoma cells (LMH), CYP3A37 mRNA was increased after treatment with typical CYP3A inducers, such as metyrapone, phenobarbital, dexamethasone, and pregnenolone 16alpha-carbonitrile, but not rifampicin. Rifampin 241-251 cytochrome P450 family 3 subfamily A member 5 Gallus gallus 72-79 10772626-6 2000 Although the responses to dexamethasone and rifampicin were both increased by both receptors, dexamethasone activation of CYP3A4 was similar for both the hGR and the hPXR, whereas rifampicin-dependent activation favored the hPXR. Rifampin 180-190 nuclear receptor subfamily 1 group I member 2 Homo sapiens 224-228 10727519-2 2000 Recently, rifampicin has been reported to activate the glucocorticoid receptor (GR) in human hepatocytes. Rifampin 10-20 nuclear receptor subfamily 3 group C member 1 Homo sapiens 55-78 10727519-2 2000 Recently, rifampicin has been reported to activate the glucocorticoid receptor (GR) in human hepatocytes. Rifampin 10-20 nuclear receptor subfamily 3 group C member 1 Homo sapiens 80-82 10727519-3 2000 Because there is evidence that increased levels of glucocorticoids may induce cognitive impairment, sometimes culminating in depression, the side effects of rifampicin may result from GR activation in central nerve cells. Rifampin 157-167 nuclear receptor subfamily 3 group C member 1 Homo sapiens 184-186 10727519-6 2000 Rifampicin was also inactive in the same HepG2 cell line that was originally used to demonstrate the effect of rifampicin on GR. Rifampin 111-121 nuclear receptor subfamily 3 group C member 1 Homo sapiens 125-127 10706865-4 2000 Finally, in the MAIPA assay, the rifampicin-dependent antibody almost completely cross-blocked the binding of the anti-GPIX mAb (SZ1) to platelets. Rifampin 33-43 glycoprotein IX platelet Homo sapiens 119-123 10706865-6 2000 This finding not only confirms that the epitope of the rifampicin-dependent antibody is on GPIX but it is also identical to or located in close proximity to that of the quinine-dependent antibody and SZ1. Rifampin 55-65 glycoprotein IX platelet Homo sapiens 91-95 10751073-0 2000 Slow N-acetyltransferase 2 genotype affects the incidence of isoniazid and rifampicin-induced hepatotoxicity. Rifampin 75-85 N-acetyltransferase 2 Homo sapiens 5-26 10751073-2 2000 OBJECTIVE: To elucidate the relationship between N-acetyltransferase 2 (NAT2) genotype and the incidence of isoniazid + rifampicin-induced hepatotoxicity. Rifampin 120-130 N-acetyltransferase 2 Homo sapiens 49-70 10751073-2 2000 OBJECTIVE: To elucidate the relationship between N-acetyltransferase 2 (NAT2) genotype and the incidence of isoniazid + rifampicin-induced hepatotoxicity. Rifampin 120-130 N-acetyltransferase 2 Homo sapiens 72-76 10648470-6 2000 Cyclosporin A, rifamycin SV, rifampicin, and glibenclamide cis-inhibited Bsep-mediated bile salt transport to similar extents as ATP-dependent taurocholate transport in cLPM vesicles. Rifampin 29-39 ATP binding cassette subfamily B member 11 Homo sapiens 73-77 10673364-4 2000 Using an RT-competitive PCR (RT-cPCR) with beta-actin as the standard in this study, the constitutive and rifampicin (RFP)-induced expression of CYP3A4, CYP2C9, CYP2E1, and CYP1A2 mRNA in the HepG2 cells could be quantitatively and reproducibly determined. Rifampin 106-116 POTE ankyrin domain family member F Homo sapiens 43-53 10673364-4 2000 Using an RT-competitive PCR (RT-cPCR) with beta-actin as the standard in this study, the constitutive and rifampicin (RFP)-induced expression of CYP3A4, CYP2C9, CYP2E1, and CYP1A2 mRNA in the HepG2 cells could be quantitatively and reproducibly determined. Rifampin 106-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 145-151 10673364-4 2000 Using an RT-competitive PCR (RT-cPCR) with beta-actin as the standard in this study, the constitutive and rifampicin (RFP)-induced expression of CYP3A4, CYP2C9, CYP2E1, and CYP1A2 mRNA in the HepG2 cells could be quantitatively and reproducibly determined. Rifampin 106-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 10673364-4 2000 Using an RT-competitive PCR (RT-cPCR) with beta-actin as the standard in this study, the constitutive and rifampicin (RFP)-induced expression of CYP3A4, CYP2C9, CYP2E1, and CYP1A2 mRNA in the HepG2 cells could be quantitatively and reproducibly determined. Rifampin 106-116 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 161-167 10673364-4 2000 Using an RT-competitive PCR (RT-cPCR) with beta-actin as the standard in this study, the constitutive and rifampicin (RFP)-induced expression of CYP3A4, CYP2C9, CYP2E1, and CYP1A2 mRNA in the HepG2 cells could be quantitatively and reproducibly determined. Rifampin 106-116 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 173-179 10772631-0 2000 Rabbit pregnane X receptor is activated by rifampicin. Rifampin 43-53 nuclear receptor subfamily 1, group I, member 2 Mus musculus 7-26 10772631-7 2000 Rifampicin treatment of CV-1 cells cotransfected with a rabbit PXR expression plasmid and a luciferase reporter construct containing two copies of the DR3 enhancer from CYP3A23 produced a 6-fold induction of luciferase activity. Rifampin 0-10 nuclear receptor subfamily 1 group I member 2 Oryctolagus cuniculus 63-66 10803787-0 2000 Rifampicin suppresses hepatic CYP2E1 expression and minimizes DNA injury caused by carbon tetrachloride in perivenular hepatocytes of mice. Rifampin 0-10 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 30-36 10803787-1 2000 BACKGROUND: Rifampicin has been shown to increase during activities of serum transaminases and to decrease in cytochrome P-450-mediated monooxygenase activities in livers of mice treated with carbon tetrachloride (CCl4). Rifampin 12-22 chemokine (C-C motif) ligand 4 Mus musculus 214-218 10803787-2 2000 Although these findings suggest that rifampicin prevents hepatocyte damage caused by CCl4, detailed information on the protective effects is not available. Rifampin 37-47 chemokine (C-C motif) ligand 4 Mus musculus 85-89 10803787-6 2000 RESULTS: Rifampicin prevented the denaturation and fragmentation of DNA caused by CCl4 in perivenular hepatocytes except for those located within two or three cell layers surrounding the central venule. Rifampin 9-19 chemokine (C-C motif) ligand 4 Mus musculus 82-86 10803787-7 2000 Furthermore, CYP2E1 decreased in liver homogenates or microsomes from rifampicin-treated animals. Rifampin 70-80 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 13-19 10803787-8 2000 It is therefore likely that rifampicin suppresses expression of CYP2E1 and protects CCl4-mediated DNA damage of hepatocytes by inhibiting formation of free radicals. Rifampin 28-38 cytochrome P450, family 2, subfamily e, polypeptide 1 Mus musculus 64-70 10803787-8 2000 It is therefore likely that rifampicin suppresses expression of CYP2E1 and protects CCl4-mediated DNA damage of hepatocytes by inhibiting formation of free radicals. Rifampin 28-38 chemokine (C-C motif) ligand 4 Mus musculus 84-88 10803787-9 2000 In addition, perivenular hepatocytes except for those surrounding the venule showed negative immunoreaction for p53 and bcl2 in rifampicin+CCl4-treated animals. Rifampin 128-138 B cell leukemia/lymphoma 2 Mus musculus 120-124 10729196-12 2000 Upon incubation with phenobarbital and rifampin (rifampicin), human hepatocytes increased CYP 2B6, 3A4, and 3A5 among others. Rifampin 39-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-97 10729196-12 2000 Upon incubation with phenobarbital and rifampin (rifampicin), human hepatocytes increased CYP 2B6, 3A4, and 3A5 among others. Rifampin 49-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-97 10681370-1 2000 Cytochrome P-4503A, CYP2B, and P-450 reductase are induced by glucocorticoids, antiglucocorticoids such as pregnenolone 16alpha-carbonitrile, and drugs such as rifampin and phenobarbital. Rifampin 160-168 cytochrome P450, family 2, subfamily b, polypeptide 10 Mus musculus 20-25 10681370-8 2000 Similarly, rifampin induced CYP3A in either wild-type or glucocorticoid receptor-null mice. Rifampin 11-19 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 28-33 10681370-8 2000 Similarly, rifampin induced CYP3A in either wild-type or glucocorticoid receptor-null mice. Rifampin 11-19 nuclear receptor subfamily 3, group C, member 1 Mus musculus 57-80 10681370-9 2000 Despite reports that rifampin is a nonsteroidal ligand for the human glucocorticoid receptor, rifampin failed to induce tyrosine aminotransferase in mice regardless of glucocorticoid receptor genotype, and rifampin did not compete for ligand binding to either mouse or human glucocorticoid receptor. Rifampin 21-29 nuclear receptor subfamily 3 group C member 1 Homo sapiens 69-92 10752642-8 2000 The capacity of troglitazone to induce CYP3A4 was between that of rifampin (EC50 = 0.8 microM) and dexamethasone (40-50 microM). Rifampin 66-74 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 39-45 10706193-9 2000 Moreover, induction of intestinal P-glycoprotein by rifampin has now been identified as the major underlying mechanism of reduced digoxin plasma concentrations during concomitant rifampin therapy. Rifampin 52-60 ATP binding cassette subfamily B member 1 Homo sapiens 34-48 10620362-12 2000 Finally, in both chicken embryo liver and chicken hepatoma cells (LMH), CYP3A37 mRNA was increased after treatment with typical CYP3A inducers, such as metyrapone, phenobarbital, dexamethasone, and pregnenolone 16alpha-carbonitrile, but not rifampicin. Rifampin 241-251 cytochrome P450 family 3 subfamily A member 5 Gallus gallus 72-77 10611146-5 2000 The pregnane X receptor (PXR), which has recently been shown to mediate the transcriptional induction of CYP3A4 expression in response to rifampicin, was activated by metyrapone in CV-1 cells transiently cotransfected with an expression vector encoding the human PXR and a reporter construct containing the everted repeat sequence that confers CYP3A4 induction responsiveness to inducers within its promoter. Rifampin 138-148 nuclear receptor subfamily 1 group I member 2 Homo sapiens 25-28 10611140-5 2000 The amplitude of the increase in CYP3A4 protein and activity by 25 microM eletriptan was significantly lower, with a mean of 19 (P =.0015) and 26% (P =.0002), respectively, of that observed in response to 25 microM rifampicin. Rifampin 215-225 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 33-39 10611140-6 2000 CYP2A6, a protein with minor pharmacological implication, also was induced by eletriptan and rifampicin in two cultures but was not detected in the others. Rifampin 93-103 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 0-6 10611146-5 2000 The pregnane X receptor (PXR), which has recently been shown to mediate the transcriptional induction of CYP3A4 expression in response to rifampicin, was activated by metyrapone in CV-1 cells transiently cotransfected with an expression vector encoding the human PXR and a reporter construct containing the everted repeat sequence that confers CYP3A4 induction responsiveness to inducers within its promoter. Rifampin 138-148 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-111 10654647-3 2000 Mutations in the rpoB gene identified by LiPA that confer resistance to rifampicin (RMP) were confirmed by DNA sequencing, while susceptibilities were confirmed by the proportion method and BACTEC. Rifampin 72-82 lipase A, lysosomal acid type Homo sapiens 41-45 10654647-3 2000 Mutations in the rpoB gene identified by LiPA that confer resistance to rifampicin (RMP) were confirmed by DNA sequencing, while susceptibilities were confirmed by the proportion method and BACTEC. Rifampin 84-87 lipase A, lysosomal acid type Homo sapiens 41-45 10570062-0 1999 The orphan human pregnane X receptor mediates the transcriptional activation of CYP3A4 by rifampicin through a distal enhancer module. Rifampin 90-100 nuclear receptor subfamily 1 group I member 2 Homo sapiens 17-36 10628745-8 2000 For example, the macrolide antibiotic rifampicin, the antidiabetic drug troglitazone, and the hypocholesterolemic drug SR12813 are efficacious activators of the human and rabbit PXR but have little activity on the rat and mouse PXR. Rifampin 38-48 nuclear receptor subfamily 1 group I member 2 Oryctolagus cuniculus 178-181 10628745-8 2000 For example, the macrolide antibiotic rifampicin, the antidiabetic drug troglitazone, and the hypocholesterolemic drug SR12813 are efficacious activators of the human and rabbit PXR but have little activity on the rat and mouse PXR. Rifampin 38-48 nuclear receptor subfamily 1, group I, member 2 Mus musculus 228-231 10570062-0 1999 The orphan human pregnane X receptor mediates the transcriptional activation of CYP3A4 by rifampicin through a distal enhancer module. Rifampin 90-100 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 80-86 10570062-1 1999 Cytochrome P-450 3A4 (CYP3A4), the predominant cytochrome P-450 expressed in adult human liver, is subject to transcriptional induction by a variety of structurally unrelated xenobiotics, including the antibiotic rifampicin. Rifampin 213-223 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-20 10570062-1 1999 Cytochrome P-450 3A4 (CYP3A4), the predominant cytochrome P-450 expressed in adult human liver, is subject to transcriptional induction by a variety of structurally unrelated xenobiotics, including the antibiotic rifampicin. Rifampin 213-223 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 22-28 10570062-4 1999 Rifampicin-inducible transcription of the reporter gene was observed only with the longest construct, which encompassed bases -13000 to +53 of CYP3A4 (3-fold induction). Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 143-149 10570062-5 1999 The responsive region was functional regardless of its position or orientation relative to the proximal promoter of CYP3A4 and was capable of conferring rifampicin-inducible expression on a heterologous promoter. Rifampin 153-163 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-122 10570062-9 1999 Cotransfection of responsive constructs with a hPXR expression vector substantially increased the rifampicin-inducibility to approximately 50-fold. Rifampin 98-108 nuclear receptor subfamily 1 group I member 2 Homo sapiens 47-51 10628900-9 1999 The results indicate that the inductive effect of rifampicin is likely to be of clinical relevance particularly when used concomitantly with drugs metabolized by CYP3A4. Rifampin 50-60 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 162-168 10634135-4 1999 Four received ALP after a 2-day pretreatment with ketoconazole, an inhibitor of CYP3A4, and four normal volunteers received ALP after 4 days of rifampin, an inducer of CYP3A4. Rifampin 144-152 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 168-174 10563691-1 1999 Rifampicin has been successfully used as an adjunct to vancomycin therapy in several clinical conditions of MRSA infections such as endocarditis, ventriculoperitoneal shunts and septicaemia. Rifampin 0-10 solute carrier family 9 member A6 Homo sapiens 108-112 10545214-7 1999 The expression of CYP2A15 mRNA was slightly induced by treatment with either rifampicin or 3-methylcholanthrene. Rifampin 77-87 cytochrome P450 2A15 Cricetulus griseus 18-25 10563691-3 1999 The present prospective study was conducted to evaluate the efficacy of rifampicin as an adjunct therapy in burn cases with MRSA septicaemia not responding well to vancomycin. Rifampin 72-82 solute carrier family 9 member A6 Homo sapiens 124-128 10563691-11 1999 Institution of rifampicin, as an adjunct to vancomycin therapy to which the MRSA isolates were susceptible, showed a dramatic clinical response and survival of grafts. Rifampin 15-25 solute carrier family 9 member A6 Homo sapiens 76-80 10563691-13 1999 The present study thus confirms the efficacy of clinical use of rifampicin as an adjunct in vancomycin nonresponding cases of MRSA septicaemia in burns. Rifampin 64-74 solute carrier family 9 member A6 Homo sapiens 126-130 10486655-9 1999 Following explanation can be hypothesis: several antihypertensive drugs are liver-metabolised by microsomal cytochrome P450 3A4 isoform that could explain a significantly decreased half-life in association with enzymatic inducers, such as rifampicine or antiepileptic drugs (phenobarbital, phenytoin or carbamazepine). Rifampin 239-250 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 108-127 10506631-2 1999 RIF treatment with combined pentoxifylline (PTX) and tocopherol (Vit E) was prompted by recent advances in cellular and molecular biology that have improved researchers" understanding of radiation-induced late-injury mechanisms and by the excellent results from our previous human and animal studies. Rifampin 0-3 vitrin Homo sapiens 65-68 10591535-16 1999 Thus, induction of both phase I pathways (CYP3A4/1A2) and phase II pathways (glucuronidation, sulfation) of propafenone by rifampicin resulted in a clinically relevant metabolic drug interaction which was more pronounced in extensive metabolizers than in poor metabolizers with regard to percentage decrease in bioavailability of propafenone. Rifampin 123-133 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-52 10462436-3 1999 Roles for three "orphan" nuclear receptor superfamily members, designated CAR, PXR, and PPAR, in respectively mediating the induction of hepatic P450s belonging to families CYP2, CYP3, and CYP4 in response to the prototypical inducers phenobarbital (CAR), pregnenolone 16alpha-carbonitrile and rifampicin (PXR), and clofibric acid (PPAR) have now been established. Rifampin 294-304 CXADR pseudogene 1 Homo sapiens 74-77 10462436-3 1999 Roles for three "orphan" nuclear receptor superfamily members, designated CAR, PXR, and PPAR, in respectively mediating the induction of hepatic P450s belonging to families CYP2, CYP3, and CYP4 in response to the prototypical inducers phenobarbital (CAR), pregnenolone 16alpha-carbonitrile and rifampicin (PXR), and clofibric acid (PPAR) have now been established. Rifampin 294-304 nuclear receptor subfamily 1 group I member 2 Homo sapiens 79-82 10462436-3 1999 Roles for three "orphan" nuclear receptor superfamily members, designated CAR, PXR, and PPAR, in respectively mediating the induction of hepatic P450s belonging to families CYP2, CYP3, and CYP4 in response to the prototypical inducers phenobarbital (CAR), pregnenolone 16alpha-carbonitrile and rifampicin (PXR), and clofibric acid (PPAR) have now been established. Rifampin 294-304 peroxisome proliferator activated receptor alpha Homo sapiens 88-92 10462436-3 1999 Roles for three "orphan" nuclear receptor superfamily members, designated CAR, PXR, and PPAR, in respectively mediating the induction of hepatic P450s belonging to families CYP2, CYP3, and CYP4 in response to the prototypical inducers phenobarbital (CAR), pregnenolone 16alpha-carbonitrile and rifampicin (PXR), and clofibric acid (PPAR) have now been established. Rifampin 294-304 peptidylprolyl isomerase F Homo sapiens 179-183 10462436-3 1999 Roles for three "orphan" nuclear receptor superfamily members, designated CAR, PXR, and PPAR, in respectively mediating the induction of hepatic P450s belonging to families CYP2, CYP3, and CYP4 in response to the prototypical inducers phenobarbital (CAR), pregnenolone 16alpha-carbonitrile and rifampicin (PXR), and clofibric acid (PPAR) have now been established. Rifampin 294-304 CXADR pseudogene 1 Homo sapiens 250-253 10462436-3 1999 Roles for three "orphan" nuclear receptor superfamily members, designated CAR, PXR, and PPAR, in respectively mediating the induction of hepatic P450s belonging to families CYP2, CYP3, and CYP4 in response to the prototypical inducers phenobarbital (CAR), pregnenolone 16alpha-carbonitrile and rifampicin (PXR), and clofibric acid (PPAR) have now been established. Rifampin 294-304 nuclear receptor subfamily 1 group I member 2 Homo sapiens 306-309 10462436-3 1999 Roles for three "orphan" nuclear receptor superfamily members, designated CAR, PXR, and PPAR, in respectively mediating the induction of hepatic P450s belonging to families CYP2, CYP3, and CYP4 in response to the prototypical inducers phenobarbital (CAR), pregnenolone 16alpha-carbonitrile and rifampicin (PXR), and clofibric acid (PPAR) have now been established. Rifampin 294-304 peroxisome proliferator activated receptor alpha Homo sapiens 332-336 10470557-2 1999 Penicillins, certain cephalosporins, carbapenems, fluoroquinolones, vancomycin, and rifampin provide the highest ratios of CSF levels to the MBC for common infecting organisms. Rifampin 84-92 colony stimulating factor 2 Homo sapiens 123-126 10421615-5 1999 The maximal CYP3A activity detected after treatment with Taxol or rifampicin was similar in six separate human hepatocyte cultures, suggesting that the cultures have achieved a limit of maximally inducible CYP3A. Rifampin 66-76 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 12-17 10704137-2 1999 The resolution of ternary mixtures of rifampicin, isoniazid and pyrazinamide has been accomplished by using partial least squares (PLS-1) regression analysis. Rifampin 38-48 plastin 1 Homo sapiens 131-136 10403778-5 1999 Using electrophoretic mobility shift assays and cotransfection experiments we show that this element is able to bind the PXR:RXR complex and transactivates the expression of a down stream promoter in response to rifampicin, clotrimazole, and RU-486, three compounds known to specifically activate the human PXR. Rifampin 212-222 nuclear receptor subfamily 1 group I member 2 Homo sapiens 121-124 10403778-5 1999 Using electrophoretic mobility shift assays and cotransfection experiments we show that this element is able to bind the PXR:RXR complex and transactivates the expression of a down stream promoter in response to rifampicin, clotrimazole, and RU-486, three compounds known to specifically activate the human PXR. Rifampin 212-222 retinoid X receptor alpha Homo sapiens 125-128 10403778-5 1999 Using electrophoretic mobility shift assays and cotransfection experiments we show that this element is able to bind the PXR:RXR complex and transactivates the expression of a down stream promoter in response to rifampicin, clotrimazole, and RU-486, three compounds known to specifically activate the human PXR. Rifampin 212-222 nuclear receptor subfamily 1 group I member 2 Homo sapiens 307-310 10493554-3 1999 We describe a case of rifampicin-associated acute renal failure, with biopsy findings of tubulointerstitial nephritis; inflammatory cells were characterized by immunohistochemistry, which showed immunoreactivity for CD3 and CD5 (T lymphocytes) and for CD68 (macrophages). Rifampin 22-32 CD5 molecule Homo sapiens 224-227 10493554-3 1999 We describe a case of rifampicin-associated acute renal failure, with biopsy findings of tubulointerstitial nephritis; inflammatory cells were characterized by immunohistochemistry, which showed immunoreactivity for CD3 and CD5 (T lymphocytes) and for CD68 (macrophages). Rifampin 22-32 CD68 molecule Homo sapiens 252-256 10411543-0 1999 The role of intestinal P-glycoprotein in the interaction of digoxin and rifampin. Rifampin 72-80 ATP binding cassette subfamily B member 1 Homo sapiens 23-37 10411543-2 1999 On the basis of clinical observations of patients in whom digoxin levels decreased considerably when treated with rifampin, we hypothesized that concomitant rifampin therapy may affect digoxin disposition in humans by induction of P-gp. Rifampin 114-122 ATP binding cassette subfamily B member 1 Homo sapiens 231-235 10411543-2 1999 On the basis of clinical observations of patients in whom digoxin levels decreased considerably when treated with rifampin, we hypothesized that concomitant rifampin therapy may affect digoxin disposition in humans by induction of P-gp. Rifampin 157-165 ATP binding cassette subfamily B member 1 Homo sapiens 231-235 10381771-5 1999 Oatp1-mediated gadoxetate uptake (100 microM) could be inhibited by 10 microM bromosulfophthalein (45%), 200 microM taurocholate (92%), 100 microM rifamycin SV (97%), and 100 microM rifampicin (51%). Rifampin 182-192 solute carrier organic anion transporter family, member 1a1 Rattus norvegicus 0-5 10348770-2 1999 Here it is shown that the Escherichia coli msbB mutant, which elaborates defective, penta-acylated lipid A, is practically as resistant to a representative set of hydrophobic solutes (rifampin, fusidic acid, erythromycin, clindamycin, and azithromycin) as the parent-type control strain. Rifampin 184-192 lipid A biosynthesis (KDO)2-(lauroyl)-lipid IVA acyltransferase Escherichia coli 43-47 10463384-3 1999 We examined the 6beta-OHF/F ratio in urine of common marmosets administered with rifampicin, a potent inducer of CYP3A, to evaluate the usefulness of common marmosets for the prediction of CYP3A induction. Rifampin 81-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 113-118 10354960-9 1999 The identification of CYP3A4 as a low-affinity nortriptyline E-10-hydroxylase explains the ability of poor metabolizers of debrisoquin to hydroxylate nortriptyline, as well as the increased in vivo clearance via this pathway caused by CYP3A4-inducing drugs such as pentobarbital, carbamazepine, and rifampin. Rifampin 299-307 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 22-28 10354960-9 1999 The identification of CYP3A4 as a low-affinity nortriptyline E-10-hydroxylase explains the ability of poor metabolizers of debrisoquin to hydroxylate nortriptyline, as well as the increased in vivo clearance via this pathway caused by CYP3A4-inducing drugs such as pentobarbital, carbamazepine, and rifampin. Rifampin 299-307 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 235-241 10220562-1 1999 It has recently been reported that rifampicin activates the glucocorticoid receptor and acts as an immunosuppressive drug. Rifampin 35-45 nuclear receptor subfamily 3 group C member 1 Homo sapiens 60-83 10591535-0 1999 Consequences of rifampicin treatment on propafenone disposition in extensive and poor metabolizers of CYP2D6. Rifampin 16-26 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 102-108 10591535-3 1999 However, non-CYP2D6-dependent pathways may be induced as a case report described dramatically lowered plasma concentrations of propafenone with loss of dysrhythmia control associated with rifampicin treatment. Rifampin 188-198 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 13-19 10591535-4 1999 Therefore, this study aimed to investigate induction properties of rifampicin on propafenone disposition in extensive metabolizers and poor metabolizers of CYP2D6. Rifampin 67-77 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 156-162 10408915-0 1999 Inhibition of multidrug resistance-associated protein (MRP) activity by rifampicin in human multidrug-resistant lung tumor cells. Rifampin 72-82 ATP binding cassette subfamily C member 3 Homo sapiens 14-53 10408915-0 1999 Inhibition of multidrug resistance-associated protein (MRP) activity by rifampicin in human multidrug-resistant lung tumor cells. Rifampin 72-82 ATP binding cassette subfamily C member 3 Homo sapiens 55-58 10408915-2 1999 In order to look for compounds that can lead to reversal of such a resistance, the antituberculosis compound rifampicin, belonging to the chemical class of rifamycins, was examined for its effect on MRP activity in human multidrug resistant lung cancer GLC4/ADR cells. Rifampin 109-119 ATP binding cassette subfamily C member 3 Homo sapiens 199-202 10408915-3 1999 Rifampicin was shown to increase accumulation of the MRP substrate calcein in GLC4/ADR cells in a dose-dependent manner by inhibiting its MRP-mediated efflux from the cells; it also enhanced intracellular retention of another substrate of MRP such as the anticancer drug vincristine in the resistant cells. Rifampin 0-10 ATP binding cassette subfamily C member 3 Homo sapiens 53-56 10408915-3 1999 Rifampicin was shown to increase accumulation of the MRP substrate calcein in GLC4/ADR cells in a dose-dependent manner by inhibiting its MRP-mediated efflux from the cells; it also enhanced intracellular retention of another substrate of MRP such as the anticancer drug vincristine in the resistant cells. Rifampin 0-10 ATP binding cassette subfamily C member 3 Homo sapiens 138-141 10408915-3 1999 Rifampicin was shown to increase accumulation of the MRP substrate calcein in GLC4/ADR cells in a dose-dependent manner by inhibiting its MRP-mediated efflux from the cells; it also enhanced intracellular retention of another substrate of MRP such as the anticancer drug vincristine in the resistant cells. Rifampin 0-10 ATP binding cassette subfamily C member 3 Homo sapiens 138-141 10408915-6 1999 These results therefore indicate that rifamycins, including rifampicin, probably constitute a new chemical class of modulators down-regulating MRP-mediated drug transport. Rifampin 60-70 ATP binding cassette subfamily C member 3 Homo sapiens 143-146 10220562-3 1999 We have used reporter gene assays to measure the trans-activating and trans-repressing capacity of the glucocorticoid receptor after treating A549 human alveolar cells with rifampicin. Rifampin 173-183 nuclear receptor subfamily 3 group C member 1 Homo sapiens 103-126 10379985-8 1999 Rifampicin induced (2-3-fold) cortisol 6beta-hydroxylase, S-mephenytoin 4"-hydroxylase, S-mephenytoin N-demethylase and tolbutamide hydroxylase activities, the O-dealkylations of methoxy-, ethoxy- and propoxyresorufin and CYP2C- and CYP3A-immunorelated proteins in monkey. Rifampin 0-10 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 58-86 10943389-7 1999 In this temperature-sensitive strain, the recovery of active aldehyde dehydrogenase, in the presence of rifampicin but not of chloramphenicol, when cells grown at 37 degrees C were shifted to 30 degrees C indicated that this mutation affected the folding process of the protein at the restrictive temperature. Rifampin 104-114 Aldehyde dehydrogenase Escherichia coli 61-83 10223773-2 1999 Rifampin (INN, rifampicin) is a potent inducer of CYP3A4 and some other CYP enzymes. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-56 10223773-2 1999 Rifampin (INN, rifampicin) is a potent inducer of CYP3A4 and some other CYP enzymes. Rifampin 15-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-56 10223773-14 1999 Concomitant use of rifampin or other potent inducers of CYP3A4 with ondansetron may result in a reduced antiemetic effect, particularly after oral administration of ondansetron. Rifampin 19-27 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 56-62 10037683-5 1999 In addition to PBREM, CAR also transactivates the steroid/rifampicin-response element of the human CYP3A4 gene in HepG2 cells. Rifampin 58-68 CXADR pseudogene 1 Homo sapiens 22-25 10037683-5 1999 In addition to PBREM, CAR also transactivates the steroid/rifampicin-response element of the human CYP3A4 gene in HepG2 cells. Rifampin 58-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-105 10339711-3 1999 He became asymptomatic on treatment with tetracycline, rifampicin and streptomycin with significant CSF response. Rifampin 55-65 colony stimulating factor 2 Homo sapiens 100-103 9987705-3 1999 A pharmacokinetic interaction with rifampin, an antituberculosis agent and potent inducer of CYP3A4 and P-glycoprotein, and tacrolimus was evaluated in six healthy male volunteers. Rifampin 35-43 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-99 10219967-9 1999 Induction of CYP3A4-dependent reporter gene expression and enhancement of the induction by the glucocorticoid receptor was also observed with pregnenolone-16alpha-carbonitrile (PCN), rifampicin, phenytoin, carbamazepine, phenylbutazone and phenobarbitone, all known in vivo inducers of CYP3A4 in man. Rifampin 183-193 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 13-19 10219967-9 1999 Induction of CYP3A4-dependent reporter gene expression and enhancement of the induction by the glucocorticoid receptor was also observed with pregnenolone-16alpha-carbonitrile (PCN), rifampicin, phenytoin, carbamazepine, phenylbutazone and phenobarbitone, all known in vivo inducers of CYP3A4 in man. Rifampin 183-193 nuclear receptor subfamily 3 group C member 1 Homo sapiens 95-118 9987705-3 1999 A pharmacokinetic interaction with rifampin, an antituberculosis agent and potent inducer of CYP3A4 and P-glycoprotein, and tacrolimus was evaluated in six healthy male volunteers. Rifampin 35-43 ATP binding cassette subfamily B member 1 Homo sapiens 104-118 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. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 114-119 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. Rifampin 0-8 ATP binding cassette subfamily B member 1 Homo sapiens 124-138 10069527-10 1999 Neither in PMNs nor in lymphocytes, induction of CYP 3A protein expression was observed after rifampicin treatment despite overall induction of CYP 3A activity assessed by the urinary excretion of 6beta-hydroxycortisol. Rifampin 94-104 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 49-55 10803271-3 1999 The strongest immunosuppression effect was demonstrated using rifampicin (39 ng/ml IFN-gamma) (Control: 123 +/- 29 ng/ml IFN-gamma, p < 0.05). Rifampin 62-72 interferon gamma Mus musculus 83-92 10803271-3 1999 The strongest immunosuppression effect was demonstrated using rifampicin (39 ng/ml IFN-gamma) (Control: 123 +/- 29 ng/ml IFN-gamma, p < 0.05). Rifampin 62-72 interferon gamma Mus musculus 121-130 10803271-6 1999 Our results suggest that rifampicin, ticarcillin, cephalosporins and aminoglycosides act as inhibitors of production IFN-gamma. Rifampin 25-35 interferon gamma Mus musculus 117-126 10548883-11 1999 Many of the PXR activators are widely used drugs such as dexamethasone, lovastatin, and rifampicin, whose induction of CYP3A levels causes them to promote the metabolism of other drugs, often with adverse consequences. Rifampin 88-98 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 119-124 9825726-1 1998 Cultured rabbit hepatocytes were used to compare the relative activities of cytokines to inhibit the constitutive or rifampicin (RIF)-induced expression of the cytochrome P450 3A6 gene (CYP3A6). Rifampin 117-127 cytochrome P450 3A6 Oryctolagus cuniculus 160-179 9862241-1 1998 AIMS: The N-deethylation of lignocaine to monoethylglycinexylidide (MEGX) is partially catalysed by the rifampicin inducible P-450 isoenzyme CYP3A4. Rifampin 104-114 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 141-147 9862241-13 1998 CONCLUSION: An insignificant increase of MEGX plasma concentrations was found in 10 volunteers after induction of CYP3A4 activity by rifampicin. Rifampin 133-143 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 114-120 9883615-2 1998 0.3-0.5 g).rifampicin (RFP. Rifampin 11-21 tripartite motif containing 27 Homo sapiens 23-26 9950072-0 1998 Induction of hepatic mrp2 (cmrp/cmoat) gene expression in nonhuman primates treated with rifampicin or tamoxifen. Rifampin 89-99 ATP binding cassette subfamily C member 2 Macaca mulatta 21-25 9950072-3 1998 Based on previous observations in rat hepatocytes, the inducibility of mrp2 gene expression in primate liver was investigated in rhesus monkeys treated with tamoxifen or rifampicin. Rifampin 170-180 ATP binding cassette subfamily C member 2 Rattus norvegicus 71-75 9950072-4 1998 It was found that treatment with tamoxifen (25 mg/kg per day; over 7 days) or rifampicin (15 mg/kg per day; over 7 days) leading to an induction of cytochrome P450 3A4, resulted in a strong increase in mrp2 mRNA in the liver of male and female rhesus monkeys. Rifampin 78-88 ATP binding cassette subfamily C member 2 Macaca mulatta 202-206 9871429-1 1998 BACKGROUND: Rifampin (INN, rifampicin) is a potent inducer of cytochrome P450 (CYP) enzymes involved in drug metabolism and therefore causes many drug interactions. Rifampin 12-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 9871429-1 1998 BACKGROUND: Rifampin (INN, rifampicin) is a potent inducer of cytochrome P450 (CYP) enzymes involved in drug metabolism and therefore causes many drug interactions. Rifampin 12-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 9871429-1 1998 BACKGROUND: Rifampin (INN, rifampicin) is a potent inducer of cytochrome P450 (CYP) enzymes involved in drug metabolism and therefore causes many drug interactions. Rifampin 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 9871429-1 1998 BACKGROUND: Rifampin (INN, rifampicin) is a potent inducer of cytochrome P450 (CYP) enzymes involved in drug metabolism and therefore causes many drug interactions. Rifampin 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 9871429-9 1998 CONCLUSIONS: Rifampin markedly reduces the plasma concentrations of tamoxifen and toremifene by inducing their CYP3A4-mediated metabolism. Rifampin 13-21 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 111-117 9871429-10 1998 Concomitant use of rifampin or other potent inducers of CYP3A4 with tamoxifen and toremifene may reduce the efficacy of these antiestrogens. Rifampin 19-27 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 56-62 9825726-1 1998 Cultured rabbit hepatocytes were used to compare the relative activities of cytokines to inhibit the constitutive or rifampicin (RIF)-induced expression of the cytochrome P450 3A6 gene (CYP3A6). Rifampin 117-127 cytochrome P450 3A6 Oryctolagus cuniculus 186-192 9825726-1 1998 Cultured rabbit hepatocytes were used to compare the relative activities of cytokines to inhibit the constitutive or rifampicin (RIF)-induced expression of the cytochrome P450 3A6 gene (CYP3A6). Rifampin 129-132 cytochrome P450 3A6 Oryctolagus cuniculus 160-179 9825726-1 1998 Cultured rabbit hepatocytes were used to compare the relative activities of cytokines to inhibit the constitutive or rifampicin (RIF)-induced expression of the cytochrome P450 3A6 gene (CYP3A6). Rifampin 129-132 cytochrome P450 3A6 Oryctolagus cuniculus 186-192 9812178-19 1998 Rifampicin (rifampin), a potent CYP3A inducer, decreased the AUC of ritonavir by only 35%. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-37 9770465-3 1998 hPAR was found to be efficiently activated by pregnanes and by clinically used drugs including rifampicin, an antibiotic known to selectively induce human but not murine CYP3A expression. Rifampin 95-105 nuclear receptor subfamily 1 group I member 2 Homo sapiens 0-4 9770465-3 1998 hPAR was found to be efficiently activated by pregnanes and by clinically used drugs including rifampicin, an antibiotic known to selectively induce human but not murine CYP3A expression. Rifampin 95-105 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 170-175 9812178-19 1998 Rifampicin (rifampin), a potent CYP3A inducer, decreased the AUC of ritonavir by only 35%. Rifampin 12-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-37 9784933-16 1998 Inducers of cytochrome P450 (CYP) 3A4, e.g. rifampicin (rifampin) and phenytoin should not be combined with nisoldipine CC, as they may reduce its bioavailability and result in a loss of efficacy. Rifampin 44-54 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 12-37 9806107-7 1998 Hyperuricaemia, which was reported only during the intensive phase, elevated ALT and AST levels and neutropenia were the most common treatment-related adverse events reported in patients receiving rifapentine- or rifampicin-containing regimens for tuberculosis in 1 Western study. Rifampin 213-223 solute carrier family 17 member 5 Homo sapiens 85-88 9732934-0 1998 Unexpected clinical remission of cholestasis after rifampicin therapy in patients with normal or slightly increased levels of gamma-glutamyl transpeptidase. Rifampin 51-61 inactive glutathione hydrolase 2 Homo sapiens 126-155 9732934-12 1998 CONCLUSION: Rifampicin may induce clinical remission, and perhaps prevent clinical relapses of intrahepatic cholestasis with normal or slightly increased levels of gamma-glutamyl transpeptidase. Rifampin 12-22 inactive glutathione hydrolase 2 Homo sapiens 164-193 9784933-16 1998 Inducers of cytochrome P450 (CYP) 3A4, e.g. rifampicin (rifampin) and phenytoin should not be combined with nisoldipine CC, as they may reduce its bioavailability and result in a loss of efficacy. Rifampin 56-64 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 12-37 9650537-5 1998 Rifampin induced CYP3A, reflected in a mean percent (+/- standard deviation) increase in EBT values of 86 +/- 30%. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 17-22 9753209-3 1998 Known inducers of CYP3A, such as rifampin, phenytoin, carbamazepine, and phenobarbital, increase the urinary excretion of 6beta-hydroxycortisol and the ratio of 6beta-hydroxycortisol to cortisol. Rifampin 33-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 18-23 9675018-6 1998 Taxol was almost as effective as rifampicin in inducing CYP3A in two of the cultures, but less effective than rifampicin in two other cultures. Rifampin 33-43 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 56-61 9661035-1 1998 The contributions of 23 insertion, deletion, or missense mutations within an 81-bp fragment of rpoB, the gene encoding the beta-subunit of the DNA-dependent RNA polymerase of Mycobacterium tuberculosis, to the development of resistance to rifamycins (rifampin, rifabutin, rifapentine, and KRM-1648) in 29 rifampin-resistant clinical isolates were defined. Rifampin 251-259 DNA-directed RNA polymerase subunit beta Mycobacterium tuberculosis H37Rv 95-99 9661035-1 1998 The contributions of 23 insertion, deletion, or missense mutations within an 81-bp fragment of rpoB, the gene encoding the beta-subunit of the DNA-dependent RNA polymerase of Mycobacterium tuberculosis, to the development of resistance to rifamycins (rifampin, rifabutin, rifapentine, and KRM-1648) in 29 rifampin-resistant clinical isolates were defined. Rifampin 305-313 DNA-directed RNA polymerase subunit beta Mycobacterium tuberculosis H37Rv 95-99 9661035-2 1998 Specific mutant rpoB alleles led to the development of cross-resistance to all rifamycins tested, while a subset of mutations were associated with resistance to rifampin and rifapentine but not to KRM-1648 or rifabutin. Rifampin 161-169 DNA-directed RNA polymerase subunit beta Mycobacterium tuberculosis H37Rv 16-20 9663807-3 1998 CYP2C9 activity in vivo is inducible by rifampicin. Rifampin 40-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 9677642-3 1998 CASE REPORT: Three patients developed a pruritic rash 1 hour after the first dose of isoniazide, rifampicine, pyrazinamide and ethambutol given simultaneously. Rifampin 97-108 HRas proto-oncogene, GTPase Homo sapiens 49-55 9667078-0 1998 Characterization of cytochrome P450 (CYP3A12) induction by rifampicin in dog liver. Rifampin 59-69 cytochrome P450 3A12 Canis lupus familiaris 37-44 9667078-2 1998 Effects of rifampicin (Rif) on the contents of cytochrome P450 (P450) enzymes (CYP1A1/2, 2B11, 2C21 and 3A12) assessed by enzyme-linked immunosorbent assay and catalytic activities (ethoxyresorufin O-deethylase, and testosterone 6 beta-, 16 alpha- and 16 beta-hydroxylase; 6 beta-, 16 alpha- and 16 beta-OHT) in dog liver microsomes were compared between liver lobes of both the male and female dogs. Rifampin 11-21 Cytochrome P450 1A1 Canis lupus familiaris 79-93 9667078-13 1998 These results indicate that Rif treatment induces the expression of CYP3A12 protein, and correlates well with the elevation of its catalytic activity (6 beta- and 16 beta-OHT), and that the female dog is more responsive to Rif treatment as compared with the male. Rifampin 28-31 cytochrome P450 3A12 Canis lupus familiaris 68-75 9625027-3 1998 Other medications, e.g., rifampin and phenobarbital, which also induce p450 3A activity, have been reported to significantly decrease cyclosporine (CsA) concentrations. Rifampin 25-33 ERCC excision repair 8, CSA ubiquitin ligase complex subunit Homo sapiens 148-151 9556634-8 1998 Nicked DNA was significantly generated, and the frequency of mutations resistant to rifampicin was increased by 30-fold, depending upon the expression of Bax. Rifampin 84-94 BCL2 associated X, apoptosis regulator Homo sapiens 154-157 9578186-8 1998 CONCLUSIONS: The strong interaction between rifampicin and buspirone is probably mostly due to enhanced CYP3A4-mediated first-pass metabolism of buspirone. Rifampin 44-54 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-110 9489590-0 1998 The induction effect of rifampicin on activity of mephenytoin 4"-hydroxylase related to M1 mutation of CYP2C19 and gene dose. Rifampin 24-34 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 50-76 9514072-4 1998 In male rats, western blot analyses showed that rifampicin and dexamethasone caused 50% and 5-fold increases in Pgp levels, respectively. Rifampin 48-58 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 112-115 9514072-5 1998 RNase protection assays using gene-specific probes for the three Pgp isoforms revealed a 3-fold increase in mdr2 mRNA levels after dexamethasone administration and a 2-fold increase following rifampicin treatment. Rifampin 192-202 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 65-68 9454825-5 1998 When hepatocytes were treated with inducers, marked increases in the specific activities of CYP1A1/2 by 3-methylcholanthrene and CYP3A4 by rifampicin were observed, and these inductive effects were greatly reduced in the presence of HGF. Rifampin 139-149 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 92-98 9454825-5 1998 When hepatocytes were treated with inducers, marked increases in the specific activities of CYP1A1/2 by 3-methylcholanthrene and CYP3A4 by rifampicin were observed, and these inductive effects were greatly reduced in the presence of HGF. Rifampin 139-149 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 129-135 9454825-7 1998 The observed changes in the activity and protein levels of CYP1A2 and CYP3A4 correlated with a reduction in the specific messenger RNA levels both in control, 3-methylcholanthrene-treated (for CYP1A2) and rifampicin-treated (for CYP3A4) hepatocytes, which thus suggested that HGF could down-regulate CYP expression at a pretranslational level. Rifampin 205-215 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 59-65 9454825-7 1998 The observed changes in the activity and protein levels of CYP1A2 and CYP3A4 correlated with a reduction in the specific messenger RNA levels both in control, 3-methylcholanthrene-treated (for CYP1A2) and rifampicin-treated (for CYP3A4) hepatocytes, which thus suggested that HGF could down-regulate CYP expression at a pretranslational level. Rifampin 205-215 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 70-76 9454825-7 1998 The observed changes in the activity and protein levels of CYP1A2 and CYP3A4 correlated with a reduction in the specific messenger RNA levels both in control, 3-methylcholanthrene-treated (for CYP1A2) and rifampicin-treated (for CYP3A4) hepatocytes, which thus suggested that HGF could down-regulate CYP expression at a pretranslational level. Rifampin 205-215 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 229-235 9454825-7 1998 The observed changes in the activity and protein levels of CYP1A2 and CYP3A4 correlated with a reduction in the specific messenger RNA levels both in control, 3-methylcholanthrene-treated (for CYP1A2) and rifampicin-treated (for CYP3A4) hepatocytes, which thus suggested that HGF could down-regulate CYP expression at a pretranslational level. Rifampin 205-215 hepatocyte growth factor Homo sapiens 276-279 9454825-7 1998 The observed changes in the activity and protein levels of CYP1A2 and CYP3A4 correlated with a reduction in the specific messenger RNA levels both in control, 3-methylcholanthrene-treated (for CYP1A2) and rifampicin-treated (for CYP3A4) hepatocytes, which thus suggested that HGF could down-regulate CYP expression at a pretranslational level. Rifampin 205-215 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 9489590-0 1998 The induction effect of rifampicin on activity of mephenytoin 4"-hydroxylase related to M1 mutation of CYP2C19 and gene dose. Rifampin 24-34 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 103-110 9402948-5 1997 In addition, the expression and inducibility of CYP proteins of the CYP1, CYP2 and CYP3 families in response to their prototypical inducers including 2,3,7,8-tetrachlorodibenzo(p)dioxin and rifampicin, have been evaluated by immunoblot analysis of microsomes or cell lysates. Rifampin 190-200 cytochrome P450 family 27 subfamily B member 1 Homo sapiens 68-72 9427613-0 1998 The antibiotic rifampicin is a nonsteroidal ligand and activator of the human glucocorticoid receptor. Rifampin 15-25 nuclear receptor subfamily 3 group C member 1 Homo sapiens 78-101 9385296-1 1997 Rifampin is a potent inducer of cytochrome P-450 oxidative enzymes. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 9402947-2 1997 Our study here represents the first reported study that directly compares the cytochrome P450 isozyme 3A (CYP3A) induction potential of three antimicrobials derived from rifamycin B, namely, rifampin, rifapentine and rifabutin. Rifampin 191-199 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 78-104 9402947-4 1997 Results obtained with hepatocytes from four different human donors show consistently that rifampin and rifapentine are potent inducers of CYP3A, while a significantly lower induction potential is observed for rifabutin. Rifampin 90-98 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 138-143 9402948-5 1997 In addition, the expression and inducibility of CYP proteins of the CYP1, CYP2 and CYP3 families in response to their prototypical inducers including 2,3,7,8-tetrachlorodibenzo(p)dioxin and rifampicin, have been evaluated by immunoblot analysis of microsomes or cell lysates. Rifampin 190-200 peptidylprolyl isomerase F Homo sapiens 83-87 9402949-9 1997 Likewise, Cyp3A and Cyp2A6 mRNAs can be shown to be induced in primary human hepatocytes cultured on collagen-coated plates and treated with rifampin for 72 h. By contrast, mRNA levels for Cyp1A1 and Cyp2E1 were not increased by rifampin treatment. Rifampin 141-149 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 10-15 9402949-9 1997 Likewise, Cyp3A and Cyp2A6 mRNAs can be shown to be induced in primary human hepatocytes cultured on collagen-coated plates and treated with rifampin for 72 h. By contrast, mRNA levels for Cyp1A1 and Cyp2E1 were not increased by rifampin treatment. Rifampin 141-149 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 20-26 9402949-9 1997 Likewise, Cyp3A and Cyp2A6 mRNAs can be shown to be induced in primary human hepatocytes cultured on collagen-coated plates and treated with rifampin for 72 h. By contrast, mRNA levels for Cyp1A1 and Cyp2E1 were not increased by rifampin treatment. Rifampin 141-149 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 189-195 9402949-9 1997 Likewise, Cyp3A and Cyp2A6 mRNAs can be shown to be induced in primary human hepatocytes cultured on collagen-coated plates and treated with rifampin for 72 h. By contrast, mRNA levels for Cyp1A1 and Cyp2E1 were not increased by rifampin treatment. Rifampin 141-149 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 200-206 9402949-9 1997 Likewise, Cyp3A and Cyp2A6 mRNAs can be shown to be induced in primary human hepatocytes cultured on collagen-coated plates and treated with rifampin for 72 h. By contrast, mRNA levels for Cyp1A1 and Cyp2E1 were not increased by rifampin treatment. Rifampin 229-237 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 10-15 9402949-9 1997 Likewise, Cyp3A and Cyp2A6 mRNAs can be shown to be induced in primary human hepatocytes cultured on collagen-coated plates and treated with rifampin for 72 h. By contrast, mRNA levels for Cyp1A1 and Cyp2E1 were not increased by rifampin treatment. Rifampin 229-237 cytochrome P450 family 2 subfamily A member 6 Homo sapiens 20-26 9402949-9 1997 Likewise, Cyp3A and Cyp2A6 mRNAs can be shown to be induced in primary human hepatocytes cultured on collagen-coated plates and treated with rifampin for 72 h. By contrast, mRNA levels for Cyp1A1 and Cyp2E1 were not increased by rifampin treatment. Rifampin 229-237 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 189-195 9413920-7 1997 Compared with control (dimethyl sulphoxide only treated) liver slice microsomes, rifampicin increased levels of CYP3A4 but had no effect on CYP1A2. Rifampin 81-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 112-118 9413920-9 1997 These results demonstrate that rifampicin induces CYP3A isoforms, but not CYP1A2, in cultured human liver slices. Rifampin 31-41 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-55 9353130-11 1997 Rifampicin treatment (50 micromol/L) for 7 days resulted in a 3.7-fold induction of CYP 3A4 at day 9 in culture. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 84-91 9353130-14 1997 These results demonstrate that rat and human hepatocytes preserve the major forms of CYP isozymes and phase II activities and respond to inducing drugs such as rifampicin. Rifampin 160-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 9391787-0 1997 Relationship between cytochrome P-450 induction by rifampicin, hepatic volume and portal blood flow in man. Rifampin 51-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 9391787-10 1997 CONCLUSION: A fourfold increase of urinary 6-beta-hydroxycortisol output after induction of cytochrome P-450 by rifampicin is associated with a significant but less than 10% increase in human liver volume. Rifampin 112-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 9350037-9 1997 Whereas ATA caused increased stability of PR isoforms, Rifampicin induced a upshift in the mobility of PR in SDS gels-a phenomenon associated with hyperphosphorylation of steroid receptors (SRs). Rifampin 55-65 progesterone receptor Homo sapiens 103-105 9350037-12 1997 We propose that the inhibition of T47D cell proliferation by phenanthroline, Rifampicin and ATA results from a number of cellular changes that include regulation of p53 and PR. Rifampin 77-87 tumor protein p53 Homo sapiens 165-168 9350037-12 1997 We propose that the inhibition of T47D cell proliferation by phenanthroline, Rifampicin and ATA results from a number of cellular changes that include regulation of p53 and PR. Rifampin 77-87 progesterone receptor Homo sapiens 173-175 9596814-1 1996 OBJECTIVE: To evaluate rpoB gene mutation in rifampin-resistant Mycobacterium tuberculosis and its relationship with rifampin resistance. Rifampin 45-53 ER membrane protein complex subunit 3 Rattus norvegicus 23-27 9298257-8 1997 Rifampin induction of CYP3A, a known effect of rifampin in vivo, was also reproduced in primary human hepatocytes. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 22-27 9298257-11 1997 To demonstrate that the rifampin-induction of testosterone 6 beta-hydroxylation could be generalized to other CYP3A4 substrates, we evaluated the metabolism of another known substrate of CYP3A4, lidocaine. Rifampin 24-32 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 187-193 9157990-5 1997 Rifampin (1 microM, 96-h exposure) was a particularly potent inducer of ifosfamide and cyclophosphamide 4-hydroxylation, as well as of CYP3A protein levels and CYP3A-dependent testosterone 6beta-hydroxylation. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 135-140 9157990-5 1997 Rifampin (1 microM, 96-h exposure) was a particularly potent inducer of ifosfamide and cyclophosphamide 4-hydroxylation, as well as of CYP3A protein levels and CYP3A-dependent testosterone 6beta-hydroxylation. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 160-165 9141435-3 1997 In the absence of EGF, the accumulation of CYP3A4 and CYP1A2 messenger RNAs (mRNAs) in response to their respective inducers (rifampicin and dioxin) was dramatically decreased in subconfluent culture with respect to confluent cultures. Rifampin 126-136 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 43-49 9141435-3 1997 In the absence of EGF, the accumulation of CYP3A4 and CYP1A2 messenger RNAs (mRNAs) in response to their respective inducers (rifampicin and dioxin) was dramatically decreased in subconfluent culture with respect to confluent cultures. Rifampin 126-136 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 54-60 9084959-20 1997 Recognised enzyme inducers of CYP3A, which are likely to be prescribed for patients with HIV disease, include rifampicin (rifampin) [treatment of pulmonary tuberculosis], rifabutin (treatment and prophylaxis of Mycobacterium avium complex), phenobarbital (phenobarbitone), phenytoin and carbamazepine (treatment of seizures secondary to cerebral toxoplasmosis or cerebral lymphoma). Rifampin 110-120 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 30-35 9084959-20 1997 Recognised enzyme inducers of CYP3A, which are likely to be prescribed for patients with HIV disease, include rifampicin (rifampin) [treatment of pulmonary tuberculosis], rifabutin (treatment and prophylaxis of Mycobacterium avium complex), phenobarbital (phenobarbitone), phenytoin and carbamazepine (treatment of seizures secondary to cerebral toxoplasmosis or cerebral lymphoma). Rifampin 122-130 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 30-35 9187114-8 1997 By the introduction of recombinant hMMH, the rate of mutation, the formation of rifampicin-resistant revertants, was reduced by 4-7 fold. Rifampin 80-90 8-oxoguanine DNA glycosylase Homo sapiens 35-39 9157990-7 1997 In one human hepatocyte culture that contained the polymorphically expressed CYP3A5 in addition to the more widely expressed CYP3A4, only CYP3A4 was induced by cyclophosphamide, ifosfamide, and rifampin. Rifampin 194-202 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 77-83 9157990-7 1997 In one human hepatocyte culture that contained the polymorphically expressed CYP3A5 in addition to the more widely expressed CYP3A4, only CYP3A4 was induced by cyclophosphamide, ifosfamide, and rifampin. Rifampin 194-202 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 138-144 9145822-0 1997 Effect of combination therapy of rifampicin and azithromycin on TNF levels during a rat model of chronic osteomyelitis. Rifampin 33-43 tumor necrosis factor Rattus norvegicus 64-67 9178371-4 1997 Seven different possible metabolites were identified in the microsomal preparations from rats treated with rifampin or dexamethasone whereas the microsomes from the control rats failed to produce the mono-demethylated and monohydroxylated metabolite of TAC (TAC+2, m/z = 805.5). Rifampin 107-115 tachykinin precursor 3 Rattus norvegicus 253-256 9148761-1 1997 Rifampicin and its analogues, p-benzoquinone and hydroquinone, inhibited the toxicity of preformed aggregates of human islet amyloid polypeptide, amylin, to rat pheochromocytoma PC12 cells, when preincubated with the aggregated peptide before addition to cell cultures. Rifampin 0-10 islet amyloid polypeptide Homo sapiens 146-152 9024169-2 1997 Rifampin (rifampicin) is a potent inducer of CYP3A4 and it is known to markedly reduce plasma concentrations and effects of drugs such as midazolam. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 45-51 9024169-2 1997 Rifampin (rifampicin) is a potent inducer of CYP3A4 and it is known to markedly reduce plasma concentrations and effects of drugs such as midazolam. Rifampin 10-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 45-51 9024169-12 1997 This is most likely due to increased metabolism of triazolam after induction of CYP3A4 in the gut wall and liver by rifampin. Rifampin 116-124 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 80-86 9010637-5 1997 The inducers of CYP3A and/or CYP2C9 (e.g. rifampicin and phenytoin) are likely to alter the disposition of seratrodast. Rifampin 42-52 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 16-21 9010637-5 1997 The inducers of CYP3A and/or CYP2C9 (e.g. rifampicin and phenytoin) are likely to alter the disposition of seratrodast. Rifampin 42-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 9131486-5 1996 CYP3A activity can also be readily modulated by inducers like rifampicin and several anticonvulsant agents, and many potent inhibitors exist such as azole antifungal agents and macrolide antibiotics. Rifampin 62-72 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-5 8855178-2 1996 CYP3A4 for example is present and inducible by rifampin in epithelial cells of the gastrointestinal tract. Rifampin 47-55 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 8883230-14 1996 Prophylactic ciprofloxacin and rifampin is a well-tolerated and highly effective combination that effectively decreases the risk of both gram-positive and gram-negative bacterial infection following HDC/ASCR. Rifampin 31-39 prion protein Homo sapiens 203-207 8843295-13 1996 A comparison of equal doses of rifampin and rifabutin administered to healthy volunteers for 2 weeks indicates that induction of CYP1A2, as measured by theophylline clearance, is significantly less following rifabutin treatment than it is following rifampin treatment. Rifampin 31-39 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 129-135 8753820-0 1996 The fetal specific gene CYP3A7 is inducible by rifampicin in adult human hepatocytes in primary culture. Rifampin 47-57 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 24-30 8753820-5 1996 However, when the cells were treated for 48 hours with 25 microM rifampicin, both CYP3A4 and CYP3A7 mRNAs were strongly induced in the 3 and in 2 of 3 cultures examined, respectively. Rifampin 65-75 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 82-88 8753820-5 1996 However, when the cells were treated for 48 hours with 25 microM rifampicin, both CYP3A4 and CYP3A7 mRNAs were strongly induced in the 3 and in 2 of 3 cultures examined, respectively. Rifampin 65-75 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 93-99 8753820-8 1996 These results show for the first time that CYP3A7 and CYP3A4 mRNAs, but not the proteins, are co-inducible by rifampicin in adult human hepatocytes in culture. Rifampin 110-120 cytochrome P450 family 3 subfamily A member 7 Homo sapiens 43-49 8753820-8 1996 These results show for the first time that CYP3A7 and CYP3A4 mRNAs, but not the proteins, are co-inducible by rifampicin in adult human hepatocytes in culture. Rifampin 110-120 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 54-60 8843295-3 1996 This study, involving 12 adult male volunteers, compared the effects of 14-day courses of rifampin and rifabutin on clearance of theophylline, a substrate for the hepatic microsomal enzyme CYP1A2. Rifampin 90-98 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 189-195 8819303-7 1996 The oral treatment of male dogs with phenobarbital (PB), rifampicin (Rif) or beta-naphthoflavone (beta-NF) induced significant increases in the contents of CYP1A1/2 (12-fold by beta-NF), 2B11 (16-fold by PB), 2C21 (2-fold by PB) and 3A12 (5-fold by PB and Rif), resulting in marked proportional alterations of the P450 enzymes in dog liver microsomes. Rifampin 57-67 Cytochrome P450 1A1 Canis lupus familiaris 156-162 8670846-6 1996 Expression of CDG or TDG in Escherichia coli causes 4- to 100-fold increases in the yield of rifampicin-resistant mutants. Rifampin 93-103 thymine DNA glycosylase Homo sapiens 21-24 8700101-1 1996 Interindividual variation in the spontaneous and in the glucocorticoid-or rifampicin-inducible expression of the CYP3A cytochromes P450, the dominant froms of this supergene family that catalyze the oxidation of numerous drugs and environmental chemicals in human liver, remains largely unexplained, due in part to the lack of a validated animal model. Rifampin 74-84 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 113-118 8819303-7 1996 The oral treatment of male dogs with phenobarbital (PB), rifampicin (Rif) or beta-naphthoflavone (beta-NF) induced significant increases in the contents of CYP1A1/2 (12-fold by beta-NF), 2B11 (16-fold by PB), 2C21 (2-fold by PB) and 3A12 (5-fold by PB and Rif), resulting in marked proportional alterations of the P450 enzymes in dog liver microsomes. Rifampin 69-72 Cytochrome P450 1A1 Canis lupus familiaris 156-162 8819303-7 1996 The oral treatment of male dogs with phenobarbital (PB), rifampicin (Rif) or beta-naphthoflavone (beta-NF) induced significant increases in the contents of CYP1A1/2 (12-fold by beta-NF), 2B11 (16-fold by PB), 2C21 (2-fold by PB) and 3A12 (5-fold by PB and Rif), resulting in marked proportional alterations of the P450 enzymes in dog liver microsomes. Rifampin 256-259 Cytochrome P450 1A1 Canis lupus familiaris 156-162 8690981-3 1996 This was the motive behind the state of Sao Paulo"s Health Department"s decision to carry out a study that would evaluate not only the incidence rate of adverse effects of rifampin in relation to kidney function but also in relation to the use of WHO/MDT in general. Rifampin 172-180 solute carrier family 4 member 1 (Diego blood group) Homo sapiens 40-43 8621783-8 1996 During normal Na diet, RIF PGE2 and cGMP increased in response to angiotensin II. Rifampin 23-26 angiotensinogen Rattus norvegicus 66-80 8836814-0 1996 Inducing properties of rifampicin and rifabutin for selected enzyme activities of the cytochrome P-450 and UDP-glucuronosyltransferase superfamilies in female rat liver. Rifampin 23-33 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 86-102 8836814-0 1996 Inducing properties of rifampicin and rifabutin for selected enzyme activities of the cytochrome P-450 and UDP-glucuronosyltransferase superfamilies in female rat liver. Rifampin 23-33 UDP glycosyltransferase 2 family, polypeptide B Rattus norvegicus 107-134 8836814-3 1996 In the present study a strong induction of cytochrome P-450 3A-dependent enzyme activities was observed in female rat liver microsomes after high dose treatment (> or = 250 mg/kg/day for 9 days) with rifampicin, resulting in an up to 30-fold enhanced hydroxylation rate of testosterone in the 2 beta-, 6 beta- and 15 beta-position in vitro. Rifampin 203-213 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 43-59 8836814-5 1996 A steep increase in cytochrome P-450 3A activity on a moderate elevation of the dose administered, together with the previously observed lack of efficient induction with doses below 200 mg/kg/day demonstrated that there is a threshold in enzyme induction by rifampicin. Rifampin 258-268 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 20-36 8836814-7 1996 Induction by rifabutin showed an isoenzyme-selectivity profile similar to that produced by rifampicin, but the maximally achievable induction of cytochrome P-450 3A by rifabutin was about two-fold lower compared with rifampicin. Rifampin 217-227 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 145-161 8619627-5 1996 A putative CYP3A protein is expressed in PLHC-1 cells but its content was not altered by exposure of cultures to the prototypical mammalian CYP3A inducers dexamethasone (DEX), pregnenolone-16 alpha-carbonitrile (PCN), or rifampicin (RIF). Rifampin 221-231 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 11-16 8619627-5 1996 A putative CYP3A protein is expressed in PLHC-1 cells but its content was not altered by exposure of cultures to the prototypical mammalian CYP3A inducers dexamethasone (DEX), pregnenolone-16 alpha-carbonitrile (PCN), or rifampicin (RIF). Rifampin 233-236 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 11-16 8619627-11 1996 Two other glucocorticoid receptor (GR) agonists, cortisol and prednisone, also produced a strong potentiation of CYP1A induction, but other mammalian CYP3A inducers that are not GR agonists, such as the anti-glucocorticoid PCN, the anti-mineralocorticoid spironolactone, or the macrolide antibiotics RIF and troleandomycin, did not potentiate the CYP1A induction in PLHC-1 cells. Rifampin 300-303 nuclear receptor subfamily 3 group C member 1 Homo sapiens 10-33 8619627-11 1996 Two other glucocorticoid receptor (GR) agonists, cortisol and prednisone, also produced a strong potentiation of CYP1A induction, but other mammalian CYP3A inducers that are not GR agonists, such as the anti-glucocorticoid PCN, the anti-mineralocorticoid spironolactone, or the macrolide antibiotics RIF and troleandomycin, did not potentiate the CYP1A induction in PLHC-1 cells. Rifampin 300-303 nuclear receptor subfamily 3 group C member 1 Homo sapiens 35-37 8633005-0 1996 P-glycoprotein: a major determinant of rifampicin-inducible expression of cytochrome P4503A in mice and humans. Rifampin 39-49 phosphoglycolate phosphatase Mus musculus 0-14 8633005-0 1996 P-glycoprotein: a major determinant of rifampicin-inducible expression of cytochrome P4503A in mice and humans. Rifampin 39-49 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 74-80 8633005-3 1996 The magnitude of CYP3A induction by rifampicin was compared in the human parental colon carcinoma cell line LS 180/WT (wild type) and in two derivative clones overexpressing the human multidrug resistance gene MDR1 (also designated PGY1) because of either drug selection (LS 180/ADR) or transfection with MDRI cDNA (LS 180/MDR). Rifampin 36-46 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 17-22 8633005-4 1996 In both MDR1 cDNA-overexpressing clones, rifampicin induction of CYP3A mRNA and protein was decreased and required greater rifampicin concentrations compared with parental cells. Rifampin 41-51 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 8-12 8633005-4 1996 In both MDR1 cDNA-overexpressing clones, rifampicin induction of CYP3A mRNA and protein was decreased and required greater rifampicin concentrations compared with parental cells. Rifampin 41-51 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 65-70 8633005-4 1996 In both MDR1 cDNA-overexpressing clones, rifampicin induction of CYP3A mRNA and protein was decreased and required greater rifampicin concentrations compared with parental cells. Rifampin 123-133 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 65-70 8633005-6 1996 Oral treatment with increasing doses of rifampicin resulted in elevated drug levels in the livers of mdr1a (-/-) mice compared with mdr1a (+/+) mice at all doses. Rifampin 40-50 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 101-106 8633005-6 1996 Oral treatment with increasing doses of rifampicin resulted in elevated drug levels in the livers of mdr1a (-/-) mice compared with mdr1a (+/+) mice at all doses. Rifampin 40-50 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 132-137 8633005-7 1996 Consistent with the enhanced accumulation of rifampicin in mdr1a (-/-) mice, lower doses of rifampicin were required for induction of CYP3A proteins, and the magnitude of CYP3A induction was greater at all doses of rifampicin in mdr1a (-/-) mice compared with mdr1a (+/+) mice. Rifampin 45-55 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 59-64 8633005-7 1996 Consistent with the enhanced accumulation of rifampicin in mdr1a (-/-) mice, lower doses of rifampicin were required for induction of CYP3A proteins, and the magnitude of CYP3A induction was greater at all doses of rifampicin in mdr1a (-/-) mice compared with mdr1a (+/+) mice. Rifampin 92-102 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 134-139 8633005-7 1996 Consistent with the enhanced accumulation of rifampicin in mdr1a (-/-) mice, lower doses of rifampicin were required for induction of CYP3A proteins, and the magnitude of CYP3A induction was greater at all doses of rifampicin in mdr1a (-/-) mice compared with mdr1a (+/+) mice. Rifampin 92-102 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 229-234 8633005-7 1996 Consistent with the enhanced accumulation of rifampicin in mdr1a (-/-) mice, lower doses of rifampicin were required for induction of CYP3A proteins, and the magnitude of CYP3A induction was greater at all doses of rifampicin in mdr1a (-/-) mice compared with mdr1a (+/+) mice. Rifampin 92-102 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 229-234 8633005-7 1996 Consistent with the enhanced accumulation of rifampicin in mdr1a (-/-) mice, lower doses of rifampicin were required for induction of CYP3A proteins, and the magnitude of CYP3A induction was greater at all doses of rifampicin in mdr1a (-/-) mice compared with mdr1a (+/+) mice. Rifampin 92-102 cytochrome P450, family 3, subfamily a, polypeptide 11 Mus musculus 134-139 8633005-7 1996 Consistent with the enhanced accumulation of rifampicin in mdr1a (-/-) mice, lower doses of rifampicin were required for induction of CYP3A proteins, and the magnitude of CYP3A induction was greater at all doses of rifampicin in mdr1a (-/-) mice compared with mdr1a (+/+) mice. Rifampin 92-102 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 229-234 8633005-7 1996 Consistent with the enhanced accumulation of rifampicin in mdr1a (-/-) mice, lower doses of rifampicin were required for induction of CYP3A proteins, and the magnitude of CYP3A induction was greater at all doses of rifampicin in mdr1a (-/-) mice compared with mdr1a (+/+) mice. Rifampin 92-102 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 229-234 8636108-6 1996 These results suggest that at least one mechanism of rifampicin-mediated inhibition of A beta aggregation and neurotoxicity involves scavenging of free radicals and that rifampicin and/or appropriate hydroxyl radical scavengers may have therapeutic potential for Alzheimer"s disease. Rifampin 53-63 amyloid beta precursor protein Rattus norvegicus 87-93 8549036-2 1996 Rifampin is a potent enzyme inducer that may seriously interact with some substrates of CYP3A4. Rifampin 0-8 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-94 8640817-8 1996 Docetaxel metabolism was induced in vitro in human hepatocytes by dexamethasone and rifampicin, both classical CYP3A inducers. Rifampin 84-94 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 111-116 8632764-3 1996 P-glycoprotein and CYP3A4 were constitutively expressed in both LS180/AD50 and LS180/WT cells, and both proteins were up-regulated after treatment with many drugs, including rifampicin, phenobarbital, clotrimazole, reserpine, and isosafrole. Rifampin 174-184 ATP binding cassette subfamily B member 1 Homo sapiens 0-14 8632764-3 1996 P-glycoprotein and CYP3A4 were constitutively expressed in both LS180/AD50 and LS180/WT cells, and both proteins were up-regulated after treatment with many drugs, including rifampicin, phenobarbital, clotrimazole, reserpine, and isosafrole. Rifampin 174-184 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-25 8549036-10 1996 CONCLUSIONS: The observed substantial decrease in plasma concentrations and effects of midazolam most likely results from induction of CYP3A4 by rifampin in both the gut wall and the liver. Rifampin 145-153 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 135-141 7585637-5 1995 Pretreatment of the cells with 3-methylcholanthrene or rifampicin, inducers of CYP1A2 and CYP3A4, respectively, caused a significant increase in AFB1 metabolism. Rifampin 55-65 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 79-85 7585637-5 1995 Pretreatment of the cells with 3-methylcholanthrene or rifampicin, inducers of CYP1A2 and CYP3A4, respectively, caused a significant increase in AFB1 metabolism. Rifampin 55-65 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-96 8786635-1 1995 Rifampicin conferred significant protection against carbon tetrachloride (CCl4)-induced liver injury. Rifampin 0-10 chemokine (C-C motif) ligand 4 Mus musculus 74-78 8786635-2 1995 Serum alanine transaminase (ALT) and aspartate transaminase (AST) activities were not markedly altered and only hepatocellular fatty degeneration was found in mice pretreated with rifampicin (200 mg/kg), whereas severe centrilobular necrosis was observed and serum ALT and AST activities were as high as 281 and 271 I.U./l, respectively, in the control group following administration of CCl4 (400 microliters/kg). Rifampin 180-190 glutamic pyruvic transaminase, soluble Mus musculus 28-31 8786635-2 1995 Serum alanine transaminase (ALT) and aspartate transaminase (AST) activities were not markedly altered and only hepatocellular fatty degeneration was found in mice pretreated with rifampicin (200 mg/kg), whereas severe centrilobular necrosis was observed and serum ALT and AST activities were as high as 281 and 271 I.U./l, respectively, in the control group following administration of CCl4 (400 microliters/kg). Rifampin 180-190 solute carrier family 17 (anion/sugar transporter), member 5 Mus musculus 61-64 8786635-2 1995 Serum alanine transaminase (ALT) and aspartate transaminase (AST) activities were not markedly altered and only hepatocellular fatty degeneration was found in mice pretreated with rifampicin (200 mg/kg), whereas severe centrilobular necrosis was observed and serum ALT and AST activities were as high as 281 and 271 I.U./l, respectively, in the control group following administration of CCl4 (400 microliters/kg). Rifampin 180-190 glutamic pyruvic transaminase, soluble Mus musculus 265-268 8786635-2 1995 Serum alanine transaminase (ALT) and aspartate transaminase (AST) activities were not markedly altered and only hepatocellular fatty degeneration was found in mice pretreated with rifampicin (200 mg/kg), whereas severe centrilobular necrosis was observed and serum ALT and AST activities were as high as 281 and 271 I.U./l, respectively, in the control group following administration of CCl4 (400 microliters/kg). Rifampin 180-190 solute carrier family 17 (anion/sugar transporter), member 5 Mus musculus 273-276 8786635-2 1995 Serum alanine transaminase (ALT) and aspartate transaminase (AST) activities were not markedly altered and only hepatocellular fatty degeneration was found in mice pretreated with rifampicin (200 mg/kg), whereas severe centrilobular necrosis was observed and serum ALT and AST activities were as high as 281 and 271 I.U./l, respectively, in the control group following administration of CCl4 (400 microliters/kg). Rifampin 180-190 chemokine (C-C motif) ligand 4 Mus musculus 387-391 8786635-4 1995 CCl4-mediated malondialdehyde (MDA) formation was increased in rifampicin-treated liver microsomes, demonstrating that rifampicin was capable of increasing the NADPH-dependent metabolism of CCl4 catalyzed by P-450 2E1 to produce free radicals. Rifampin 63-73 chemokine (C-C motif) ligand 4 Mus musculus 0-4 8786635-4 1995 CCl4-mediated malondialdehyde (MDA) formation was increased in rifampicin-treated liver microsomes, demonstrating that rifampicin was capable of increasing the NADPH-dependent metabolism of CCl4 catalyzed by P-450 2E1 to produce free radicals. Rifampin 63-73 chemokine (C-C motif) ligand 4 Mus musculus 190-194 8786635-4 1995 CCl4-mediated malondialdehyde (MDA) formation was increased in rifampicin-treated liver microsomes, demonstrating that rifampicin was capable of increasing the NADPH-dependent metabolism of CCl4 catalyzed by P-450 2E1 to produce free radicals. Rifampin 119-129 chemokine (C-C motif) ligand 4 Mus musculus 0-4 8786635-4 1995 CCl4-mediated malondialdehyde (MDA) formation was increased in rifampicin-treated liver microsomes, demonstrating that rifampicin was capable of increasing the NADPH-dependent metabolism of CCl4 catalyzed by P-450 2E1 to produce free radicals. Rifampin 119-129 chemokine (C-C motif) ligand 4 Mus musculus 190-194 8786635-5 1995 However, MDA formation was obviously depressed by rifampicin at varying concentrations from 2 to 32 x 10(-6) M in an in vitro cytochrome P-450 (P-450) enzyme system. Rifampin 50-60 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 126-142 8786635-7 1995 Therefore, the protective effect of rifampicin against CCl4 hepatotoxicity appeared to result from the direct inhibition of lipid peroxidation generated by CCl4-derived free radicals. Rifampin 36-46 chemokine (C-C motif) ligand 4 Mus musculus 55-59 8786635-7 1995 Therefore, the protective effect of rifampicin against CCl4 hepatotoxicity appeared to result from the direct inhibition of lipid peroxidation generated by CCl4-derived free radicals. Rifampin 36-46 chemokine (C-C motif) ligand 4 Mus musculus 156-160 8591727-9 1995 Treatment of cells with inducers of CYP1A (beta-naphthoflavone) and CYP3A (rifampicin and phenobarbital) greatly increased the rate of production of this metabolite. Rifampin 75-85 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 68-73 7646562-4 1995 This activity was specifically increased four-fold upon addition of beta-naphthoflavone into culture medium but not by rifampicine or clofibrate and was related to a biosynthesis of UDP-glucuronosyltransferase 1*6 (UGT1*6). Rifampin 119-130 UDP glucuronosyltransferase family 1 member A6 Homo sapiens 182-213 7574728-7 1995 In all three cultures, the increases in CYP3A after treatment with ethanol were less than those observed after treatment with rifampicin, a highly effective inducer of CYP3A in human hepatocytes. Rifampin 126-136 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 168-173 7574728-10 1995 In this latter culture, the steady-state levels of CYP3A3/4 mRNA increased by 0.1 mM isopentanol and 1 microM rifampicin were similar. Rifampin 110-120 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 51-57 7557864-6 1995 When cells were treated with the CYP inducer alone, large increases in the expression of CYP1A1 and CYP1A2 by beta-naphthoflavone and of CYP3A4 by rifampicin were observed at messenger RNA (mRNA) and protein levels, by ribonuclease protection and immunoblotting, respectively. Rifampin 147-157 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 89-95 7557864-6 1995 When cells were treated with the CYP inducer alone, large increases in the expression of CYP1A1 and CYP1A2 by beta-naphthoflavone and of CYP3A4 by rifampicin were observed at messenger RNA (mRNA) and protein levels, by ribonuclease protection and immunoblotting, respectively. Rifampin 147-157 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 100-106 7557864-6 1995 When cells were treated with the CYP inducer alone, large increases in the expression of CYP1A1 and CYP1A2 by beta-naphthoflavone and of CYP3A4 by rifampicin were observed at messenger RNA (mRNA) and protein levels, by ribonuclease protection and immunoblotting, respectively. Rifampin 147-157 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 137-143 7557864-10 1995 In cells preinduced with beta-naphthoflavone or rifampicin, the decay with time of the levels of the CYP1A2 or CYP3A4 proteins, after the removal of the inducer, was not affected by cytokines. Rifampin 48-58 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 101-107 7557864-10 1995 In cells preinduced with beta-naphthoflavone or rifampicin, the decay with time of the levels of the CYP1A2 or CYP3A4 proteins, after the removal of the inducer, was not affected by cytokines. Rifampin 48-58 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 111-117 7763306-1 1995 Rifampicin, a semi-synthetic antibiotic used in the treatment of tuberculosis and belonging to the chemical class of rifamycins, was examined for its effect on anti-cancer drug accumulation and activity in multidrug resistant cells overexpressing P-glycoprotein (P-gp). Rifampin 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 247-261 7614688-7 1995 Treatment of cynomolgus monkeys with rifampicin induced hepatic cytochromes P450 related to human CYP3A4 and CYP2C9/10 without inducing CYP1A1 or CYP1A2. Rifampin 37-47 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 98-104 7614688-7 1995 Treatment of cynomolgus monkeys with rifampicin induced hepatic cytochromes P450 related to human CYP3A4 and CYP2C9/10 without inducing CYP1A1 or CYP1A2. Rifampin 37-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 7614688-12 1995 Since cytochromes P4503A and/or P4502C are constitutively expressed in cynomolgus monkey hepatic microsomes, and upon induction with rifampicin are associated with an increased metabolic activation of IQ but not MeIQx, it appears that CYP3A and/or CYP2C are the isoform(s) showing the selective substrate specificity in the metabolic activation of IQ over MeIQx. Rifampin 133-143 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 235-240 7614688-12 1995 Since cytochromes P4503A and/or P4502C are constitutively expressed in cynomolgus monkey hepatic microsomes, and upon induction with rifampicin are associated with an increased metabolic activation of IQ but not MeIQx, it appears that CYP3A and/or CYP2C are the isoform(s) showing the selective substrate specificity in the metabolic activation of IQ over MeIQx. Rifampin 133-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 248-253 7636727-2 1995 We report the effect of a known cytochrome P450 (CYP) inducer, rifampicin, on the metabolism of lidocaine by primary human hepatocytes. Rifampin 63-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 7636727-2 1995 We report the effect of a known cytochrome P450 (CYP) inducer, rifampicin, on the metabolism of lidocaine by primary human hepatocytes. Rifampin 63-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 7636727-3 1995 Rifampicin has been shown to induce CYP3A4, a major human hepatic CYP isozyme that is known to metabolize lidocaine to its primary metabolite, monoethylglycinexylidide. Rifampin 0-10 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 36-42 7636727-3 1995 Rifampicin has been shown to induce CYP3A4, a major human hepatic CYP isozyme that is known to metabolize lidocaine to its primary metabolite, monoethylglycinexylidide. Rifampin 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 7745704-4 1995 p32 was associated with vaccinia virus membranes at all stages of virion assembly, starting with the viral crescents, as well as with the membranes which accumulated during the inhibition of assembly by rifampin. Rifampin 203-211 complement C1q binding protein Homo sapiens 0-3 7548905-4 1995 RESULTS: With therapy of TB (with isoniazid [INH], rifampin [RIF], ethambutol and/or pyrazinamide), TBG increased above control values and T3RU decreased (P < 0.001). Rifampin 51-59 serpin family A member 7 Homo sapiens 100-103 7548905-4 1995 RESULTS: With therapy of TB (with isoniazid [INH], rifampin [RIF], ethambutol and/or pyrazinamide), TBG increased above control values and T3RU decreased (P < 0.001). Rifampin 61-64 serpin family A member 7 Homo sapiens 100-103 7763306-1 1995 Rifampicin, a semi-synthetic antibiotic used in the treatment of tuberculosis and belonging to the chemical class of rifamycins, was examined for its effect on anti-cancer drug accumulation and activity in multidrug resistant cells overexpressing P-glycoprotein (P-gp). Rifampin 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 263-267 7763306-6 1995 Rifampicin was also demonstrated to inhibit P-gp radiolabeling by the photoactivable P-gp ligand azidopine, thereby suggesting that the antituberculosis compound can interfere directly with P-gp drug binding sites. Rifampin 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 44-48 7763306-6 1995 Rifampicin was also demonstrated to inhibit P-gp radiolabeling by the photoactivable P-gp ligand azidopine, thereby suggesting that the antituberculosis compound can interfere directly with P-gp drug binding sites. Rifampin 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 85-89 7763306-6 1995 Rifampicin was also demonstrated to inhibit P-gp radiolabeling by the photoactivable P-gp ligand azidopine, thereby suggesting that the antituberculosis compound can interfere directly with P-gp drug binding sites. Rifampin 0-10 ATP binding cassette subfamily B member 1 Homo sapiens 85-89 7763306-7 1995 These results thus indicate that rifampicin was able to down-modulate P-gp-associated resistance through inhibition of P-gp function. Rifampin 33-43 ATP binding cassette subfamily B member 1 Homo sapiens 70-74 7763306-7 1995 These results thus indicate that rifampicin was able to down-modulate P-gp-associated resistance through inhibition of P-gp function. Rifampin 33-43 ATP binding cassette subfamily B member 1 Homo sapiens 119-123 8068867-5 1994 An increase to 72 +/- 25 mL min-1 in the CLH of cortisol to 6 beta-hydroxycortisol was calculated following rifampicin treatment. Rifampin 108-118 CD59 molecule (CD59 blood group) Homo sapiens 28-33 7628309-1 1995 We previously demonstrated that induction of hepatic cytochrome P4503A (CYP3A) immunoreactive protein is a response in rats, but not rabbits, treated with the antiglucocorticoid, pregnenolone 16 alpha-carbonitrile and in rabbits, but not rats, treated with rifampicin. Rifampin 257-267 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 53-59 7628309-1 1995 We previously demonstrated that induction of hepatic cytochrome P4503A (CYP3A) immunoreactive protein is a response in rats, but not rabbits, treated with the antiglucocorticoid, pregnenolone 16 alpha-carbonitrile and in rabbits, but not rats, treated with rifampicin. Rifampin 257-267 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 72-77 7628309-4 1995 As expected from their effects in vivo, incubations of cultures with medium containing pregnenolone 16 alpha-carbonitrile or rifampicin induced CYP3A mRNA to high levels exclusively in rat hepatocytes or rabbit hepatocytes, respectively. Rifampin 125-135 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 144-149 7628309-5 1995 Pregnenolone 16 alpha-carbonitrile treatment also did not induce CYP3A immunoreactive protein in rabbit hepatocytes, although rifampicin treatment did increase CYP3A immunoreactive protein levels in rat hepatocyte cultures. Rifampin 126-136 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 160-165 8844805-8 1995 In addition, Rifampicin-induced rabbit liver microsomes which are enriched for CYP3A6, show enhanced activation of MeIQ in the presence of alpha-napthoflavone. Rifampin 13-23 cytochrome P450 3A6 Oryctolagus cuniculus 79-85 7853151-1 1994 The suppositories of rifampicin (RFP) containing sodium para-aminosalicylate dihydrate (PAS-Na) were prepared in order to enhance the rectal absorption of RFP. Rifampin 21-31 calcium voltage-gated channel subunit alpha1 D Homo sapiens 88-94 8169844-5 1994 Omeprazole, omeprazole sulfone and lansoprazole induced CYP3A in approximately 50% of tested cultures, whereas 100% of tested cultures responded to omeprazole and to rifampicin in terms of CYP1A and CYP3A induction, respectively. Rifampin 166-176 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 199-204 8347575-2 1993 Addition of rifampicin to inhibit the E. coli RNA polymerase after induction of the T7 RNA polymerase gene resulted in about 30% of newly synthesized protein being alpha 1-antitrypsin. Rifampin 12-22 serpin family A member 1 Homo sapiens 164-183 8049635-2 1994 Debrisoquine metabolism is controlled by a cytochrome P-450 isozyme encoded at the CYP2D6 locus, which is inducible by antipyrine and rifampicin. Rifampin 134-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 8049635-2 1994 Debrisoquine metabolism is controlled by a cytochrome P-450 isozyme encoded at the CYP2D6 locus, which is inducible by antipyrine and rifampicin. Rifampin 134-144 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 83-89 8126890-3 1994 Antibiotics, such as macrolides, new quinolones and rifampicin, are very active against Legionella spp. Rifampin 52-62 histocompatibility minor 13 Homo sapiens 99-102 8119047-8 1993 By contrast, rifampicin (rifampin) and rifabutin induce several cytochromes P450, including CYP3A4, and hence can enhance the metabolism of many other drugs. Rifampin 13-23 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 92-98 8119047-8 1993 By contrast, rifampicin (rifampin) and rifabutin induce several cytochromes P450, including CYP3A4, and hence can enhance the metabolism of many other drugs. Rifampin 25-33 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 92-98 8091338-11 1993 Some P450 (3A, 1A, 2E, ...) are inducible by compounds such as phenobarbital, rifampicin, aromatic hydrocarbon, ethanol, or omeprazole. Rifampin 78-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 8325897-12 1993 However, when neonatal rabbits were treated with either dexamethasone or rifampicin, UGT2B13 mRNA levels were induced. Rifampin 73-83 UDP-glucuronosyltransferase 2B13 Oryctolagus cuniculus 85-92 8161275-2 1993 The rifampicin MIC with respect to 50 strains of the plague microbe of different origin in the tests on the Hottinger agar ranged from 1.6 to 6.4 micrograms/ml. Rifampin 4-14 microphthalmia Japan Mus musculus 15-18 1460703-1 1992 The objective of the present study was to assess renal damage, if any, by non-invasive technique, viz NAG activity in urine and GFR in patients on continuous and intermittent rifampicin therapy. Rifampin 175-185 N-acetyl-alpha-glucosaminidase Homo sapiens 102-105 8335500-1 1993 Rifampicin is used to treat neurosurgical shunt infections because of its excellent in-vitro activity against staphylococci and its adequate penetration into the CSF. Rifampin 0-10 colony stimulating factor 2 Rattus norvegicus 162-165 8503179-5 1993 Temperature-sensitive mutants C6, C17, and C43 were found to be rifampicin hypersensitive. Rifampin 64-74 cytokine like 1 Homo sapiens 34-37 8503179-9 1993 Further, the finding that the hypersensitive mutants C6 and C17 contain a wild-type D13L ORF suggests a possible role of other viral functions in the interaction of rifampicin with vaccinia virus. Rifampin 165-175 cytokine like 1 Homo sapiens 60-63 8508626-0 1993 Increased insulin requirement in a patient with type 1 diabetes on rifampicin. Rifampin 67-77 insulin Homo sapiens 10-17 1430211-0 1992 Identification of rifampin-inducible P450IIIA4 (CYP3A4) in human small bowel enterocytes. Rifampin 18-26 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-54 1430211-6 1992 Rifampin treatment resulted in a five- or eightfold mean increase (P < 0.05) in the biopsy concentration of P450IIIA4 mRNA when normalized for content of sucrase isomaltase or intestinal fatty acid binding protein mRNAs, respectively. Rifampin 0-8 sucrase-isomaltase Homo sapiens 157-175 1546459-1 1992 Specific missense mutations of the vaccinia virus D13L gene confer resistance to the effects of rifampicin on virion morphogenesis. Rifampin 96-106 rifampicin target Vaccinia virus 50-54 1454750-1 1992 A previous study has demonstrated that the urinary level of 6 beta-hydroxycortisol is a marker of liver CYP3A content after induction by rifampicin. Rifampin 137-147 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-109 1418078-0 1992 Fibrinogen and antithrombin III blood levels fluctuations during isoniazid or isoniazid plus rifampicin administration. Rifampin 93-103 fibrinogen beta chain Homo sapiens 0-10 1418078-0 1992 Fibrinogen and antithrombin III blood levels fluctuations during isoniazid or isoniazid plus rifampicin administration. Rifampin 93-103 serpin family C member 1 Homo sapiens 15-31 1495829-3 1992 EF4 bacteria were susceptible to aminopenicillins, carboxypenicillins, ureidopenicillins, third-generation cephalosporins, fluoroquinolones, rifampicin, and trimethoprime-sulfamethoxazole. Rifampin 141-151 GTP binding elongation factor GUF1 Homo sapiens 0-3 1768567-8 1991 In contrast, rifampicin treatment produced a larger and a differential increase in the oral clearance of R-(-) hexobarbitone in young and elderly subjects; an 89 fold change in the young (15.6 +/- 16.4 to 1146.7 +/- 1478.0 ml min-1 kg-1) and a 19 fold change (10.3 +/- 3.0 to 199.9 +/- 98.1 ml min-1 kg-1) in the elderly. Rifampin 13-23 CD59 molecule (CD59 blood group) Homo sapiens 226-236 1768567-8 1991 In contrast, rifampicin treatment produced a larger and a differential increase in the oral clearance of R-(-) hexobarbitone in young and elderly subjects; an 89 fold change in the young (15.6 +/- 16.4 to 1146.7 +/- 1478.0 ml min-1 kg-1) and a 19 fold change (10.3 +/- 3.0 to 199.9 +/- 98.1 ml min-1 kg-1) in the elderly. Rifampin 13-23 CD59 molecule (CD59 blood group) Homo sapiens 294-304