PMID-sentid Pub_year Sent_text comp_official_name comp_offset protein_name organism prot_offset 8461288-2 1993 With the exception of chlorophyll b, which is known to be associated with the complexes comprising the outer antenna (LHCII, CP24, CP26, CP29), the spectral forms occur with similar absorption maxima and are present in rather similar amounts in each of the antenna complexes. chlorophyll b 22-35 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 131-135 33233443-0 2020 CYP26A1 Is a Novel Biomarker for Betel Quid-Related Oral and Pharyngeal Cancers. betel 33-38 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 1864384-3 1991 Based on the assumption that there is 1 cytochrome b559 per reaction centre it has been found that oxygen-evolving complexes containing CP26 and CP29 bind 42 chlorophyll molecules. Oxygen 99-105 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 136-140 1864384-3 1991 Based on the assumption that there is 1 cytochrome b559 per reaction centre it has been found that oxygen-evolving complexes containing CP26 and CP29 bind 42 chlorophyll molecules. Chlorophyll 158-169 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 136-140 1864384-4 1991 When CP26 and CP29 are stripped away, the resulting PSII cores bind 30 chlorophyll molecules while CP43-less cores bind approximately 18 chlorophylls. Chlorophyll 71-82 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 5-9 1864384-4 1991 When CP26 and CP29 are stripped away, the resulting PSII cores bind 30 chlorophyll molecules while CP43-less cores bind approximately 18 chlorophylls. cp43 99-103 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 5-9 33233443-9 2020 We found that CYP26A1 was downregulated as the arecoline dose increased. Arecoline 47-56 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 14-21 33233443-11 2020 Arecoline appears to modulate CYP26A1 expression through specific pathways. Arecoline 0-9 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 30-37 34389675-5 2021 There were two unexpected findings: 1) a strong induction of CYPs involved in activation of fatty acids (CYP4), and in inactivation of calcitriol (CYP24A1) and retinoic acid (CYP26A1); and 2) a marked down-regulation of FOS, FRA1, and JUN, known tethering partners of ERbeta. Calcitriol 135-145 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 175-182 19525031-0 2009 Retinoic acid 4-hydroxylase inducibility and clinical response to isotretinoin in patients with acne. Isotretinoin 66-78 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-27 19525031-2 2009 The CYP26 enzyme acts specifically on tRA, but not 13-cis RA (isotretinoin), a retinoid used to treat severe acne. Isotretinoin 62-74 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-9 19525031-2 2009 The CYP26 enzyme acts specifically on tRA, but not 13-cis RA (isotretinoin), a retinoid used to treat severe acne. Retinoids 79-87 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-9 19525031-3 2009 However, 13-cis RA can isomerize to tRA, which can then be metabolized by CYP26. Tretinoin 16-18 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 74-79 19525031-5 2009 We then investigated whether response to oral 13-cis RA among patients with acne correlates with variability in CYP26 expression. Tretinoin 53-55 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 112-117 34389675-5 2021 There were two unexpected findings: 1) a strong induction of CYPs involved in activation of fatty acids (CYP4), and in inactivation of calcitriol (CYP24A1) and retinoic acid (CYP26A1); and 2) a marked down-regulation of FOS, FRA1, and JUN, known tethering partners of ERbeta. Tretinoin 160-173 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 175-182 33472949-3 2021 Here we found that experimental YAP activation in 5FU-sensitive and 5FU-resistant HT29 CRC cells enhanced nuclear YAP localization and the transcript levels of the retinoic acid (RA) receptors RARalpha/gamma and RAR target genes CYP26A1, ALDH1A3 and LGR5 through RA Response Elements (RAREs). Tretinoin 164-177 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 229-236 35565751-0 2022 CRABPs Alter all-trans-Retinoic Acid Metabolism by CYP26A1 via Protein-Protein Interactions. Tretinoin 13-36 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 51-58 35565751-3 2022 We hypothesized that CRABP1 and CRABP2 also alter atRA metabolism and clearance by CYP26A1, the third key atRA-metabolizing enzyme in the CYP26 family. Tretinoin 50-54 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 83-90 35565751-3 2022 We hypothesized that CRABP1 and CRABP2 also alter atRA metabolism and clearance by CYP26A1, the third key atRA-metabolizing enzyme in the CYP26 family. Tretinoin 106-110 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 83-90 35565751-5 2022 The unbound atRA Km values for 4-OH-atRA formation by CYP26A1 were 4.7 +- 0.8 nM with atRA, 6.8 +- 1.7 nM with holo-CRABP1 and 6.1 +- 2.7 nM with holo-CRABP2 as a substrate. 4-oh-atra 31-40 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 54-61 35565751-5 2022 The unbound atRA Km values for 4-OH-atRA formation by CYP26A1 were 4.7 +- 0.8 nM with atRA, 6.8 +- 1.7 nM with holo-CRABP1 and 6.1 +- 2.7 nM with holo-CRABP2 as a substrate. Tretinoin 86-90 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 54-61 33909340-2 2021 BRIEF METHODS: In the present study we have tested human CYPs for metabolization of five proluciferin ester substrates which had previously only been known to be hydroxylated by CYP26A1. proluciferin ester 89-107 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 178-185 33724958-5 2021 Specifically, high expression of the ATRA-catabolizing enzyme, CYP26A1, in human IDCM could dampen prospects for an ATRA-based therapy. Tretinoin 37-41 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 63-70 33724958-5 2021 Specifically, high expression of the ATRA-catabolizing enzyme, CYP26A1, in human IDCM could dampen prospects for an ATRA-based therapy. Tretinoin 116-120 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 63-70 33472949-3 2021 Here we found that experimental YAP activation in 5FU-sensitive and 5FU-resistant HT29 CRC cells enhanced nuclear YAP localization and the transcript levels of the retinoic acid (RA) receptors RARalpha/gamma and RAR target genes CYP26A1, ALDH1A3 and LGR5 through RA Response Elements (RAREs). Fluorouracil 50-53 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 229-236 33472949-3 2021 Here we found that experimental YAP activation in 5FU-sensitive and 5FU-resistant HT29 CRC cells enhanced nuclear YAP localization and the transcript levels of the retinoic acid (RA) receptors RARalpha/gamma and RAR target genes CYP26A1, ALDH1A3 and LGR5 through RA Response Elements (RAREs). Fluorouracil 68-71 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 229-236 34122632-4 2021 The results of cell proliferation assay and reverse transcription-PCR demonstrated that ATRA may exert synergistic effects with the SphK1 inhibitor SKI 5C or the pan-SphK inhibitor SKI II to inhibit the proliferation of K562 cells and upregulate the expression levels of the ATRA-inducible enzyme cytochrome P450 26A1 (CYP26A1). Tretinoin 88-92 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 297-317 34122632-4 2021 The results of cell proliferation assay and reverse transcription-PCR demonstrated that ATRA may exert synergistic effects with the SphK1 inhibitor SKI 5C or the pan-SphK inhibitor SKI II to inhibit the proliferation of K562 cells and upregulate the expression levels of the ATRA-inducible enzyme cytochrome P450 26A1 (CYP26A1). Tretinoin 88-92 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 319-326 34122632-4 2021 The results of cell proliferation assay and reverse transcription-PCR demonstrated that ATRA may exert synergistic effects with the SphK1 inhibitor SKI 5C or the pan-SphK inhibitor SKI II to inhibit the proliferation of K562 cells and upregulate the expression levels of the ATRA-inducible enzyme cytochrome P450 26A1 (CYP26A1). Tretinoin 275-279 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 297-317 34122632-4 2021 The results of cell proliferation assay and reverse transcription-PCR demonstrated that ATRA may exert synergistic effects with the SphK1 inhibitor SKI 5C or the pan-SphK inhibitor SKI II to inhibit the proliferation of K562 cells and upregulate the expression levels of the ATRA-inducible enzyme cytochrome P450 26A1 (CYP26A1). Tretinoin 275-279 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 319-326 35565751-7 2022 In addition, increasing concentrations in apo-CRABPs decreased the 4-OH-atRA formation rates by CYP26A1. 4-oh-atra 67-76 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 96-103 33472949-3 2021 Here we found that experimental YAP activation in 5FU-sensitive and 5FU-resistant HT29 CRC cells enhanced nuclear YAP localization and the transcript levels of the retinoic acid (RA) receptors RARalpha/gamma and RAR target genes CYP26A1, ALDH1A3 and LGR5 through RA Response Elements (RAREs). Tretinoin 179-181 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 229-236 32151018-0 2020 Regulating Retinoic Acid Availability during Development and Regeneration: The Role of the CYP26 Enzymes. Tretinoin 11-24 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 91-96 31903789-3 2020 In vivo, retinoids induce CYP26 enzyme production in the liver, enhancing their own rapid metabolic clearance, while retinoid resistance in tumour cells themselves is considered to be due in part to increased CYP26 production. Retinoids 9-18 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 26-31 31903789-3 2020 In vivo, retinoids induce CYP26 enzyme production in the liver, enhancing their own rapid metabolic clearance, while retinoid resistance in tumour cells themselves is considered to be due in part to increased CYP26 production. Retinoids 9-17 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 26-31 31903789-4 2020 Retinoic acid metabolism blocking agents (RAMBAs), which inhibit CYP26 enzymes, can improve retinoic acid (RA) pharmacokinetics in pre-clinical neuroblastoma models. Tretinoin 0-13 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 65-70 31903789-4 2020 Retinoic acid metabolism blocking agents (RAMBAs), which inhibit CYP26 enzymes, can improve retinoic acid (RA) pharmacokinetics in pre-clinical neuroblastoma models. Tretinoin 92-105 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 65-70 31903789-4 2020 Retinoic acid metabolism blocking agents (RAMBAs), which inhibit CYP26 enzymes, can improve retinoic acid (RA) pharmacokinetics in pre-clinical neuroblastoma models. Tretinoin 42-44 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 65-70 32251704-2 2020 The proposed MIE for azole fungicides is CYP26 inhibition with retinoic acid (RA) local increase, triggering key events leading to craniofacial defects. Azoles 21-26 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 41-46 32251704-2 2020 The proposed MIE for azole fungicides is CYP26 inhibition with retinoic acid (RA) local increase, triggering key events leading to craniofacial defects. Tretinoin 63-76 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 41-46 32251704-2 2020 The proposed MIE for azole fungicides is CYP26 inhibition with retinoic acid (RA) local increase, triggering key events leading to craniofacial defects. Tretinoin 78-80 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 41-46 32251704-9 2020 In silico results confirmed azole-related CYP26 inhibition and a weak general VPA inhibition on the tested HDACs. Azoles 28-33 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 42-47 32251704-10 2020 Unexpectedly, VPA showed also a weak, but not marginal, capability to enter the CYP 26A1 and CYP 26C1 catalytic sites, suggesting a possible role of VPA in decreasing RA catabolism, acting as an additional MIE. Valproic Acid 14-17 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 80-88 32251704-10 2020 Unexpectedly, VPA showed also a weak, but not marginal, capability to enter the CYP 26A1 and CYP 26C1 catalytic sites, suggesting a possible role of VPA in decreasing RA catabolism, acting as an additional MIE. Valproic Acid 149-152 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 80-88 32251704-10 2020 Unexpectedly, VPA showed also a weak, but not marginal, capability to enter the CYP 26A1 and CYP 26C1 catalytic sites, suggesting a possible role of VPA in decreasing RA catabolism, acting as an additional MIE. Tretinoin 167-169 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 80-88 32251704-13 2020 VPA MIEs (HDAC and CYP26 inhibition) impinge on the two converging AOPs. Valproic Acid 0-3 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 19-24 33294638-0 2020 New luciferin-based probe substrates for human CYP26A1. D-luciferin 4-13 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 47-54 33294638-1 2020 Activity of human CYP26A1 towards six proluciferin probe substrates and their ester derivatives was monitored. proluciferin 38-50 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 18-25 33294638-1 2020 Activity of human CYP26A1 towards six proluciferin probe substrates and their ester derivatives was monitored. Esters 78-83 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 18-25 33294638-6 2020 Taken together, we describe eleven new probe substrates for CYP26A1 and demonstrate for the first time that CYP26A1 does not only accept acid substrates but can also metabolize esters. Esters 177-183 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 60-67 33294638-6 2020 Taken together, we describe eleven new probe substrates for CYP26A1 and demonstrate for the first time that CYP26A1 does not only accept acid substrates but can also metabolize esters. Esters 177-183 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 108-115 32258025-1 2020 Organogenesis, including renal development, requires an appropriate retinoic acid concentration, which is established by differential expression of aldehyde dehydrogenase 1 family member A2 (ALDH1A2) and cytochrome P450 family 26 subfamily A/B/C member 1 (CYP26A1/B1/C1). Tretinoin 68-81 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 256-263 32151018-1 2020 This review focuses on the role of the Cytochrome p450 subfamily 26 (CYP26) retinoic acid (RA) degrading enzymes during development and regeneration. Tretinoin 76-89 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 39-67 32151018-1 2020 This review focuses on the role of the Cytochrome p450 subfamily 26 (CYP26) retinoic acid (RA) degrading enzymes during development and regeneration. Tretinoin 76-89 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 69-74 32151018-1 2020 This review focuses on the role of the Cytochrome p450 subfamily 26 (CYP26) retinoic acid (RA) degrading enzymes during development and regeneration. Tretinoin 91-93 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 39-67 32151018-1 2020 This review focuses on the role of the Cytochrome p450 subfamily 26 (CYP26) retinoic acid (RA) degrading enzymes during development and regeneration. Tretinoin 91-93 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 69-74 32151018-2 2020 Cyp26 enzymes, along with retinoic acid synthesising enzymes, are absolutely required for RA homeostasis in these processes by regulating availability of RA for receptor binding and signalling. Tretinoin 26-39 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 32151018-2 2020 Cyp26 enzymes, along with retinoic acid synthesising enzymes, are absolutely required for RA homeostasis in these processes by regulating availability of RA for receptor binding and signalling. Tretinoin 90-92 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 32151018-2 2020 Cyp26 enzymes, along with retinoic acid synthesising enzymes, are absolutely required for RA homeostasis in these processes by regulating availability of RA for receptor binding and signalling. Tretinoin 154-156 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 32151018-3 2020 Cyp26 enzymes are necessary to generate RA gradients and to protect specific tissues from RA signalling. Tretinoin 40-42 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 32151018-3 2020 Cyp26 enzymes are necessary to generate RA gradients and to protect specific tissues from RA signalling. Tretinoin 90-92 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 32151018-5 2020 Here, the function of CYP26 enzymes is discussed in the context of the RA signalling pathway, enzymatic structure and biochemistry, human genetic disease, and function in development and regeneration as elucidated from animal model studies. Tretinoin 71-73 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 22-27 32359660-0 2020 Using the human CYP26A1 gene promoter as a suitable tool for the determination of RAR-mediated retinoid activity. Retinoids 95-103 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 16-23 31419517-2 2019 The three CYP26 family enzymes, CYP26A1, CYP26B1 and CYP26C1 have all been shown to metabolize all-trans-retinoic acid (atRA) it"s 9-cisRA and 13-cisRA isomers and primary metabolites 4-OH-RA and 4-oxo-RA with high efficiency. Tretinoin 99-118 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 10-15 31744925-0 2020 Correction to "Induction of CYP26A1 by Metabolites of Retinoic Acid: Evidence That CYP26A1 Is an Important Enzyme in the Elimination of Active Retinoids". Tretinoin 54-67 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 28-35 31744925-0 2020 Correction to "Induction of CYP26A1 by Metabolites of Retinoic Acid: Evidence That CYP26A1 Is an Important Enzyme in the Elimination of Active Retinoids". Tretinoin 54-67 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 83-90 31744925-0 2020 Correction to "Induction of CYP26A1 by Metabolites of Retinoic Acid: Evidence That CYP26A1 Is an Important Enzyme in the Elimination of Active Retinoids". Retinoids 143-152 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 28-35 31744925-0 2020 Correction to "Induction of CYP26A1 by Metabolites of Retinoic Acid: Evidence That CYP26A1 Is an Important Enzyme in the Elimination of Active Retinoids". Retinoids 143-152 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 83-90 31419517-2 2019 The three CYP26 family enzymes, CYP26A1, CYP26B1 and CYP26C1 have all been shown to metabolize all-trans-retinoic acid (atRA) it"s 9-cisRA and 13-cisRA isomers and primary metabolites 4-OH-RA and 4-oxo-RA with high efficiency. Tretinoin 99-118 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 32-39 31419517-2 2019 The three CYP26 family enzymes, CYP26A1, CYP26B1 and CYP26C1 have all been shown to metabolize all-trans-retinoic acid (atRA) it"s 9-cisRA and 13-cisRA isomers and primary metabolites 4-OH-RA and 4-oxo-RA with high efficiency. Tretinoin 120-124 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 10-15 31419517-2 2019 The three CYP26 family enzymes, CYP26A1, CYP26B1 and CYP26C1 have all been shown to metabolize all-trans-retinoic acid (atRA) it"s 9-cisRA and 13-cisRA isomers and primary metabolites 4-OH-RA and 4-oxo-RA with high efficiency. Tretinoin 120-124 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 32-39 31419517-2 2019 The three CYP26 family enzymes, CYP26A1, CYP26B1 and CYP26C1 have all been shown to metabolize all-trans-retinoic acid (atRA) it"s 9-cisRA and 13-cisRA isomers and primary metabolites 4-OH-RA and 4-oxo-RA with high efficiency. Isotretinoin 133-138 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 10-15 31419517-2 2019 The three CYP26 family enzymes, CYP26A1, CYP26B1 and CYP26C1 have all been shown to metabolize all-trans-retinoic acid (atRA) it"s 9-cisRA and 13-cisRA isomers and primary metabolites 4-OH-RA and 4-oxo-RA with high efficiency. Isotretinoin 133-138 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 32-39 31419517-2 2019 The three CYP26 family enzymes, CYP26A1, CYP26B1 and CYP26C1 have all been shown to metabolize all-trans-retinoic acid (atRA) it"s 9-cisRA and 13-cisRA isomers and primary metabolites 4-OH-RA and 4-oxo-RA with high efficiency. Isotretinoin 146-151 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 10-15 31419517-2 2019 The three CYP26 family enzymes, CYP26A1, CYP26B1 and CYP26C1 have all been shown to metabolize all-trans-retinoic acid (atRA) it"s 9-cisRA and 13-cisRA isomers and primary metabolites 4-OH-RA and 4-oxo-RA with high efficiency. Isotretinoin 146-151 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 32-39 31419517-2 2019 The three CYP26 family enzymes, CYP26A1, CYP26B1 and CYP26C1 have all been shown to metabolize all-trans-retinoic acid (atRA) it"s 9-cisRA and 13-cisRA isomers and primary metabolites 4-OH-RA and 4-oxo-RA with high efficiency. 4-oh-ra 184-191 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 10-15 31419517-2 2019 The three CYP26 family enzymes, CYP26A1, CYP26B1 and CYP26C1 have all been shown to metabolize all-trans-retinoic acid (atRA) it"s 9-cisRA and 13-cisRA isomers and primary metabolites 4-OH-RA and 4-oxo-RA with high efficiency. 4-oh-ra 184-191 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 32-39 31419517-2 2019 The three CYP26 family enzymes, CYP26A1, CYP26B1 and CYP26C1 have all been shown to metabolize all-trans-retinoic acid (atRA) it"s 9-cisRA and 13-cisRA isomers and primary metabolites 4-OH-RA and 4-oxo-RA with high efficiency. 4-oxoretinoic acid 196-204 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 10-15 31419517-2 2019 The three CYP26 family enzymes, CYP26A1, CYP26B1 and CYP26C1 have all been shown to metabolize all-trans-retinoic acid (atRA) it"s 9-cisRA and 13-cisRA isomers and primary metabolites 4-OH-RA and 4-oxo-RA with high efficiency. 4-oxoretinoic acid 196-204 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 32-39 31419517-4 2019 All three CYP26 enzymes are inducible by treatment with atRA in various prenatal and postnatal tissues and cell types. Tretinoin 56-60 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 10-15 31419517-6 2019 In humans and in animal models the expression patterns of CYP26 enzymes have been shown to be tissue and cell type specific, and the expression of the CYP26 enzymes is believed to regulate the formation of critical atRA concentration gradients in various tissue types. Tretinoin 215-219 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 58-63 31419517-6 2019 In humans and in animal models the expression patterns of CYP26 enzymes have been shown to be tissue and cell type specific, and the expression of the CYP26 enzymes is believed to regulate the formation of critical atRA concentration gradients in various tissue types. Tretinoin 215-219 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 151-156 31419517-9 2019 Interestingly, some of these genetic variants result in increased activity of the CYP26 enzymes potentially leading to complex gene-environment interactions due to variability in dietary intake of retinoids. Retinoids 197-206 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 82-87 31419517-10 2019 This review highlights the current knowledge of structure-function of CYP26 enzymes and focuses on their role in human retinoid metabolism in different tissues. Retinoids 119-127 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 70-75 31039331-0 2019 CYP26A1 gene promoter is a useful tool for reporting RAR-mediated retinoid activity. Retinoids 66-74 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 31283017-6 2019 We found increases in mRNAs of differentiation-associated genes (Hoxa1, Cyp26a1, and RARbeta2) upon EtOH treatment of WT and Acss2-/- ESCs, but not Aldh2-/- ESCs. Ethanol 100-104 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 72-79 31039331-1 2019 Of numerous genes regulated by retinoic acid (RA), CYP26A1 is the most inducible gene by RA. Tretinoin 31-44 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 51-58 31039331-2 2019 In this study, we have used a shortened construct form, E4, of the CYP26A1 gene promoter, in a promoter-less vector with either luciferase or red fluorescent protein (RFP) as the reporter gene and have tested its responses to retinoids in transfected HepG2 and HEK293T cells. Retinoids 226-235 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 67-74 30250298-5 2019 Moreover, we demonstrated that pharmacological inhibition of 15-PGDH enhanced CYP26A1 expression, leading to depletion of all-trans retinoic acid (ATRA) and expansion of the ALDH1-positive subset in both human PDAC cells and tumor cells of KrasLSL-G12D/+; Ptf1aCre/+ (KC) mice. Tretinoin 147-151 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 78-85 30885956-4 2019 In this study, we demonstrate that enhancing RA degradation in the host and to a lesser extent donor hematopoietic cells by overexpressing the RA-catabolizing enzyme CYP26A1 reduced GVHD. Tretinoin 45-47 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 166-173 30885956-4 2019 In this study, we demonstrate that enhancing RA degradation in the host and to a lesser extent donor hematopoietic cells by overexpressing the RA-catabolizing enzyme CYP26A1 reduced GVHD. Tretinoin 143-145 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 166-173 30737277-6 2019 As shown with CRISPR/Cas9 knockout lines, the retinol dehydrogenase gene Rdh10 and a functional RARE in the ROL transporter stimulated by retinoic acid 6 (Stra6) gene are required for EtOH induction of Hoxa1 and Cyp26a1 We conclude that EtOH stimulates stem cell differentiation by increasing the influx and metabolism of ROL for downstream RARgamma-dependent transcription. Ethanol 184-188 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 212-219 30250298-5 2019 Moreover, we demonstrated that pharmacological inhibition of 15-PGDH enhanced CYP26A1 expression, leading to depletion of all-trans retinoic acid (ATRA) and expansion of the ALDH1-positive subset in both human PDAC cells and tumor cells of KrasLSL-G12D/+; Ptf1aCre/+ (KC) mice. Tretinoin 132-145 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 78-85 31288815-12 2019 Our studies demonstrated that five PRGs including Bcl2, FOXO1A, SCGB2A2, CYP26a1 and MMP11 exhibited significant progesterone-hyper-responsiveness in human PrMyoF cells as compared to PrMyoN cells (P < 0.05). Progesterone 113-125 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 73-80 30737277-5 2019 We also report that EtOH-mediated increases in homeobox A1 (Hoxa1) and cytochrome P450 family 26 subfamily A member 1 (Cyp26a1) transcripts, direct RA target genes, require the expression of the RA-synthesizing enzyme, aldehyde dehydrogenase 1 family member A2 (Aldh1a2), suggesting that EtOH-mediated induction of Hoxa1 and Cyp26a1 requires ROL from the serum. Ethanol 20-24 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 71-117 30737277-5 2019 We also report that EtOH-mediated increases in homeobox A1 (Hoxa1) and cytochrome P450 family 26 subfamily A member 1 (Cyp26a1) transcripts, direct RA target genes, require the expression of the RA-synthesizing enzyme, aldehyde dehydrogenase 1 family member A2 (Aldh1a2), suggesting that EtOH-mediated induction of Hoxa1 and Cyp26a1 requires ROL from the serum. Ethanol 20-24 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 119-126 30737277-5 2019 We also report that EtOH-mediated increases in homeobox A1 (Hoxa1) and cytochrome P450 family 26 subfamily A member 1 (Cyp26a1) transcripts, direct RA target genes, require the expression of the RA-synthesizing enzyme, aldehyde dehydrogenase 1 family member A2 (Aldh1a2), suggesting that EtOH-mediated induction of Hoxa1 and Cyp26a1 requires ROL from the serum. Ethanol 20-24 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 325-332 30737277-5 2019 We also report that EtOH-mediated increases in homeobox A1 (Hoxa1) and cytochrome P450 family 26 subfamily A member 1 (Cyp26a1) transcripts, direct RA target genes, require the expression of the RA-synthesizing enzyme, aldehyde dehydrogenase 1 family member A2 (Aldh1a2), suggesting that EtOH-mediated induction of Hoxa1 and Cyp26a1 requires ROL from the serum. Tretinoin 148-150 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 71-117 30737277-5 2019 We also report that EtOH-mediated increases in homeobox A1 (Hoxa1) and cytochrome P450 family 26 subfamily A member 1 (Cyp26a1) transcripts, direct RA target genes, require the expression of the RA-synthesizing enzyme, aldehyde dehydrogenase 1 family member A2 (Aldh1a2), suggesting that EtOH-mediated induction of Hoxa1 and Cyp26a1 requires ROL from the serum. Tretinoin 148-150 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 119-126 29984664-6 2018 RESULTS: RT-qPCR data analysis showed that after 12 h of exposure of HepG2/C3A cells to genistein (5 and 50 microM) there was an upregulation of CYP1A1 and CYP1B1 and downregulation of CYP2D6, CYP26A1 and CYP26B1 mRNA levels. Genistein 88-97 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 193-200 29337257-5 2018 Evaluating also specific gene responses related with the retinoic acid and sterol biosynthesis pathways, which represent the toxicological and fungicidal mode of action of azoles respectively in the WEC and EST, we observed that the differential regulation of Dhrs3 and Msmo1 reached higher magnitudes in both systems compared to Cyp26a1 and Cyp51. Tretinoin 57-70 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 330-337 29337257-5 2018 Evaluating also specific gene responses related with the retinoic acid and sterol biosynthesis pathways, which represent the toxicological and fungicidal mode of action of azoles respectively in the WEC and EST, we observed that the differential regulation of Dhrs3 and Msmo1 reached higher magnitudes in both systems compared to Cyp26a1 and Cyp51. Sterols 75-81 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 330-337 29337257-5 2018 Evaluating also specific gene responses related with the retinoic acid and sterol biosynthesis pathways, which represent the toxicological and fungicidal mode of action of azoles respectively in the WEC and EST, we observed that the differential regulation of Dhrs3 and Msmo1 reached higher magnitudes in both systems compared to Cyp26a1 and Cyp51. Azoles 172-178 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 330-337 29990529-4 2018 Trans-resveratrol also altered expression levels of developmental regulator genes involved in embryonic patterning, such as Wnt3a, Tbx6, and Cyp26a1. Resveratrol 0-17 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 141-148 29990529-7 2018 By contrast, a reduction in the DNA replication rate with aphidicolin (0.4 muM) or hydroxyurea (40 muM) created smaller and rounder EBs and altered the expression levels of Wnt3a, Tbx6, and Cyp26a1 in a manner similar to trans-resveratrol. Aphidicolin 58-69 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 190-197 29990529-7 2018 By contrast, a reduction in the DNA replication rate with aphidicolin (0.4 muM) or hydroxyurea (40 muM) created smaller and rounder EBs and altered the expression levels of Wnt3a, Tbx6, and Cyp26a1 in a manner similar to trans-resveratrol. Hydroxyurea 83-94 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 190-197 29476041-1 2018 The clearance of retinoic acid (RA) and its metabolites is believed to be regulated by the CYP26 enzymes, but the specific roles of CYP26A1, CYP26B1, and CYP26C1 in clearing active vitamin A metabolites have not been defined. Vitamin A 181-190 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 132-139 29330906-4 2018 The developing embryo contains regions that produce RA, and specific intracellular concentrations of RA are created through local degradation mediated by Cyp26 enzymes. Tretinoin 101-103 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 154-159 29177441-7 2018 These variants altered the expression of CYP26A1, a direct CDX2 target encoding the major retinoic acid (RA)-degrading enzyme. Tretinoin 90-103 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 41-48 29177441-7 2018 These variants altered the expression of CYP26A1, a direct CDX2 target encoding the major retinoic acid (RA)-degrading enzyme. Tretinoin 105-107 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 41-48 28754309-6 2017 Here, we discuss accumulating evidence suggesting that cytochrome P450 (CYP26), the primary retinoid-inactivating enzyme, plays a critical role in the integration of two of these molecular programs: the retinoid and Hedgehog pathways. Retinoids 92-100 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 72-77 28735443-8 2017 In situ hybridization analysis showed that RA is able to alter cyp26a1, sox9, tgfbeta2, and id2 spatial distribution; to increase rarbeta, meis2, and hoxb5 expression levels; and has a very modest effect on sox2, fgf10, fgfr2, and shh expression levels. Tretinoin 43-45 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 63-70 29066753-4 2017 Exploration of the orientational phase-space revealed that the placement of central carotenoids minimizes their interaction with the nearest chlorophylls in the plant antenna complexes LHCII, CP26, CP29 and LHCI. Carotenoids 84-95 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 192-196 28754309-6 2017 Here, we discuss accumulating evidence suggesting that cytochrome P450 (CYP26), the primary retinoid-inactivating enzyme, plays a critical role in the integration of two of these molecular programs: the retinoid and Hedgehog pathways. Retinoids 203-211 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 72-77 28754309-7 2017 Induction of stromal CYP26 by either one of these pathways limits retinoic acid concentration in the stem cell niche, with profound effects on tissue homeostasis and drug resistance. Tretinoin 66-79 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 21-26 28087752-6 2017 Localisation of RA synthetic (RALDH-1) and degrading (cytochrome P450 subfamily 26 A1 (CYP26A1)) enzymes in human lung was determined by immunofluorescence. Tretinoin 16-18 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 54-85 28643469-5 2017 In antler chondrocytes, CYP26A1 and CYP26B1 weakened the sensitivity of ATRA to COL9A1. Tretinoin 72-76 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 24-31 28087752-13 2017 CONCLUSIONS: RA regulates lung microvascular angiogenesis; the endothelium produces CYP26A1 which is increased in emphysema, possibly leading to reduced RA availability. Tretinoin 153-155 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 84-91 28087752-6 2017 Localisation of RA synthetic (RALDH-1) and degrading (cytochrome P450 subfamily 26 A1 (CYP26A1)) enzymes in human lung was determined by immunofluorescence. Tretinoin 16-18 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 87-94 28275201-6 2017 Of these, ketoconazole and liarozole have shown some benefits, but their usage is limited by side effects and low potency toward the cytochrome P450 26A1 isoform (CYP26A1), the main atRA hydroxylase. Ketoconazole 10-22 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 133-153 28275201-6 2017 Of these, ketoconazole and liarozole have shown some benefits, but their usage is limited by side effects and low potency toward the cytochrome P450 26A1 isoform (CYP26A1), the main atRA hydroxylase. Ketoconazole 10-22 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 163-170 28275201-9 2017 The population-based PBPK model of atRA disposition incorporated saturable metabolic clearance of atRA, induction of CYP26A1 by atRA, and the absorption and distribution kinetics of atRA. Tretinoin 35-39 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 117-124 28087565-5 2017 Embryonic tissues expressing Cyp26a1 show reduced efficiency of RA clearance. Tretinoin 64-66 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 29-36 27589347-7 2016 The 79 deleted genes included CYP26A1 and C1, both major RA-metabolizing enzymes. Tretinoin 57-59 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 30-37 28361034-10 2017 Two genes comprising OGS (CYP26A1 and RDH10) are strongly associated with ALDH1A2 in the retinoic acid (RA) pathways, suggesting a major role of RA signaling in early PCa progression. Tretinoin 89-102 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 26-33 27775549-3 2016 We previously showed that expression of CYP26 in BM stromal cells maintains a retinoic acid-low (RA-low) microenvironment that prevents the differentiation of normal and malignant hematopoietic cells. Tretinoin 78-91 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 40-45 27775549-3 2016 We previously showed that expression of CYP26 in BM stromal cells maintains a retinoic acid-low (RA-low) microenvironment that prevents the differentiation of normal and malignant hematopoietic cells. Radium 97-99 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 40-45 27775549-5 2016 CYP26-mediated inactivation of RA within the BM microenvironment prevented plasma cell differentiation and promoted a B cell-like, BTZ-resistant phenotype in human MM cells that were cocultured on BM stroma. Radium 31-33 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 27775549-5 2016 CYP26-mediated inactivation of RA within the BM microenvironment prevented plasma cell differentiation and promoted a B cell-like, BTZ-resistant phenotype in human MM cells that were cocultured on BM stroma. Bortezomib 131-134 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 27589347-11 2016 CYP26A1 and C1 haploinsufficiency may contribute to the elevated retinoic acid concentrations and clinical findings of the patient, although this phenotype has not been reported in other patients with similar deletions, suggesting that other unknown genetic or environmental factors may also contribute. Tretinoin 65-78 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 27193728-7 2016 PAMAM-50% C12 at 1 mug/mL altered the expression level of the oxidative stress-related genes, ROR1, CYP26A1, and TGFB1, which is a DNA damage response gene. (Z)-14-methylpentadec-2-enoic acid 10-13 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 100-107 27242163-3 2016 Immunocytochemistry was used to localize different components of RA signaling within sections of the retina and optic tectum, namely, the synthetic enzyme retinaldehyde dehydrogenase (RALDH), the RA binding proteins CRABPI and II, the retinoic acid receptors RARalpha, beta and gamma, and finally the catabolic enzyme CYP26A1. Tretinoin 65-67 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 318-325 27366899-2 2016 Cytochrome P450 oxidoreductase (POR) is the only obligate electron donor for all of the microsomal cytochrome P450 enzymes including CYP26A1 which is highly specific for ATRA metabolism and efficacy in human myeloid leukaemia cells. Tretinoin 170-174 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 133-140 26937021-0 2016 Identification of Tazarotenic Acid as the First Xenobiotic Substrate of Human Retinoic Acid Hydroxylase CYP26A1 and CYP26B1. tazarotenic acid 18-34 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 104-111 26937021-1 2016 Cytochrome P450 (CYP) 26A1 and 26B1 are heme-containing enzymes responsible for metabolizing all-trans retinoic acid (at-RA). Heme 40-44 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-35 26747727-6 2016 Additionally, Wnt3A attenuated ATRA-induced Cyp26a1 expression, inhibiting the degradation of ATRA into its oxidative forms. Tretinoin 31-35 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 44-51 27193728-7 2016 PAMAM-50% C12 at 1 mug/mL altered the expression level of the oxidative stress-related genes, ROR1, CYP26A1, and TGFB1, which is a DNA damage response gene. Poly(amidoamine) 0-5 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 100-107 27590114-6 2016 Consistent with these findings, increased colonic expression of the atRA-catabolizing enzyme, CYP26A1, correlated with reduced frequencies of tumoral cytotoxic CD8(+) T cells and with worse disease prognosis in human CRC. Tretinoin 68-72 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 94-101 27366899-7 2016 KEY FINDINGS: All-trans-retinoic acid treatment caused the expression of POR upregulation and CYP26A1 downregulation in dose- and time-dependent manners. Tretinoin 14-37 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 94-101 26374207-3 2016 The catabolic enzymes Cyp26a1 and Cyp26b1 have been studied in detail in the embryo, where they limit gradients of RA that form patterns of gene expression, crucial for morphogenesis. Tretinoin 115-117 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 22-29 26374207-7 2016 In vivo use of a new and potent inhibitor of the Cyp26 enzymes, ser 2-7, demonstrated a function for endogenous Cyp26 in the brain and that hippocampal RA levels can be raised by ser 2-7, altering the effect of RA on differential patterning of cell proliferation in the hippocampal region of neurogenesis, the subgranular zone. Serine 64-67 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 49-54 26374207-7 2016 In vivo use of a new and potent inhibitor of the Cyp26 enzymes, ser 2-7, demonstrated a function for endogenous Cyp26 in the brain and that hippocampal RA levels can be raised by ser 2-7, altering the effect of RA on differential patterning of cell proliferation in the hippocampal region of neurogenesis, the subgranular zone. Serine 64-67 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 112-117 26374207-7 2016 In vivo use of a new and potent inhibitor of the Cyp26 enzymes, ser 2-7, demonstrated a function for endogenous Cyp26 in the brain and that hippocampal RA levels can be raised by ser 2-7, altering the effect of RA on differential patterning of cell proliferation in the hippocampal region of neurogenesis, the subgranular zone. Tretinoin 152-154 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 49-54 26374207-7 2016 In vivo use of a new and potent inhibitor of the Cyp26 enzymes, ser 2-7, demonstrated a function for endogenous Cyp26 in the brain and that hippocampal RA levels can be raised by ser 2-7, altering the effect of RA on differential patterning of cell proliferation in the hippocampal region of neurogenesis, the subgranular zone. Serine 179-182 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 49-54 26374207-7 2016 In vivo use of a new and potent inhibitor of the Cyp26 enzymes, ser 2-7, demonstrated a function for endogenous Cyp26 in the brain and that hippocampal RA levels can be raised by ser 2-7, altering the effect of RA on differential patterning of cell proliferation in the hippocampal region of neurogenesis, the subgranular zone. Tretinoin 211-213 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 49-54 26937021-1 2016 Cytochrome P450 (CYP) 26A1 and 26B1 are heme-containing enzymes responsible for metabolizing all-trans retinoic acid (at-RA). Tretinoin 103-116 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-35 26937021-6 2016 The CYP26A1 and CYP26B1 homology models predicted that the benzothiopyranyl moiety of tazarotenic acid would be oriented toward the heme of each enzyme and suggested that tazarotenic acid would be a substrate of CYP26A1 and CYP26B1. tazarotenic acid 86-102 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-11 26937021-6 2016 The CYP26A1 and CYP26B1 homology models predicted that the benzothiopyranyl moiety of tazarotenic acid would be oriented toward the heme of each enzyme and suggested that tazarotenic acid would be a substrate of CYP26A1 and CYP26B1. tazarotenic acid 86-102 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 212-219 26937021-6 2016 The CYP26A1 and CYP26B1 homology models predicted that the benzothiopyranyl moiety of tazarotenic acid would be oriented toward the heme of each enzyme and suggested that tazarotenic acid would be a substrate of CYP26A1 and CYP26B1. Heme 132-136 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-11 26937021-6 2016 The CYP26A1 and CYP26B1 homology models predicted that the benzothiopyranyl moiety of tazarotenic acid would be oriented toward the heme of each enzyme and suggested that tazarotenic acid would be a substrate of CYP26A1 and CYP26B1. Heme 132-136 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 212-219 26937021-6 2016 The CYP26A1 and CYP26B1 homology models predicted that the benzothiopyranyl moiety of tazarotenic acid would be oriented toward the heme of each enzyme and suggested that tazarotenic acid would be a substrate of CYP26A1 and CYP26B1. tazarotenic acid 171-187 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-11 26937021-6 2016 The CYP26A1 and CYP26B1 homology models predicted that the benzothiopyranyl moiety of tazarotenic acid would be oriented toward the heme of each enzyme and suggested that tazarotenic acid would be a substrate of CYP26A1 and CYP26B1. tazarotenic acid 171-187 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 212-219 26937021-7 2016 Metabolite identification experiments indicated that CYP26A1 and CYP26B1 oxidatively metabolized tazarotenic acid on the predicted moiety, with in vitro rates of metabolite formation by CYP26A1 and CYP26B1 being the highest across a panel of enzymes. tazarotenic acid 97-113 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 53-60 26937021-7 2016 Metabolite identification experiments indicated that CYP26A1 and CYP26B1 oxidatively metabolized tazarotenic acid on the predicted moiety, with in vitro rates of metabolite formation by CYP26A1 and CYP26B1 being the highest across a panel of enzymes. tazarotenic acid 97-113 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 186-193 26937021-9 2016 Overall, the homology models presented herein describe the enzyme characteristics leading to the metabolism of tazarotenic acid by CYP26A1 and CYP26B1 and support a potential role for the CYP26 enzymes in the metabolism of xenobiotics. tazarotenic acid 111-127 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 131-138 26937021-9 2016 Overall, the homology models presented herein describe the enzyme characteristics leading to the metabolism of tazarotenic acid by CYP26A1 and CYP26B1 and support a potential role for the CYP26 enzymes in the metabolism of xenobiotics. tazarotenic acid 111-127 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 131-136 26918322-1 2016 Cytochrome P450 CYP26 enzymes are responsible for all-trans-retinoic acid (atRA) clearance. Tretinoin 50-73 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 16-21 26918322-1 2016 Cytochrome P450 CYP26 enzymes are responsible for all-trans-retinoic acid (atRA) clearance. Tretinoin 75-79 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 16-21 26918322-2 2016 Inhibition of CYP26 enzymes will increase endogenous atRA concentrations and is an attractive therapeutic target. Tretinoin 53-57 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 14-19 26918322-3 2016 However, the selectivity and potency of the existing atRA metabolism inhibitors toward CYP26A1 and CYP26B1 is unknown, and no selective CYP26A1 or CYP26B1 inhibitors have been developed. Tretinoin 53-57 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 87-94 26918322-5 2016 A series of nonazole CYP26A1 selective inhibitors was identified with low nM potency. nonazole 12-20 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 21-28 26999080-3 2016 Furthermore, the docking capabilities of the model were assessed by docking of the natural substrate all-trans-retinoic acid (atRA), and a group of known azole- and tetralone-based CYP26A1 inhibitors. Tretinoin 126-130 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 181-188 26999080-3 2016 Furthermore, the docking capabilities of the model were assessed by docking of the natural substrate all-trans-retinoic acid (atRA), and a group of known azole- and tetralone-based CYP26A1 inhibitors. Azoles 154-159 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 181-188 26999080-3 2016 Furthermore, the docking capabilities of the model were assessed by docking of the natural substrate all-trans-retinoic acid (atRA), and a group of known azole- and tetralone-based CYP26A1 inhibitors. Tetralones 165-174 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 181-188 26009309-1 2016 Retinoic acid (RA)-metabolizing enzyme CYP26A1 has been shown to have increased expression levels in breast cancers and to effectively promote the survival of breast carcinoma cells, implying a potential oncogenic function. Tretinoin 0-13 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 39-46 26009309-1 2016 Retinoic acid (RA)-metabolizing enzyme CYP26A1 has been shown to have increased expression levels in breast cancers and to effectively promote the survival of breast carcinoma cells, implying a potential oncogenic function. Tretinoin 15-17 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 39-46 26058854-0 2015 The retinoic acid-metabolizing enzyme CYP26A1 upregulates fascin and promotes the malignant behavior of breast carcinoma cells. Tretinoin 4-17 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 38-45 26365710-0 2015 Design, synthesis, and biological evaluation of amide imidazole derivatives as novel metabolic enzyme CYP26A1 inhibitors. amide imidazole 48-63 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 102-109 26365710-2 2015 CYP26A1 enzyme, induced by ATRA in liver and target tissues, metabolizes ATRA into 4-hydroxyl-RA. Tretinoin 27-31 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 26365710-2 2015 CYP26A1 enzyme, induced by ATRA in liver and target tissues, metabolizes ATRA into 4-hydroxyl-RA. Tretinoin 73-77 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 26365710-2 2015 CYP26A1 enzyme, induced by ATRA in liver and target tissues, metabolizes ATRA into 4-hydroxyl-RA. 4-hydroxyl-ra 83-96 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 26365710-4 2015 Herein, we describe the design, synthesis and biological evaluation of a series of new amide imidazole derivatives as retinoic acid metabolism blocking agents (RAMBAs) toward CYP26A1 enzyme. amide imidazole 87-102 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 175-182 26365710-4 2015 Herein, we describe the design, synthesis and biological evaluation of a series of new amide imidazole derivatives as retinoic acid metabolism blocking agents (RAMBAs) toward CYP26A1 enzyme. Tretinoin 118-131 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 175-182 26365710-11 2015 Both compounds 20 and 23 showed higher selectivity for CYP26A1 over other CYPs (CYP2D6, CYP3A4) when compared to liarozole. liarozole 113-122 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 55-62 30695588-3 2016 We showed that ALDHA1 and RDH10 expression were inversely related with expression of a key gene for all-trans-retinoic acid catabolism, CYP26A1, and correlated with expression of RARalpha and PPARbeta/ genes. retinoic 110-118 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 136-143 26058854-1 2015 The retinoic acid (RA)-metabolizing enzyme CYP26A1 has been shown to efficiently enhance the oncogenic potential of breast cancer, suggesting a potential oncogenic function. Tretinoin 4-17 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 43-50 26058854-1 2015 The retinoic acid (RA)-metabolizing enzyme CYP26A1 has been shown to efficiently enhance the oncogenic potential of breast cancer, suggesting a potential oncogenic function. Tretinoin 19-21 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 43-50 26058854-9 2015 These data suggest that fascin expression is modulated by the intracellular RA status regulated by the expression of CYP26A1 and plays a significant role in the malignant behavior of CYP26A1-expressing breast carcinoma cells. Tretinoin 76-78 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 117-124 25684424-0 2015 Synthesis and biological evaluation of 3-phenyl-3-aryl carboxamido propanoic acid derivatives as small molecule inhibitors of retinoic acid 4-hydroxylase (CYP26A1). 3-phenyl-3-aryl carboxamido propanoic acid 39-81 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 155-162 25915157-3 2015 We showed that bone marrow (BM) stromal cytochrome P450 (CYP)26 enzymes protect normal hematopoietic stem cells (HSCs) from the pro-differentiation effects of retinoic acid. Tretinoin 159-172 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 40-63 26047326-4 2015 The microenvironment expression of cytochrome P450 (CYP)26, a retinoid-metabolizing enzyme was shown to determine normal hematopoietic stem cell fate. Retinoids 62-70 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 35-58 26047326-8 2015 Inhibition of CYP26 rescued atRA levels and AML cell sensitivity in the presence of stroma. Tretinoin 28-32 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 14-19 26047326-9 2015 Our data suggest that stromal CYP26 activity creates retinoid low sanctuaries in the BM that protect AML cells from systemic atRA therapy. Retinoids 53-61 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 30-35 26047326-9 2015 Our data suggest that stromal CYP26 activity creates retinoid low sanctuaries in the BM that protect AML cells from systemic atRA therapy. Tretinoin 125-129 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 30-35 26047326-10 2015 Inhibition of CYP26 provides new opportunities to expand the clinical activity of atRA in both APL and non-APL AML. Tretinoin 82-86 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 14-19 25684424-6 2015 The most promising compound 32, with a CYP26A1 IC50 value of 1.36muM (compared to liarozole (IC50=2.45muM) and S8 (IC50=3.21muM)) displayed strong inhibitory and differentiation activity against HL60 cells. liarozole 82-91 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 39-46 25684424-7 2015 In addition, the study focused on the effect of beta-phenylalanine, which forms the coordination bond with the heme of CYP26A1. beta-phenylalanine 48-66 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 119-126 25684424-7 2015 In addition, the study focused on the effect of beta-phenylalanine, which forms the coordination bond with the heme of CYP26A1. Heme 111-115 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 119-126 25541526-0 2015 Molecular recognition of CYP26A1 binding pockets and structure-activity relationship studies for design of potent and selective retinoic acid metabolism blocking agents. Tretinoin 128-141 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 25-32 25541526-2 2015 However, ATRA is very easy to be metabolized into 4-hydroxyl-RA in vivo by CYP26A1, an inducible cytochrome P450 enzyme, eventually into more polar metabolites. Tretinoin 9-13 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 75-82 25541526-2 2015 However, ATRA is very easy to be metabolized into 4-hydroxyl-RA in vivo by CYP26A1, an inducible cytochrome P450 enzyme, eventually into more polar metabolites. 4-hydroxyl-ra 50-63 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 75-82 25541526-3 2015 Therefore, it is vital to develop specific retinoic acid metabolism blocking agents (RAMBAs) to inhibit the metabolic enzyme CYP26A1 in the treatment of relevant diseases aforementioned. Tretinoin 43-56 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 125-132 25541526-4 2015 In this study, CYP26A1 and its interactions with retinoic acid-competitive metabolism blocking agents were investigated by a combined ligand- and structure-based approach. Tretinoin 49-62 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 15-22 25492813-12 2015 These data support the role of CYP26A1 to clear bioactive retinoids, and suggest that the enzyme forming active 4-oxo-atRA may be important in modulating retinoid action. Retinoids 58-67 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 31-38 26233912-0 2015 Role of Retinoic Acid-Metabolizing Cytochrome P450s, CYP26, in Inflammation and Cancer. Tretinoin 8-21 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 53-58 26233912-3 2015 Concentrations of atRA are tightly regulated in tissues, predominantly by the availability of retinol, synthesis of atRA by ALDH1A enzymes and metabolism and clearance of atRA by CYP26 enzymes. Tretinoin 18-22 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 179-184 26233912-5 2015 In the immune system, the ALDH1A and CYP26 enzymes appear to modulate RA concentrations. Radium 70-72 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 37-42 26233912-10 2015 Inhibition of the CYP26 enzymes to increase atRA concentrations and combat therapy resistance has been pursued as a drug target in these cancers. Tretinoin 44-48 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 18-23 25492813-0 2015 Induction of CYP26A1 by metabolites of retinoic acid: evidence that CYP26A1 is an important enzyme in the elimination of active retinoids. Tretinoin 39-52 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 13-20 25492813-12 2015 These data support the role of CYP26A1 to clear bioactive retinoids, and suggest that the enzyme forming active 4-oxo-atRA may be important in modulating retinoid action. 4-oxoretinoic acid 112-122 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 31-38 25492813-0 2015 Induction of CYP26A1 by metabolites of retinoic acid: evidence that CYP26A1 is an important enzyme in the elimination of active retinoids. Tretinoin 39-52 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 68-75 25492813-0 2015 Induction of CYP26A1 by metabolites of retinoic acid: evidence that CYP26A1 is an important enzyme in the elimination of active retinoids. Retinoids 128-137 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 13-20 25492813-12 2015 These data support the role of CYP26A1 to clear bioactive retinoids, and suggest that the enzyme forming active 4-oxo-atRA may be important in modulating retinoid action. Retinoids 58-66 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 31-38 25492813-0 2015 Induction of CYP26A1 by metabolites of retinoic acid: evidence that CYP26A1 is an important enzyme in the elimination of active retinoids. Retinoids 128-137 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 68-75 24667328-2 2014 RA levels in embryos are dampened by Cyp26 enzymes, which metabolize RA into easily degraded derivatives. Tretinoin 0-2 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 37-42 25492813-2 2015 The primary hydroxylated metabolites formed from atRA by CYP26A1, and the subsequent metabolite 4-oxo-atRA, bind to RARs and potentially have biologic activity. 4-oxoretinoic acid 96-106 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 57-64 25492813-3 2015 Hence, CYP26A1, the main atRA hydroxylase, may function either to deplete bioactive retinoids or to form active metabolites. Retinoids 84-93 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 7-14 25492813-5 2015 After treatment of HepG2 cells with atRA, (4S)-OH-atRA, (4R)-OH-atRA, 4-oxo-atRA, and 18-OH-atRA, mRNAs of CYP26A1 and RARbeta were increased 300- to 3000-fold, with 4-oxo-atRA and atRA being the most potent inducers. Tretinoin 36-40 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 107-114 25492813-5 2015 After treatment of HepG2 cells with atRA, (4S)-OH-atRA, (4R)-OH-atRA, 4-oxo-atRA, and 18-OH-atRA, mRNAs of CYP26A1 and RARbeta were increased 300- to 3000-fold, with 4-oxo-atRA and atRA being the most potent inducers. (4S)-OH-atRA 42-54 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 107-114 25492813-5 2015 After treatment of HepG2 cells with atRA, (4S)-OH-atRA, (4R)-OH-atRA, 4-oxo-atRA, and 18-OH-atRA, mRNAs of CYP26A1 and RARbeta were increased 300- to 3000-fold, with 4-oxo-atRA and atRA being the most potent inducers. (4r)-oh-atra 56-68 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 107-114 25492813-5 2015 After treatment of HepG2 cells with atRA, (4S)-OH-atRA, (4R)-OH-atRA, 4-oxo-atRA, and 18-OH-atRA, mRNAs of CYP26A1 and RARbeta were increased 300- to 3000-fold, with 4-oxo-atRA and atRA being the most potent inducers. 4-oxoretinoic acid 70-80 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 107-114 25492813-5 2015 After treatment of HepG2 cells with atRA, (4S)-OH-atRA, (4R)-OH-atRA, 4-oxo-atRA, and 18-OH-atRA, mRNAs of CYP26A1 and RARbeta were increased 300- to 3000-fold, with 4-oxo-atRA and atRA being the most potent inducers. 4-oxoretinoic acid 166-176 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 107-114 25492813-5 2015 After treatment of HepG2 cells with atRA, (4S)-OH-atRA, (4R)-OH-atRA, 4-oxo-atRA, and 18-OH-atRA, mRNAs of CYP26A1 and RARbeta were increased 300- to 3000-fold, with 4-oxo-atRA and atRA being the most potent inducers. Tretinoin 50-54 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 107-114 25492813-7 2015 In human hepatocytes, atRA, 4-OH-atRA, and 4-oxo-atRA induced CYP26A1 and 4-oxo-atRA formation was observed from 4-OH-atRA. Tretinoin 22-26 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 62-69 25492813-7 2015 In human hepatocytes, atRA, 4-OH-atRA, and 4-oxo-atRA induced CYP26A1 and 4-oxo-atRA formation was observed from 4-OH-atRA. 4-oh-atra 28-37 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 62-69 25492813-7 2015 In human hepatocytes, atRA, 4-OH-atRA, and 4-oxo-atRA induced CYP26A1 and 4-oxo-atRA formation was observed from 4-OH-atRA. 4-oxoretinoic acid 43-53 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 62-69 25492813-10 2015 Although 4-oxo-atRA was not formed by CYP26A1, it was depleted by CYP26A1 (Km = 63 nM and intrinsic clearance = 90 mul/min per pmol). 4-oxoretinoic acid 9-19 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 66-73 25492813-11 2015 Similarly, CYP26A1 depleted 18-OH-atRA and the 4-OH-atRA enantiomers. 18-oh-atra 28-38 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 11-18 25492813-11 2015 Similarly, CYP26A1 depleted 18-OH-atRA and the 4-OH-atRA enantiomers. 4-oh-atra 47-56 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 11-18 25839051-1 2015 BQ chewing may produce significant amounts of reactive oxygen species (ROS), resulting in oral mucosa damage, and ROS may be metabolized by CYP26 families. bulaquine 0-2 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 140-145 24819304-1 2014 CYP26A1 expression is very highly induced by retinoic acid (RA) in the liver, compared to most other tissues, suggesting that a liver-enriched factor may be required for its physiological transcriptional response. Tretinoin 45-58 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 24819304-1 2014 CYP26A1 expression is very highly induced by retinoic acid (RA) in the liver, compared to most other tissues, suggesting that a liver-enriched factor may be required for its physiological transcriptional response. Tretinoin 60-62 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 24819304-3 2014 In this study, we hypothesized that HNF4alpha and RARs may cooperate in an RA-dependent manner to induce a high level of CYP26A1 expression in liver cells. Tretinoin 50-52 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 121-128 24819304-4 2014 Partial inhibition of endogenous HNF4alpha by siRNA reduced the level of RA-induced CYP26A1 mRNA in HepG2 cells. Tretinoin 73-75 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 84-91 24819304-5 2014 Cotransfection of HNF4alpha, with or without RARs, demonstrated RA-dependent activation of a human CYP26A1 promoter-luciferase construct. Tretinoin 45-47 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 99-106 24819304-7 2014 In EMSA and ChIP analyses HNF4alpha and RARs binding in the proximal and distal CYP26A1 promoter regions was significantly higher in RA-treated cells. Tretinoin 40-42 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 80-87 24819304-9 2014 Our results indicate that HNF4alpha coordinates with RARs in an RA-dependent manner to strongly induce CYP26A1 gene expression in the liver, which may explain the high level of response to RA observed in vivo. Tretinoin 53-55 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 103-110 24819304-9 2014 Our results indicate that HNF4alpha coordinates with RARs in an RA-dependent manner to strongly induce CYP26A1 gene expression in the liver, which may explain the high level of response to RA observed in vivo. Tretinoin 64-66 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 103-110 24799257-5 2014 These three natural RAR agonists with diterpene structures, while structurally different from ATRA, were able to increase the mRNA levels of the constitutive androstane receptor in HepG2 cells, induce F9 cell differentiation followed by Cyp26a1 mRNA expression, and differentiate HL-60 cells via RAR activation in a different manner from ATRA. Diterpenes 38-47 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 237-244 24667328-9 2014 Our results suggest that localized expression of Cyp26 enzymes cell non-autonomously defines the boundaries between the cardiac and VP fields within the ALPM through regulating RA levels, which ensures a proper balance of myocardial and endothelial lineages. Tretinoin 177-179 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 49-54 25839051-1 2015 BQ chewing may produce significant amounts of reactive oxygen species (ROS), resulting in oral mucosa damage, and ROS may be metabolized by CYP26 families. Reactive Oxygen Species 114-117 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 140-145 25236354-6 2014 PCR analysis revealed a significant upregulation of retinoid-regulated genes such as CYP26A1 and STRA6 in LRAT KD cells, suggesting their possible involvement in mediating retinoid resistance in melanoma cells. Retinoids 52-60 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 85-92 25236354-6 2014 PCR analysis revealed a significant upregulation of retinoid-regulated genes such as CYP26A1 and STRA6 in LRAT KD cells, suggesting their possible involvement in mediating retinoid resistance in melanoma cells. Retinoids 172-180 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 85-92 25294402-0 2014 Increased expression of the retinoic acid-metabolizing enzyme CYP26A1 during the progression of cervical squamous neoplasia and head and neck cancer. Tretinoin 28-41 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 62-69 24821725-6 2014 Importantly, RA treatment differentially mediates the removal of HDACs from the Hoxa1, Cyp26a1, and RARbeta2 genes and promotes the deposition of the H3K27ac mark at these genes. Tretinoin 13-15 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 87-94 24667328-2 2014 RA levels in embryos are dampened by Cyp26 enzymes, which metabolize RA into easily degraded derivatives. Tretinoin 69-71 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 37-42 24642534-3 2014 Inside the cells 1,25D is degraded to calcitrioic acid by a mitochondrial enzyme CYP24A1, while ATRA is degraded to several polar metabolites by CYP26. Tretinoin 96-100 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 145-150 24608339-4 2014 Immunohistochemistry was performed on the tissue microarray using monoclonal antibodies which we have developed to the retinoic acid metabolising enzymes CYP26A1, CYP26B1, CYP26C1 and lecithin retinol acyl transferase (LRAT) using a semi-quantitative scoring scheme to assess expression. Tretinoin 119-132 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 154-161 24608339-12 2014 This study has shown that the retinoic acid metabolising enzymes CYP26A1, CYP26B1 and LRAT are significantly overexpressed in colorectal cancer and that CYP26B1 and LRAT are significantly associated with prognosis both in the total cohort and in those tumours which are mismatch repair proficient. Tretinoin 30-43 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 65-72 23601821-0 2013 2-(2-Methylfuran-3-carboxamido)-3-phenylpropanoic acid, a potential CYP26A1 inhibitor to enhance all-trans retinoic acid-induced leukemia cell differentiation based on virtual screening and biological evaluation. 2-(2-methylfuran-3-carboxamido)-3-phenylpropanoic acid 0-54 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 68-75 25217584-2 2014 We have studied the effect of activation of the retinoic A receptor, at the RARE-promoter chromatin of CASP9 and CYP26A1 genes, 15 and 45 min following RA exposure, and we found that histone H3 lysines 4 and 9 are demethylated by the lysine-specific demethylase, LSD1 and by the JMJ-domain containing demethylase, D2A. Radium 76-78 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 113-120 23946489-3 2013 We show here that the cytochrome P450 enzyme Cyp26a1, which metabolizes all-trans-retinoic acid (RA) and thereby reduces RA levels, plays a crucial role in specifying MN columnar subtypes. Tretinoin 72-95 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 45-52 23946489-3 2013 We show here that the cytochrome P450 enzyme Cyp26a1, which metabolizes all-trans-retinoic acid (RA) and thereby reduces RA levels, plays a crucial role in specifying MN columnar subtypes. Tretinoin 97-99 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 45-52 23946489-3 2013 We show here that the cytochrome P450 enzyme Cyp26a1, which metabolizes all-trans-retinoic acid (RA) and thereby reduces RA levels, plays a crucial role in specifying MN columnar subtypes. Tretinoin 121-123 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 45-52 24043786-9 2013 Accordingly, we found that bone marrow stromal cell CYP26 was also able to inactivate retinoids in serum, preventing RA signaling. Retinoids 86-95 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 52-57 24043786-9 2013 Accordingly, we found that bone marrow stromal cell CYP26 was also able to inactivate retinoids in serum, preventing RA signaling. Tretinoin 117-119 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 52-57 23601821-0 2013 2-(2-Methylfuran-3-carboxamido)-3-phenylpropanoic acid, a potential CYP26A1 inhibitor to enhance all-trans retinoic acid-induced leukemia cell differentiation based on virtual screening and biological evaluation. Tretinoin 107-120 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 68-75 23053054-2 2012 Cytochrome P450 26A1 (Cyp26a1), an RA-metabolizing enzyme, is involved in mammalian early pregnancy. Tretinoin 35-37 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-20 23441061-6 2013 Finally, overexpression of SKI in human cells results in reduced levels of CYP26A1 mRNA, a known target of retinoic acid signaling. Tretinoin 107-120 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 75-82 23688132-6 2013 Since the initial identification of inhibitors of RA metabolism, CYP26 enzymes have been characterized as the main enzymes responsible for RA clearance. Tretinoin 50-52 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 65-70 23688132-6 2013 Since the initial identification of inhibitors of RA metabolism, CYP26 enzymes have been characterized as the main enzymes responsible for RA clearance. Tretinoin 139-141 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 65-70 23688132-8 2013 The basic principle of development of CYP26 inhibitors is that endogenous RA concentrations will be increased in the presence of a CYP26 inhibitor, thus, potentiating the activity of endogenous RA in a cell-type specific manner. Tretinoin 74-76 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 38-43 23688132-8 2013 The basic principle of development of CYP26 inhibitors is that endogenous RA concentrations will be increased in the presence of a CYP26 inhibitor, thus, potentiating the activity of endogenous RA in a cell-type specific manner. Tretinoin 74-76 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 131-136 23688132-8 2013 The basic principle of development of CYP26 inhibitors is that endogenous RA concentrations will be increased in the presence of a CYP26 inhibitor, thus, potentiating the activity of endogenous RA in a cell-type specific manner. Tretinoin 194-196 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 38-43 23688132-8 2013 The basic principle of development of CYP26 inhibitors is that endogenous RA concentrations will be increased in the presence of a CYP26 inhibitor, thus, potentiating the activity of endogenous RA in a cell-type specific manner. Tretinoin 194-196 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 131-136 23071109-2 2012 atRA is metabolized mainly by CYP26A1, but other P450 enzymes such as CYP2C8 and CYP3As also contribute to atRA 4-hydroxylation. Tretinoin 0-4 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 30-37 23071109-5 2012 CYP26A1 was found to form predominantly (4S)-OH-RA. (4s)-oh-ra 40-50 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 23071109-6 2012 This stereoselectivity was rationalized via docking of atRA in the active site of a CYP26A1 homology model. Tretinoin 55-59 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 84-91 23071109-10 2012 Both (4S)- and (4R)-OH-RA were substrates of CYP26A1 but (4S)-OH-RA was cleared 3-fold faster than (4R)-OH-RA. (4s)- and 5-14 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 45-52 23071109-10 2012 Both (4S)- and (4R)-OH-RA were substrates of CYP26A1 but (4S)-OH-RA was cleared 3-fold faster than (4R)-OH-RA. (4r)-oh-ra 15-25 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 45-52 23071109-13 2012 The stereoselectivity observed in CYP26A1 function will aid in better understanding of the active site features of the enzyme and the disposition of biologically active retinoids. Retinoids 169-178 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 34-41 23053054-2 2012 Cytochrome P450 26A1 (Cyp26a1), an RA-metabolizing enzyme, is involved in mammalian early pregnancy. Tretinoin 35-37 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 22-29 21428449-0 2011 Small molecule inhibitors of retinoic acid 4-hydroxylase (CYP26): synthesis and biological evaluation of imidazole methyl 3-(4-(aryl-2-ylamino)phenyl)propanoates. imidazole methyl 3-(4-(aryl-2-ylamino)phenyl)propanoates 105-161 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 29-56 22542753-5 2012 Functionality of the co-cultures was assayed using as endpoints the retinol-dependent secretion of RBP4 and the retinoic acid-dependent induction of CYP26A1 in hepatocytes. Tretinoin 112-125 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 149-156 22542753-7 2012 HepaRG hepatocytes were also shown to up-regulate the expression of CYP26A1 mRNA in response to retinoid treatment. Retinoids 96-104 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 68-75 22727372-0 2012 Novel retinoic acid 4-hydroxylase (CYP26) inhibitors based on a 3-(1H-imidazol- and triazol-1-yl)-2,2-dimethyl-3-(4-(phenylamino)phenyl)propyl scaffold. 3-(1h-imidazol- and triazol-1-yl)-2,2-dimethyl-3-(4-(phenylamino)phenyl)propyl scaffold 64-151 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 6-33 22727372-0 2012 Novel retinoic acid 4-hydroxylase (CYP26) inhibitors based on a 3-(1H-imidazol- and triazol-1-yl)-2,2-dimethyl-3-(4-(phenylamino)phenyl)propyl scaffold. 3-(1h-imidazol- and triazol-1-yl)-2,2-dimethyl-3-(4-(phenylamino)phenyl)propyl scaffold 64-151 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 35-40 22727372-3 2012 RA metabolism inhibitors (RAMBAs or CYP26 inhibitors) are attracting increasing interest as an alternative method for enhancing endogenous levels of retinoic acid in the treatment of hyperproliferative disease. Tretinoin 0-2 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 36-41 22727372-3 2012 RA metabolism inhibitors (RAMBAs or CYP26 inhibitors) are attracting increasing interest as an alternative method for enhancing endogenous levels of retinoic acid in the treatment of hyperproliferative disease. Tretinoin 149-162 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 36-41 22727372-4 2012 Here the synthesis and inhibitory activity of novel 3-(1H-imidazol- and triazol-1-yl)-2,2-dimethyl-3-(4-(phenylamino)phenyl)propyl derivatives in a MCF-7 CYP26A1 microsomal assay are described. 3-(1h-imidazol- and triazol-1-yl)-2,2-dimethyl-3-(4-(phenylamino)phenyl)propyl derivatives 52-142 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 154-161 22640768-10 2012 A few well-validated, specific predictors such as OPRM1, ADH1B, ALDH2, CHRNA5, and CYP26 have been identified and can provide some specific guidance, for example, to understand alcohol-related flushing and upper GI cancer risk (ADH1B and AKLDH2), variation in nicotine metabolism (CYP26), and, potentially, naltrexone treatment response (OPRM1). Alcohols 177-184 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 83-88 22640768-10 2012 A few well-validated, specific predictors such as OPRM1, ADH1B, ALDH2, CHRNA5, and CYP26 have been identified and can provide some specific guidance, for example, to understand alcohol-related flushing and upper GI cancer risk (ADH1B and AKLDH2), variation in nicotine metabolism (CYP26), and, potentially, naltrexone treatment response (OPRM1). Alcohols 177-184 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 281-286 22640768-10 2012 A few well-validated, specific predictors such as OPRM1, ADH1B, ALDH2, CHRNA5, and CYP26 have been identified and can provide some specific guidance, for example, to understand alcohol-related flushing and upper GI cancer risk (ADH1B and AKLDH2), variation in nicotine metabolism (CYP26), and, potentially, naltrexone treatment response (OPRM1). Nicotine 260-268 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 83-88 22640768-10 2012 A few well-validated, specific predictors such as OPRM1, ADH1B, ALDH2, CHRNA5, and CYP26 have been identified and can provide some specific guidance, for example, to understand alcohol-related flushing and upper GI cancer risk (ADH1B and AKLDH2), variation in nicotine metabolism (CYP26), and, potentially, naltrexone treatment response (OPRM1). Naltrexone 307-317 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 83-88 22179182-0 2011 Enhanced expression of retinoic acid-metabolizing enzyme CYP26A1 in sunlight-damaged human skin. Tretinoin 23-36 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 57-64 22179182-2 2011 CYP26A1, the gene encoding a cytochrome P450 enzyme specifically involved in metabolic inactivation of retinoic acid (RA), the most active vitamin A derivative, has been shown to result in a state of functional VAD of the cell. Tretinoin 103-116 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 22179182-2 2011 CYP26A1, the gene encoding a cytochrome P450 enzyme specifically involved in metabolic inactivation of retinoic acid (RA), the most active vitamin A derivative, has been shown to result in a state of functional VAD of the cell. Tretinoin 118-120 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 22179182-2 2011 CYP26A1, the gene encoding a cytochrome P450 enzyme specifically involved in metabolic inactivation of retinoic acid (RA), the most active vitamin A derivative, has been shown to result in a state of functional VAD of the cell. Vitamin A 139-148 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 21906690-9 2011 DISCUSSION: CYP26 enzymes are functionally expressed in microsomal fractions of insect cells and stably bind radiolabeled RA isomers with affinities respecting their substrate specificities. Tretinoin 122-124 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 12-17 21838328-0 2011 Synthesis and biological evaluation of 3-(1H-imidazol- and triazol-1-yl)-2,2-dimethyl-3-[4-(naphthalen-2-ylamino)phenyl]propyl derivatives as small molecule inhibitors of retinoic acid 4-hydroxylase (CYP26). 3-(1h-imidazol 39-53 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 171-198 21838328-0 2011 Synthesis and biological evaluation of 3-(1H-imidazol- and triazol-1-yl)-2,2-dimethyl-3-[4-(naphthalen-2-ylamino)phenyl]propyl derivatives as small molecule inhibitors of retinoic acid 4-hydroxylase (CYP26). 3-(1h-imidazol 39-53 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 200-205 21838328-0 2011 Synthesis and biological evaluation of 3-(1H-imidazol- and triazol-1-yl)-2,2-dimethyl-3-[4-(naphthalen-2-ylamino)phenyl]propyl derivatives as small molecule inhibitors of retinoic acid 4-hydroxylase (CYP26). triazol-1-yl)-2,2-dimethyl-3-[4-(naphthalen-2-ylamino)phenyl]propyl derivatives 59-138 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 171-198 21838328-0 2011 Synthesis and biological evaluation of 3-(1H-imidazol- and triazol-1-yl)-2,2-dimethyl-3-[4-(naphthalen-2-ylamino)phenyl]propyl derivatives as small molecule inhibitors of retinoic acid 4-hydroxylase (CYP26). triazol-1-yl)-2,2-dimethyl-3-[4-(naphthalen-2-ylamino)phenyl]propyl derivatives 59-138 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 200-205 21838328-1 2011 The synthesis and potent inhibitory activity of novel 3-(1H-imidazol- and triazol-1-yl)-2,2-dimethyl-3-(4-(naphthalen-2-ylamino)phenyl)propyl derivatives vs a MCF-7 CYP26A1 microsomal assay is described. 3-(1h-imidazol- and triazol-1-yl)-2,2-dimethyl-3-(4-(naphthalen-2-ylamino)phenyl)propyl derivatives 54-153 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 165-172 21838328-4 2011 Compounds with CYP26 inhibitory IC(50) values <=50 nM enhanced the biological activity of exogenous ATRA, as evidenced by a 3.7-5.8-fold increase in CYP26A1 mRNA in SH-SY5Y neuroblastoma cells as compared with ATRA alone. Tretinoin 103-107 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 15-20 21838328-4 2011 Compounds with CYP26 inhibitory IC(50) values <=50 nM enhanced the biological activity of exogenous ATRA, as evidenced by a 3.7-5.8-fold increase in CYP26A1 mRNA in SH-SY5Y neuroblastoma cells as compared with ATRA alone. Tretinoin 103-107 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 152-159 21838328-4 2011 Compounds with CYP26 inhibitory IC(50) values <=50 nM enhanced the biological activity of exogenous ATRA, as evidenced by a 3.7-5.8-fold increase in CYP26A1 mRNA in SH-SY5Y neuroblastoma cells as compared with ATRA alone. Tretinoin 213-217 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 15-20 21521770-2 2011 The hepatic clearance of atRA is mediated primarily by CYP26A1, but design of CYP26A1 inhibitors is hindered by lack of information on CYP26A1 structure and structure-activity relationships of its ligands. Tretinoin 25-29 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 55-62 21521770-3 2011 The aim of this study was to identify the primary metabolites of atRA formed by CYP26A1 and to characterize the ligand selectivity and ligand interactions of CYP26A1. Tretinoin 65-69 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 80-87 21521770-3 2011 The aim of this study was to identify the primary metabolites of atRA formed by CYP26A1 and to characterize the ligand selectivity and ligand interactions of CYP26A1. Tretinoin 65-69 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 158-165 21521770-4 2011 On the basis of high-resolution tandem mass spectrometry data, four metabolites formed from atRA by CYP26A1 were identified as 4-OH-RA, 4-oxo-RA, 16-OH-RA and 18-OH-RA. 4-oh-ra 127-134 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 100-107 21521770-4 2011 On the basis of high-resolution tandem mass spectrometry data, four metabolites formed from atRA by CYP26A1 were identified as 4-OH-RA, 4-oxo-RA, 16-OH-RA and 18-OH-RA. 4-oxoretinoic acid 136-144 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 100-107 21521770-4 2011 On the basis of high-resolution tandem mass spectrometry data, four metabolites formed from atRA by CYP26A1 were identified as 4-OH-RA, 4-oxo-RA, 16-OH-RA and 18-OH-RA. 16-oh-ra 146-154 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 100-107 21521770-4 2011 On the basis of high-resolution tandem mass spectrometry data, four metabolites formed from atRA by CYP26A1 were identified as 4-OH-RA, 4-oxo-RA, 16-OH-RA and 18-OH-RA. 18-oh-ra 159-167 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 100-107 21521770-5 2011 9-cis-RA and 13-cis-RA were also substrates of CYP26A1. Alitretinoin 0-8 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 47-54 21521770-5 2011 9-cis-RA and 13-cis-RA were also substrates of CYP26A1. Isotretinoin 13-22 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 47-54 22989911-0 2012 Synthesis and CYP26A1 inhibitory activity of novel methyl 3-[4-(arylamino)phenyl]-3-(azole)-2,2-dimethylpropanoates. methyl 3-[4-(arylamino)phenyl]-3-(azole)-2,2-dimethylpropanoates 51-115 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 14-21 22989911-2 2012 The use of ATRA or prodrugs as pharmacological agents is limited by a short half-life in vivo resulting from the activity of specific ATRA hydroxylases, CYP26 enzymes, induced by ATRA in liver and target tissues. Tretinoin 11-15 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 153-158 22989911-4 2012 The synthesis, CYP26A1 inhibitory activity and molecular modeling studies of novel methyl 3-[4-(arylamino)phenyl]-3-(azole)-2,2-dimethylpropanoates are presented. 3-[4-(arylamino)phenyl]-3-(azole)-2,2-dimethylpropanoates 90-147 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 15-22 22989911-6 2012 Both the methylenedioxyphenyl imidazole (17, IC(50) = 8 nM) and triazole (18, IC(50) = 6.7 nM) derivatives were potent inhibitors with additional binding interactions between the methylenedioxy moiety and the CYP26 active site likely to be the main factor. methylenedioxyphenyl imidazole 9-39 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 209-214 22989911-6 2012 Both the methylenedioxyphenyl imidazole (17, IC(50) = 8 nM) and triazole (18, IC(50) = 6.7 nM) derivatives were potent inhibitors with additional binding interactions between the methylenedioxy moiety and the CYP26 active site likely to be the main factor. Triazoles 64-72 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 209-214 22415012-1 2012 All-trans retinoic acid, controlled by cytochrome P450, family 26 (CYP26) enzymes, potentially has beneficial effects in atherosclerosis treatment. Tretinoin 10-23 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 39-65 22415012-1 2012 All-trans retinoic acid, controlled by cytochrome P450, family 26 (CYP26) enzymes, potentially has beneficial effects in atherosclerosis treatment. Tretinoin 10-23 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 67-72 22020119-2 2012 The cytochrome P450 enzymes CYP26 are believed to partially regulate cellular concentrations of atRA via oxidative metabolism and hence affect retinoid homeostasis and signaling. Tretinoin 96-100 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 28-33 22020119-8 2012 Qualitatively, recombinant CYP26A1 and CYP26B1 formed the same primary and sequential metabolites from atRA. Tretinoin 103-107 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 27-34 22020119-10 2012 The major atRA metabolites 4-OH-RA, 18-OH-RA and 4-oxo-RA were all substrates of CYP26A1 and CYP26B1, and CYP26A1 had a 2-10-fold higher catalytic activity towards all substrates tested. Tretinoin 10-14 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 81-88 22020119-10 2012 The major atRA metabolites 4-OH-RA, 18-OH-RA and 4-oxo-RA were all substrates of CYP26A1 and CYP26B1, and CYP26A1 had a 2-10-fold higher catalytic activity towards all substrates tested. Tretinoin 10-14 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 106-113 22020119-10 2012 The major atRA metabolites 4-OH-RA, 18-OH-RA and 4-oxo-RA were all substrates of CYP26A1 and CYP26B1, and CYP26A1 had a 2-10-fold higher catalytic activity towards all substrates tested. 4-oh-ra 27-34 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 81-88 22020119-10 2012 The major atRA metabolites 4-OH-RA, 18-OH-RA and 4-oxo-RA were all substrates of CYP26A1 and CYP26B1, and CYP26A1 had a 2-10-fold higher catalytic activity towards all substrates tested. 4-oh-ra 27-34 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 106-113 22020119-10 2012 The major atRA metabolites 4-OH-RA, 18-OH-RA and 4-oxo-RA were all substrates of CYP26A1 and CYP26B1, and CYP26A1 had a 2-10-fold higher catalytic activity towards all substrates tested. 18-oh-ra 36-44 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 81-88 22020119-10 2012 The major atRA metabolites 4-OH-RA, 18-OH-RA and 4-oxo-RA were all substrates of CYP26A1 and CYP26B1, and CYP26A1 had a 2-10-fold higher catalytic activity towards all substrates tested. 18-oh-ra 36-44 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 106-113 22020119-10 2012 The major atRA metabolites 4-OH-RA, 18-OH-RA and 4-oxo-RA were all substrates of CYP26A1 and CYP26B1, and CYP26A1 had a 2-10-fold higher catalytic activity towards all substrates tested. 4-oxo-ra 49-57 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 81-88 22020119-10 2012 The major atRA metabolites 4-OH-RA, 18-OH-RA and 4-oxo-RA were all substrates of CYP26A1 and CYP26B1, and CYP26A1 had a 2-10-fold higher catalytic activity towards all substrates tested. 4-oxo-ra 49-57 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 106-113 22020119-12 2012 CYP26A1 has higher catalytic activity than CYP26B1 and seems to be responsible for metabolism of atRA in tissues that function as a barrier for atRA exposure. Tretinoin 97-101 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 21906690-1 2011 INTRODUCTION: The cytochrome P450 CYP26 family of retinoic acid (RA) metabolizing enzymes, comprising CYP26A1, CYP26B1, and CYP26C1 is critical for establishing patterns of RA distribution during embryonic development and retinoid homeostasis in the adult. Tretinoin 50-63 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 34-39 21906690-1 2011 INTRODUCTION: The cytochrome P450 CYP26 family of retinoic acid (RA) metabolizing enzymes, comprising CYP26A1, CYP26B1, and CYP26C1 is critical for establishing patterns of RA distribution during embryonic development and retinoid homeostasis in the adult. Tretinoin 50-63 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 102-109 21906690-1 2011 INTRODUCTION: The cytochrome P450 CYP26 family of retinoic acid (RA) metabolizing enzymes, comprising CYP26A1, CYP26B1, and CYP26C1 is critical for establishing patterns of RA distribution during embryonic development and retinoid homeostasis in the adult. Tretinoin 65-67 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 34-39 21906690-1 2011 INTRODUCTION: The cytochrome P450 CYP26 family of retinoic acid (RA) metabolizing enzymes, comprising CYP26A1, CYP26B1, and CYP26C1 is critical for establishing patterns of RA distribution during embryonic development and retinoid homeostasis in the adult. Tretinoin 65-67 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 102-109 21906690-1 2011 INTRODUCTION: The cytochrome P450 CYP26 family of retinoic acid (RA) metabolizing enzymes, comprising CYP26A1, CYP26B1, and CYP26C1 is critical for establishing patterns of RA distribution during embryonic development and retinoid homeostasis in the adult. Tretinoin 173-175 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 34-39 21906690-1 2011 INTRODUCTION: The cytochrome P450 CYP26 family of retinoic acid (RA) metabolizing enzymes, comprising CYP26A1, CYP26B1, and CYP26C1 is critical for establishing patterns of RA distribution during embryonic development and retinoid homeostasis in the adult. Tretinoin 173-175 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 102-109 21529158-5 2011 The CYP26A1 gene is regulated by multiple RA response elements. Tretinoin 42-44 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-11 21529158-7 2011 Enzyme kinetic studies have demonstrated that several CYP proteins are capable of metabolizing at-RA; however, it is likely that CYP26A1 plays a major role in RA clearance. Tretinoin 98-100 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 129-136 21521770-9 2011 The retinoic acid receptor (RAR) gamma agonist CD1530 was as potent an inhibitor of CYP26A1 as ketoconazole with an IC(50) of 530 nM, whereas the RARalpha and RARbeta agonists tested did not significantly inhibit CYP26A1. 4-(6-hydroxy-7-tricyclo(3.3.1.13,7)dec-1-yl-2-naphthalenyl)benzoic acid 47-53 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 84-91 21521770-9 2011 The retinoic acid receptor (RAR) gamma agonist CD1530 was as potent an inhibitor of CYP26A1 as ketoconazole with an IC(50) of 530 nM, whereas the RARalpha and RARbeta agonists tested did not significantly inhibit CYP26A1. 4-(6-hydroxy-7-tricyclo(3.3.1.13,7)dec-1-yl-2-naphthalenyl)benzoic acid 47-53 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 213-220 21641329-0 2011 Minor complexes at work: light-harvesting by carotenoids in the photosystem II antenna complexes CP24 and CP26. Carotenoids 45-56 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 106-110 21641329-5 2011 In CP26 fast transfer (80 fs) occurs from the carotenoid S(2) state to chlorophylls a absorbing at 675 and 678 nm, whereas transfer from the hot S(1) state to the lowest energy chlorophylls is observed in <1 ps. Carotenoids 46-56 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 3-7 21641329-5 2011 In CP26 fast transfer (80 fs) occurs from the carotenoid S(2) state to chlorophylls a absorbing at 675 and 678 nm, whereas transfer from the hot S(1) state to the lowest energy chlorophylls is observed in <1 ps. chlorophylls a 71-85 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 3-7 21428449-0 2011 Small molecule inhibitors of retinoic acid 4-hydroxylase (CYP26): synthesis and biological evaluation of imidazole methyl 3-(4-(aryl-2-ylamino)phenyl)propanoates. imidazole methyl 3-(4-(aryl-2-ylamino)phenyl)propanoates 105-161 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 58-63 21428449-1 2011 The synthesis and potent inhibitory activity of novel imidazole methyl 3-(4-(aryl-2-ylamino)phenyl)propanoates in a MCF-7 CYP26A1 microsomal assay is described. imidazole 54-63 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 122-129 21428449-1 2011 The synthesis and potent inhibitory activity of novel imidazole methyl 3-(4-(aryl-2-ylamino)phenyl)propanoates in a MCF-7 CYP26A1 microsomal assay is described. 3-(4-(aryl-2-ylamino)phenyl)propanoates 71-110 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 122-129 21428449-2 2011 The induction of CYP26A1 mRNA was used to evaluate the ability of the compounds to enhance the biological effects of all-trans retinoic acid (ATRA) in a retinoid-responsive neuroblastoma cell line. Tretinoin 142-146 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 17-24 22347780-0 2011 2 D - QSAR studies on CYP26A1 inhibitory activity of 1-[benzofuran-2-yl-(4-alkyl/aryl-phenyl)-methyl]- 1 H-triazoles. 1-[benzofuran-2-yl-(4-alkyl/aryl-phenyl)-methyl]- 1 h-triazoles 53-116 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 22-29 20513361-0 2010 The relative importance of CYP26A1 in hepatic clearance of all-trans retinoic acid. Tretinoin 69-82 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 27-34 20606468-1 2011 AIM: The cytochrome P450 enzymes of the CYP26 family are involved in the catabolism of the biologically active retinoid all-trans-retinoic acid (atRA). Retinoids 111-119 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 40-45 20606468-1 2011 AIM: The cytochrome P450 enzymes of the CYP26 family are involved in the catabolism of the biologically active retinoid all-trans-retinoic acid (atRA). Tretinoin 120-143 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 40-45 20606468-1 2011 AIM: The cytochrome P450 enzymes of the CYP26 family are involved in the catabolism of the biologically active retinoid all-trans-retinoic acid (atRA). Tretinoin 145-149 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 40-45 20606468-2 2011 Since it is possible that an increased local CYP26 activity would reduce the effects of retinoids in vascular injury, we investigated the role of CYP26 in the regulation of atRA levels in human aortic smooth muscle cells (AOSMCs). Tretinoin 173-177 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 146-151 20606468-7 2011 The CYP26 inhibitor also induced expression of atRA-responsive genes. Tretinoin 47-51 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-9 20606468-8 2011 Therefore, atRA-induced CYP26 expression accelerated atRA inactivation in AOSMCs, giving rise to an atRA-CYP26 feedback loop. Tretinoin 11-15 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 24-29 20606468-8 2011 Therefore, atRA-induced CYP26 expression accelerated atRA inactivation in AOSMCs, giving rise to an atRA-CYP26 feedback loop. Tretinoin 11-15 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 105-110 20606468-8 2011 Therefore, atRA-induced CYP26 expression accelerated atRA inactivation in AOSMCs, giving rise to an atRA-CYP26 feedback loop. Tretinoin 53-57 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 24-29 20606468-8 2011 Therefore, atRA-induced CYP26 expression accelerated atRA inactivation in AOSMCs, giving rise to an atRA-CYP26 feedback loop. Tretinoin 53-57 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 105-110 20606468-9 2011 Inhibition of this loop with a CYP26 inhibitor increased retinoid signaling. Retinoids 57-65 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 31-36 20513361-2 2010 Several P450 enzymes including CYP26A1, CYP2C8, and CYP3A4 have been proposed to be responsible for RA clearance in the liver but their quantitative importance has not been demonstrated. Tretinoin 100-102 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 31-38 20513361-6 2010 CYP2C8, CYP3A4, CYP3A5 and CYP3A7 metabolized RA with unbound K(m) values of 3.4-7.2microM and V(max) values of 2.3-4.9pmol/min/pmol P450, but were less efficient than CYP26A1 in clearing RA. Tretinoin 46-48 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 168-175 20513361-7 2010 Simulations performed for livers with varying P450 expression levels over a range of RA concentrations demonstrated that at both endogenous and therapeutic concentrations of RA, CYP26A1 is the primary enzyme responsible for 4-OH RA formation clearance. Tretinoin 85-87 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 178-185 20513361-7 2010 Simulations performed for livers with varying P450 expression levels over a range of RA concentrations demonstrated that at both endogenous and therapeutic concentrations of RA, CYP26A1 is the primary enzyme responsible for 4-OH RA formation clearance. Tretinoin 174-176 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 178-185 20513361-7 2010 Simulations performed for livers with varying P450 expression levels over a range of RA concentrations demonstrated that at both endogenous and therapeutic concentrations of RA, CYP26A1 is the primary enzyme responsible for 4-OH RA formation clearance. 4-oh ra 224-231 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 178-185 20513361-8 2010 HLM incubation data showed that 4-OH RA formation velocity varied from 0.2 to 15.3pmol/min/mg microsomal protein and velocity in HLMs was significantly correlated (p<0.01) to CYP26A1, CYP3A4, and CYP3A5 protein content, but not to CYP2C8. 4-oh 32-36 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 178-185 20682464-0 2010 Multiple retinoic acid response elements cooperate to enhance the inducibility of CYP26A1 gene expression in liver. Tretinoin 9-22 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 82-89 20682464-1 2010 CYP26A1, which catalyzes the oxidation of all-trans (at)-retinoic acid (RA), is induced moderately by RA in numerous tissues, but is highly responsive in liver. Tretinoin 52-70 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 20682464-1 2010 CYP26A1, which catalyzes the oxidation of all-trans (at)-retinoic acid (RA), is induced moderately by RA in numerous tissues, but is highly responsive in liver. Tretinoin 72-74 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 20682464-5 2010 In DNA binding and chromatin immunoprecipitation assays, the binding of RARs to the proximal RARE1 and distal RARE2, -3, and -4 regions of the CYP26A1 promoter was increased in RA-treated HepG2 cells, and greater in VA-sufficient than VA-deficient liver. Vitamin A 216-218 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 143-150 20682464-7 2010 The results support a cooperative model in which the functioning of multiple RAREs may account for the strong inducibility of CYP26A1 in liver, which, in turn, may be important physiologically for restoring retinoid homeostasis when the concentration of RA rises. Retinoids 207-215 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 126-133 20682464-7 2010 The results support a cooperative model in which the functioning of multiple RAREs may account for the strong inducibility of CYP26A1 in liver, which, in turn, may be important physiologically for restoring retinoid homeostasis when the concentration of RA rises. Tretinoin 77-79 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 126-133 20682464-1 2010 CYP26A1, which catalyzes the oxidation of all-trans (at)-retinoic acid (RA), is induced moderately by RA in numerous tissues, but is highly responsive in liver. Tretinoin 102-104 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 20513361-8 2010 HLM incubation data showed that 4-OH RA formation velocity varied from 0.2 to 15.3pmol/min/mg microsomal protein and velocity in HLMs was significantly correlated (p<0.01) to CYP26A1, CYP3A4, and CYP3A5 protein content, but not to CYP2C8. Tretinoin 37-39 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 178-185 20513361-10 2010 These findings suggest that CYP26A1 is the P450 isoform that should be targeted when designing RA metabolism blocking agents. Tretinoin 95-97 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 28-35 20409737-1 2010 CONTEXT: Cytochrome P450 oxidoreductase (POR) is an electron donor for all microsomal P450 enzymes including CYP26 involved in inactivation of all-trans retinoic acid (atRA). Tretinoin 153-166 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 109-114 20658618-6 2010 RESULTS: Treatment of NB cells with retinoids induced expression of several genes including the retinoid metabolizing enzymes CYP26A1 and CYP26B1. Retinoids 36-45 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 126-133 20658618-6 2010 RESULTS: Treatment of NB cells with retinoids induced expression of several genes including the retinoid metabolizing enzymes CYP26A1 and CYP26B1. Retinoids 36-44 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 126-133 20409737-1 2010 CONTEXT: Cytochrome P450 oxidoreductase (POR) is an electron donor for all microsomal P450 enzymes including CYP26 involved in inactivation of all-trans retinoic acid (atRA). Tretinoin 168-172 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 109-114 19884280-7 2010 Classic cytochrome P450 inducers did not affect CYP26 transcription, whereas the peroxisome proliferator-activated receptor (PPAR) gamma agonists pioglitazone and rosiglitazone up-regulated CYP26B1 transcription by as much as 209- +/- 80-fold and CYP26A1 by 10-fold. Pioglitazone 146-158 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 247-254 19935721-2 2010 Here, we found that CYP26A1, the gene encoding a cytochrome P450 enzyme specifically involved in metabolic inactivation of retinoic acid (RA), the most active vitamin A derivative, is highly expressed in 42% (27/65) of primary breast cancers. Tretinoin 123-136 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 20-27 19935721-2 2010 Here, we found that CYP26A1, the gene encoding a cytochrome P450 enzyme specifically involved in metabolic inactivation of retinoic acid (RA), the most active vitamin A derivative, is highly expressed in 42% (27/65) of primary breast cancers. Tretinoin 138-140 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 20-27 19935721-2 2010 Here, we found that CYP26A1, the gene encoding a cytochrome P450 enzyme specifically involved in metabolic inactivation of retinoic acid (RA), the most active vitamin A derivative, is highly expressed in 42% (27/65) of primary breast cancers. Vitamin A 159-168 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 20-27 19935721-5 2010 Suppression of CYP26A1 significantly reversed the CYP26A1-mediated oncogenic characteristics, suggesting a direct link between intracellular RA status and tumorigenicity. Tretinoin 141-143 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 15-22 19935721-5 2010 Suppression of CYP26A1 significantly reversed the CYP26A1-mediated oncogenic characteristics, suggesting a direct link between intracellular RA status and tumorigenicity. Tretinoin 141-143 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 50-57 20200558-6 2010 Co-treatment with a pharmacologic inhibitor of PRKCD and RA resulted in the induction of an RA responsive reporter construct, as well as the endogenous RA target genes, CEBPE, CYP26A1 and RIG-I. Tretinoin 57-59 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 176-183 19884280-7 2010 Classic cytochrome P450 inducers did not affect CYP26 transcription, whereas the peroxisome proliferator-activated receptor (PPAR) gamma agonists pioglitazone and rosiglitazone up-regulated CYP26B1 transcription by as much as 209- +/- 80-fold and CYP26A1 by 10-fold. Rosiglitazone 163-176 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 247-254 19294396-1 2009 The biosynthesis of retinoic acid (RA) from retinol is controlled by several enzymes, e.g. dehydrogenases (RalDH2, RoDH-4) and retinol-esterifying enzyme (LRAT), whereas its degradation mainly involves CYP26 enzymes. Vitamin A 44-51 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 202-207 19853565-8 2009 Concomitant regulation of Cyp26a1 expression, restraining retinoic acid signaling away from the posterior growth zone, may likewise play a role in timing the trunk-tail transition. Tretinoin 58-71 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 26-33 19294396-0 2009 Both all-trans retinoic acid and cytochrome P450 (CYP26) inhibitors affect the expression of vitamin A metabolizing enzymes and retinoid biomarkers in organotypic epidermis. Vitamin A 93-102 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 50-55 19294396-0 2009 Both all-trans retinoic acid and cytochrome P450 (CYP26) inhibitors affect the expression of vitamin A metabolizing enzymes and retinoid biomarkers in organotypic epidermis. Retinoids 128-136 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 50-55 19294396-1 2009 The biosynthesis of retinoic acid (RA) from retinol is controlled by several enzymes, e.g. dehydrogenases (RalDH2, RoDH-4) and retinol-esterifying enzyme (LRAT), whereas its degradation mainly involves CYP26 enzymes. Tretinoin 20-33 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 202-207 19294396-1 2009 The biosynthesis of retinoic acid (RA) from retinol is controlled by several enzymes, e.g. dehydrogenases (RalDH2, RoDH-4) and retinol-esterifying enzyme (LRAT), whereas its degradation mainly involves CYP26 enzymes. Tretinoin 35-37 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 202-207 19294396-3 2009 Here, we examined the effects of RA and the CYP26 inhibitors, liarozole and talarozole, on retinoid metabolism and RA-regulated genes in organotypic epidermis. Retinoids 91-99 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 44-49 19294396-4 2009 RA induced the expression of CYP26 enzymes already after 8 h, whereas LRAT exhibited a later response and peaked at 48 h, indicating a feedback induction of retinol esterification. Tretinoin 0-2 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 29-34 19294396-8 2009 Cellular accumulation of exogenous [3H]RA was higher after talarozole than after liarozole, probably indicating a greater CYP26-inhibitory potency of the former drug. Tritium 36-38 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 122-127 19294396-8 2009 Cellular accumulation of exogenous [3H]RA was higher after talarozole than after liarozole, probably indicating a greater CYP26-inhibitory potency of the former drug. Tretinoin 39-41 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 122-127 19294396-9 2009 The present study shows that CYP26A1 expression is extremely sensitive to both exogenous RA and increased endogenous RA levels, i.e. due to CYP26 inhibition, and thus an excellent biomarker for retinoid signalling in organotypic epidermis. Tretinoin 89-91 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 29-36 19294396-9 2009 The present study shows that CYP26A1 expression is extremely sensitive to both exogenous RA and increased endogenous RA levels, i.e. due to CYP26 inhibition, and thus an excellent biomarker for retinoid signalling in organotypic epidermis. Tretinoin 89-91 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 29-34 19294396-9 2009 The present study shows that CYP26A1 expression is extremely sensitive to both exogenous RA and increased endogenous RA levels, i.e. due to CYP26 inhibition, and thus an excellent biomarker for retinoid signalling in organotypic epidermis. Tretinoin 117-119 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 29-36 19294396-9 2009 The present study shows that CYP26A1 expression is extremely sensitive to both exogenous RA and increased endogenous RA levels, i.e. due to CYP26 inhibition, and thus an excellent biomarker for retinoid signalling in organotypic epidermis. Tretinoin 117-119 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 29-34 19294396-9 2009 The present study shows that CYP26A1 expression is extremely sensitive to both exogenous RA and increased endogenous RA levels, i.e. due to CYP26 inhibition, and thus an excellent biomarker for retinoid signalling in organotypic epidermis. Retinoids 194-202 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 29-36 19294396-9 2009 The present study shows that CYP26A1 expression is extremely sensitive to both exogenous RA and increased endogenous RA levels, i.e. due to CYP26 inhibition, and thus an excellent biomarker for retinoid signalling in organotypic epidermis. Retinoids 194-202 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 29-34 19519282-4 2009 The CYP26 enzymes have been shown to metabolize RA efficiently and they are also inducible by RA in selected systems. Tretinoin 94-96 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-9 19519282-4 2009 The CYP26 enzymes have been shown to metabolize RA efficiently and they are also inducible by RA in selected systems. Tretinoin 48-50 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-9 19519282-0 2009 The role of CYP26 enzymes in retinoic acid clearance. Tretinoin 29-42 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 12-17 19519282-8 2009 To further the understanding of how CYP26 enzymes contribute to the regulation of RA homeostasis, structural information of the CYP26s, commercially available recombinant enzymes and good specific and sensitive antibodies are needed. Tretinoin 82-84 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 36-41 19519282-3 2009 It has been proposed that cytochrome P450 family 26 (CYP26) enzymes have a role in determining the cellular exposure to RA by inactivating RA in cells that do not need RA. Tretinoin 120-122 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 26-51 19519282-3 2009 It has been proposed that cytochrome P450 family 26 (CYP26) enzymes have a role in determining the cellular exposure to RA by inactivating RA in cells that do not need RA. Tretinoin 120-122 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 53-58 19147277-8 2009 The effect of EBBP on retinoid-responsive transcription appeared to be limited to genes with the retinoic acid response element (betaRARE) regulatory sequence, such as CYP26A1. Retinoids 22-30 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 168-175 19519282-3 2009 It has been proposed that cytochrome P450 family 26 (CYP26) enzymes have a role in determining the cellular exposure to RA by inactivating RA in cells that do not need RA. Tretinoin 139-141 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 26-51 19519282-3 2009 It has been proposed that cytochrome P450 family 26 (CYP26) enzymes have a role in determining the cellular exposure to RA by inactivating RA in cells that do not need RA. Tretinoin 139-141 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 53-58 19519282-3 2009 It has been proposed that cytochrome P450 family 26 (CYP26) enzymes have a role in determining the cellular exposure to RA by inactivating RA in cells that do not need RA. Tretinoin 139-141 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 26-51 19519282-3 2009 It has been proposed that cytochrome P450 family 26 (CYP26) enzymes have a role in determining the cellular exposure to RA by inactivating RA in cells that do not need RA. Tretinoin 139-141 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 53-58 19147277-8 2009 The effect of EBBP on retinoid-responsive transcription appeared to be limited to genes with the retinoic acid response element (betaRARE) regulatory sequence, such as CYP26A1. Tretinoin 97-110 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 168-175 19171200-1 2009 All-trans retinoic acid (RA) levels are controlled by enzymes of the vitamin A metabolism (RDH16, RalDH2, and LRAT) and RA catabolism (CYP26 and CYP2S1). 2-octenal 4-9 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 135-140 18608743-4 2009 The benzooxazol-2-yl-[phenyl-imidazol-1-yl-methyl)phenyl]amines with small substituents in the phenyl ring were moderately potent CYP26A1 inhibitors (IC(50) 8 and 12 microM) and comparable with liarozole (IC(50) 7 microM). benzooxazol-2-yl-[phenyl-imidazol-1-yl-methyl)phenyl]amines 4-63 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 130-137 18608743-1 2009 The design of N-phenylbenzo[d]oxazolamines as CYP26A1 inhibitors involved ligand docking experiments using molecular modeling (FlexX) and analysis of ligand interactions at the binding domain. n-phenylbenzo[d]oxazolamines 14-42 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 46-53 19171200-7 2009 Thus CYP26B1 appears essential for RA catabolism under physiological conditions, whereas CYP26A1 might play a greater role during RA excess. Tretinoin 130-132 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 89-96 19171200-1 2009 All-trans retinoic acid (RA) levels are controlled by enzymes of the vitamin A metabolism (RDH16, RalDH2, and LRAT) and RA catabolism (CYP26 and CYP2S1). Tretinoin 10-23 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 135-140 19171200-1 2009 All-trans retinoic acid (RA) levels are controlled by enzymes of the vitamin A metabolism (RDH16, RalDH2, and LRAT) and RA catabolism (CYP26 and CYP2S1). Tretinoin 25-27 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 135-140 19171200-4 2009 RA (1 microM) induced CYP26A1, CYP26B1, CYP2S1, CRABPII and LRAT mRNA. Tretinoin 0-2 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 22-29 19171200-5 2009 The CYP26 inhibitor talarozole altered CYP26A1 and LRAT mRNA expression in a similar way as RA, increased the cellular accumulation of [(3)H]RA, and induced a punctate CRABPII staining, also observed after siRNA knock-down of CYP26B1 (but not after RA exposure). R 115866 20-30 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-9 19171200-5 2009 The CYP26 inhibitor talarozole altered CYP26A1 and LRAT mRNA expression in a similar way as RA, increased the cellular accumulation of [(3)H]RA, and induced a punctate CRABPII staining, also observed after siRNA knock-down of CYP26B1 (but not after RA exposure). R 115866 20-30 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 39-46 19120344-0 2009 The CYP26 inhibitor R115866 potentiates the effects of all-trans retinoic acid on cultured human epidermal keratinocytes. R 115866 20-27 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-9 19120344-4 2009 R115866 is a specific inhibitor of the cytochrome P450 isoform CYP26, which is involved in the metabolic inactivation pathway of RA. Tretinoin 129-131 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 63-68 19171200-5 2009 The CYP26 inhibitor talarozole altered CYP26A1 and LRAT mRNA expression in a similar way as RA, increased the cellular accumulation of [(3)H]RA, and induced a punctate CRABPII staining, also observed after siRNA knock-down of CYP26B1 (but not after RA exposure). Tretinoin 52-54 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-9 18992717-3 2009 Enzymes in the CYP26 family are thought to be responsible for the elimination of RA, and CYP26A1 appears to serve the most critical functions in this family. Tretinoin 81-83 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 89-96 19120344-0 2009 The CYP26 inhibitor R115866 potentiates the effects of all-trans retinoic acid on cultured human epidermal keratinocytes. all-trans 55-64 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-9 19120344-0 2009 The CYP26 inhibitor R115866 potentiates the effects of all-trans retinoic acid on cultured human epidermal keratinocytes. Tretinoin 65-78 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-9 19100254-6 2009 We further show that fibroblast growth factor (FGF) signalling, induces the expression of the retinoic acid degrading enzyme cytochrome P450 (cyp) 26a1, and that one of its products, 4-oxo-RA, mimics the action of the RARalpha agonist in the differentiation of the NPCs into cholinergic neurons. Tretinoin 94-107 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 125-151 18992717-6 2009 Heme incorporation was determined by carbon monoxide difference spectrum and a type 1 spectrum was observed with RA binding to CYP26A1. Heme 0-4 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 127-134 18992717-6 2009 Heme incorporation was determined by carbon monoxide difference spectrum and a type 1 spectrum was observed with RA binding to CYP26A1. Tretinoin 113-115 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 127-134 18992717-7 2009 We found that RA is a tight binding ligand of CYP26A1 with low nM binding affinity. Tretinoin 14-16 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 46-53 18992717-8 2009 CYP26A1 oxidized RA efficiently (depletion K(m) 9.4+/-3.3nM and V(max) 11.3+/-4.3pmolesmin(-1)pmoleP450(-1)) when supplemented with P450 oxidoreductase and NADPH but was independent of cytochrome b5. Tretinoin 17-19 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 18992717-8 2009 CYP26A1 oxidized RA efficiently (depletion K(m) 9.4+/-3.3nM and V(max) 11.3+/-4.3pmolesmin(-1)pmoleP450(-1)) when supplemented with P450 oxidoreductase and NADPH but was independent of cytochrome b5. NADP 156-161 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 18990705-7 2009 We also show that lutein radical cation formation in CP26 is dependent on binding of zeaxanthin to the L2 domain, implying that zeaxanthin acts as an allosteric effector of charge transfer quenching involving lutein in the L1 domain. Zeaxanthins 85-95 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 53-57 18992717-10 2009 4-OH-RA was further metabolized by CYP26A1 to more polar metabolites and this sequential metabolism of RA occurred in part without 4-OH-RA leaving the active site of CYP26A1. 4-oh-ra 0-7 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 35-42 18992717-10 2009 4-OH-RA was further metabolized by CYP26A1 to more polar metabolites and this sequential metabolism of RA occurred in part without 4-OH-RA leaving the active site of CYP26A1. 4-oh-ra 0-7 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 166-173 18992717-10 2009 4-OH-RA was further metabolized by CYP26A1 to more polar metabolites and this sequential metabolism of RA occurred in part without 4-OH-RA leaving the active site of CYP26A1. Tretinoin 5-7 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 35-42 18992717-10 2009 4-OH-RA was further metabolized by CYP26A1 to more polar metabolites and this sequential metabolism of RA occurred in part without 4-OH-RA leaving the active site of CYP26A1. Tretinoin 5-7 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 166-173 18992717-11 2009 The high efficiency of CYP26A1 in eliminating both RA and its potentially active metabolites supports the major role of this enzyme in regulating RA clearance in vivo. Tretinoin 51-53 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 23-30 18992717-11 2009 The high efficiency of CYP26A1 in eliminating both RA and its potentially active metabolites supports the major role of this enzyme in regulating RA clearance in vivo. Tretinoin 146-148 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 23-30 19016711-0 2009 Topical treatment with CYP26 inhibitor talarozole (R115866) dose dependently alters the expression of retinoid-regulated genes in normal human epidermis. Retinoids 102-110 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 23-28 19197536-6 2009 A significant decrease in the mRNA expression of KRT2 and TNF-alpha, and trends toward increased expression of KRT4 and CYP26A1 were observed in liarozole-treated patients, consistent with an increased retinoid stimulation of epidermis. liarozole 145-154 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 120-127 19016711-6 2009 RESULTS: Talarozole treatment increased the mRNA expression of CRABP2, KRT4, CYP26A1 and CYP26B1 dose dependently, and decreased the expression of KRT2 and IL-1alpha compared with vehicle-treated skin. R 115866 9-19 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 77-84 19016711-0 2009 Topical treatment with CYP26 inhibitor talarozole (R115866) dose dependently alters the expression of retinoid-regulated genes in normal human epidermis. R 115866 39-49 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 23-28 18927157-8 2008 Together with data on the expression of osteoblast markers, our results indicate that temporal and spatial restriction of RA signaling by Cyp26 enzymes is required to attenuate osteoblast maturation and/or activity in vivo. Tretinoin 122-124 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 138-143 18722776-0 2008 Novel azolyl-(phenylmethyl)]aryl/heteroarylamines: potent CYP26 inhibitors and enhancers of all-trans retinoic acid activity in neuroblastoma cells. azolyl-(phenylmethyl)]aryl/heteroarylamines 6-49 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 58-63 18769122-8 2008 Several retinoid-regulated genes exhibited much higher mRNA levels in SKRC06 than in SKRC39, even in the absence of drugs; these included crabp2, rargamma and cyp26A1. Retinoids 8-16 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 159-166 18816852-3 2008 These have shown local control of synthesis and degradation, and computational models suggest that degradation by the Cyp26 enzymes plays a critical role in the formation of a morphogen gradient as well as its ability to compensate for fluctuations in RA levels. Radium 252-254 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 118-123 18722776-1 2008 The synthesis and potent inhibitory activity of novel 4-[(imidazol-1-yl and triazol-1-yl)(phenyl)methyl]aryl-and heteroaryl amines versus a MCF-7 CYP26A1 cell assay is described. 4-[(imidazol-1-yl and triazol-1-yl)(phenyl)methyl]aryl-and heteroaryl amines 54-130 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 146-153 18606996-8 2008 These extracts contained retinoids (5.4 nM) as assessed by RA-inducible Cyp26A1-promoter luciferase reporter cell lines. Retinoids 25-34 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 72-79 18606996-8 2008 These extracts contained retinoids (5.4 nM) as assessed by RA-inducible Cyp26A1-promoter luciferase reporter cell lines. Radium 59-61 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 72-79 18502188-6 2008 And during P19 cell neural differentiation induced by all trans-retinoic acid (ATRA) and cell aggregate formation, RALDH1, RALDH2, CYP26A1, and CYP26B1 could be notably upregulated by ATRA, and keeping the high-level expression of RALDH1 and RALDH2 was dependent on the further neural differentiation, but not continuous ATRA induction. Tretinoin 58-77 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 131-138 18502188-6 2008 And during P19 cell neural differentiation induced by all trans-retinoic acid (ATRA) and cell aggregate formation, RALDH1, RALDH2, CYP26A1, and CYP26B1 could be notably upregulated by ATRA, and keeping the high-level expression of RALDH1 and RALDH2 was dependent on the further neural differentiation, but not continuous ATRA induction. Tretinoin 184-188 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 131-138 18059332-9 2008 CYP26A1 expression was increased synergistically by RA and lithocholic acid (P<0.05). Tretinoin 52-54 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 26621242-0 2008 Homology Models and Molecular Modeling of Human Retinoic Acid Metabolizing Enzymes Cytochrome P450 26A1 (CYP26A1) and P450 26B1 (CYP26B1). Tretinoin 48-61 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 83-103 26621242-0 2008 Homology Models and Molecular Modeling of Human Retinoic Acid Metabolizing Enzymes Cytochrome P450 26A1 (CYP26A1) and P450 26B1 (CYP26B1). Tretinoin 48-61 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 105-112 26621242-6 2008 Residues involved in hydrophobic interactions with atRA were Pro113, Phe222, Phe299, Val370, Pro371, and Phe374 in CYP26A1 and Leu88, Pro118, Phe222, Phe295, Ile368, and Tyr272 in CYP26B1. Tretinoin 51-55 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 115-122 26621242-7 2008 Hydrogen bonding interactions were observed between the atRA carboxylate group and Arg 90 in CYP26A1 and with Arg76, Arg95, and Ser369 in CYP26B1. Hydrogen 0-8 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 93-100 26621242-7 2008 Hydrogen bonding interactions were observed between the atRA carboxylate group and Arg 90 in CYP26A1 and with Arg76, Arg95, and Ser369 in CYP26B1. atra carboxylate 56-72 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 93-100 26621242-7 2008 Hydrogen bonding interactions were observed between the atRA carboxylate group and Arg 90 in CYP26A1 and with Arg76, Arg95, and Ser369 in CYP26B1. Arginine 83-86 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 93-100 18059332-0 2008 A novel role for the retinoic acid-catabolizing enzyme CYP26A1 in Barrett"s associated adenocarcinoma. Tretinoin 21-34 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 55-62 18059332-1 2008 Vitamin A deficiency is associated with carcinogenesis, and upregulation of CYP26A1, a major retinoic acid (RA)-catabolizing enzyme, has recently been shown in cancer. Tretinoin 93-106 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 76-83 18059332-1 2008 Vitamin A deficiency is associated with carcinogenesis, and upregulation of CYP26A1, a major retinoic acid (RA)-catabolizing enzyme, has recently been shown in cancer. Tretinoin 108-110 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 76-83 18059332-9 2008 CYP26A1 expression was increased synergistically by RA and lithocholic acid (P<0.05). Lithocholic Acid 59-75 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 18059332-7 2008 CYP26A1 was stably overexpressed in GihTERT cells, which were evaluated for gene-expression changes (pathway array and quantitative RT-PCR), cellular proliferation (cytometric DNA profile and colorimetric assay) and invasion (in vitro matrigel assay) with or without the CYP inhibitor ketaconazole. ketaconazole 285-297 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 18059332-9 2008 CYP26A1 expression was increased synergistically by RA and lithocholic acid (P<0.05). p< 77-81 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 18059332-8 2008 RA levels decreased progressively with the degree of dysplasia (P<0.05) and were inversely correlated with CYP26A1 gene levels and activity (P<0.01). Tretinoin 0-2 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 110-117 18059332-11 2008 Functional effects of CYP26A1 overexpression were increased proliferation (P<0.01) and invasion in vitro (P<0.01), which were inhibited by ketaconazole. ketaconazole 145-157 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 22-29 18059332-12 2008 Overexpression of CYP26A1 causes intracellular RA depletion and drives the cell into a highly proliferative and invasive state with induction of other known oncogenes. Tretinoin 47-49 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 18-25 18318834-5 2008 We postulate that nonphotochemical quenching occurs within a structural locus comprising the PsbS subunit and components of the light-harvesting antenna, CP26, CP24, CP29 and LHCIIb (the major trimeric light-harvesting complex), formed in response to protonation and controlled by the xanthophyll cycle carotenoids. Xanthophylls 285-296 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 154-158 18241852-3 2008 CYP26a1(-/-) ES cells had a 11.0+/-3.2-fold higher intracellular RA concentration than Wt ES cells after RA treatment for 48 h. RA-treated CYP26A1(-/-) ES cells exhibited 2-3 fold higher mRNA levels of Hoxa1, a primary RA target gene, than Wt ES cells. Tretinoin 105-107 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 139-146 18241852-3 2008 CYP26a1(-/-) ES cells had a 11.0+/-3.2-fold higher intracellular RA concentration than Wt ES cells after RA treatment for 48 h. RA-treated CYP26A1(-/-) ES cells exhibited 2-3 fold higher mRNA levels of Hoxa1, a primary RA target gene, than Wt ES cells. Tretinoin 105-107 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 18241852-3 2008 CYP26a1(-/-) ES cells had a 11.0+/-3.2-fold higher intracellular RA concentration than Wt ES cells after RA treatment for 48 h. RA-treated CYP26A1(-/-) ES cells exhibited 2-3 fold higher mRNA levels of Hoxa1, a primary RA target gene, than Wt ES cells. Tretinoin 105-107 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 139-146 18241852-4 2008 Despite increased intracellular RA levels, CYP26a1(-/-) ES cells were more resistant than Wt ES cells to RA-induced proliferation arrest. Tretinoin 105-107 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 43-50 18241852-6 2008 Microarray analyses revealed that RA-treated CYP26A1(-/-) ES cells exhibited lower mRNA levels than Wt ES cells for genes involved in differentiation, particularly in neural (Epha4, Pmp22, Nrp1, Gap43, Ndn) and smooth muscle differentiation (Madh3, Nrp1, Tagln Calponin, Caldesmon1). Tretinoin 34-36 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 45-52 18241852-7 2008 In contrast, genes involved in the stress response (e.g. Tlr2, Stk2, Fcgr2b, Bnip3, Pdk1) were expressed at higher levels in CYP26A1(-/-) than in Wt ES cells without RA. Tretinoin 166-168 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 125-132 18241852-8 2008 Collectively, our results show that CYP26A1 activity regulates intracellular RA levels, cell proliferation, transcriptional regulation of primary RA target genes, and ES cell differentiation to parietal endoderm. Tretinoin 77-79 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 36-43 18241852-0 2008 CYP26A1 knockout embryonic stem cells exhibit reduced differentiation and growth arrest in response to retinoic acid. Tretinoin 103-116 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 18241852-1 2008 CYP26A1, a cytochrome P450 enzyme, metabolizes all-trans-retinoic acid (RA) into polar metabolites, e.g. 4-oxo-RA and 4-OH-RA. Tretinoin 47-70 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 18241852-1 2008 CYP26A1, a cytochrome P450 enzyme, metabolizes all-trans-retinoic acid (RA) into polar metabolites, e.g. 4-oxo-RA and 4-OH-RA. Tretinoin 72-74 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 18241852-1 2008 CYP26A1, a cytochrome P450 enzyme, metabolizes all-trans-retinoic acid (RA) into polar metabolites, e.g. 4-oxo-RA and 4-OH-RA. 4-oxoretinoic acid 105-113 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 18241852-1 2008 CYP26A1, a cytochrome P450 enzyme, metabolizes all-trans-retinoic acid (RA) into polar metabolites, e.g. 4-oxo-RA and 4-OH-RA. 4-oh-ra 118-125 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 18241852-3 2008 CYP26a1(-/-) ES cells had a 11.0+/-3.2-fold higher intracellular RA concentration than Wt ES cells after RA treatment for 48 h. RA-treated CYP26A1(-/-) ES cells exhibited 2-3 fold higher mRNA levels of Hoxa1, a primary RA target gene, than Wt ES cells. Tretinoin 65-67 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 18241852-3 2008 CYP26a1(-/-) ES cells had a 11.0+/-3.2-fold higher intracellular RA concentration than Wt ES cells after RA treatment for 48 h. RA-treated CYP26A1(-/-) ES cells exhibited 2-3 fold higher mRNA levels of Hoxa1, a primary RA target gene, than Wt ES cells. Tretinoin 65-67 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 139-146 18241852-3 2008 CYP26a1(-/-) ES cells had a 11.0+/-3.2-fold higher intracellular RA concentration than Wt ES cells after RA treatment for 48 h. RA-treated CYP26A1(-/-) ES cells exhibited 2-3 fold higher mRNA levels of Hoxa1, a primary RA target gene, than Wt ES cells. Tretinoin 105-107 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 18318834-5 2008 We postulate that nonphotochemical quenching occurs within a structural locus comprising the PsbS subunit and components of the light-harvesting antenna, CP26, CP24, CP29 and LHCIIb (the major trimeric light-harvesting complex), formed in response to protonation and controlled by the xanthophyll cycle carotenoids. Carotenoids 303-314 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 154-158 17998248-10 2008 The loss of the forebrain-specific RA-degrading enzyme cyp26a1 causes a forebrain phenotype that mimics tgif morphants. Tretinoin 35-37 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 55-62 18093970-7 2008 Moreover, in the case of excessive maternal dietary vitamin A intake, LRAT acts together with Cyp26A1, one of the enzymes that catalyze the degradation of retinoic acid, and possibly with STRA6, the recently identified cell surface receptor for retinol-RBP, in maintaining adequate levels of retinoids in embryonic and extraembryonic tissues. Vitamin A 52-61 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 94-101 18093970-7 2008 Moreover, in the case of excessive maternal dietary vitamin A intake, LRAT acts together with Cyp26A1, one of the enzymes that catalyze the degradation of retinoic acid, and possibly with STRA6, the recently identified cell surface receptor for retinol-RBP, in maintaining adequate levels of retinoids in embryonic and extraembryonic tissues. Tretinoin 155-168 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 94-101 17663992-0 2007 Retinoid regulated association of transcriptional co-regulators and the polycomb group protein SUZ12 with the retinoic acid response elements of Hoxa1, RARbeta(2), and Cyp26A1 in F9 embryonal carcinoma cells. Retinoids 0-8 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 168-175 18050373-6 2007 The CYP26 gene, which plays a key role in retinol metabolism, was examined to define any single-nucleotide polymorphisms (SNP) associations with AS. Vitamin A 42-49 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-9 17850797-0 2007 A specific binding site for neoxanthin in the monomeric antenna proteins CP26 and CP29 of Photosystem II. neoxanthin 28-38 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 73-77 17850797-1 2007 The location of the neoxanthin binding site in CP26 and CP29 was investigated by site-directed mutagenesis. neoxanthin 20-30 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 47-51 17663992-0 2007 Retinoid regulated association of transcriptional co-regulators and the polycomb group protein SUZ12 with the retinoic acid response elements of Hoxa1, RARbeta(2), and Cyp26A1 in F9 embryonal carcinoma cells. Tretinoin 110-123 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 168-175 17714122-1 2007 BACKGROUND: R115866 (Rambazole) is a new generation all-trans retinoic acid metabolism blocking agent, highly specific against the retinoic acid 4-hydroxylase. R 115866 21-30 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 131-158 17714122-1 2007 BACKGROUND: R115866 (Rambazole) is a new generation all-trans retinoic acid metabolism blocking agent, highly specific against the retinoic acid 4-hydroxylase. Tretinoin 62-75 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 131-158 17526768-8 2007 Lastly, treatment of LNCaP with all-trans retinal (RAL) in the presence of DHT resulted in significant up-regulation of the RA-inducible, RA-metabolizing enzyme CYP26A1 mRNA compared with RAL treatment alone. Dihydrotestosterone 75-78 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 161-168 17510468-0 2007 CYP26 inhibitor R115866 increases retinoid signaling in intimal smooth muscle cells. R 115866 16-23 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 17510468-0 2007 CYP26 inhibitor R115866 increases retinoid signaling in intimal smooth muscle cells. Retinoids 34-42 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 17510468-5 2007 A key enzyme is the all-trans retinoic acid-degrading enzyme cytochrome p450 isoform 26 (CYP26). Tretinoin 20-43 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 61-87 17510468-5 2007 A key enzyme is the all-trans retinoic acid-degrading enzyme cytochrome p450 isoform 26 (CYP26). Tretinoin 20-43 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 89-94 17510468-6 2007 Thus, an alternative approach to exogenous retinoid administration could be to increase the intracellular level of all-trans retinoic acid by blocking CYP26-mediated degradation of retinoids. Retinoids 43-51 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 151-156 17510468-6 2007 Thus, an alternative approach to exogenous retinoid administration could be to increase the intracellular level of all-trans retinoic acid by blocking CYP26-mediated degradation of retinoids. 2-octenal 119-124 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 151-156 17510468-6 2007 Thus, an alternative approach to exogenous retinoid administration could be to increase the intracellular level of all-trans retinoic acid by blocking CYP26-mediated degradation of retinoids. Tretinoin 125-138 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 151-156 17510468-6 2007 Thus, an alternative approach to exogenous retinoid administration could be to increase the intracellular level of all-trans retinoic acid by blocking CYP26-mediated degradation of retinoids. Retinoids 181-190 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 151-156 17510468-8 2007 Although medial cells remained unaffected, treatment with the CYP26-inhibitor R115866 significantly increased cellular levels of all-trans retinoic acid in intimal SMCs. R 115866 78-85 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 62-67 17510468-8 2007 Although medial cells remained unaffected, treatment with the CYP26-inhibitor R115866 significantly increased cellular levels of all-trans retinoic acid in intimal SMCs. Tretinoin 139-152 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 62-67 17510468-10 2007 CONCLUSIONS: Blocking of the CYP26-mediated catabolism mimics the effects of exogenously administrated active retinoids on intimal SMCs. Retinoids 110-119 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 29-34 17070795-7 2007 It is proposed that heterogeneous local distribution of retinoids controlled through Raldh2 and Cyp26A1 is responsible for matching the fleshy and the tendinous components of each muscle belly. Retinoids 56-65 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 96-103 17276435-7 2007 Cytochrome P450 (CYP 26A1), which is responsible for retinoic acid metabolism, was highly up-regulated in leiomyomas (+5.4- +/- 0.53-fold). Tretinoin 53-66 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 17-25 17218384-0 2007 Pharmacogenomic analysis of acute promyelocytic leukemia cells highlights CYP26 cytochrome metabolism in differential all-trans retinoic acid sensitivity. Tretinoin 128-141 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 74-79 17218384-5 2007 In opposition, only high-sensitive blasts expressed the CYP26A1 gene, encoding the p450 cytochrome which is known to be involved in retinoic acid catabolism. Tretinoin 132-145 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 56-63 17218384-6 2007 In NB4 cells, ATRA treatment activates a novel signaling pathway, whereby interleukin-8 stimulates the expression of the homeobox transcription factor HOXA10v2, an effective enhancer of CYP26A1 transcription. Tretinoin 14-18 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 186-193 17244623-7 2007 Furthermore, BZLF1 expression in AGS cells is sufficient to activate DHRS9 gene expression and increases the ability of retinol to induce the RA-responsive gene, CYP26A1. Vitamin A 120-127 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 162-169 17244623-7 2007 Furthermore, BZLF1 expression in AGS cells is sufficient to activate DHRS9 gene expression and increases the ability of retinol to induce the RA-responsive gene, CYP26A1. Tretinoin 142-144 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 162-169 17460545-0 2007 The discovery of new coding alleles of human CYP26A1 that are potentially defective in the metabolism of all-trans retinoic acid and their assessment in a recombinant cDNA expression system. Tretinoin 115-128 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 45-52 17460545-3 2007 We sequenced CYP26A1 in racially diverse individuals and assessed the metabolism of retinoic acid by newly identified coding alleles of CYP26A1 in a recombinant system. Tretinoin 84-97 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 136-143 17460545-10 2007 Wild type CYP26A1 protein metabolized all-trans-retinoic acid (at-RA) to 4-oxo-RA, 4-OH-RA as well as water-soluble metabolites. Tretinoin 42-61 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 10-17 17460545-10 2007 Wild type CYP26A1 protein metabolized all-trans-retinoic acid (at-RA) to 4-oxo-RA, 4-OH-RA as well as water-soluble metabolites. Tretinoin 63-68 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 10-17 17460545-10 2007 Wild type CYP26A1 protein metabolized all-trans-retinoic acid (at-RA) to 4-oxo-RA, 4-OH-RA as well as water-soluble metabolites. 4-oxoretinoic acid 73-81 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 10-17 17460545-10 2007 Wild type CYP26A1 protein metabolized all-trans-retinoic acid (at-RA) to 4-oxo-RA, 4-OH-RA as well as water-soluble metabolites. 4-oh-ra 83-90 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 10-17 17460545-10 2007 Wild type CYP26A1 protein metabolized all-trans-retinoic acid (at-RA) to 4-oxo-RA, 4-OH-RA as well as water-soluble metabolites. Water 102-107 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 10-17 17059167-0 2006 Homology model of human retinoic acid metabolising enzyme cytochrome P450 26A1 (CYP26A1): active site architecture and ligand binding. Tretinoin 24-37 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 58-78 17164423-0 2007 Cyp26 enzymes generate the retinoic acid response pattern necessary for hindbrain development. Tretinoin 27-40 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 17164423-4 2007 Here, we show that the cytochrome p450 enzymes of the Cyp26 class, which metabolize RA into polar derivatives, function redundantly to shape RA-dependent gene-expression domains during hindbrain development. Tretinoin 84-86 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 54-59 17164423-4 2007 Here, we show that the cytochrome p450 enzymes of the Cyp26 class, which metabolize RA into polar derivatives, function redundantly to shape RA-dependent gene-expression domains during hindbrain development. Tretinoin 141-143 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 54-59 17164423-5 2007 In zebrafish embryos depleted of the orthologs of the three mammalian CYP26 genes CYP26A1, CYP26B1 and CYP26C1, the entire hindbrain expresses RA-responsive genes that are normally restricted to nested domains in the posterior hindbrain. Tretinoin 143-145 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 70-75 17164423-5 2007 In zebrafish embryos depleted of the orthologs of the three mammalian CYP26 genes CYP26A1, CYP26B1 and CYP26C1, the entire hindbrain expresses RA-responsive genes that are normally restricted to nested domains in the posterior hindbrain. Tretinoin 143-145 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 82-89 17164423-6 2007 Furthermore, we show that Cyp26 enzymes are essential for exogenous RA to rescue hindbrain patterning in RA-depleted embryos. Tretinoin 68-70 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 26-31 17164423-6 2007 Furthermore, we show that Cyp26 enzymes are essential for exogenous RA to rescue hindbrain patterning in RA-depleted embryos. Tretinoin 105-107 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 26-31 16778795-0 2006 Skin retinoid concentrations are modulated by CYP26AI expression restricted to basal keratinocytes in normal human skin and differentiated 3D skin models. Retinoids 5-13 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 46-51 16778795-9 2006 In light of our prior studies documenting the functional activity of RA metabolites, expression of CYP26 in the sebaceous gland epithelium supports the suggestion that altered RA metabolism may be involved in the pathogenesis of acne. Tretinoin 176-178 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 99-104 17059167-0 2006 Homology model of human retinoic acid metabolising enzyme cytochrome P450 26A1 (CYP26A1): active site architecture and ligand binding. Tretinoin 24-37 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 80-87 16837774-8 2006 Furthermore, the retinoic acid-catabolizing enzyme CYP26A1 is expressed at higher levels in medial SMCs compared to intimal SMCs. Tretinoin 17-30 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 51-58 16458519-0 2006 Synthesis and CYP26A1 inhibitory activity of 1-[benzofuran-2-yl-(4-alkyl/aryl-phenyl)-methyl]-1H-triazoles. 1-[benzofuran-2-yl-(4-alkyl/aryl-phenyl)-methyl]-1h-triazoles 45-106 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 14-21 16458519-3 2006 All the triazole derivatives were evaluated for CYP26A1 inhibitory activity using a MCF-7 cell-based assay. Triazoles 8-16 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 48-55 16458519-4 2006 The 4-ethyl and 4-phenyl-1,2,4-triazole derivatives displayed inhibitory activity (IC(50) 4.5 and 7 microM, respectively) comparable with that of the CYP26 inhibitor liarozole (IC(50) 7 microM). liarozole 166-175 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 150-155 16458519-5 2006 Using a CYP26A1 homology model (based on CYP3A4) template, docking experiments were performed with MOE with multiple hydrophobic interactions observed in addition to coordination between the triazole nitrogen and the haem transition metal. Triazoles 191-199 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 8-15 16458519-5 2006 Using a CYP26A1 homology model (based on CYP3A4) template, docking experiments were performed with MOE with multiple hydrophobic interactions observed in addition to coordination between the triazole nitrogen and the haem transition metal. Nitrogen 200-208 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 8-15 16933217-1 2006 BACKGROUND: CYP26A1, together with CYP26B1 and CYP26C1, are key enzymes of all-trans retinoic acid (RA) inactivation and their specific and restricted expression in developing embryos participate in the fine tuning RA levels. Tretinoin 85-98 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 12-19 16933217-1 2006 BACKGROUND: CYP26A1, together with CYP26B1 and CYP26C1, are key enzymes of all-trans retinoic acid (RA) inactivation and their specific and restricted expression in developing embryos participate in the fine tuning RA levels. Tretinoin 100-102 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 12-19 16933217-1 2006 BACKGROUND: CYP26A1, together with CYP26B1 and CYP26C1, are key enzymes of all-trans retinoic acid (RA) inactivation and their specific and restricted expression in developing embryos participate in the fine tuning RA levels. Tretinoin 215-217 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 12-19 16279770-0 2005 Novel tetralone-derived retinoic acid metabolism blocking agents: synthesis and in vitro evaluation with liver microsomal and MCF-7 CYP26A1 cell assays. Tetralones 6-15 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 132-139 16279770-0 2005 Novel tetralone-derived retinoic acid metabolism blocking agents: synthesis and in vitro evaluation with liver microsomal and MCF-7 CYP26A1 cell assays. Tretinoin 24-37 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 132-139 16279770-1 2005 The potent inhibitory activity of novel 2-benzyltetralone and 2-benzylidenetetralone derivatives vs liver microsomal retinoic acid metabolizing enzymes and a MCF-7 CYP26A1 cell assay is described. 2-benzyltetralone 40-57 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 164-171 16279770-1 2005 The potent inhibitory activity of novel 2-benzyltetralone and 2-benzylidenetetralone derivatives vs liver microsomal retinoic acid metabolizing enzymes and a MCF-7 CYP26A1 cell assay is described. 2-benzylidenetetralone 62-84 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 164-171 16279770-3 2005 In the MCF-7 CYP26A1 cell assay, the 2-(4-hydroxybenzyl)-6-methoxytetralone 5 and unsaturated benzylidene precursor 6 were found to be the most potent (IC50 = 7 and 5 microM, respectively), which was comparable with liarozole (7 microM) but considerably less active than R115866 (IC50 = 5 nM). 2-(4-hydroxybenzyl)-6-methoxytetralone 37-75 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 13-20 16279770-3 2005 In the MCF-7 CYP26A1 cell assay, the 2-(4-hydroxybenzyl)-6-methoxytetralone 5 and unsaturated benzylidene precursor 6 were found to be the most potent (IC50 = 7 and 5 microM, respectively), which was comparable with liarozole (7 microM) but considerably less active than R115866 (IC50 = 5 nM). unsaturated benzylidene 82-105 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 13-20 16279770-4 2005 With a CYP26A1 homology model, the tetralones were shown to be positioned in a hydrophobic tunnel with additional interactions, e.g., transition metal coordination and hydrogen-bonding interactions with GLY300, observed for the potent 4-hydroxyphenyl substituted inhibitors. Metals 145-150 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 7-14 16504501-1 2006 3-[6-(2-Dimethylamino-1-imidazol-1-yl-butyl)-naphthalen-2-yloxy]-2,2-dimethyl-propionic acid and analogs were designed and synthesized as highly potent and selective CYP26 inhibitors, serving as retinoic acid metabolic blocking agents (RAMBAs), with demonstrated in vivo efficacy to increase the half-life of exogenous atRA. 3-(6-(2-dimethylamino-1-imidazol-1-yl-butyl)naphthalen-2-yloxy)-2,2-dimethyl-propionic acid 0-92 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 166-171 16463413-1 2006 BACKGROUND: The retinoic acid (RA)-catabolizing enzyme Cyp26a1 plays an important role in protecting tailbud tissues from inappropriate exposure to RA. Tretinoin 16-29 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 55-62 16463413-1 2006 BACKGROUND: The retinoic acid (RA)-catabolizing enzyme Cyp26a1 plays an important role in protecting tailbud tissues from inappropriate exposure to RA. Tretinoin 31-33 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 55-62 16463413-1 2006 BACKGROUND: The retinoic acid (RA)-catabolizing enzyme Cyp26a1 plays an important role in protecting tailbud tissues from inappropriate exposure to RA. Tretinoin 148-150 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 55-62 16279770-4 2005 With a CYP26A1 homology model, the tetralones were shown to be positioned in a hydrophobic tunnel with additional interactions, e.g., transition metal coordination and hydrogen-bonding interactions with GLY300, observed for the potent 4-hydroxyphenyl substituted inhibitors. Tetralones 35-45 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 7-14 16194896-1 2005 The recently identified retinoic acid (RA)-metabolizing cytochrome P450RAI-1 (CYP26A1) has been implicated in accelerated metabolism and rapid clearance of all-trans-retinoic acid (ATRA) during prolonged oral administration in patients with acute promyelocytic leukemia (APL), leading to a progressive decline in plasma drug levels. Tretinoin 24-37 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 78-85 16194896-1 2005 The recently identified retinoic acid (RA)-metabolizing cytochrome P450RAI-1 (CYP26A1) has been implicated in accelerated metabolism and rapid clearance of all-trans-retinoic acid (ATRA) during prolonged oral administration in patients with acute promyelocytic leukemia (APL), leading to a progressive decline in plasma drug levels. Tretinoin 39-41 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 78-85 16194896-1 2005 The recently identified retinoic acid (RA)-metabolizing cytochrome P450RAI-1 (CYP26A1) has been implicated in accelerated metabolism and rapid clearance of all-trans-retinoic acid (ATRA) during prolonged oral administration in patients with acute promyelocytic leukemia (APL), leading to a progressive decline in plasma drug levels. Tretinoin 156-179 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 78-85 16194896-1 2005 The recently identified retinoic acid (RA)-metabolizing cytochrome P450RAI-1 (CYP26A1) has been implicated in accelerated metabolism and rapid clearance of all-trans-retinoic acid (ATRA) during prolonged oral administration in patients with acute promyelocytic leukemia (APL), leading to a progressive decline in plasma drug levels. Tretinoin 181-185 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 78-85 16194896-3 2005 ATRA rapidly induced upregulation of CYP26A1 mRNA expression in a dose-dependent manner. Tretinoin 0-4 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 37-44 16194896-4 2005 Other retinoids (retinol, 9-cis-RA, and 13-cis-RA) also induced significant CYP26A1 expression in HepG2 and NB4 cells. Retinoids 6-15 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 76-83 16194896-4 2005 Other retinoids (retinol, 9-cis-RA, and 13-cis-RA) also induced significant CYP26A1 expression in HepG2 and NB4 cells. Vitamin A 17-24 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 76-83 16194896-4 2005 Other retinoids (retinol, 9-cis-RA, and 13-cis-RA) also induced significant CYP26A1 expression in HepG2 and NB4 cells. Alitretinoin 26-34 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 76-83 16194896-4 2005 Other retinoids (retinol, 9-cis-RA, and 13-cis-RA) also induced significant CYP26A1 expression in HepG2 and NB4 cells. Isotretinoin 40-49 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 76-83 16194896-5 2005 CYP26A1 mRNA expression in HepG2 cells returned to baseline in 48 h upon removal of ATRA from the culture medium, suggesting that the expression is reversible and requires the presence of ATRA. Tretinoin 84-88 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 16194896-5 2005 CYP26A1 mRNA expression in HepG2 cells returned to baseline in 48 h upon removal of ATRA from the culture medium, suggesting that the expression is reversible and requires the presence of ATRA. Tretinoin 188-192 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-7 16194896-6 2005 In endothelial cells, however, a higher concentration of ATRA (10 microM) was required to induce expression of CYP26A1. Tretinoin 57-61 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 111-118 16194896-7 2005 A specific RA receptor-alpha antagonist totally inhibited ATRA-induced expression of CYP26A1, indicating that RA receptor-alpha plays a major role in CYP26A1 expression in HepG2 cells. Tretinoin 58-62 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 85-92 16194896-7 2005 A specific RA receptor-alpha antagonist totally inhibited ATRA-induced expression of CYP26A1, indicating that RA receptor-alpha plays a major role in CYP26A1 expression in HepG2 cells. Tretinoin 58-62 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 150-157 16194896-10 2005 L-ATRA induced lower CYP26A1 expression and metabolic activity in HepG2 and NB4 cells when compared with free ATRA. Tretinoin 2-6 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 21-28 16194896-12 2005 Our data suggest that upregulation of CYP26A1 expression in intestinal, endothelial, liver, and APL cells and metabolism of ATRA may play a role in rapid clearance of ATRA after continuous oral administration. Tretinoin 167-171 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 38-45 15597402-9 2004 In addition to these inhibitors of CYP5, CYP17, and CYP19, liarozole, known to be a potent inhibitor of CYP26 (retinoic acid-4-hydroxylase) and ATRA (all-trans-retinoic acid) metabolism, was able to reduce tumor cell adhesion to 51% of the initial rate. liarozole 59-68 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 104-109 15896339-2 2005 Among the cytochrome P450s (CYPs) involved in ATRA metabolism, the ATRA-inducible cytochrome P450 26A1 (CYP26A1) is particularly active although the molecular mechanisms involved in its regulation are not well defined in the target leukemia cells. Tretinoin 46-50 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 82-102 15896339-2 2005 Among the cytochrome P450s (CYPs) involved in ATRA metabolism, the ATRA-inducible cytochrome P450 26A1 (CYP26A1) is particularly active although the molecular mechanisms involved in its regulation are not well defined in the target leukemia cells. Tretinoin 46-50 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 104-111 15896339-2 2005 Among the cytochrome P450s (CYPs) involved in ATRA metabolism, the ATRA-inducible cytochrome P450 26A1 (CYP26A1) is particularly active although the molecular mechanisms involved in its regulation are not well defined in the target leukemia cells. Tretinoin 67-71 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 82-102 15896339-2 2005 Among the cytochrome P450s (CYPs) involved in ATRA metabolism, the ATRA-inducible cytochrome P450 26A1 (CYP26A1) is particularly active although the molecular mechanisms involved in its regulation are not well defined in the target leukemia cells. Tretinoin 67-71 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 104-111 15896339-4 2005 CYP26A1 constitutive expression was barely detectable in NB4 cells, but ATRA could induce high levels of CYP26A1 expression, which reached a maximum at 72h. Tretinoin 72-76 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 105-112 15714209-7 2005 Both ATRA and 13cisRA dramatically induced the expression of CYP26A1 in SH-SY5Y cells, and treatment with R116010, but not acitretin, potentiated the RA-induced expression of a reporter gene and CYP26A1. Tretinoin 5-9 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 61-68 15714209-7 2005 Both ATRA and 13cisRA dramatically induced the expression of CYP26A1 in SH-SY5Y cells, and treatment with R116010, but not acitretin, potentiated the RA-induced expression of a reporter gene and CYP26A1. Tretinoin 5-9 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 195-202 15714209-7 2005 Both ATRA and 13cisRA dramatically induced the expression of CYP26A1 in SH-SY5Y cells, and treatment with R116010, but not acitretin, potentiated the RA-induced expression of a reporter gene and CYP26A1. Isotretinoin 14-21 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 61-68 15714209-7 2005 Both ATRA and 13cisRA dramatically induced the expression of CYP26A1 in SH-SY5Y cells, and treatment with R116010, but not acitretin, potentiated the RA-induced expression of a reporter gene and CYP26A1. Isotretinoin 14-21 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 195-202 16143075-7 2005 CONCLUSION: ATRA has remarkable synergic effect with IFN-alpha on HL-60 cells, probably because IFN-alpha inhibits CYP26 mRNA expression and thus reduces the metabolism of ATRA. Tretinoin 12-16 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 115-120 16012519-4 2005 The pattern of retinoid responsiveness for six of 13 target genes (RARbeta2, CYP26A1, CRBP1, RGS16, DUSP6, EGR1) correlated with phenotypic retinoid sensitivity, across a panel of retinoid-sensitive or -resistant lung and breast cancer cell lines. Retinoids 15-23 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 77-84 16012519-4 2005 The pattern of retinoid responsiveness for six of 13 target genes (RARbeta2, CYP26A1, CRBP1, RGS16, DUSP6, EGR1) correlated with phenotypic retinoid sensitivity, across a panel of retinoid-sensitive or -resistant lung and breast cancer cell lines. Retinoids 140-148 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 77-84 16012519-4 2005 The pattern of retinoid responsiveness for six of 13 target genes (RARbeta2, CYP26A1, CRBP1, RGS16, DUSP6, EGR1) correlated with phenotypic retinoid sensitivity, across a panel of retinoid-sensitive or -resistant lung and breast cancer cell lines. Retinoids 140-148 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 77-84 16012519-6 2005 In retinoid-sensitive neuroblastoma, lung and breast cancer cell lines, direct inhibition of retinoid-induced RARbeta2 expression blocked induction of only one of eight retinoid target genes (CYP26A1). Retinoids 93-101 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 192-199 16012519-6 2005 In retinoid-sensitive neuroblastoma, lung and breast cancer cell lines, direct inhibition of retinoid-induced RARbeta2 expression blocked induction of only one of eight retinoid target genes (CYP26A1). Retinoids 93-101 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 192-199 16012519-7 2005 DNA demethylation, histone acetylation, and exogenous overexpression of RARbeta2 partially restored retinoid-responsive CYP26A1 expression in RA-resistant MDA-MB-231 breast, but not SK-MES-1 lung, cancer cells. Retinoids 100-108 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 120-127 15703382-0 2005 Expression of the retinoic acid-metabolizing enzyme CYP26A1 limits programmed cell death. Tretinoin 18-31 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 52-59 15703382-3 2005 On the other hand, we show here that various cell lines overexpressing CYP26A1, a cytochrome P450 enzyme specifically involved in the catabolic inactivation of RA, exhibit increased resistance to various apoptogenic factors, including death receptor ligands such as tumor necrosis factor-related apoptosis-inducing ligand. Tretinoin 160-162 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 71-78 15703382-5 2005 In addition, synthetic retinoids Am80 (4[(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)carbamoyl]benzoic acid) and Am580 [4(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphtamido)benzoic acid], which are resistant to CYP26A1 metabolism, can restore the sensitivity of these cells to apoptogens. Retinoids 23-32 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 223-230 15703382-5 2005 In addition, synthetic retinoids Am80 (4[(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)carbamoyl]benzoic acid) and Am580 [4(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphtamido)benzoic acid], which are resistant to CYP26A1 metabolism, can restore the sensitivity of these cells to apoptogens. tamibarotene 33-37 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 223-230 15745819-1 2005 A series of [2-imidazol-1-yl-2-(6-alkoxy-naphthalen-2-yl)-1-methyl-ethyl]-dimethyl-amines were designed and synthesized as CYP26 inhibitors, serving as retinoic acid metabolic blocking agents (RAMBA"s). [2-imidazol-1-yl-2-(6-alkoxy-naphthalen-2-yl)-1-methyl-ethyl]-dimethyl-amines 12-89 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 123-128 15745819-1 2005 A series of [2-imidazol-1-yl-2-(6-alkoxy-naphthalen-2-yl)-1-methyl-ethyl]-dimethyl-amines were designed and synthesized as CYP26 inhibitors, serving as retinoic acid metabolic blocking agents (RAMBA"s). Tretinoin 152-165 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 123-128 15589975-6 2005 In addition, organochlorine pesticides strongly induce cytochrome P450RAI1 (P450RAI1), a key factor of retinoic acid level regulation in many tissues and whose expression and activity are strongly induced by retinoic acid. Hydrocarbons, Chlorinated 13-27 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 66-74 15589975-6 2005 In addition, organochlorine pesticides strongly induce cytochrome P450RAI1 (P450RAI1), a key factor of retinoic acid level regulation in many tissues and whose expression and activity are strongly induced by retinoic acid. Hydrocarbons, Chlorinated 13-27 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 76-84 15589975-6 2005 In addition, organochlorine pesticides strongly induce cytochrome P450RAI1 (P450RAI1), a key factor of retinoic acid level regulation in many tissues and whose expression and activity are strongly induced by retinoic acid. Tretinoin 103-116 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 66-74 15589975-6 2005 In addition, organochlorine pesticides strongly induce cytochrome P450RAI1 (P450RAI1), a key factor of retinoic acid level regulation in many tissues and whose expression and activity are strongly induced by retinoic acid. Tretinoin 103-116 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 76-84 15589975-6 2005 In addition, organochlorine pesticides strongly induce cytochrome P450RAI1 (P450RAI1), a key factor of retinoic acid level regulation in many tissues and whose expression and activity are strongly induced by retinoic acid. Tretinoin 208-221 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 66-74 15589975-6 2005 In addition, organochlorine pesticides strongly induce cytochrome P450RAI1 (P450RAI1), a key factor of retinoic acid level regulation in many tissues and whose expression and activity are strongly induced by retinoic acid. Tretinoin 208-221 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 76-84 15597402-9 2004 In addition to these inhibitors of CYP5, CYP17, and CYP19, liarozole, known to be a potent inhibitor of CYP26 (retinoic acid-4-hydroxylase) and ATRA (all-trans-retinoic acid) metabolism, was able to reduce tumor cell adhesion to 51% of the initial rate. liarozole 59-68 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 111-138 15281009-0 2004 The effect of cellular retinoic acid binding protein-I expression on the CYP26-mediated catabolism of all-trans retinoic acid and cell proliferation in head and neck squamous cell carcinoma. Tretinoin 23-36 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 73-78 15451545-1 2004 All trans-retinoic acid (RA) plays a role in regulation of P450RAI gene expression. Tretinoin 4-23 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 59-66 15448016-0 2004 Identification of the fenretinide metabolite 4-oxo-fenretinide present in human plasma and formed in human ovarian carcinoma cells through induction of cytochrome P450 26A1. Fenretinide 22-33 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 152-172 15448016-0 2004 Identification of the fenretinide metabolite 4-oxo-fenretinide present in human plasma and formed in human ovarian carcinoma cells through induction of cytochrome P450 26A1. 4-oxofenretinide 45-62 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 152-172 14532297-4 2004 We have previously identified two cytochrome P450s, P450RAI-1 and P450RAI-2 (herein named CYP26A1 and CYP26B1), which were shown to be responsible for catabolism of atRA both in the embryo and the adult. Tretinoin 165-169 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 52-61 15291358-8 2004 Although ATRA and 4-HPR induced expression of CYP26, an ATRA-inducible gene encoding a cytochrome P450 enzyme, in HL-60 cells, both retinoids failed to induce CYP26 in HL-60R cells. Tretinoin 9-13 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 46-51 15291358-8 2004 Although ATRA and 4-HPR induced expression of CYP26, an ATRA-inducible gene encoding a cytochrome P450 enzyme, in HL-60 cells, both retinoids failed to induce CYP26 in HL-60R cells. Tretinoin 56-60 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 46-51 14532297-4 2004 We have previously identified two cytochrome P450s, P450RAI-1 and P450RAI-2 (herein named CYP26A1 and CYP26B1), which were shown to be responsible for catabolism of atRA both in the embryo and the adult. Tretinoin 165-169 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 90-97 14532297-9 2004 Specifically, CYP26C1 can also recognize and metabolize 9-cis-RA and is much less sensitive than the other CYP26 family members to the inhibitory effects of ketoconazole. Tretinoin 56-64 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 14-19 14532297-9 2004 Specifically, CYP26C1 can also recognize and metabolize 9-cis-RA and is much less sensitive than the other CYP26 family members to the inhibitory effects of ketoconazole. Ketoconazole 157-169 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 14-19 14532297-11 2004 This third CYP26 member may play a specific role in catabolizing both all-trans and 9-cis isomers of RA. Radium 101-103 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 11-16 14704332-6 2004 Additionally, an RA-inducible cytochrome P450, P450RAI or CYP26, is down-regulated in liver during vitamin A deficiency and up-regulated dose-dependently by dietary vitamin A and exogenous RA. Tretinoin 17-19 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 47-54 14704332-6 2004 Additionally, an RA-inducible cytochrome P450, P450RAI or CYP26, is down-regulated in liver during vitamin A deficiency and up-regulated dose-dependently by dietary vitamin A and exogenous RA. Tretinoin 17-19 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 58-63 14704332-6 2004 Additionally, an RA-inducible cytochrome P450, P450RAI or CYP26, is down-regulated in liver during vitamin A deficiency and up-regulated dose-dependently by dietary vitamin A and exogenous RA. Vitamin A 99-108 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 47-54 14704332-7 2004 Based on these results, we propose that LRAT and CYP26 serve as two molecular mechanisms, coordinately regulated by all-trans-RA, to control the availability of retinol and RA, respectively. Vitamin A 161-168 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 49-54 12054474-1 2002 A novel retinoic acid (RA)-inducible cytochrome P450 (P450 RAI or CYP26), previously cloned from human, zebra fish, and mouse, functions in the metabolism of all-trans-RA to polar metabolites including 4-hydroxy-RA and 4-oxo-RA. Tretinoin 8-21 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 54-62 12870655-6 2003 The same ATRA treatment also resulted in the detection of CYP26A1 but not CYP1A1, CYP2B6, CYP2C8, CY2C9, CYP3A4, or CYP3A5 mRNA. Tretinoin 9-13 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 58-65 12711469-7 2003 We showed that all-trans retinoic acid is metabolised by CYP2S1, which has higher cutaneous expression than CYP26, previously described as the specific cutaneous P450 retinoic-acid-metabolising enzyme. Tretinoin 25-38 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 108-113 12711469-7 2003 We showed that all-trans retinoic acid is metabolised by CYP2S1, which has higher cutaneous expression than CYP26, previously described as the specific cutaneous P450 retinoic-acid-metabolising enzyme. Tretinoin 167-180 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 108-113 12044165-8 2002 We also found that in CP26 three chlorophyll binding sites are selective for Chl a, one of them being essential for the folding of the pigment-protein complex. Chlorophyll 33-44 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 22-26 14704332-6 2004 Additionally, an RA-inducible cytochrome P450, P450RAI or CYP26, is down-regulated in liver during vitamin A deficiency and up-regulated dose-dependently by dietary vitamin A and exogenous RA. Vitamin A 99-108 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 58-63 14692511-2 2003 Several of the compounds were weak inhibitors of the non-specific rat liver microsomal P450 enzymes and moderate inhibitors of the RA-induced enzymes in cultured human genital fibroblasts, where the RA-specific enzyme CYP26 is probably expressed. Radium 131-133 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 218-223 14692511-2 2003 Several of the compounds were weak inhibitors of the non-specific rat liver microsomal P450 enzymes and moderate inhibitors of the RA-induced enzymes in cultured human genital fibroblasts, where the RA-specific enzyme CYP26 is probably expressed. Radium 199-201 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 218-223 12054474-1 2002 A novel retinoic acid (RA)-inducible cytochrome P450 (P450 RAI or CYP26), previously cloned from human, zebra fish, and mouse, functions in the metabolism of all-trans-RA to polar metabolites including 4-hydroxy-RA and 4-oxo-RA. Tretinoin 8-21 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 66-71 12054474-1 2002 A novel retinoic acid (RA)-inducible cytochrome P450 (P450 RAI or CYP26), previously cloned from human, zebra fish, and mouse, functions in the metabolism of all-trans-RA to polar metabolites including 4-hydroxy-RA and 4-oxo-RA. Tretinoin 23-25 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 54-62 12054474-1 2002 A novel retinoic acid (RA)-inducible cytochrome P450 (P450 RAI or CYP26), previously cloned from human, zebra fish, and mouse, functions in the metabolism of all-trans-RA to polar metabolites including 4-hydroxy-RA and 4-oxo-RA. Tretinoin 23-25 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 66-71 12054474-1 2002 A novel retinoic acid (RA)-inducible cytochrome P450 (P450 RAI or CYP26), previously cloned from human, zebra fish, and mouse, functions in the metabolism of all-trans-RA to polar metabolites including 4-hydroxy-RA and 4-oxo-RA. Tretinoin 59-61 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 66-71 12054474-1 2002 A novel retinoic acid (RA)-inducible cytochrome P450 (P450 RAI or CYP26), previously cloned from human, zebra fish, and mouse, functions in the metabolism of all-trans-RA to polar metabolites including 4-hydroxy-RA and 4-oxo-RA. 4-hydroxyretinoic acid 202-214 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 54-62 12054474-1 2002 A novel retinoic acid (RA)-inducible cytochrome P450 (P450 RAI or CYP26), previously cloned from human, zebra fish, and mouse, functions in the metabolism of all-trans-RA to polar metabolites including 4-hydroxy-RA and 4-oxo-RA. 4-hydroxyretinoic acid 202-214 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 66-71 12054474-1 2002 A novel retinoic acid (RA)-inducible cytochrome P450 (P450 RAI or CYP26), previously cloned from human, zebra fish, and mouse, functions in the metabolism of all-trans-RA to polar metabolites including 4-hydroxy-RA and 4-oxo-RA. 4-oxoretinoic acid 219-227 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 54-62 12054474-1 2002 A novel retinoic acid (RA)-inducible cytochrome P450 (P450 RAI or CYP26), previously cloned from human, zebra fish, and mouse, functions in the metabolism of all-trans-RA to polar metabolites including 4-hydroxy-RA and 4-oxo-RA. 4-oxoretinoic acid 219-227 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 66-71 12054474-3 2002 The nucleotide sequence predicts a 497-amino-acid protein whose sequence is 95% identical to mouse and 91% homologous to human CYP26. amino 39-44 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 127-132 11994980-0 2002 all-trans-Retinoic acid-induced expression and regulation of retinoic acid 4-hydroxylase (CYP26) in human promyelocytic leukemia. Tretinoin 0-23 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 61-88 11994980-0 2002 all-trans-Retinoic acid-induced expression and regulation of retinoic acid 4-hydroxylase (CYP26) in human promyelocytic leukemia. Tretinoin 0-23 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 90-95 11994980-4 2002 We found that treatment of NB4 cells with a pharmacological concentration of ATRA (1 microM) induced rapid and dose-dependent expression of CYP26 mRNA. Tretinoin 77-81 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 140-145 11994980-5 2002 The CYP26 expression returned to pretreatment levels in both cells after ATRA was removed from the media. Tretinoin 73-77 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 4-9 11994980-6 2002 Retinoic acid receptor-alpha (RARalpha) specific antagonist (CD2503) totally abolished the ATRA-induced expression of CYP26 mRNA in HL-60 and NB4 cells. Tretinoin 91-95 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 118-123 11994980-8 2002 ATRA-induced expression of CYP26 was restored in HL-60R cells retrovirally transduced with RARalpha, but not in those cells transduced with the other retinoid receptors. Tretinoin 0-4 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 27-32 11994980-9 2002 In conclusion, ATRA induces expression of CYP26 in myeloid and promyelocytic leukemia cells and this expression is modulated by RARalpha. Tretinoin 15-19 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 42-47 11994980-10 2002 The induction of CYP26 expression by ATRA treatment might be related to a substrate-driven feedback mechanism to regulate intracellular concentrations of ATRA and its over expression in some clones may be partly responsible for reduced sensitivity or resistance to ATRA therapy. Tretinoin 37-41 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 17-22 11994980-10 2002 The induction of CYP26 expression by ATRA treatment might be related to a substrate-driven feedback mechanism to regulate intracellular concentrations of ATRA and its over expression in some clones may be partly responsible for reduced sensitivity or resistance to ATRA therapy. Tretinoin 154-158 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 17-22 11994980-10 2002 The induction of CYP26 expression by ATRA treatment might be related to a substrate-driven feedback mechanism to regulate intracellular concentrations of ATRA and its over expression in some clones may be partly responsible for reduced sensitivity or resistance to ATRA therapy. Tretinoin 154-158 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 17-22 11606945-2 2001 Presented is a summary of recent research that examines the role of some of the enzymes involved in RA distribution, particularly those involved in RA catabolism (P450RAI). Tretinoin 100-102 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 163-170 11953746-4 2002 Cyp26a1(-/-) mouse fetuses have lethal morphogenetic phenotypes mimicking those generated by excess retinoic acid administration, indicating that human CYP26A1 may be essential in controlling retinoic acid levels during development. Tretinoin 100-113 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 152-159 11953746-4 2002 Cyp26a1(-/-) mouse fetuses have lethal morphogenetic phenotypes mimicking those generated by excess retinoic acid administration, indicating that human CYP26A1 may be essential in controlling retinoic acid levels during development. Tretinoin 192-205 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 152-159 11912557-0 2002 Retinoic acid 4-hydroxylase-mediated catabolism of all-trans retinoic acid and the cell proliferation in head and neck squamous cell carcinoma. Tretinoin 61-74 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-27 11912557-2 2002 The aim of this study was to confirm if retinoic acid 4-hydroxylase (CYP26) is a P450 induced by RA and to investigate the role of cellular RA binding proteins (CRABPs), using a slow catabolizer, AMC-HN-4, and a rapid catabolizer, AMC-HN-6. Tretinoin 97-99 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 40-67 11912557-2 2002 The aim of this study was to confirm if retinoic acid 4-hydroxylase (CYP26) is a P450 induced by RA and to investigate the role of cellular RA binding proteins (CRABPs), using a slow catabolizer, AMC-HN-4, and a rapid catabolizer, AMC-HN-6. Tretinoin 97-99 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 69-74 11912557-5 2002 The sensitivity to RA was variable by the amount of CYP26, and the rapid catabolism by CYP26 made AMC-HN-6 resistant to RA in vitro. Tretinoin 19-21 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 52-57 11912557-5 2002 The sensitivity to RA was variable by the amount of CYP26, and the rapid catabolism by CYP26 made AMC-HN-6 resistant to RA in vitro. Tretinoin 120-122 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 87-92 11912557-7 2002 Conclusively, the CYP26 activity might be one essential factor for the RA sensitivity, but in cells showing induction of CYP26, the RA sensitivity is inversely related to the rate of RA catabolism. Tretinoin 71-73 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 18-23 11912557-7 2002 Conclusively, the CYP26 activity might be one essential factor for the RA sensitivity, but in cells showing induction of CYP26, the RA sensitivity is inversely related to the rate of RA catabolism. Tretinoin 132-134 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 121-126 11912557-7 2002 Conclusively, the CYP26 activity might be one essential factor for the RA sensitivity, but in cells showing induction of CYP26, the RA sensitivity is inversely related to the rate of RA catabolism. Tretinoin 132-134 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 121-126 11766909-4 2001 RA also induces the expression in liver of the mRNA for CYP26, the enzyme that disposes of excess RA by oxidizing RA to 4-oxo-RA. Tretinoin 0-2 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 56-61 11766909-4 2001 RA also induces the expression in liver of the mRNA for CYP26, the enzyme that disposes of excess RA by oxidizing RA to 4-oxo-RA. Tretinoin 98-100 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 56-61 11766909-4 2001 RA also induces the expression in liver of the mRNA for CYP26, the enzyme that disposes of excess RA by oxidizing RA to 4-oxo-RA. Tretinoin 98-100 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 56-61 11766909-4 2001 RA also induces the expression in liver of the mRNA for CYP26, the enzyme that disposes of excess RA by oxidizing RA to 4-oxo-RA. 4-oxoretinoic acid 120-128 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 56-61 11766909-5 2001 CYP26 is downregulated upon vitamin A depletion. Vitamin A 28-37 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 11302942-0 2001 Effects of receptor-selective retinoids on CYP26 gene expression and metabolism of all-trans-retinoic acid in intestinal cells. Tretinoin 86-106 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 43-48 11690641-0 2001 Phytanic acid and docosahexaenoic acid increase the metabolism of all-trans-retinoic acid and CYP26 gene expression in intestinal cells. Phytanic Acid 0-13 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 94-99 11690641-0 2001 Phytanic acid and docosahexaenoic acid increase the metabolism of all-trans-retinoic acid and CYP26 gene expression in intestinal cells. Docosahexaenoic Acids 18-38 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 94-99 11690641-6 2001 However, in combination with retinoic acid receptor (RAR)-ligands (all-trans-RA or synthetic Am580) phytanic acid enhanced the induction of CYP26 and RA-metabolism in comparison to treatments with all-trans-RA or Am580 alone. Phytanic Acid 100-113 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 140-145 11690641-7 2001 Also treatment with DHA did not affect CYP26 gene expression and RA-metabolism but cotreatment of the cells with DHA and all-trans-RA or Am580 enhanced the induction of CYP26, in comparison to the induction caused by all-trans-RA or Am580 alone. Docosahexaenoic Acids 113-116 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 169-174 11690641-8 2001 This study indicates that food compounds such as phytanic acid and DHA that are RXR-agonists and have an impact on intestinal CYP26 gene expression and metabolism of all-trans-RA in intestinal cells. Phytanic Acid 49-62 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 126-131 11690641-8 2001 This study indicates that food compounds such as phytanic acid and DHA that are RXR-agonists and have an impact on intestinal CYP26 gene expression and metabolism of all-trans-RA in intestinal cells. Docosahexaenoic Acids 67-70 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 126-131 11690641-8 2001 This study indicates that food compounds such as phytanic acid and DHA that are RXR-agonists and have an impact on intestinal CYP26 gene expression and metabolism of all-trans-RA in intestinal cells. Tretinoin 176-178 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 126-131 11471143-0 2001 Differential expression of chicken CYP26 in anterior versus posterior limb bud in response to retinoic acid. Tretinoin 94-107 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 35-40 11471143-4 2001 Recently, enzymes capable of catabolizing RA were found to constitute a new family, called CYP26, within the cytochrome P450 superfamily. Tretinoin 42-44 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 91-96 11302942-2 2001 The complex mechanistic pattern of retinoid-induced effects on gene expression of CYP26 and intestinal metabolism of all-trans-retinoic acid (RA) was investigated here by studying the effects of retinoid ligands with relative selectivity for binding and transactivation of the retinoid acid receptors, RARs and RXRs, in human intestinal Caco-2 cells. Retinoids 35-43 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 82-87 11302942-2 2001 The complex mechanistic pattern of retinoid-induced effects on gene expression of CYP26 and intestinal metabolism of all-trans-retinoic acid (RA) was investigated here by studying the effects of retinoid ligands with relative selectivity for binding and transactivation of the retinoid acid receptors, RARs and RXRs, in human intestinal Caco-2 cells. Retinoids 195-203 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 82-87 11302942-10 2001 Receptor-selective retinoids showed enhanced effects on induction of CYP26 gene expression and all-trans-retinoic acid metabolism. Retinoids 19-28 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 69-74 11309353-5 2001 Furthermore, the mRNA expression of the RA-specific 4-hydroxylase, CYP26A1, was dramatically increased after RA-induction in the two HNSCC cell lines with the highest metabolism, was undetectable in normal keratinocytes, and was not inducible by RA. Tretinoin 40-42 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 67-74 11309353-5 2001 Furthermore, the mRNA expression of the RA-specific 4-hydroxylase, CYP26A1, was dramatically increased after RA-induction in the two HNSCC cell lines with the highest metabolism, was undetectable in normal keratinocytes, and was not inducible by RA. Tretinoin 109-111 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 67-74 11309353-6 2001 Next, introduction of CYP26A1 cDNA in a low-metabolizing HNSCC cell line resulted in an 11-fold higher turnover rate of RA and a 12-fold increase in the amount of polar metabolites, but it did not change sensitivity to RA. Tretinoin 120-122 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 22-29 11522124-11 2001 One known morphogen, retinoic acid, has the enzymes retinal dehydrogenase (RALDH) and CYP26 maintaining its actions, the former responsible for its generation and the latter for its elimination. Tretinoin 21-34 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 86-91 10953040-5 2000 CYP26 mRNA can be induced by the RAR-selective retinoid 4-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-anthracenyl)-benzoic acid (TTAB) but not by the RXR-selective retinoid SR11217 or the anti-activator-protein 1-selective retinoid SR11302. Retinoids 47-55 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 11082432-10 2000 The specific retinoic acid metabolizing CYP26 was induced after RA treatment in Caco-2 cells. Tretinoin 13-26 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 40-45 11082432-10 2000 The specific retinoic acid metabolizing CYP26 was induced after RA treatment in Caco-2 cells. Tretinoin 64-66 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 40-45 11025452-8 2000 Strong RA induction of the RA hydroxylase P450RAI (CYP26) was confined to ERalpha-positive T47D and MCF-7 breast cancer cells and did not appear to explain the lack of detectable RA levels in these cells since RA remained undetectable when the cells were treated with 5-10 microM liarozole, a P450RAI inhibitor. liarozole 280-289 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 42-49 11025452-8 2000 Strong RA induction of the RA hydroxylase P450RAI (CYP26) was confined to ERalpha-positive T47D and MCF-7 breast cancer cells and did not appear to explain the lack of detectable RA levels in these cells since RA remained undetectable when the cells were treated with 5-10 microM liarozole, a P450RAI inhibitor. Tretinoin 7-9 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 42-49 11025452-8 2000 Strong RA induction of the RA hydroxylase P450RAI (CYP26) was confined to ERalpha-positive T47D and MCF-7 breast cancer cells and did not appear to explain the lack of detectable RA levels in these cells since RA remained undetectable when the cells were treated with 5-10 microM liarozole, a P450RAI inhibitor. Tretinoin 7-9 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 51-56 11025452-8 2000 Strong RA induction of the RA hydroxylase P450RAI (CYP26) was confined to ERalpha-positive T47D and MCF-7 breast cancer cells and did not appear to explain the lack of detectable RA levels in these cells since RA remained undetectable when the cells were treated with 5-10 microM liarozole, a P450RAI inhibitor. Tretinoin 7-9 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 293-300 11025452-8 2000 Strong RA induction of the RA hydroxylase P450RAI (CYP26) was confined to ERalpha-positive T47D and MCF-7 breast cancer cells and did not appear to explain the lack of detectable RA levels in these cells since RA remained undetectable when the cells were treated with 5-10 microM liarozole, a P450RAI inhibitor. Tretinoin 27-29 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 42-49 11025452-8 2000 Strong RA induction of the RA hydroxylase P450RAI (CYP26) was confined to ERalpha-positive T47D and MCF-7 breast cancer cells and did not appear to explain the lack of detectable RA levels in these cells since RA remained undetectable when the cells were treated with 5-10 microM liarozole, a P450RAI inhibitor. Tretinoin 27-29 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 51-56 11025452-8 2000 Strong RA induction of the RA hydroxylase P450RAI (CYP26) was confined to ERalpha-positive T47D and MCF-7 breast cancer cells and did not appear to explain the lack of detectable RA levels in these cells since RA remained undetectable when the cells were treated with 5-10 microM liarozole, a P450RAI inhibitor. Tretinoin 27-29 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 293-300 11025452-8 2000 Strong RA induction of the RA hydroxylase P450RAI (CYP26) was confined to ERalpha-positive T47D and MCF-7 breast cancer cells and did not appear to explain the lack of detectable RA levels in these cells since RA remained undetectable when the cells were treated with 5-10 microM liarozole, a P450RAI inhibitor. liarozole 280-289 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 51-56 11041855-6 2000 It is concluded that the extrinsic subunits keep the S-LHCII and CP29 subunits in proper positions at some distance from the central part of the photosystem II core dimer to ensure a directed transfer of excitation energy through the monomeric peripheral antenna proteins CP26 and CP29 and/or to maintain sequestered domains of inorganic cofactors required for oxygen evolution. Oxygen 361-367 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 272-276 11023996-10 2000 We conclude that CYP26 expression and RA metabolism are regulated in adult liver not only acutely by RA administration, as may be relevant to retinoid therapy, but under chronic dietary conditions relevant to vitamin A nutrition in humans. Tretinoin 101-103 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 17-22 11023996-10 2000 We conclude that CYP26 expression and RA metabolism are regulated in adult liver not only acutely by RA administration, as may be relevant to retinoid therapy, but under chronic dietary conditions relevant to vitamin A nutrition in humans. Retinoids 142-150 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 17-22 11023996-10 2000 We conclude that CYP26 expression and RA metabolism are regulated in adult liver not only acutely by RA administration, as may be relevant to retinoid therapy, but under chronic dietary conditions relevant to vitamin A nutrition in humans. Vitamin A 209-218 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 17-22 10953040-5 2000 CYP26 mRNA can be induced by the RAR-selective retinoid 4-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-anthracenyl)-benzoic acid (TTAB) but not by the RXR-selective retinoid SR11217 or the anti-activator-protein 1-selective retinoid SR11302. Retinoids 162-170 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 10953040-5 2000 CYP26 mRNA can be induced by the RAR-selective retinoid 4-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-anthracenyl)-benzoic acid (TTAB) but not by the RXR-selective retinoid SR11217 or the anti-activator-protein 1-selective retinoid SR11302. SR 11302 230-237 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 10953040-6 2000 RARalpha-, beta-, and gamma-selective retinoids are able to induce CYP26; this induction is inhibited by the RARalpha-selective antagonist Ro41-5253. Retinoids 38-47 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 67-72 10953040-6 2000 RARalpha-, beta-, and gamma-selective retinoids are able to induce CYP26; this induction is inhibited by the RARalpha-selective antagonist Ro41-5253. Ro 41-5253 139-148 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 67-72 10953040-7 2000 TTAB is able to induce CYP26 mRNA expression in only a few of the lung carcinoma cell lines tested. 4-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-anthracenyl)benzoic acid 0-4 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 23-28 10953040-9 2000 The induction of CYP26 correlated with increased metabolism of RA into 18-hydroxy-, 4-oxo-, and 4-hydroxy-RA. Tretinoin 63-65 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 17-22 10953040-9 2000 The induction of CYP26 correlated with increased metabolism of RA into 18-hydroxy-, 4-oxo-, and 4-hydroxy-RA. 18-hydroxy-, 4-oxo-, and 4-hydroxy-ra 71-108 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 17-22 10862743-7 2000 Furthermore, RA upregulates P450RAI expression, suggesting the presence of feedback mechanisms controlling RA availability. Tretinoin 13-15 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 28-35 9228041-0 1997 CYP26, a novel mammalian cytochrome P450, is induced by retinoic acid and defines a new family. Tretinoin 56-69 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 10702251-9 2000 We also propose that atRA induces its own oxidative metabolism via a cytochrome P450 (CYP26) and is further biotransformed into glucuronides via UGT-mediated pathways. Tretinoin 21-25 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 86-91 10540859-5 1999 A refined control mechanism for intracellular level of retinoic acid is also discussed with retinal dehydrogenase II and cytochrome P450 RAI (CYP26). Tretinoin 55-68 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 142-147 10479456-5 1999 P19 embryonal carcinoma (EC) cell lines stably expressing hCYP26 undergo extensive and rapid neuronal differentiation in monolayer at already low concentrations of RA, while normally P19 cells under these conditions differentiate only in endoderm-like cells. Tretinoin 164-166 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 58-64 9740237-0 1998 Retinoic acid receptors regulate expression of retinoic acid 4-hydroxylase that specifically inactivates all-trans retinoic acid in human keratinocyte HaCaT cells. 2-octenal 109-114 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 47-74 9716180-6 1998 The RA-induced CYP26 was shown to be highly specific for the hydroxylation of all-trans-RA and did not recognize the 13-cis and 9-cis isomers. Tretinoin 4-6 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 15-20 10953040-1 2000 In this study, the expression of CYP26 is examined in relation to retinoid-induced mucosecretory differentiation in human tracheobronchial epithelial (HTBE) cells and compared with that in human lung carcinoma cell lines. Retinoids 66-74 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 33-38 10953040-4 2000 RA is required but not sufficient to induce RARbeta, CYP26, and MUC2 mRNA because induction is only observed in confluent but not in logarithmic cultures, suggesting that additional factors are critical in their regulation. Tretinoin 0-2 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 53-58 10953040-5 2000 CYP26 mRNA can be induced by the RAR-selective retinoid 4-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-anthracenyl)-benzoic acid (TTAB) but not by the RXR-selective retinoid SR11217 or the anti-activator-protein 1-selective retinoid SR11302. retinoid 4-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-anthracenyl)-benzoic acid 47-125 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 10953040-5 2000 CYP26 mRNA can be induced by the RAR-selective retinoid 4-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-anthracenyl)-benzoic acid (TTAB) but not by the RXR-selective retinoid SR11217 or the anti-activator-protein 1-selective retinoid SR11302. 4-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-anthracenyl)benzoic acid 127-131 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 10727725-1 2000 CYP26 (P450RAI) catalyzes catabolic retinoic acid (RA) hydroxylation and thereby appears to play a critical role in retinoid signaling pathways during development. Tretinoin 36-49 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 10727725-1 2000 CYP26 (P450RAI) catalyzes catabolic retinoic acid (RA) hydroxylation and thereby appears to play a critical role in retinoid signaling pathways during development. Tretinoin 36-49 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 7-14 10727725-1 2000 CYP26 (P450RAI) catalyzes catabolic retinoic acid (RA) hydroxylation and thereby appears to play a critical role in retinoid signaling pathways during development. Tretinoin 11-13 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 10727725-1 2000 CYP26 (P450RAI) catalyzes catabolic retinoic acid (RA) hydroxylation and thereby appears to play a critical role in retinoid signaling pathways during development. Retinoids 116-124 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 10727725-1 2000 CYP26 (P450RAI) catalyzes catabolic retinoic acid (RA) hydroxylation and thereby appears to play a critical role in retinoid signaling pathways during development. Retinoids 116-124 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 7-14 10421060-8 1999 [3H]Retinoic acid showed a more rapid metabolism to 4-hydroxy/4-keto-retinoic acid in HaCaT cells than in HEK, which could be explained by a higher expression of cytochrome p450RAI in the former cells. Tritium 1-3 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 162-180 10421060-8 1999 [3H]Retinoic acid showed a more rapid metabolism to 4-hydroxy/4-keto-retinoic acid in HaCaT cells than in HEK, which could be explained by a higher expression of cytochrome p450RAI in the former cells. Tretinoin 4-17 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 162-180 10421060-8 1999 [3H]Retinoic acid showed a more rapid metabolism to 4-hydroxy/4-keto-retinoic acid in HaCaT cells than in HEK, which could be explained by a higher expression of cytochrome p450RAI in the former cells. 4-hydroxy/4-keto-retinoic acid 52-82 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 162-180 9716180-1 1998 We report on the isolation of a cytochrome P450 (CYP)-like retinoic acid (RA) 4-hydroxylase cDNA from T-47D human breast cancer cells that is identical to the recently cloned hCYP26, which is involved in the metabolic breakdown of RA. Tretinoin 74-76 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 175-181 9716180-2 1998 Northern analysis showed that this novel human CYP26 is induced within 1 h upon RA treatment in RA-sensitive T-47D breast carcinoma cells but not in RA-resistant MDA-MB-231 breast cancer cells and HCT 116 colon cancer cells. Tretinoin 80-82 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 47-52 9716180-2 1998 Northern analysis showed that this novel human CYP26 is induced within 1 h upon RA treatment in RA-sensitive T-47D breast carcinoma cells but not in RA-resistant MDA-MB-231 breast cancer cells and HCT 116 colon cancer cells. Tretinoin 96-98 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 47-52 9716180-3 1998 Stable introduction of different RA receptor (RAR) subtypes in HCT 116 cells showed that CYP26 expression is dependent on RARalpha and RARgamma and, to a lesser extent, on RARbeta and closely paralleled RA metabolism, suggesting that it represents the major RA 4-hydroxylase in these human cells. Tretinoin 33-35 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 89-94 9716180-5 1998 Interestingly, CYP26 activity was efficiently inhibited by liarozole, an inhibitor of RA metabolism, leading to enhanced growth inhibition by RA. liarozole 59-68 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 15-20 9716180-5 1998 Interestingly, CYP26 activity was efficiently inhibited by liarozole, an inhibitor of RA metabolism, leading to enhanced growth inhibition by RA. Tretinoin 86-88 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 15-20 9716180-5 1998 Interestingly, CYP26 activity was efficiently inhibited by liarozole, an inhibitor of RA metabolism, leading to enhanced growth inhibition by RA. Tretinoin 142-144 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 15-20 9564181-0 1998 The cloning and characterization of a novel cytochrome P450 family, CYP26, with specificity toward retinoic acid. Tretinoin 99-112 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 68-73 9564181-2 1998 CYP26 displays specificity toward retinoic acid and it may function as an important regulator or differentiation and a possible modulator of disease states by controlling retinoid concentration and homeostasis. Tretinoin 34-47 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 9564181-2 1998 CYP26 displays specificity toward retinoic acid and it may function as an important regulator or differentiation and a possible modulator of disease states by controlling retinoid concentration and homeostasis. Retinoids 171-179 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-5 9857269-4 1998 Recently, a novel cytochrome P450 enzyme (CYP26) with specific RA 4-hydroxylase activity, which is rapidly induced by RA, has been cloned from man, mouse and zebra fish, fullfilling all requirements of an enzyme which could be of crucial importance in controlling steady-state levels of active retinoids in cells and target tissues, thus protecting against excessive exposure. Tretinoin 63-65 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 42-47 9857269-4 1998 Recently, a novel cytochrome P450 enzyme (CYP26) with specific RA 4-hydroxylase activity, which is rapidly induced by RA, has been cloned from man, mouse and zebra fish, fullfilling all requirements of an enzyme which could be of crucial importance in controlling steady-state levels of active retinoids in cells and target tissues, thus protecting against excessive exposure. Retinoids 294-303 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 42-47 9857269-5 1998 Besides the putative role of this newly identified CYP26 in contributing to susceptibility of cancer cells to retinoids, the possible function of this gene in early embryonic development is discussed. Retinoids 110-119 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 51-56 9228017-0 1997 cDNA cloning of human retinoic acid-metabolizing enzyme (hP450RAI) identifies a novel family of cytochromes P450. Tretinoin 22-35 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 57-65 9200700-14 1997 In contrast, the Triton X-100-solubilized LHCII is highly depleted in CP26 (CP 29 type I gene product) and is contaminated by a variety of unidentified polypeptides. Octoxynol 17-29 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 70-74 9228017-11 1997 In this paper, we describe the cloning and characterization of the first mammalian retinoic acid-inducible retinoic acid-metabolizing cytochrome P450 (hP450RAI), which belongs to a novel class of cytochromes (CYP26). Tretinoin 83-96 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 145-159 9228017-12 1997 We demonstrate that hP450RAI is responsible for generation of several hydroxylated forms of RA, including 4-OH-RA, 4-oxo-RA, and 18-OH-RA. 4-oh-ra 106-113 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 20-28 9228017-12 1997 We demonstrate that hP450RAI is responsible for generation of several hydroxylated forms of RA, including 4-OH-RA, 4-oxo-RA, and 18-OH-RA. 4-oxoretinoic acid 115-123 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 20-28 9228017-12 1997 We demonstrate that hP450RAI is responsible for generation of several hydroxylated forms of RA, including 4-OH-RA, 4-oxo-RA, and 18-OH-RA. 18-oh-ra 129-137 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 20-28 8757760-9 1996 These data demonstrate that, at doses used here, liarozole, although an effective inhibitor of retinoic acid 4-hydroxylase, cannot function alone like a retinoid in vivo, probably because of retinoic acid 4-hydroxylase induction. liarozole 49-58 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 95-122 9228017-13 1997 We also show that hP450RAI mRNA expression is highly induced by RA in certain human tumor cell lines and further show that RA-inducible RA metabolism may correlate with P450RAI expression. Tretinoin 64-66 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 18-26 9228017-13 1997 We also show that hP450RAI mRNA expression is highly induced by RA in certain human tumor cell lines and further show that RA-inducible RA metabolism may correlate with P450RAI expression. Tretinoin 64-66 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 19-26 9228017-13 1997 We also show that hP450RAI mRNA expression is highly induced by RA in certain human tumor cell lines and further show that RA-inducible RA metabolism may correlate with P450RAI expression. Tretinoin 64-66 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 18-26 9228017-13 1997 We also show that hP450RAI mRNA expression is highly induced by RA in certain human tumor cell lines and further show that RA-inducible RA metabolism may correlate with P450RAI expression. Tretinoin 64-66 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 19-26 8757760-0 1996 Liarozole inhibits human epidermal retinoic acid 4-hydroxylase activity and differentially augments human skin responses to retinoic acid and retinol in vivo. liarozole 0-9 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 35-62 8757760-2 1996 We investigated the effects of liarozole on the retinoic acid 4-hydroxylase activity of human epidermis and its ability to modify in vivo human skin responses to retinoic acid and all-trans retinol. liarozole 31-40 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 48-75 8757760-3 1996 Retinoic acid 4-hydroxylase activity induced in vivo by 4 d treatment with retinoic acid (0.1%) was inhibited in vitro by liarozole in a concentration-dependent manner. Tretinoin 75-88 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-27 8757760-3 1996 Retinoic acid 4-hydroxylase activity induced in vivo by 4 d treatment with retinoic acid (0.1%) was inhibited in vitro by liarozole in a concentration-dependent manner. liarozole 122-131 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 0-27 8757760-9 1996 These data demonstrate that, at doses used here, liarozole, although an effective inhibitor of retinoic acid 4-hydroxylase, cannot function alone like a retinoid in vivo, probably because of retinoic acid 4-hydroxylase induction. liarozole 49-58 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 191-218 8601734-1 1996 Application of all-trans retinoic acid to human skin for 4 d under occlusion produces a marked increase in retinoic acid 4-hydroxylase activity. 2-octenal 19-24 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 107-134 8601734-1 1996 Application of all-trans retinoic acid to human skin for 4 d under occlusion produces a marked increase in retinoic acid 4-hydroxylase activity. Tretinoin 25-38 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 107-134 8601734-3 1996 Application of 0.1% all-trans, 0.1% 9-cis, and 0.1% 13-cis retinoic acid to human skin for 2 d resulted in induction of only all-trans retinoic acid 4-hydroxylase activity. Isotretinoin 52-72 cytochrome P450 family 26 subfamily A member 1 Homo sapiens 135-162