PMID-sentid Pub_year Sent_text comp_official_name comp_offset protein_name organism prot_offset 31734656-2 2019 Aromatase is an enzyme that plays an important role in converting testosterone to estradiol and androstenedione to estrogen. Testosterone 66-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31734656-2 2019 Aromatase is an enzyme that plays an important role in converting testosterone to estradiol and androstenedione to estrogen. Estradiol 82-91 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31734656-2 2019 Aromatase is an enzyme that plays an important role in converting testosterone to estradiol and androstenedione to estrogen. Androstenedione 96-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31734656-2 2019 Aromatase is an enzyme that plays an important role in converting testosterone to estradiol and androstenedione to estrogen. Estrogens 115-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31734656-3 2019 Aromatase inhibitors can increase testosterone and androgen production without increasing the amount of estrogen in circulation. Testosterone 34-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31734656-3 2019 Aromatase inhibitors can increase testosterone and androgen production without increasing the amount of estrogen in circulation. Androgens 51-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31734656-4 2019 The aim of this study was to evaluate the effect of aromatase inhibitor letrozole on the male infertility. letrozole 72-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-61 31734656-14 2019 CONCLUSION: The ratio of testosterone to estradiol levels in infertile men treated with aromatase inhibitor improved and caused changes in sperm parameters. Testosterone 25-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 31734656-14 2019 CONCLUSION: The ratio of testosterone to estradiol levels in infertile men treated with aromatase inhibitor improved and caused changes in sperm parameters. Estradiol 41-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 31265900-4 2019 Treatment with bexarotene increased estradiol levels as well as estrogen-synthesizing enzymes and CYP19 expression in hippocampal slice cultures. Bexarotene 15-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-103 31265900-7 2019 The RXR antagonists HX531 and UVI3003 and the CYP19 inhibitor letrozole abolished the neuroprotection elicited by bexarotene, indicating that estradiol produced by RXR stimulation protects neurons from ischemic insult. Letrozole 62-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-51 31265900-7 2019 The RXR antagonists HX531 and UVI3003 and the CYP19 inhibitor letrozole abolished the neuroprotection elicited by bexarotene, indicating that estradiol produced by RXR stimulation protects neurons from ischemic insult. Bexarotene 114-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-51 31265900-7 2019 The RXR antagonists HX531 and UVI3003 and the CYP19 inhibitor letrozole abolished the neuroprotection elicited by bexarotene, indicating that estradiol produced by RXR stimulation protects neurons from ischemic insult. Estradiol 142-151 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-51 31265900-8 2019 The human brain-specific CYP19 promoter had 6 RXR half sites, and 2 of 6 half sites were responsible for CYP19 expression induced by bexarotene. Bexarotene 133-143 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-30 31265900-8 2019 The human brain-specific CYP19 promoter had 6 RXR half sites, and 2 of 6 half sites were responsible for CYP19 expression induced by bexarotene. Bexarotene 133-143 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 105-110 31393836-0 2019 Aromatase (CYP19) inhibition by biflavonoids obtained from the branches of Garcinia gardneriana (Clusiaceae). Biflavonoids 32-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31681690-2 2019 A genetic variation at the level of aromatase enzyme gene (CYP19 gene) and/or androgen receptors with subsequent increased ovarian androgen was suggested in PCOS. Androgens 131-139 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-64 31393836-6 2019 This demonstrates that biflavonoids are an important source of scaffolds for the development of new aromatase inhibitors, focusing on the development of new anticancer agents. Biflavonoids 23-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-109 31393836-3 2019 Although several flavonoids had already demonstrated the capacity of inhibiting aromatase activity, the role of biflavonoids as aromatase inhibitors is poorly studied. Flavonoids 17-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 80-89 31393836-3 2019 Although several flavonoids had already demonstrated the capacity of inhibiting aromatase activity, the role of biflavonoids as aromatase inhibitors is poorly studied. Biflavonoids 112-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 128-137 31393836-4 2019 In this work, the biflavonoids isolated from Garcinia gardneriana, morelloflavone (1), Gb-2a (2) and Gb-2a-7-O-glucose (3) were submitted to in vitro assay to evaluate the aromatase modulatory effect. Biflavonoids 18-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 172-181 31351863-10 2019 Analysis of a different subset of proteins that contain embedded metal ions as reference points revealed that many Met-Aro bridges are at/near protein surfaces. Metals 65-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 119-122 31426637-6 2019 In addition, it was found that FCF inhibited the proliferation of granulosa cells (GCs) and H295R cells, as well as downregulated the expression of CYP17A1 and CYP19A1 in estradiol and progesterone production, resulting in decreased steroidogenesis in GCs and H295R cells. Progesterone 185-197 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 160-167 31525235-5 2019 Additionally, variants in ESR1, SRD5A1, CYP19A1, and RXRG are suggested to be associated with response to dutasteride. Dutasteride 106-117 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 40-47 31325085-0 2019 Bone and body composition response to testosterone therapy vary according to polymorphisms in the CYP19A1 gene. Testosterone 38-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-105 31325085-1 2019 PURPOSE: To evaluate the influence of single nucleotide polymorphisms (SNPs) of CYP19A1 on the response and susceptibility to side effects from testosterone therapy. Testosterone 144-156 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 80-87 31325085-10 2019 CONCLUSIONS: For the first time, we demonstrated that CYP19A1 SNPs influence response to testosterone therapy in hypogonadal men, highlighting the importance of genetic profiling in therapeutics even for common clinical conditions. Testosterone 89-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 54-61 31330226-2 2019 The aim of this study was to mechanistically investigate the endocrine disrupting potential of four commonly used azole antifungal drugs; clotrimazole, miconazole, ketoconazole and fluconazole in vitro using the H295R cell assay and two recombinant, CYP17A1 and CYP19A1 (aromatase), assays. Azoles 114-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 262-269 31330226-2 2019 The aim of this study was to mechanistically investigate the endocrine disrupting potential of four commonly used azole antifungal drugs; clotrimazole, miconazole, ketoconazole and fluconazole in vitro using the H295R cell assay and two recombinant, CYP17A1 and CYP19A1 (aromatase), assays. Fluconazole 181-192 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 262-269 31330226-6 2019 Clotrimazole inhibited steroid production in a dose-dependent manner with IC50 values for CYP17A1 and CYP19A1 in the range 0.017-0.184 muM. Steroids 23-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 102-109 31701078-3 2019 We previously reported that FOXL2C134W stimulates CYP19 transcription synergistically with SMAD3, leading to elevated estradiol synthesis in a human granulosa cell line (HGrC1). Estradiol 118-127 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-55 31075703-4 2019 Results showed that BPA at 100 muM decreased estradiol level and CYP19A1 mRNA, but increased progesterone production, steroidogenic acute regulatory protein (STAR) and peroxisome proliferator-activated receptor gamma (PPARgamma) mRNA expression after 48 h. Shorter (6 h) exposure to BPA elevated PPARgamma mRNA level in hCGC. bisphenol A 20-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-72 31199532-4 2019 Furthermore, the reaction of 1OOCm with 2,6-di-tert-butylphenol derivatives produces the corresponding phenoxyl radical species (ArO. ) 2,6-di-tert-butylphenol 40-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 31199532-4 2019 Furthermore, the reaction of 1OOCm with 2,6-di-tert-butylphenol derivatives produces the corresponding phenoxyl radical species (ArO. ) phenoxy radical 103-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 31174145-8 2019 PFUA, PFNA, PFDA, PFHxS, and PFASs were associated with higher P450arom levels. perfluorononanoic acid 6-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-72 31174145-8 2019 PFUA, PFNA, PFDA, PFHxS, and PFASs were associated with higher P450arom levels. perfluorodecanoic acid 12-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-72 31002529-0 2019 Mammary adipose stromal cells derived from obese women reduce sensitivity to the aromatase inhibitor anastrazole in an organotypic breast model. Anastrozole 101-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-90 30951242-6 2019 Addition of total PDE inhibitor 3-isobutyl-1-methylxanthine prevented T-2 toxin"s action on LHCGR, STAR, and CYP19A1 mRNA expression in FSH-stimulated human cumulus granulosa cells. 1-Methyl-3-isobutylxanthine 32-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 109-116 31002529-2 2019 To compare how stromal cells derived from women who are lean or obese influence response to the aromatase inhibitor (anastrazole), we incorporated patient-derived stroma in a previously characterized MCF7-derived in vitro duct model. Anastrozole 117-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-105 31002529-4 2019 The effects of T were inhibited by the ER antagonist tamoxifen and aromatase inhibitor anastrazole and were increased by the aromatase inducer dexamethasone. Anastrozole 87-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-76 31002529-4 2019 The effects of T were inhibited by the ER antagonist tamoxifen and aromatase inhibitor anastrazole and were increased by the aromatase inducer dexamethasone. Dexamethasone 143-156 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-134 31175839-5 2019 RESULTS Seven key target genes for formononetin in the treatment of patients with AD were identified, including estrogen receptor alpha (ESR1), peroxisome proliferator-activated receptor gamma (PPARG), tumor protein p53 (TP53), sirtuin 1 (SIRT1), tumor necrosis factor (TNF), cytochrome P450 19A1 (CYP19A1), and nuclear factor (erythroid-derived 2)-like 2 (NFE2L2). formononetin 35-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 276-296 31344684-9 2019 Furthermore, we treated the human granulosa cell line KGN with different ethanol concentrations (15, 30, 60, 120 mM) and found that the expression of IGF1 signaling pathway components, 3beta-HSD, and P450arom, as well as the production of E2, was increased. Ethanol 73-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 200-208 31175839-5 2019 RESULTS Seven key target genes for formononetin in the treatment of patients with AD were identified, including estrogen receptor alpha (ESR1), peroxisome proliferator-activated receptor gamma (PPARG), tumor protein p53 (TP53), sirtuin 1 (SIRT1), tumor necrosis factor (TNF), cytochrome P450 19A1 (CYP19A1), and nuclear factor (erythroid-derived 2)-like 2 (NFE2L2). formononetin 35-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 298-305 30959089-7 2019 In vitro, nicotine decreased P450arom expression and estradiol production in human granulosa cell line KGN. Nicotine 10-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-37 31170164-8 2019 Furthermore, treatment with TNF-alpha promoted the production of testosterone in KGN granulosa cells by reducing CYP19A1 expression, whereas ETA treatment blocked this process. Testosterone 65-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-120 30478740-5 2019 RESULTS: CdCl2 exposure of HUVECs induced a significant increase of ERbeta and Cyp19a1 at both mRNA and protein levels, while a drastic dose-dependent decrease of AR expression level was observed after 24 h of exposure. Cadmium Chloride 9-14 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-86 30868236-2 2019 This study investigated the significance of genetic polymorphism in CYP19A1, which encodes aromatase that catalyzes androgens into estrogens, among men treated with primary ADT for metastatic prostate cancer. 1,4-androstadiene-3,17-dione 173-176 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-75 30084008-2 2019 The discovery that the brain expresses the enzyme aromatase, which produces estradiol from testosterone, expanded this traditional concept. Estradiol 76-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-59 30084008-2 2019 The discovery that the brain expresses the enzyme aromatase, which produces estradiol from testosterone, expanded this traditional concept. Testosterone 91-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-59 30868236-6 2019 When adjusted by Gleason score, initial PSA, M-stage and serum testosterone level during ADT, heterozygous/homozygous variant (AC/CC) in the CYP19A1 gene was associated with a lower risk of progression to castration resistance [hazard ratio (95% confidence interval), 0.53 [0.29-0.92], p = 0.025], but not to any-cause death [hazard ratio (95% confidence interval), 0.74 [0.36-1.49], p = 0.40]. Testosterone 63-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 141-148 30868236-6 2019 When adjusted by Gleason score, initial PSA, M-stage and serum testosterone level during ADT, heterozygous/homozygous variant (AC/CC) in the CYP19A1 gene was associated with a lower risk of progression to castration resistance [hazard ratio (95% confidence interval), 0.53 [0.29-0.92], p = 0.025], but not to any-cause death [hazard ratio (95% confidence interval), 0.74 [0.36-1.49], p = 0.40]. 1,4-androstadiene-3,17-dione 89-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 141-148 30868236-7 2019 CONCLUSIONS: These findings suggest that genetic variation in CYP19A1 (rs1870050) might affect the prognosis of patients with metastatic prostate cancer when treated with ADT by regulating serum testosterone levels. 1,4-androstadiene-3,17-dione 171-174 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-69 30868236-7 2019 CONCLUSIONS: These findings suggest that genetic variation in CYP19A1 (rs1870050) might affect the prognosis of patients with metastatic prostate cancer when treated with ADT by regulating serum testosterone levels. Testosterone 195-207 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-69 30896833-7 2019 The mRNA and protein expression of CYP17A1, HSD17B3 and CYP19A1, which are associated with the synthesis of dehydroepiandrosterone (DHEA) and E2, was elevated at different gestational ages in human placenta. Dehydroepiandrosterone 108-130 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-63 30541132-9 2019 RESULTS: FSH and GB treatment increased CYP19A1 promoter activity, mRNA, and protein levels as well as estradiol when compared with cells treated with FSH only. gb 17-19 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 40-47 30541132-13 2019 CONCLUSION: The combination of GDF9 and BMP15 potently stimulates the effect of FSH and cAMP on CYP19a1 promoter activity and mRNA/protein levels. Cyclic AMP 88-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-103 30896833-7 2019 The mRNA and protein expression of CYP17A1, HSD17B3 and CYP19A1, which are associated with the synthesis of dehydroepiandrosterone (DHEA) and E2, was elevated at different gestational ages in human placenta. Dehydroepiandrosterone 132-136 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-63 29804490-0 2019 Aromatase inhibition by 2-methyl indole hydrazone derivatives evaluated via molecular docking and in vitro activity studies. 2-methyl indole hydrazone 24-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 30896833-7 2019 The mRNA and protein expression of CYP17A1, HSD17B3 and CYP19A1, which are associated with the synthesis of dehydroepiandrosterone (DHEA) and E2, was elevated at different gestational ages in human placenta. Estradiol 142-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-63 29804490-3 2019 Melatonin, a natural indolic hormone, is reported to prevent free radical induced carcinogenesis and block local estrogen synthesis in breast tissue via aromatase inhibition. Melatonin 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 153-162 29804490-7 2019 In biological activity assays monochloro substituted indole hydrazones were found to have stronger aromatase inhibitory activity among all tested derivatives and were more active than melatonin. monochloro substituted indole hydrazones 30-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-108 29804490-5 2019 In the present study aromatase inhibiting potential of 2-methyl indole hydrazones are investigated, and compared with melatonin, by two in vitro models; a cell-free assay using a fluorescence substrate and a cell-based assay where cell proliferation was determined in ER + human breast cancer cells (MCF-7 BUS) in the absence of estrogen and the presence of testosterone. 2-methyl indole hydrazones 55-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 29804490-7 2019 In biological activity assays monochloro substituted indole hydrazones were found to have stronger aromatase inhibitory activity among all tested derivatives and were more active than melatonin. Melatonin 184-193 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-108 29804490-9 2019 These results may be useful in the design and synthesis of novel melatonin analogues with higher inhibitory potency against aromatase. Melatonin 65-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 124-133 31028061-0 2019 Effects of the Dibutyl Phthalate (DBP) on the Expression and Activity of Aromatase in Human Granulosa Cell Line KGN. Dibutyl Phthalate 15-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-82 30997616-0 2019 Acupuncture and Vitamin D for the Management of Aromatase Inhibitor-Induced Arthralgia. Vitamin D 16-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-57 30979376-16 2019 We also found that melatonin could up-regulates aromatase mRNA, VEGF mRNA expression and down-regulates iNOS mRNA expression in the granulosa cells. Melatonin 19-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-57 31006191-0 2019 [Effects of metformin on the expression of estrogen synthetase and ER mRNA in uterine leiomyoma tissues]. Metformin 12-21 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-62 31006191-1 2019 Objective: To elucidate whether metformin could regulate the mRNA expression level of estrogen synthetase and ER in human uterine leiomyoma tissues. Metformin 32-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-105 31006191-11 2019 (2) After cultured with different concentrations of metformin (10, 50 and 100 mumol/L), the P450arom mRNA levels in the uterine leiomyoma tissues were 9+-4, 8+-5 and 8+-3 respectively in the treatment groups and was 16+-5 in the control group. Metformin 52-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-100 30982255-6 2019 We demonstrate BKM120 inhibits growth, generation of drug-resistant derivatives and SC formation in BCSC-CYP19 and MCF-7-CYP19. NVP-BKM120 15-21 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 105-110 30982255-6 2019 We demonstrate BKM120 inhibits growth, generation of drug-resistant derivatives and SC formation in BCSC-CYP19 and MCF-7-CYP19. NVP-BKM120 15-21 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 121-126 30982255-7 2019 Result: BKM120 could inhibit BCSC-CYP19 growth by dose-dependence, reduce migration and colony formation of BCSC-CYP19, and also significantly reduced expression of PI3K, Akt1 and S6. NVP-BKM120 8-14 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-39 30982255-7 2019 Result: BKM120 could inhibit BCSC-CYP19 growth by dose-dependence, reduce migration and colony formation of BCSC-CYP19, and also significantly reduced expression of PI3K, Akt1 and S6. NVP-BKM120 8-14 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-118 30982255-8 2019 Combined BKM120 and letrozolecaninhibit BCSC-CYP19 growth and proliferation, make BCSC-CYP19 stayed in G0-G1 phase increasing significantly to induce early apoptosis, and down-regulate expression of PI3K, Akt1 and S6. NVP-BKM120 9-15 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-50 30982255-8 2019 Combined BKM120 and letrozolecaninhibit BCSC-CYP19 growth and proliferation, make BCSC-CYP19 stayed in G0-G1 phase increasing significantly to induce early apoptosis, and down-regulate expression of PI3K, Akt1 and S6. NVP-BKM120 9-15 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-92 31028061-0 2019 Effects of the Dibutyl Phthalate (DBP) on the Expression and Activity of Aromatase in Human Granulosa Cell Line KGN. Dibutyl Phthalate 34-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-82 31028061-4 2019 Here the effect of DBP on P450 aromatase, a rate-limiting enzyme stimulated by FSH in the estradiol synthesis, was investigated in human granulosa cell line KGN. Estradiol 90-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 31028061-6 2019 Then the expression of aromatase was assessed, and the synthesis of estradiol was detected to reflect aromatase activity. Estradiol 68-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 102-111 30353958-0 2019 A 5alpha-reductase (SRD5A2) polymorphism is associated with serum testosterone and sex hormone-binding globulin in men, while aromatase (CYP19A1) polymorphisms are associated with oestradiol and luteinizing hormone reciprocally. Estradiol 180-190 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 137-144 29536789-9 2019 We aimed to emphasize that neurological side effect like hallucination may rarely occur during the treatment of anastrozole and in case of aromatase inhibitor-related hallucinations, switching to another aromatase inhibitor (letrozole) can be a treatment option. Anastrozole 112-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 204-213 29536789-9 2019 We aimed to emphasize that neurological side effect like hallucination may rarely occur during the treatment of anastrozole and in case of aromatase inhibitor-related hallucinations, switching to another aromatase inhibitor (letrozole) can be a treatment option. Letrozole 225-234 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-148 29536789-9 2019 We aimed to emphasize that neurological side effect like hallucination may rarely occur during the treatment of anastrozole and in case of aromatase inhibitor-related hallucinations, switching to another aromatase inhibitor (letrozole) can be a treatment option. Letrozole 225-234 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 204-213 30518737-4 2019 Knockdown of Clock gene by siRNA resulted in a significant reduction of estradiol production by inhibiting P450arom expression, while it induced a significant increase of progesterone production by upregulating 3betaHSD in KGN cells treated with forskolin. Estradiol 72-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-115 30518737-4 2019 Knockdown of Clock gene by siRNA resulted in a significant reduction of estradiol production by inhibiting P450arom expression, while it induced a significant increase of progesterone production by upregulating 3betaHSD in KGN cells treated with forskolin. Colforsin 246-255 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-115 30353958-9 2019 CYP19A1 polymorphisms were associated with higher serum E2 and lower LH in reciprocal fashion, from which the two-copy haplotype rs10046 = T/rs2899470 = G/rs11575899 = I/rs700518 = G/rs17703883 = T was associated with higher E2 (63.4 vs. 56.5 pmol/L, P = 0.001) and lower LH (3.9 vs. 4.5 IU/L, P = 0.001) compared to null copies. Estradiol 56-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-7 30353958-9 2019 CYP19A1 polymorphisms were associated with higher serum E2 and lower LH in reciprocal fashion, from which the two-copy haplotype rs10046 = T/rs2899470 = G/rs11575899 = I/rs700518 = G/rs17703883 = T was associated with higher E2 (63.4 vs. 56.5 pmol/L, P = 0.001) and lower LH (3.9 vs. 4.5 IU/L, P = 0.001) compared to null copies. Estradiol 225-227 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-7 30442903-4 2018 We performed a positron emission tomography (PET) study in 21 healthy subjects using 11C-cetrozole, which has high selectivity and affinity for aromatase. cetrozole 85-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 144-153 30425102-4 2019 Here we report the X-ray crystal structure of human CYP11B1 (at 2.1 A resolution) in complex with fadrozole, a racemic compound normally used to treat breast cancer by inhibiting estrogen-producing CYP19A1. Fadrozole 98-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 198-205 30366299-5 2019 Regardless, BADGE 2HCl and BADGE showed higher cytotoxicity than BADGE H2O, which was the only compound that significantly inhibited CYP19 activity (IC50 49 +- 5 muM). Water 71-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 133-138 30366299-8 2019 The results evidence the ability of BADGE and derivatives to affect placental lipid handling and to modulate placental CYP19 activity (BADGE H2O) and highlights the need to monitor human exposure to these compounds, at least as intensely as BPA is monitored. Water 141-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 119-124 30020241-7 2019 In E-PE placenta, the productions and activities of 17beta-hydroxysteroid dehydrogenases 3 and aromatase, the essential enzymes for testosterone and estradiol synthesis, were compromised. Testosterone 132-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-104 30020241-7 2019 In E-PE placenta, the productions and activities of 17beta-hydroxysteroid dehydrogenases 3 and aromatase, the essential enzymes for testosterone and estradiol synthesis, were compromised. Estradiol 149-158 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-104 30118172-2 2019 We have evaluated in vitro the effects of two synthetic isoflavones, methoxyisoflavone and ipriflavone, on the catalytic activity of human aromatase (CYP19), the enzyme catalyzing the conversion of androgens (ie, testosterone or androstenedione) to estrogens (ie, estradiol and estrone). Isoflavones 56-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 150-155 30118172-2 2019 We have evaluated in vitro the effects of two synthetic isoflavones, methoxyisoflavone and ipriflavone, on the catalytic activity of human aromatase (CYP19), the enzyme catalyzing the conversion of androgens (ie, testosterone or androstenedione) to estrogens (ie, estradiol and estrone). methoxyisoflavone 69-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 150-155 30118172-2 2019 We have evaluated in vitro the effects of two synthetic isoflavones, methoxyisoflavone and ipriflavone, on the catalytic activity of human aromatase (CYP19), the enzyme catalyzing the conversion of androgens (ie, testosterone or androstenedione) to estrogens (ie, estradiol and estrone). ipriflavone 91-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 150-155 30118172-2 2019 We have evaluated in vitro the effects of two synthetic isoflavones, methoxyisoflavone and ipriflavone, on the catalytic activity of human aromatase (CYP19), the enzyme catalyzing the conversion of androgens (ie, testosterone or androstenedione) to estrogens (ie, estradiol and estrone). Testosterone 213-225 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 150-155 30118172-2 2019 We have evaluated in vitro the effects of two synthetic isoflavones, methoxyisoflavone and ipriflavone, on the catalytic activity of human aromatase (CYP19), the enzyme catalyzing the conversion of androgens (ie, testosterone or androstenedione) to estrogens (ie, estradiol and estrone). Androstenedione 229-244 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 150-155 30118172-2 2019 We have evaluated in vitro the effects of two synthetic isoflavones, methoxyisoflavone and ipriflavone, on the catalytic activity of human aromatase (CYP19), the enzyme catalyzing the conversion of androgens (ie, testosterone or androstenedione) to estrogens (ie, estradiol and estrone). Estradiol 264-273 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 150-155 30118172-2 2019 We have evaluated in vitro the effects of two synthetic isoflavones, methoxyisoflavone and ipriflavone, on the catalytic activity of human aromatase (CYP19), the enzyme catalyzing the conversion of androgens (ie, testosterone or androstenedione) to estrogens (ie, estradiol and estrone). Estrone 278-285 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 150-155 30688541-6 2019 RESULTS: Patients with the variant homozygous genotype AA of CYP19A1 (rs10046) showed increased serum concentrations of estradiol when compared to patients with other genotypes (p = 0.005). Estradiol 120-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-68 30688541-9 2019 CONCLUSION: The CYP19A1 variant is associated with an estradiol imbalance in serum. Estradiol 54-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-23 30284180-11 2018 Moreover, the cell proliferation of high CYP19A1-expressing KKU-213 cells was more profoundly suppressed by CYP19A1 inhibitors (exemestane and letrozole) than low CYP19A1-expressing KKU-100 cells. exemestane 128-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-48 30284180-11 2018 Moreover, the cell proliferation of high CYP19A1-expressing KKU-213 cells was more profoundly suppressed by CYP19A1 inhibitors (exemestane and letrozole) than low CYP19A1-expressing KKU-100 cells. exemestane 128-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-115 30284180-11 2018 Moreover, the cell proliferation of high CYP19A1-expressing KKU-213 cells was more profoundly suppressed by CYP19A1 inhibitors (exemestane and letrozole) than low CYP19A1-expressing KKU-100 cells. exemestane 128-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-115 30284180-11 2018 Moreover, the cell proliferation of high CYP19A1-expressing KKU-213 cells was more profoundly suppressed by CYP19A1 inhibitors (exemestane and letrozole) than low CYP19A1-expressing KKU-100 cells. Letrozole 143-152 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-48 30284180-11 2018 Moreover, the cell proliferation of high CYP19A1-expressing KKU-213 cells was more profoundly suppressed by CYP19A1 inhibitors (exemestane and letrozole) than low CYP19A1-expressing KKU-100 cells. Letrozole 143-152 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-115 30284180-11 2018 Moreover, the cell proliferation of high CYP19A1-expressing KKU-213 cells was more profoundly suppressed by CYP19A1 inhibitors (exemestane and letrozole) than low CYP19A1-expressing KKU-100 cells. Letrozole 143-152 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-115 29723543-1 2018 The cytochrome P450 family 19 subfamily A member 1 (CYP19A1) gene, encodes aromatase, a key enzyme in estradiol (E2) synthesis, and is down-regulated during porcine follicular atresia. Estradiol 102-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-50 29723543-1 2018 The cytochrome P450 family 19 subfamily A member 1 (CYP19A1) gene, encodes aromatase, a key enzyme in estradiol (E2) synthesis, and is down-regulated during porcine follicular atresia. Estradiol 102-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-59 29723543-7 2018 These findings provide further insight into the mechanisms of CYP19A1 in steroid hormone synthesis and GC apoptosis and provide molecular targets for exploring methods of treatment for steroid-dependent reproductive disorders. Steroids 73-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-69 29723543-7 2018 These findings provide further insight into the mechanisms of CYP19A1 in steroid hormone synthesis and GC apoptosis and provide molecular targets for exploring methods of treatment for steroid-dependent reproductive disorders. Steroids 73-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-69 29287126-7 2018 However, the aromatase inhibitors had almost equivalent impacts on high-density liportein cholesterol, low-density lipoprotein cholesterol, and triglyceride after long-term aromatase inhibitor treatment. high-density liportein cholesterol 67-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 29287126-7 2018 However, the aromatase inhibitors had almost equivalent impacts on high-density liportein cholesterol, low-density lipoprotein cholesterol, and triglyceride after long-term aromatase inhibitor treatment. Triglycerides 144-156 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 30386297-1 2018 Diverse cognitive functions in many vertebrate species are influenced by local conversion of androgens to 17beta-estradiol (E2) by aromatase. Estradiol 106-122 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 131-140 29131373-7 2018 Effect of overexpression of miR-210 was analyzed on granulosa cell marker genes (CYP19A1 and PCNA) which were significantly downregulated (P < 0.05). mir-210 28-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-88 30114613-3 2018 To determine if evodiamine might be useful in the treatment of thyroid cancer and its cytotoxic mechanism, we analyzed the impact of evodiamine treatment on differential protein expression in human thyroid cancer cell line ARO using lysine-labeling two-dimensional difference gel electrophoresis (2D-DIGE) combined with mass spectrometry (MS). evodiamine 133-143 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 223-226 30216511-0 2018 Let-7g inhibits synthesis of estradiol by downregulating activity of aromatase in JEG3 cells. Estradiol 29-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-78 30374289-1 2018 Aromatase is the enzyme responsible for converting testosterone to estradiol. Testosterone 51-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 30374289-1 2018 Aromatase is the enzyme responsible for converting testosterone to estradiol. Estradiol 67-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 29718108-7 2018 The ability of LAM to bind to CYP17A1, CYP19A1, and CYP21A2 was investigated using docking and molecular dynamics simulations. Lamotrigine 15-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-46 29718108-9 2018 The molecular dynamics simulations also suggested binding of LAM to the heme iron in the CYP19A1 active site. Lamotrigine 61-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 89-96 29718108-9 2018 The molecular dynamics simulations also suggested binding of LAM to the heme iron in the CYP19A1 active site. Heme 72-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 89-96 29718108-9 2018 The molecular dynamics simulations also suggested binding of LAM to the heme iron in the CYP19A1 active site. Iron 77-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 89-96 29921733-6 2018 Liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis revealed that aromatase is basally acetylated on several lysine residues (108, 169, 242, 262, 334, 352, and 354) in MCF-7 cells, and treatment with a SIRT-1 inhibitor induced additional acetylation (376, 390, 440, and 448). Lysine 124-130 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-90 29921733-7 2018 These acetylated lysine residues are in regions critical for aromatase activity. Lysine 17-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-70 29773500-0 2018 An analysis on distribution and inter-relationships of biomarkers under rivaroxaban in Japanese patients with non-valvular atrial fibrillation (CVI ARO 1). Rivaroxaban 72-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 148-153 30043503-4 2018 Aromatase, the enzyme responsible for the last step of estrogen biosynthesis converting testosterone and androgen to estrogen, was previously reported to be more abundant in endometriotic tissues than in the normal endometrium, leading to an increased local estrogen concentration. Testosterone 88-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 29873684-1 2018 Background: The German rectal cancer trial CAO/ARO/AIO-04 has shown a significant benefit in 3-year disease-free survival (DFS) of adding oxaliplatin to a standard preoperative 5-fluorouracil (5-FU)-based chemoradiotherapy (CRT) and adjuvant chemotherapy in patients with locally advanced rectal cancer. Oxaliplatin 138-149 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-50 29476820-0 2018 Testosterone complex and non-steroidal ligands of human aromatase. Testosterone 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-65 29976866-4 2018 Comparing the samples from fibroblasts cultured with CsA and those cultured without, up-regulated changes of CYP19A1, 1B1, 7A1, 7F1, 17A1 and down-regulated 2D6 gene expression were observed. Cyclosporine 53-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 109-116 29476820-2 2018 Previously crystal structures of human AROM in complex with the substrate androstenedione, and inhibitors exemestane, as well as the newly designed steroidal compounds, have been reported. Androstenedione 74-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-43 29476820-10 2018 Interestingly, polyethylene glycol exhibits weak inhibition of AROM enzyme activity in a time dependent manner. Polyethylene Glycols 15-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-67 29476820-2 2018 Previously crystal structures of human AROM in complex with the substrate androstenedione, and inhibitors exemestane, as well as the newly designed steroidal compounds, have been reported. exemestane 106-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-43 29476820-3 2018 Here we report the first crystal structure of testosterone complex of human placental AROM. Testosterone 46-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-90 29476820-6 2018 The heme proximal region comprises the intermolecular interface in AROM, and also the putative interaction surface of its redox partner cytochrome P450 reductase. Heme 4-8 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-71 29476820-8 2018 Using five best X-ray data sets from androstenedione and testosterone complexes of AROM, we now unequivocally show the presence of an unexplained ligand electron density inside the proximal cavity. Androstenedione 37-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-87 29476820-8 2018 Using five best X-ray data sets from androstenedione and testosterone complexes of AROM, we now unequivocally show the presence of an unexplained ligand electron density inside the proximal cavity. Testosterone 57-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-87 29397057-2 2018 Activation of the cAMP/protein kinase A (PKA)/cAMP response element-binding protein (CREB) pathway plays a crucial role in FSH regulation of CYP19A1 in human ovarian granulosa cells. Cyclic AMP 18-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 141-148 29654831-8 2018 This was mirrored by significant downregulating of the expression of estrogen and catechol estrogen-synthesizing enzymes (CYP19, CYP1A1 and CYP1B1) within prostate cancer cells. catechol 82-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-127 29397057-2 2018 Activation of the cAMP/protein kinase A (PKA)/cAMP response element-binding protein (CREB) pathway plays a crucial role in FSH regulation of CYP19A1 in human ovarian granulosa cells. Cyclic AMP 46-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 141-148 29266331-5 2018 The CYP19-RT-LAMP assay rapidly identified the LPS-induced downregulation of the CYP19 gene within 30 min at 63 C in a water bath. Water 119-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-86 29701941-0 2018 Effects of Neonicotinoid Pesticides on Promoter-Specific Aromatase (CYP19) Expression in Hs578t Breast Cancer Cells and the Role of the VEGF Pathway. Neonicotinoids 11-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-73 29797697-0 2018 Aromatase inhibitors (letrozole) for subfertile women with polycystic ovary syndrome. Letrozole 22-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 30364888-3 2018 These benefits are mediated primarily through conversion of a small amount of testosterone to estradiol by the catalytic activity of estrogen synthetase (aromatase cytochrome P450 enzyme). Testosterone 78-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 133-152 30364888-3 2018 These benefits are mediated primarily through conversion of a small amount of testosterone to estradiol by the catalytic activity of estrogen synthetase (aromatase cytochrome P450 enzyme). Estradiol 94-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 133-152 29622348-1 2018 The steroidogenic enzyme P450 aromatase (ARO) has a key role in the conversion of testosterone (T) into estrogens (E), expressed as 17beta-estradiol. Testosterone 82-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-44 29622348-1 2018 The steroidogenic enzyme P450 aromatase (ARO) has a key role in the conversion of testosterone (T) into estrogens (E), expressed as 17beta-estradiol. Estradiol 132-148 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-44 29546425-4 2018 Human cytotrophoblasts cultured in 20% O2 spontaneously differentiate to syncytiotrophoblast with induction of hCYP191A/aromatase, a marker of differentiation. Oxygen 39-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-129 29266331-5 2018 The CYP19-RT-LAMP assay rapidly identified the LPS-induced downregulation of the CYP19 gene within 30 min at 63 C in a water bath. Water 119-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-9 29474975-4 2018 The cAMP inducer forskolin (10 muM) and protein kinase C stimulant phorbol myristate acetate (1 muM) increased CYP19A1 mRNA levels by 1.8- and 1.6-fold, respectively, as well as inducing aromatase catalytic activity. Cyclic AMP 4-8 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-118 29474975-4 2018 The cAMP inducer forskolin (10 muM) and protein kinase C stimulant phorbol myristate acetate (1 muM) increased CYP19A1 mRNA levels by 1.8- and 1.6-fold, respectively, as well as inducing aromatase catalytic activity. Colforsin 17-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-118 29474975-4 2018 The cAMP inducer forskolin (10 muM) and protein kinase C stimulant phorbol myristate acetate (1 muM) increased CYP19A1 mRNA levels by 1.8- and 1.6-fold, respectively, as well as inducing aromatase catalytic activity. Tetradecanoylphorbol Acetate 67-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-118 29474975-5 2018 Dexamethasone (100 nM) and vascular endothelial growth factor (5 ng/mL) decreased CYP19A1 mRNA levels, while having no effect on aromatase activity. Dexamethasone 0-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-89 29182028-5 2018 However, positive correlation between CYP19A1 and LPAR1 accounts for supporting role of LPA acting via LPAR1 in intratumoral DHT concentration and the ethiology of endometrial cancer progression. Dihydrotestosterone 125-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-45 28854867-3 2018 We demonstrated that spontaneously differentiated murine EBs (mEBs) and human EBs (hEBs) displayed ovarian GC markers, such as aromatase (CYP19A1), FOXL2, AMHR2, FSHR, and GJA1. hebs 83-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-145 29701941-5 2018 OBJECTIVE: We aimed to determine the effects of two neonicotinoids (thiacloprid and imidacloprid) on promoter-specific CYP19 expression in Hs578t breast cancer cells and understand the signaling pathways involved. Neonicotinoids 52-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 119-124 29701941-5 2018 OBJECTIVE: We aimed to determine the effects of two neonicotinoids (thiacloprid and imidacloprid) on promoter-specific CYP19 expression in Hs578t breast cancer cells and understand the signaling pathways involved. thiacloprid 68-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 119-124 29701941-5 2018 OBJECTIVE: We aimed to determine the effects of two neonicotinoids (thiacloprid and imidacloprid) on promoter-specific CYP19 expression in Hs578t breast cancer cells and understand the signaling pathways involved. imidacloprid 84-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 119-124 29701941-6 2018 METHODS: Hs578t cells were exposed to various signaling pathway stimulants or neonicotinoids for 24 h. Promoter-specific expression of CYP19 was determined by real-time quantitative polymerase chain reaction and catalytic activity of aromatase by tritiated water release assay. Water 257-262 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-140 29701941-9 2018 Exposure of Hs578t cells to environmental concentrations of imidacloprid and thiacloprid resulted in a switch in CYP19 promoter usage, involving inhibition of I.4 promoter activity and an increase of PII, I.3, and I.7 promoter-mediated CYP19 expression and aromatase catalytic activity. imidacloprid 60-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-118 29701941-9 2018 Exposure of Hs578t cells to environmental concentrations of imidacloprid and thiacloprid resulted in a switch in CYP19 promoter usage, involving inhibition of I.4 promoter activity and an increase of PII, I.3, and I.7 promoter-mediated CYP19 expression and aromatase catalytic activity. imidacloprid 60-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 236-241 29701941-9 2018 Exposure of Hs578t cells to environmental concentrations of imidacloprid and thiacloprid resulted in a switch in CYP19 promoter usage, involving inhibition of I.4 promoter activity and an increase of PII, I.3, and I.7 promoter-mediated CYP19 expression and aromatase catalytic activity. thiacloprid 77-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-118 29701941-9 2018 Exposure of Hs578t cells to environmental concentrations of imidacloprid and thiacloprid resulted in a switch in CYP19 promoter usage, involving inhibition of I.4 promoter activity and an increase of PII, I.3, and I.7 promoter-mediated CYP19 expression and aromatase catalytic activity. thiacloprid 77-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 236-241 29701941-12 2018 CONCLUSIONS: We demonstrated in vitro that neonicotinoids may stimulate a change in CYP19 promoter usage similar to that observed in patients with hormone-dependent breast cancer. Neonicotinoids 43-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-89 29484793-2 2018 The bulky phenol ligand ArOH (Ar=2,6-Dipp2 C6 H3 , Dipp=2,6-diisopropylphenyl) was utilized to construct low-coordinate lanthanide compound [(ArO)Ln(OAr")] (Ar"=6-Dipp-2-(2"-i Pr-6"-CHMe(CH2- )C6 H3 )C6 H3 O- ; Ln=Tb, Dy, Ho, Er, Tm). Phenol 10-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-27 29765553-0 2018 Acquired resistance to everolimus in aromatase inhibitor-resistant breast cancer. Everolimus 23-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-46 29484793-2 2018 The bulky phenol ligand ArOH (Ar=2,6-Dipp2 C6 H3 , Dipp=2,6-diisopropylphenyl) was utilized to construct low-coordinate lanthanide compound [(ArO)Ln(OAr")] (Ar"=6-Dipp-2-(2"-i Pr-6"-CHMe(CH2- )C6 H3 )C6 H3 O- ; Ln=Tb, Dy, Ho, Er, Tm). 2,6-dipp2 33-42 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-27 29484793-2 2018 The bulky phenol ligand ArOH (Ar=2,6-Dipp2 C6 H3 , Dipp=2,6-diisopropylphenyl) was utilized to construct low-coordinate lanthanide compound [(ArO)Ln(OAr")] (Ar"=6-Dipp-2-(2"-i Pr-6"-CHMe(CH2- )C6 H3 )C6 H3 O- ; Ln=Tb, Dy, Ho, Er, Tm). 2,6-diisopropylphenyl 56-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-27 29484793-2 2018 The bulky phenol ligand ArOH (Ar=2,6-Dipp2 C6 H3 , Dipp=2,6-diisopropylphenyl) was utilized to construct low-coordinate lanthanide compound [(ArO)Ln(OAr")] (Ar"=6-Dipp-2-(2"-i Pr-6"-CHMe(CH2- )C6 H3 )C6 H3 O- ; Ln=Tb, Dy, Ho, Er, Tm). Lanthanoid Series Elements 120-130 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-27 29484793-2 2018 The bulky phenol ligand ArOH (Ar=2,6-Dipp2 C6 H3 , Dipp=2,6-diisopropylphenyl) was utilized to construct low-coordinate lanthanide compound [(ArO)Ln(OAr")] (Ar"=6-Dipp-2-(2"-i Pr-6"-CHMe(CH2- )C6 H3 )C6 H3 O- ; Ln=Tb, Dy, Ho, Er, Tm). 6-dipp 35-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-27 28859903-5 2018 In addition, bazedoxifene enhanced expression of ERalpha-regulated Cyp19a1 mRNA. bazedoxifene 13-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-74 29474090-4 2018 This new reaction was successfully utilized in the synthesis of the potent CYP19 aromatase inhibitor and late-stage functionalization on the bioactive complex estrone. Estrone 159-166 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-80 29630096-0 2018 Effects of testosterone on PPARgamma and P450arom expression in polycystic ovary syndrome patients and related mechanisms. Testosterone 11-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-49 29630096-2 2018 This study recruited PCOS patients and adopted testosterone therapy to analyze its effect on expression of nuclear peroxisome proliferation activated receptor gamma (PPARgamma), and cytochrome P450 aromatase (P450arom) in ovary granular cells of patients. Testosterone 47-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 209-217 29630096-6 2018 The treatment of 10 nmol/l testosterone significantly elevated PPARgamma mRNA and protein expression, and decreased P450arom expression (p < 0.05). Testosterone 27-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-124 29630096-8 2018 CONCLUSIONS: Testosterone causes hyperandrogenism microenvironment of PCOS patients, with a correlated increase of PPARgamma and reduction of P450arom. Testosterone 13-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 142-150 28610873-3 2018 It has been reported that 1alpha,25-dihydroxyvitamin D3 down-regulates the expression of the CYP19A1 gene, encoding the aromatase enzyme that catalyzes the synthesis of estradiol. Calcitriol 26-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-100 28610873-3 2018 It has been reported that 1alpha,25-dihydroxyvitamin D3 down-regulates the expression of the CYP19A1 gene, encoding the aromatase enzyme that catalyzes the synthesis of estradiol. Estradiol 169-178 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-100 28610873-6 2018 We have investigated the molecular mechanisms for the 1alpha,25-dihydroxyvitamin D3-mediated down-regulation of CYP19A1 and ERalpha gene expression in human MCF-7 breast cancer cells and found that Williams syndrome transcription factor (WSTF) plays a key role by binding to the promoters of CYP19A1 and ERalpha. 25-dihydroxyvitamin d3 61-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 112-119 28610873-6 2018 We have investigated the molecular mechanisms for the 1alpha,25-dihydroxyvitamin D3-mediated down-regulation of CYP19A1 and ERalpha gene expression in human MCF-7 breast cancer cells and found that Williams syndrome transcription factor (WSTF) plays a key role by binding to the promoters of CYP19A1 and ERalpha. 25-dihydroxyvitamin d3 61-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 292-299 28610873-9 2018 When cells were treated with 1alpha,25-dihydroxyvitamin D3, WSTF was dissociated from the promoters and the promoter activities of CYP19A1 and ERalpha were decreased. Calcitriol 29-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 131-138 29228205-9 2018 CYP19A1 was the only gene harboring SNPs that were significantly associated with risk in both the placebo and finasteride arms. Finasteride 110-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-7 29228121-7 2018 In steroid hormone synthesis, two specific enzymes are important: P450scc (CYP11A1) converts cholesterol to pregnenolone and aromatase (CYP19) induces androgen conversion to estrogen.To determine the effects of a low dose of BPA on hormone synthesis in the placenta, we used JEG-3 cells as a model. Steroids 3-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-141 29228121-8 2018 We found that the steroidogenic genes CYP11A1 and CYP19 were downregulated in human tissues by detectable concentrations of BPA (1-1000 nM), which do not affect cell viability. bisphenol A 124-127 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-55 29150338-11 2018 3beta-HSD and P450arom demonstrated mediating effects in the association between prenatal TCS exposure and testosterone concentrations in cord blood. Testosterone 107-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-22 29843142-2 2018 Aromatase is an enzyme that catalyzes estrogen biosynthesis from androgen precursors, and seladin-1 is an enzyme that converts desmosterol to cholesterol, which is the precursor of all hormones. Desmosterol 127-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 29375481-11 2017 At the same time upregulation of estradiol synthesis and CYP19 mRNA expression increased steroid output profoundly. Steroids 89-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-62 29150338-11 2018 3beta-HSD and P450arom demonstrated mediating effects in the association between prenatal TCS exposure and testosterone concentrations in cord blood. Triclosan 90-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-22 29349513-5 2018 For the quantitative description of the effects of the hydrophobic contact and nitrogen-heme-iron coordination on aromatase inhibition, the hydrophobicity density field model and the smallest dual descriptor Deltaf(r) S were introduced, respectively. Nitrogen 79-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 114-123 29349513-5 2018 For the quantitative description of the effects of the hydrophobic contact and nitrogen-heme-iron coordination on aromatase inhibition, the hydrophobicity density field model and the smallest dual descriptor Deltaf(r) S were introduced, respectively. Iron 93-97 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 114-123 28059043-5 2018 In this review, the therapeutic potentials of plant derived natural products belonging to secondary metabolites, namely alkaloids, flavonoids and terpenoids as anticancer molecules, involving various strategies of treatment, have been discussed with special reference to topoisomerases (Topo), cyclooxygenases (COX), lipoxygenase (LOX) and aromatase as enzymatic targets for various types of cancers. Alkaloids 120-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 340-349 29953993-7 2018 Folpet, captan, and captafol inhibited human CYP19A1 with inhibitory concentration (IC50) values of 3.55, 10.68, and 1.14 mumol/L respectively. Captan 8-14 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-52 29953993-7 2018 Folpet, captan, and captafol inhibited human CYP19A1 with inhibitory concentration (IC50) values of 3.55, 10.68, and 1.14 mumol/L respectively. captafol 20-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-52 29953993-9 2018 Molecular docking simulation analysis showed that they tended to bind to the steroid-binding pocket of the CYP19A1. Steroids 77-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-114 29953993-11 2018 CONCLUSION: Folpet, captan, and captafol are potential inhibitors of human CYP19A1. Captan 20-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-82 29953993-11 2018 CONCLUSION: Folpet, captan, and captafol are potential inhibitors of human CYP19A1. captafol 32-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-82 29843142-2 2018 Aromatase is an enzyme that catalyzes estrogen biosynthesis from androgen precursors, and seladin-1 is an enzyme that converts desmosterol to cholesterol, which is the precursor of all hormones. Cholesterol 142-153 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 28649695-3 2017 To identify a potential cellular source of local estrogen, here we examined the expression of aromatase, the enzyme that catalyzes the conversion of testosterone to estradiol. Testosterone 149-161 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-103 28709873-6 2017 (ii) Overexpression of exogenous FZD3 in human granulosa cell COV434 impaired long-term FSH incubation-induced CYP19A1 transactivation and the recruitment of beta-Catenin onto CYP19A1 promoter, and subsequently compromised FSH-stimulated estrogen production. cov434 62-68 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-118 28709873-6 2017 (ii) Overexpression of exogenous FZD3 in human granulosa cell COV434 impaired long-term FSH incubation-induced CYP19A1 transactivation and the recruitment of beta-Catenin onto CYP19A1 promoter, and subsequently compromised FSH-stimulated estrogen production. cov434 62-68 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 176-183 28649695-3 2017 To identify a potential cellular source of local estrogen, here we examined the expression of aromatase, the enzyme that catalyzes the conversion of testosterone to estradiol. Estradiol 165-174 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-103 29153737-0 2017 Structure-activity relationships and docking studies of synthetic 2-arylindole derivatives determined with aromatase and quinone reductase 1. 2-arylindole 66-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-116 29153737-1 2017 In our ongoing effort of discovering anticancer and chemopreventive agents, a series of 2-arylindole derivatives were synthesized and evaluated toward aromatase and quinone reductase 1 (QR1). 2-arylindole 88-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 151-160 29153737-5 2017 Computational docking studies using CDOCKER (Discovery Studio 3.5) provided insight in regard to the potential binding modes of 2-arylindoles within the aromatase active site. 2-arylindoles 128-141 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 153-162 28992603-4 2017 Moreover, two SNPs (CYP17 -34 T:C (MSP AI) and CYP19 T:C (Trp:Arg)) of cytochrome P450, which is involved in steroid metabolism pathways, were analysed between the groups. Tryptophan 58-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-52 28992603-4 2017 Moreover, two SNPs (CYP17 -34 T:C (MSP AI) and CYP19 T:C (Trp:Arg)) of cytochrome P450, which is involved in steroid metabolism pathways, were analysed between the groups. Arginine 62-65 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-52 28992603-4 2017 Moreover, two SNPs (CYP17 -34 T:C (MSP AI) and CYP19 T:C (Trp:Arg)) of cytochrome P450, which is involved in steroid metabolism pathways, were analysed between the groups. Steroids 109-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-52 28649695-8 2017 A few aromatase neurons express Fos after cheek injection of capsaicin, formalin, or chloroquine. Capsaicin 61-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 6-15 28649695-8 2017 A few aromatase neurons express Fos after cheek injection of capsaicin, formalin, or chloroquine. Formaldehyde 72-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 6-15 28573719-12 2017 Moreover, the present data showed that MEN and DEN down-regulated the protein expression of both CYP19A1 and 21A2 in both livers and testes of male rabbits. Diethylnitrosamine 47-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 97-104 28649695-8 2017 A few aromatase neurons express Fos after cheek injection of capsaicin, formalin, or chloroquine. Chloroquine 85-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 6-15 28866654-5 2017 We observed upregulation of Cyp19a1, Hsd17b1, Cyp1a1, Sult1e1 in the DEN-treated livers compared to the control-treated livers using real time PCR. Diethylnitrosamine 69-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-35 29113261-1 2017 It has been hypothesized that single nucleotide polymorphisms in CYP19A1 gene may alter aromatase activity and circulating steroid hormone levels in females. Steroids 123-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-72 28888137-2 2017 In this study, we investigated to what extent the aromatase (CYP19A1) gene, which encodes an enzyme in converting testosterone into estrogen, contributes to subjective well-being and in another self-related disposition: independent and interdependent self-construal. Testosterone 114-126 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-68 28888137-3 2017 In study 1, a meta-analysis showed that the GG genotype of CYP19A1 (a G/A substitution at Val80, rs700518) was associated with higher testosterone and lower estradiol. Testosterone 134-146 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-66 28888137-3 2017 In study 1, a meta-analysis showed that the GG genotype of CYP19A1 (a G/A substitution at Val80, rs700518) was associated with higher testosterone and lower estradiol. Estradiol 157-166 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-66 28977593-9 2017 CB NPs rapidly activated extracellular signal-regulated kinase 1 and 2 in KGN cells and pharmacological inhibition of this signaling pathway using PD 98059 significantly attenuated the inhibitory effects of CB NPs on CYP19A1 gene expression and aromatase activity. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 147-155 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 217-224 28429243-2 2017 Two major reactions contribute to E1 plasma concentrations, aromatase (CYP19A1) catalyzed E1 synthesis from androstenedione and steroid sulfatase (STS) catalyzed hydrolysis of estrone conjugates (E1Cs). Androstenedione 108-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-78 27289045-7 2017 Furthermore, incubation with estradiol induced a significant decrease of NF2 expression in both HA22T and Huh7 cells (over 54% and 22%, respectively), while no change could be observed in HepG2 cells, this effect being inversely related to aromatase expression and activity in HCC cell lines. Estradiol 29-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 240-249 28970799-6 2017 We found that proteins carrying mutations A115V, T142A located close to the FMN binding site had reduced flavin content compared to WT POR and lost almost all activity to metabolize androstenedione via CYP19A1 and showed reduced CYP3A4 activity. Androstenedione 182-197 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 202-209 28977209-9 2017 Antiepileptic drugs (lamotrigine, phenobarbital, and phenytoin) also inhibit aromatase. Lamotrigine 21-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-86 28977209-9 2017 Antiepileptic drugs (lamotrigine, phenobarbital, and phenytoin) also inhibit aromatase. Phenobarbital 34-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-86 28977209-9 2017 Antiepileptic drugs (lamotrigine, phenobarbital, and phenytoin) also inhibit aromatase. Phenytoin 53-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-86 28679145-6 2017 Excessive aromatase activity in adipose tissue leads to increased conversion of androgens into estrogens, eventually results in a reduction of testosterone levels and is the underlying reason for obesity-related infertility. Testosterone 143-155 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-19 28740157-2 2017 DHA stimulates the retinoid X receptor, which reportedly regulates the expression of cytochrome P450 aromatase (P450arom). Docosahexaenoic Acids 0-3 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 112-120 28740157-4 2017 Dietary supplementation with DHA increased not only the expression of P450arom, but also 17beta-estradiol in the cerebral cortex. Docosahexaenoic Acids 29-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 70-78 28740157-6 2017 A P450arom inhibitor, letrozole, reduced 17beta-estradiol levels and completely suppressed the elongation of seizure latency elicited by DHA. Letrozole 22-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 2-10 28740157-6 2017 A P450arom inhibitor, letrozole, reduced 17beta-estradiol levels and completely suppressed the elongation of seizure latency elicited by DHA. Estradiol 41-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 2-10 28740157-6 2017 A P450arom inhibitor, letrozole, reduced 17beta-estradiol levels and completely suppressed the elongation of seizure latency elicited by DHA. Docosahexaenoic Acids 137-140 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 2-10 28977209-10 2017 Concomitant use of phenobarbital or valproate has a synergistic effect on aromatase inhibition. Phenobarbital 19-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 74-83 28977209-10 2017 Concomitant use of phenobarbital or valproate has a synergistic effect on aromatase inhibition. Valproic Acid 36-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 74-83 28429243-2 2017 Two major reactions contribute to E1 plasma concentrations, aromatase (CYP19A1) catalyzed E1 synthesis from androstenedione and steroid sulfatase (STS) catalyzed hydrolysis of estrone conjugates (E1Cs). Estrone 176-183 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-78 28512028-5 2017 These inhibitors were found to be comparable to the existing CYP19 inhibitor exemestane (42.5nM). exemestane 77-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-66 28827987-0 2017 Aromatase inhibitory activity of 1,4-naphthoquinone derivatives and QSAR study. 1,4-naphthoquinone 33-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 28611384-7 2017 Compared to the high tacrolimus group, expression levels of six cytochrome P450 enzymes, CYP1A1, CYP2B6, CYP3A5, CYP4A11, CYP19A1, and CYP17A1 were significantly higher in the low tacrolimus group. Tacrolimus 180-190 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-129 28400072-8 2017 Compared with the control group, the mRNA abundances of HES1, MYC, BAX, BCL2 and CYP19A1 in DAPT-treated groups was lower (P<0.05), respectively; whereas, the expression of CCND2, CDKN1A and TP53 mRNA showed no remarkable difference compared with control group. dapt 92-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-88 28827987-7 2017 The study suggests the 1,4-naphthoquinone derivatives as promising compounds to be further developed as a novel class of aromatase inhibitors. 1,4-naphthoquinone 23-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 121-130 28177277-0 2017 Prevalence of bisphosphonate therapy in women with breast cancer treated with aromatase inhibitors in Germany : . Diphosphonates 14-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-87 28423275-6 2017 Here, by applying free energy methods, for a cumulative simulation time of 20 mus, we provide detailed atomistic characterization and free energy profiles of the entry/exit routes preferentially followed by a substrate (androstenedione) and a last-generation inhibitor (letrozole) to/from the catalytic site of CYP19A1 (the human aromatase (HA) enzyme), a key clinical target against breast cancer, studied here as prototypical CYP450. Androstenedione 220-235 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 311-318 28423275-6 2017 Here, by applying free energy methods, for a cumulative simulation time of 20 mus, we provide detailed atomistic characterization and free energy profiles of the entry/exit routes preferentially followed by a substrate (androstenedione) and a last-generation inhibitor (letrozole) to/from the catalytic site of CYP19A1 (the human aromatase (HA) enzyme), a key clinical target against breast cancer, studied here as prototypical CYP450. Letrozole 270-279 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 311-318 28177277-1 2017 PURPOSE: The aim of this study is to analyze the prevalence of bisphosphonate therapy in women with breast cancer (BC) treated with aromatase inhibitors (AI) in Germany. Diphosphonates 63-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 132-141 28603632-0 2017 Exemestane potency is unchanged by common nonsynonymous polymorphisms in CYP19A1: results of a novel anti-aromatase activity assay examining exemestane and its derivatives. exemestane 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-80 28452938-4 2017 We found that LH induced predominant pERK1/2 and pAKT activation STARD1, CCND2 and anti-apoptotic XIAP gene expression, while hCG mediated more potent CREB phosphorylation, expression of CYP19A1 and procaspase 3 cleavage than LH. Luteinizing Hormone 14-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 187-194 28603632-1 2017 Exemestane (EXE) treats estrogen receptor positive (ER+) breast cancer in postmenopausal women by inhibiting the estrogen-synthesizing cytochrome P450 CYP19A1. exemestane 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 151-158 28487654-1 2017 After menopause, estradiol is primarily synthesized in peripheral tissues by the enzyme aromatase, encoded by CYP19A1. Estradiol 17-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 110-117 27863301-5 2017 EPR spectroscopy shows an environment of four aryloxide (ArO-) groups in the equatorial plane of the VO moiety, both in solution and in the solid complex. aryloxide 46-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-60 28178667-0 2017 Anti-GD2-ch14.18/CHO coated nanoparticles mediate glioblastoma (GBM)-specific delivery of the aromatase inhibitor, Letrozole, reducing proliferation, migration and chemoresistance in patient-derived GBM tumor cells. CAV protocol 17-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-103 28178667-0 2017 Anti-GD2-ch14.18/CHO coated nanoparticles mediate glioblastoma (GBM)-specific delivery of the aromatase inhibitor, Letrozole, reducing proliferation, migration and chemoresistance in patient-derived GBM tumor cells. Letrozole 115-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-103 28178667-3 2017 In this study, aromatase inhibition was achieved using third generation inhibitor, Letrozole, encapsulated within the core of biodegradable poly lactic-co-glycolic acid (PLGA) nanoparticles (NPs). Letrozole 83-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-24 28178667-3 2017 In this study, aromatase inhibition was achieved using third generation inhibitor, Letrozole, encapsulated within the core of biodegradable poly lactic-co-glycolic acid (PLGA) nanoparticles (NPs). Polylactic Acid-Polyglycolic Acid Copolymer 140-168 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-24 27871977-5 2017 The aim of the present study was to investigate the effects of calcitriol upon gene expression of several steroid enzymes such as cytochrome P450scc (CYP11A1), type 1 3beta-hydroxysteroid dehydrogenase(3beta-HSDI), 17beta-HSD3, 17alpha-hydroxylase/17,20 lyase (CYP17A1) and aromatase (CYP19A1) in primary cultures of human placental cells. Calcitriol 63-73 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 285-292 28126832-8 2017 We found that aromatase activity and the CYP19A1 gene transcript were both upregulated in THP-1 cells in the presence of Dex. Dexamethasone 121-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-48 30023623-2 2017 In the present study, we have designed and synthesized three C2-symmetric bisubstituted bisbenzimidazole naphthalenediimide (NDI) ligands, ALI-C3 , BBZ-ARO, and BBZ-AROCH2 , which stabilize human telomeric G-quadruplex DNA with high affinity. naphthalenediimide 125-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 152-155 30023623-5 2017 The binding equilibrium constants obtained from the microcalorimetry studies of BBZ-ARO, ALI-C3 , and BBZ-AROCH2 were 8.47, 6.35, and 3.41 muM, respectively, with h-telo 22-mer quadruplex. telo 165-169 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-87 30023623-9 2017 MD simulation results showed the highest hydrogen bond occupancy and van der Waals interactions were between the side chains of BBZ-ARO and the DNA grooves. Hydrogen 41-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 132-135 28959637-3 2017 Using this in vitro model it was shown that these depsidones inhibit the growth of T47D tumor cells most likely via inhibition of aromatase (CYP19) activity. depsidone 50-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 141-146 27747906-0 2017 Polymorphisms in ABCB1 and CYP19A1 genes affect anastrozole plasma concentrations and clinical outcomes in postmenopausal breast cancer patients. Anastrozole 48-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-34 28065637-0 2017 The steroid metabolite 16(beta)-OH-androstenedione generated by CYP21A2 serves as a substrate for CYP19A1. Steroids 4-11 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-105 28065637-0 2017 The steroid metabolite 16(beta)-OH-androstenedione generated by CYP21A2 serves as a substrate for CYP19A1. 16(beta)-oh-androstenedione 23-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-105 27890559-3 2017 Norfluoxetine decreased estrogen secretion in the feto-placental co-culture and competitively inhibited catalytic CYP19 activity in BeWo cells. norfluoxetine 0-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 114-119 27277477-7 2017 RESULTS AND LIMITATIONS: Seven SNPs located in or nearby genes implicated in steroid hormone metabolism were significantly associated with prostate volume: HSD17B2 (rs1119933), ESR2 (rs8006145), SULT2B1 (rs279451), NQO1 (rs2917670), ESR1 (rs1569788), GSTP1 (rs1138272), and CYP19A1 (rs17523880). Steroids 77-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 274-281 28144163-2 2017 OBJECTIVE: The objective of this study was to understand the relationship between CYP19 TTTA repetition polymorphism and telomere length and its effects on serum estradiol, estrone and follicle-stimulating hormone (FSH). Estradiol 162-171 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-87 28144163-8 2017 CYP19 TTTA repeat polymorphism was associated with serum levels of estradiol and estrone in both groups, being higher in those with >10 repeats. ttta 6-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-5 28144163-8 2017 CYP19 TTTA repeat polymorphism was associated with serum levels of estradiol and estrone in both groups, being higher in those with >10 repeats. Estradiol 67-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-5 28144163-8 2017 CYP19 TTTA repeat polymorphism was associated with serum levels of estradiol and estrone in both groups, being higher in those with >10 repeats. Estrone 81-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-5 28144163-12 2017 The CYP19 TTTA repeat polymorphism is associated with serum levels of estradiol and estrone in both healthy women and BC patients, being higher in those with polymorphism carriers >10 repeats. Estradiol 70-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-9 27769104-6 2017 Leydig cells were isolated by Laser Capture Microdissection (LCM) and CYP19A1 mRNA expression was quantified by SYBR Green-based qPCR. sybr green 112-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 70-77 27769104-8 2017 Relative to control group, CYP19A1 was overexpressed more than twofold in 10/19 cases (2.3-12.2-fold increase), showing a significant increment in cases with low T/LH ratio (T/LH < 2) compared to cases with T/LH >= 2 (p = 0.038, REST ). Luteinizing Hormone 164-166 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-34 27769104-8 2017 Relative to control group, CYP19A1 was overexpressed more than twofold in 10/19 cases (2.3-12.2-fold increase), showing a significant increment in cases with low T/LH ratio (T/LH < 2) compared to cases with T/LH >= 2 (p = 0.038, REST ). Luteinizing Hormone 176-178 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-34 27769104-8 2017 Relative to control group, CYP19A1 was overexpressed more than twofold in 10/19 cases (2.3-12.2-fold increase), showing a significant increment in cases with low T/LH ratio (T/LH < 2) compared to cases with T/LH >= 2 (p = 0.038, REST ). Luteinizing Hormone 176-178 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-34 29376079-0 2017 Triclocarban and Triclosan Inhibit Human Aromatase via Different Mechanisms. triclocarban 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-50 29376079-0 2017 Triclocarban and Triclosan Inhibit Human Aromatase via Different Mechanisms. Triclosan 17-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-50 29376079-6 2017 Triclocarban and triclosan inhibited human CYP19A1 with IC50 values of 15.81 and 6.26 muM, respectively. triclocarban 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-50 29376079-6 2017 Triclocarban and triclosan inhibited human CYP19A1 with IC50 values of 15.81 and 6.26 muM, respectively. Triclosan 17-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-50 29376079-7 2017 Triclosan competitively inhibited CYP19A1, while triclocarban noncompetitively inhibited this enzyme. Triclosan 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-41 29376079-8 2017 Docking study showed that triclosan bound to the steroid-binding pocket of CYP19A1, while triclocarban was off this target, suggesting a different mechanism. Triclosan 26-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-82 29376079-8 2017 Docking study showed that triclosan bound to the steroid-binding pocket of CYP19A1, while triclocarban was off this target, suggesting a different mechanism. Steroids 49-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-82 29376079-9 2017 In conclusion, triclocarban and triclosan are inhibitors of human CYP19A1. triclocarban 15-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-73 29376079-9 2017 In conclusion, triclocarban and triclosan are inhibitors of human CYP19A1. Triclosan 32-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-73 27863301-5 2017 EPR spectroscopy shows an environment of four aryloxide (ArO-) groups in the equatorial plane of the VO moiety, both in solution and in the solid complex. Vanadium(II) oxide 101-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-60 27477115-8 2016 Moreover, the expression of BCL-2, CYP17A1, CYP19A1, SOD1, and GPX4 were up-regulated by 0.01ng/mL melatonin or combined with IIK7, but decreased for the mRNA levels of BAX, P53, and CASPASE-3, as compared with control or groups treated with Luzindole or 4P-PDOT in the presence of melatonin. Melatonin 99-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-51 27616271-0 2017 Effect of sildenafil on human aromatase activity: From in vitro structural analysis to catalysis and inhibition in cells. Sildenafil Citrate 10-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-39 27616271-2 2017 As it has been suggested that its activity is affected by inhibitors of phosphodiesterase-5, this work investigates the potential interaction of sildenafil with aromatase. Sildenafil Citrate 145-155 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 161-170 27616271-4 2017 Sildenafil is found to bind to aromatase with a KD of 0.58+-0.05muM acting as a partial and mixed inhibitor with a maximal inhibition of 35+-2%. Sildenafil Citrate 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 27984130-5 2016 The combined effect of IR with 32muM Cd in granulosa cells demonstrated significant decrease in expression of StAR, CYP11A1, CYP19A1, 17beta-HSD, 3beta-HSD, FSH-R and LH-R. Cadmium 37-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-132 27825388-10 2016 CONCLUSION: The CYP19A1 rs10046 variant T/T favors lower incidence of hot flashes/sweating under exemestane + OFS treatment, suggesting endocrine-mediated effects. exemestane 97-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-23 27825388-10 2016 CONCLUSION: The CYP19A1 rs10046 variant T/T favors lower incidence of hot flashes/sweating under exemestane + OFS treatment, suggesting endocrine-mediated effects. ofs 110-113 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-23 28293075-6 2016 In this study we used in silico approach by modeling CYP19A1 protein the strcture was subjected to protein preparation wizard; to add hydrogen and optimize the protonation states of Thr310 and Ser478 and Asp309 residues. Hydrogen 134-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-60 27737328-2 2016 The POR gene encodes a flavor protein that transfers electrons from nicotinamide adenine dinucleotide phosphate (NADPH) to all microsomal cytochrome P450 type II (including 21-hydroxylase, 17alpha-hydroxylase 17,20 lyase and aromatase), which is fundamental for their enzymatic activity. NADP 68-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 189-234 27477115-8 2016 Moreover, the expression of BCL-2, CYP17A1, CYP19A1, SOD1, and GPX4 were up-regulated by 0.01ng/mL melatonin or combined with IIK7, but decreased for the mRNA levels of BAX, P53, and CASPASE-3, as compared with control or groups treated with Luzindole or 4P-PDOT in the presence of melatonin. Melatonin 282-291 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-51 27032764-1 2017 Cytochrome P450 aromatase (CYP19A1), in human placenta metabolizes androgens to estrogens and uses reduced nicotinamide adenine dinucleotide phosphate through cytochrome P450 oxidoreductase (POR) for the energy requirements of its metabolic activities. NADP 107-150 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-34 27854074-4 2017 Resveratrol at the doses of 1 and 5 micromol/L increased the level of CYP19 transcript and protein level, while 5MS reduced mRNA transcript of both CYP19 and STS genes. Resveratrol 0-11 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 70-75 28637041-2 2017 In human placenta, 3beta-hydroxysteroid dehydrogenase 1 (HSD3B1) catalyzes pregnenolone to form progesterone, and aromatase (CYP19A1) catalyzes testosterone into estradiol. Pregnenolone 75-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-132 28637041-2 2017 In human placenta, 3beta-hydroxysteroid dehydrogenase 1 (HSD3B1) catalyzes pregnenolone to form progesterone, and aromatase (CYP19A1) catalyzes testosterone into estradiol. Progesterone 96-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-132 28637041-2 2017 In human placenta, 3beta-hydroxysteroid dehydrogenase 1 (HSD3B1) catalyzes pregnenolone to form progesterone, and aromatase (CYP19A1) catalyzes testosterone into estradiol. Testosterone 144-156 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-132 28637041-2 2017 In human placenta, 3beta-hydroxysteroid dehydrogenase 1 (HSD3B1) catalyzes pregnenolone to form progesterone, and aromatase (CYP19A1) catalyzes testosterone into estradiol. Estradiol 162-171 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-132 27690428-0 2016 4-(((4-Iodophenyl)methyl)-4H-1,2,4-triazol-4-ylamino)-benzonitrile: A Potential Imaging Agent for Aromatase. 4-(((4-iodophenyl)methyl)-4H-1,2,4-triazol-4-ylamino)benzonitrile 0-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-107 27690428-4 2016 Two examples of these derivatives, 4-(((4-iodophenyl)methyl)-4H-1,2,4-triazol-4-yl-amino)-benzonitrile (5) and 4-((1H-imidazol-1-yl)(4-iodobenzyl)amino)benzonitrile (11), displayed potent binding affinities to human aromatase (IC50 = 0.17 and 0.04 nM, respectively). 4-(((4-iodophenyl)methyl)-4H-1,2,4-triazol-4-ylamino)benzonitrile 35-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 216-225 27690428-4 2016 Two examples of these derivatives, 4-(((4-iodophenyl)methyl)-4H-1,2,4-triazol-4-yl-amino)-benzonitrile (5) and 4-((1H-imidazol-1-yl)(4-iodobenzyl)amino)benzonitrile (11), displayed potent binding affinities to human aromatase (IC50 = 0.17 and 0.04 nM, respectively). 4-((1h-imidazol-1-yl)(4-iodobenzyl)amino)benzonitrile 111-164 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 216-225 27737328-2 2016 The POR gene encodes a flavor protein that transfers electrons from nicotinamide adenine dinucleotide phosphate (NADPH) to all microsomal cytochrome P450 type II (including 21-hydroxylase, 17alpha-hydroxylase 17,20 lyase and aromatase), which is fundamental for their enzymatic activity. NADP 113-118 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 189-234 27477115-8 2016 Moreover, the expression of BCL-2, CYP17A1, CYP19A1, SOD1, and GPX4 were up-regulated by 0.01ng/mL melatonin or combined with IIK7, but decreased for the mRNA levels of BAX, P53, and CASPASE-3, as compared with control or groups treated with Luzindole or 4P-PDOT in the presence of melatonin. luzindole 242-251 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-51 27580802-7 2016 In a human granulosa cell line, COV434, a NKB agonist, senktide, also increased CYP11A1 and CYP19A1 mRNA levels and enhanced aromatase protein levels and activities. cov434 32-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-99 27582109-13 2016 DCI is an insulin-sensitizer that counteracts this action by reducing the expression of the genes CYP19A1, P450scc and IGF-1R. dci 0-3 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-105 27071740-5 2016 On the other hand, the 6-chloro derivative 1-(1H-imidazol-1-yl)methyl-6-chloro-9H-xanthen-9-one (1 b) was found to be a fairly potent and somewhat selective CYP19 inhibitor, confirming the versatility of the scaffold. 6-chloro 23-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 157-162 27071740-5 2016 On the other hand, the 6-chloro derivative 1-(1H-imidazol-1-yl)methyl-6-chloro-9H-xanthen-9-one (1 b) was found to be a fairly potent and somewhat selective CYP19 inhibitor, confirming the versatility of the scaffold. 1-(1h-imidazol-1-yl)methyl-6-chloro-9h-xanthen-9-one 43-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 157-162 27002602-4 2016 Additionally, to evaluate the differences in binding modes of these enantiomers, docking and molecular dynamics (MD) simulations of S-omeprazole (S-OME) and R-omeprazole (R-OME) in CYP17A1, CYP19A1 and CYP21A2 were carried out. Omeprazole 132-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 190-197 27296990-8 2016 In human pulmonary arterial smooth muscle cells, metformin decreased proliferation and decreased estrogen synthesis by decreasing aromatase activity through the PII promoter site of Cyp19a1 Thus, we report for the first time that metformin can reverse pulmonary hypertension through inhibition of aromatase and estrogen synthesis in a manner likely to be mediated by AMP-activated protein kinase. Metformin 49-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 182-189 27296990-8 2016 In human pulmonary arterial smooth muscle cells, metformin decreased proliferation and decreased estrogen synthesis by decreasing aromatase activity through the PII promoter site of Cyp19a1 Thus, we report for the first time that metformin can reverse pulmonary hypertension through inhibition of aromatase and estrogen synthesis in a manner likely to be mediated by AMP-activated protein kinase. Metformin 230-239 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 182-189 27002602-4 2016 Additionally, to evaluate the differences in binding modes of these enantiomers, docking and molecular dynamics (MD) simulations of S-omeprazole (S-OME) and R-omeprazole (R-OME) in CYP17A1, CYP19A1 and CYP21A2 were carried out. r-omeprazole 157-169 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 190-197 27346174-2 2016 Second-order rate constants (ks and kr) for reactions of ArO( ) and alpha-Toc( ) radicals with the above antioxidants were measured in 2-propanol/water (5:1, v/v) solution at 25.0 C, using single- and double-mixing stopped-flow spectrophotometries, respectively. 2-Propanol 135-145 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-60 27467582-10 2016 CP-31398, a p53 rescue compound, suppressed levels of Aha1, Hsp90 ATPase activity, levels of PKM2 and HIF-1alpha, and aromatase expression in LFS stromal cells. CP 31398 0-8 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 118-127 27346174-5 2016 The ultraviolet-visible absorption of alpha-Toc( ) (lambdamax = 428 nm), which was produced by the reaction of alpha-tocopherol (alpha-TocH) with ArO( ), disappeared under the coexistence of CAs due to the alpha-TocH-regeneration reaction. alpha-Tocopherol 111-127 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 146-149 27346174-5 2016 The ultraviolet-visible absorption of alpha-Toc( ) (lambdamax = 428 nm), which was produced by the reaction of alpha-tocopherol (alpha-TocH) with ArO( ), disappeared under the coexistence of CAs due to the alpha-TocH-regeneration reaction. Catecholamines 191-194 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 146-149 27112698-10 2016 Similarly, 3beta-HSD, CYP11A1 and CYP19A1 protein levels were reduced after administration of 20 microg/ml BPA. bisphenol A 107-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-41 27220457-0 2016 Zeranol stimulates proliferation and aromatase activation in human breast preadipocytes. Zeranol 0-7 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-46 27220457-4 2016 The present study hypothesized that aromatase expression and activity may be elevated by low dose zeranol exposure, providing a source of estrogens that may stimulate cell proliferation. Zeranol 98-105 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-45 27220457-7 2016 The results demonstrated that low dose zeranol (2-50 nM) was able to significantly promote cell proliferation, aromatase mRNA expression, aromatase activity and estrogen production in primary cultured human breast preadipocytes, thus suggesting that zeranol may act as an aromatase activator. Zeranol 39-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-120 27290646-7 2016 In addition, PGE2, NF-kappaB and leptin influenced the expression of aromatase, as observed in women. Dinoprostone 13-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-78 27220457-7 2016 The results demonstrated that low dose zeranol (2-50 nM) was able to significantly promote cell proliferation, aromatase mRNA expression, aromatase activity and estrogen production in primary cultured human breast preadipocytes, thus suggesting that zeranol may act as an aromatase activator. Zeranol 39-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-147 27220457-7 2016 The results demonstrated that low dose zeranol (2-50 nM) was able to significantly promote cell proliferation, aromatase mRNA expression, aromatase activity and estrogen production in primary cultured human breast preadipocytes, thus suggesting that zeranol may act as an aromatase activator. Zeranol 39-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-147 27220457-8 2016 The findings of the present study suggest that zeranol promotes breast cancer cell growth by stimulating aromatase activation and increasing estrogen biosynthesis in adipose tissue. Zeranol 47-54 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 105-114 27252161-1 2016 Aromatase (estrogen synthetase; EC 1.14.14.1) catalyzes the demethylation of androgens" carbon 19, producing phenolic 18-carbon estrogens. Carbon 88-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 27252161-1 2016 Aromatase (estrogen synthetase; EC 1.14.14.1) catalyzes the demethylation of androgens" carbon 19, producing phenolic 18-carbon estrogens. Carbon 88-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-30 27252161-1 2016 Aromatase (estrogen synthetase; EC 1.14.14.1) catalyzes the demethylation of androgens" carbon 19, producing phenolic 18-carbon estrogens. Carbon 121-127 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 27252161-1 2016 Aromatase (estrogen synthetase; EC 1.14.14.1) catalyzes the demethylation of androgens" carbon 19, producing phenolic 18-carbon estrogens. Carbon 121-127 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-30 27257473-7 2016 We documented multiple nonsynonymous substitutions in genes associated with steroid synthesis, transport, and cellular action (SRD5A2, CYP19A1, SHBG, and AR) and with anti-Mullerian hormone (AMH and AMHR2) in callitrichines. Steroids 76-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-142 27257473-8 2016 In the only callitrichine to produce single infants (Callimico), two genes contained nonsynonymous substitutions relative to twinning callitrichines (CYP19A1 and AMRHR2); these substitutions were identical with nontwinning Saimiri and humans, suggesting a reversion to an ancestral sequence. callitrichine 12-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 150-157 26472556-8 2016 Treatment of the cultures with 17beta-estradiol results in phosphorylation of the enzyme and increased aromatase protein expression, which suggests that estradiol synthesis in hippocampal neurons is regulated in an autocrine manner. Estradiol 31-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 103-112 26689671-3 2016 The so-called third generation aromatase inhibitors (AIs) letrozole, anastrozole, and the steroidal exemestane were approved in the U.S. in the late 1990s for estrogen-dependent postmenopausal breast cancer. Letrozole 58-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 26689671-3 2016 The so-called third generation aromatase inhibitors (AIs) letrozole, anastrozole, and the steroidal exemestane were approved in the U.S. in the late 1990s for estrogen-dependent postmenopausal breast cancer. Anastrozole 69-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 26472556-5 2016 Aromatase becomes inactivated as soon as it is phosphorylated by Ca(2+)-dependent kinases upon calcium release from internal stores. Calcium 95-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 26472556-8 2016 Treatment of the cultures with 17beta-estradiol results in phosphorylation of the enzyme and increased aromatase protein expression, which suggests that estradiol synthesis in hippocampal neurons is regulated in an autocrine manner. Estradiol 38-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 103-112 26472556-6 2016 Accordingly, thapsigargin dephosphorylates aromatase and stimulates estradiol synthesis by depletion of internal Ca(2+) stores. Thapsigargin 13-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-52 27262800-0 2016 The individual or combinational effects of Hesperetin and Letrozole on the activity and expression of aromatase in MCF-7 cells. hesperetin 43-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 102-111 26472556-7 2016 Vice versa, letrozole, an aromatase inhibitor, phosphorylates aromatase and reduces estradiol synthesis. Letrozole 12-21 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-35 26472556-7 2016 Vice versa, letrozole, an aromatase inhibitor, phosphorylates aromatase and reduces estradiol synthesis. Letrozole 12-21 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-71 26472556-7 2016 Vice versa, letrozole, an aromatase inhibitor, phosphorylates aromatase and reduces estradiol synthesis. Estradiol 84-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-35 27067917-5 2016 There were responses in that only diphthalates with 4-7 carbon atoms were competitive HSD3B1 inhibitors and diphthalates with 6 carbon atoms were CYP19A1 inhibitors. diphthalates 108-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 146-153 27067917-7 2016 DCHP and BBOP inhibited CYP19A1, with IC50s of 64.70 and 56.47muM. O,O'-dimethyl-O-(6-chlorobicyclo(3.2.0)heptadiene-1,5-yl)phosphate 0-4 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-31 27067917-7 2016 DCHP and BBOP inhibited CYP19A1, with IC50s of 64.70 and 56.47muM. bbop 9-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-31 27155741-0 2016 Pictilisib for oestrogen receptor-positive, aromatase inhibitor-resistant, advanced or metastatic breast cancer (FERGI): a randomised, double-blind, placebo-controlled, phase 2 trial. 2-(1H-indazol-4-yl)-6-(4-methanesulfonylpiperazin-1-ylmethyl)-4-morpholin-4-ylthieno(3,2-d)pyrimidine 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-53 27262800-0 2016 The individual or combinational effects of Hesperetin and Letrozole on the activity and expression of aromatase in MCF-7 cells. Letrozole 58-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 102-111 27262800-4 2016 This study was carried out to investigate the effect of hesperetin on the activity and expression of aromatase and compare this property with letrozole as an aromatase inhibitor in MCF-7 breast cancer cell line. hesperetin 56-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-110 27262800-4 2016 This study was carried out to investigate the effect of hesperetin on the activity and expression of aromatase and compare this property with letrozole as an aromatase inhibitor in MCF-7 breast cancer cell line. Letrozole 142-151 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 158-167 27262800-6 2016 Aromatase activity assay, based on 17beta-Estradiol (E2) production from testosterone, revealed that hesperetin had no effect. Estradiol 35-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 27262800-6 2016 Aromatase activity assay, based on 17beta-Estradiol (E2) production from testosterone, revealed that hesperetin had no effect. Estradiol 53-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 27262800-6 2016 Aromatase activity assay, based on 17beta-Estradiol (E2) production from testosterone, revealed that hesperetin had no effect. Testosterone 73-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 27262800-7 2016 Real-time PCR results indicated that treatment with 1muM concentration of hesperetin for 48 h significantly decreased relative aromatase expression (P =0.004). hesperetin 74-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-136 27262800-11 2016 In conclusion, the present study showed that hesperetin could decrease expression of aromatase at low concentrations in MCF-7 breast cancer cells. hesperetin 45-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 85-94 27102200-0 2016 Alcohol-related breast cancer in postmenopausal women - effect of CYP19A1, PPARG and PPARGC1A polymorphisms on female sex-hormone levels and interaction with alcohol consumption and NSAID usage in a nested case-control study and a randomised controlled trial. Alcohols 0-7 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-73 27050599-12 2016 Within the CAO/ARO/AIO-94 cohort (n = 398), 19% of patients with ypT1/2 (ypT1 = 22%; ypT2 = 18%) had ypN+ status compared with 43% with ypT3/4 cancers (ypT3 = 40%; ypT4 = 73%). ypn+ 101-105 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-18 26897397-4 2016 Polymorphisms in the CYP19 gene are associated with circulating estrogen levels and estrogen/testosterone ratio. Testosterone 93-105 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-26 26463708-7 2016 CONCLUSIONS: Variants in CYP19A1 are associated with decreases in triglycerides and variable changes in HDL-C in postmenopausal women on adjuvant aromatase inhibitors. Triglycerides 66-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-32 26593510-2 2016 Influence of various substituents at the ortho, meta and para positions of the aromatic core of phenols on the bond dissociation enthalpy of the ArO-H bond was evaluated using a DFT method B3LYP/6-311++G(2d,2p)//B3LYP/6-311G(d,p). Phenols 96-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 145-148 26593510-6 2016 Reaction mechanisms involving the evolution of the primary phenoxyl radical ArO are proposed to rationalise these effects. phenoxyl 59-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-79 26574572-1 2016 Candidate gene studies have reported CYP19A1 variants to be associated with endometrial cancer and with estradiol (E2) concentrations. Estradiol 104-113 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-44 26709042-6 2016 In contrast, (-) gossypol moderately inhibited CYP19A1 activity with IC50 of 23 muM, while (+) gossypol had no inhibition when the highest concentration (100 muM) was tested. Gossypol 17-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-54 26709042-8 2016 (-) Gossypol competitively inhibited CYP19A1 against substrate testosterone. Gossypol 4-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-44 26709042-8 2016 (-) Gossypol competitively inhibited CYP19A1 against substrate testosterone. Testosterone 63-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-44 26709042-9 2016 Gossypol enantiomers showed different potency related to their inhibition on human HSD3B1 and CYP19A1. Gossypol 0-8 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-101 26857876-8 2016 Human cartilage, freshly isolated chondrocytes, and chondrocytes cultured for 2h expressed an insignificant amount of aromatase; however, expression arose after 48 h of culture and remained increased thereafter. Deuterium 78-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 118-127 26134065-0 2016 Tadalafil modulates aromatase activity and androgen receptor expression in a human osteoblastic cell in vitro model. Tadalafil 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-29 26134065-2 2016 We aimed to characterize, in a cellular model system in vitro, the potential modulation of aromatase and sex steroid hormone receptors upon exposure to tadalafil (TAD). Tadalafil 152-161 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-100 26134065-2 2016 We aimed to characterize, in a cellular model system in vitro, the potential modulation of aromatase and sex steroid hormone receptors upon exposure to tadalafil (TAD). Tadalafil 163-166 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-100 26134065-8 2016 Also, TAD inhibited ARO mRNA and protein expression leading to an increase in testosterone levels in the supernatants. Testosterone 78-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-23 26601608-11 2016 An RCT of copper surfaces in an ICU demonstrated 58% reduction in HCAI (P = 0.013) and 64% reduction in ARO transmission (P = 0.063) but was considered low-quality evidence due to improper randomization and incomplete blinding. Copper 10-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 104-107 26526982-3 2016 In this study, we investigated the effect of c-fos on the steady-state levels of CYP17 and CYP19 in human ovarian granulosa-like tumor cell line (KGN) by inhibiting MEK/ERK pathway with PD98059. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 186-193 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-96 26526982-6 2016 In conclusion, factors such as c-fos may play a crucial role in the down-regulation of CYP17 and up-regulation of CYP19 in granulosa cells, thereby suppressing androstenedione synthesis. Androstenedione 160-175 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 114-119 26432395-1 2016 Biosynthesis of steroid hormones in vertebrates involves three cytochrome P450 hydroxylases, CYP11A1, CYP17A1 and CYP19A1, which catalyze sequential steps in steroidogenesis. Steroids 16-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 114-121 29629743-0 2016 [Effect of free fatty acids on CYP19A1 (aromatase) gene expression in human adipose tissue stromal vascular fraction cells]. Fatty Acids, Nonesterified 11-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-38 26735933-0 2016 Effects of Methoxychlor and Its Metabolite Hydroxychlor on Human Placental 3beta-Hydroxysteroid Dehydrogenase 1 and Aromatase in JEG-3 Cells. Methoxychlor 11-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-125 26735933-2 2016 In the human placenta, 3beta-hydroxysteroid dehydrogenase 1 (HSD3B1) is responsible for the formation of progesterone from pregnenolone and aromatase (CYP19A1) for the production of estradiol from androgen. Progesterone 105-117 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 151-158 26735933-2 2016 In the human placenta, 3beta-hydroxysteroid dehydrogenase 1 (HSD3B1) is responsible for the formation of progesterone from pregnenolone and aromatase (CYP19A1) for the production of estradiol from androgen. Pregnenolone 123-135 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 151-158 26735933-2 2016 In the human placenta, 3beta-hydroxysteroid dehydrogenase 1 (HSD3B1) is responsible for the formation of progesterone from pregnenolone and aromatase (CYP19A1) for the production of estradiol from androgen. Estradiol 182-191 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 151-158 26735933-10 2016 When testosterone was used, HPTE was a mixed inhibitor of CYP19A1. Testosterone 5-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-65 26735933-10 2016 When testosterone was used, HPTE was a mixed inhibitor of CYP19A1. 2,2-bis(4-hydroxyphenyl)-1,1,1-trichloroethane 28-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-65 26735933-11 2016 In conclusion, MXC and HPTE are potent inhibitors of human HSD3B1, and HPTE is a weak CYP19A1 inhibitor. 2,2-bis(4-hydroxyphenyl)-1,1,1-trichloroethane 71-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-93 29629743-3 2016 Estrogens are foundto be protective for metabolism homeostasis,and cardiovascular system.Disturbed dietary and endogenousfatty acids (FAs) turnover is responsiblefor development of metabolicsyndrome and it complications.Aim of the work was to investigatethe effect of physiological concentrationsof acids: arachidonic (AA), oleic(OA), palmitynoic (PA) and eikozapentaenoic(EPA) on CYP19A1 expressionin differentiating human SVF, able toform adipocytes as well as endothelialcells. ammonium ferrous sulfate 134-137 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 381-388 29629743-6 2016 Changes in the CYP19A1expression induced by 24hs incubationin the presence of FAs (10 - 30muM )were monitored by the Real timePCR (qRT -PCR). ammonium ferrous sulfate 78-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-22 26536870-10 2015 In the exemestane-treated patients, SNPs in ESR1 (Rs2813543) and CYP19A1 (Rs6493497) were associated with decreased bone density. exemestane 7-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-72 26464060-0 2016 Effects of Neonicotinoids on Promoter-Specific Expression and Activity of Aromatase (CYP19) in Human Adrenocortical Carcinoma (H295R) and Primary Umbilical Vein Endothelial (HUVEC) Cells. Neonicotinoids 11-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 74-83 26464060-0 2016 Effects of Neonicotinoids on Promoter-Specific Expression and Activity of Aromatase (CYP19) in Human Adrenocortical Carcinoma (H295R) and Primary Umbilical Vein Endothelial (HUVEC) Cells. Neonicotinoids 11-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 85-90 26464060-5 2016 Exposure to certain pesticides, such as atrazine, is associated with increased CYP19 expression, but little is known about the effects of neonicotinoid insecticides on CYP19. Atrazine 40-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-84 26464060-6 2016 We developed sensitive and robust RT-qPCR methods to detect the promoter-specific expression of CYP19 in human adrenocortical carcinoma (H295R) and primary umbilical vein endothelial (HUVEC) cells, and determined the potential promoter-specific disruption of CYP19 expression by atrazine and the commonly used neonicotinoids imidacloprid, thiacloprid, and thiamethoxam. Atrazine 279-287 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-101 26464060-6 2016 We developed sensitive and robust RT-qPCR methods to detect the promoter-specific expression of CYP19 in human adrenocortical carcinoma (H295R) and primary umbilical vein endothelial (HUVEC) cells, and determined the potential promoter-specific disruption of CYP19 expression by atrazine and the commonly used neonicotinoids imidacloprid, thiacloprid, and thiamethoxam. Neonicotinoids 310-324 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-101 26464060-6 2016 We developed sensitive and robust RT-qPCR methods to detect the promoter-specific expression of CYP19 in human adrenocortical carcinoma (H295R) and primary umbilical vein endothelial (HUVEC) cells, and determined the potential promoter-specific disruption of CYP19 expression by atrazine and the commonly used neonicotinoids imidacloprid, thiacloprid, and thiamethoxam. imidacloprid 325-337 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-101 26631750-10 2015 This work completes the structural and functional characterization of mono- and di-domain ARO/CYCs in bacterial type II polyketide synthases and lays the groundwork for engineered biosynthesis of new bioactive polyketides. Polyketides 210-221 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-93 26464060-6 2016 We developed sensitive and robust RT-qPCR methods to detect the promoter-specific expression of CYP19 in human adrenocortical carcinoma (H295R) and primary umbilical vein endothelial (HUVEC) cells, and determined the potential promoter-specific disruption of CYP19 expression by atrazine and the commonly used neonicotinoids imidacloprid, thiacloprid, and thiamethoxam. thiacloprid 339-350 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-101 26464060-6 2016 We developed sensitive and robust RT-qPCR methods to detect the promoter-specific expression of CYP19 in human adrenocortical carcinoma (H295R) and primary umbilical vein endothelial (HUVEC) cells, and determined the potential promoter-specific disruption of CYP19 expression by atrazine and the commonly used neonicotinoids imidacloprid, thiacloprid, and thiamethoxam. Thiamethoxam 356-368 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-101 26464060-7 2016 In H295R cells, atrazine concentration-dependently increased PII- and I.3-mediated CYP19 expression and aromatase catalytic activity. Atrazine 16-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-88 26464060-7 2016 In H295R cells, atrazine concentration-dependently increased PII- and I.3-mediated CYP19 expression and aromatase catalytic activity. Atrazine 16-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 104-113 26464060-8 2016 Thiacloprid and thiamethoxam induced PII- and I.3-mediated CYP19 expression and aromatase activity at relatively low concentrations (0.1-1.0 microM), exhibiting non-monotonic concentration-response curves with a decline in gene induction and catalytic activity at higher concentrations. thiacloprid 0-11 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-64 26464060-8 2016 Thiacloprid and thiamethoxam induced PII- and I.3-mediated CYP19 expression and aromatase activity at relatively low concentrations (0.1-1.0 microM), exhibiting non-monotonic concentration-response curves with a decline in gene induction and catalytic activity at higher concentrations. thiacloprid 0-11 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 80-89 26464060-8 2016 Thiacloprid and thiamethoxam induced PII- and I.3-mediated CYP19 expression and aromatase activity at relatively low concentrations (0.1-1.0 microM), exhibiting non-monotonic concentration-response curves with a decline in gene induction and catalytic activity at higher concentrations. Thiamethoxam 16-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-64 26464060-8 2016 Thiacloprid and thiamethoxam induced PII- and I.3-mediated CYP19 expression and aromatase activity at relatively low concentrations (0.1-1.0 microM), exhibiting non-monotonic concentration-response curves with a decline in gene induction and catalytic activity at higher concentrations. Thiamethoxam 16-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 80-89 26464060-9 2016 In HUVEC cells, atrazine slightly induced overall (promoter-indistinct) CYP19 expression (30 microM) and aromatase activity (>= 3 microM), without increasing I.1 promoter activity. Atrazine 16-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 72-77 26464060-9 2016 In HUVEC cells, atrazine slightly induced overall (promoter-indistinct) CYP19 expression (30 microM) and aromatase activity (>= 3 microM), without increasing I.1 promoter activity. Atrazine 16-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 105-114 26631750-3 2015 The aromatase/cyclases (ARO/CYCs) are responsible for regiospecific cyclization of bacterial polyketides. Polyketides 93-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-27 26631750-7 2015 Di-domain ARO/CYCs can further be separated into two subclasses: "nonreducing" ARO/CYCs, which act on nonreduced poly-beta-ketones, and "reducing" ARO/CYCs, which act on cyclized C9 reduced poly-beta-ketones. poly-beta-ketones 113-130 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-13 26631750-7 2015 Di-domain ARO/CYCs can further be separated into two subclasses: "nonreducing" ARO/CYCs, which act on nonreduced poly-beta-ketones, and "reducing" ARO/CYCs, which act on cyclized C9 reduced poly-beta-ketones. poly-beta-ketones 113-130 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-82 26631750-7 2015 Di-domain ARO/CYCs can further be separated into two subclasses: "nonreducing" ARO/CYCs, which act on nonreduced poly-beta-ketones, and "reducing" ARO/CYCs, which act on cyclized C9 reduced poly-beta-ketones. poly-beta-ketones 113-130 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-82 26631750-7 2015 Di-domain ARO/CYCs can further be separated into two subclasses: "nonreducing" ARO/CYCs, which act on nonreduced poly-beta-ketones, and "reducing" ARO/CYCs, which act on cyclized C9 reduced poly-beta-ketones. poly-beta-ketones 190-207 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-13 26631750-7 2015 Di-domain ARO/CYCs can further be separated into two subclasses: "nonreducing" ARO/CYCs, which act on nonreduced poly-beta-ketones, and "reducing" ARO/CYCs, which act on cyclized C9 reduced poly-beta-ketones. poly-beta-ketones 190-207 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-82 26631750-7 2015 Di-domain ARO/CYCs can further be separated into two subclasses: "nonreducing" ARO/CYCs, which act on nonreduced poly-beta-ketones, and "reducing" ARO/CYCs, which act on cyclized C9 reduced poly-beta-ketones. poly-beta-ketones 190-207 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-82 26323233-10 2015 RESULTS: After adjustment for multiple comparisons, we found that rs4441215 and rs936306 in CYP19A1 and rs4888202 and rs2955160 in HSD17B2 were associated with differences in serum estrone level. Estrone 181-188 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-99 26170243-7 2015 Protein expression in CLs was investigated for CYP11A1, CYP17A1, CYP19A1 and HSD3B. Chlorine 22-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-72 26196865-0 2015 Testosterone and Adult Male Bone: Actions Independent of 5alpha-Reductase and Aromatase. Testosterone 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-87 26361012-3 2015 Here we directly compared the human (h-) and porcine gonadal (p(g)-) P450arom, as p(g)-P450arom has very low catalytic efficiency, with a ten-fold higher affinity (Km) for a substrate (androstenedione) and ten-fold reduction in turnover (Vmax). Androstenedione 185-200 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-77 26361012-3 2015 Here we directly compared the human (h-) and porcine gonadal (p(g)-) P450arom, as p(g)-P450arom has very low catalytic efficiency, with a ten-fold higher affinity (Km) for a substrate (androstenedione) and ten-fold reduction in turnover (Vmax). Androstenedione 185-200 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-95 26512648-6 2015 All three EDCs-PCB 153, phthalates, and BPA influenced five common genes-CYP19A1, EGFR, ESR2, FOS, and IGF1-in breast cancer as well as in endometriosis. 2,2',4,4',5,5'-Hexachlorobiphenyl 15-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-80 26213324-6 2015 Therefore, we tested the hypothesis if Trichostatin A (TSA), a HDAC inhibitor, can attenuate LPS induced pro-inflammatory cytokine gene expression and can prevent LPS mediated down-regulation of CYP19A1 expression and E2 in GCs. trichostatin A 39-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 195-202 26213324-6 2015 Therefore, we tested the hypothesis if Trichostatin A (TSA), a HDAC inhibitor, can attenuate LPS induced pro-inflammatory cytokine gene expression and can prevent LPS mediated down-regulation of CYP19A1 expression and E2 in GCs. trichostatin A 55-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 195-202 26213324-8 2015 Additionally, TSA pre-treatment reversed the inhibitory effect of LPS on CYP19A1 expression and E2 production. trichostatin A 14-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-80 26213324-9 2015 CHIP analyses of H3 (Lys 9/14) acetylation of ovary specific CYP19A1 proximal promoter (PII) showed that TSA pre-treatment prevented the LPS mediated H3 deacetylation, thereby increased the acetylation of PII and restored CYP19A1 expression and E2 production. Lysine 21-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-68 26213324-9 2015 CHIP analyses of H3 (Lys 9/14) acetylation of ovary specific CYP19A1 proximal promoter (PII) showed that TSA pre-treatment prevented the LPS mediated H3 deacetylation, thereby increased the acetylation of PII and restored CYP19A1 expression and E2 production. trichostatin A 105-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-68 26213324-9 2015 CHIP analyses of H3 (Lys 9/14) acetylation of ovary specific CYP19A1 proximal promoter (PII) showed that TSA pre-treatment prevented the LPS mediated H3 deacetylation, thereby increased the acetylation of PII and restored CYP19A1 expression and E2 production. trichostatin A 105-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 222-229 26213324-10 2015 The present study demonstrated that TSA pre-treatment attenuated- LPS induced immune response involving NF-kappaB and HDACs, and thus prevented inhibition of CYP19A1 expression and E2 production through chromatin remodeling. trichostatin A 36-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 158-165 26135543-0 2015 Valproic acid-induced histone acetylation suppresses CYP19 gene expression and inhibits the growth and survival of endometrial stromal cells. Valproic Acid 0-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-58 26096372-0 2015 QM/MM modeling of the hydroxylation of the androstenedione substrate catalyzed by cytochrome P450 aromatase (CYP19A1). Androstenedione 43-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 109-116 26135543-7 2015 The results revealed that valproic acid (VPA) significantly promoted histone acetylation in the ESCs, which inhibited histone acetylation in the promoter region of the CYP19 gene, thus suppressing its expression. Valproic Acid 26-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 168-173 26135543-7 2015 The results revealed that valproic acid (VPA) significantly promoted histone acetylation in the ESCs, which inhibited histone acetylation in the promoter region of the CYP19 gene, thus suppressing its expression. Valproic Acid 41-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 168-173 25628423-7 2015 Aromatase (CYP19A1) expression in cumulus cells was also inhibited by miR-378, leading to a significant decrease in estradiol production. Estradiol 116-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-18 26058395-2 2015 Metformin is known to suppress prostaglandin E2 (PGE2)-induced CYP19A1 messenger RNA (mRNA) expression in human endometriotic stromal cells (ESCs). Metformin 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-70 26058395-2 2015 Metformin is known to suppress prostaglandin E2 (PGE2)-induced CYP19A1 messenger RNA (mRNA) expression in human endometriotic stromal cells (ESCs). Dinoprostone 31-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-70 26058395-2 2015 Metformin is known to suppress prostaglandin E2 (PGE2)-induced CYP19A1 messenger RNA (mRNA) expression in human endometriotic stromal cells (ESCs). Dinoprostone 49-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-70 26058395-6 2015 The binding of the cyclic AMP response element-binding (CREB) protein to CYP19A1 promoter II (PII) was assessed by chromatin immunoprecipitation assay. Cyclic AMP 19-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-80 26058395-9 2015 CONCLUSION: Metformin could inhibit PGE2-induced CYP19A1 mRNA expression and aromatase activity via AMPK activation and inhibition of CREB to CYP19A1 PII in human ESCs. Metformin 12-21 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-56 26058395-9 2015 CONCLUSION: Metformin could inhibit PGE2-induced CYP19A1 mRNA expression and aromatase activity via AMPK activation and inhibition of CREB to CYP19A1 PII in human ESCs. Metformin 12-21 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 142-149 26058395-9 2015 CONCLUSION: Metformin could inhibit PGE2-induced CYP19A1 mRNA expression and aromatase activity via AMPK activation and inhibition of CREB to CYP19A1 PII in human ESCs. Dinoprostone 36-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-56 26058395-9 2015 CONCLUSION: Metformin could inhibit PGE2-induced CYP19A1 mRNA expression and aromatase activity via AMPK activation and inhibition of CREB to CYP19A1 PII in human ESCs. Dinoprostone 36-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 142-149 25701839-9 2015 alpha-MEM cells were estrogenic and expressed the CYP19 gene. alpha-mem 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-55 26246878-2 2015 The present study was designed to investigate the ef- fect of EPA on the expression levels of peroxisome proliferator-activated receptor gamma (PPARgamma) and cytochrome P450 aromatase (encoded by the CYP-19) in primary cultured granulosa cells (GC) from patients undergoing in vitro fertilization (IVF), and also to compare these effects with those in GC of PCOS patients. Eicosapentaenoic Acid 62-65 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 201-207 26246878-6 2015 High doses of EPA in the presence of rFSH produced a stimulatory effect on expres- sion level of PPARgamma (2.15-fold, P=0.001) and a suppressive effect (0.56-fold, P=0.01) on the expression level of CYP-19, only in the PCOS GC. Eicosapentaenoic Acid 14-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 200-206 26246878-8 2015 Altered FSH-induced PPARgamma activity in PCOS GC may modulate the CYP-19 gene expression in response to EPA, and possibly modulates the subsequent steroidogenesis of these cells. Eicosapentaenoic Acid 105-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-73 25935582-11 2015 CYP19A1 rs700518 variants showed associations with BCFI, DRFI, in tamoxifen treated patients and musculoskeletal AEs regardless of treatment. Tamoxifen 66-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-7 25678418-8 2015 CYP17A1, CYP19A1 and CYP27A1 catalyzed steroid synthesis, including hydroxylation at 17alpha, 19 and 27 positions, respectively. Steroids 39-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-16 25773531-2 2015 The expression of the CYP19A1 gene (that encodes aromatase) was compared in cumulus cells and oestradiol concentrations in the follicular fluid of infertile women with and without endometriosis submitted to ovarian stimulation for intracytoplasmic sperm injection. Estradiol 94-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 22-29 25773531-2 2015 The expression of the CYP19A1 gene (that encodes aromatase) was compared in cumulus cells and oestradiol concentrations in the follicular fluid of infertile women with and without endometriosis submitted to ovarian stimulation for intracytoplasmic sperm injection. Estradiol 94-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-58 25773531-5 2015 A lower expression of the CYP19A1 in the cumulus cells of infertile women with endometriosis was observed compared with controls (0.17 +- 0.13 and 0.56 +- 0.12, respectively), and no significant difference in the follicular fluid oestradiol concentrations was observed between groups. Estradiol 230-240 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-33 25676362-3 2015 These results suggested that Cu(II)-catalyzed phenol autoxidation by activating O2 and phenol in terms of a phenoxy radical (ArO)-Cu(II)-superoxide ternary complex, whereas selectivity between oxygenation and coupling depends mainly on the electronic structure of ArO. cu(ii) 29-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-128 25676362-3 2015 These results suggested that Cu(II)-catalyzed phenol autoxidation by activating O2 and phenol in terms of a phenoxy radical (ArO)-Cu(II)-superoxide ternary complex, whereas selectivity between oxygenation and coupling depends mainly on the electronic structure of ArO. cu(ii) 29-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 264-267 25676362-3 2015 These results suggested that Cu(II)-catalyzed phenol autoxidation by activating O2 and phenol in terms of a phenoxy radical (ArO)-Cu(II)-superoxide ternary complex, whereas selectivity between oxygenation and coupling depends mainly on the electronic structure of ArO. Phenol 46-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-128 25676362-3 2015 These results suggested that Cu(II)-catalyzed phenol autoxidation by activating O2 and phenol in terms of a phenoxy radical (ArO)-Cu(II)-superoxide ternary complex, whereas selectivity between oxygenation and coupling depends mainly on the electronic structure of ArO. Phenol 46-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 264-267 25676362-3 2015 These results suggested that Cu(II)-catalyzed phenol autoxidation by activating O2 and phenol in terms of a phenoxy radical (ArO)-Cu(II)-superoxide ternary complex, whereas selectivity between oxygenation and coupling depends mainly on the electronic structure of ArO. Oxygen 80-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-128 25676362-3 2015 These results suggested that Cu(II)-catalyzed phenol autoxidation by activating O2 and phenol in terms of a phenoxy radical (ArO)-Cu(II)-superoxide ternary complex, whereas selectivity between oxygenation and coupling depends mainly on the electronic structure of ArO. Oxygen 80-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 264-267 25676362-3 2015 These results suggested that Cu(II)-catalyzed phenol autoxidation by activating O2 and phenol in terms of a phenoxy radical (ArO)-Cu(II)-superoxide ternary complex, whereas selectivity between oxygenation and coupling depends mainly on the electronic structure of ArO. Phenol 87-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-128 25676362-3 2015 These results suggested that Cu(II)-catalyzed phenol autoxidation by activating O2 and phenol in terms of a phenoxy radical (ArO)-Cu(II)-superoxide ternary complex, whereas selectivity between oxygenation and coupling depends mainly on the electronic structure of ArO. Phenol 87-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 264-267 25676362-3 2015 These results suggested that Cu(II)-catalyzed phenol autoxidation by activating O2 and phenol in terms of a phenoxy radical (ArO)-Cu(II)-superoxide ternary complex, whereas selectivity between oxygenation and coupling depends mainly on the electronic structure of ArO. phenoxy radical 108-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-128 25676362-3 2015 These results suggested that Cu(II)-catalyzed phenol autoxidation by activating O2 and phenol in terms of a phenoxy radical (ArO)-Cu(II)-superoxide ternary complex, whereas selectivity between oxygenation and coupling depends mainly on the electronic structure of ArO. cu(ii)-superoxide 130-147 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-128 25670541-1 2015 PURPOSE: To report the long-term results of the ARO 95-06 randomized trial comparing hyperfractionated accelerated chemoradiation with mitomycin C/5-fluorouracil (C-HART) with hyperfractionated accelerated radiation therapy (HART) alone in locally advanced head and neck cancer. Mitomycin 135-146 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-51 26188906-0 2015 Zearalenone metabolism in human placental subcellular organelles, JEG-3 cells, and recombinant CYP19A1. Zearalenone 0-11 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-102 26189067-0 2015 Oxaliplatin added to fluorouracil-based preoperative chemoradiotherapy and postoperative chemotherapy of locally advanced rectal cancer (the German CAO/ARO/AIO-04 study): final results of the multicentre, open-label, randomised, phase 3 trial. Oxaliplatin 0-11 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 152-155 26189067-1 2015 BACKGROUND: Preoperative chemoradiotherapy with infusional fluorouracil, total mesorectal excision surgery, and postoperative chemotherapy with fluorouracil was established by the German CAO/ARO/AIO-94 trial as a standard combined modality treatment for locally advanced rectal cancer. Fluorouracil 144-156 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 191-194 26189067-14 2015 INTERPRETATION: Adding oxaliplatin to fluorouracil-based neoadjuvant chemoradiotherapy and adjuvant chemotherapy (at the doses and intensities used in this trial) significantly improved disease-free survival of patients with clinically staged cT3-4 or cN1-2 rectal cancer compared with our former fluorouracil-based combined modality regimen (based on CAO/ARO/AIO-94). Oxaliplatin 23-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 356-359 26189067-14 2015 INTERPRETATION: Adding oxaliplatin to fluorouracil-based neoadjuvant chemoradiotherapy and adjuvant chemotherapy (at the doses and intensities used in this trial) significantly improved disease-free survival of patients with clinically staged cT3-4 or cN1-2 rectal cancer compared with our former fluorouracil-based combined modality regimen (based on CAO/ARO/AIO-94). Fluorouracil 38-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 356-359 26067721-10 2015 Seven studies analyzed the association between AEs with different polymorphisms of CYP19A1. aes 47-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-90 25611452-3 2015 By analyzing the 10-year outcome data of the German CAO/ARO/AIO-94 phase 3 trial, the authors demonstrated that significantly fewer patients had poor prognostic features (eg, ypT3-4, ypN1-2) after preoperative 5-fluorouracil-based chemoradiotherapy. Fluorouracil 210-224 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-59 25765776-7 2015 Interestingly, mRNAs encoding the steroidogenic enzymes Hsd17b1, Cyp17a1 and Cyp19a1 were increased in all DBP-treated groups. Dibutyl Phthalate 107-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-84 25839436-0 2015 Aromatase inhibitor (anastrozole) affects growth of endometrioma cells in culture. Anastrozole 21-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 25839436-1 2015 OBJECTIVE: To study the effects of aromatase inhibitor (anastrozole) on the growth and estradiol secretion of endometrioma cells in culture. Anastrozole 56-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 35-44 25839436-4 2015 INTERVENTIONS: Testosterone at a concentration of 10 mug/mL was added as a substrate for the intracellular aromatase. Testosterone 15-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-116 25839436-9 2015 CONCLUSION: Aromatase inhibitor (anastrozole) may be an effective treatment for endometriosis due to inhibition of cellular aromatase. Anastrozole 33-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-21 25839436-9 2015 CONCLUSION: Aromatase inhibitor (anastrozole) may be an effective treatment for endometriosis due to inhibition of cellular aromatase. Anastrozole 33-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 124-133 25837259-4 2015 Furthermore, treatment with the aromatase substrate, testosterone, inhibited suspension culture-induced apoptosis whereas an aromatase inhibitor attenuated the effect of testosterone suggesting that suspended circulating ERalpha positive breast cancer cells may up-regulate intracrine estrogen activity for survival. Testosterone 53-65 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-41 25837259-4 2015 Furthermore, treatment with the aromatase substrate, testosterone, inhibited suspension culture-induced apoptosis whereas an aromatase inhibitor attenuated the effect of testosterone suggesting that suspended circulating ERalpha positive breast cancer cells may up-regulate intracrine estrogen activity for survival. Testosterone 170-182 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-134 25625755-0 2015 New cell culture model for aromatase inhibitor-resistant breast cancer shows sensitivity to fulvestrant treatment and cross-resistance between letrozole and exemestane. Letrozole 143-152 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-36 25625755-0 2015 New cell culture model for aromatase inhibitor-resistant breast cancer shows sensitivity to fulvestrant treatment and cross-resistance between letrozole and exemestane. exemestane 157-167 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-36 25625755-3 2015 Cell culture models for acquired resistance to the three clinically relevant AIs letrozole, anastrozole and exemestane were developed by selection and expansion of colonies of MCF-7 breast cancer cells surviving long-term AI treatment under conditions where endogenous aromatase-mediated conversion of androgen to estrogen was required for growth. exemestane 108-118 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 269-278 25450981-6 2015 In vitro experiments were performed using H23 and A549 NSCLC cell lines and exemestane was used for aromatase inhibition. exemestane 76-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-109 25275756-2 2015 The aim of our study was to investigate the expression and localization of ghrelin and its active receptor, growth hormone secretagogue receptor type 1a (GHS-R1a) in buffalo ovarian follicles of different follicular size and to investigate role of ghrelin on estradiol (E2) secretion, aromatase (CYP19A1), proliferating cell nuclear antigen (PCNA) and apoptosis regulator Bax gene expression on granulosa cell culture. Ghrelin 75-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 296-303 25462231-4 2015 Compound 23, 2-(1H-imidazol-1-yl)-1-(4-{[3(trifluoromethoxy)benzyl]oxy}phenyl) ethanone, was the most interesting inhibitor of the series displaying a high potency at CYP11B1 (IC50 = 15 nM), increased selectivities over CYP11B2 (SIB = 33), CYP19 (SIB = 390) and CYP17 (5% inhibition at 2.5 muM concentration). 2-(1h-imidazol-1-yl)-1-(4-{[3(trifluoromethoxy)benzyl]oxy}phenyl) ethanone 13-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 240-245 25612679-0 2015 Inhibition of Testosterone Aromatization by the Indole-3-carbinol Derivative CTet in CYP19A1-overexpressing MCF-7 Breast Cancer Cells. Testosterone 14-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 85-92 25612679-0 2015 Inhibition of Testosterone Aromatization by the Indole-3-carbinol Derivative CTet in CYP19A1-overexpressing MCF-7 Breast Cancer Cells. indole-3-carbinol 48-65 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 85-92 25612679-0 2015 Inhibition of Testosterone Aromatization by the Indole-3-carbinol Derivative CTet in CYP19A1-overexpressing MCF-7 Breast Cancer Cells. ctet 77-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 85-92 26100049-0 2015 A translational bioinformatic approach in identifying and validating an interaction between Vitamin A and CYP19A1. Vitamin A 92-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 106-113 26100049-8 2015 RESULTS: From a total of 868 interactions we were able to identify an interesting interaction between retinoic acid (i.e. Vitamin A) and the aromatase gene (i.e. CYP19A1). Tretinoin 102-115 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 162-169 26100049-8 2015 RESULTS: From a total of 868 interactions we were able to identify an interesting interaction between retinoic acid (i.e. Vitamin A) and the aromatase gene (i.e. CYP19A1). Vitamin A 122-131 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 162-169 26100049-9 2015 Our experimental results showed that retinoic acid at physiological concentration significantly influenced CYP19A1 gene expressions. Tretinoin 37-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-114 26635974-1 2015 Vorozole and letrozole are third-generation aromatase (cytochrome P450 19A1) inhibitors. vorozole 0-8 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 55-75 26635974-1 2015 Vorozole and letrozole are third-generation aromatase (cytochrome P450 19A1) inhibitors. Letrozole 13-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 55-75 25223798-7 2015 The present study clearly indicates that TBBPA alters JEG-3 cells estrogen synthesis due to its action on CYP19 protein expression and thus this compound may interfere with normal placental development during early pregnancy. tetrabromobisphenol A 41-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 106-111 25425647-0 2015 Evidence for an elevated aspartate pK(a) in the active site of human aromatase. Aspartic Acid 25-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-78 25425647-9 2015 Aromatase binding to anastrozole is pH-independent, consistent with the hypothesis that this ligand exploits a distinct set of interactions in the active site. Anastrozole 21-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 26434836-7 2015 The present study demonstrated that three flavonoids synergistically inhibit estrogen biosynthesis through aromatase inhibition, decrease CYP19 mRNA, and induce transcriptional suppression. Flavonoids 42-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-143 25275756-10 2015 Ghrelin treatment significantly (P<0.05) inhibited E2 secretion, CYP19A1 expression, apoptosis and promoted cell proliferation. Ghrelin 0-7 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-75 24702656-0 2015 Molecular docking and 3D-QSAR-based virtual screening of flavonoids as potential aromatase inhibitors against estrogen-dependent breast cancer. Flavonoids 57-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-90 24702656-1 2015 Aromatase, catalyzing final step of estrogen biosynthesis, is considered a key target for the development of drug against estrogen-dependent breast cancer (EDBC). ethyl vinyl ether 156-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 24702656-3 2015 Thus, a three-dimensional quantitative structure-activity relationship, using comparative molecular field analysis (CoMFA) was done on a series of 45 flavonoids against human aromatase. Flavonoids 150-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 175-184 24702656-8 2015 Thus, a 25 ns MD simulation analysis revealed high stability and effective binding of 7-hydroxyflavanone beta-D-glucopyranoside within the active site of aromatase. 7-Hydroxyflavanone beta-D-glucopyranoside 86-127 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-163 24702656-9 2015 To the best of our knowledge, this is the first report of CoMFA-based QSAR model for virtual screening of flavonoids as inhibitors of aromatase. Flavonoids 106-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 134-143 25234182-5 2015 There were found some remarkable differences in the effect of SFN at a dose of 5 micromol/L on CYP19 expression: in ER(+) MCF7 significant reduction, in ER(-) MDA-MB-231 an increased expression and unchanged expression in MCF10A cell line. sulforaphane 62-65 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-100 26535448-3 2015 Cytochrome P450 aromatase (CYP19) catalyses the conversion of androstenedione and testosterone into oestrone and oestradiol, respectively. Androstenedione 62-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-32 26535448-3 2015 Cytochrome P450 aromatase (CYP19) catalyses the conversion of androstenedione and testosterone into oestrone and oestradiol, respectively. Testosterone 82-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-32 26535448-3 2015 Cytochrome P450 aromatase (CYP19) catalyses the conversion of androstenedione and testosterone into oestrone and oestradiol, respectively. Estrone 100-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-32 26535448-3 2015 Cytochrome P450 aromatase (CYP19) catalyses the conversion of androstenedione and testosterone into oestrone and oestradiol, respectively. Estradiol 113-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-32 25358120-2 2014 Irradiation of aryl imidazylates and of the corresponding imidazolium salts mainly caused homolysis of the ArO-S bond. imidazolium salts 58-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-110 25516776-7 2014 Rodel et al reported on the CAO/ARO/AIO-04 randomized phase III trial that compared standard treatment with 5-FU and radiation therapy, to oxaliplatin plus 5-FU in association with radiation therapy. Fluorouracil 108-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-35 25516776-7 2014 Rodel et al reported on the CAO/ARO/AIO-04 randomized phase III trial that compared standard treatment with 5-FU and radiation therapy, to oxaliplatin plus 5-FU in association with radiation therapy. Oxaliplatin 139-150 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-35 25516776-7 2014 Rodel et al reported on the CAO/ARO/AIO-04 randomized phase III trial that compared standard treatment with 5-FU and radiation therapy, to oxaliplatin plus 5-FU in association with radiation therapy. Fluorouracil 156-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-35 25203739-6 2014 This paper reviews the role of genetic polymorphisms of CYP19A1, CYP3A4, and UGT1A4 in the responses of female hormone receptor-positive postmenopausal breast cancer patients to anastrozole. Anastrozole 178-189 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-63 25243857-2 2014 Aromatase inhibition by the triazole compounds letrozole (LTZ) and anastrozole is a prevalent therapy for estrogen-dependent postmenopausal breast cancer. Triazoles 28-36 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 25243857-2 2014 Aromatase inhibition by the triazole compounds letrozole (LTZ) and anastrozole is a prevalent therapy for estrogen-dependent postmenopausal breast cancer. Letrozole 47-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 25243857-2 2014 Aromatase inhibition by the triazole compounds letrozole (LTZ) and anastrozole is a prevalent therapy for estrogen-dependent postmenopausal breast cancer. Letrozole 58-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 25243857-2 2014 Aromatase inhibition by the triazole compounds letrozole (LTZ) and anastrozole is a prevalent therapy for estrogen-dependent postmenopausal breast cancer. Anastrozole 67-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 25243857-6 2014 We have evaluated the inhibitory effects of 3 common fungicides, bifonazole, imazalil, and flusilazole, in human aromatase purified from placenta and compared them with LTZ, the most potent inhibitor of aromatase. flusilazole 91-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-122 25243857-6 2014 We have evaluated the inhibitory effects of 3 common fungicides, bifonazole, imazalil, and flusilazole, in human aromatase purified from placenta and compared them with LTZ, the most potent inhibitor of aromatase. Letrozole 169-172 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 203-212 25243857-10 2014 We interpret the inhibition mechanism in the context of the x-ray structure of aromatase-androstenedione complex. Androstenedione 89-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-88 25243857-11 2014 Structural data show that aromatase has 3 binding pockets in relation to the heme. Heme 77-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-35 25243857-12 2014 The substrate-binding cavity at the heme-distal site closely compliments the structures of the natural substrate, androstenedione, and steroidal aromatase inhibitors. Heme 36-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 145-154 25243857-12 2014 The substrate-binding cavity at the heme-distal site closely compliments the structures of the natural substrate, androstenedione, and steroidal aromatase inhibitors. Androstenedione 114-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 145-154 25226294-2 2014 These tumors overexpress aromatase (CYP19A1), which leads to increased conversion of delta-4-androstenedione to estrone and testosterone to estradiol. Androstenedione 85-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-43 25226294-2 2014 These tumors overexpress aromatase (CYP19A1), which leads to increased conversion of delta-4-androstenedione to estrone and testosterone to estradiol. Estrone 112-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-43 25226294-2 2014 These tumors overexpress aromatase (CYP19A1), which leads to increased conversion of delta-4-androstenedione to estrone and testosterone to estradiol. Testosterone 124-136 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-43 25226294-2 2014 These tumors overexpress aromatase (CYP19A1), which leads to increased conversion of delta-4-androstenedione to estrone and testosterone to estradiol. Estradiol 140-149 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-43 24787658-10 2014 These results suggest that a maternal LP diet in pregnancy and lactation elevated serum 17beta-estradiol level by activating CYP19A1 through miRNA-mediated mechanism, and induced granulosa apoptosis in graafian follicles through ER-activated Fas/FasL-caspase 3 pathway. Estradiol 88-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-132 25252141-1 2014 Aromatase is the cytochrome P450 enzyme that cleaves the C10-C19 carbon-carbon bond of androgens to form estrogens, in a three-step process. Carbon 65-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 25252141-1 2014 Aromatase is the cytochrome P450 enzyme that cleaves the C10-C19 carbon-carbon bond of androgens to form estrogens, in a three-step process. Carbon 72-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 25252141-3 2014 Incubation of purified aromatase with its 19-deutero-19-oxo androgen substrate was performed in the presence of (18)O2, and the products were derivatized using a novel diazo reagent. 19-deutero-19-oxo 42-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 23-32 25252141-3 2014 Incubation of purified aromatase with its 19-deutero-19-oxo androgen substrate was performed in the presence of (18)O2, and the products were derivatized using a novel diazo reagent. Oxygen 116-118 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 23-32 25252141-3 2014 Incubation of purified aromatase with its 19-deutero-19-oxo androgen substrate was performed in the presence of (18)O2, and the products were derivatized using a novel diazo reagent. diazo reagent 168-181 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 23-32 25252141-7 2014 Thus, the evidence supports Compound I and not ferric peroxide as the active iron species in the third step of the steroid aromatase reaction. Iron 77-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 123-132 25130462-4 2014 We investigated the effect of 7days testosterone treatment (100nmol/l) on glucose transport and gene expression levels of hormone receptors and enzymes involved in the synthesis and conversion of testosterone (HSD17B1, HSD17B2, CYP19A1, SRD5A1-2, AR, ER-alpha, HSD17B6 and AKR1-3) in myotubes from ten patients with PCOS and ten matched controls. Testosterone 196-208 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 228-235 25155384-0 2014 Discovery of a new class of cinnamyl-triazole as potent and selective inhibitors of aromatase (cytochrome P450 19A1). cinnamyl-triazole 28-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-115 24814680-2 2014 A small number of xenobiotic metabolizing CYP enzymes are constantly expressed in placenta, those include CYP19A1 which mainly converts androgens to estrogens, and CYP1A1 whose substrates include steroid hormones and xenobiotics. Steroids 196-212 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 106-113 25010478-4 2014 Transcriptome analyses (microarrays, northern blots and quantitative PCR) of testes discovered that dexamethasone treatment decreased concentrations of glucocorticoid receptor alpha (NR3C1), alpha actinin 4 (ACTN4), luteinizing hormone receptor (LHCGR), squalene epoxidase (SQLE), 24-dehydrocholesterol reductase (DHCR24), glutathione S-transferase A3 (GSTA3) and aromatase (CYP19A1) mRNAs. Dexamethasone 100-113 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 375-382 25010478-9 2014 In conclusion, dexamethasone acutely decreased the expression of genes involved in hormone signaling (NR3C1, ACTN4 and LHCGR), cholesterol synthesis (SQLE and DHCR24) and steroidogenesis (GSTA3 and CYP19A1) along with testosterone production. Dexamethasone 15-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 198-205 24853760-0 2014 Two natural products, trans-phytol and (22E)-ergosta-6,9,22-triene-3beta,5alpha,8alpha-triol, inhibit the biosynthesis of estrogen in human ovarian granulosa cells by aromatase (CYP19). 8alpha-triol 80-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 178-183 24836190-0 2014 Aromatase induction in tamoxifen-resistant breast cancer: Role of phosphoinositide 3-kinase-dependent CREB activation. Tamoxifen 23-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 24836190-4 2014 Here, we found that the basal expression and activity of aromatase were significantly increased in tamoxifen (TAM)-resistant human breast cancer (TAMR-MCF-7) cells compared to control MCF-7 cells. Tamoxifen 99-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-66 24836190-4 2014 Here, we found that the basal expression and activity of aromatase were significantly increased in tamoxifen (TAM)-resistant human breast cancer (TAMR-MCF-7) cells compared to control MCF-7 cells. Tamoxifen 110-113 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-66 24836190-5 2014 We further revealed that aromatase immunoreactivity in tumor tissues was increased in recurrence group after TAM therapy compared to non-recurrence group after TAM therapy. Tamoxifen 109-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-34 24836190-5 2014 We further revealed that aromatase immunoreactivity in tumor tissues was increased in recurrence group after TAM therapy compared to non-recurrence group after TAM therapy. Tamoxifen 160-163 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-34 24836190-9 2014 Our findings suggest that aromatase expression is up-regulated in TAM-resistant breast cancer via PI3K/Akt-dependent CREB activation. Tamoxifen 66-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-35 24997347-1 2014 CYP19A1, or human aromatase catalyzes the conversion of androgens to estrogens in a three-step reaction through the formation of 19-hydroxy and 19-aldehyde intermediates. 19-hydroxy and 19-aldehyde 129-155 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-7 24853760-0 2014 Two natural products, trans-phytol and (22E)-ergosta-6,9,22-triene-3beta,5alpha,8alpha-triol, inhibit the biosynthesis of estrogen in human ovarian granulosa cells by aromatase (CYP19). trans-phytol 22-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 178-183 24853760-0 2014 Two natural products, trans-phytol and (22E)-ergosta-6,9,22-triene-3beta,5alpha,8alpha-triol, inhibit the biosynthesis of estrogen in human ovarian granulosa cells by aromatase (CYP19). (22e)-ergosta-6,9,22-triene-3beta 39-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 178-183 24853760-0 2014 Two natural products, trans-phytol and (22E)-ergosta-6,9,22-triene-3beta,5alpha,8alpha-triol, inhibit the biosynthesis of estrogen in human ovarian granulosa cells by aromatase (CYP19). Bromocriptine 73-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 178-183 24737458-6 2014 METHODS: A genetic score predicting 17beta-estradiol was developed in 237 young Chinese women (university students) from Hong Kong based on a parsimonious set of genetic polymorphisms (ESR1 (rs2175898) and CYP19A1 (rs1008805)). Estradiol 36-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 206-213 24969759-2 2014 Forskolin, a PKA activator, significantly increased [(3)H]DHEAS uptake and the mRNA expression levels of organic anion transporter (OAT) 4 and CYP19A1 in choriocarcinoma JEG-3 cells, while other steroid sulfate transporters present in the placenta showed no change in expression level. Colforsin 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 143-150 25097392-4 2014 The frequency distribution of CYP19 polymorphism was assessed by polymerase chain reaction confronting two pair primers (PCR-CTPP). ctpp 125-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-35 25097392-5 2014 RESULTS: CYP19 polymorphism at codon 39 Trp/Arg (W39R) results in three genotypes TT, TC, and CC, but in the present study CC genotype was not found in breast cancer cases as well as in controls. Technetium 86-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-14 24873989-2 2014 The latter complex, in combination with B(C6F5)3, reacts with ArO-H substrates via formal hydrogen-atom transfer and exhibits dramatically increased reaction rates over the Mn(V)(O) starting material. tris(pentafluorophenyl)borane 40-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-65 24873989-2 2014 The latter complex, in combination with B(C6F5)3, reacts with ArO-H substrates via formal hydrogen-atom transfer and exhibits dramatically increased reaction rates over the Mn(V)(O) starting material. Hydrogen 90-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-65 24486430-6 2014 Atrazine and prochloraz had cell-type specific effects on CYP19 activity and estrogen production in co-culture. Atrazine 0-8 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-63 24705958-3 2014 The substrate androstenedione was incubated with human CYP 19A1 supersomes in the presence of NADPH for 30 min, and estrone formation was determined by LC-MS/MS analysis. Androstenedione 14-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 55-63 24705958-3 2014 The substrate androstenedione was incubated with human CYP 19A1 supersomes in the presence of NADPH for 30 min, and estrone formation was determined by LC-MS/MS analysis. NADP 94-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 55-63 24584631-1 2014 Carbon-carbon bond cleavage reactions are catalyzed by, among others, lanosterol 14-demethylase (CYP51), cholesterol side-chain cleavage enzyme (CYP11), sterol 17beta-lyase (CYP17), and aromatase (CYP19). Carbon 0-6 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 197-202 24584631-1 2014 Carbon-carbon bond cleavage reactions are catalyzed by, among others, lanosterol 14-demethylase (CYP51), cholesterol side-chain cleavage enzyme (CYP11), sterol 17beta-lyase (CYP17), and aromatase (CYP19). Carbon 7-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 197-202 24584631-1 2014 Carbon-carbon bond cleavage reactions are catalyzed by, among others, lanosterol 14-demethylase (CYP51), cholesterol side-chain cleavage enzyme (CYP11), sterol 17beta-lyase (CYP17), and aromatase (CYP19). Cholesterol 105-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 197-202 23696345-13 2014 Attenuation of activated ERK/MAPK levels, reportedly responsible for reduced human cyp19 transcription, was also observed in ketamine-treated embryos. Ketamine 125-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-88 24684533-5 2014 Moreover, testosterone inhibits aromatase activity. Testosterone 10-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-41 24684533-8 2014 A hormone involved in aromatase activity is vitamin D, which downregulates aromatase in human RA macrophages. Vitamin D 44-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 22-31 24684533-8 2014 A hormone involved in aromatase activity is vitamin D, which downregulates aromatase in human RA macrophages. Vitamin D 44-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-84 24684533-9 2014 Collectively, evidence suggests a key role of aromatase in sex hormone balance during chronic inflammation and points to the importance of vitamin D as a possible new tool for aromatase modulation. Vitamin D 139-148 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 176-185 24676756-0 2014 11C-cetrozole: an improved C-11C-methylated PET probe for aromatase imaging in the brain. cetrozole 0-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-67 24676756-0 2014 11C-cetrozole: an improved C-11C-methylated PET probe for aromatase imaging in the brain. c-11c 27-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-67 24676756-2 2014 To investigate the physiologic and pathologic importance of aromatase in the brain, including in humans, we here report the development of a novel PET probe for aromatase, (11)C-cetrozole, which allows noninvasive quantification of aromatase expression. cetrozole 178-187 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-69 24676756-2 2014 To investigate the physiologic and pathologic importance of aromatase in the brain, including in humans, we here report the development of a novel PET probe for aromatase, (11)C-cetrozole, which allows noninvasive quantification of aromatase expression. cetrozole 178-187 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 161-170 24676756-2 2014 To investigate the physiologic and pathologic importance of aromatase in the brain, including in humans, we here report the development of a novel PET probe for aromatase, (11)C-cetrozole, which allows noninvasive quantification of aromatase expression. cetrozole 178-187 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 161-170 24676756-6 2014 RESULTS: In vitro and in vivo studies using (11)C-cetrozole showed its superiority in brain aromatase imaging in terms of specificity and selectivity, compared with previously developed (11)C-vorozole. cetrozole 44-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 24676756-9 2014 CONCLUSION: These results suggest that PET imaging with newly developed (11)C-cetrozole is suitable for quantifying the expression of brain aromatase in vivo, possibly providing critical information regarding the functional roles of aromatase in human neurologic and emotional disorders. cetrozole 72-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 140-149 24725461-2 2014 The conversion by aromatase (CYP19A1) of androstenedione into estrone (E1) and of testosterone into 17beta-estradiol (E2) plays a key role in the endogenous synthesis of estrogens in tissue. Androstenedione 41-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-36 24725461-2 2014 The conversion by aromatase (CYP19A1) of androstenedione into estrone (E1) and of testosterone into 17beta-estradiol (E2) plays a key role in the endogenous synthesis of estrogens in tissue. Estrone 62-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-36 24725461-2 2014 The conversion by aromatase (CYP19A1) of androstenedione into estrone (E1) and of testosterone into 17beta-estradiol (E2) plays a key role in the endogenous synthesis of estrogens in tissue. Estradiol 100-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-36 24630560-3 2014 Androstenedione was used as a substrate and incubated with R. verniciflua extract in cDNA-expressed CYP19 supersomes in the presence of NADPH, and estrone formation was measured using liquid chromatography-tandem mass spectrometry. Androstenedione 0-15 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-105 24630560-3 2014 Androstenedione was used as a substrate and incubated with R. verniciflua extract in cDNA-expressed CYP19 supersomes in the presence of NADPH, and estrone formation was measured using liquid chromatography-tandem mass spectrometry. NADP 136-141 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-105 24486430-6 2014 Atrazine and prochloraz had cell-type specific effects on CYP19 activity and estrogen production in co-culture. prochloraz 13-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-63 24486430-7 2014 Atrazine induced CYP19 activity and estrogen production in H295R cells only, but did not affect overall estrogen production in co-culture, whereas prochloraz inhibited CYP19 activity exclusively in BeWo cells and reduced estrogen production in co-culture by almost 90%. Atrazine 0-8 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-22 24486430-7 2014 Atrazine induced CYP19 activity and estrogen production in H295R cells only, but did not affect overall estrogen production in co-culture, whereas prochloraz inhibited CYP19 activity exclusively in BeWo cells and reduced estrogen production in co-culture by almost 90%. prochloraz 147-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 168-173 23676512-3 2014 We report the first case of cutaneous vasculitis clearly associated with the use of aromatase inhibitor letrozole. Letrozole 104-113 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-93 24590773-2 2014 We investigated the relationship between these specific AEs and single nucleotide polymorphisms (SNPs) in the CYP19A1 gene in postmenopausal, hormone receptor-positive early breast cancer (BC) patients treated with adjuvant exemestane for 5 years. alanylglutamic acid 56-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 110-117 24590773-2 2014 We investigated the relationship between these specific AEs and single nucleotide polymorphisms (SNPs) in the CYP19A1 gene in postmenopausal, hormone receptor-positive early breast cancer (BC) patients treated with adjuvant exemestane for 5 years. exemestane 224-234 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 110-117 24361490-5 2014 The PAH also increased mRNA expressions of other trophoblastic differentiation markers, including those of the steroid metabolism enzymes CYP19A1 and HSD11B2 and of the fusogenic protein syncytin-2; in parallel, it triggered syncytialisation of BeWo cells. Steroids 111-118 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-145 24456285-0 2014 An innovative LC-MS/MS-based method for determining CYP 17 and CYP 19 activity in the adipose tissue of pre- and postmenopausal and ovariectomized women using 13C-labeled steroid substrates. 13c 159-162 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-69 24456285-0 2014 An innovative LC-MS/MS-based method for determining CYP 17 and CYP 19 activity in the adipose tissue of pre- and postmenopausal and ovariectomized women using 13C-labeled steroid substrates. Steroids 171-178 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-69 24456285-12 2014 CONCLUSION: Our findings suggest that adipose tissue acts as an endocrine organ, with CYP17 and CYP19 activity playing an essential role in sex steroid hormone biosynthesis. Steroids 144-159 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-101 24481325-9 2014 GPER-induced aromatase upregulation was probably responsible for this phenomenon, as TAM- and G1-induced CYP19A1 gene expression was reduced by GPER knockdown and G15, AG1478, and U0126 administration. U 0126 180-185 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 105-112 24678481-3 2014 These rapid changes in StAR and Cyp19a1 gene expression after the LH surge efficiently facilitate progesterone production, which plays a crucial role in ovulation and the following luteinization. Progesterone 98-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-39 24014089-0 2014 Everolimus in combination with letrozole inhibit human breast cancer MCF-7/Aro stem cells via PI3K/mTOR pathway: an experimental study. Everolimus 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-78 24552606-12 2014 This duplication, occurring within the aromatase alpha-helix, would be likely to disrupt substrate (androgen) and cofactor (protoporphyrin IX) binding, resulting in a lack of oestrogen synthesis. protoporphyrin IX 124-141 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-48 24274332-6 2014 Inhibition of other CYPs, such as aromatase (CYP19), can lead to numerous toxicological effects, which are also evident from high dose human exposures to therapeutic azoles. Azoles 166-172 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-50 24014089-0 2014 Everolimus in combination with letrozole inhibit human breast cancer MCF-7/Aro stem cells via PI3K/mTOR pathway: an experimental study. Letrozole 31-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-78 24014089-1 2014 This study evaluated the effects of an mTOR inhibitor everolimus alone or in combination with letrozole on MCF-7/Aro (MCF-7 cells stably transfected with CYP19) in vitro and in vivo. Everolimus 54-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-159 24014089-8 2014 In vitro, compared with MCF-7/Aro NSCs, there were greater resistance to the standard treatment doses of letrozole and everolimus in MCF-7/Aro SCs (17- and 15-fold, respectively). Letrozole 105-114 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-142 24014089-8 2014 In vitro, compared with MCF-7/Aro NSCs, there were greater resistance to the standard treatment doses of letrozole and everolimus in MCF-7/Aro SCs (17- and 15-fold, respectively). Everolimus 119-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-33 24014089-8 2014 In vitro, compared with MCF-7/Aro NSCs, there were greater resistance to the standard treatment doses of letrozole and everolimus in MCF-7/Aro SCs (17- and 15-fold, respectively). Everolimus 119-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-142 24729854-8 2014 The stable phenoxyl radical 2,4,6- t Bu3C6H2O (ArO ) forms a stronger bond to (TMP)FeII to irreversibly make a similar FeIII(OR) complex. phenoxyl radical 2,4,6- t bu3c6h2o 11-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-51 24434719-0 2014 Aromatase inhibitors in the breast cancer clinic: focus on exemestane. exemestane 59-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 24434719-3 2014 The main anti-estrogen treatments in use consist of selective estrogen-receptor modulators, such as tamoxifen, and third-generation aromatase inhibitors (AIs), such as exemestane, letrozole, and anastrozole. exemestane 168-178 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 132-141 24729854-8 2014 The stable phenoxyl radical 2,4,6- t Bu3C6H2O (ArO ) forms a stronger bond to (TMP)FeII to irreversibly make a similar FeIII(OR) complex. tmp 80-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-51 24398390-6 2014 Lung tumors from both males and females also express CYP19 (aromatase), the rate-limiting enzyme in estrogen synthesis that converts testosterone to estrone and beta-estradiol. Testosterone 133-145 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-58 24782009-8 2014 In granulosa cells from small antral follicles, FSH stimulates the synthesis of aromatase (Cyp19) which catalyzes the conversion of theca-derived androstenedione to estradiol. Androstenedione 146-161 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-96 24782009-8 2014 In granulosa cells from small antral follicles, FSH stimulates the synthesis of aromatase (Cyp19) which catalyzes the conversion of theca-derived androstenedione to estradiol. Estradiol 165-174 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-96 25002878-4 2014 A tetranucleotide (TTTA)n microsatellite repeat polymorphism, at intron 4 of the CYP19 (aromatase) gene, has been previously associated with higher lumbar spine bone mineral density (LS-BMD) and lower risk of spine fracture in postmenopausal women. tetranucleotide 2-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-86 25002878-4 2014 A tetranucleotide (TTTA)n microsatellite repeat polymorphism, at intron 4 of the CYP19 (aromatase) gene, has been previously associated with higher lumbar spine bone mineral density (LS-BMD) and lower risk of spine fracture in postmenopausal women. ttta 19-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-86 24603137-0 2014 Human granulosa cells: insulin and insulin-like growth factor-1 receptors and aromatase expression modulation by metformin. Metformin 113-122 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-87 24603137-3 2014 The aim of this study was to evaluate gene and protein expression of an insulin receptor (IR), insulin-like growth factor-1 (IGF1) receptor (IGF1R) and aromatase in granulosa cells treated with metformin and insulin. Metformin 194-203 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 152-161 24603137-7 2014 Aromatase mRNA expression was significantly reduced in metformin-incubated cells following stimulation with insulin for 30 min. Metformin 55-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 24603137-9 2014 CONCLUSION: A direct effect of metformin on the gene expression of IGF1R, IR and aromatase was observed. Metformin 31-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-90 24398390-6 2014 Lung tumors from both males and females also express CYP19 (aromatase), the rate-limiting enzyme in estrogen synthesis that converts testosterone to estrone and beta-estradiol. Estrone 149-156 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-58 24398390-6 2014 Lung tumors from both males and females also express CYP19 (aromatase), the rate-limiting enzyme in estrogen synthesis that converts testosterone to estrone and beta-estradiol. Estradiol 161-175 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-58 24148837-3 2013 Virtual screening by molecular docking against clinical targets of steroidal anticancer drugs (ERalpha, AR, Aromatase and CYP17A1) suggests that 17(E)-picolinylidene, but not 17alpha-picolyl androstanes could specifically interact with CYP17A1 (17alpha-hydroxylase) with similar geometry and affinity as Abiraterone, a 17-pyridinyl androstane drug clinically used in the treatment of prostate cancer. 17(e)-picolinylidene 145-165 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-117 24049095-7 2013 RESULTS: We identified a novel genetic region at 15q21.2 (rs2414095 in CYP19A1), which was significantly associated with oestradiol and FSH in the Chinese population at a genome-wide significant level (p=6.54x10(-31) and 1.59x10(-16), respectively). Estradiol 121-131 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-78 24049095-8 2013 Another single nucleotide polymorphism in CYP19A1 gene was significantly associated with oestradiol level (rs2445762, p=7.75x10(-28)). Estradiol 89-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-49 24113062-0 2013 Investigation of aryl halides as ketone bioisosteres: refinement of potent and selective inhibitors of human cytochrome P450 19A1 (aromatase). aryl halides 17-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 109-129 23850856-1 2013 BACKGROUND: Exposure to bisphenol A (BPA), a chemical widely used in consumer products, has been associated with in vitro Cyp19 gene expression. bisphenol A 24-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-127 23850856-1 2013 BACKGROUND: Exposure to bisphenol A (BPA), a chemical widely used in consumer products, has been associated with in vitro Cyp19 gene expression. bisphenol A 37-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-127 23850856-4 2013 METHODS: Mixed effect models were used to evaluate the association of urinary BPA concentrations with granulosa cell Cyp19 mRNA expression. bisphenol A 78-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 117-122 24113062-0 2013 Investigation of aryl halides as ketone bioisosteres: refinement of potent and selective inhibitors of human cytochrome P450 19A1 (aromatase). Ketones 33-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 109-129 23846817-4 2013 OBJECTIVE: The aim of this study was to investigate metformin"s interaction with the FSH/cAMP/protein kinase A pathway, which is the primary signaling pathway controlling CYP19A1 (aromatase) expression in the ovary. Cyclic AMP 89-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 171-178 24012714-0 2013 QSAR modeling of aromatase inhibitory activity of 1-substituted 1,2,3-triazole analogs of letrozole. 1-substituted 1,2,3-triazole 50-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 24012714-0 2013 QSAR modeling of aromatase inhibitory activity of 1-substituted 1,2,3-triazole analogs of letrozole. Letrozole 90-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 24025069-1 2013 A small library of both [2,3-h] and [3,2-f] novel pyrroloquinolines equipped with an azolylmethyl group was designed and synthesized as nonsteroidal CYP19 aromatase inhibitors. pyrroloquinoline 50-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 149-154 24065098-9 2013 The findings of this study indicate that rs4646 polymorphism in the CYP19A1 gene may serve as a prognostic maker of the response to anastrozole in patients with MBC who are treated with anastrozole. Anastrozole 186-197 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-75 26217113-5 2013 However, randomised phase III trials, evaluating the addition of oxaliplatin at low doses plus preoperative fluoropyrimidine-based chemoradiotherapy (CRT), have in the main failed to show a significant improvement on early pathological response, with the exception of the German CAO/ARO/AIO-04 study. Oxaliplatin 65-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 283-286 23683135-9 2013 In addition, the enzyme aromatase CYP19 inhibitory activity of benzofurans having -OH and -OMe were greater than that observed for the reference arimidex. Benzofurans 63-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-39 23683135-9 2013 In addition, the enzyme aromatase CYP19 inhibitory activity of benzofurans having -OH and -OMe were greater than that observed for the reference arimidex. -oh and -ome 82-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-39 23683135-9 2013 In addition, the enzyme aromatase CYP19 inhibitory activity of benzofurans having -OH and -OMe were greater than that observed for the reference arimidex. Anastrozole 145-153 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-39 23178495-7 2013 The anchorage-independent growth of MCF-10A(arom) cells can be completely abolished by pre-treatment with the aromatase inhibitor, letrozole. Letrozole 131-140 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 110-119 24065098-3 2013 The aim was to investigate the impact of the genetic polymorphisms, CYP19A1, 17-beta-HSD-1 and FTO, on the response to anastrozole in metastatic breast carcinoma (MBC) and to evaluate the impact of those polymorphisms on various clinicopathologic features. Anastrozole 119-130 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-75 24065098-9 2013 The findings of this study indicate that rs4646 polymorphism in the CYP19A1 gene may serve as a prognostic maker of the response to anastrozole in patients with MBC who are treated with anastrozole. Anastrozole 132-143 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-75 23824607-7 2013 The inhibitory abilities of E-, mixed, and Z-norendoxifen against recombinant aromatase (CYP19), CYP1A2, CYP3A4, CYP3A5, and CYP2C19 were tested using microsomal incubations. N,N-didesmethyl-4-hydroxytamoxifen 43-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 89-94 23824607-9 2013 E-Norendoxifen had a 9.3-fold-higher inhibitory ability than Z-norendoxifen against CYP19, while E- and Z-norendoxifen had similar potencies against CYP1A2, CYP3A4, CYP3A5, and CYP2C19. E-Norendoxifen 0-14 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-89 23824607-9 2013 E-Norendoxifen had a 9.3-fold-higher inhibitory ability than Z-norendoxifen against CYP19, while E- and Z-norendoxifen had similar potencies against CYP1A2, CYP3A4, CYP3A5, and CYP2C19. N,N-didesmethyl-4-hydroxytamoxifen 61-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-89 23846817-4 2013 OBJECTIVE: The aim of this study was to investigate metformin"s interaction with the FSH/cAMP/protein kinase A pathway, which is the primary signaling pathway controlling CYP19A1 (aromatase) expression in the ovary. Metformin 52-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 171-178 25157205-4 2013 Recent studies in our laboratory indicate that brain aromatase activity is rapidly inhibited by an increase in intracellular calcium concentration resulting from potassium-induced depolarization or from the activation of glutamatergic receptors. Calcium 125-132 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 23727336-10 2013 In summary, TCDD could induce CYP19 transcription in brain cells. Polychlorinated Dibenzodioxins 12-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-35 23859149-2 2013 Via a combination of ligand- and structure-based approaches, a series of dual inhibitors were designed leading to the 2-(3-pyridyl)naphthalenes 10 and 11 with strong inhibition of both enzymes (IC50 values around 20 nM) and excellent selectivities over CYP11B1, CYP19, and CYP3A4. 2-(3-pyridyl)naphthalenes 118-143 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 262-267 23090135-0 2013 Modulation of CYP19 expression by cabbage juices and their active components: indole-3-carbinol and 3,3"-diindolylmethene in human breast epithelial cell lines. indole-3-carbinol 78-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-19 23090135-0 2013 Modulation of CYP19 expression by cabbage juices and their active components: indole-3-carbinol and 3,3"-diindolylmethene in human breast epithelial cell lines. 3,3"-diindolylmethene 100-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-19 23090135-1 2013 PURPOSE: The aim of this study was to evaluate the effect of white cabbage and sauerkraut juices of different origin and indole-3-carbinol (I3C) and diindolylmethane (DIM) on expression of CYP19 gene encoding aromatase, the key enzyme of estrogen synthesis. indole-3-carbinol 121-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 189-194 23090135-1 2013 PURPOSE: The aim of this study was to evaluate the effect of white cabbage and sauerkraut juices of different origin and indole-3-carbinol (I3C) and diindolylmethane (DIM) on expression of CYP19 gene encoding aromatase, the key enzyme of estrogen synthesis. 3,3'-diindolylmethane 149-165 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 189-194 23090135-4 2013 RESULTS: Remarkable differences in the effect on CYP19 transcript and protein level were found between the cabbage juices (in 2.5-25 mL/L concentrations) and indoles (in 2.5-50 muM doses) in the three cell lines. Indoles 158-165 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-54 23851904-7 2013 Administration of the CYP3A4 inducer rifampicin produced distinct differences in CYP3A4 and CYP11B1, CYP19A1, HSD11B, and HSD17B, which were indicated by their heat map-visualized steroid signatures. Rifampin 37-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-108 23851904-7 2013 Administration of the CYP3A4 inducer rifampicin produced distinct differences in CYP3A4 and CYP11B1, CYP19A1, HSD11B, and HSD17B, which were indicated by their heat map-visualized steroid signatures. Steroids 180-187 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-108 23777398-1 2013 Measurements of aroxyl radical (ArO( ))-scavenging rate constants (ks(AOH)) of antioxidants (AOHs) (alpha-tocopherol (alpha-TocH), ubiquinol-10 (UQ10H2), and sodium ascorbate (Na(+)AsH(-))) were performed in 2-propanol/water (2-PrOH/H2O, 5/1, v/v) solution using stopped-flow spectrophotometry. aroxyl radical 16-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-35 23777398-1 2013 Measurements of aroxyl radical (ArO( ))-scavenging rate constants (ks(AOH)) of antioxidants (AOHs) (alpha-tocopherol (alpha-TocH), ubiquinol-10 (UQ10H2), and sodium ascorbate (Na(+)AsH(-))) were performed in 2-propanol/water (2-PrOH/H2O, 5/1, v/v) solution using stopped-flow spectrophotometry. Potassium 67-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-35 23777398-1 2013 Measurements of aroxyl radical (ArO( ))-scavenging rate constants (ks(AOH)) of antioxidants (AOHs) (alpha-tocopherol (alpha-TocH), ubiquinol-10 (UQ10H2), and sodium ascorbate (Na(+)AsH(-))) were performed in 2-propanol/water (2-PrOH/H2O, 5/1, v/v) solution using stopped-flow spectrophotometry. alternariol 93-97 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-35 23777398-1 2013 Measurements of aroxyl radical (ArO( ))-scavenging rate constants (ks(AOH)) of antioxidants (AOHs) (alpha-tocopherol (alpha-TocH), ubiquinol-10 (UQ10H2), and sodium ascorbate (Na(+)AsH(-))) were performed in 2-propanol/water (2-PrOH/H2O, 5/1, v/v) solution using stopped-flow spectrophotometry. alpha-Tocopherol 100-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-35 23777398-1 2013 Measurements of aroxyl radical (ArO( ))-scavenging rate constants (ks(AOH)) of antioxidants (AOHs) (alpha-tocopherol (alpha-TocH), ubiquinol-10 (UQ10H2), and sodium ascorbate (Na(+)AsH(-))) were performed in 2-propanol/water (2-PrOH/H2O, 5/1, v/v) solution using stopped-flow spectrophotometry. ubiquinol-10 131-143 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-35 25157205-4 2013 Recent studies in our laboratory indicate that brain aromatase activity is rapidly inhibited by an increase in intracellular calcium concentration resulting from potassium-induced depolarization or from the activation of glutamatergic receptors. Potassium 162-171 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 23402939-1 2013 Effects of 96 h aflatoxin B1 (AFB1) exposure at concentrations from 0.2 muM to 6 muM on the mRNA and protein expression levels of the following transporters ABCB1/B4, ABCC1, ABCC2, ABCG2, OAT4 and the mRNA expression of steroid-metabolizing enzymes CYP1A1, CYP19A1, HSD3B1 and HSD17B1, and conjugating enzyme family UGT1A were evaluated in trophoblastic JEG-3 cells. Aflatoxin B1 16-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 257-264 23669293-1 2013 OBJECTIVES: To investigate the association between polymorphisms of aromatase (encoded by the CYP19A1 gene and a key enzyme in biosynthesis of oestradiol) and the risk of lung cancer, and whether there were differences stratified by sex and smoking history. Estradiol 143-153 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-101 23402939-1 2013 Effects of 96 h aflatoxin B1 (AFB1) exposure at concentrations from 0.2 muM to 6 muM on the mRNA and protein expression levels of the following transporters ABCB1/B4, ABCC1, ABCC2, ABCG2, OAT4 and the mRNA expression of steroid-metabolizing enzymes CYP1A1, CYP19A1, HSD3B1 and HSD17B1, and conjugating enzyme family UGT1A were evaluated in trophoblastic JEG-3 cells. Aflatoxin B1 30-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 257-264 23402939-6 2013 When an inhibitor of CYP19A1, finrozole, was dosed simultaneously with AFB1, no increases in the transcripts of transporters or steroid hydroxylases or CYP19A1 were observed. finrozole 30-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-28 23485457-4 2013 The NFkappaB inhibitor BAY-11-7082 dose-dependently inhibited transcription of EGR2 and EGR3 mRNA as well as total and PI.4-specific CYP19A1 mRNA. 3-(4-methylphenylsulfonyl)-2-propenenitrile 23-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 133-140 23415678-11 2013 Diethylstilbestrol and tetrabromobisphenol A inhibited CYP17 but not CYP19 activity, indicating different mechanisms for the inhibition of these enzymes. tetrabromobisphenol A 23-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-74 23450050-2 2013 Prostaglandin E2 (PGE2) stimulates aromatase (P450arom) expression in endometrioma stromal cells (ESCs) and increases the production of estrogens. Dinoprostone 0-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-54 23450050-2 2013 Prostaglandin E2 (PGE2) stimulates aromatase (P450arom) expression in endometrioma stromal cells (ESCs) and increases the production of estrogens. Dinoprostone 18-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-54 23450050-8 2013 Dehydroepiandrosterone (DHEA) was added to the culture, and then the combined enzyme activity of HSD3B2, which converts DHEA to androstenedione, and P450arom, which converts androstenedione to estrone, was examined by measuring estrone concentration in the supernatants with a specific enzyme immunoassay. Dehydroepiandrosterone 0-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 149-157 23450050-8 2013 Dehydroepiandrosterone (DHEA) was added to the culture, and then the combined enzyme activity of HSD3B2, which converts DHEA to androstenedione, and P450arom, which converts androstenedione to estrone, was examined by measuring estrone concentration in the supernatants with a specific enzyme immunoassay. Dehydroepiandrosterone 24-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 149-157 23450050-8 2013 Dehydroepiandrosterone (DHEA) was added to the culture, and then the combined enzyme activity of HSD3B2, which converts DHEA to androstenedione, and P450arom, which converts androstenedione to estrone, was examined by measuring estrone concentration in the supernatants with a specific enzyme immunoassay. Androstenedione 174-189 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 149-157 23450050-12 2013 IL-4 had no effect on the expression of P450arom mRNA, whereas PGE2 increased the expression of P450arom mRNA. Dinoprostone 63-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-104 23001364-2 2013 In contrast, reaction of this boryl triflate with aryloxides (ArO(-)) in THF resulted in insertion of THF with ring opening in between the NHC-boryl electrophile and the aryloxide to give products dipp-Imd-BH(2)O(CH(2))(4)OAr. trifluoromethanesulfonate 36-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-65 23340254-2 2013 In breast cancer, aberrant expression of the CYP19A1 gene, which encodes the aromatase enzyme, contributes to increased intratumoral levels of estradiol. Estradiol 143-152 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-52 22990756-4 2013 RESULTS: In all, 42.4% of aromatase inhibitor-treated breast cancer patients were dissatisfied with their sex life in general, and 50.0% reported low sexual interest; this was significantly more common than in tamoxifen-treated patients and controls (P < 0.05). Tamoxifen 210-219 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-35 23281812-3 2013 By combining decisive structural features of CYP11B2 and CYP19 inhibitors into a common template, a series of pyridinylmethyl substituted 1,2,5,6-tetrahydro-pyrrolo[3,2,1-ij]quinolin-4-ones were designed and synthesized. pyridinylmethyl substituted 1,2,5,6-tetrahydro-pyrrolo[3,2,1-ij]quinolin-4-ones 110-189 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-62 23208609-7 2013 Expression of particular steroidogenic genes, including the one encoding for aromatase (CYP19A1), was significantly upregulated after incubation of H295R cells with 2-ITX, 4-ITX, and 2,4-diethyl-TX. 2-itx 165-170 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-95 23208609-7 2013 Expression of particular steroidogenic genes, including the one encoding for aromatase (CYP19A1), was significantly upregulated after incubation of H295R cells with 2-ITX, 4-ITX, and 2,4-diethyl-TX. 4-itx 172-177 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-95 23208609-7 2013 Expression of particular steroidogenic genes, including the one encoding for aromatase (CYP19A1), was significantly upregulated after incubation of H295R cells with 2-ITX, 4-ITX, and 2,4-diethyl-TX. 2,4-diethyl-tx 183-197 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-95 22414444-3 2013 Specifically, aromatase; the enzyme responsible for the conversion of testosterone to estradiol, is upregulated in the avian and mammalian brain following disruption of the neuropil by multiple forms of perturbation including mechanical injury, ischemia and excitotoxicity. Testosterone 70-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 22414444-3 2013 Specifically, aromatase; the enzyme responsible for the conversion of testosterone to estradiol, is upregulated in the avian and mammalian brain following disruption of the neuropil by multiple forms of perturbation including mechanical injury, ischemia and excitotoxicity. Estradiol 86-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 23001364-2 2013 In contrast, reaction of this boryl triflate with aryloxides (ArO(-)) in THF resulted in insertion of THF with ring opening in between the NHC-boryl electrophile and the aryloxide to give products dipp-Imd-BH(2)O(CH(2))(4)OAr. aryloxides 50-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-65 23001364-2 2013 In contrast, reaction of this boryl triflate with aryloxides (ArO(-)) in THF resulted in insertion of THF with ring opening in between the NHC-boryl electrophile and the aryloxide to give products dipp-Imd-BH(2)O(CH(2))(4)OAr. tetrahydrofuran 73-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-65 23001364-2 2013 In contrast, reaction of this boryl triflate with aryloxides (ArO(-)) in THF resulted in insertion of THF with ring opening in between the NHC-boryl electrophile and the aryloxide to give products dipp-Imd-BH(2)O(CH(2))(4)OAr. tetrahydrofuran 102-105 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-65 23001364-2 2013 In contrast, reaction of this boryl triflate with aryloxides (ArO(-)) in THF resulted in insertion of THF with ring opening in between the NHC-boryl electrophile and the aryloxide to give products dipp-Imd-BH(2)O(CH(2))(4)OAr. aryloxide 50-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-65 23001364-2 2013 In contrast, reaction of this boryl triflate with aryloxides (ArO(-)) in THF resulted in insertion of THF with ring opening in between the NHC-boryl electrophile and the aryloxide to give products dipp-Imd-BH(2)O(CH(2))(4)OAr. dipp-imd-bh 197-208 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-65 23085504-0 2013 Vitamin D analog EB1089 inhibits aromatase expression by dissociation of comodulator WSTF from the CYP19A1 promoter-a new regulatory pathway for aromatase. Vitamin D 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-106 23748068-9 2013 In the genetic analysis, homozygous R375H guanine-adenine (G-A) mutation was detected in the CYP19A1 gene, and a diagnosis of AD was reached. guanine-adenine 42-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-100 23586996-1 2013 Aromatase (CYP19) is involved in steroidogenesis, catalyzing the conversion of androgens into estrogens through a unique reaction that causes the aromatization of the A ring of the steroid. Steroids 33-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 23586996-1 2013 Aromatase (CYP19) is involved in steroidogenesis, catalyzing the conversion of androgens into estrogens through a unique reaction that causes the aromatization of the A ring of the steroid. Steroids 33-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-16 23129173-0 2013 Plasma estrone sulfate concentrations and genetic variation at the CYP19A1 locus in postmenopausal women with early breast cancer treated with letrozole. Letrozole 143-152 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-74 23129173-2 2013 We assessed the association between plasma estrone sulfate (ES), letrozole treatment, and four SNPs of CYP19A1 gene (rs10046 C>T, rs4646 G>T, rs749292 C>T, rs727479 T>G) which seem to be related to circulating estrogen levels. estrone sulfate 43-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 103-110 23129173-2 2013 We assessed the association between plasma estrone sulfate (ES), letrozole treatment, and four SNPs of CYP19A1 gene (rs10046 C>T, rs4646 G>T, rs749292 C>T, rs727479 T>G) which seem to be related to circulating estrogen levels. estrone sulfate 60-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 103-110 23129173-3 2013 Patients were enrolled into a prospective, Italian multi-center clinical trial (Gruppo Italiano Mammella, GIM-5) testing the association of CYP19A1 SNPs with the efficacy of letrozole adjuvant therapy, in postmenopausal early BC patients. Letrozole 174-183 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 140-147 23748068-9 2013 In the genetic analysis, homozygous R375H guanine-adenine (G-A) mutation was detected in the CYP19A1 gene, and a diagnosis of AD was reached. g-a 59-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-100 23133418-0 2012 Potential contribution of aromatase inhibition to the effects of nicotine and related compounds on the brain. Nicotine 65-73 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-35 23142464-8 2013 Gene expression analysis of StAR, Bzrp, CYP11A, CYP17, CYP21 and CYP19 mRNA showed a decrease in Bzrp mRNA levels for 8:2 monoPAPS and 8:2 FTOH indicating interference with cholesterol transport to the inner mitochondria. Cholesterol 173-184 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-70 23142464-10 2013 In accordance with these data CYP19 gene expression increased with 8:2 diPAPS, 8:2 monoPAPS and 8:2 FTOH exposures indicating that this is a contributing factor to the decreased androgen and the increased estrogen levels. dipaps 71-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-35 23142464-10 2013 In accordance with these data CYP19 gene expression increased with 8:2 diPAPS, 8:2 monoPAPS and 8:2 FTOH exposures indicating that this is a contributing factor to the decreased androgen and the increased estrogen levels. ftoh 100-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-35 22963808-11 2012 The patient"s menorrhagia, which had previously been resistant to progesterone IUD therapy, resolved with the aromatase inhibitor. Progesterone 66-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 110-119 23342035-4 2013 The rs10046 polymorphism on the CYP19 gene has been related to levels of circulating estradiol and to the estradiol/testosterone ratio. Estradiol 85-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-37 23342035-4 2013 The rs10046 polymorphism on the CYP19 gene has been related to levels of circulating estradiol and to the estradiol/testosterone ratio. Estradiol 106-115 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-37 23342035-4 2013 The rs10046 polymorphism on the CYP19 gene has been related to levels of circulating estradiol and to the estradiol/testosterone ratio. Testosterone 116-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-37 22951915-10 2012 In addition, mRNA transcripts of factors involved in steroid metabolism, such as CYP11A1, CYP1B1, CYP19A1 and CYP2B7P1, were deregulated in PCOS CCs, and this could explain the abnormal steroidogenesis observed in these women. Steroids 53-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-105 23001776-1 2012 STUDY QUESTION: Does synergism between AR(CAG)(n) and CYP19(TTTA)(n) polymorphisms influence the quality of sperm? ttta 60-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 54-59 23116507-2 2012 The CYP19A1 (formerly CYP19) gene encodes the cytochrome P450 enzyme aromatase (estrogen synthetase), which converts testosterone to estrogen. Testosterone 117-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-11 23116507-2 2012 The CYP19A1 (formerly CYP19) gene encodes the cytochrome P450 enzyme aromatase (estrogen synthetase), which converts testosterone to estrogen. Testosterone 117-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-9 23116507-2 2012 The CYP19A1 (formerly CYP19) gene encodes the cytochrome P450 enzyme aromatase (estrogen synthetase), which converts testosterone to estrogen. Testosterone 117-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 80-99 23133418-7 2012 Several lines of evidence suggest that many of the reported sex differences related to cigarette smoking may stem from the inhibitory effects of nicotine and other tobacco alkaloids on estrogen synthesis via the enzyme aromatase (cyp19a gene product). Nicotine 145-153 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 219-228 23133418-7 2012 Several lines of evidence suggest that many of the reported sex differences related to cigarette smoking may stem from the inhibitory effects of nicotine and other tobacco alkaloids on estrogen synthesis via the enzyme aromatase (cyp19a gene product). Alkaloids 172-181 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 219-228 23133418-9 2012 This review provides a summary of experimental evidence supporting brain aromatase as a potential mediator and/or modulator of nicotine actions in the brain, contributing to sex differences in smoking behavior. Nicotine 127-135 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-82 23253631-8 2012 RESULTS: In E2 untreated condition, 1,25(OH)2D3 reduced P450-aromatase synthesis and CYP19A1 gene expression in cultured cells. Calcitriol 36-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 85-92 23158130-4 2012 We assayed the concentrations of PGE2 in FF, the production of E2 and progesterone in FF and in culture medium, and the expression of steroidogenic acute regulatory protein (StAR) and CYP19A1 in GLCs with the intervention of PGE2. Dinoprostone 225-229 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 184-191 22929011-3 2012 Prostaglandin E2 (PGE2), a factor secreted by tumours, is known to stimulate CYP19A1 expression in human BAFs. Dinoprostone 0-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-84 23158130-10 2012 In contrast, the E2 and CYP19A1 are decreased in GLCs, which may delay the development of the follicles and cause an imbalance in the follicular steroid hormone levels. Steroids 145-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-31 22929011-3 2012 Prostaglandin E2 (PGE2), a factor secreted by tumours, is known to stimulate CYP19A1 expression in human BAFs. Dinoprostone 18-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-84 22878119-6 2012 Our results show that application of medroxyprogestrone acetate, dydrogesterone and dienogest significantly decreases HSD17B1 and CYP19A1 expression and significantly increases HSD17B2 expression. medroxyprogestrone acetate 37-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-137 22878119-6 2012 Our results show that application of medroxyprogestrone acetate, dydrogesterone and dienogest significantly decreases HSD17B1 and CYP19A1 expression and significantly increases HSD17B2 expression. Dydrogesterone 65-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-137 22634420-3 2012 The expression levels of aromatase (ARO) and steroidogenic acute regulatory protein (StAR) were positively correlated with estrogens and testosterone concentrations, respectively. Testosterone 137-149 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-39 23009063-7 2012 The significantly decreased Lewis acidity of the Zr metal center in anions 4a,b (versus neutral analogues) due to the anionic charge and a likely substantial electronic pi donation of the four Zr-O(ArO) oxygens to the Zr metal center may rationalize the moderate polymerization activity. Zirconium 49-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 198-201 23009063-7 2012 The significantly decreased Lewis acidity of the Zr metal center in anions 4a,b (versus neutral analogues) due to the anionic charge and a likely substantial electronic pi donation of the four Zr-O(ArO) oxygens to the Zr metal center may rationalize the moderate polymerization activity. Oxygen 203-210 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 198-201 23009063-7 2012 The significantly decreased Lewis acidity of the Zr metal center in anions 4a,b (versus neutral analogues) due to the anionic charge and a likely substantial electronic pi donation of the four Zr-O(ArO) oxygens to the Zr metal center may rationalize the moderate polymerization activity. Zirconium 193-195 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 198-201 22634420-7 2012 Our results suggest that the status of ARO and StAR contribute to estrogen synthesis in situ, especially for the regulation of elastic fiber formation, and to testosterone synthesis, which may be associated with hair growth, respectively. Testosterone 159-171 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-42 22787115-0 2012 Pioglitazone, a PPARgamma agonist, suppresses CYP19 transcription: evidence for involvement of 15-hydroxyprostaglandin dehydrogenase and BRCA1. Pioglitazone 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-51 22324859-0 2012 Higher than expected estradiol levels in aromatase inhibitor-treated, postmenopausal breast cancer patients. Estradiol 21-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-50 22324859-3 2012 However, in clinical practice, no precise estimation has been performed of circulating estrogens and aromatase index in postmenopausal breast cancer patients on long-lasting AI or tamoxifen treatment. Tamoxifen 180-189 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-110 22787115-3 2012 Prostaglandin E(2) (PGE(2)) stimulates the cAMP protein kinase A (PKA) pathway leading to increased CYP19 transcription and elevated aromatase activity in inflamed white adipose tissue. Cyclic AMP 43-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-105 22787115-3 2012 Prostaglandin E(2) (PGE(2)) stimulates the cAMP protein kinase A (PKA) pathway leading to increased CYP19 transcription and elevated aromatase activity in inflamed white adipose tissue. Dinoprostone 0-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-105 22787115-3 2012 Prostaglandin E(2) (PGE(2)) stimulates the cAMP protein kinase A (PKA) pathway leading to increased CYP19 transcription and elevated aromatase activity in inflamed white adipose tissue. Dinoprostone 20-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-105 22787115-7 2012 Pioglitazone also inhibited cAMP PKA signaling leading to reduced interaction between phosphorylated cAMP responsive element-binding protein, p300, and CYP19 I.3/II promoter. Pioglitazone 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 152-157 22814869-1 2012 New titanium complexes of general formula [(ArO)(n)Ti(Oi-Pr)((4-n))] were synthesized and used as pre-catalysts for the selective dimerization of ethylene to 1-butene. Titanium 4-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-47 22787115-7 2012 Pioglitazone also inhibited cAMP PKA signaling leading to reduced interaction between phosphorylated cAMP responsive element-binding protein, p300, and CYP19 I.3/II promoter. Cyclic AMP 28-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 152-157 22787115-7 2012 Pioglitazone also inhibited cAMP PKA signaling leading to reduced interaction between phosphorylated cAMP responsive element-binding protein, p300, and CYP19 I.3/II promoter. Cyclic AMP 101-105 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 152-157 22787115-8 2012 BRCA1, a repressor of CYP19 transcription, was induced by pioglitazone. Pioglitazone 58-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 22-27 22814869-1 2012 New titanium complexes of general formula [(ArO)(n)Ti(Oi-Pr)((4-n))] were synthesized and used as pre-catalysts for the selective dimerization of ethylene to 1-butene. naltrindole 49-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-47 22814869-1 2012 New titanium complexes of general formula [(ArO)(n)Ti(Oi-Pr)((4-n))] were synthesized and used as pre-catalysts for the selective dimerization of ethylene to 1-butene. ethylene 146-154 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-47 22814869-1 2012 New titanium complexes of general formula [(ArO)(n)Ti(Oi-Pr)((4-n))] were synthesized and used as pre-catalysts for the selective dimerization of ethylene to 1-butene. 1-butene 158-166 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-47 22814869-4 2012 For alkyl-substituted phenols, it was demonstrated that large steric hindrance at both ortho and ortho" positions selectively yielded the mono-substituted complexes [(ArO)Ti(Oi-Pr)(3)]. alkyl-substituted phenols 4-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 167-170 22773874-0 2012 Oxidation of dihydrotestosterone by human cytochromes P450 19A1 and 3A4. Dihydrotestosterone 13-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-71 22407818-5 2012 Among the elevated miRNAs following letrozole treatment, computational analysis identified let-7f, a tumour-suppressor miRNA which targeted the aromatase gene (CYP19A1) expression. Letrozole 36-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 144-153 22843875-5 2012 Aromatase immunoreactivity tended to be positively associated with the intratumoral concentration of estrogens (n=53), and in particular, the concentration of estradiol was significantly higher (p=0.02) in aromatase-positive cases in men. Estradiol 159-168 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 22843875-5 2012 Aromatase immunoreactivity tended to be positively associated with the intratumoral concentration of estrogens (n=53), and in particular, the concentration of estradiol was significantly higher (p=0.02) in aromatase-positive cases in men. Estradiol 159-168 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 206-215 22659248-10 2012 CONCLUSIONS: The LHR gene is expressed in GC of human antral follicles throughout the follicular phase and is significantly associated with expression of the CYP19a1 gene and with the corresponding FF concentrations of estradiol and progesterone. Progesterone 233-245 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 158-165 22712891-5 2012 Reversible (Anastrozole) and irreversible (Exemestane) Aromatase inhibitors are selected and performed molecular docking studies of each drug against each variant to see the binding affinity impact on protein variant and drugs. exemestane 43-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 55-64 22659248-10 2012 CONCLUSIONS: The LHR gene is expressed in GC of human antral follicles throughout the follicular phase and is significantly associated with expression of the CYP19a1 gene and with the corresponding FF concentrations of estradiol and progesterone. Estradiol 219-228 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 158-165 22407818-5 2012 Among the elevated miRNAs following letrozole treatment, computational analysis identified let-7f, a tumour-suppressor miRNA which targeted the aromatase gene (CYP19A1) expression. Letrozole 36-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 160-167 21683476-6 2012 Additionally, short-term administration of estradiol is associated with decreased expression of CYP17A1, STS, SULT2B1, and AROMATASE, consistent with a downregulation not only of estrogen synthesis from circulating DHEA, but also of de novo DHEA synthesis within the hippocampus. Estradiol 43-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 123-132 21814747-0 2012 The tamoxifen metabolite norendoxifen is a potent and selective inhibitor of aromatase (CYP19) and a potential lead compound for novel therapeutic agents. Tamoxifen 4-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-93 24367197-0 2012 Aromatase inhibitor-associated bone loss and its management with bisphosphonates in patients with breast cancer. Diphosphonates 65-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 22699893-1 2012 Inhibitors of aromatase, the final enzyme of estradiol synthesis, are suspected of inducing memory deficits in women. Estradiol 45-54 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 22025366-6 2012 Genes involved downstream in steroid hormone generation, including CYP17A1 and CYP19A1 were also induced. Steroids 29-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-86 22627104-2 2012 With the aim of improving disease-free survival (DFS), this phase 3 study (CAO/ARO/AIO-04) integrated oxaliplatin into standard treatment. Oxaliplatin 102-113 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-82 22418701-1 2012 Single nucleotide polymorphisms (SNPs) in the gene encoding aromatase (CYP19A1) have been associated with differential benefit from letrozole treatment in metastatic breast cancer (mBC) patients, but validation is lacking. Letrozole 132-141 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-78 22418701-6 2012 Patients with >7 TTTA repeats on either allele of CYP19A1 intron 4 had a lower risk of failure than those with a smaller repeat size [HR = 0.84 per >7 TTTA repeats (0.7-0.99); P = 0.04]. ttta 20-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-60 22434523-0 2012 Vitamin D threshold to prevent aromatase inhibitor-related bone loss: the B-ABLE prospective cohort study. Vitamin D 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 22300440-6 2012 Also, CYP19A1 arginine allele in homozygosity or heterozygosity (TC/CC) was associated with a significant increased risk for breast cancer when associated to GSTM1 null genotype (OR=6.158; 95% CI=2.676-14.171; p<0.001) and GSTT1 null genotype (OR=4.870; 95% CI=2.216-10.700; p<0.001). Arginine 14-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 6-13 22336992-3 2012 We investigated maternal functional polymorphisms of CYP17, CYP19, and CYP1B1, which control three major enzymatic steps in sex steroid biosynthesis and metabolism, in relation to childhood behaviors. Steroids 128-135 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-65 21814747-0 2012 The tamoxifen metabolite norendoxifen is a potent and selective inhibitor of aromatase (CYP19) and a potential lead compound for novel therapeutic agents. N,N-didesmethyl-4-hydroxytamoxifen 25-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-93 21814747-3 2012 The ability of ten tamoxifen metabolites to inhibit recombinant aromatase (CYP19) was tested using microsomal incubations. Tamoxifen 19-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-80 22755322-10 2012 We present a case of recurrent and treatment refractory GCT in a postmenopausal patient, managed by aromatase inhibitor Anastrozole with reasonable efficacy. Anastrozole 120-131 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-109 22350385-4 2012 Aromatase (cytochrome P450 19 enzyme, CYP19) inhibitory activity was measured by a tritiated water release assay and by direct measurement of bio-transformed steroids using the tritium labeled substrate (3)H-androst-4-ene-3,17-dione. Water 93-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-43 22350385-4 2012 Aromatase (cytochrome P450 19 enzyme, CYP19) inhibitory activity was measured by a tritiated water release assay and by direct measurement of bio-transformed steroids using the tritium labeled substrate (3)H-androst-4-ene-3,17-dione. Steroids 158-166 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-43 22350385-4 2012 Aromatase (cytochrome P450 19 enzyme, CYP19) inhibitory activity was measured by a tritiated water release assay and by direct measurement of bio-transformed steroids using the tritium labeled substrate (3)H-androst-4-ene-3,17-dione. Tritium 177-184 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-43 22350385-4 2012 Aromatase (cytochrome P450 19 enzyme, CYP19) inhibitory activity was measured by a tritiated water release assay and by direct measurement of bio-transformed steroids using the tritium labeled substrate (3)H-androst-4-ene-3,17-dione. Androstenedione 207-232 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-43 22301800-3 2012 We report an LCT in a 29-year-old male with elevated plasma oestradiol caused by enhanced CYP19 transcription. Estradiol 60-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-95 22432862-3 2012 Especially important in aromatic polyketide biosynthesis is the regiospecific cyclization of a linear, preassembled polyketide chain catalyzed by aromatase/cyclase (ARO/CYC), which serves as a key control point in aromatic ring formation. Polyketides 33-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 165-168 22245270-14 2012 In conclusion, result of the present study indicated that that in the absence of consensus CRE (cAMP response element); GATA-4 could be a downstream effector of cAMP/PKA pathway in regulation of CYP19 gene during folliculogenesis and luteinization. Cyclic AMP 96-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 195-200 22245270-14 2012 In conclusion, result of the present study indicated that that in the absence of consensus CRE (cAMP response element); GATA-4 could be a downstream effector of cAMP/PKA pathway in regulation of CYP19 gene during folliculogenesis and luteinization. Cyclic AMP 161-165 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 195-200 22361454-0 2012 Steroidal pyrazolines evaluated as aromatase and quinone reductase-2 inhibitors for chemoprevention of cancer. pyrazolines 10-21 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 35-44 22361454-1 2012 The aromatase and quinone reductase-2 inhibition of synthesized heterocyclic pyrazole derivatives fused with steroidal structure for chemoprevention of cancer is reported herein. pyrazole 77-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-13 22361454-3 2012 The aromatase inhibitory activities of these compounds were much more potent than the lead compound resveratrol, which has an IC(50) of 80 muM. Resveratrol 100-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-13 22432862-3 2012 Especially important in aromatic polyketide biosynthesis is the regiospecific cyclization of a linear, preassembled polyketide chain catalyzed by aromatase/cyclase (ARO/CYC), which serves as a key control point in aromatic ring formation. Polyketides 116-126 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 165-168 22432862-6 2012 The WhiE ARO/CYC catalyzes the regiospecific C9-C14 and C7-C16 cyclization and aromatization of a 24-carbon polyketide chain. carbon polyketide 101-118 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-12 22386564-0 2012 Optimization of the aromatase inhibitory activities of pyridylthiazole analogues of resveratrol. pyridylthiazole 55-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-29 22327052-0 2012 Effects of bisphenol A on the expression of cytochrome P450 aromatase (CYP19) in human fetal osteoblastic and granulosa cell-like cell lines. bisphenol A 11-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-76 22327052-2 2012 CYP19 enzyme activity was suppressed in the presence of BPA in a dose-dependent fashion in both cell lines. bisphenol A 56-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-5 22327052-4 2012 These data and the previous finding that BPA induced the down-regulation of promoter I.1 activity within the human placental cell line suggest that there may be a conserved signaling pathway that down-regulates CYP19 expression in response to BPA in both cell lines. bisphenol A 41-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 211-216 22327052-4 2012 These data and the previous finding that BPA induced the down-regulation of promoter I.1 activity within the human placental cell line suggest that there may be a conserved signaling pathway that down-regulates CYP19 expression in response to BPA in both cell lines. bisphenol A 243-246 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 211-216 22386564-0 2012 Optimization of the aromatase inhibitory activities of pyridylthiazole analogues of resveratrol. Resveratrol 84-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-29 22386564-2 2012 The moderate and non-selective aromatase inhibitory activity of resveratrol (1) was improved about 100-fold by replacement of the ethylenic bridge with a thiadiazole and the phenyl rings with pyridines (e.g., compound 3). Resveratrol 64-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 22386564-2 2012 The moderate and non-selective aromatase inhibitory activity of resveratrol (1) was improved about 100-fold by replacement of the ethylenic bridge with a thiadiazole and the phenyl rings with pyridines (e.g., compound 3). Thiadiazoles 154-165 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 22386564-2 2012 The moderate and non-selective aromatase inhibitory activity of resveratrol (1) was improved about 100-fold by replacement of the ethylenic bridge with a thiadiazole and the phenyl rings with pyridines (e.g., compound 3). Pyridines 192-201 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 22237979-8 2012 Treatment of BAFs and CAFs with MLT resulted in significant suppression of CYP19A1 transcription and aromatase activity at pharmacological, physiological and sub-physiological concentrations. bafs 13-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-82 22386564-3 2012 The aromatase inhibitory activity was enhanced over 6000-fold by using a 1,3-thiazole as the central ring and modifying the substituents on the "A" ring to target the Met374 residue of aromatase. Thiazoles 73-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-13 21824047-2 2012 This study aimed to correlate polymorphisms of genes involved in the biosynthesis and metabolism of steroids and insulin action (CYP17A1, CYP19A1, AR, ESR1, ESR2, INSR, IGF2 and PAI1) with clinical and biochemical parameters of PCOS. Steroids 100-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-145 22237979-10 2012 Stimulation of CYP19A1 PII-mRNA and aromatase activity by prostaglandin E(2) (PGE(2)) were significantly attenuated by physiological doses of MLT. Dinoprostone 58-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-22 22237979-10 2012 Stimulation of CYP19A1 PII-mRNA and aromatase activity by prostaglandin E(2) (PGE(2)) were significantly attenuated by physiological doses of MLT. Prostaglandins E 78-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-22 22576212-5 2012 We provide evidence that cyclooxygenase (COX)-2-derived PGE(2) stimulates the cyclic AMP (cAMP) PKA signal transduction pathway that activates CYP19 transcription, resulting in increased aromatase expression and elevated progesterone receptor levels in breast tissues from overweight and obese women. Prostaglandins E 56-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 145-150 22576212-5 2012 We provide evidence that cyclooxygenase (COX)-2-derived PGE(2) stimulates the cyclic AMP (cAMP) PKA signal transduction pathway that activates CYP19 transcription, resulting in increased aromatase expression and elevated progesterone receptor levels in breast tissues from overweight and obese women. Cyclic AMP 78-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 145-150 22576212-5 2012 We provide evidence that cyclooxygenase (COX)-2-derived PGE(2) stimulates the cyclic AMP (cAMP) PKA signal transduction pathway that activates CYP19 transcription, resulting in increased aromatase expression and elevated progesterone receptor levels in breast tissues from overweight and obese women. Cyclic AMP 90-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 145-150 22386365-3 2012 Therefore, we studied the effect of sodium butyrate (NaBu) on the CYP19A1 transcript and protein levels and on the conversion of A to E1 in HT29, DLD-1 and LoVo CRC cells. sethoxydim 53-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-73 22386365-4 2012 We found that NaBu significantly downregulated CYP19A1 transcript and protein levels, a phenomenon that was associated with reduced conversion of A to E1 in HT29, DLD-1 and LoVo cells. sethoxydim 14-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-54 22132829-1 2012 The aromatase inhibitor anastrazole proved effective in the treatment of endometrial hyperplasia and postmenopausal bleeding in an obese 65-year-old woman with high operative risk. Anastrozole 24-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-13 22014381-0 2012 Immediate Administration of Zoledronic Acid Reduces Aromatase Inhibitor-Associated Bone Loss in Postmenopausal Women With Early Breast Cancer: 12-month analysis of the E-ZO-FAST trial. Zoledronic Acid 28-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-61 22132829-6 2012 This case demonstrates that long-term aromatase inhibitor treatment can result in a refractory status of the endometrium and the estradiol produced in the adipose tissue does not exert a proliferative effect. Estradiol 129-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-47 22014381-2 2012 As with other aromatase inhibitors (AIs), long-term letrozole administration is associated with decreased bone mineral density (BMD) and increased fracture risk. Letrozole 52-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 22128327-0 2012 Methylseleninic acid is a novel suppressor of aromatase expression. methylselenic acid 0-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-55 22128327-5 2012 In obese postmenopausal women, increased peripheral aromatase is primarily attributed to the activity of the glucocorticoid-stimulated promoter, PI.4, and the cAMP-stimulated promoter, PII. Cyclic AMP 159-163 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-61 22128327-6 2012 In the present study, we show that methylseleninic acid (MSA), a second-generation selenium compound, can effectively suppress aromatase activation by dexamethasone, a synthetic glucocorticoid, and forskolin, a specific activator of adenylate cyclase. methylselenic acid 35-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-136 22128327-6 2012 In the present study, we show that methylseleninic acid (MSA), a second-generation selenium compound, can effectively suppress aromatase activation by dexamethasone, a synthetic glucocorticoid, and forskolin, a specific activator of adenylate cyclase. methylselenic acid 57-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-136 22128327-6 2012 In the present study, we show that methylseleninic acid (MSA), a second-generation selenium compound, can effectively suppress aromatase activation by dexamethasone, a synthetic glucocorticoid, and forskolin, a specific activator of adenylate cyclase. Selenium 83-91 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-136 22128327-6 2012 In the present study, we show that methylseleninic acid (MSA), a second-generation selenium compound, can effectively suppress aromatase activation by dexamethasone, a synthetic glucocorticoid, and forskolin, a specific activator of adenylate cyclase. Dexamethasone 151-164 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-136 22128327-6 2012 In the present study, we show that methylseleninic acid (MSA), a second-generation selenium compound, can effectively suppress aromatase activation by dexamethasone, a synthetic glucocorticoid, and forskolin, a specific activator of adenylate cyclase. Colforsin 198-207 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-136 21918127-6 2012 In experiment 1, partitioning of P450c17, P450arom, and 3betaHSD markedly decreased estrone synthesis in comparison to cells coexpressing enzymes in different combinations. Estrone 84-91 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-50 23110082-1 2012 BACKGROUND: The objective of the study was to investigate the role of genes (HSD3B1, CYP17A1, CYP19A1, HSD17B2, HSD17B1) involved in the steroid hormone biosynthesis pathway and progesterone receptor (PGR) in the etiology of gastric cancer in a population-based two-phase genetic association study. Steroids 137-152 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-101 21918127-2 2012 Human cytochromes 17alpha-hydroxylase/17,20-lyase (P450c17) and aromatase (P450arom), always coexpressed with their redox partner NADPH-P450 oxidoreductase (CPR) and 3beta-hydroxysteroid dehydrogenase/delta5-4 isomerase (3betaHSD; types 1 or 2), were compartmentally expressed in different cell populations or coexpressed together with pregnenolone (100 nM) as substrate. Pregnenolone 336-348 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-83 22079757-0 2012 Discovery of a novel class of aldol-derived 1,2,3-triazoles: potent and selective inhibitors of human cytochrome P450 19A1 (aromatase). 1,2,3-triazoles 44-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 102-122 23110082-5 2012 RESULTS: Of the 108 SNPs in steroid hormone biosynthesis pathway related genes and PGR analyzed in the discovery phase, 23 SNPs in PGR in the recessive model and 10 SNPs in CYP19A1 in the recessive or additive models were significantly associated with increased gastric cancer risk (p<0.05). Steroids 28-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 173-180 22957105-4 2012 Treatments with salmon pituitary homogenates (female) or human chorionic gonadotrophin (male), known to turn on steroid production in immature eels, strongly stimulated cyp19a1 messenger and protein expression in radial glial cells and pituitary cells. Steroids 112-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-176 23029074-15 2012 CONCLUSIONS/SIGNIFICANCE: Puerarin suppresses proliferation of ESCs induced by E(2)-BSA partly via impeding a rapid, non-genomic, membrane-initiated ERK pathway, and down-regulation of Cyclin D1, Cox-2 and Cyp19 are involved in the process. puerarin 26-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 206-211 21952832-3 2011 In the present study 33 tag polymorphisms were genotyped across the CYP19A1 gene in a population of 1,185 Caucasian postmenopausal women to test the association between sequence variations, total DXA hip aBMD, and circulating 17beta-estradiol levels. Estradiol 226-242 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-75 22493800-4 2012 This review is focused on the natural compounds that exhibit the aromatase inhibition, which include flavonoids, xanthones, coumarins, and sesquiterpenes. Flavonoids 101-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 22493800-4 2012 This review is focused on the natural compounds that exhibit the aromatase inhibition, which include flavonoids, xanthones, coumarins, and sesquiterpenes. Xanthones 113-122 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 22493800-4 2012 This review is focused on the natural compounds that exhibit the aromatase inhibition, which include flavonoids, xanthones, coumarins, and sesquiterpenes. Coumarins 124-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 22493800-4 2012 This review is focused on the natural compounds that exhibit the aromatase inhibition, which include flavonoids, xanthones, coumarins, and sesquiterpenes. Sesquiterpenes 139-153 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 22119029-6 2012 RESULTS: Three novel findings are elucidated: first, that up-regulation of CYP19 expression is associated with exposure to PCB groupings containing dioxin-like, potentially anti-estrogenic, immunotoxic congeners, including PCB IUPAC #74, #105, #118, #138, #156, #157, #158, #167, and #170 from this cohort. Polychlorinated Biphenyls 123-126 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-80 22119029-6 2012 RESULTS: Three novel findings are elucidated: first, that up-regulation of CYP19 expression is associated with exposure to PCB groupings containing dioxin-like, potentially anti-estrogenic, immunotoxic congeners, including PCB IUPAC #74, #105, #118, #138, #156, #157, #158, #167, and #170 from this cohort. Dioxins 148-154 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-80 22119029-6 2012 RESULTS: Three novel findings are elucidated: first, that up-regulation of CYP19 expression is associated with exposure to PCB groupings containing dioxin-like, potentially anti-estrogenic, immunotoxic congeners, including PCB IUPAC #74, #105, #118, #138, #156, #157, #158, #167, and #170 from this cohort. Polychlorinated Biphenyls 223-226 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-80 22087767-4 2011 The rate laws for oxidation of the four coupling products by [IrCl(6)](2-) have the same form as those for the oxidation of phenol itself: -d[Ir(IV)]/dt = {(k(ArOH) + k(ArO(-))K(a)/[H(+)])/(1 + K(a)/[H(+)])}[ArOH](tot)[Ir(IV)]. Phenol 124-130 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 159-162 21953762-5 2011 Naringenin was able to inhibit CYP19, CYP2C9, and CYP2C19 with IC50 values below 5 muM. naringenin 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-36 22000946-8 2011 Also, the present study showed that all the tested steroid derivatives exhibited significant depletion with various intensities in gene expression of breast cancer related genes (VEGF, CYP19 and hAP-2gamma). Steroids 51-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 185-190 21442439-0 2011 Single nucleotide polymorphisms of CYP19A1 predict clinical outcomes and adverse events associated with letrozole in patients with metastatic breast cancer. Letrozole 104-113 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 35-42 21442439-2 2011 We evaluated the efficacy of the aromatase inhibitor letrozole in patients with metastatic breast cancer (MBC) as related to DNA polymorphisms of CYP19A1. Letrozole 53-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 146-153 21442439-10 2011 CONCLUSION: The genetic variations of CYP19A1 were significantly associated with clinical efficacy, suggesting potential predictive markers for letrozole treatment in patients with metastatic breast cancer. Letrozole 144-153 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-45 22001251-0 2011 Halowax 1051 affects steroidogenesis, 17beta-hydroxysteroid dehydrogenase (17beta-HSD) and cytochrome P450arom (CYP19) activity, and protein expression in porcine ovarian follicles. halowax 1051 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-110 22001251-0 2011 Halowax 1051 affects steroidogenesis, 17beta-hydroxysteroid dehydrogenase (17beta-HSD) and cytochrome P450arom (CYP19) activity, and protein expression in porcine ovarian follicles. halowax 1051 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 112-117 22001251-2 2011 17beta-HSD and CYP19 activity were determined indirectly by measuring the conversion of androstenedione (A4) to T and T to E2, respectively. Androstenedione 88-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-20 22001251-3 2011 Additionally, CYP19 activity by dibenzylfluorescein assay. dibenzylfluorescein 32-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-19 21953762-7 2011 Whereas (S)-naringenin was 2-fold more potent as an inhibitor of CYP19 and CYP2C19 than (R)-naringenin, (R)-naringenin was 2-fold more potent for CYP2C9 and CYP3A. naringenin 8-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-70 21871953-0 2011 Congener-specific action of PBDEs on steroid secretion, CYP17, 17beta-HSD and CYP19 activity and protein expression in porcine ovarian follicles. Halogenated Diphenyl Ethers 28-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-83 21620560-5 2011 Testosterone, the primary circulating androgen in men, can also be metabolized via CYP19/aromatase into the potent estrogen, estradiol-17beta. Testosterone 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-88 21620560-5 2011 Testosterone, the primary circulating androgen in men, can also be metabolized via CYP19/aromatase into the potent estrogen, estradiol-17beta. Estradiol 125-141 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-88 21514383-7 2011 Modeling of the amphioxus aromatase substrate binding site and simulated docking of androstenedione in comparison to the human aromatase shows that the substrate binding site is conserved and predicts that androstenedione could be a substrate for amphioxus CYP19. Androstenedione 84-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 257-262 21514383-7 2011 Modeling of the amphioxus aromatase substrate binding site and simulated docking of androstenedione in comparison to the human aromatase shows that the substrate binding site is conserved and predicts that androstenedione could be a substrate for amphioxus CYP19. Androstenedione 206-221 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 257-262 21858332-0 2011 Direct spectroscopic evidence for binding of anastrozole to the iron heme of human aromatase. Anastrozole 45-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-92 21956398-1 2011 The design and the synthesis of a new family of hydroxy-4-thiaflavanes, in which the reactive phenolic OH is ortho to the sulfur atom of the benzofused oxathiin ring, allowed to prepare antioxidants that show rate constants for the reaction with peroxyl radicals (k(inh)), and bond dissociation energies (BDE), of the ArO-H group identical to those of alpha-tocopherol, the main component of vitamin E and the most effective lipophilic antioxidant known in nature. hydroxy-4-thiaflavanes 48-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 318-321 21956398-1 2011 The design and the synthesis of a new family of hydroxy-4-thiaflavanes, in which the reactive phenolic OH is ortho to the sulfur atom of the benzofused oxathiin ring, allowed to prepare antioxidants that show rate constants for the reaction with peroxyl radicals (k(inh)), and bond dissociation energies (BDE), of the ArO-H group identical to those of alpha-tocopherol, the main component of vitamin E and the most effective lipophilic antioxidant known in nature. Sulfur 122-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 318-321 21956398-1 2011 The design and the synthesis of a new family of hydroxy-4-thiaflavanes, in which the reactive phenolic OH is ortho to the sulfur atom of the benzofused oxathiin ring, allowed to prepare antioxidants that show rate constants for the reaction with peroxyl radicals (k(inh)), and bond dissociation energies (BDE), of the ArO-H group identical to those of alpha-tocopherol, the main component of vitamin E and the most effective lipophilic antioxidant known in nature. Oxathiins 152-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 318-321 21956398-1 2011 The design and the synthesis of a new family of hydroxy-4-thiaflavanes, in which the reactive phenolic OH is ortho to the sulfur atom of the benzofused oxathiin ring, allowed to prepare antioxidants that show rate constants for the reaction with peroxyl radicals (k(inh)), and bond dissociation energies (BDE), of the ArO-H group identical to those of alpha-tocopherol, the main component of vitamin E and the most effective lipophilic antioxidant known in nature. perhydroxyl radical 246-262 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 318-321 21956398-1 2011 The design and the synthesis of a new family of hydroxy-4-thiaflavanes, in which the reactive phenolic OH is ortho to the sulfur atom of the benzofused oxathiin ring, allowed to prepare antioxidants that show rate constants for the reaction with peroxyl radicals (k(inh)), and bond dissociation energies (BDE), of the ArO-H group identical to those of alpha-tocopherol, the main component of vitamin E and the most effective lipophilic antioxidant known in nature. alpha-Tocopherol 352-368 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 318-321 21956398-1 2011 The design and the synthesis of a new family of hydroxy-4-thiaflavanes, in which the reactive phenolic OH is ortho to the sulfur atom of the benzofused oxathiin ring, allowed to prepare antioxidants that show rate constants for the reaction with peroxyl radicals (k(inh)), and bond dissociation energies (BDE), of the ArO-H group identical to those of alpha-tocopherol, the main component of vitamin E and the most effective lipophilic antioxidant known in nature. Vitamin E 392-401 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 318-321 21858332-4 2011 Using hyperfine sublevel correlation (HYSCORE) spectroscopy we provide the first experimental evidence of the binding of anastrozole to the iron heme of human aromatase. Anastrozole 121-132 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 159-168 21948762-0 2011 Association of serum sex steroid levels and bone mineral density with CYP17 and CYP19 gene polymorphisms in postmenopausal women in Turkey. Steroids 25-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 80-85 21741436-0 2011 Dietary flavones and flavonones display differential effects on aromatase (CYP19) transcription in the breast cancer cells MCF-7. Flavones 8-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-80 21741436-0 2011 Dietary flavones and flavonones display differential effects on aromatase (CYP19) transcription in the breast cancer cells MCF-7. flavonones 21-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-80 21870821-3 2011 The aromatase/cyclases (ARO/CYCs) are an important component of the type II polyketide synthase (PKS) and help fold the polyketide for regiospecific cyclizations of the first ring and/or aromatization, promoting two commonly observed first-ring cyclization patterns for the bacterial type II PKSs: C7-C12 and C9-C14. Polyketides 76-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-27 21870821-6 2011 In this work, we present the 2.4 A crystal structure of ZhuI, a C7-C12-specific first-ring ARO/CYC from the type II PKS pathway responsible for the production of the R1128 polyketides. Polyketides 172-183 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-94 21320576-1 2011 Estradiol biosynthesis is catalyzed by the enzyme aromatase, the product of the CYP19A1 gene. Estradiol 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 80-87 21948762-9 2011 Testosterone and DHEA-SO(4) levels in the CYP19 GG genotype were higher compared to those of the other genotypes in patients but not in controls. Testosterone 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-47 21948762-9 2011 Testosterone and DHEA-SO(4) levels in the CYP19 GG genotype were higher compared to those of the other genotypes in patients but not in controls. dhea-so 17-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-47 20354812-0 2011 Aromatase inhibitor anastrozole as a second-line hormonal treatment to a recurrent low-grade endometrial stromal sarcoma: a case report. Anastrozole 20-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21826442-2 2011 Although it has long been known that aromatase, the final enzyme in estrogen synthesis, is expressed in the hippocampus, a new paradigm emerged when it was shown that estradiol is actually synthesized de novo in this part of the brain. Estradiol 167-176 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-46 20354812-8 2011 Aromatase inhibitor anastrozole was then given at a daily dose of 1 mg with partial response (the tumor size decreased to 7 cm in diameter) for a duration of 9 months. Anastrozole 20-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21739987-2 2011 The measurement of the UV-vis absorption spectrum of alpha-tocopheroxyl (alpha-Toc( )) radical was performed by reacting aroxyl (ArO( )) radical with alpha-tocopherol (alpha-TocH) in acetonitrile solution including four kinds of alkali and alkaline earth metal salts (MX or MX(2)) (LiClO(4), LiI, NaClO(4), and Mg(ClO(4))(2)), using stopped-flow spectrophotometry. alpha-tocopheroxyl 53-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 21414724-0 2011 The TTTAn aromatase (CYP19A1) polymorphism is associated with compulsive craving of male patients during alcohol withdrawal. Alcohols 105-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-28 21757507-0 2011 Estrogen-related receptor gamma (ERRgamma) mediates oxygen-dependent induction of aromatase (CYP19) gene expression during human trophoblast differentiation. Oxygen 52-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-98 21757507-3 2011 Treatment with the selective ERRgamma agonist, DY131, or overexpression of ERRgamma, stimulated hCYP19 expression in syncytiotrophoblast. N'-((1E)-(4-(diethylamino)phenyl)methylene)-4-hydroxybenzohydrazide 47-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-102 21739987-5 2011 By reacting ArO( ) with alpha-TocH in acetonitrile, the absorption of ArO( ) disappeared rapidly, while that of alpha-Toc( ) appeared and then decreased gradually as a result of the bimolecular self-reaction of alpha-Toc( ) after passing through the maximum. acetonitrile 38-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 70-73 21739987-6 2011 The second-order rate constants (k(s)) obtained for the reaction of alpha-TocH with ArO( ) increased linearly with an increasing concentration of metal salts. Metals 146-151 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-87 21739987-7 2011 The results indicate that the hydrogen transfer reaction of alpha-TocH proceeds via an electron transfer intermediate from alpha-TocH to ArO( ) radicals followed by proton transfer. Hydrogen 30-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 137-140 21739987-2 2011 The measurement of the UV-vis absorption spectrum of alpha-tocopheroxyl (alpha-Toc( )) radical was performed by reacting aroxyl (ArO( )) radical with alpha-tocopherol (alpha-TocH) in acetonitrile solution including four kinds of alkali and alkaline earth metal salts (MX or MX(2)) (LiClO(4), LiI, NaClO(4), and Mg(ClO(4))(2)), using stopped-flow spectrophotometry. alpha-toc( )) radical 73-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 21739987-7 2011 The results indicate that the hydrogen transfer reaction of alpha-TocH proceeds via an electron transfer intermediate from alpha-TocH to ArO( ) radicals followed by proton transfer. alpha-Tocopherol 60-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 137-140 21739987-2 2011 The measurement of the UV-vis absorption spectrum of alpha-tocopheroxyl (alpha-Toc( )) radical was performed by reacting aroxyl (ArO( )) radical with alpha-tocopherol (alpha-TocH) in acetonitrile solution including four kinds of alkali and alkaline earth metal salts (MX or MX(2)) (LiClO(4), LiI, NaClO(4), and Mg(ClO(4))(2)), using stopped-flow spectrophotometry. alpha-Tocopherol 150-166 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 21739987-7 2011 The results indicate that the hydrogen transfer reaction of alpha-TocH proceeds via an electron transfer intermediate from alpha-TocH to ArO( ) radicals followed by proton transfer. alpha-Tocopherol 123-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 137-140 21739987-2 2011 The measurement of the UV-vis absorption spectrum of alpha-tocopheroxyl (alpha-Toc( )) radical was performed by reacting aroxyl (ArO( )) radical with alpha-tocopherol (alpha-TocH) in acetonitrile solution including four kinds of alkali and alkaline earth metal salts (MX or MX(2)) (LiClO(4), LiI, NaClO(4), and Mg(ClO(4))(2)), using stopped-flow spectrophotometry. alpha-Tocopherol 168-178 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 21739987-8 2011 Both the coordination of metal cations to the one-electron reduced anions of ArO( ) (ArO:(-)) and the coordination of counteranions to the one-electron oxidized cations of alpha-TocH (alpha-TocH( )(+)) may stabilize the intermediate, resulting in the acceleration of electron transfer. Metals 25-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-80 21739987-2 2011 The measurement of the UV-vis absorption spectrum of alpha-tocopheroxyl (alpha-Toc( )) radical was performed by reacting aroxyl (ArO( )) radical with alpha-tocopherol (alpha-TocH) in acetonitrile solution including four kinds of alkali and alkaline earth metal salts (MX or MX(2)) (LiClO(4), LiI, NaClO(4), and Mg(ClO(4))(2)), using stopped-flow spectrophotometry. acetonitrile 183-195 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 21739987-2 2011 The measurement of the UV-vis absorption spectrum of alpha-tocopheroxyl (alpha-Toc( )) radical was performed by reacting aroxyl (ArO( )) radical with alpha-tocopherol (alpha-TocH) in acetonitrile solution including four kinds of alkali and alkaline earth metal salts (MX or MX(2)) (LiClO(4), LiI, NaClO(4), and Mg(ClO(4))(2)), using stopped-flow spectrophotometry. alkali and 229-239 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 21739987-2 2011 The measurement of the UV-vis absorption spectrum of alpha-tocopheroxyl (alpha-Toc( )) radical was performed by reacting aroxyl (ArO( )) radical with alpha-tocopherol (alpha-TocH) in acetonitrile solution including four kinds of alkali and alkaline earth metal salts (MX or MX(2)) (LiClO(4), LiI, NaClO(4), and Mg(ClO(4))(2)), using stopped-flow spectrophotometry. alkaline earth metal salts 240-266 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 21739987-2 2011 The measurement of the UV-vis absorption spectrum of alpha-tocopheroxyl (alpha-Toc( )) radical was performed by reacting aroxyl (ArO( )) radical with alpha-tocopherol (alpha-TocH) in acetonitrile solution including four kinds of alkali and alkaline earth metal salts (MX or MX(2)) (LiClO(4), LiI, NaClO(4), and Mg(ClO(4))(2)), using stopped-flow spectrophotometry. 3-chloro-4-(dichloromethyl)-5-hydroxy-2(5H)-furanone 268-270 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 21739987-2 2011 The measurement of the UV-vis absorption spectrum of alpha-tocopheroxyl (alpha-Toc( )) radical was performed by reacting aroxyl (ArO( )) radical with alpha-tocopherol (alpha-TocH) in acetonitrile solution including four kinds of alkali and alkaline earth metal salts (MX or MX(2)) (LiClO(4), LiI, NaClO(4), and Mg(ClO(4))(2)), using stopped-flow spectrophotometry. 3-chloro-4-(dichloromethyl)-5-hydroxy-2(5H)-furanone 274-276 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 21739987-2 2011 The measurement of the UV-vis absorption spectrum of alpha-tocopheroxyl (alpha-Toc( )) radical was performed by reacting aroxyl (ArO( )) radical with alpha-tocopherol (alpha-TocH) in acetonitrile solution including four kinds of alkali and alkaline earth metal salts (MX or MX(2)) (LiClO(4), LiI, NaClO(4), and Mg(ClO(4))(2)), using stopped-flow spectrophotometry. liclo 282-287 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 21739987-2 2011 The measurement of the UV-vis absorption spectrum of alpha-tocopheroxyl (alpha-Toc( )) radical was performed by reacting aroxyl (ArO( )) radical with alpha-tocopherol (alpha-TocH) in acetonitrile solution including four kinds of alkali and alkaline earth metal salts (MX or MX(2)) (LiClO(4), LiI, NaClO(4), and Mg(ClO(4))(2)), using stopped-flow spectrophotometry. lii 292-295 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 21739987-2 2011 The measurement of the UV-vis absorption spectrum of alpha-tocopheroxyl (alpha-Toc( )) radical was performed by reacting aroxyl (ArO( )) radical with alpha-tocopherol (alpha-TocH) in acetonitrile solution including four kinds of alkali and alkaline earth metal salts (MX or MX(2)) (LiClO(4), LiI, NaClO(4), and Mg(ClO(4))(2)), using stopped-flow spectrophotometry. Sodium Hypochlorite 297-302 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 21739987-2 2011 The measurement of the UV-vis absorption spectrum of alpha-tocopheroxyl (alpha-Toc( )) radical was performed by reacting aroxyl (ArO( )) radical with alpha-tocopherol (alpha-TocH) in acetonitrile solution including four kinds of alkali and alkaline earth metal salts (MX or MX(2)) (LiClO(4), LiI, NaClO(4), and Mg(ClO(4))(2)), using stopped-flow spectrophotometry. Magnesium 311-313 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 21739987-2 2011 The measurement of the UV-vis absorption spectrum of alpha-tocopheroxyl (alpha-Toc( )) radical was performed by reacting aroxyl (ArO( )) radical with alpha-tocopherol (alpha-TocH) in acetonitrile solution including four kinds of alkali and alkaline earth metal salts (MX or MX(2)) (LiClO(4), LiI, NaClO(4), and Mg(ClO(4))(2)), using stopped-flow spectrophotometry. clo 284-287 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 21739987-5 2011 By reacting ArO( ) with alpha-TocH in acetonitrile, the absorption of ArO( ) disappeared rapidly, while that of alpha-Toc( ) appeared and then decreased gradually as a result of the bimolecular self-reaction of alpha-Toc( ) after passing through the maximum. acetonitrile 38-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-15 21608133-0 2011 Aromatase and dual aromatase-steroid sulfatase inhibitors from the letrozole and vorozole templates. Letrozole 67-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21608133-0 2011 Aromatase and dual aromatase-steroid sulfatase inhibitors from the letrozole and vorozole templates. vorozole 81-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21608133-0 2011 Aromatase and dual aromatase-steroid sulfatase inhibitors from the letrozole and vorozole templates. Letrozole 67-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 19-28 21608133-0 2011 Aromatase and dual aromatase-steroid sulfatase inhibitors from the letrozole and vorozole templates. vorozole 81-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 19-28 21693046-1 2011 Aromatase inhibitors effectively delay epiphysial maturation in boys and improve testosterone levels in adult men Therefore, aromatase inhibitors may be used to increase adult height in boys with gonadotropin-independent precocious puberty, idiopathic short stature and constitutional delay of puberty. Testosterone 81-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-134 21608133-2 2011 Three aromatase inhibitors with sub-nanomolar potency, better than the benchmark agent letrozole, were designed. Letrozole 87-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 6-15 21608133-4 2011 Amongst achiral and racemic compounds, 2-bromo-4-(2-(4-cyanophenyl)-2-(1H-1,2,4-triazol-1-yl)ethyl)phenyl sulfamate is the most potent DASI (aromatase: IC50 =0.87 nM; STS: IC50 =593 nM). 2-bromo-4-(2-(4-cyanophenyl)-2-(1h-1,2,4-triazol-1-yl)ethyl)phenyl sulfamate 39-115 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 141-150 21615185-7 2011 We developed the version of temperature derivative spectroscopy to monitor kinetics of autoxidation of cytochromes P450 and applied it to study the properties of the oxy-ferrous complex of a human membrane bound P450, CYP19A1 (aromatase), and that of a bacterial soluble P450, CYP101 when bound with their most common substrates, androstenedione (AD) and camphor, respectively. Androstenedione 330-345 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 218-225 21615185-7 2011 We developed the version of temperature derivative spectroscopy to monitor kinetics of autoxidation of cytochromes P450 and applied it to study the properties of the oxy-ferrous complex of a human membrane bound P450, CYP19A1 (aromatase), and that of a bacterial soluble P450, CYP101 when bound with their most common substrates, androstenedione (AD) and camphor, respectively. Camphor 355-362 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 218-225 21479914-5 2011 Variants of CYP19A1 were associated with CIMP+ and CIMP+/KRAS2-mutated tumors. cimp+ 41-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-19 21738342-6 2011 The thyroid cancer cell lines, TPC-1, FTC-133, NPA, FRO, and ARO, displayed growth inhibition in response to genistein, resveratrol, quercetin. Genistein 109-118 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-64 21738342-6 2011 The thyroid cancer cell lines, TPC-1, FTC-133, NPA, FRO, and ARO, displayed growth inhibition in response to genistein, resveratrol, quercetin. Resveratrol 120-131 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-64 21738342-6 2011 The thyroid cancer cell lines, TPC-1, FTC-133, NPA, FRO, and ARO, displayed growth inhibition in response to genistein, resveratrol, quercetin. Quercetin 133-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-64 20888390-1 2011 Aromatase, estrone sulfatase, and 17beta-hydroxysteroid dehydrogenase type 1 are involved in the key steps of 17beta-estradiol biosynthesis. Estradiol 110-126 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21521281-6 2011 Aromatase enzyme activity was completely lost when the mutant p.N411S protein was expressed in COS-1 cells. carbonyl sulfide 95-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21392519-3 2011 Recent studies demonstrated that estrogens are synthesized in situ in both male and female lung cancers through aromatase, suggesting that sex steroid may contribute to the pathogenesis and development of lung carcinoma. Steroids 143-150 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 112-121 21693046-1 2011 Aromatase inhibitors effectively delay epiphysial maturation in boys and improve testosterone levels in adult men Therefore, aromatase inhibitors may be used to increase adult height in boys with gonadotropin-independent precocious puberty, idiopathic short stature and constitutional delay of puberty. Testosterone 81-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 24900349-4 2011 Most interesting was the naphth-1-yl compound 23 (IC50 = 42 nM), showing a 49-fold selectivity toward the highly homologous CYP11B2 (1: 18-fold) as well as selectivity toward the androgen and estrogen forming enzymes CYP17 and CYP19, respectively. naphth-1-yl 25-36 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 227-232 21296134-0 2011 Aflatoxin B1--a potential endocrine disruptor--up-regulates CYP19A1 in JEG-3 cells. Aflatoxin B1 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-67 21282199-1 2011 Aromatase is a key enzyme involved in estradiol and estrone biosynthesis. Estradiol 38-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21282199-1 2011 Aromatase is a key enzyme involved in estradiol and estrone biosynthesis. Estrone 52-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21282199-2 2011 Given that polymorphisms of the CYP19A1 gene encoding aromatase have been correlated with plasma testosterone levels, CYP19A1 may therefore act as a genetic modifier of the hyperandrogenic phenotype of polycystic ovary syndrome (PCOS). Testosterone 97-109 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-39 21282199-2 2011 Given that polymorphisms of the CYP19A1 gene encoding aromatase have been correlated with plasma testosterone levels, CYP19A1 may therefore act as a genetic modifier of the hyperandrogenic phenotype of polycystic ovary syndrome (PCOS). Testosterone 97-109 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 54-63 21282199-2 2011 Given that polymorphisms of the CYP19A1 gene encoding aromatase have been correlated with plasma testosterone levels, CYP19A1 may therefore act as a genetic modifier of the hyperandrogenic phenotype of polycystic ovary syndrome (PCOS). Testosterone 97-109 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 118-125 20685100-0 2011 Retinol inhibits aromatase activity and expression in vitro. Vitamin A 0-7 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 20685100-3 2011 We investigated the effect of retinol and selected retinoids on the activity and expression of aromatase in two human carcinoma cell lines in vitro. Vitamin A 30-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-104 20685100-3 2011 We investigated the effect of retinol and selected retinoids on the activity and expression of aromatase in two human carcinoma cell lines in vitro. Retinoids 51-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-104 20685100-4 2011 Retinol (ROH) and all-trans retinoic acid (ATRA) significantly inhibited aromatase activity in a concentration-dependent manner in microsomes isolated from JEG-3 human placental carcinoma cells, whereas 9-cis and 13-cis retinoic acid had significant inhibitory activity only at the highest concentrations tested. Vitamin A 0-7 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-82 20685100-4 2011 Retinol (ROH) and all-trans retinoic acid (ATRA) significantly inhibited aromatase activity in a concentration-dependent manner in microsomes isolated from JEG-3 human placental carcinoma cells, whereas 9-cis and 13-cis retinoic acid had significant inhibitory activity only at the highest concentrations tested. roh 9-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-82 20685100-4 2011 Retinol (ROH) and all-trans retinoic acid (ATRA) significantly inhibited aromatase activity in a concentration-dependent manner in microsomes isolated from JEG-3 human placental carcinoma cells, whereas 9-cis and 13-cis retinoic acid had significant inhibitory activity only at the highest concentrations tested. Tretinoin 18-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-82 20685100-4 2011 Retinol (ROH) and all-trans retinoic acid (ATRA) significantly inhibited aromatase activity in a concentration-dependent manner in microsomes isolated from JEG-3 human placental carcinoma cells, whereas 9-cis and 13-cis retinoic acid had significant inhibitory activity only at the highest concentrations tested. Tretinoin 43-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-82 20685100-7 2011 In addition, ROH suppressed both the basal and cAMP-induced expression of aromatase mRNA in MCF-7 cells and inhibited transcription controlled by a cAMP-responsive element. Cyclic AMP 47-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 74-83 21296134-1 2011 Previous studies have indicated that aromatase (CYP19A1) is involved in the metabolism of aflatoxin B1 (AFB1). Aflatoxin B1 90-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-55 21296134-1 2011 Previous studies have indicated that aromatase (CYP19A1) is involved in the metabolism of aflatoxin B1 (AFB1). Aflatoxin B1 104-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-55 21296134-4 2011 JEG-3 cells, as model for placental cells, were exposed alone and in combination to AFB1 and estrogen receptor ligands for 24-96 h. AFB1 (0.3-1.0 muM) induced the expression of CYP19A1 by 163%-339% compared to control at the 96 h time point, although no induction was observed at 24 h. AFB1 concentrations higher than 1 muM were cytotoxic to JEG-3 cells, and the cytotoxicity was inhibited by the aromatase inhibitor, finrozole. finrozole 418-427 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 177-184 21296134-5 2011 AFB1 was metabolized to aflatoxicol (AFL) by JEG-3 cells and CYP19A1 recombinant protein. Aflatoxin B1 0-4 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-68 21353673-8 2011 CONCLUSION(S): These results indicate that the release of negative feedback inhibition of estradiol on the hypothalamic-pituitary axis in normal women by aromatase inhibitors creates an amplitude-related increase in endogenous hypothalamic-pituitary drive. Estradiol 90-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-163 21296658-0 2011 Differential accumulation of HCBz and PeCBz in porcine ovarian follicles and their opposing actions on steroid secretion and CYP11, CYP17, 17beta-HSD and CYP19 protein expression. Hexachlorobenzene 29-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-159 21492245-0 2011 Aromatase inhibitor-induced skin adverse reactions: exemestane-related cutaneous vasculitis. exemestane 52-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21492245-1 2011 BACKGROUND: Three aromatase inhibitors, namely anastrozole, letrozole and exemestane, which reduce circulating oestrogen, are used to treat breast cancer patients; the therapeutic use of such aromatase inhibitors is quickly increasing. Anastrozole 47-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 18-27 21492245-1 2011 BACKGROUND: Three aromatase inhibitors, namely anastrozole, letrozole and exemestane, which reduce circulating oestrogen, are used to treat breast cancer patients; the therapeutic use of such aromatase inhibitors is quickly increasing. Anastrozole 47-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 192-201 21492245-1 2011 BACKGROUND: Three aromatase inhibitors, namely anastrozole, letrozole and exemestane, which reduce circulating oestrogen, are used to treat breast cancer patients; the therapeutic use of such aromatase inhibitors is quickly increasing. Letrozole 60-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 18-27 21492245-1 2011 BACKGROUND: Three aromatase inhibitors, namely anastrozole, letrozole and exemestane, which reduce circulating oestrogen, are used to treat breast cancer patients; the therapeutic use of such aromatase inhibitors is quickly increasing. Letrozole 60-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 192-201 21492245-1 2011 BACKGROUND: Three aromatase inhibitors, namely anastrozole, letrozole and exemestane, which reduce circulating oestrogen, are used to treat breast cancer patients; the therapeutic use of such aromatase inhibitors is quickly increasing. exemestane 74-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 18-27 21277123-0 2011 Aromatase inhibitor-induced arthralgia: is vitamin D deficiency responsible? Vitamin D 43-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21413907-0 2011 Aromatase inhibition: a potential target for the management of recurrent or metastatic endometrial cancer by letrozole: more questions than answers? Letrozole 109-118 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21413907-9 2011 EXPERT OPINION: Further understanding of the relevance of aromatase and estrogen receptor and their interplay with other growth pathways will be necessary to guide further development of letrozole. Letrozole 187-196 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-67 21492245-1 2011 BACKGROUND: Three aromatase inhibitors, namely anastrozole, letrozole and exemestane, which reduce circulating oestrogen, are used to treat breast cancer patients; the therapeutic use of such aromatase inhibitors is quickly increasing. exemestane 74-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 192-201 21492245-2 2011 OBJECTIVE: We intended (i) to review aromatase inhibitor-induced cutaneous adverse effects and (ii) to describe a recently observed case of cutaneous vasculitis triggered by exemestane. exemestane 174-184 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-46 21492245-7 2011 CONCLUSION: As aromatase inhibitors (e.g. exemestane) are increasingly incorporated into the treatment strategy of breast cancer patients, it is important to recognize possible cutaneous adverse effects. exemestane 42-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-24 21094255-3 2011 Certain members of the P-450 family, including CYP17 and CYP19, catalyze, at the same active site, not only the hydroxylation process but also an acyl-carbon bond cleavage reaction which has been interpreted to involve the nucleophilic attack of the ferric-peroxyanion, Fe(III)-O-O(-), on the acyl carbon to furnish a tetrahedral intermediate which fragments, leading to acyl-carbon cleavage. Carbon 151-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-62 21094255-3 2011 Certain members of the P-450 family, including CYP17 and CYP19, catalyze, at the same active site, not only the hydroxylation process but also an acyl-carbon bond cleavage reaction which has been interpreted to involve the nucleophilic attack of the ferric-peroxyanion, Fe(III)-O-O(-), on the acyl carbon to furnish a tetrahedral intermediate which fragments, leading to acyl-carbon cleavage. ferric-peroxyanion 250-268 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-62 21094255-3 2011 Certain members of the P-450 family, including CYP17 and CYP19, catalyze, at the same active site, not only the hydroxylation process but also an acyl-carbon bond cleavage reaction which has been interpreted to involve the nucleophilic attack of the ferric-peroxyanion, Fe(III)-O-O(-), on the acyl carbon to furnish a tetrahedral intermediate which fragments, leading to acyl-carbon cleavage. fe(iii)-o-o 270-281 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-62 21094255-3 2011 Certain members of the P-450 family, including CYP17 and CYP19, catalyze, at the same active site, not only the hydroxylation process but also an acyl-carbon bond cleavage reaction which has been interpreted to involve the nucleophilic attack of the ferric-peroxyanion, Fe(III)-O-O(-), on the acyl carbon to furnish a tetrahedral intermediate which fragments, leading to acyl-carbon cleavage. Carbon 298-304 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-62 21094255-3 2011 Certain members of the P-450 family, including CYP17 and CYP19, catalyze, at the same active site, not only the hydroxylation process but also an acyl-carbon bond cleavage reaction which has been interpreted to involve the nucleophilic attack of the ferric-peroxyanion, Fe(III)-O-O(-), on the acyl carbon to furnish a tetrahedral intermediate which fragments, leading to acyl-carbon cleavage. Carbon 298-304 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-62 21296658-0 2011 Differential accumulation of HCBz and PeCBz in porcine ovarian follicles and their opposing actions on steroid secretion and CYP11, CYP17, 17beta-HSD and CYP19 protein expression. pentachlorobenzene 38-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-159 21296658-5 2011 Immunoblot analyses showed an inhibitory effect of HCBz on CYP17, 17beta-HSD and CYP19, a stimulatory effect of PeCBz on CYP17 and CYP19 and no effect on 17beta-HSD protein expression. Hexachlorobenzene 51-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-86 21232533-2 2011 In this study, we measured the reaction rates of vitamin E (alpha-, beta-, gamma-, delta-tocopherols, TocH) and tocol with aroxyl radical (ArO ) as model lipid peroxyl radicals in membranes by stopped-flow spectrophotometry. Vitamin E 49-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-142 21215765-0 2011 Synthesis of some novel androstanes as potential aromatase inhibitors. Androstanes 24-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-58 21215765-1 2011 Novel pyrazole and isoxazole derivatives (6-9) were synthesized as a aromatase inhibitors. pyrazole 6-14 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-78 21215765-1 2011 Novel pyrazole and isoxazole derivatives (6-9) were synthesized as a aromatase inhibitors. Isoxazoles 19-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-78 21215765-5 2011 Aromatase inhibitory activity for compound (6) having pyrazole ring at 2,3 position showed highest activity followed by nitrile derivative (9). pyrazole 54-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21215765-5 2011 Aromatase inhibitory activity for compound (6) having pyrazole ring at 2,3 position showed highest activity followed by nitrile derivative (9). Nitriles 120-127 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21341743-0 2011 Design, synthesis, and biological evaluation of imidazolyl derivatives of 4,7-disubstituted coumarins as aromatase inhibitors selective over 17-alpha-hydroxylase/C17-20 lyase. imidazolyl 48-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 105-114 21341743-0 2011 Design, synthesis, and biological evaluation of imidazolyl derivatives of 4,7-disubstituted coumarins as aromatase inhibitors selective over 17-alpha-hydroxylase/C17-20 lyase. 4,7-disubstituted coumarins 74-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 105-114 21341743-1 2011 The design, synthesis, and biological evaluation of a series of new aromatase (AR, CYP19) inhibitors bearing an imidazole ring linked to a 7-substituted coumarin scaffold at position 4 (or 3) are reported. imidazole 112-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-77 21341743-1 2011 The design, synthesis, and biological evaluation of a series of new aromatase (AR, CYP19) inhibitors bearing an imidazole ring linked to a 7-substituted coumarin scaffold at position 4 (or 3) are reported. imidazole 112-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-81 21341743-1 2011 The design, synthesis, and biological evaluation of a series of new aromatase (AR, CYP19) inhibitors bearing an imidazole ring linked to a 7-substituted coumarin scaffold at position 4 (or 3) are reported. imidazole 112-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-88 21341743-1 2011 The design, synthesis, and biological evaluation of a series of new aromatase (AR, CYP19) inhibitors bearing an imidazole ring linked to a 7-substituted coumarin scaffold at position 4 (or 3) are reported. coumarin 153-161 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-77 21341743-1 2011 The design, synthesis, and biological evaluation of a series of new aromatase (AR, CYP19) inhibitors bearing an imidazole ring linked to a 7-substituted coumarin scaffold at position 4 (or 3) are reported. coumarin 153-161 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-81 21341743-1 2011 The design, synthesis, and biological evaluation of a series of new aromatase (AR, CYP19) inhibitors bearing an imidazole ring linked to a 7-substituted coumarin scaffold at position 4 (or 3) are reported. coumarin 153-161 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-88 21341743-3 2011 The most potent AR inhibitor was the 7-(3,4-difluorophenoxy)-4-imidazolylmethyl coumarin 24 endowed with an IC(50) = 47 nM. 7-(3,4-difluorophenoxy)-4-imidazolylmethyl coumarin 37-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-18 21217768-8 2011 The higher frequency of short CYP19 alleles in oligospermic men compared to normozoospermic men (43.3 and 28.3%, respectively; P<0.01) was primarily due to the distribution of the CYP19 (TTTA)(7) allele. ttta 190-194 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-35 21217768-8 2011 The higher frequency of short CYP19 alleles in oligospermic men compared to normozoospermic men (43.3 and 28.3%, respectively; P<0.01) was primarily due to the distribution of the CYP19 (TTTA)(7) allele. ttta 190-194 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 183-188 21217768-9 2011 The CYP19 (TTTA)(7) allele was associated with lower sperm concentration in normozoospermic men (P<0.01) and in the total study population (P<0.01); it was also associated with lower sperm motility in normozoospermic men (P<0.05) and in the total study population (P<0.01). ttta 11-15 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-9 21217768-10 2011 In conclusion, the CYP19 (TTTA)(7) allele probably impairs aromatase activity, which in turn alters aromatase and oestrogen levels in the testis, leading to decreased sperm concentration and motility. ttta 26-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 19-24 19921206-1 2011 PURPOSE: The known importance of testosterone for the development of benign prostatic hyperplasia (BPH) prompted us to test the hypothesis whether polymorphisms of two genes (CYP19A1 and CYP3A4) involved in testosterone metabolism are associated with clinical BPH-parameters. Testosterone 207-219 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 175-182 21232533-2 2011 In this study, we measured the reaction rates of vitamin E (alpha-, beta-, gamma-, delta-tocopherols, TocH) and tocol with aroxyl radical (ArO ) as model lipid peroxyl radicals in membranes by stopped-flow spectrophotometry. toch 102-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-142 21232533-2 2011 In this study, we measured the reaction rates of vitamin E (alpha-, beta-, gamma-, delta-tocopherols, TocH) and tocol with aroxyl radical (ArO ) as model lipid peroxyl radicals in membranes by stopped-flow spectrophotometry. aroxyl radical 123-137 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-142 21232533-2 2011 In this study, we measured the reaction rates of vitamin E (alpha-, beta-, gamma-, delta-tocopherols, TocH) and tocol with aroxyl radical (ArO ) as model lipid peroxyl radicals in membranes by stopped-flow spectrophotometry. lipid peroxyl radicals 154-176 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-142 25961250-3 2011 RESULTS: Following 24 h and 72 h incubation of liver tissues or cells with testosterone, human HCC tissues and HepG2 hepatoma cells showed elevated Aro activity (estrogen formation, respectively, of 20% and 52%-99%). Testosterone 75-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 148-151 21106711-6 2011 The CYP19 R264C variant was related to the duration of survival after docetaxel in patients who were >70 years old (median survival difference =10.6 months; P=0.041). Docetaxel 70-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-9 21176111-8 2011 Acute exposure (6 hours) to both PDE5i tadalafil and sildenafil increased ARO mRNA expression by 4.7- and 2.8-fold, respectively (P < 0.001). Tadalafil 39-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 74-77 21176111-8 2011 Acute exposure (6 hours) to both PDE5i tadalafil and sildenafil increased ARO mRNA expression by 4.7- and 2.8-fold, respectively (P < 0.001). Sildenafil Citrate 53-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 74-77 21176111-12 2011 Such effect was reversed by the co-incubation with the specific ARO-inhibitor letrozole. Letrozole 78-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-67 21212407-4 2011 Prostaglandin E(2) (PGE(2)) stimulates the cyclic AMP (cAMP) protein kinase A (PKA) cAMP responsive element binding protein (CREB) pathway leading to increased CYP19 transcription. Dinoprostone 0-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 164-169 21212407-4 2011 Prostaglandin E(2) (PGE(2)) stimulates the cyclic AMP (cAMP) protein kinase A (PKA) cAMP responsive element binding protein (CREB) pathway leading to increased CYP19 transcription. Dinoprostone 20-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 164-169 21212407-4 2011 Prostaglandin E(2) (PGE(2)) stimulates the cyclic AMP (cAMP) protein kinase A (PKA) cAMP responsive element binding protein (CREB) pathway leading to increased CYP19 transcription. Cyclic AMP 43-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 164-169 21212407-4 2011 Prostaglandin E(2) (PGE(2)) stimulates the cyclic AMP (cAMP) protein kinase A (PKA) cAMP responsive element binding protein (CREB) pathway leading to increased CYP19 transcription. Cyclic AMP 55-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 164-169 21212407-7 2011 Silencing of PGT in preadipocytes increased PGE(2) levels in the extracellular medium, thereby stimulating the cAMP PKA pathway resulting in enhanced interaction between pCREB, p300, and the CYP19 I.3/II promoter. Cyclic AMP 111-115 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 193-198 21212407-14 2011 Collectively, these results suggest that PGT modulates adipogenesis and thereby PGE(2)-mediated activation of the cAMP PKA CREB pathway leading to altered CYP19 transcription and aromatase activity. Dinoprostone 80-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 159-164 21212407-14 2011 Collectively, these results suggest that PGT modulates adipogenesis and thereby PGE(2)-mediated activation of the cAMP PKA CREB pathway leading to altered CYP19 transcription and aromatase activity. Cyclic AMP 114-118 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 159-164 21106711-2 2011 OBJECTIVE: We hypothesized that polymorphisms in estrogen synthesis and estrogen targets (i.e., CYP19 and ERalpha) would be linked to interindividual variation in CRPC risk, docetaxel response, and overall survival in men with CRPC. Docetaxel 174-183 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-101 21125689-2 2011 These catalysts in the presence of nonracemic imine as an additive provided beta-amino alcohol in excellent yield (99%) and chiral purity (enantiomeric excess (ee) up to 99%) for the ARO of meso-stilbene oxide with aniline. Imines 46-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 183-186 21125689-2 2011 These catalysts in the presence of nonracemic imine as an additive provided beta-amino alcohol in excellent yield (99%) and chiral purity (enantiomeric excess (ee) up to 99%) for the ARO of meso-stilbene oxide with aniline. beta-amino alcohol 76-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 183-186 21125689-2 2011 These catalysts in the presence of nonracemic imine as an additive provided beta-amino alcohol in excellent yield (99%) and chiral purity (enantiomeric excess (ee) up to 99%) for the ARO of meso-stilbene oxide with aniline. stilbene oxide 190-209 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 183-186 21125689-2 2011 These catalysts in the presence of nonracemic imine as an additive provided beta-amino alcohol in excellent yield (99%) and chiral purity (enantiomeric excess (ee) up to 99%) for the ARO of meso-stilbene oxide with aniline. aniline 215-222 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 183-186 21125689-3 2011 The same protocol was less effective for the ARO of cyclic epoxides; however, when triphenylphosphine was used as an additive, there was a significant improvement in catalyst performance for the ARO of cyclohexene oxide (yield, 85-90%; ee, 63-67%). cyclic epoxides 52-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-48 21125689-3 2011 The same protocol was less effective for the ARO of cyclic epoxides; however, when triphenylphosphine was used as an additive, there was a significant improvement in catalyst performance for the ARO of cyclohexene oxide (yield, 85-90%; ee, 63-67%). triphenylphosphine 83-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-48 21125689-3 2011 The same protocol was less effective for the ARO of cyclic epoxides; however, when triphenylphosphine was used as an additive, there was a significant improvement in catalyst performance for the ARO of cyclohexene oxide (yield, 85-90%; ee, 63-67%). triphenylphosphine 83-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 195-198 21125689-3 2011 The same protocol was less effective for the ARO of cyclic epoxides; however, when triphenylphosphine was used as an additive, there was a significant improvement in catalyst performance for the ARO of cyclohexene oxide (yield, 85-90%; ee, 63-67%). cyclohexene oxide 202-219 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-48 21125689-3 2011 The same protocol was less effective for the ARO of cyclic epoxides; however, when triphenylphosphine was used as an additive, there was a significant improvement in catalyst performance for the ARO of cyclohexene oxide (yield, 85-90%; ee, 63-67%). cyclohexene oxide 202-219 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 195-198 20526739-6 2011 Immunostaining for aromatase employed the streptavidin-biotin amplification method and #677 mouse monoclonal antibody. Biotin 55-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 19-28 22335023-2 2011 Polymorphic variation in the CYP19 gene can affect estrogen synthesis by increasing aromatase activity resulting in elevated levels of estrone and estradiol. Estrone 135-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-34 22335023-2 2011 Polymorphic variation in the CYP19 gene can affect estrogen synthesis by increasing aromatase activity resulting in elevated levels of estrone and estradiol. Estradiol 147-156 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-34 21366040-1 2011 Flavones such as chrysin show structural similarities to androgens, the substrates of human aromatase, which converts androgens to estrogens. Flavones 0-8 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 21047944-5 2011 In addition, we measured aromatase activity by assaying estrone levels after administration of its precursor, androstenedione. Estrone 56-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-34 21047944-5 2011 In addition, we measured aromatase activity by assaying estrone levels after administration of its precursor, androstenedione. Androstenedione 110-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-34 21366040-1 2011 Flavones such as chrysin show structural similarities to androgens, the substrates of human aromatase, which converts androgens to estrogens. chrysin 17-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 21366040-3 2011 Flavone-based aromatase inhibitors are of growing interest, and chrysin in particular provides a (natural) lead structure. flavone 0-7 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 21366040-5 2011 A Mannich-type reaction was used to synthesize a range of mono- and disubstituted chrysin derivatives, some of which are more effective aromatase inhibitors than the benchmark compound, aminoglutethimide. chrysin 82-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-145 21366040-6 2011 Similarly, the reaction of chrysin with various isonitriles and acetylene dicarboxylates results in a new class of flavone derivatives, tricyclic pyrano-flavones which also inhibit human aromatase. isonitriles 48-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 187-196 21366040-6 2011 Similarly, the reaction of chrysin with various isonitriles and acetylene dicarboxylates results in a new class of flavone derivatives, tricyclic pyrano-flavones which also inhibit human aromatase. acetylene dicarboxylates 64-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 187-196 21366040-6 2011 Similarly, the reaction of chrysin with various isonitriles and acetylene dicarboxylates results in a new class of flavone derivatives, tricyclic pyrano-flavones which also inhibit human aromatase. flavone 115-122 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 187-196 21366040-6 2011 Similarly, the reaction of chrysin with various isonitriles and acetylene dicarboxylates results in a new class of flavone derivatives, tricyclic pyrano-flavones which also inhibit human aromatase. tricyclic pyrano-flavones 136-161 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 187-196 20967895-7 2011 Only eight mutants showed growth hypersensitivity to all the three treatments and harboured deletions in genes important for aromatic amino acid biosynthesis (ARO1, ARO7) or mitochondrial protein synthesis and respiration (ATP5, ISA1, RML2, GET2, SLS1, MRPL38). Amino Acids, Aromatic 125-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 159-163 21054398-2 2010 In this study, a series of androstenedione derivatives with CYP19 inhibitory activity was subjected to a molecular docking study followed by quantitative structure-activity relationship (QSAR) analyses in search of ideal physicochemical characteristics of potential aromatase inhibitors. Androstenedione 27-42 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-65 22127008-7 2011 In addition, a down-regulation of Cyp19A1, the rate-limiting enzyme of 17beta-estradiol synthesis, was detected in CC cultures after 5h. Estradiol 71-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-41 21187493-0 2010 Aromatase expression in leptin-pretreated human breast pre-adipocytes is enhanced by zeranol and suppressed by (-)-gossypol. Zeranol 85-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21187493-0 2010 Aromatase expression in leptin-pretreated human breast pre-adipocytes is enhanced by zeranol and suppressed by (-)-gossypol. Gossypol 111-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21187493-5 2010 The effects of Z and gossypol on aromatase expression of leptin-pretreated HNBPADs were evaluated by RT-PCR. Gossypol 21-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-42 21187493-8 2010 Moreover, (-)-gossypol counteracted Z- and leptin-induced cell proliferation and aromatase expression. Gossypol 10-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-90 21143665-8 2010 In agreement with the fact that brain aromatase activity is correlated with sex steroid levels, the high expression of cyp19a1b is due to an autoregulatory loop through which estrogens and aromatizable androgens upregulate aromatase expression. Steroids 80-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-47 20430378-8 2010 In control cells, (Bu)(2)cAMP and PF-E increased P(450)Arom and SF-1 expression (but not COUP-TF expression) in a dose-dependent way, an effect not observed with PF-C, adsorbed PF-E, or 10(-5) M indomethacin. Cyclic AMP 25-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-59 20546769-0 2010 4- and 6-(p-Sulphamoylphenyl)androstenediones: Studies of aromatase inhibitor-based oestrone sulphatase inhibition. 4- and 6-(p-sulphamoylphenyl)androstenediones 0-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-67 20546769-1 2010 4-(p-Sulphamoylphenyl)androstenedione (3) and 6alpha-p-sulphamoylphenyl analogues 12-14 were synthesised and tested as aromatase inhibitors as well as oestrone sulphatase inhibitors in human placental microsomes. 4-(p-sulphamoylphenyl)androstenedione 0-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 119-128 20558190-1 2010 Carbon-11-labeled casimiroin analogues were first designed and synthesized as new potential PET agents for imaging of quinone reductase (QR) 2 and aromatase expression in breast cancer. Carbon-11 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 147-156 20558190-0 2010 Synthesis of carbon-11-labeled casimiroin analogues as new potential PET agents for imaging of quinone reductase 2 and aromatase expression in breast cancer. Carbon-11 13-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 119-128 20184498-6 2010 The genotypes obtained for COMT and CYP19 were connected with higher 17&#x03B2;-estradiol production, which was confirmed by significantly elevated serum levels of 17&#x03B2;-estradiol in male patients. 17& 69-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-41 20184498-6 2010 The genotypes obtained for COMT and CYP19 were connected with higher 17&#x03B2;-estradiol production, which was confirmed by significantly elevated serum levels of 17&#x03B2;-estradiol in male patients. Estradiol 84-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-41 20184498-6 2010 The genotypes obtained for COMT and CYP19 were connected with higher 17&#x03B2;-estradiol production, which was confirmed by significantly elevated serum levels of 17&#x03B2;-estradiol in male patients. 17& 168-174 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-41 20184498-6 2010 The genotypes obtained for COMT and CYP19 were connected with higher 17&#x03B2;-estradiol production, which was confirmed by significantly elevated serum levels of 17&#x03B2;-estradiol in male patients. Estradiol 183-192 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-41 20668023-4 2010 IGF-I enhanced the dexamethasone (DEX)-induced aromatase activity by 30% in serum-starved THP-1 cells. Dexamethasone 19-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 20842717-0 2010 Unique distribution of aromatase in the human brain: in vivo studies with PET and [N-methyl-11C]vorozole. vorozole 82-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 23-32 20842717-3 2010 Using positron emission tomography (PET) with the radiolabeled aromatase inhibitor [N-methyl-(11)C]vorozole, we characterized the tracer distribution and kinetics in the living human brain. vorozole 83-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-72 20842717-6 2010 Specificity was confirmed by pretreatment with a pharmacological (2.5 mg) dose of the aromatase inhibitor letrozole. Letrozole 106-115 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-95 20668023-9 2010 In the presence of pepstatin A, DEX caused an increase in the amount of aromatase in both microsomes and lysosomes, and IGF-I attenuated the DEX-induced accumulation of aromatase in lysosomes and, conversely, enhanced its accumulation in the microsomes. Dexamethasone 32-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 72-81 20668023-4 2010 IGF-I enhanced the dexamethasone (DEX)-induced aromatase activity by 30% in serum-starved THP-1 cells. Dexamethasone 34-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 20668023-9 2010 In the presence of pepstatin A, DEX caused an increase in the amount of aromatase in both microsomes and lysosomes, and IGF-I attenuated the DEX-induced accumulation of aromatase in lysosomes and, conversely, enhanced its accumulation in the microsomes. Dexamethasone 32-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-178 20668023-9 2010 In the presence of pepstatin A, DEX caused an increase in the amount of aromatase in both microsomes and lysosomes, and IGF-I attenuated the DEX-induced accumulation of aromatase in lysosomes and, conversely, enhanced its accumulation in the microsomes. Dexamethasone 141-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 72-81 20668023-9 2010 In the presence of pepstatin A, DEX caused an increase in the amount of aromatase in both microsomes and lysosomes, and IGF-I attenuated the DEX-induced accumulation of aromatase in lysosomes and, conversely, enhanced its accumulation in the microsomes. Dexamethasone 141-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-178 20668023-13 2010 Finally, we observed that IGF-I enhanced the aromatase activity by 50% as early as 1 h after treatment; furthermore, rapamycin, an enhancer of autophagy, completely negated the effect of IGF-I on the enzyme. Sirolimus 117-126 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-54 20668023-7 2010 We identified pepstatin A as the most effective inhibitor of aromatase degradation by in vitro assay. pepstatin 14-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-70 20668023-9 2010 In the presence of pepstatin A, DEX caused an increase in the amount of aromatase in both microsomes and lysosomes, and IGF-I attenuated the DEX-induced accumulation of aromatase in lysosomes and, conversely, enhanced its accumulation in the microsomes. pepstatin 19-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 72-81 20668023-9 2010 In the presence of pepstatin A, DEX caused an increase in the amount of aromatase in both microsomes and lysosomes, and IGF-I attenuated the DEX-induced accumulation of aromatase in lysosomes and, conversely, enhanced its accumulation in the microsomes. pepstatin 19-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-178 20505544-3 2010 We evaluated the relationship between steroid hormone metabolism genotypes at COMT, CYP1A2, CYP1B1, CYP3A4, CYP19, SULT1A1, and SULT1E1 with hormone levels and menopausal features. Steroids 38-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-113 20564173-4 2010 In addition, the expression of cyp19 gene, which is involved both in neuronal differentiation and stress response, was evaluated; cyp19 gene expression was enhanced by ATRA treatment and significantly enhanced further by ELF-MF exposure combined with ATRA. Tretinoin 168-172 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-36 20564173-4 2010 In addition, the expression of cyp19 gene, which is involved both in neuronal differentiation and stress response, was evaluated; cyp19 gene expression was enhanced by ATRA treatment and significantly enhanced further by ELF-MF exposure combined with ATRA. Tretinoin 168-172 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-135 20564173-4 2010 In addition, the expression of cyp19 gene, which is involved both in neuronal differentiation and stress response, was evaluated; cyp19 gene expression was enhanced by ATRA treatment and significantly enhanced further by ELF-MF exposure combined with ATRA. Tretinoin 251-255 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-36 20564173-4 2010 In addition, the expression of cyp19 gene, which is involved both in neuronal differentiation and stress response, was evaluated; cyp19 gene expression was enhanced by ATRA treatment and significantly enhanced further by ELF-MF exposure combined with ATRA. Tretinoin 251-255 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-135 20503248-0 2010 Anti-proliferative effects of evodiamine on human thyroid cancer cell line ARO. evodiamine 30-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-78 20503248-11 2010 These results suggested that evodiamine inhibited the growth of the ARO cells, arrested them at M phase, and induced apoptosis through caspases signaling. evodiamine 29-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-71 20503248-6 2010 In the present study, the mechanism by which evodiamine inhibited the undifferentiated thyroid cancer cell line ARO was examined. evodiamine 45-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 112-115 20503248-8 2010 According to the flow cytometric analysis, evodiamine treatment resulted in G2/M arrest and DNA fragmentation in ARO cells. evodiamine 43-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-116 20410453-0 2010 Methadone: a substrate and mechanism-based inhibitor of CYP19 (aromatase). Methadone 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-61 20521760-2 2010 A time-resolved kinetic study on the reactions of alkoxyl radicals with trialkyl and triaryl phosphites ((RO)(3)P: R = Me, Et, i-Pr, t-Bu; (ArO)(3)P: Ar = C(6)H(5), 2,4-(t-Bu)(2)C(6)H(3)) has been carried out. alkoxyl radical 50-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 140-143 20410453-7 2010 Preincubation of methadone, EDDP, or EMDP with CYP19 resulted in time- and concentration-dependent inhibition, indicating a mechanism-based reaction that destroys CYP19 activity. Methadone 17-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-52 20410453-7 2010 Preincubation of methadone, EDDP, or EMDP with CYP19 resulted in time- and concentration-dependent inhibition, indicating a mechanism-based reaction that destroys CYP19 activity. Methadone 17-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 163-168 20410453-7 2010 Preincubation of methadone, EDDP, or EMDP with CYP19 resulted in time- and concentration-dependent inhibition, indicating a mechanism-based reaction that destroys CYP19 activity. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 28-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-52 20410453-7 2010 Preincubation of methadone, EDDP, or EMDP with CYP19 resulted in time- and concentration-dependent inhibition, indicating a mechanism-based reaction that destroys CYP19 activity. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 28-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 163-168 20410453-10 2010 Methadone is metabolized by CYP19 and may act as a potent inhibitor of CYP19 in vivo. Methadone 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-33 20410453-10 2010 Methadone is metabolized by CYP19 and may act as a potent inhibitor of CYP19 in vivo. Methadone 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-76 19906189-2 2010 In testis, P450-aromatase, encoded by CYP19, is responsible for the conversion of testosterone to oestradiol. Testosterone 82-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-25 19906189-2 2010 In testis, P450-aromatase, encoded by CYP19, is responsible for the conversion of testosterone to oestradiol. Testosterone 82-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-43 19906189-2 2010 In testis, P450-aromatase, encoded by CYP19, is responsible for the conversion of testosterone to oestradiol. Estradiol 98-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-25 19906189-2 2010 In testis, P450-aromatase, encoded by CYP19, is responsible for the conversion of testosterone to oestradiol. Estradiol 98-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-43 19906189-13 2010 However, an altered intratesticular oestradiol/testosterone ratio in some patients with complete SCOS suggests that aromatase is increased, which might contribute to Leydig cell dysfunction. Estradiol 36-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-125 19906189-13 2010 However, an altered intratesticular oestradiol/testosterone ratio in some patients with complete SCOS suggests that aromatase is increased, which might contribute to Leydig cell dysfunction. Testosterone 47-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-125 20527908-6 2010 An O-H group in the para position of ArO(*) or ArCH(2)(+) becomes more acidic than in the parent molecules and hence forms stronger HBs with hydrogen bond acceptors (HBAs) in the meta position. Hydrogen 141-149 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-40 20521760-2 2010 A time-resolved kinetic study on the reactions of alkoxyl radicals with trialkyl and triaryl phosphites ((RO)(3)P: R = Me, Et, i-Pr, t-Bu; (ArO)(3)P: Ar = C(6)H(5), 2,4-(t-Bu)(2)C(6)H(3)) has been carried out. triaryl phosphites 85-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 140-143 20521760-7 2010 CumO(*) reacts with triaryl phosphites (ArO)(3)P to give phenoxyl radicals, with rate constants that are influenced to a limited extent by substitution of the aromatic rings. phenoxy radical 57-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 40-43 20052540-5 2010 It can be concluded that potentially functional CYP19_630 3-bp del/Ins polymorphism and CYP19_681 (TTTA)(n) polymorphisms may play a low penetrance role in breast cancer susceptibility in an ethnicity-specific manner. ttta 99-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-93 20211687-6 2010 However, inhibition of DNA methylation with 5-aza-2"-deoxycytidine resulted in a significant approximately 40-fold induction in CYP19 mRNA expression in BAFs and breast cell lines. Decitabine 44-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 128-133 20173016-7 2010 RESULTS: Several polymorphisms in SHBG, ESR2, AR, CYP19A1, and LHB were significantly associated with circulating levels of SHBG, LH, total, free, and bioavailable testosterone and estradiol, the LH x testosterone product, and indices of insulin sensitivity. Testosterone 164-176 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-57 19962423-0 2010 2,3,7,8-Tetrachlorodibenzo-para-dioxin increases aromatase (CYP19) mRNA stability in MCF-7 cells. 2,3,7,8-tetrachlorodibenzo-para-dioxin 0-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-65 19962423-9 2010 Further investigation indicated that TCDD slowed down the CYP19 mRNA degradation with concurrent activation of ERK. Polychlorinated Dibenzodioxins 37-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-63 20359372-12 2010 Treatment with calcium phosphate nanoparticles at concentrations of 10-100 microM didn"t significantly change either the progesterone or estradiol levels in culture fluid, and the expression levels of mRNAs encoding P450scc, P450arom and StAR after 48 h and 72 h period of treatment. calcium phosphate 15-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 225-233 20121156-2 2010 C-O insertion was found to be controlled by Pd-HOMO ArO-LUMO interaction, where C-Cl insertion is facilitated by the lower C-Cl TS distortion energy. Cefaclor 80-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-55 20121156-2 2010 C-O insertion was found to be controlled by Pd-HOMO ArO-LUMO interaction, where C-Cl insertion is facilitated by the lower C-Cl TS distortion energy. Carbon 0-1 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-55 21551958-10 2011 CLI clearly demonstrated RI uptake in thyroid gland and xenografted ARO-NIS cells in mice, which was further confirmed by NI. iodine monochloride 0-3 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-71 20173016-7 2010 RESULTS: Several polymorphisms in SHBG, ESR2, AR, CYP19A1, and LHB were significantly associated with circulating levels of SHBG, LH, total, free, and bioavailable testosterone and estradiol, the LH x testosterone product, and indices of insulin sensitivity. Estradiol 181-190 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-57 19952760-2 2010 This study examined whether the genetic polymorphisms of estrogen receptors (ESR-alpha and ESR-beta) and cytochrome P450 19A1 (CYP19A1) modified the protective effect of isoflavones against prostate cancer. Isoflavones 170-181 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-134 20173016-7 2010 RESULTS: Several polymorphisms in SHBG, ESR2, AR, CYP19A1, and LHB were significantly associated with circulating levels of SHBG, LH, total, free, and bioavailable testosterone and estradiol, the LH x testosterone product, and indices of insulin sensitivity. Testosterone 201-213 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-57 19952760-8 2010 The combination of the TTTA short repeat and those homozygous for the major allele of rs10046 in CYP19A1 was low risk despite less than 60 mg isoflavones/day. Isoflavones 142-153 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 97-104 19808095-5 2010 We have determined the crystal structure of the highly active aromatase purified from human placenta, in complex with its natural substrate androstenedione. Androstenedione 140-155 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-71 20186154-4 2010 The distribution of CYP19A1 (TTTA)(n) polymorphisms was such that the most frequent allele was (TTTA)(7) (66%), followed by (TTTA)(11) (30%), (TTTA)(12) (3%) and (TTTA)(13) (1%). ttta 29-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-27 20186154-4 2010 The distribution of CYP19A1 (TTTA)(n) polymorphisms was such that the most frequent allele was (TTTA)(7) (66%), followed by (TTTA)(11) (30%), (TTTA)(12) (3%) and (TTTA)(13) (1%). ttta 96-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-27 20186154-4 2010 The distribution of CYP19A1 (TTTA)(n) polymorphisms was such that the most frequent allele was (TTTA)(7) (66%), followed by (TTTA)(11) (30%), (TTTA)(12) (3%) and (TTTA)(13) (1%). ttta 96-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-27 20186154-4 2010 The distribution of CYP19A1 (TTTA)(n) polymorphisms was such that the most frequent allele was (TTTA)(7) (66%), followed by (TTTA)(11) (30%), (TTTA)(12) (3%) and (TTTA)(13) (1%). ttta 96-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-27 20186154-4 2010 The distribution of CYP19A1 (TTTA)(n) polymorphisms was such that the most frequent allele was (TTTA)(7) (66%), followed by (TTTA)(11) (30%), (TTTA)(12) (3%) and (TTTA)(13) (1%). ttta 96-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-27 19808095-7 2010 Hydrogen bond-forming interactions and tight packing hydrophobic side chains that complement the puckering of the steroid backbone provide the molecular basis for the exclusive androgenic specificity of aromatase. Hydrogen 0-8 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 203-212 19808095-7 2010 Hydrogen bond-forming interactions and tight packing hydrophobic side chains that complement the puckering of the steroid backbone provide the molecular basis for the exclusive androgenic specificity of aromatase. Steroids 114-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 203-212 19931613-10 2010 Real-time PCR analysis demonstrates that the sulfonanilide analogs decrease aromatase gene transcription in breast cells. sulfonanilide 45-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 19931613-11 2010 Furthermore, the sulfonanilide compounds selectively decrease aromatase gene expression in several breast cancer cells, without exhibiting cytotoxic or apoptotic effects at low micromole concentrations. sulfonanilide 17-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-71 19931613-12 2010 A ligand-based pharmacophore model for selective aromatase modulation (SAM) by the novel sulfonanilides identified an aromatic ring, two hydrogen bond acceptors, and a hydrophobic function as four key chemical features. sulfonanilides 89-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-58 19931613-12 2010 A ligand-based pharmacophore model for selective aromatase modulation (SAM) by the novel sulfonanilides identified an aromatic ring, two hydrogen bond acceptors, and a hydrophobic function as four key chemical features. Hydrogen 137-145 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-58 19931613-15 2010 Thus, these results suggest that the novel sulfonanilides and the siRNAs targeting aromatase expression may be valuable tools for selective regulation of aromatase in breast cancer. sulfonanilides 43-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-163 19815003-4 2010 In both the normal and epileptic hippocampus, aromatase was detected in numerous CA1-CA3 pyramidal neurons, in granule cells of the dentate gyrus and in interneurons that co-expressed the calcium-binding proteins calbindin, calretinin or parvalbumin. Calcium 188-195 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-55 20144226-4 2010 We evaluate a series of polymorphisms of CYP19A1 and their effect on response to neoadjuvant letrozole in early BC. Letrozole 93-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-48 19360465-3 2010 Of the analyzed genes, ERCC2, XRCC1, XRCC2, XRCC3 and Lig4 participate in DNA repair, TP53 in cell cycle check point control, AIB1, AR, COMT, CYP11A1, CYP17A1, CYP19A1, HSD17 and PGR in steroid hormone biosynthesis/metabolism/signaling, TYMS in folate metabolism and HER2, IL6, LRP1, TGFB and TGFBR1 affect cell growth. Steroids 186-201 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 160-167 19308726-9 2010 Treatment with the histone deacetylase inhibitor trichostatin A (TSA) induced SULT1E1 and CYP19 mRNA but suppressed CYP1B1, STS, COMT, 17betaHSD1, and 17betaHSD2 mRNA in MCF10A lineage cell lines. trichostatin A 49-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-95 19308726-9 2010 Treatment with the histone deacetylase inhibitor trichostatin A (TSA) induced SULT1E1 and CYP19 mRNA but suppressed CYP1B1, STS, COMT, 17betaHSD1, and 17betaHSD2 mRNA in MCF10A lineage cell lines. trichostatin A 65-68 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-95 19308726-10 2010 In MCF7 cells, TSA treatment suppressed ERalpha, CYP1B1, STS, COMT, SULT1A1, and SULT2B1 but induced ERbeta, CYP19 and SULT2A1 mRNA expression. trichostatin A 15-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 109-114 19703265-12 2010 Behavior pattern differences observed between aromatase expression, present in normal tissues and absent in meningiomas, and estrogen and androgen hormone receptors, absent in normal tissues and present in meningiomas, suggest that there is heterogeneity in modulation by sex steroids in the development of these tumors. Steroids 276-284 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-55 20702940-3 2010 The reaction with NH(3) in the DRC system provides an eligible technique to determine Cr, because of a greater improvement in the signal/noise (S/N) ratio due to an effective elimination of interferences arising from Ar (ArC, ArN and ArO), and makes it possible to analyze Cr even at sub-microg L(-1) levels. Chromium 86-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 234-237 19360465-3 2010 Of the analyzed genes, ERCC2, XRCC1, XRCC2, XRCC3 and Lig4 participate in DNA repair, TP53 in cell cycle check point control, AIB1, AR, COMT, CYP11A1, CYP17A1, CYP19A1, HSD17 and PGR in steroid hormone biosynthesis/metabolism/signaling, TYMS in folate metabolism and HER2, IL6, LRP1, TGFB and TGFBR1 affect cell growth. Folic Acid 245-251 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 160-167 20205115-1 2010 Aromatase is a member of the cytochrome P450 superfamily that catalyzes the conversion of androgens (C(19)), namely testosterone and androstenedione, into oestrogens (C(18)), oestradiol, and oestrone, respectively. Testosterone 116-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 19906814-0 2010 Tissue-selective regulation of aromatase expression by calcitriol: implications for breast cancer therapy. Calcitriol 55-65 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 19906814-2 2010 We show that calcitriol, the hormonally active form of vitamin D, regulates the expression of aromatase in a tissue-selective manner. Calcitriol 13-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-103 19906814-2 2010 We show that calcitriol, the hormonally active form of vitamin D, regulates the expression of aromatase in a tissue-selective manner. Vitamin D 55-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-103 19906814-3 2010 Calcitriol significantly decreased aromatase expression in human BCa cells and adipocytes and caused substantial increases in human osteosarcoma cells (a bone cell model exhibiting osteoblast phenotype in culture) and modest increases in ovarian cancer cells. Calcitriol 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 35-44 19906814-4 2010 Calcitriol administration to immunocompromised mice bearing human BCa xenografts decreased aromatase mRNA levels in the tumors and the surrounding mammary adipose tissue but did not alter ovarian aromatase expression. Calcitriol 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-100 19906814-5 2010 In BCa cells, calcitriol also reduced the levels of prostaglandins (PGs), major stimulators of aromatase transcription, by suppressing the expression of cyclooxygenase-2 (which catalyzes PG synthesis) and increasing that of 15-hydroxyprostaglandin dehydrogenase (which catalyzes PG degradation). Calcitriol 14-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-104 19906814-5 2010 In BCa cells, calcitriol also reduced the levels of prostaglandins (PGs), major stimulators of aromatase transcription, by suppressing the expression of cyclooxygenase-2 (which catalyzes PG synthesis) and increasing that of 15-hydroxyprostaglandin dehydrogenase (which catalyzes PG degradation). Prostaglandins 52-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-104 19906814-5 2010 In BCa cells, calcitriol also reduced the levels of prostaglandins (PGs), major stimulators of aromatase transcription, by suppressing the expression of cyclooxygenase-2 (which catalyzes PG synthesis) and increasing that of 15-hydroxyprostaglandin dehydrogenase (which catalyzes PG degradation). Prostaglandins 68-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-104 20205115-1 2010 Aromatase is a member of the cytochrome P450 superfamily that catalyzes the conversion of androgens (C(19)), namely testosterone and androstenedione, into oestrogens (C(18)), oestradiol, and oestrone, respectively. Androstenedione 133-148 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 19906814-5 2010 In BCa cells, calcitriol also reduced the levels of prostaglandins (PGs), major stimulators of aromatase transcription, by suppressing the expression of cyclooxygenase-2 (which catalyzes PG synthesis) and increasing that of 15-hydroxyprostaglandin dehydrogenase (which catalyzes PG degradation). Prostaglandins 68-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-104 20205115-1 2010 Aromatase is a member of the cytochrome P450 superfamily that catalyzes the conversion of androgens (C(19)), namely testosterone and androstenedione, into oestrogens (C(18)), oestradiol, and oestrone, respectively. Estradiol 175-185 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 19906814-6 2010 The mechanism of aromatase down-regulation by calcitriol in BCa cells is therefore 2-fold: a direct repression of aromatase transcription via promoter II through the vitamin D-response elements identified in this promoter and an indirect suppression by reducing the levels of PGs. Calcitriol 46-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 19906814-6 2010 The mechanism of aromatase down-regulation by calcitriol in BCa cells is therefore 2-fold: a direct repression of aromatase transcription via promoter II through the vitamin D-response elements identified in this promoter and an indirect suppression by reducing the levels of PGs. Calcitriol 46-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 114-123 20205115-1 2010 Aromatase is a member of the cytochrome P450 superfamily that catalyzes the conversion of androgens (C(19)), namely testosterone and androstenedione, into oestrogens (C(18)), oestradiol, and oestrone, respectively. Estrone 191-199 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 19906814-6 2010 The mechanism of aromatase down-regulation by calcitriol in BCa cells is therefore 2-fold: a direct repression of aromatase transcription via promoter II through the vitamin D-response elements identified in this promoter and an indirect suppression by reducing the levels of PGs. Vitamin D 166-175 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 19906814-6 2010 The mechanism of aromatase down-regulation by calcitriol in BCa cells is therefore 2-fold: a direct repression of aromatase transcription via promoter II through the vitamin D-response elements identified in this promoter and an indirect suppression by reducing the levels of PGs. Prostaglandins 276-279 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 20133979-11 2010 The risk possesed by the TC varient of CYP19 was reduced when evaluated with A1, the protective allele of CYP17 (p=0.082). Technetium 25-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-44 19906814-9 2010 In addition to augmenting the ability of AIs to inhibit BCa growth, calcitriol acting as a selective aromatase modulator that increases aromatase expression in bone would reduce the estrogen deprivation in bone caused by the AIs, thus ameliorating the AI-induced side effect of osteoporosis. Calcitriol 68-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-110 19906814-9 2010 In addition to augmenting the ability of AIs to inhibit BCa growth, calcitriol acting as a selective aromatase modulator that increases aromatase expression in bone would reduce the estrogen deprivation in bone caused by the AIs, thus ameliorating the AI-induced side effect of osteoporosis. Calcitriol 68-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-145 25961968-4 2010 Furthermore, the human gene encoding aromatase, the enzyme that synthesizes estradiol, is under the control of different tissue-specific promoters, and it is therefore conceivable that selective aromatase modulators can be developed that will enhance the expression of the enzyme and the consequent increase in estrogen formation in the brain but not in other tissues. Estradiol 76-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-46 25961968-4 2010 Furthermore, the human gene encoding aromatase, the enzyme that synthesizes estradiol, is under the control of different tissue-specific promoters, and it is therefore conceivable that selective aromatase modulators can be developed that will enhance the expression of the enzyme and the consequent increase in estrogen formation in the brain but not in other tissues. Estradiol 76-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 195-204 19818337-1 2010 BACKGROUND: Single nucleotide polymorphism (SNP) rs2470152 of the gene CYP19A1 is associated with serum estradiol (E2) levels in Caucasian men. Estradiol 104-113 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-78 19818337-3 2010 We determined whether 2 CYP19A1 SNPs tagging different haploblocks (rs2470152 and rs2899470) are associated with sex steroid levels in Chinese men. Steroids 117-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-31 20588246-0 2010 Evaluation of the association between the CYP19 Tetranucleotide (TTTA)n polymorphism and polycystic ovarian syndrome(PCOS) in Han Chinese women. tetranucleotide 48-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-47 20574914-10 2010 In addition, PCB 126 upregulated CYP19, 3beta-HSD2, StAR, and HMGR mRNA levels at the highest concentration tested, and downregulated CYP21 at 40 nM. Polychlorinated Biphenyls 13-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-38 20588246-0 2010 Evaluation of the association between the CYP19 Tetranucleotide (TTTA)n polymorphism and polycystic ovarian syndrome(PCOS) in Han Chinese women. ttta 65-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-47 20588246-2 2010 The present study aims to evaluate the association between tetranucleotide TTTA repeat polymorphism in the CYP19 gene and PCOS among Han Chinese women. tetranucleotide 59-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-112 20588246-4 2010 The CYP19 tetranucleotide TTTA repeat polymorphism was genotyped with a protocol of PCR and fluorescent capillary electrophoresis. tetranucleotide ttta 10-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-9 20588246-5 2010 RESULTS: Common allele of the CYP19 tetranucleotide TTTA repeat polymorphism in this population of Han Chinese women was 11R. tetranucleotide 36-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-35 20588246-9 2010 CONCLUSIONS: The most common allele of the tetranucleotide TTTA repeat polymorphism in the forth intron of CYP19 gene in Han Chinese women is 11R, which was different with the previous study in European Caucasians. tetranucleotide 43-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-112 20588246-9 2010 CONCLUSIONS: The most common allele of the tetranucleotide TTTA repeat polymorphism in the forth intron of CYP19 gene in Han Chinese women is 11R, which was different with the previous study in European Caucasians. ttta 59-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-112 20588246-12 2010 This CYP19 tetranucleotide TTTA repeat polymorphism is an ethnic and racial variant and moderately contributes to the pathogenesis of PCOS in the population of Han Chinese women. tetranucleotide 11-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 5-10 20003221-9 2009 The CYP19A1 mRNA level at exposure to rosiglitazone alone showed a drop, but was not significantly reduced comparing to control. Rosiglitazone 38-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-11 19679108-8 2009 Human placental microsomal CYP19/aromatase was the major isozyme responsible for the biotransformation of glyburide to predominantly M5. Glyburide 106-115 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-32 19820017-3 2009 OBJECTIVE: The objective of the study was to assess the effects of aromatase inhibition on BMD in older men with low testosterone levels. Testosterone 117-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-76 19822197-0 2009 Mono-(2-ethylhexyl) phthalate induces NR4A subfamily and GIOT-1 gene expression, and suppresses CYP19 expression in human granulosa-like tumor cell line KGN. mono-(2-ethylhexyl)phthalate 0-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-101 19822197-1 2009 The mechanism for transcriptional suppression of CYP19 by mono-ethylhexyl phthalate (MEHP) in a human ovarian granulosa cell line (KGN) was investigated. mono-(2-ethylhexyl)phthalate 58-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-54 19822197-1 2009 The mechanism for transcriptional suppression of CYP19 by mono-ethylhexyl phthalate (MEHP) in a human ovarian granulosa cell line (KGN) was investigated. mono-(2-ethylhexyl)phthalate 85-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-54 19822197-3 2009 MEHP was found to significantly suppress Forskolin (FSK)-induced CYP19 gene transcription, CYP19 promoter II activity and CYP19 enzyme activity in a dose-dependent manner. mono-(2-ethylhexyl)phthalate 0-4 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-70 19822197-3 2009 MEHP was found to significantly suppress Forskolin (FSK)-induced CYP19 gene transcription, CYP19 promoter II activity and CYP19 enzyme activity in a dose-dependent manner. mono-(2-ethylhexyl)phthalate 0-4 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-96 19822197-3 2009 MEHP was found to significantly suppress Forskolin (FSK)-induced CYP19 gene transcription, CYP19 promoter II activity and CYP19 enzyme activity in a dose-dependent manner. mono-(2-ethylhexyl)phthalate 0-4 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-96 19822197-3 2009 MEHP was found to significantly suppress Forskolin (FSK)-induced CYP19 gene transcription, CYP19 promoter II activity and CYP19 enzyme activity in a dose-dependent manner. Colforsin 41-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-70 19822197-3 2009 MEHP was found to significantly suppress Forskolin (FSK)-induced CYP19 gene transcription, CYP19 promoter II activity and CYP19 enzyme activity in a dose-dependent manner. Colforsin 41-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-96 19822197-3 2009 MEHP was found to significantly suppress Forskolin (FSK)-induced CYP19 gene transcription, CYP19 promoter II activity and CYP19 enzyme activity in a dose-dependent manner. Colforsin 41-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-96 19822197-3 2009 MEHP was found to significantly suppress Forskolin (FSK)-induced CYP19 gene transcription, CYP19 promoter II activity and CYP19 enzyme activity in a dose-dependent manner. Colforsin 52-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-70 19822197-3 2009 MEHP was found to significantly suppress Forskolin (FSK)-induced CYP19 gene transcription, CYP19 promoter II activity and CYP19 enzyme activity in a dose-dependent manner. Colforsin 52-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-96 19822197-3 2009 MEHP was found to significantly suppress Forskolin (FSK)-induced CYP19 gene transcription, CYP19 promoter II activity and CYP19 enzyme activity in a dose-dependent manner. Colforsin 52-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-96 19854050-0 2009 Lead optimization of COX-2 inhibitor nimesulide analogs to overcome aromatase inhibitor resistance in breast cancer cells. nimesulide 37-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-77 19820017-13 2009 CONCLUSIONS: In older men, aromatase inhibition increases testosterone levels, decreases estradiol levels, and appears to decrease BMD. Testosterone 58-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-36 19820017-13 2009 CONCLUSIONS: In older men, aromatase inhibition increases testosterone levels, decreases estradiol levels, and appears to decrease BMD. Estradiol 89-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-36 20112494-1 2009 OBJECTIVE: To investigate the effect of yikun neiyi wan (YKNYW) and gestrinone on the expression of aromatase P450 (P450arom), cyclo-oxygenase-2 (COX-2) and estrogen receptor (ER) in isolated ectopic and normal endometrial stroma cells in vitro. Gestrinone 68-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 110-114 20112494-1 2009 OBJECTIVE: To investigate the effect of yikun neiyi wan (YKNYW) and gestrinone on the expression of aromatase P450 (P450arom), cyclo-oxygenase-2 (COX-2) and estrogen receptor (ER) in isolated ectopic and normal endometrial stroma cells in vitro. Gestrinone 68-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-124 20112494-5 2009 RESULTS: The expression levels of P450arom, COX-2 in glandular epithelium cells in vitro were decreased significantly by YKNYW compared with gestrinone (P < 0.05). Gestrinone 141-151 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-42 19930708-3 2009 In the current study we investigated the activity of the aromatase inhibitor exemestane in human NSCLC cell lines H23 and A549. exemestane 77-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-66 19484778-1 2009 Prostate require high levels of steroidogenic enzymes such as 5alpha-reductase (5alpha-r) and Aromatase (Aro) for the formation of active steroids. Steroids 138-146 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-103 19484778-1 2009 Prostate require high levels of steroidogenic enzymes such as 5alpha-reductase (5alpha-r) and Aromatase (Aro) for the formation of active steroids. Steroids 138-146 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-97 19484778-2 2009 Dihydrotestosterone (DHT), the prostate dominant androgen, is converted from testosterone (T) by the action of 5alpha-r. Aro provides an alternative pathway for estrogen, via T aromatization. Dihydrotestosterone 0-19 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 121-124 19484778-2 2009 Dihydrotestosterone (DHT), the prostate dominant androgen, is converted from testosterone (T) by the action of 5alpha-r. Aro provides an alternative pathway for estrogen, via T aromatization. Dihydrotestosterone 21-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 121-124 19484778-2 2009 Dihydrotestosterone (DHT), the prostate dominant androgen, is converted from testosterone (T) by the action of 5alpha-r. Aro provides an alternative pathway for estrogen, via T aromatization. Testosterone 7-19 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 121-124 19484778-4 2009 Data obtained 1 day after the 30 consecutive days of enzymatic inhibition with Finasteride (5alpha-r inhibitor) and Letrozole (Aro inhibitor) demonstrated a marked stromal remodeling, with an increased deposition of extracellular matrix (ECM) proteins besides androgen receptor (AR) overexpression in the three phases of postnatal development analyzed. Letrozole 116-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-130 19930708-14 2009 Exemestane may be more effective in cells with higher aromatase levels. exemestane 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 54-63 19861537-3 2009 We examined whether the heat shock protein 90 (HSP90) inhibitor 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin (17-DMAG) can inhibit the growth of aromatase inhibitor-resistant breast cancers and the mechanisms by which 17-DMAG affects proliferation. 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin 64-117 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-163 19861537-3 2009 We examined whether the heat shock protein 90 (HSP90) inhibitor 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin (17-DMAG) can inhibit the growth of aromatase inhibitor-resistant breast cancers and the mechanisms by which 17-DMAG affects proliferation. 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin 119-126 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-163 19559719-0 2009 Design and synthesis of carbon-11-labeled dual aromatase-steroid sulfatase inhibitors as new potential PET agents for imaging of aromatase and steroid sulfatase expression in breast cancer. Carbon-11 24-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 19559719-0 2009 Design and synthesis of carbon-11-labeled dual aromatase-steroid sulfatase inhibitors as new potential PET agents for imaging of aromatase and steroid sulfatase expression in breast cancer. Carbon-11 24-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-138 19559719-2 2009 New carbon-11-labeled sulfamate derivatives were first designed and synthesized as potential PET dual aromatase-steroid sulfatase inhibitor (DASSI) radiotracers for imaging of aromatase and STS expression in breast cancer. Carbon-11 4-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 102-111 19559719-2 2009 New carbon-11-labeled sulfamate derivatives were first designed and synthesized as potential PET dual aromatase-steroid sulfatase inhibitor (DASSI) radiotracers for imaging of aromatase and STS expression in breast cancer. Carbon-11 4-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 176-185 19559719-2 2009 New carbon-11-labeled sulfamate derivatives were first designed and synthesized as potential PET dual aromatase-steroid sulfatase inhibitor (DASSI) radiotracers for imaging of aromatase and STS expression in breast cancer. sulfamic acid 22-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 102-111 19559719-2 2009 New carbon-11-labeled sulfamate derivatives were first designed and synthesized as potential PET dual aromatase-steroid sulfatase inhibitor (DASSI) radiotracers for imaging of aromatase and STS expression in breast cancer. sulfamic acid 22-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 176-185 19500885-2 2009 Aromatase inhibition is usually achieved with steroids structurally related to the substrate of catalysis or, alternatively, with azole non-steroid compounds. Steroids 46-54 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 19739633-1 2009 The synthesis of a potentially redox active tripodal ligand containing a tris(aryloxide) functionalized mesitylene anchor, (((tBu)ArOH)(3)mes) (1), and its metalation with low-valent uranium to form [(((tBu)ArO)(3)mes)U] (1-U) is reported. Tromethamine 73-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-133 19739633-1 2009 The synthesis of a potentially redox active tripodal ligand containing a tris(aryloxide) functionalized mesitylene anchor, (((tBu)ArOH)(3)mes) (1), and its metalation with low-valent uranium to form [(((tBu)ArO)(3)mes)U] (1-U) is reported. 2-(N-morpholino)ethanesulfonic acid 138-141 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-133 19739633-3 2009 Comparison to the previously synthesized complex, [(((tBu)ArO)(3)tacn)U] (2-U), featuring the redox-innocent triazacyclononane anchor reveals that changing the anchor from the flexible triazacyclononane to a rigid mesityl fragment increases the structural flexibility of the aryloxide substituents in complexes of 1. triazacyclononane 109-126 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-61 19739633-3 2009 Comparison to the previously synthesized complex, [(((tBu)ArO)(3)tacn)U] (2-U), featuring the redox-innocent triazacyclononane anchor reveals that changing the anchor from the flexible triazacyclononane to a rigid mesityl fragment increases the structural flexibility of the aryloxide substituents in complexes of 1. triazacyclononane 185-202 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-61 19739633-3 2009 Comparison to the previously synthesized complex, [(((tBu)ArO)(3)tacn)U] (2-U), featuring the redox-innocent triazacyclononane anchor reveals that changing the anchor from the flexible triazacyclononane to a rigid mesityl fragment increases the structural flexibility of the aryloxide substituents in complexes of 1. aryloxide 275-284 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-61 19754085-1 2009 A kinetic study of the aroxyl (ArO*) radical-scavenging reaction of alpha-tocopherol (alpha-TocH) has been performed in the presence of six kinds of alkali and alkaline earth metal salts (LiI, LiClO(4), NaI, NaClO(4), KI, and Mg(ClO(4))(2)) in methanol solution, using stopped-flow spectrophotometry. alpha-Tocopherol 68-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-35 19754085-1 2009 A kinetic study of the aroxyl (ArO*) radical-scavenging reaction of alpha-tocopherol (alpha-TocH) has been performed in the presence of six kinds of alkali and alkaline earth metal salts (LiI, LiClO(4), NaI, NaClO(4), KI, and Mg(ClO(4))(2)) in methanol solution, using stopped-flow spectrophotometry. alpha-Tocopherol 86-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-35 19754085-1 2009 A kinetic study of the aroxyl (ArO*) radical-scavenging reaction of alpha-tocopherol (alpha-TocH) has been performed in the presence of six kinds of alkali and alkaline earth metal salts (LiI, LiClO(4), NaI, NaClO(4), KI, and Mg(ClO(4))(2)) in methanol solution, using stopped-flow spectrophotometry. Metals 175-180 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-35 19754085-1 2009 A kinetic study of the aroxyl (ArO*) radical-scavenging reaction of alpha-tocopherol (alpha-TocH) has been performed in the presence of six kinds of alkali and alkaline earth metal salts (LiI, LiClO(4), NaI, NaClO(4), KI, and Mg(ClO(4))(2)) in methanol solution, using stopped-flow spectrophotometry. lii 188-191 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-35 19754085-1 2009 A kinetic study of the aroxyl (ArO*) radical-scavenging reaction of alpha-tocopherol (alpha-TocH) has been performed in the presence of six kinds of alkali and alkaline earth metal salts (LiI, LiClO(4), NaI, NaClO(4), KI, and Mg(ClO(4))(2)) in methanol solution, using stopped-flow spectrophotometry. liclo 193-198 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-35 19754085-1 2009 A kinetic study of the aroxyl (ArO*) radical-scavenging reaction of alpha-tocopherol (alpha-TocH) has been performed in the presence of six kinds of alkali and alkaline earth metal salts (LiI, LiClO(4), NaI, NaClO(4), KI, and Mg(ClO(4))(2)) in methanol solution, using stopped-flow spectrophotometry. Sodium Iodide 203-206 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-35 19754085-1 2009 A kinetic study of the aroxyl (ArO*) radical-scavenging reaction of alpha-tocopherol (alpha-TocH) has been performed in the presence of six kinds of alkali and alkaline earth metal salts (LiI, LiClO(4), NaI, NaClO(4), KI, and Mg(ClO(4))(2)) in methanol solution, using stopped-flow spectrophotometry. Sodium Hypochlorite 208-213 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-35 19754085-1 2009 A kinetic study of the aroxyl (ArO*) radical-scavenging reaction of alpha-tocopherol (alpha-TocH) has been performed in the presence of six kinds of alkali and alkaline earth metal salts (LiI, LiClO(4), NaI, NaClO(4), KI, and Mg(ClO(4))(2)) in methanol solution, using stopped-flow spectrophotometry. Magnesium 226-228 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-35 19754085-1 2009 A kinetic study of the aroxyl (ArO*) radical-scavenging reaction of alpha-tocopherol (alpha-TocH) has been performed in the presence of six kinds of alkali and alkaline earth metal salts (LiI, LiClO(4), NaI, NaClO(4), KI, and Mg(ClO(4))(2)) in methanol solution, using stopped-flow spectrophotometry. clo 195-198 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-35 19754085-1 2009 A kinetic study of the aroxyl (ArO*) radical-scavenging reaction of alpha-tocopherol (alpha-TocH) has been performed in the presence of six kinds of alkali and alkaline earth metal salts (LiI, LiClO(4), NaI, NaClO(4), KI, and Mg(ClO(4))(2)) in methanol solution, using stopped-flow spectrophotometry. Methanol 244-252 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-35 19754085-2 2009 The decay rate of the ArO* for the reaction of alpha-TocH with ArO* increased linearly with increasing concentration of metal salts. Metals 120-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 22-26 19754085-2 2009 The decay rate of the ArO* for the reaction of alpha-TocH with ArO* increased linearly with increasing concentration of metal salts. Metals 120-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-67 19754085-3 2009 The second-order rate constants (k(s)) for the reaction of alpha-TocH with ArO* increased in the order of no metal salt < KI approximately NaClO(4) approximately NaI <or= LiClO(4) < Mg(ClO(4))(2) < LiI at the same concentration of metal salts. metal salt 109-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-79 19754085-3 2009 The second-order rate constants (k(s)) for the reaction of alpha-TocH with ArO* increased in the order of no metal salt < KI approximately NaClO(4) approximately NaI <or= LiClO(4) < Mg(ClO(4))(2) < LiI at the same concentration of metal salts. Sodium Hypochlorite 142-147 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-79 19754085-3 2009 The second-order rate constants (k(s)) for the reaction of alpha-TocH with ArO* increased in the order of no metal salt < KI approximately NaClO(4) approximately NaI <or= LiClO(4) < Mg(ClO(4))(2) < LiI at the same concentration of metal salts. Sodium Iodide 165-168 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-79 19754085-3 2009 The second-order rate constants (k(s)) for the reaction of alpha-TocH with ArO* increased in the order of no metal salt < KI approximately NaClO(4) approximately NaI <or= LiClO(4) < Mg(ClO(4))(2) < LiI at the same concentration of metal salts. liclo 177-182 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-79 19754085-3 2009 The second-order rate constants (k(s)) for the reaction of alpha-TocH with ArO* increased in the order of no metal salt < KI approximately NaClO(4) approximately NaI <or= LiClO(4) < Mg(ClO(4))(2) < LiI at the same concentration of metal salts. Magnesium 191-193 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-79 19754085-3 2009 The second-order rate constants (k(s)) for the reaction of alpha-TocH with ArO* increased in the order of no metal salt < KI approximately NaClO(4) approximately NaI <or= LiClO(4) < Mg(ClO(4))(2) < LiI at the same concentration of metal salts. clo 144-147 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-79 19754085-3 2009 The second-order rate constants (k(s)) for the reaction of alpha-TocH with ArO* increased in the order of no metal salt < KI approximately NaClO(4) approximately NaI <or= LiClO(4) < Mg(ClO(4))(2) < LiI at the same concentration of metal salts. Metals 109-114 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-79 19754085-6 2009 Effects of metal cations on the UV-vis absorption spectra of the alpha-Toc* (and ArO*) radical were negligible in methanol solution, suggesting that the complex formation between the alpha-Toc* (and ArO*) radical molecule and metal cations is hindered by the hydrogen bond between radical and methanol molecules. Metals 11-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-85 19754085-6 2009 Effects of metal cations on the UV-vis absorption spectra of the alpha-Toc* (and ArO*) radical were negligible in methanol solution, suggesting that the complex formation between the alpha-Toc* (and ArO*) radical molecule and metal cations is hindered by the hydrogen bond between radical and methanol molecules. Metals 11-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 199-203 19754085-6 2009 Effects of metal cations on the UV-vis absorption spectra of the alpha-Toc* (and ArO*) radical were negligible in methanol solution, suggesting that the complex formation between the alpha-Toc* (and ArO*) radical molecule and metal cations is hindered by the hydrogen bond between radical and methanol molecules. Methanol 114-122 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-85 19754085-6 2009 Effects of metal cations on the UV-vis absorption spectra of the alpha-Toc* (and ArO*) radical were negligible in methanol solution, suggesting that the complex formation between the alpha-Toc* (and ArO*) radical molecule and metal cations is hindered by the hydrogen bond between radical and methanol molecules. Hydrogen 259-267 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-85 19754085-6 2009 Effects of metal cations on the UV-vis absorption spectra of the alpha-Toc* (and ArO*) radical were negligible in methanol solution, suggesting that the complex formation between the alpha-Toc* (and ArO*) radical molecule and metal cations is hindered by the hydrogen bond between radical and methanol molecules. Hydrogen 259-267 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 199-203 19754085-6 2009 Effects of metal cations on the UV-vis absorption spectra of the alpha-Toc* (and ArO*) radical were negligible in methanol solution, suggesting that the complex formation between the alpha-Toc* (and ArO*) radical molecule and metal cations is hindered by the hydrogen bond between radical and methanol molecules. radical 87-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-85 19754085-6 2009 Effects of metal cations on the UV-vis absorption spectra of the alpha-Toc* (and ArO*) radical were negligible in methanol solution, suggesting that the complex formation between the alpha-Toc* (and ArO*) radical molecule and metal cations is hindered by the hydrogen bond between radical and methanol molecules. radical 87-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 199-203 19754085-6 2009 Effects of metal cations on the UV-vis absorption spectra of the alpha-Toc* (and ArO*) radical were negligible in methanol solution, suggesting that the complex formation between the alpha-Toc* (and ArO*) radical molecule and metal cations is hindered by the hydrogen bond between radical and methanol molecules. Methanol 293-301 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-85 19754085-6 2009 Effects of metal cations on the UV-vis absorption spectra of the alpha-Toc* (and ArO*) radical were negligible in methanol solution, suggesting that the complex formation between the alpha-Toc* (and ArO*) radical molecule and metal cations is hindered by the hydrogen bond between radical and methanol molecules. Methanol 293-301 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 199-203 19754085-7 2009 The results indicate that the hydrogen transfer reaction of alpha-TocH proceeds via an electron transfer intermediate from alpha-TocH to ArO* radicals followed by proton transfer. Hydrogen 30-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 137-141 19754085-7 2009 The results indicate that the hydrogen transfer reaction of alpha-TocH proceeds via an electron transfer intermediate from alpha-TocH to ArO* radicals followed by proton transfer. alpha-Tocopherol 60-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 137-141 19754085-7 2009 The results indicate that the hydrogen transfer reaction of alpha-TocH proceeds via an electron transfer intermediate from alpha-TocH to ArO* radicals followed by proton transfer. alpha-Tocopherol 123-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 137-141 19754085-8 2009 Both the coordinations of metal cations to the one-electron reduced anions of ArO* (ArO: (-)) and of counteranions to the one-electron oxidized cations of alpha-TocH (alpha-TocH(+)*) may stabilize the intermediate, resulting in the acceleration of electron transfer. Metals 26-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-82 19789370-3 2009 Here we report on findings from a comprehensive study of genetic variation at the CYP19A1 locus in relation to prostate cancer risk and to circulating steroid hormone concentrations in men by the Breast and Prostate Cancer Cohort Consortium (BPC3), a large collaborative prospective study. Steroids 151-166 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-89 19500885-0 2009 An efficient steroid pharmacophore-based strategy to identify new aromatase inhibitors. Steroids 13-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-75 19500885-2 2009 Aromatase inhibition is usually achieved with steroids structurally related to the substrate of catalysis or, alternatively, with azole non-steroid compounds. Azoles 130-135 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 19500885-2 2009 Aromatase inhibition is usually achieved with steroids structurally related to the substrate of catalysis or, alternatively, with azole non-steroid compounds. Steroids 46-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 19500885-3 2009 Substituted androstenedione derivatives with Delta(1), Delta(6) and Delta(1,6) unsaturations and 6-alkyl/6-phenyl aliphatic substitutions, are among the most potent steroid aromatase inhibitors known to date. Androstenedione 12-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 173-182 19539015-0 2009 Bisphenol A downregulates CYP19 transcription in JEG-3 cells. bisphenol A 0-11 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-31 19556341-2 2009 Here, we show that 17beta-estradiol (E2) can rapidly enhance aromatase enzymatic activity through an increase of aromatase protein phosphorylation in breast cancer cell lines. Estradiol 19-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-70 19556341-2 2009 Here, we show that 17beta-estradiol (E2) can rapidly enhance aromatase enzymatic activity through an increase of aromatase protein phosphorylation in breast cancer cell lines. Estradiol 19-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-122 19556341-3 2009 In vivo labeling experiments and site-directed mutagenesis studies demonstrated that phosphorylation of the 361-tyrosine residue is crucial in the up-regulation of aromatase activity under E2 exposure. Tyrosine 112-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 164-173 19556341-6 2009 These findings show, for the first time, that tyrosine phosphorylation processes play a key role in the rapid changes induced by E2 in aromatase enzymatic activity, revealing the existence of a short nongenomic autocrine loop between E2 and aromatase in breast cancer cells. Tyrosine 46-54 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 19539015-6 2009 Cells treated with bisphenol A displayed a reduced aromatase activity. bisphenol A 19-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 51-60 19591804-0 2009 The critical iron-oxygen intermediate in human aromatase. Iron 13-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 19560483-9 2009 Disruptions of steroidogenesis related-gene expression such as Star, Cyp19a1, Hsd17b8, and Nr4a3 were observed in the DTPE group, but not in the DNTPE group. (Ethane-1,2-diyl)bis[bis(4-methylphenyl)phosphane] 118-122 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-76 19591804-0 2009 The critical iron-oxygen intermediate in human aromatase. Oxygen 18-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 19591804-2 2009 To better understand the CYP19 reaction, the oxy-ferrous complex of CYP19 with androstenedione substrate was cryotrapped, characterized by UV-vis spectroscopy, and cryoreduced to generate the next reaction cycle intermediate. ammonium ferrous sulfate 49-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-30 19591804-2 2009 To better understand the CYP19 reaction, the oxy-ferrous complex of CYP19 with androstenedione substrate was cryotrapped, characterized by UV-vis spectroscopy, and cryoreduced to generate the next reaction cycle intermediate. ammonium ferrous sulfate 49-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-73 19591804-2 2009 To better understand the CYP19 reaction, the oxy-ferrous complex of CYP19 with androstenedione substrate was cryotrapped, characterized by UV-vis spectroscopy, and cryoreduced to generate the next reaction cycle intermediate. Androstenedione 79-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-30 19591804-2 2009 To better understand the CYP19 reaction, the oxy-ferrous complex of CYP19 with androstenedione substrate was cryotrapped, characterized by UV-vis spectroscopy, and cryoreduced to generate the next reaction cycle intermediate. Androstenedione 79-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-73 19505075-7 2009 Comparatively, oxidative addition of ArO-Ac to Ni(0) is a more facile process (barrier = +14.2 kcal/mol) than oxidative addition of Ar-OAc to Ni(0). ar-oac 132-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-40 19520161-1 2009 To gain the structure-activity relationship of Delta(1)-androstenediones (Delta(1)-ADs) as mechanism-based inactivator of aromatase, series of 2-alkyl- and 2-alkoxy-substituted Delta(1)-ADs (6 and 9) as well as 2-bromo-Delta(1)-AD (14) were synthesized and tested. delta(1)-androstenediones 47-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-131 19520161-5 2009 All of the alkyl steroids 6 along with the alkoxy steroid 9, except for the ethyl and n-propyl compounds 6b and 6c, caused a time-dependent inactivation of aromatase. alkyl steroids 11-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 156-165 19520161-5 2009 All of the alkyl steroids 6 along with the alkoxy steroid 9, except for the ethyl and n-propyl compounds 6b and 6c, caused a time-dependent inactivation of aromatase. Steroids 17-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 156-165 19520161-5 2009 All of the alkyl steroids 6 along with the alkoxy steroid 9, except for the ethyl and n-propyl compounds 6b and 6c, caused a time-dependent inactivation of aromatase. ethyl and n-propyl compounds 76-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 156-165 19520161-8 2009 The results indicate that the 2-hexyl compound 6f act as the most powerful mechanism-based inactivator of aromatase among Delta(1)-AD analogs and may be submitted to the preclinical study in estrogen-dependent breast cancer. 2-hexyl 30-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 106-115 19639193-9 2009 These results suggest that 3-bp I/D polymorphisms of the CYP19 gene may be associated with breast cancer and that the (TTTA)n repeat genotype would be useful in selecting candidates for tamoxifen therapy, as well as predicting breast cancer risk in Korean women. Tamoxifen 186-195 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-62 19661043-2 2009 Herein, we report deterioration of visual acuity in 2 Chinese patients with breast cancer and preexisting myopia who had recently commenced adjuvant hormonal therapy using the aromatase inhibitor exemestane. exemestane 196-206 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 176-185 19745796-9 2009 Aromatase expression, a possible intrinsic survival factor for endometrial tissue, is inducible in human endometrial fragments by androstenedione at physiological concentrations found in peritoneal fluid. Androstenedione 130-145 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 19278756-0 2009 CoMFA, LeapFrog and blind docking studies on sulfonanilide derivatives acting as selective aromatase expression regulators. sulfonanilide 45-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-100 19278756-2 2009 In this manuscript, the structure-based drug design approach of sulfonanilide analogues as potential selective aromatase expression regulators (SAERs) is described. sulfonanilide 64-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-120 18941892-2 2009 METHODS: A validated immunohistochemical assay for aromatase was applied to samples from the P024 neoadjuvant endocrine therapy trial that compared tamoxifen and letrozole. Tamoxifen 148-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 51-60 18941892-2 2009 METHODS: A validated immunohistochemical assay for aromatase was applied to samples from the P024 neoadjuvant endocrine therapy trial that compared tamoxifen and letrozole. Letrozole 162-171 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 51-60 19598235-12 2009 In the sex steroid group, this included CYP17A1 and CYP19A1. Steroids 11-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-59 19435862-0 2009 Aromatase explains why testosterone increases breast cancer rate. Testosterone 23-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 19489632-6 2009 CYP3A4, CYP3A5, CYP3A7, and CYP19 (aromatase) inhibition to the log concentration of beta-acid content was significant with r(2) > 0.37, suggesting that these components can account for some of the variation in inhibition of CYP metabolism. 6-Hydroxy-2-oxo-1,2-dihydroquinoline-4-carboxylic acid 85-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-33 19389994-6 2009 The other approach initially sought to block adrenal function as a treatment for breast cancer but led to the serendipitous finding that a nonsteroidal P450 steroidogenesis inhibitor, aminoglutethimide, served as a potent but nonselective aromatase inhibitor. Aminoglutethimide 184-201 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 239-248 19389994-7 2009 Proof of the therapeutic concept of aromatase inhibition involved a variety of studies with aminoglutethimide and the selective steroidal inhibitor, formestane. Aminoglutethimide 92-109 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-45 19389994-7 2009 Proof of the therapeutic concept of aromatase inhibition involved a variety of studies with aminoglutethimide and the selective steroidal inhibitor, formestane. formestane 149-159 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-45 19299445-2 2009 In this study, trophoblast differentiation and associated induction of CYP19 expression were prevented when cytotrophoblasts were cultured in phenol red-free medium containing charcoal-stripped serum or with the estrogen receptor (ER) antagonist, ICI 182,780, suggesting a stimulatory role of estrogen/ER. Phenolsulfonphthalein 142-152 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-76 19428940-0 2009 Modulation of Delta9-tetrahydrocannabinol-induced MCF-7 breast cancer cell growth by cyclooxygenase and aromatase. Dronabinol 14-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 104-113 18602794-0 2009 Trichostatin A down-regulates CYP19 transcript and protein levels in MCF-7 breast cancer cells. trichostatin A 0-14 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-35 18602794-3 2009 Using quantitative real-time PCR and Western blot analysis, we demonstrated that trichostatin A (TSA) histone deacetylase inhibitor significantly reduced CYP19 transcript and protein contents in MCF-7 breast cancer cells. trichostatin A 81-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-159 18602794-3 2009 Using quantitative real-time PCR and Western blot analysis, we demonstrated that trichostatin A (TSA) histone deacetylase inhibitor significantly reduced CYP19 transcript and protein contents in MCF-7 breast cancer cells. trichostatin A 97-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-159 19428940-7 2009 However, the growth of MCF-7 cells induced by Delta(9)-THC was not stimulated by PGE(2), and the expression of aromatase was not affected by COX-2 inhibitors, arachidonic acid, and PGE(2), suggesting that there is a disconnection between COX-2 (PGE(2)) and aromatase in Delta(9)-THC-mediated MCF-7 cell proliferation. Dronabinol 270-282 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-120 19428940-8 2009 On the other hand, Delta(9)-THC-induced MCF-7 cell growth was elevated by two kinds of aromatase inhibitors. Dronabinol 19-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-96 19549443-6 2009 Del/Del homozygous patients with shorter TTTA repeats exhibited decreased ovarian FSH sensitivity in ovarian stimulation, which may reflect variations in aromatase gene expression during early antral follicle development. ttta 41-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-163 18817841-2 2009 After incubation with 4-androstene-3,17-dione (4-dione), there are no estrogens, estrone (E1) and estradiol (E2), detected suggesting that the pathway of aromatase is not important in these cell lines. Androstenedione 47-54 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-163 19420978-4 2009 The German Rectal Cancer study CAO/ARO/AIO-94 showed a full remission rate of 8% after a 5-fluorouracil (5-FU)based chemotherapy added to a conventional fractional radiation therapy (50.4 Gy). Fluorouracil 89-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 35-38 19420978-4 2009 The German Rectal Cancer study CAO/ARO/AIO-94 showed a full remission rate of 8% after a 5-fluorouracil (5-FU)based chemotherapy added to a conventional fractional radiation therapy (50.4 Gy). Fluorouracil 105-109 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 35-38 19116788-7 2009 The cell specific regulation of CYP19A1 by vitamin D, dexamethasone, and mifepristone opens the possibility for cellular selective modulation of estrogen biosynthesis within the brain. Vitamin D 43-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-39 19116788-7 2009 The cell specific regulation of CYP19A1 by vitamin D, dexamethasone, and mifepristone opens the possibility for cellular selective modulation of estrogen biosynthesis within the brain. Dexamethasone 54-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-39 19116788-7 2009 The cell specific regulation of CYP19A1 by vitamin D, dexamethasone, and mifepristone opens the possibility for cellular selective modulation of estrogen biosynthesis within the brain. Mifepristone 73-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-39 19026713-4 2009 Western immunoblotting demonstrated a marked induction of the CYP19 protein of characteristic size after only a short (6h) treatment period with VIP or forskolin. Colforsin 152-161 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-67 19250198-5 2009 Because of the membrane-bound character and heme-binding instability, no crystal structure of aromatase has been reported so far. Heme 44-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-103 18629629-7 2009 The CYP19 variant TC + TT genotypes were associated with both overall cancer risk (TT + TC vs. TT aOR = 1.73, 95% CI = 1.13-2.65) and premenopausal cancer risk (TT + TC vs. TT aOR = 1.78, 95% CI = 1.03-3.09), particularly for ER +/PR + tumors. Technetium 18-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-9 18629629-7 2009 The CYP19 variant TC + TT genotypes were associated with both overall cancer risk (TT + TC vs. TT aOR = 1.73, 95% CI = 1.13-2.65) and premenopausal cancer risk (TT + TC vs. TT aOR = 1.78, 95% CI = 1.03-3.09), particularly for ER +/PR + tumors. Technetium 88-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-9 19026713-3 2009 The expression of CYP19 transcripts and protein were markedly induced in the H295 adrenocortical carcinoma cell line after treatment with either forskolin or vasoactive intestinal peptide (VIP). Colforsin 145-154 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 18-23 19065667-5 2009 In the present study the licorice flavonoid isoliquiritigenin (ILN) was shown to be an aromatase inhibitor in recombinant protein and MCF-7 cells stably transfected with CYP19 (MCF-7aro). licorice flavonoid 25-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 170-175 19065667-5 2009 In the present study the licorice flavonoid isoliquiritigenin (ILN) was shown to be an aromatase inhibitor in recombinant protein and MCF-7 cells stably transfected with CYP19 (MCF-7aro). isoliquiritigenin 44-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 170-175 19065667-5 2009 In the present study the licorice flavonoid isoliquiritigenin (ILN) was shown to be an aromatase inhibitor in recombinant protein and MCF-7 cells stably transfected with CYP19 (MCF-7aro). isoliquiritigenin 63-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 170-175 19065667-10 2009 Subsequently, the flavonoid"s effect on CYP19 transcriptional control in vitro was also investigated. Flavonoids 18-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 40-45 19250204-2 2009 Following 24 and 72 h incubation of malignant human liver cell lines HepG2, HuH7, and HA22T cells with testosterone (T) used as androgen precursor, we observed an increasingly high proportion of T oxidation to androstenedione, with aromatase being the prevalent enzyme activity in HepG2 and HuH7 cells at 72 h, while no aromatase could be detected in HA22T cells. Testosterone 103-115 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 232-241 19250204-2 2009 Following 24 and 72 h incubation of malignant human liver cell lines HepG2, HuH7, and HA22T cells with testosterone (T) used as androgen precursor, we observed an increasingly high proportion of T oxidation to androstenedione, with aromatase being the prevalent enzyme activity in HepG2 and HuH7 cells at 72 h, while no aromatase could be detected in HA22T cells. Testosterone 103-115 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 320-329 19331152-5 2009 RESULTS: The highest radioiodide uptake of ARO and HepG2 clones which stably expressed hNIS gene were 87- and 208-fold higher than that of parental cells, respectively. radioiodide 21-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-46 19469636-5 2009 CYP19 (Trp39Arg) is a rare polymorphism and all the cases were homozygous for the wild type Trp allele (100%); this was also the case for 99.2% of the controls. Tryptophan 7-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-5 19630952-6 2009 The CYP19A1 rs936306 TT genotype was associated with 6.2% lower mammographic density than the TC/CC genotype (P = 0.02). Technetium 94-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-11 18974231-5 2009 Tissue aromatase activity was determined by the tritiated ((3)H)-water method. Tritium 58-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 7-16 18297393-1 2009 BACKGROUND: Decrement of endometrial thickness was recorded following short-term aromatase inhibitor treatment in breast cancer patients previously treated with tamoxifen. Tamoxifen 161-170 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-90 18297393-4 2009 RESULTS: There was a significant decrement of endometrial thickness, following 36.5 +/- 15.7 months of tamoxifen treatment, from a mean value of 8.7 +/- 5.2 mm, measured at the last ultrasonographic measurement performed before discontinuation of tamoxifen treatment, down to a mean value of 6.2 +/- 4.6 mm, measured following 5.3 +/- 4.8 months of aromatase inhibitor therapy (P < 0.001). Tamoxifen 103-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 349-358 18974231-5 2009 Tissue aromatase activity was determined by the tritiated ((3)H)-water method. Water 65-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 7-16 19006385-1 2008 One-electron oxidation of phenol, 2,4,6-trimethylphenol, and 2,6-dimethylphenol by [IrCl(6)](2-) in aqueous solution has a simple pH dependence, indicating slow bimolecular oxidation of ArOH and faster oxidation of ArO(-). Phenol 26-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 186-189 20072949-3 2009 In the present work the distribution of genotypes and frequency of alleles of the T/C polymorphism in promoter region of CYP17 and Trp/Arg polymorphism in codon 39 of CYP19 gene in breast cancer women were investigated. Tryptophan 131-134 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 167-172 20072949-3 2009 In the present work the distribution of genotypes and frequency of alleles of the T/C polymorphism in promoter region of CYP17 and Trp/Arg polymorphism in codon 39 of CYP19 gene in breast cancer women were investigated. Arginine 135-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 167-172 20072949-7 2009 However, the distribution of the genotypes of the Trp/Arg polymorphism of CYP19 in both control and patients did not differ significantly (p > 0.05) from those predicted by the Hardy-Weinberg distribution. Tryptophan 50-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 74-79 20072949-7 2009 However, the distribution of the genotypes of the Trp/Arg polymorphism of CYP19 in both control and patients did not differ significantly (p > 0.05) from those predicted by the Hardy-Weinberg distribution. Arginine 54-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 74-79 19172223-8 2009 Men with CYP19 (TTTA)(7-3)/ERalpha (TA)(19) allele combination had significantly lower lumbar spine BMD (P = 0.02) and those with CYP19 (TTTA)(7-3)/ERalpha (TA)(21) allele combination had significantly lower BMD for all three measurements, i.e. lumbar spine (P = 0.02), femoral neck (P = 0.02) and total hip (P = 0.008). ttta 16-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-14 19172223-8 2009 Men with CYP19 (TTTA)(7-3)/ERalpha (TA)(19) allele combination had significantly lower lumbar spine BMD (P = 0.02) and those with CYP19 (TTTA)(7-3)/ERalpha (TA)(21) allele combination had significantly lower BMD for all three measurements, i.e. lumbar spine (P = 0.02), femoral neck (P = 0.02) and total hip (P = 0.008). ttta 137-141 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-135 19495700-3 2009 This chapter describes methods for the measurement of the expression and catalytic activity of three key cytochrome P450 (CYP) enzymes involved in the production of progesterone and estrogens, aromatase (CYP19), steroid 17-hydroxylase/17,20-lyase (CYP17), and cholesterol side-chain cleavage (CYP11A). Progesterone 165-177 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 204-209 19495700-3 2009 This chapter describes methods for the measurement of the expression and catalytic activity of three key cytochrome P450 (CYP) enzymes involved in the production of progesterone and estrogens, aromatase (CYP19), steroid 17-hydroxylase/17,20-lyase (CYP17), and cholesterol side-chain cleavage (CYP11A). Cholesterol 260-271 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 204-209 19006385-1 2008 One-electron oxidation of phenol, 2,4,6-trimethylphenol, and 2,6-dimethylphenol by [IrCl(6)](2-) in aqueous solution has a simple pH dependence, indicating slow bimolecular oxidation of ArOH and faster oxidation of ArO(-). 2,6-xylenol 61-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 186-189 19006385-1 2008 One-electron oxidation of phenol, 2,4,6-trimethylphenol, and 2,6-dimethylphenol by [IrCl(6)](2-) in aqueous solution has a simple pH dependence, indicating slow bimolecular oxidation of ArOH and faster oxidation of ArO(-). ircl 84-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 186-189 18991378-8 2008 In phenols, electronic effects of the substituents and intramolecular H-bonds have a large influence on the activation energies and on the ArO-H BDEs. Phenols 3-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-142 18991378-13 2008 Significant deviations between experimental and DFT calculated ArO-H BDEs were found, however, when an intramolecular H-bond to the O(*) center was present in the phenoxyl radical, e.g., in ortho semiquinone radicals. phenoxy radical 163-179 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-66 18991378-13 2008 Significant deviations between experimental and DFT calculated ArO-H BDEs were found, however, when an intramolecular H-bond to the O(*) center was present in the phenoxyl radical, e.g., in ortho semiquinone radicals. ortho semiquinone radicals 190-216 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-66 18181018-6 2008 Aromatase is expressed in SGBS cells and its expression and activity are strongly stimulated by forskolin (FSK) and phorbol 12-myristate-13-acetate (PMA) treatment. Colforsin 96-105 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 18181018-6 2008 Aromatase is expressed in SGBS cells and its expression and activity are strongly stimulated by forskolin (FSK) and phorbol 12-myristate-13-acetate (PMA) treatment. Colforsin 107-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 18782656-0 2008 Aromatase inhibitor-induced joint pain: melatonin"s role. Melatonin 40-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 18181018-6 2008 Aromatase is expressed in SGBS cells and its expression and activity are strongly stimulated by forskolin (FSK) and phorbol 12-myristate-13-acetate (PMA) treatment. Tetradecanoylphorbol Acetate 116-147 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 18181018-6 2008 Aromatase is expressed in SGBS cells and its expression and activity are strongly stimulated by forskolin (FSK) and phorbol 12-myristate-13-acetate (PMA) treatment. Tetradecanoylphorbol Acetate 149-152 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 18181018-7 2008 Consistent with this, FSK and PMA treatment also increased activation of the proximal aromatase promoter, promoter II. Colforsin 22-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-95 18181018-7 2008 Consistent with this, FSK and PMA treatment also increased activation of the proximal aromatase promoter, promoter II. Tetradecanoylphorbol Acetate 30-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-95 18410552-2 2008 Several genes involved in steroid synthesis and metabolism, such as 11beta-hydroxysteroid dehydrogenase type 1 and aromatase, are known to be expressed within adipose tissue, thus modulating local steroid levels; however, our knowledge of which genes are expressed and at what level is incomplete. Steroids 26-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-124 18820009-6 2008 Pairwise analysis showed that combinations of the ERalpha G allele with the homozygous Trp genotype of CYP19A1 codon 39 (rs2236722), the methionine (Met) allele of COMT codon 158 (rs4680) or Pro allele of p53 codon 72 (rs1042522) were more frequent in ER-positive than ER-negative breast cancer, especially in patients less than 50-year old. Tryptophan 87-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 103-110 18437511-5 2008 For CYP19A1, risk was lower among women homozygous for the 3-bp deletion (rs11575899) in exon 4 (adjusted OR = 0.44, 95% CI 0.26-0.76), while the number of [TTTA]( n ) repeats was not significantly related to risk: the adjusted OR for n = 7/7 repeats versus n > 7/>7 repeats was 0.81 (95% CI 0.54-1.23). ttta 157-161 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-11 18937411-4 2008 The active sequence of Yb < Eu < Sm for metal and MeC5H4 < ArO < N(TMS)2 for ligand around the metal was observed for both reactions. mec5h4 56-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-71 18639541-0 2008 Effects of lactone derivatives on aromatase (CYP19) activity in H295R human adrenocortical and (anti)androgenicity in transfected LNCaP human prostate cancer cells. Lactones 11-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-50 18822378-0 2008 Aromatase is phosphorylated in situ at serine-118. Serine 39-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 18822378-2 2008 Herein, we demonstrate that aromatase serine-118 is a potential phosphorylation site in mammalian cells. Serine 38-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-37 18822378-3 2008 The amino acid context surrounding S118 is highly conserved among diverse animal species and suggests that an AGC-like kinase may phosphorylate aromatase. s118 35-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 144-153 18822378-5 2008 Mutation of S118 to either Ala or Asp destabilized aromatase, indicating an important structural role for S118. Alanine 27-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 51-60 18822378-5 2008 Mutation of S118 to either Ala or Asp destabilized aromatase, indicating an important structural role for S118. Aspartic Acid 34-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 51-60 18639541-12 2008 These lactones also induced pII promoter-specific CYP19 transcripts. Lactones 6-14 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-55 18640836-3 2008 The racemate, 4-[(1H-imidazol-1-yl)(1H-indol-4-yl)methyl]benzonitrile 9, showed high level of inhibitory activity towards CYP19 (IC(50)=11.5 nM). racemate 4-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-127 18541361-12 2008 In addition, PMA inhibited FRO, ARO and ML-1 cell migration toward serum. Tetradecanoylphorbol Acetate 13-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-35 18758106-4 2008 The isonicotinyl derivatives of 2- and 4-aminoestrone, compounds 5c and 10c, were fairly potent competitive inhibitors of aromatase (K(i), 2.1+/-0.14 and 1.53+/-0.08 microM for 5c and 10c, respectively) and other compounds did not show, to a significant extent, the aromatase inhibitory activity. isonicotinyl 4-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-131 18758106-4 2008 The isonicotinyl derivatives of 2- and 4-aminoestrone, compounds 5c and 10c, were fairly potent competitive inhibitors of aromatase (K(i), 2.1+/-0.14 and 1.53+/-0.08 microM for 5c and 10c, respectively) and other compounds did not show, to a significant extent, the aromatase inhibitory activity. isonicotinyl 4-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 266-275 18758106-4 2008 The isonicotinyl derivatives of 2- and 4-aminoestrone, compounds 5c and 10c, were fairly potent competitive inhibitors of aromatase (K(i), 2.1+/-0.14 and 1.53+/-0.08 microM for 5c and 10c, respectively) and other compounds did not show, to a significant extent, the aromatase inhibitory activity. 2- and 4-aminoestrone 32-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-131 18758106-4 2008 The isonicotinyl derivatives of 2- and 4-aminoestrone, compounds 5c and 10c, were fairly potent competitive inhibitors of aromatase (K(i), 2.1+/-0.14 and 1.53+/-0.08 microM for 5c and 10c, respectively) and other compounds did not show, to a significant extent, the aromatase inhibitory activity. 2- and 4-aminoestrone 32-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 266-275 18640836-3 2008 The racemate, 4-[(1H-imidazol-1-yl)(1H-indol-4-yl)methyl]benzonitrile 9, showed high level of inhibitory activity towards CYP19 (IC(50)=11.5 nM). CHEMBL468410 14-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-127 18475209-3 2008 METHODS: Colony formation assay was used to determine the effect of Gefitinib, a small molecule inhibitor of EGFR, on anaplastic (ARO) and follicular (WRO) thyroid cancer cell lines. Gefitinib 68-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-133 18475209-8 2008 Inhibition of EGFR kinase activity by Gefitinib resulted in a dose-dependent decrease in colony formation in both ARO and WRO thyroid cancer cell lines. Gefitinib 38-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 114-117 18475209-9 2008 Addition of Gefitinib in combination with ionizing radiation reduced cell proliferation in ARO (P = .0084) and WRO (P = .0252) as measured by colony formation assay. Gefitinib 12-21 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-94 18483177-1 2008 Aromatase (product of CYP19 gene), the critical enzyme in estrogen biosynthesis, is up-regulated in 70% of all breast cancers and is highly correlated with cyclooxygenase 2 (COX-2), the rate-determining enzyme in prostanoid biosynthesis. Prostaglandins 213-223 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 22-27 18486175-0 2008 Aromatase inhibiting and combined estrogenic effects of parabens and estrogenic effects of other additives in cosmetics. Parabens 56-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 18486175-7 2008 In addition, the catalytic inhibitory properties of parabens on an enzyme involved in a rate limiting step in steroid genesis (aromatase) were studied in human placental microsomes. Parabens 52-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-136 18486175-9 2008 In addition, it was found that parabens inhibit aromatase. Parabens 31-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-57 18482580-1 2008 In this communication, we document the self-assembly of heterologously expressed truncated human aromatase (CYP19) into nanometer scale phospholipids bilayers (Nanodiscs). Phospholipids 136-149 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-113 18482580-2 2008 The resulting P450 CYP19 preparation is stable and can tightly associate with the substrate androstenedione to form a nearly complete high-spin ferric protein. Androstenedione 92-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 19-24 18482580-3 2008 Ferrous CYP19 in Nanodiscs was mixed anaerobically in a rapid-scan stopped-flow with atmospheric dioxygen and the formation of the ferrous-oxy complex observed. Oxygen 97-105 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 8-13 18480557-0 2008 Aromatase in human breast carcinoma as a key regulator of intratumoral sex steroid concentrations. Steroids 75-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 18480557-7 2008 In addition, aromatase suppressed in situ production of bioactive androgen, 5alpha-dihydrotestosterone (DHT), in breast carcinoma. Dihydrotestosterone 76-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 18480557-7 2008 In addition, aromatase suppressed in situ production of bioactive androgen, 5alpha-dihydrotestosterone (DHT), in breast carcinoma. Dihydrotestosterone 104-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 18480557-8 2008 Aromatase inhibitors may thus have additional antiproliferative effects through increasing local DHT concentration with estrogen deprivation. Dihydrotestosterone 97-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 18462857-9 2008 Instead the phytochemical reduced the amount of ERK activated by estradiol, which could be the pathway responsible for Promoter I.1 transactivation and the induced CYP19 expression. Estradiol 65-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 164-169 18445666-2 2008 Several studies have detected associations between variations in genes encoding estrogen receptors alpha (ESR1) and beta (ESR2), and enzyme aromatase (CYP19A1), which regulates the estrogen to testosterone ratio, and cardiovascular phenotypes in the Framingham Heart Study. Testosterone 193-205 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 151-158 18445666-5 2008 RESULTS: In men, significant associations were detected between CYP19A1 polymorphisms and estradiol and testosterone levels, and the estradiol to testosterone ratio (P ranges 0.0005-0.01). Estradiol 90-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-71 18445666-5 2008 RESULTS: In men, significant associations were detected between CYP19A1 polymorphisms and estradiol and testosterone levels, and the estradiol to testosterone ratio (P ranges 0.0005-0.01). Testosterone 104-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-71 18445666-9 2008 CONCLUSIONS: Our findings suggest that variations in CYP19A1 correlate with steroid hormone levels in men. Steroids 76-91 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-60 18483761-5 2008 The Cys allele of CYP19 gene was also associated with statistically significant increased risk of prostate cancer (OR; 2.28, 95% CI, 1.20-4.35, P = 0.012). Cysteine 4-7 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 18-23 18462857-10 2008 The present study illustrated that resveratrol impeded the non-genomic induction of estrogen on CYP19. Resveratrol 35-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-101 18505907-9 2008 Importantly, PCOS women with long SHBG-short CYP19 alleles had the lowest SHBG levels (P=0.02) and the highest total testosterone (P=0.008), free androgen index (P=0.002), DHEAS (P=0.02), and testosterone/estradiol ratio (P=0.03), compared with those with other genotypes. Dehydroepiandrosterone 172-177 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-50 18465862-1 2008 The relation between the hydrogen atom transfer (HAT) and proton-coupled electron transfer (PCET) mechanisms is discussed and is illustrated by multiconfigurational electronic structure calculations on the ArOH + R(*) --> ArO(*) + RH reactions. Hydrogen 25-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 206-209 18493807-13 2008 Twelve of the marine water samples completely inhibited the expression of CYP19 without affecting E2 production; inhibition of CYP17 expression was observed only in one of the samples while expression of CYP11beta2 was induced as much as five- and ninefold after exposure of cells to extracts from two locations. Water 21-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 74-79 18509002-1 2008 In this study, we evaluated the activity of two novel pyrazolopyrimidine derivatives (Si 34 and Si 35) against ARO cells, a human anaplastic thyroid cancer cell line. 1H-pyrazolo[4,3-d]pyrimidine 54-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-114 18509002-1 2008 In this study, we evaluated the activity of two novel pyrazolopyrimidine derivatives (Si 34 and Si 35) against ARO cells, a human anaplastic thyroid cancer cell line. Silicon 86-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-114 18505907-9 2008 Importantly, PCOS women with long SHBG-short CYP19 alleles had the lowest SHBG levels (P=0.02) and the highest total testosterone (P=0.008), free androgen index (P=0.002), DHEAS (P=0.02), and testosterone/estradiol ratio (P=0.03), compared with those with other genotypes. Testosterone 192-204 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-50 18505907-9 2008 Importantly, PCOS women with long SHBG-short CYP19 alleles had the lowest SHBG levels (P=0.02) and the highest total testosterone (P=0.008), free androgen index (P=0.002), DHEAS (P=0.02), and testosterone/estradiol ratio (P=0.03), compared with those with other genotypes. Estradiol 205-214 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-50 18336853-6 2008 In comparison, 10(-7) to 2 x 10(-5) M PNMPP with 1 mM 8-Br-cAMP for 48 h decreased concentrations of estradiol-17beta, increased progesterone levels, but did not affect testosterone and cortisol secretion due to the significant suppression of CYP17 and the non-significant but obvious suppression of CYP19. pnmpp 38-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 300-305 18398871-8 2008 Aromatase and Cyp1A inhibitory potential and cytotoxicity toward HCT116 colon cancer cells increased with increasing content in procyanidins. Proanthocyanidins 128-140 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 18336853-6 2008 In comparison, 10(-7) to 2 x 10(-5) M PNMPP with 1 mM 8-Br-cAMP for 48 h decreased concentrations of estradiol-17beta, increased progesterone levels, but did not affect testosterone and cortisol secretion due to the significant suppression of CYP17 and the non-significant but obvious suppression of CYP19. 8-Bromo Cyclic Adenosine Monophosphate 54-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 300-305 17662285-15 2008 To determine the role of epigenetic modification of aromatase gene expression in endometriotic cells, endometrial cells were treated with 5-aza-deoxycytidine, which markedly enhanced aromatase mRNA expression, depending on the same proximal promoters as those in endometriotic cells. Decitabine 138-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-61 17662285-15 2008 To determine the role of epigenetic modification of aromatase gene expression in endometriotic cells, endometrial cells were treated with 5-aza-deoxycytidine, which markedly enhanced aromatase mRNA expression, depending on the same proximal promoters as those in endometriotic cells. Decitabine 138-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 183-192 18201740-0 2008 Aromatase inhibition by synthetic lactones and flavonoids in human placental microsomes and breast fibroblasts--a comparative study. Lactones 34-42 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 18515735-0 2008 Integrated analysis of zoledronic acid for prevention of aromatase inhibitor-associated bone loss in postmenopausal women with early breast cancer receiving adjuvant letrozole. Zoledronic Acid 23-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-66 18515735-11 2008 CONCLUSIONS: The results of this analysis strengthen the statistical validity of the preliminary results of the Z-FAST and ZO-FAST studies, showing that upfront zoledronic acid prevents aromatase inhibitor-associated bone loss more effectively than delayed-start zoledronic acid in postmenopausal women with early-stage breast cancer receiving letrozole. Zoledronic Acid 161-176 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 186-195 18201740-0 2008 Aromatase inhibition by synthetic lactones and flavonoids in human placental microsomes and breast fibroblasts--a comparative study. Flavonoids 47-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 18201740-2 2008 Aromatase is the key enzyme in the biosynthesis of steroids, as it converts androgens to estrogens. Steroids 51-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 18201740-3 2008 Certain flavonoids, plant derived chemicals, are known catalytic aromatase inhibitors. Flavonoids 8-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 18201740-7 2008 In this study aromatase inhibitory properties of fadrozole, 8-prenylnaringenin and a synthetic lactone (TM-7) were determined in human placental microsomes and in human primary breast fibroblasts. Fadrozole 49-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 18367396-1 2008 This paper describes the clinical evidence for using the aromatase inhibitors (AIs), anastrozole, letrozole and exemestane, as adjuvant therapy for postmenopausal women with early breast cancer. Letrozole 98-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-66 18388203-0 2008 Crystal structure and functional analysis of tetracenomycin ARO/CYC: implications for cyclization specificity of aromatic polyketides. 5838 DNI 45-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-63 18388203-0 2008 Crystal structure and functional analysis of tetracenomycin ARO/CYC: implications for cyclization specificity of aromatic polyketides. aromatic polyketides 113-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-63 18388203-2 2008 Polyketide cyclization, promoted by the aromatase/cyclase (ARO/CYC), helps diversify aromatic polyketides. aromatic polyketides 85-105 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-62 18388203-10 2008 The chemical insights gleaned from this work pave the foundation toward defining the molecular rules for the ARO/CYC cyclization specificity, whose rational control will be important for future endeavors in the engineered biosynthesis of novel anticancer and antibiotic aromatic polyketides. Polyketides 279-290 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 109-112 18367396-1 2008 This paper describes the clinical evidence for using the aromatase inhibitors (AIs), anastrozole, letrozole and exemestane, as adjuvant therapy for postmenopausal women with early breast cancer. exemestane 112-122 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-66 18335325-8 2008 CONCLUSION: Oral contraceptives containing gestodene are effective in decreasing aromatase expression in the eutopic endometrium of patients with endometriosis. Gestodene 43-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-90 17928820-6 2008 A trend toward a decrease in the expression of several steroid hormone-metabolizing enzymes, including CYP19A1, was detected. Steroids 55-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 103-110 18448329-2 2008 Human congenital estrogen deficiency, due to an inactivating mutation of the aromatase gene, leads to the lack of the estrogen synthesis, with gonadotropins and circulating testosterone ranging from normal to elevated. Testosterone 173-185 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-86 18236493-5 2008 Docking of representative compounds into a homology model of human aromatase assists in the rationalisation of the SAR derived from the in vitro biological results and supports a crucial role for a cyano group on the "A" phenyl ring, which is accessible to hydrogen bond interactions with Ser 478. Hydrogen 257-265 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-76 18236493-5 2008 Docking of representative compounds into a homology model of human aromatase assists in the rationalisation of the SAR derived from the in vitro biological results and supports a crucial role for a cyano group on the "A" phenyl ring, which is accessible to hydrogen bond interactions with Ser 478. Serine 289-292 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-76 18022659-5 2008 Recently, our laboratory focused on the possible effects on steroidogenesis of PBDEs and OH-PBDEs, e.g. in the human adrenocortical carcinoma (H295R) cell line indicating that some OH-PBDEs can significantly influence steroidogenic enzymes like CYP19 (aromatase) and CYP17. Halogenated Diphenyl Ethers 79-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 245-250 17963179-3 2008 This is accomplished through inhibition of the endometrial expression of enzymes related to the biosynthesis of prostaglandin and oestrogen, particularly cyclooxygenase type II (Cox-2) and aromatase. Prostaglandins 112-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 189-198 18022659-5 2008 Recently, our laboratory focused on the possible effects on steroidogenesis of PBDEs and OH-PBDEs, e.g. in the human adrenocortical carcinoma (H295R) cell line indicating that some OH-PBDEs can significantly influence steroidogenic enzymes like CYP19 (aromatase) and CYP17. oh-pbdes 181-189 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 245-250 18022659-5 2008 Recently, our laboratory focused on the possible effects on steroidogenesis of PBDEs and OH-PBDEs, e.g. in the human adrenocortical carcinoma (H295R) cell line indicating that some OH-PBDEs can significantly influence steroidogenic enzymes like CYP19 (aromatase) and CYP17. oh-pbdes 89-97 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 245-250 18083712-4 2008 The main objective of this study was to identify the receptors (EP) for prostaglandin E(2) (PGE(2)) that mediate the induction of CYP19 transcription in human adipocytes and breast cancer cells. Dinoprostone 72-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-135 18083712-4 2008 The main objective of this study was to identify the receptors (EP) for prostaglandin E(2) (PGE(2)) that mediate the induction of CYP19 transcription in human adipocytes and breast cancer cells. Prostaglandins E 92-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-135 18083712-13 2008 Taken together, these results suggest that PGE(2) via EP(2) and EP(4) activates the cAMP-->PKA-->CREB pathway leading to enhanced CYP19 transcription and increased aromatase activity. Prostaglandins E 43-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-141 18083712-13 2008 Taken together, these results suggest that PGE(2) via EP(2) and EP(4) activates the cAMP-->PKA-->CREB pathway leading to enhanced CYP19 transcription and increased aromatase activity. Cyclic AMP 84-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-141 17761019-10 2008 Since genistein is a major metabolite of biochanin A, it might contribute to biochanin A"s suppressive effect on CYP19 expression. Genistein 6-15 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-118 18190199-6 2008 Energy-resolved vis fluorescence spectra show the 2 1Sigma+-->1 1Sigma+(ArO(1S)-->ArO(1D)) transition from argon oxide as well as the vibrational progression A "3Delta u(nu"=0)-->X 3Sigmag*(nu") of O2 indicating that molecular oxygen dissociates and occasionally recombines depending on the experimental conditions. argon oxide 113-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-78 17959350-5 2008 The drugs inhibiting CYP19 were: lamotrigine, oxcarbazepine, tiagabine, phenobarbital, phenytoin, ethosuximide, and valproate. Lamotrigine 33-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-26 17959350-5 2008 The drugs inhibiting CYP19 were: lamotrigine, oxcarbazepine, tiagabine, phenobarbital, phenytoin, ethosuximide, and valproate. Oxcarbazepine 46-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-26 17959350-5 2008 The drugs inhibiting CYP19 were: lamotrigine, oxcarbazepine, tiagabine, phenobarbital, phenytoin, ethosuximide, and valproate. Tiagabine 61-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-26 17959350-5 2008 The drugs inhibiting CYP19 were: lamotrigine, oxcarbazepine, tiagabine, phenobarbital, phenytoin, ethosuximide, and valproate. Phenobarbital 72-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-26 17959350-5 2008 The drugs inhibiting CYP19 were: lamotrigine, oxcarbazepine, tiagabine, phenobarbital, phenytoin, ethosuximide, and valproate. Phenytoin 87-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-26 17959350-5 2008 The drugs inhibiting CYP19 were: lamotrigine, oxcarbazepine, tiagabine, phenobarbital, phenytoin, ethosuximide, and valproate. Ethosuximide 98-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-26 17959350-5 2008 The drugs inhibiting CYP19 were: lamotrigine, oxcarbazepine, tiagabine, phenobarbital, phenytoin, ethosuximide, and valproate. Valproic Acid 116-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-26 18245543-1 2008 PURPOSE: To evaluate the efficacy of treatment with the aromatase inhibitor letrozole in breast cancer patients segregated with respect to DNA polymorphisms of the aromatase gene CYP19. Letrozole 76-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 179-184 18245543-12 2008 CONCLUSION: In patients with hormone receptor-positive metastatic breast cancer treated with the aromatase inhibitor letrozole, the presence of a SNP in the 3" untranslated region of the CYP19 aromatase gene is associated with improved treatment efficacy. Letrozole 117-126 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 187-192 18506592-0 2008 Pharmacophore mapping of flavone derivatives for aromatase inhibition. flavone 25-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-58 18506592-3 2008 Besides the development of synthetic compounds, several naturally occurring and synthetic flavonoids, which are ubiquitous natural phenolic compounds and mediate the host of biological activities, are found to demonstrate inhibitory effects on aromatase. Flavonoids 90-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 244-253 18190199-8 2008 The O2+ decays via dissociative recombination and either reacts with Ar resulting in electronically excited ArO or it recombines to O2 within the Ar cage. Oxygen 4-7 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-111 18190199-8 2008 The O2+ decays via dissociative recombination and either reacts with Ar resulting in electronically excited ArO or it recombines to O2 within the Ar cage. Oxygen 4-6 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-111 18190199-6 2008 Energy-resolved vis fluorescence spectra show the 2 1Sigma+-->1 1Sigma+(ArO(1S)-->ArO(1D)) transition from argon oxide as well as the vibrational progression A "3Delta u(nu"=0)-->X 3Sigmag*(nu") of O2 indicating that molecular oxygen dissociates and occasionally recombines depending on the experimental conditions. argon oxide 113-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-91 18190199-6 2008 Energy-resolved vis fluorescence spectra show the 2 1Sigma+-->1 1Sigma+(ArO(1S)-->ArO(1D)) transition from argon oxide as well as the vibrational progression A "3Delta u(nu"=0)-->X 3Sigmag*(nu") of O2 indicating that molecular oxygen dissociates and occasionally recombines depending on the experimental conditions. Oxygen 207-209 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-78 18190199-6 2008 Energy-resolved vis fluorescence spectra show the 2 1Sigma+-->1 1Sigma+(ArO(1S)-->ArO(1D)) transition from argon oxide as well as the vibrational progression A "3Delta u(nu"=0)-->X 3Sigmag*(nu") of O2 indicating that molecular oxygen dissociates and occasionally recombines depending on the experimental conditions. Oxygen 236-242 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-78 18190199-7 2008 Both the emission from ArO and O2 as well the vuv quenching by oxygen are found to depend on the excitation energy, providing evidence that the energy transfer from the photoexcited cluster to the embedded oxygen proceeds via the O2+ ground state. Oxygen 206-212 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 23-26 18190199-7 2008 Both the emission from ArO and O2 as well the vuv quenching by oxygen are found to depend on the excitation energy, providing evidence that the energy transfer from the photoexcited cluster to the embedded oxygen proceeds via the O2+ ground state. Oxygen 230-232 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 23-26 19281057-1 2008 The third-generation aromatase inhibitors (AIs), letrozole, anastrozole and exemestane, are becoming the first choice endocrine drugs for post-menopausal women with breast cancer, since they present greater efficacy when compared with tamoxifen in both adjuvant and metastatic setting. Letrozole 49-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 18601603-10 2008 However, alleles presenting [TTTA](7) repeats in intron 4 of CYP19 were more frequent in the control group (p=0.0550). ttta 29-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-66 18611244-8 2008 RESULTS: Both MCF7 and T47D cells, as well as MCF7/Aro and T47D/Aro, exhibited sensitivity to inhibition of 17beta-estradiol dependent proliferation by NVP-AEW541. 17beta 108-114 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 51-54 18611244-8 2008 RESULTS: Both MCF7 and T47D cells, as well as MCF7/Aro and T47D/Aro, exhibited sensitivity to inhibition of 17beta-estradiol dependent proliferation by NVP-AEW541. 17beta 108-114 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-67 19281057-1 2008 The third-generation aromatase inhibitors (AIs), letrozole, anastrozole and exemestane, are becoming the first choice endocrine drugs for post-menopausal women with breast cancer, since they present greater efficacy when compared with tamoxifen in both adjuvant and metastatic setting. Anastrozole 60-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 19281057-1 2008 The third-generation aromatase inhibitors (AIs), letrozole, anastrozole and exemestane, are becoming the first choice endocrine drugs for post-menopausal women with breast cancer, since they present greater efficacy when compared with tamoxifen in both adjuvant and metastatic setting. exemestane 76-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 19281057-1 2008 The third-generation aromatase inhibitors (AIs), letrozole, anastrozole and exemestane, are becoming the first choice endocrine drugs for post-menopausal women with breast cancer, since they present greater efficacy when compared with tamoxifen in both adjuvant and metastatic setting. Tamoxifen 235-244 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 17910019-0 2007 Combining computational and biochemical studies for a rationale on the anti-aromatase activity of natural polyphenols. Polyphenols 106-117 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 17827443-1 2007 Certain polyphenols inhibit the activity of aromatase, a critical enzyme in estrogen synthesis that is coded by the CYP19A1 gene. Polyphenols 8-19 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-123 17827443-2 2007 Consumption of polyphenol-rich foods and beverages, thus, may interact with CYP19A1 genetic polymorphisms in the development of endometrial cancer. Polyphenols 15-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-83 17827443-9 2007 The authors" findings suggest that tea polyphenols may modify the effect of CYP19A1 genetic polymorphisms on the development of endometrial cancer. Polyphenols 39-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-83 18086758-2 2007 We have genotyped nine putative functional single-nucleotide polymorphisms (SNP) in genes involved in steroid hormone synthesis (SRD5A2, CYP19A1, HSB17B1, and HSD17B4) and DNA repair (XRCC2, XRCC3, BRCA2, and RAD52) using two Australian ovarian cancer case-control studies, comprising a total of 1,466 cases and 1,821 controls of Caucasian origin. Steroids 102-117 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 137-144 17661084-1 2008 PURPOSE: Intratumoral estradiol levels in postmenopausal women with breast cancer are thought to be mainly regulated by the aromatase-mediated conversion from androgens and estrogen sulfotransferase (EST)-mediated reduction of bioavailability. Estradiol 22-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 124-133 17661084-8 2008 Lower stromal aromatase expression in preinvasive breast cancer and lower EST levels in high-grade DCIS suggest that the net effect of intratumoral estradiol (E2)-modulating enzymes results in lower local E2 levels in earlier stages of breast tumorigenesis. Estradiol 148-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 17703364-1 2008 Aromatase (P450AROM) converts testosterone to estrogen. Testosterone 30-42 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17703364-1 2008 Aromatase (P450AROM) converts testosterone to estrogen. Testosterone 30-42 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-19 17910019-3 2007 Strong aromatase inhibitors including flavones, flavanones, resveratrol, and oleuropein, with activities comparable to that of the reference anti-aromatase drug aminoglutethimide, were identified. Flavones 38-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 7-16 17910019-3 2007 Strong aromatase inhibitors including flavones, flavanones, resveratrol, and oleuropein, with activities comparable to that of the reference anti-aromatase drug aminoglutethimide, were identified. Resveratrol 60-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 7-16 17910019-3 2007 Strong aromatase inhibitors including flavones, flavanones, resveratrol, and oleuropein, with activities comparable to that of the reference anti-aromatase drug aminoglutethimide, were identified. Aminoglutethimide 161-178 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 7-16 17910019-3 2007 Strong aromatase inhibitors including flavones, flavanones, resveratrol, and oleuropein, with activities comparable to that of the reference anti-aromatase drug aminoglutethimide, were identified. Aminoglutethimide 161-178 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 146-155 17975261-7 2007 Such a mixed character of adrenal and gonadal steroid production in human BMCs was supported by the expressions of P450scc, 3beta-hydroxysteroid dehydrogenase (3beta-HSD), P450c21, P450c11, P450c17, 17beta-HSD, and P450arom mRNAs. Steroids 46-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 215-223 17822730-5 2007 Amoxicillin up-regulated the expression of CYP11beta2 and CYP19 by more than 2-fold and induced estradiol production up to almost 3-fold. Amoxicillin 0-11 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-63 17822730-8 2007 The binary mixture of cyproterone and salbutamol significantly down-regulated expression of CYP19, while a mixture of ethynylestradiol and trenbolone, increased E2 production 3.7-fold. Cyproterone 22-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-97 17822730-8 2007 The binary mixture of cyproterone and salbutamol significantly down-regulated expression of CYP19, while a mixture of ethynylestradiol and trenbolone, increased E2 production 3.7-fold. Albuterol 38-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-97 17595315-3 2007 POR supported CYP19A1 activity with a calculated Km of 126 nm for androstenedione and a Vmax of 1.7 pmol/min. Androstenedione 66-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-21 17904790-3 2007 Type I aromatase inhibitors such as exemestane are steroidal inhibitors, which have androstenedione like structure and bind to androgen receptor with low affinity. exemestane 36-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 7-16 17904790-3 2007 Type I aromatase inhibitors such as exemestane are steroidal inhibitors, which have androstenedione like structure and bind to androgen receptor with low affinity. Androstenedione 84-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 7-16 17904790-4 2007 Type II aromatase inhibitors such as anastrozole and letrozole are known as non-steroidal inhibitors, which are non-competitive inhibitors of aromatase. Anastrozole 37-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 8-17 17904790-4 2007 Type II aromatase inhibitors such as anastrozole and letrozole are known as non-steroidal inhibitors, which are non-competitive inhibitors of aromatase. Anastrozole 37-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 142-151 17904790-4 2007 Type II aromatase inhibitors such as anastrozole and letrozole are known as non-steroidal inhibitors, which are non-competitive inhibitors of aromatase. Letrozole 53-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 8-17 17904790-4 2007 Type II aromatase inhibitors such as anastrozole and letrozole are known as non-steroidal inhibitors, which are non-competitive inhibitors of aromatase. Letrozole 53-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 142-151 17845332-0 2007 Aromatase inhibitor-induced bone mineral loss and its prevention by bisphosphonate administration in postmenopausal breast cancer patients. Diphosphonates 68-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17845332-1 2007 AIM: To investigate aromatase inhibitor-induced bone mineral loss and its prevention by bisphosphonate administration in postmenopausal breast cancer patients. Diphosphonates 88-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-29 17845343-0 2007 Role of aromatase inhibitor in patients with poor response to clomiphene citrate needs further evaluation. Clomiphene 62-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 8-17 17624765-0 2007 Aromatase inhibition by bioavailable methylated flavones. Flavones 48-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17624765-1 2007 Previous studies have shown chrysin, 7-hydroxyflavone and 7,4"-dihydroxyflavone to be the most potent flavonoid inhibitors of aromatase. 7-hydroxyflavone 37-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 126-135 17624765-1 2007 Previous studies have shown chrysin, 7-hydroxyflavone and 7,4"-dihydroxyflavone to be the most potent flavonoid inhibitors of aromatase. 7,4'-dihydroxyflavone 58-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 126-135 17624765-1 2007 Previous studies have shown chrysin, 7-hydroxyflavone and 7,4"-dihydroxyflavone to be the most potent flavonoid inhibitors of aromatase. Flavonoids 102-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 126-135 17624765-4 2007 In this study, we examined these fully methylated flavones as potential aromatase inhibitors for the prevention and/or treatment of hormone-dependent cancers. Flavones 50-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 72-81 17316633-9 2007 Application of medroxyprogesterone acetate, dydrogesterone, danazol, and the aromatase inhibitor finrozole significantly inhibited aromatase transcription. Medroxyprogesterone Acetate 15-42 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 131-140 17316633-9 2007 Application of medroxyprogesterone acetate, dydrogesterone, danazol, and the aromatase inhibitor finrozole significantly inhibited aromatase transcription. Dydrogesterone 44-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 131-140 17316633-9 2007 Application of medroxyprogesterone acetate, dydrogesterone, danazol, and the aromatase inhibitor finrozole significantly inhibited aromatase transcription. Danazol 60-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 131-140 17316633-9 2007 Application of medroxyprogesterone acetate, dydrogesterone, danazol, and the aromatase inhibitor finrozole significantly inhibited aromatase transcription. finrozole 97-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-86 17316633-9 2007 Application of medroxyprogesterone acetate, dydrogesterone, danazol, and the aromatase inhibitor finrozole significantly inhibited aromatase transcription. finrozole 97-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 131-140 17595315-7 2007 Mutations C569Y and V608F in the NADPH binding domain of POR had 49 and 28% of activity of CYP19A1 compared with normal reductase and were more sensitive to the amount of NADPH available for supporting CYP19A1 activity. NADP 33-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-98 17595315-7 2007 Mutations C569Y and V608F in the NADPH binding domain of POR had 49 and 28% of activity of CYP19A1 compared with normal reductase and were more sensitive to the amount of NADPH available for supporting CYP19A1 activity. NADP 33-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 202-209 17595315-7 2007 Mutations C569Y and V608F in the NADPH binding domain of POR had 49 and 28% of activity of CYP19A1 compared with normal reductase and were more sensitive to the amount of NADPH available for supporting CYP19A1 activity. NADP 171-176 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 202-209 17595315-9 2007 Similar effects were obtained at low/high (5.5/8.5) pH, but using octanol to limit the flux of electrons from POR to CYP19A1 inhibited activity supported by all variants. Octanols 66-73 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 117-124 17595315-10 2007 High molar ratios of KCl also reduced the CYP19A1 supporting activities of C569Y and V608F mutants of POR to a greater extent compared to normal POR and A287P mutant. Potassium Chloride 21-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-49 17512696-2 2007 PCBs may cause these abnormalities by altering expression of the aromatase gene CYP19. Polychlorinated Biphenyls 0-4 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 80-85 17614108-0 2007 Aromatase expression in a human osteoblastic cell line increases in response to prostaglandin E(2) in a dexamethasone-dependent fashion. Dinoprostone 80-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17614108-0 2007 Aromatase expression in a human osteoblastic cell line increases in response to prostaglandin E(2) in a dexamethasone-dependent fashion. Dexamethasone 104-117 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Colforsin 36-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Colforsin 36-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-92 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Colforsin 36-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 317-326 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Colforsin 36-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 328-333 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Colforsin 47-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Colforsin 47-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-92 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Colforsin 47-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 317-326 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Colforsin 47-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 328-333 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Dexamethasone 155-168 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Dexamethasone 155-168 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-92 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Dexamethasone 170-173 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Dexamethasone 170-173 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-92 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Dexamethasone 170-173 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 317-326 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Dexamethasone 170-173 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 328-333 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. ohno 247-251 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. ohno 247-251 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-92 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Colforsin 266-275 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Colforsin 266-275 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-92 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Dexamethasone 280-293 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 17614108-2 2007 In a previous report, we found that forskolin (FSK) synergistically induces aromatase (CYP19: a rate-limiting enzyme for estrogen synthesis) expression in dexamethasone (Dex) dependent manner in a human osteoblastic cell line, SV-HFO [Watanabe M, Ohno S, Nakajin S. Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Dexamethasone 280-293 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-92 17614108-5 2007 We found PGE(2) up-regulates aromatase activity synergistically, but this up-regulation depends on Dex. Prostaglandins E 9-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-38 17614108-6 2007 CYP19 gene expression was also increased synergistically by Dex and PGE(2). Dexamethasone 60-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-5 17614108-6 2007 CYP19 gene expression was also increased synergistically by Dex and PGE(2). Prostaglandins E 68-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-5 17614108-8 2007 PGE(2) receptor, EP(1), EP(2) and EP(4) were involved in the up-regulation of aromatase activity in response to PGE(2) in a Dex-dependent manner. Prostaglandins E 0-3 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-87 17614108-8 2007 PGE(2) receptor, EP(1), EP(2) and EP(4) were involved in the up-regulation of aromatase activity in response to PGE(2) in a Dex-dependent manner. Dexamethasone 124-127 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-87 17614108-9 2007 The cAMP-PKA pathway and Ca(2+) signaling pathway were involved in the up-regulation of aromatase activity in response to PGE(2). Cyclic AMP 4-8 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 17614108-9 2007 The cAMP-PKA pathway and Ca(2+) signaling pathway were involved in the up-regulation of aromatase activity in response to PGE(2). Prostaglandins E 122-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 17512696-6 2007 Antagonizing the AhR activity of H295R by an inhibitor abolished PCB126-elicited CYP19 induction. 3,4,5,3',4'-pentachlorobiphenyl 65-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-86 17512696-7 2007 However, PCB126 elevated basal CYP19 expression and aromatase activity in a slow progressive manner contrary to the sharp induction of the classic AhR target gene CYP1A1. 3,4,5,3',4'-pentachlorobiphenyl 9-15 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-36 17512696-8 2007 Exposure of H295R to PCBs with different AhR activation abilities also varied CYP19 and CYP1A1 expression in dissimilar patterns, although the CYP19 mRNA levels were in line with the AhR activation abilities of the congeners. Polychlorinated Biphenyls 21-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-83 17512696-8 2007 Exposure of H295R to PCBs with different AhR activation abilities also varied CYP19 and CYP1A1 expression in dissimilar patterns, although the CYP19 mRNA levels were in line with the AhR activation abilities of the congeners. Polychlorinated Biphenyls 21-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 143-148 17512696-9 2007 In contrast to PCB126, PCB39, which could not activate AhR and lacked effect on CYP1A1, significantly reduced CYP19 mRNA expression. pcb39 23-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 110-115 17574328-7 2007 Finally, ectopic expression of Nur77 markedly suppressed forskolin-induced transcriptional activation of promoters I.3 and II of the CYP19 gene. Colforsin 57-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 133-138 17574328-3 2007 In the present study, we investigated the effects of phthalate esters on aromatase (CYP19) activity and on its gene expression in a human adrenocortical carcinoma cell line, NCI-H295R. phthalate esters 53-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-89 17524972-5 2007 The product ion spectra of the [M+H](+) ions of simple phenols were dominated by ions representing the DMIS and dimethylimidazole moieties, whereas product ion spectra of the DMIS derivatives of OHPAHs with three or more fused aromatic rings showed prominent ArO(+) ions, the relative intensity of which increased with the number of rings. Phenols 55-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 259-262 17397026-5 2007 Aromatase (CYP19) activity in mammary fat was measured by (3)H(2)O release from (3)H-1beta-androstenedione. )h-1beta-androstenedione 82-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-16 17643984-4 2007 The aim of this work was to study the effect of the prenylflavonoids xanthohumol, isoxanthohumol and 8-prenylnaringenin on the breast cancer Sk-Br-3 cell line proliferation, apoptosis and activity of the enzyme aromatase (estrogen synthase). 8-prenylnaringenin 101-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 222-239 19804020-6 2007 Bisphosphonates have demonstrated promising results in this setting, and zoledronic acid is currently under consideration by the US FDA and Europe (EMEA) for the treatment of aromatase inhibitor-associated bone loss. Zoledronic Acid 73-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 175-184 17583493-1 2007 Aromatase (CYP450(arom), CYP19) is an enzyme responsible for converting the aliphatic androgens androstenedione and testosterone to the aromatic estrogens estrone and estradiol, respectively. Androstenedione 96-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17583493-1 2007 Aromatase (CYP450(arom), CYP19) is an enzyme responsible for converting the aliphatic androgens androstenedione and testosterone to the aromatic estrogens estrone and estradiol, respectively. Androstenedione 96-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-30 17583493-1 2007 Aromatase (CYP450(arom), CYP19) is an enzyme responsible for converting the aliphatic androgens androstenedione and testosterone to the aromatic estrogens estrone and estradiol, respectively. Testosterone 116-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17583493-1 2007 Aromatase (CYP450(arom), CYP19) is an enzyme responsible for converting the aliphatic androgens androstenedione and testosterone to the aromatic estrogens estrone and estradiol, respectively. Testosterone 116-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-30 17187946-7 2007 RESULTS: A polymorphism in CYP1B1 was associated with lower dehydroepiandrosterone-sulfate (DHEA-S) and progesterone levels, while a polymorphism in CYP19 (aromatase) was associated with higher testosterone and DHEA-S levels. Testosterone 194-206 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 149-154 17583493-1 2007 Aromatase (CYP450(arom), CYP19) is an enzyme responsible for converting the aliphatic androgens androstenedione and testosterone to the aromatic estrogens estrone and estradiol, respectively. Estrone 155-162 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17507620-1 2007 Aromatase, encoded by the CYP19A1 gene, is a key enzyme in estradiol biosynthesis, which catalyzes the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Estradiol 59-68 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17583493-1 2007 Aromatase (CYP450(arom), CYP19) is an enzyme responsible for converting the aliphatic androgens androstenedione and testosterone to the aromatic estrogens estrone and estradiol, respectively. Estrone 155-162 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-30 17583493-1 2007 Aromatase (CYP450(arom), CYP19) is an enzyme responsible for converting the aliphatic androgens androstenedione and testosterone to the aromatic estrogens estrone and estradiol, respectively. Estradiol 167-176 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17583493-1 2007 Aromatase (CYP450(arom), CYP19) is an enzyme responsible for converting the aliphatic androgens androstenedione and testosterone to the aromatic estrogens estrone and estradiol, respectively. Estradiol 167-176 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-30 17507620-1 2007 Aromatase, encoded by the CYP19A1 gene, is a key enzyme in estradiol biosynthesis, which catalyzes the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Estradiol 59-68 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-33 17507620-1 2007 Aromatase, encoded by the CYP19A1 gene, is a key enzyme in estradiol biosynthesis, which catalyzes the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Androstenedione 117-132 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17507620-1 2007 Aromatase, encoded by the CYP19A1 gene, is a key enzyme in estradiol biosynthesis, which catalyzes the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Androstenedione 117-132 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-33 17507620-1 2007 Aromatase, encoded by the CYP19A1 gene, is a key enzyme in estradiol biosynthesis, which catalyzes the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Testosterone 137-149 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17507620-1 2007 Aromatase, encoded by the CYP19A1 gene, is a key enzyme in estradiol biosynthesis, which catalyzes the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Testosterone 137-149 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-33 17507620-1 2007 Aromatase, encoded by the CYP19A1 gene, is a key enzyme in estradiol biosynthesis, which catalyzes the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Estrone 153-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17507620-1 2007 Aromatase, encoded by the CYP19A1 gene, is a key enzyme in estradiol biosynthesis, which catalyzes the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Estrone 153-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-33 17507620-1 2007 Aromatase, encoded by the CYP19A1 gene, is a key enzyme in estradiol biosynthesis, which catalyzes the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Estradiol 165-174 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17507620-1 2007 Aromatase, encoded by the CYP19A1 gene, is a key enzyme in estradiol biosynthesis, which catalyzes the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Estradiol 165-174 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-33 17482958-0 2007 CYP19 TCT tri-nucleotide Del/Del genotype is a susceptibility marker for prostate cancer in a Taiwanese population. tri-nucleotide 10-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-5 17341559-12 2007 CONCLUSIONS: The primary in vivo aromatase promoter in leiomyoma tissues in non-Asian U.S. women is the prostaglandin E(2)/cAMP-responsive I.3/II region. Dinoprostone 104-122 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-42 17341559-12 2007 CONCLUSIONS: The primary in vivo aromatase promoter in leiomyoma tissues in non-Asian U.S. women is the prostaglandin E(2)/cAMP-responsive I.3/II region. Cyclic AMP 123-127 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-42 17452968-4 2007 If the fetus lacks aromatase activity, dehydroepiandrosterone sulfate produced by the fetal adrenal glands cannot be converted to estrogen by the placenta, so is converted to testosterone peripherally and results in virilization of both fetus and mother. Dehydroepiandrosterone Sulfate 39-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 19-28 17452968-4 2007 If the fetus lacks aromatase activity, dehydroepiandrosterone sulfate produced by the fetal adrenal glands cannot be converted to estrogen by the placenta, so is converted to testosterone peripherally and results in virilization of both fetus and mother. Testosterone 175-187 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 19-28 17482958-1 2007 OBJECTIVES: The CYP19 gene encodes aromatase--a key enzyme involved in the conversion of androstenedione/testosterone to estrone/estradiol. Androstenedione 89-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-21 17482958-1 2007 OBJECTIVES: The CYP19 gene encodes aromatase--a key enzyme involved in the conversion of androstenedione/testosterone to estrone/estradiol. Testosterone 105-117 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-21 17482958-1 2007 OBJECTIVES: The CYP19 gene encodes aromatase--a key enzyme involved in the conversion of androstenedione/testosterone to estrone/estradiol. Estrone 121-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-21 17482958-1 2007 OBJECTIVES: The CYP19 gene encodes aromatase--a key enzyme involved in the conversion of androstenedione/testosterone to estrone/estradiol. Estradiol 129-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-21 17325027-2 2007 In this study, we used a systematic two-step approach that included gene resequencing and a haplotype-based analysis to comprehensively survey common genetic variation across the CYP19A1 locus in relation to circulating postmenopausal steroid hormone levels and breast cancer risk. Steroids 235-250 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 179-186 17370994-1 2007 The rates of formal abstraction of phenolic hydrogen atoms by free radicals, Y* + ArOH --> YH + ArO*, are profoundly influenced by the hydrogen-bond-accepting and anion-solvation abilities of solvents, by the electron affinities and reactivities (Y-H bond dissociation enthalpies) of radicals, and by the phenol"s ring substituents. Hydrogen 44-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-103 17370994-1 2007 The rates of formal abstraction of phenolic hydrogen atoms by free radicals, Y* + ArOH --> YH + ArO*, are profoundly influenced by the hydrogen-bond-accepting and anion-solvation abilities of solvents, by the electron affinities and reactivities (Y-H bond dissociation enthalpies) of radicals, and by the phenol"s ring substituents. Free Radicals 62-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-103 17370994-1 2007 The rates of formal abstraction of phenolic hydrogen atoms by free radicals, Y* + ArOH --> YH + ArO*, are profoundly influenced by the hydrogen-bond-accepting and anion-solvation abilities of solvents, by the electron affinities and reactivities (Y-H bond dissociation enthalpies) of radicals, and by the phenol"s ring substituents. aroh 82-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-103 17370994-1 2007 The rates of formal abstraction of phenolic hydrogen atoms by free radicals, Y* + ArOH --> YH + ArO*, are profoundly influenced by the hydrogen-bond-accepting and anion-solvation abilities of solvents, by the electron affinities and reactivities (Y-H bond dissociation enthalpies) of radicals, and by the phenol"s ring substituents. H-TYR-HIS-OH 94-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-103 17370994-1 2007 The rates of formal abstraction of phenolic hydrogen atoms by free radicals, Y* + ArOH --> YH + ArO*, are profoundly influenced by the hydrogen-bond-accepting and anion-solvation abilities of solvents, by the electron affinities and reactivities (Y-H bond dissociation enthalpies) of radicals, and by the phenol"s ring substituents. Hydrogen 138-146 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-103 17370994-1 2007 The rates of formal abstraction of phenolic hydrogen atoms by free radicals, Y* + ArOH --> YH + ArO*, are profoundly influenced by the hydrogen-bond-accepting and anion-solvation abilities of solvents, by the electron affinities and reactivities (Y-H bond dissociation enthalpies) of radicals, and by the phenol"s ring substituents. Phenol 35-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-103 17234707-6 2007 A small percentage of SP cells were found in ARO (0.25%), FRO (0.1%), NPA (0.06%), and WRO (0.02%) cells but not TPC1 cells. sp 22-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-48 17066438-4 2007 Intratumoral DHT concentration was positively associated with 5alpha-reductase type 1 (5alphaRed1), and negatively correlated with aromatase. Dihydrotestosterone 13-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 131-140 17359589-2 2007 The present study was aimed at investigating the relationship between the 5" UTR polymorphism of CYP17, a tetranucleotide repeat and a trinucleotide deletion polymorphism in CYP19. tetranucleotide 106-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 174-179 17359589-2 2007 The present study was aimed at investigating the relationship between the 5" UTR polymorphism of CYP17, a tetranucleotide repeat and a trinucleotide deletion polymorphism in CYP19. trinucleotide 135-148 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 174-179 17095221-0 2007 Synthesis of carbon-11 labeled sulfonanilide analogues as new potential PET agents for imaging of aromatase in breast cancer. Carbon-11 13-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-107 17095221-0 2007 Synthesis of carbon-11 labeled sulfonanilide analogues as new potential PET agents for imaging of aromatase in breast cancer. sulfonanilide 31-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-107 17095221-2 2007 Novel carbon-11 labeled sulfonanilide analogues, N-[11C]methyl-N-(2-alkyloxy-4-nitrophenyl)-methanesulfonamides ([11C]3a-f, alkyl=propyl, isopropyl, 1-ethyl-propyl, cyclopentyl, cyclohexyl, and cyclohexylethyl), were designed and synthesized as potential PET agents for imaging of aromatase in breast cancer. Carbon-11 6-15 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 281-290 17095221-2 2007 Novel carbon-11 labeled sulfonanilide analogues, N-[11C]methyl-N-(2-alkyloxy-4-nitrophenyl)-methanesulfonamides ([11C]3a-f, alkyl=propyl, isopropyl, 1-ethyl-propyl, cyclopentyl, cyclohexyl, and cyclohexylethyl), were designed and synthesized as potential PET agents for imaging of aromatase in breast cancer. sulfonanilide 24-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 281-290 17066438-8 2007 Moreover, we examined possible regulation of DHT production by aromatase in in vitro studies. Dihydrotestosterone 45-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-72 17066438-9 2007 DHT synthesis from androstenedione in MCF-7 cells was significantly inhibited by coculture with aromatase-positive stromal cells, which was significantly reversed by addition of aromatase inhibitor exemestane. Dihydrotestosterone 0-3 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-105 17066438-9 2007 DHT synthesis from androstenedione in MCF-7 cells was significantly inhibited by coculture with aromatase-positive stromal cells, which was significantly reversed by addition of aromatase inhibitor exemestane. Dihydrotestosterone 0-3 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 178-187 17066438-9 2007 DHT synthesis from androstenedione in MCF-7 cells was significantly inhibited by coculture with aromatase-positive stromal cells, which was significantly reversed by addition of aromatase inhibitor exemestane. Androstenedione 19-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-105 17066438-9 2007 DHT synthesis from androstenedione in MCF-7 cells was significantly inhibited by coculture with aromatase-positive stromal cells, which was significantly reversed by addition of aromatase inhibitor exemestane. Androstenedione 19-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 178-187 17066438-9 2007 DHT synthesis from androstenedione in MCF-7 cells was significantly inhibited by coculture with aromatase-positive stromal cells, which was significantly reversed by addition of aromatase inhibitor exemestane. exemestane 198-208 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-105 17066438-9 2007 DHT synthesis from androstenedione in MCF-7 cells was significantly inhibited by coculture with aromatase-positive stromal cells, which was significantly reversed by addition of aromatase inhibitor exemestane. exemestane 198-208 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 178-187 17066438-10 2007 These results suggest that intratumoral DHT concentration is mainly determined by 5alphaRed1 and aromatase in breast carcinoma tissues, and antiproliferative effect of DHT may primarily occur in the cases positive for both AR and 5alphaRed1. Dihydrotestosterone 40-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 97-106 17066438-11 2007 Aromatase inhibitors may be more effective in these patients, possibly due to increasing local DHT concentration with estrogen deprivation. Dihydrotestosterone 95-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17404019-1 2007 Several studies suggested that aromatase could play an important role in tumor progression and prognosis in endometrial cancer because androstenedione is converted to estrogen by the enzyme. Androstenedione 135-150 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 18221052-6 2007 The microsomal enzyme aromatase (CYP19) transforms these androgens to estrone and estradiol. Estrone 70-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-38 17687621-8 2007 CYP19 115T>C, 240G>A, and 1531C>T polymorphisms and [TTTA]n tetranucleotide repeat polymorphisms in the CYP19 gene and their haplotypes were not significantly associated with the risk of endometriosis. ttta 62-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-118 17687621-8 2007 CYP19 115T>C, 240G>A, and 1531C>T polymorphisms and [TTTA]n tetranucleotide repeat polymorphisms in the CYP19 gene and their haplotypes were not significantly associated with the risk of endometriosis. tetranucleotide 69-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-118 18221052-6 2007 The microsomal enzyme aromatase (CYP19) transforms these androgens to estrone and estradiol. Estradiol 82-91 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-38 16996190-8 2006 To further investigate the mechanisms of aromatase induction we measured CYP19 mRNA expression and showed that methoxychlor, aldrin, chlordane and tributyltin induced the transcription of the cyp19 gene. Methoxychlor 111-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-78 17112691-2 2006 Because endometriosis is an estrogen-dependent disease and since aromatase (CYP19), a key enzyme in estrogen biosynthesis, was recently demonstrated to be expressed in endometriotic lesions, we hypothesized that dioxin-like compounds could modulate local estrogen production through an up-regulation of aromatase. Dioxins 212-218 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-81 17150002-3 2006 The highly potent and specific third-generation aromatase inhibitors (anastrozole, exemestane and letrozole) have consistently demonstrated improved efficacy over tamoxifen in large randomised neoadjuvant and adjuvant clinical trials. Anastrozole 70-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-57 17150002-3 2006 The highly potent and specific third-generation aromatase inhibitors (anastrozole, exemestane and letrozole) have consistently demonstrated improved efficacy over tamoxifen in large randomised neoadjuvant and adjuvant clinical trials. exemestane 83-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-57 17150002-3 2006 The highly potent and specific third-generation aromatase inhibitors (anastrozole, exemestane and letrozole) have consistently demonstrated improved efficacy over tamoxifen in large randomised neoadjuvant and adjuvant clinical trials. Letrozole 98-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-57 17150002-3 2006 The highly potent and specific third-generation aromatase inhibitors (anastrozole, exemestane and letrozole) have consistently demonstrated improved efficacy over tamoxifen in large randomised neoadjuvant and adjuvant clinical trials. Tamoxifen 163-172 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-57 16996190-8 2006 To further investigate the mechanisms of aromatase induction we measured CYP19 mRNA expression and showed that methoxychlor, aldrin, chlordane and tributyltin induced the transcription of the cyp19 gene. Methoxychlor 111-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 192-197 16996190-8 2006 To further investigate the mechanisms of aromatase induction we measured CYP19 mRNA expression and showed that methoxychlor, aldrin, chlordane and tributyltin induced the transcription of the cyp19 gene. Aldrin 125-131 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-78 16996190-8 2006 To further investigate the mechanisms of aromatase induction we measured CYP19 mRNA expression and showed that methoxychlor, aldrin, chlordane and tributyltin induced the transcription of the cyp19 gene. Aldrin 125-131 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 192-197 16996190-8 2006 To further investigate the mechanisms of aromatase induction we measured CYP19 mRNA expression and showed that methoxychlor, aldrin, chlordane and tributyltin induced the transcription of the cyp19 gene. Chlordan 133-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-78 16996190-8 2006 To further investigate the mechanisms of aromatase induction we measured CYP19 mRNA expression and showed that methoxychlor, aldrin, chlordane and tributyltin induced the transcription of the cyp19 gene. Chlordan 133-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 192-197 16996190-8 2006 To further investigate the mechanisms of aromatase induction we measured CYP19 mRNA expression and showed that methoxychlor, aldrin, chlordane and tributyltin induced the transcription of the cyp19 gene. tributyltin 147-158 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-78 16996190-8 2006 To further investigate the mechanisms of aromatase induction we measured CYP19 mRNA expression and showed that methoxychlor, aldrin, chlordane and tributyltin induced the transcription of the cyp19 gene. tributyltin 147-158 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 192-197 17079446-0 2006 Aromatase destabilizer: novel action of exemestane, a food and drug administration-approved aromatase inhibitor. exemestane 40-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 17079446-0 2006 Aromatase destabilizer: novel action of exemestane, a food and drug administration-approved aromatase inhibitor. exemestane 40-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 17078630-4 2006 We have found that electron-donating substituents induce the rise in the enthalpy of proton dissociation (PDE) from ArOH+* radical cation (second step in SET-PT) and in the proton affinities of phenoxide ions ArO- (reaction enthalpy of the first step in SPLET). phenolate 194-203 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-119 17079446-2 2006 We have found that exemestane treatment significantly reduces aromatase protein level. exemestane 19-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-71 17079446-3 2006 Exemestane induces aromatase degradation in a dose-responsive manner (25-200 nmol/L), and the effect can be seen in as early as 2 hours. exemestane 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 19-28 17079446-4 2006 Metabolic labeling with S(35)-methionine was used to determine the half-life (t(1/2)) of aromatase protein. s(35)-methionine 24-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 89-98 17079446-5 2006 In the presence of 200 nmol/L exemestane, the t(1/2) of aromatase was reduced to 12.5 hours from 28.2 hours in the untreated cells. exemestane 30-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-65 17079446-6 2006 Furthermore, exemestane-induced aromatase degradation can be completely blocked by 10 micromol/L MG132, indicating that the degradation is mediated by proteasome. exemestane 13-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-41 17079446-6 2006 Furthermore, exemestane-induced aromatase degradation can be completely blocked by 10 micromol/L MG132, indicating that the degradation is mediated by proteasome. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 97-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-41 17079446-7 2006 We also examined the effect of exemestane on aromatase mRNA level using real-time reverse transcription-PCR. exemestane 31-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-54 17079446-10 2006 We have found that exemestane, different from letrozole and anastrozole, can destabilize the aromatase protein. exemestane 19-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-102 17079446-10 2006 We have found that exemestane, different from letrozole and anastrozole, can destabilize the aromatase protein. Letrozole 46-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-102 17079446-10 2006 We have found that exemestane, different from letrozole and anastrozole, can destabilize the aromatase protein. Anastrozole 60-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-102 16990650-12 2006 CONCLUSIONS: The administration of estrogen to aromatase-deficient men discloses the effects of circulating estrogens on LH secretion, exerted both at pituitary level, as shown by the decrease of basal and GnRH-stimulated secretion of LH and the LH pulsed amplitude, and at hypothalamic level as shown by the reduction of the frequency of LH pulses. Luteinizing Hormone 235-237 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 17088416-5 2006 Using two ATC cell lines (CAL-62 and ARO), we show here that valproic acid (VPA), a clinically available HDAC inhibitor, enhances the activity of doxorubicin, whose anti-tumor properties involve binding to DNA and inhibiting topoisomerase II. Valproic Acid 61-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-40 17088416-5 2006 Using two ATC cell lines (CAL-62 and ARO), we show here that valproic acid (VPA), a clinically available HDAC inhibitor, enhances the activity of doxorubicin, whose anti-tumor properties involve binding to DNA and inhibiting topoisomerase II. Valproic Acid 76-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-40 17088416-5 2006 Using two ATC cell lines (CAL-62 and ARO), we show here that valproic acid (VPA), a clinically available HDAC inhibitor, enhances the activity of doxorubicin, whose anti-tumor properties involve binding to DNA and inhibiting topoisomerase II. Doxorubicin 146-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-40 17088416-6 2006 A meager 0.7 mM VPA, which corresponds to serum concentrations in patients treated for epilepsy, is able to increase the cytotoxicity of doxorubicin about threefold in CAL-62 cells and twofold in ARO cells. Valproic Acid 16-19 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 196-199 17088416-6 2006 A meager 0.7 mM VPA, which corresponds to serum concentrations in patients treated for epilepsy, is able to increase the cytotoxicity of doxorubicin about threefold in CAL-62 cells and twofold in ARO cells. Doxorubicin 137-148 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 196-199 16877370-1 2006 BACKGROUND: To compare the clinical results and the cost-effectiveness of using the aromatase inhibitor, letrozole, in conjunction with FSH and FSH alone for controlled ovarian stimulation (COS) in patients undergoing intrauterine insemination (IUI) for a variety of indications. Letrozole 105-114 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-93 16807284-5 2006 Particularly aromatase (CYP19), the enzyme that converts androgens to estrogens, has been the subject of studies into the mechanisms by which chemicals interfere with sex steroid hormone homeostasis and function, often related to (de)feminization and (de)masculinazation processes. Steroids 171-186 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-29 16990650-12 2006 CONCLUSIONS: The administration of estrogen to aromatase-deficient men discloses the effects of circulating estrogens on LH secretion, exerted both at pituitary level, as shown by the decrease of basal and GnRH-stimulated secretion of LH and the LH pulsed amplitude, and at hypothalamic level as shown by the reduction of the frequency of LH pulses. Luteinizing Hormone 121-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 16990650-12 2006 CONCLUSIONS: The administration of estrogen to aromatase-deficient men discloses the effects of circulating estrogens on LH secretion, exerted both at pituitary level, as shown by the decrease of basal and GnRH-stimulated secretion of LH and the LH pulsed amplitude, and at hypothalamic level as shown by the reduction of the frequency of LH pulses. Luteinizing Hormone 235-237 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 16990650-12 2006 CONCLUSIONS: The administration of estrogen to aromatase-deficient men discloses the effects of circulating estrogens on LH secretion, exerted both at pituitary level, as shown by the decrease of basal and GnRH-stimulated secretion of LH and the LH pulsed amplitude, and at hypothalamic level as shown by the reduction of the frequency of LH pulses. Luteinizing Hormone 235-237 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 16984518-0 2006 Role of aromatase inhibitor in ovulation induction in patients with poor response to clomiphene citrate. Clomiphene 85-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 8-17 16984518-10 2006 CONCLUSION: The aromatase inhibitor letrozole is effective for ovulation induction in anovulatory infertility in patients that failed to ovulate by CC. Letrozole 36-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-25 17045167-4 2006 However, a 1-microM all-trans retinoic acid (tRA) treatment significantly increased this 125I uptake up to approximately approximately 6.5-fold on Day 3. tRA also elevated NIS mRNA expression in ARO-NIS cells, with peaks of expression being observed on Day 3. Tretinoin 20-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 195-198 16835357-2 2006 In the present study, we evaluated the potential of 17beta-estradiol (E2), genistein (G), and 4-hydroxyta-moxifen (OHT) to regulate the expression of diverse estrogen target genes and the proliferation of human WRO, FRO, and ARO thyroid carcinoma cells, which were used as a model system. Tamoxifen 115-118 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 225-228 17045167-4 2006 However, a 1-microM all-trans retinoic acid (tRA) treatment significantly increased this 125I uptake up to approximately approximately 6.5-fold on Day 3. tRA also elevated NIS mRNA expression in ARO-NIS cells, with peaks of expression being observed on Day 3. Tretinoin 45-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 195-198 17045167-6 2006 Of 1152, genes spotted onto the microarray membrane, 18 were up-regulated (z ratio>2.0) and 33 were down-regulated (z ratio<-2.0) in ARO-NIS cells after 3 days of tRA treatment. Tretinoin 169-172 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-142 16611627-7 2006 By assaying on MCF-7 cells stably transfected with CYP19 (MCF-7aro cells), resveratrol inhibited the aromatase activity with an IC(50) value of 25 microM. Resveratrol 75-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 51-56 16949385-2 2006 We evaluated testosterone and estradiol (E2) levels among women in relation to 5 single nucleotide polymorphisms (SNPs) of the aromatase (CYP 19) gene, the cytochrome P450 enzyme that converts androgens to estrogens. Testosterone 13-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-136 16949385-2 2006 We evaluated testosterone and estradiol (E2) levels among women in relation to 5 single nucleotide polymorphisms (SNPs) of the aromatase (CYP 19) gene, the cytochrome P450 enzyme that converts androgens to estrogens. Estradiol 30-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-136 16949385-2 2006 We evaluated testosterone and estradiol (E2) levels among women in relation to 5 single nucleotide polymorphisms (SNPs) of the aromatase (CYP 19) gene, the cytochrome P450 enzyme that converts androgens to estrogens. Estradiol 30-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-144 16949385-5 2006 Compared with other genotypes of the CYP 19 rs936306 polymorphism, the TT genotype was associated with a significant difference in the testosterone to E2 (T:E2) ratio--lower testosterone and higher E2 levels--especially in African American women. Testosterone 135-147 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-43 16949385-5 2006 Compared with other genotypes of the CYP 19 rs936306 polymorphism, the TT genotype was associated with a significant difference in the testosterone to E2 (T:E2) ratio--lower testosterone and higher E2 levels--especially in African American women. Testosterone 174-186 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-43 16949385-6 2006 Japanese women with the AA genotype of the CYP 19 rs749292 polymorphism had lower testosterone and E2 levels but higher levels of sex hormone-binding globulin (SHBG) compared with Japanese women with the AG and GG genotypes. Testosterone 82-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-49 16953982-2 2006 Use of an aromatase inhibitor (AI) is expected to replace tamoxifen as standard care for many patients. Tamoxifen 58-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-19 16799915-7 2006 Aromatase/CYP19 is the placental enzyme metabolizing methadone during pregnancy. Methadone 53-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-15 16997132-0 2006 Cyclooxygenase-2 directly regulates gene expression of P450 Cyp19 aromatase promoter regions pII, pI.3 and pI.7 and estradiol production in human breast tumor cells. Estradiol 116-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-65 16895991-3 2006 Circulating levels of estradiol in women are controlled by follicle-stimulating hormone (FSH), which regulates transcription of the aromatase gene (CYP19A1) in ovarian granulosa cells. Estradiol 22-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 148-155 16895991-9 2006 The stimulatory effect of beta-catenin is mediated through functional interactions with steroidogenic factor-1 that involve four acidic residues within its ligand-binding domain, mutation of which attenuates FSH/cAMP-induced Cyp19a1 mRNA accumulation. Cyclic AMP 212-216 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 225-232 16611627-11 2006 In addition, 50 microM resveratrol significantly reduced the CYP19-encoding mRNA abundance in SK-BR-3 cells. Resveratrol 23-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-66 16755025-0 2006 Gas chromatography-mass spectrometric study of 19-oxygenation of the aromatase inhibitor 19-methylandrostenedione with human placental microsomes. 19-methylandrost-4-ene-3,17-dione 89-113 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-78 16436528-11 2006 In this regard, BMPs specifically reduced the STAR transcription, whereas the levels of CYP11A, HSD3B2, and CYP19 stimulated by forskolin as well as BtcAMP were not altered. Colforsin 128-137 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-113 16755025-1 2006 To gain insight into the catalytic function of aromatase, we studied 19-oxygenation of 19-methyl-substituted derivative of the natural substrate androstenedione (AD), compound 1, with human placental aromatase by use of gas chromatography-mass spectrometry (GC-MS). Androstenedione 145-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 16755025-1 2006 To gain insight into the catalytic function of aromatase, we studied 19-oxygenation of 19-methyl-substituted derivative of the natural substrate androstenedione (AD), compound 1, with human placental aromatase by use of gas chromatography-mass spectrometry (GC-MS). Androstenedione 145-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 200-209 16740737-3 2006 Grape seed extract (GSE) contains high levels of procyanidin dimers that have been shown in our laboratory to be potent inhibitors of aromatase. procyanidin 49-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 134-143 16641167-1 2006 Cytochrome P-450 aromatase is responsible for catalysing the conversion of androstendione into estrone, so its expression in endometriotic tissue could contribute to the development of endometriosis. Androstenedione 75-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 16641167-1 2006 Cytochrome P-450 aromatase is responsible for catalysing the conversion of androstendione into estrone, so its expression in endometriotic tissue could contribute to the development of endometriosis. Estrone 95-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 16344016-2 2006 Cytochrome P450 aromatase, which is encoded by the CYP19A1 gene, converts androgens to estradiol. Estradiol 87-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 51-58 16634644-1 2006 Specific substrate-induced structural changes in the heme pocket are proposed for human cytochrome P450 aromatase (P450arom) which undergoes three consecutive oxygen activation steps. Heme 53-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 115-123 16634644-1 2006 Specific substrate-induced structural changes in the heme pocket are proposed for human cytochrome P450 aromatase (P450arom) which undergoes three consecutive oxygen activation steps. Oxygen 159-165 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 115-123 16634644-4 2006 The nu(Fe)(-)(CO) mode was upshifted from 477 to 485 and to 490 cm(-)(1) by the binding of androstenedione and 19-aldehyde-androstenedione, substrates for the first and third steps, respectively, whereas nu(Fe)(-)(CO) was not observed for P450arom with 19-hydroxyandrostenedione, a substrate for the second step, indicating that the heme distal site is very flexible and changes its structure depending on the substrate. Androstenedione 91-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 239-247 16634644-4 2006 The nu(Fe)(-)(CO) mode was upshifted from 477 to 485 and to 490 cm(-)(1) by the binding of androstenedione and 19-aldehyde-androstenedione, substrates for the first and third steps, respectively, whereas nu(Fe)(-)(CO) was not observed for P450arom with 19-hydroxyandrostenedione, a substrate for the second step, indicating that the heme distal site is very flexible and changes its structure depending on the substrate. 19-aldehyde-androstenedione 111-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 239-247 16634644-4 2006 The nu(Fe)(-)(CO) mode was upshifted from 477 to 485 and to 490 cm(-)(1) by the binding of androstenedione and 19-aldehyde-androstenedione, substrates for the first and third steps, respectively, whereas nu(Fe)(-)(CO) was not observed for P450arom with 19-hydroxyandrostenedione, a substrate for the second step, indicating that the heme distal site is very flexible and changes its structure depending on the substrate. Iron 7-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 239-247 16634644-4 2006 The nu(Fe)(-)(CO) mode was upshifted from 477 to 485 and to 490 cm(-)(1) by the binding of androstenedione and 19-aldehyde-androstenedione, substrates for the first and third steps, respectively, whereas nu(Fe)(-)(CO) was not observed for P450arom with 19-hydroxyandrostenedione, a substrate for the second step, indicating that the heme distal site is very flexible and changes its structure depending on the substrate. Carbon Monoxide 14-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 239-247 16634644-4 2006 The nu(Fe)(-)(CO) mode was upshifted from 477 to 485 and to 490 cm(-)(1) by the binding of androstenedione and 19-aldehyde-androstenedione, substrates for the first and third steps, respectively, whereas nu(Fe)(-)(CO) was not observed for P450arom with 19-hydroxyandrostenedione, a substrate for the second step, indicating that the heme distal site is very flexible and changes its structure depending on the substrate. 19-hydroxy-4-androstene-3,17-dione 253-278 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 239-247 16634644-4 2006 The nu(Fe)(-)(CO) mode was upshifted from 477 to 485 and to 490 cm(-)(1) by the binding of androstenedione and 19-aldehyde-androstenedione, substrates for the first and third steps, respectively, whereas nu(Fe)(-)(CO) was not observed for P450arom with 19-hydroxyandrostenedione, a substrate for the second step, indicating that the heme distal site is very flexible and changes its structure depending on the substrate. Heme 333-337 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 239-247 16634644-5 2006 The 19-aldehyde-androstenedione binding could reduce the electron donation from the axial thiolate, which was evident from the low-frequency shift of nu(Fe)(-)(S) by 5 cm(-)(1) compared to that of androstenedione-bound P450arom. 19-aldehyde 4-15 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 219-227 16634644-5 2006 The 19-aldehyde-androstenedione binding could reduce the electron donation from the axial thiolate, which was evident from the low-frequency shift of nu(Fe)(-)(S) by 5 cm(-)(1) compared to that of androstenedione-bound P450arom. Androstenedione 16-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 219-227 16634644-5 2006 The 19-aldehyde-androstenedione binding could reduce the electron donation from the axial thiolate, which was evident from the low-frequency shift of nu(Fe)(-)(S) by 5 cm(-)(1) compared to that of androstenedione-bound P450arom. thiolate 90-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 219-227 16634644-5 2006 The 19-aldehyde-androstenedione binding could reduce the electron donation from the axial thiolate, which was evident from the low-frequency shift of nu(Fe)(-)(S) by 5 cm(-)(1) compared to that of androstenedione-bound P450arom. Androstenedione 197-212 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 219-227 16634644-7 2006 We, therefore, propose that the substrates can regulate the formation of alternative reaction intermediates by modulating the structure on both the heme distal and proximal sites in P450arom. Heme 148-152 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 182-190 16709062-8 2006 The 1,3-rearrangement involves excited state ArO-C bond homolysis to give para-substituted phenoxyl radicals, which can be observed directly in laser flash photolysis experiments. para-substituted phenoxyl radicals 74-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-48 16712450-5 2006 Aromatase transcript expression and activity in breast tumor tissue is greater than that in the normal breast tissue, and prostaglandins can increase CYP19 expression and aromatase activity in breast cancer cells. Prostaglandins 122-136 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 150-155 16712450-12 2006 High levels of COX-2 expression result in higher levels of prostaglandin E(2) (PGE(2)), which in turn increases CYP19 expression through increases in intracellular cyclic AMP levels and activation of promoter II. Dinoprostone 59-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 112-117 16712450-12 2006 High levels of COX-2 expression result in higher levels of prostaglandin E(2) (PGE(2)), which in turn increases CYP19 expression through increases in intracellular cyclic AMP levels and activation of promoter II. Prostaglandins E 79-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 112-117 16712450-12 2006 High levels of COX-2 expression result in higher levels of prostaglandin E(2) (PGE(2)), which in turn increases CYP19 expression through increases in intracellular cyclic AMP levels and activation of promoter II. Cyclic AMP 164-174 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 112-117 16618764-1 2006 We investigated the effects of 1-methoxy-canthin-6-one, isolated from the medicinal plant Ailanthus altissima Swingle, on apoptosis in human leukemia (Jurkat), thyroid carcinoma (ARO and NPA), and hepatocellular carcinoma (HuH7) cell lines. 1-methoxy-canthin-6-one 31-54 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 179-182 16216300-6 2006 Genes coding for enzymes involved in the later or final steps of steroid production (CYP11B1, CYP11B2, CYP19, 3beta-HSD1, 3beta-HSD2 and 17beta-HSD1) were up-regulated to various extents by most PCBs. Steroids 65-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 103-108 16216300-6 2006 Genes coding for enzymes involved in the later or final steps of steroid production (CYP11B1, CYP11B2, CYP19, 3beta-HSD1, 3beta-HSD2 and 17beta-HSD1) were up-regulated to various extents by most PCBs. Polychlorinated Biphenyls 195-199 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 103-108 16683600-4 2006 Experimental and modeling results show that monochloramine directly reacts with the phenolate form of triclosan; however, the reaction is relatively slow as evinced by the second-order rate constant k(ArO)-NH2Cl = 0.025 M(-1) s(-1). chloramine 44-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 201-204 16875543-10 2006 Data from multivariate analysis showed that the allele of SULT1A1 His and CYP19 (TTTA)10 was positively associated with the risk of breast cancer. ttta 81-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 74-79 16683600-4 2006 Experimental and modeling results show that monochloramine directly reacts with the phenolate form of triclosan; however, the reaction is relatively slow as evinced by the second-order rate constant k(ArO)-NH2Cl = 0.025 M(-1) s(-1). phenoxy radical 84-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 201-204 16683600-4 2006 Experimental and modeling results show that monochloramine directly reacts with the phenolate form of triclosan; however, the reaction is relatively slow as evinced by the second-order rate constant k(ArO)-NH2Cl = 0.025 M(-1) s(-1). Triclosan 102-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 201-204 16683600-4 2006 Experimental and modeling results show that monochloramine directly reacts with the phenolate form of triclosan; however, the reaction is relatively slow as evinced by the second-order rate constant k(ArO)-NH2Cl = 0.025 M(-1) s(-1). chloramine 206-211 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 201-204 16683600-5 2006 Kinetic modeling indicates that for pH values less than 9.5, reactions between triclosan and two monochloramine autodecomposition intermediates, hypochlorous acid (k(ArO)-HOC = 5.4 x 10(3) M(-1) s(-1)) and dichloramine (k(ArO)-NHCl2 = 60 M(-1) s(-1)), are responsible for a significant percentage of the observed triclosan decay. Triclosan 79-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 166-169 16683600-5 2006 Kinetic modeling indicates that for pH values less than 9.5, reactions between triclosan and two monochloramine autodecomposition intermediates, hypochlorous acid (k(ArO)-HOC = 5.4 x 10(3) M(-1) s(-1)) and dichloramine (k(ArO)-NHCl2 = 60 M(-1) s(-1)), are responsible for a significant percentage of the observed triclosan decay. Triclosan 79-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 222-225 16683600-5 2006 Kinetic modeling indicates that for pH values less than 9.5, reactions between triclosan and two monochloramine autodecomposition intermediates, hypochlorous acid (k(ArO)-HOC = 5.4 x 10(3) M(-1) s(-1)) and dichloramine (k(ArO)-NHCl2 = 60 M(-1) s(-1)), are responsible for a significant percentage of the observed triclosan decay. chloramine 97-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 166-169 16683600-5 2006 Kinetic modeling indicates that for pH values less than 9.5, reactions between triclosan and two monochloramine autodecomposition intermediates, hypochlorous acid (k(ArO)-HOC = 5.4 x 10(3) M(-1) s(-1)) and dichloramine (k(ArO)-NHCl2 = 60 M(-1) s(-1)), are responsible for a significant percentage of the observed triclosan decay. chloramine 97-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 222-225 16683600-5 2006 Kinetic modeling indicates that for pH values less than 9.5, reactions between triclosan and two monochloramine autodecomposition intermediates, hypochlorous acid (k(ArO)-HOC = 5.4 x 10(3) M(-1) s(-1)) and dichloramine (k(ArO)-NHCl2 = 60 M(-1) s(-1)), are responsible for a significant percentage of the observed triclosan decay. Hypochlorous Acid 145-162 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 166-169 16683600-5 2006 Kinetic modeling indicates that for pH values less than 9.5, reactions between triclosan and two monochloramine autodecomposition intermediates, hypochlorous acid (k(ArO)-HOC = 5.4 x 10(3) M(-1) s(-1)) and dichloramine (k(ArO)-NHCl2 = 60 M(-1) s(-1)), are responsible for a significant percentage of the observed triclosan decay. Hypochlorous Acid 145-162 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 222-225 16683600-5 2006 Kinetic modeling indicates that for pH values less than 9.5, reactions between triclosan and two monochloramine autodecomposition intermediates, hypochlorous acid (k(ArO)-HOC = 5.4 x 10(3) M(-1) s(-1)) and dichloramine (k(ArO)-NHCl2 = 60 M(-1) s(-1)), are responsible for a significant percentage of the observed triclosan decay. Mechlorethamine 206-218 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 166-169 16683600-5 2006 Kinetic modeling indicates that for pH values less than 9.5, reactions between triclosan and two monochloramine autodecomposition intermediates, hypochlorous acid (k(ArO)-HOC = 5.4 x 10(3) M(-1) s(-1)) and dichloramine (k(ArO)-NHCl2 = 60 M(-1) s(-1)), are responsible for a significant percentage of the observed triclosan decay. N-hydroxy-4-(4-chlorophenyl)thiazole-2(3H)-thione 227-232 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 166-169 16683600-5 2006 Kinetic modeling indicates that for pH values less than 9.5, reactions between triclosan and two monochloramine autodecomposition intermediates, hypochlorous acid (k(ArO)-HOC = 5.4 x 10(3) M(-1) s(-1)) and dichloramine (k(ArO)-NHCl2 = 60 M(-1) s(-1)), are responsible for a significant percentage of the observed triclosan decay. Triclosan 313-322 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 166-169 16455059-2 2006 CYP19/aromatase is the major placental enzyme responsible for the metabolism of methadone to 2-ethylidine-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) and BUP to norbuprenorphine (norBUP). Methadone 80-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-5 16455059-2 2006 CYP19/aromatase is the major placental enzyme responsible for the metabolism of methadone to 2-ethylidine-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) and BUP to norbuprenorphine (norBUP). 2-ethylidine-1,5-dimethyl-3,3-diphenylpyrrolidine 93-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-5 16455059-2 2006 CYP19/aromatase is the major placental enzyme responsible for the metabolism of methadone to 2-ethylidine-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) and BUP to norbuprenorphine (norBUP). 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 144-148 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-5 16455059-2 2006 CYP19/aromatase is the major placental enzyme responsible for the metabolism of methadone to 2-ethylidine-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) and BUP to norbuprenorphine (norBUP). norbuprenorphine 161-177 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-5 16455059-2 2006 CYP19/aromatase is the major placental enzyme responsible for the metabolism of methadone to 2-ethylidine-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) and BUP to norbuprenorphine (norBUP). norbuprenorphine 179-185 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-5 16455059-10 2006 The determined inhibition constants of methadone and BUP for E(3) formation by a cDNA-expressed CYP19 preparation were similar to those for placental microsomes. Methadone 39-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-101 16498361-3 2006 Aromatase transcript expression and enzyme activity in breast tumor tissue is greater than that in the normal breast tissue, and prostaglandins can increase CYP19 expression and aromatase activity in breast cancer cells. Prostaglandins 129-143 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 157-162 16542075-14 2006 The oxygen atom isotope exchange reaction involving ArO and ArO2 van der Waals complexes is also investigated; the weak binding of O or O2 to Ar has only a small effect. Oxygen 4-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-55 16380254-1 2006 The synthesis and the aromatase (CYP19) inhibitory activity of 5-[(aryl)(imidazol-1-yl)methyl]-1H-indoles were reported. 5-[(aryl)(imidazol-1-yl)methyl]-1h-indoles 63-105 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-38 16380254-2 2006 Among the tested racemate compounds, 5-[(4-chlorophenyl)(1H-imidazol-1-yl)methyl]-1H-indole 8b emerged as a potent CYP19 inhibitor (IC(50)=15.3 nM). 5-[(4-chlorophenyl)(1h-imidazol-1-yl)methyl]-1h-indole 8b 37-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 115-120 16498056-7 2006 In men, PBL aromatase mRNA values increased significantly following testosterone administration (P < 0.05). Testosterone 68-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-21 16498056-8 2006 PBL mRNA aromatase levels in women during the follicular phase and men after testosterone administration were significantly higher (one-way ANOVA; P < 0.05) than in any other group. Testosterone 77-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-18 16498056-13 2006 CONCLUSIONS: The aromatase gene is differentially expressed in PBLs from women, men, and prepubertal children, indicating a sexual dimorphism in the enzyme expression and an important role of sex steroids in the modulation of aromatase gene expression. Steroids 196-204 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 16498056-13 2006 CONCLUSIONS: The aromatase gene is differentially expressed in PBLs from women, men, and prepubertal children, indicating a sexual dimorphism in the enzyme expression and an important role of sex steroids in the modulation of aromatase gene expression. Steroids 196-204 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 226-235 16533790-9 2006 ARO cell tumor xenografts were significantly (P < 0.0001) smaller in nude mice treated with BAY 43-9006 than in control mice. Sorafenib 95-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-3 16498361-9 2006 High levels of COX-2 expression result in higher levels of prostaglandin E2 (PGE2), which in turn increases CYP19 expression through increases in intracellular cyclic AMP levels and activation of promoter II. Dinoprostone 59-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-113 16498361-9 2006 High levels of COX-2 expression result in higher levels of prostaglandin E2 (PGE2), which in turn increases CYP19 expression through increases in intracellular cyclic AMP levels and activation of promoter II. Dinoprostone 77-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-113 16498361-9 2006 High levels of COX-2 expression result in higher levels of prostaglandin E2 (PGE2), which in turn increases CYP19 expression through increases in intracellular cyclic AMP levels and activation of promoter II. Cyclic AMP 160-170 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-113 16480277-0 2006 Novel sulfonanilide analogues suppress aromatase expression and activity in breast cancer cells independent of COX-2 inhibition. sulfonanilide 6-19 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-48 16289744-9 2006 Flavones (chrysin, baicalein, and galangin), flavanones (naringenin) and isoflavones (genistein, biochanin A) inhibit the activity of aromatase (CYP19), thus decreasing estrogen biosynthesis and producing antiestrogenic effects, important in breast and prostate cancers. Flavones 0-8 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 145-150 16289744-9 2006 Flavones (chrysin, baicalein, and galangin), flavanones (naringenin) and isoflavones (genistein, biochanin A) inhibit the activity of aromatase (CYP19), thus decreasing estrogen biosynthesis and producing antiestrogenic effects, important in breast and prostate cancers. chrysin 10-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 145-150 16289744-9 2006 Flavones (chrysin, baicalein, and galangin), flavanones (naringenin) and isoflavones (genistein, biochanin A) inhibit the activity of aromatase (CYP19), thus decreasing estrogen biosynthesis and producing antiestrogenic effects, important in breast and prostate cancers. naringenin 57-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 145-150 16289744-9 2006 Flavones (chrysin, baicalein, and galangin), flavanones (naringenin) and isoflavones (genistein, biochanin A) inhibit the activity of aromatase (CYP19), thus decreasing estrogen biosynthesis and producing antiestrogenic effects, important in breast and prostate cancers. Isoflavones 73-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 145-150 16289744-9 2006 Flavones (chrysin, baicalein, and galangin), flavanones (naringenin) and isoflavones (genistein, biochanin A) inhibit the activity of aromatase (CYP19), thus decreasing estrogen biosynthesis and producing antiestrogenic effects, important in breast and prostate cancers. Genistein 86-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 145-150 16480277-2 2006 Aromatase levels in breast cancer cells are enhanced by prostaglandins and reduced by COX inhibitors. Prostaglandins 56-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 16480277-8 2006 Real-time PCR analysis demonstrates that the sulfonanilide analogues decrease aromatase gene transcription in SK-BR-3 cells. sulfonanilide 45-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-87 16480277-9 2006 These studies suggest that the novel sulfonanilide compounds suppress aromatase activity and transcription in SK-BR-3 breast cancer cells independent of COX-2 inhibition. sulfonanilide 37-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 70-79 16418790-4 2006 Aromatase, the product of the CYP19 gene, is the key enzyme in the conversion of testosterone to estradiol. Testosterone 81-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-35 16451067-3 2006 The 6-methoxy- and 6-hydroxy-substituted benzofuran derivatives were shown to be potent CYP19 inhibitors (IC(50) = 0.01-1.46 microM) with activity greater than that observed for the unsubstituted parent compounds and inhibitory activity comparable with or greater than the reference compound arimidex (IC(50) = 0.6 microM). 6-methoxy- and 6-hydroxy-substituted benzofuran 4-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-93 16451067-3 2006 The 6-methoxy- and 6-hydroxy-substituted benzofuran derivatives were shown to be potent CYP19 inhibitors (IC(50) = 0.01-1.46 microM) with activity greater than that observed for the unsubstituted parent compounds and inhibitory activity comparable with or greater than the reference compound arimidex (IC(50) = 0.6 microM). Anastrozole 292-300 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-93 16303840-8 2006 CONCLUSIONS: The importance of aromatase is relatively increased in EldCa. eldca 68-73 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 16386416-0 2006 Aromatase-independent testosterone conversion into estrogenic steroids is inhibited by a 5 alpha-reductase inhibitor. Testosterone 22-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 16386416-0 2006 Aromatase-independent testosterone conversion into estrogenic steroids is inhibited by a 5 alpha-reductase inhibitor. Steroids 62-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 16386416-1 2006 Estrogens are generated mainly by the action of aromatase, which converts testosterone to estradiol and androstenedione to estrone. Testosterone 74-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-57 16386416-1 2006 Estrogens are generated mainly by the action of aromatase, which converts testosterone to estradiol and androstenedione to estrone. Estradiol 90-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-57 16386416-1 2006 Estrogens are generated mainly by the action of aromatase, which converts testosterone to estradiol and androstenedione to estrone. Androstenedione 104-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-57 16386416-1 2006 Estrogens are generated mainly by the action of aromatase, which converts testosterone to estradiol and androstenedione to estrone. Estrone 123-130 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-57 16386416-2 2006 However, in addition to estradiol and estrone, a variety of other steroids, whose synthesis is not dependent on aromatase, can stimulate the estrogen receptor. Steroids 66-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 112-121 16386416-3 2006 Here we show that testosterone is converted into such estrogenic steroids by aromatase-negative HeLa cells. Testosterone 18-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-86 16386416-3 2006 Here we show that testosterone is converted into such estrogenic steroids by aromatase-negative HeLa cells. Steroids 65-73 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-86 16386416-4 2006 This aromatase-independent generation of estrogenic steroids is seen in aromatase-positive MCF-7 cells as well. Steroids 52-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 5-14 16386416-4 2006 This aromatase-independent generation of estrogenic steroids is seen in aromatase-positive MCF-7 cells as well. Steroids 52-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 72-81 16386416-6 2006 This finding raises the possibility that the combination of a 5 alpha-reductase inhibitor and an aromatase inhibitor may reduce estrogenic steroids in vivo more completely than an aromatase inhibitor alone. Steroids 139-147 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 97-106 16330141-0 2006 Inhibition of human CYP19 by azoles used as antifungal agents and aromatase inhibitors, using a new LC-MS/MS method for the analysis of estradiol product formation. Azoles 29-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-25 16330141-0 2006 Inhibition of human CYP19 by azoles used as antifungal agents and aromatase inhibitors, using a new LC-MS/MS method for the analysis of estradiol product formation. Estradiol 136-145 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-25 16330141-4 2006 Inhibition of CYP19 (aromatase) is the working principle for tumor therapy, but is an unwanted side effect of azoles used as fungicides or antifungal drugs. Azoles 110-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-19 16330141-5 2006 The inhibition of recombinant human CYP19 by 21 azoles in use for the three different purposes was investigated using the natural substrate testosterone. Azoles 48-54 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-41 16330141-5 2006 The inhibition of recombinant human CYP19 by 21 azoles in use for the three different purposes was investigated using the natural substrate testosterone. Testosterone 140-152 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-41 16418790-4 2006 Aromatase, the product of the CYP19 gene, is the key enzyme in the conversion of testosterone to estradiol. Estradiol 97-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-35 16418790-16 2006 One may speculate that affected CYP19 activity, resulting in altered testosterone levels during pubertal development, might contribute to the association between CYP19 polymorphisms and cortical bone size. Testosterone 69-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-37 16418790-16 2006 One may speculate that affected CYP19 activity, resulting in altered testosterone levels during pubertal development, might contribute to the association between CYP19 polymorphisms and cortical bone size. Testosterone 69-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 162-167 16357134-8 2005 Lung tumor aromatase activity was inhibited by anastrozole, an aromatase inhibitor, and treatment of tumor cells in vitro with anastrozole led to significant suppression of tumor cell growth. Anastrozole 47-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-20 16877257-8 2006 Synaptic expression of P450(17alpha), P450arom, and estrogen receptors suggests "synaptocrine" mechanisms of brain steroids, which are synthesized at synapses and act as synaptic modulators. Steroids 115-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-46 16123052-7 2006 Prostaglandin E(2) (PGE(2)) is the most potent known stimulator of both StAR and aromatase. Dinoprostone 0-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-90 16123052-7 2006 Prostaglandin E(2) (PGE(2)) is the most potent known stimulator of both StAR and aromatase. Prostaglandins E 20-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-90 16394175-0 2006 Effect of epidermal growth factor and prostaglandin on the expression of aromatase (CYP19) in human adrenocortical carcinoma cell line NCI-H295R cells. Prostaglandins 38-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-82 16394175-0 2006 Effect of epidermal growth factor and prostaglandin on the expression of aromatase (CYP19) in human adrenocortical carcinoma cell line NCI-H295R cells. Prostaglandins 38-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-89 16394175-1 2006 We investigated the effects of epidermal growth factor (EGF) and prostaglandins (PG) on the expression of aromatase (CYP19) in human adrenocortical carcinoma cell line NCI-H295R cells. Prostaglandins 65-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 106-115 16394175-1 2006 We investigated the effects of epidermal growth factor (EGF) and prostaglandins (PG) on the expression of aromatase (CYP19) in human adrenocortical carcinoma cell line NCI-H295R cells. Prostaglandins 65-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 117-122 16394175-1 2006 We investigated the effects of epidermal growth factor (EGF) and prostaglandins (PG) on the expression of aromatase (CYP19) in human adrenocortical carcinoma cell line NCI-H295R cells. Prostaglandins 81-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 106-115 16394175-1 2006 We investigated the effects of epidermal growth factor (EGF) and prostaglandins (PG) on the expression of aromatase (CYP19) in human adrenocortical carcinoma cell line NCI-H295R cells. Prostaglandins 81-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 117-122 16394175-5 2006 PGE(2) also significantly increased aromatase activity, CYP19 gene transcript, and promoter II activity. Prostaglandins E 0-3 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-45 16394175-5 2006 PGE(2) also significantly increased aromatase activity, CYP19 gene transcript, and promoter II activity. Prostaglandins E 0-3 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-61 16394175-6 2006 The results of experiments using protein kinase (PK) inhibitors suggest that the cAMP-PKA signaling pathway is involved in the up-regulation of aromatase expression by EGF. Cyclic AMP 81-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 144-153 16394175-11 2006 These results suggest that PGE(2) secondarily acts as an autocrine signal in the up-regulation of aromatase expression by EGF in NCI-H295R cells. Prostaglandins E 27-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-107 16357134-8 2005 Lung tumor aromatase activity was inhibited by anastrozole, an aromatase inhibitor, and treatment of tumor cells in vitro with anastrozole led to significant suppression of tumor cell growth. Anastrozole 47-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-72 16357134-8 2005 Lung tumor aromatase activity was inhibited by anastrozole, an aromatase inhibitor, and treatment of tumor cells in vitro with anastrozole led to significant suppression of tumor cell growth. Anastrozole 127-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-20 16357134-8 2005 Lung tumor aromatase activity was inhibited by anastrozole, an aromatase inhibitor, and treatment of tumor cells in vitro with anastrozole led to significant suppression of tumor cell growth. Anastrozole 127-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-72 16125392-0 2005 Lead optimization of 7-benzyloxy 2-(4"-pyridylmethyl)thio isoflavone aromatase inhibitors. 7-benzyloxy 2-(4"-pyridylmethyl)thio 21-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-78 16125392-2 2005 The synthesis and biological evaluation of a series of 2-(4"-pyridylmethyl)thio, 7-alkyl- or aryl-substituted isoflavones as potential aromatase inhibitors are described. 2-(4"-pyridylmethyl)thio, 7-alkyl- or aryl-substituted isoflavones 55-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 16322257-10 2005 Of the recombinant allozymes, only the double mutant (Arg39Cys264) displayed a significant change from the WT enzyme in inhibitor constant for the aromatase inhibitors exemestane and letrozole. arg39cys264 54-65 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 147-156 16322257-10 2005 Of the recombinant allozymes, only the double mutant (Arg39Cys264) displayed a significant change from the WT enzyme in inhibitor constant for the aromatase inhibitors exemestane and letrozole. exemestane 168-178 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 147-156 16322257-10 2005 Of the recombinant allozymes, only the double mutant (Arg39Cys264) displayed a significant change from the WT enzyme in inhibitor constant for the aromatase inhibitors exemestane and letrozole. Letrozole 183-192 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 147-156 16322405-0 2005 Functional analysis of the I.3, I.6, pII and I.4 promoters of CYP19 (aromatase) gene in human osteoblasts and their role in vitamin D and dexamethasone stimulation. Vitamin D 124-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-67 16322405-0 2005 Functional analysis of the I.3, I.6, pII and I.4 promoters of CYP19 (aromatase) gene in human osteoblasts and their role in vitamin D and dexamethasone stimulation. Vitamin D 124-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-78 16322405-0 2005 Functional analysis of the I.3, I.6, pII and I.4 promoters of CYP19 (aromatase) gene in human osteoblasts and their role in vitamin D and dexamethasone stimulation. Dexamethasone 138-151 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-67 16322405-0 2005 Functional analysis of the I.3, I.6, pII and I.4 promoters of CYP19 (aromatase) gene in human osteoblasts and their role in vitamin D and dexamethasone stimulation. Dexamethasone 138-151 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-78 16322405-7 2005 RESULTS: Vitamin D and dexamethasone were potent stimulators of CYP19 transcription, while testosterone and 17beta-estradiol stimulated moderately. Vitamin D 9-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-69 16322405-7 2005 RESULTS: Vitamin D and dexamethasone were potent stimulators of CYP19 transcription, while testosterone and 17beta-estradiol stimulated moderately. Dexamethasone 23-36 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-69 19079905-11 2005 Moreover, soya isoflavones cannot inhibit aromatase (CYP19), which is the enzyme responsible for oestrogen synthesis. soya isoflavones 10-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-58 16408794-1 2005 A three-dimensional (3-D) structure of human aromatase (CYP 19) was modeled on the basis of the crystal structure of rabbit CYP2C5, the first solved X-ray structure of an eukaryotic cytochrome P450 and was evaluated by docking S-fadrozole and the steroidal competitive inhibitor (19R)-10-thiiranylestr-4-ene-3,17-dione, into the enzyme active site. Fadrozole 227-238 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-62 16408794-1 2005 A three-dimensional (3-D) structure of human aromatase (CYP 19) was modeled on the basis of the crystal structure of rabbit CYP2C5, the first solved X-ray structure of an eukaryotic cytochrome P450 and was evaluated by docking S-fadrozole and the steroidal competitive inhibitor (19R)-10-thiiranylestr-4-ene-3,17-dione, into the enzyme active site. (19r)-10-thiiranylestr-4-ene-3,17-dione 279-318 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-62 16285913-4 2005 The inhibition of aromatase activity by direct interaction with the dietary phytoestrogens genistein, daidzein, chrysin, and naringenin was tested in a cell free assay. Genistein 91-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 18-27 16361981-6 2005 More recently, a combination of an oral anti-androgen (spironolactone) and an aromatase inhibitor (testolactone) decreased height velocity and improved predicted height. Testolactone 99-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-87 16361981-7 2005 A phase II study in testotoxicosis is currently underway,exploring the combination of a highly selective anti-androgen, bicalutamide, and the potent aromatase inhibitor, anastrozole. Anastrozole 170-181 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 149-158 16002112-2 2005 Aromatase is a cytochrome P-450 enzyme complex that catalyzes the conversion of androst-4-ene-3,17-dione (AD) to estrone through three sequential oxidations of the 19-methyl group. Androstenedione 80-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 16002112-2 2005 Aromatase is a cytochrome P-450 enzyme complex that catalyzes the conversion of androst-4-ene-3,17-dione (AD) to estrone through three sequential oxidations of the 19-methyl group. Estrone 113-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 16002112-3 2005 3-DeoxyAD (1) and its 5-ene isomer 4 are potent and good competitive aromatase inhibitors, which are converted by aromatase to the aldehyde derivatives 3 and 6, respectively, through 19-hydroxy intermediates 2 and 5, respectively. 3-deoxyad 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-78 16002112-3 2005 3-DeoxyAD (1) and its 5-ene isomer 4 are potent and good competitive aromatase inhibitors, which are converted by aromatase to the aldehyde derivatives 3 and 6, respectively, through 19-hydroxy intermediates 2 and 5, respectively. 3-deoxyad 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 114-123 16002112-3 2005 3-DeoxyAD (1) and its 5-ene isomer 4 are potent and good competitive aromatase inhibitors, which are converted by aromatase to the aldehyde derivatives 3 and 6, respectively, through 19-hydroxy intermediates 2 and 5, respectively. 5-ene 22-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-78 16002112-3 2005 3-DeoxyAD (1) and its 5-ene isomer 4 are potent and good competitive aromatase inhibitors, which are converted by aromatase to the aldehyde derivatives 3 and 6, respectively, through 19-hydroxy intermediates 2 and 5, respectively. 5-ene 22-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 114-123 16109788-2 2005 Indeed, the CYP19 gene encoding P450 aromatase, the key enzyme for estrogen biosynthesis, is up-regulated in breast tumors predominantly via the cAMP-responsive gonad-type PII promoter, ultimately leading to increased intratumoral estrogen production and tumor growth. Cyclic AMP 145-149 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-17 16285913-6 2005 Genistein and daidzein were inactive in the human recombinant aromatase assay whereas naringenin and chrysin inhibited aromatase activity. naringenin 86-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 119-128 16285913-7 2005 However, genistein (1 nM to 1 mM) stimulated aromatase activity in ESC whereas other phytoestrogens had no effect. Genistein 9-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-54 16285913-8 2005 Immunopositive aromatase cells were demonstrated in genistein-treated ESC but not in untreated control cultures. Genistein 52-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-24 16285913-9 2005 Taken together, our data suggest that genistein can increase aromatase activity in ESC likely via increased enzyme expression. Genistein 38-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-70 16285913-4 2005 The inhibition of aromatase activity by direct interaction with the dietary phytoestrogens genistein, daidzein, chrysin, and naringenin was tested in a cell free assay. daidzein 102-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 18-27 16002112-3 2005 3-DeoxyAD (1) and its 5-ene isomer 4 are potent and good competitive aromatase inhibitors, which are converted by aromatase to the aldehyde derivatives 3 and 6, respectively, through 19-hydroxy intermediates 2 and 5, respectively. Aldehydes 131-139 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-78 16002112-3 2005 3-DeoxyAD (1) and its 5-ene isomer 4 are potent and good competitive aromatase inhibitors, which are converted by aromatase to the aldehyde derivatives 3 and 6, respectively, through 19-hydroxy intermediates 2 and 5, respectively. Aldehydes 131-139 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 114-123 16285913-4 2005 The inhibition of aromatase activity by direct interaction with the dietary phytoestrogens genistein, daidzein, chrysin, and naringenin was tested in a cell free assay. chrysin 112-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 18-27 16285913-4 2005 The inhibition of aromatase activity by direct interaction with the dietary phytoestrogens genistein, daidzein, chrysin, and naringenin was tested in a cell free assay. naringenin 125-135 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 18-27 16077024-4 2005 By using western blot and immunofluorescence analysis on a human tumour thyroid cell line (ARO), we demonstrate that purinergic stimulation by extracellular ATP induces quick cytoplasm to nucleus translocation of the protein at early times and its neosynthesis at later times. Adenosine Triphosphate 157-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-94 16199862-1 2005 When cultured in 20% O(2), human cytotrophoblasts fuse to form the syncytiotrophoblast with marked induction of hCYP19 (aromatase) gene expression. o(2) 21-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 112-118 16199862-2 2005 When cultured in 2% O(2), cytotrophoblast fusion and induced hCYP19 expression are prevented. o(2) 20-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-67 16199862-7 2005 In cells cultured in 20% O(2), proteasome inhibitors increased USF1/2 protein levels and blocked spontaneous induction of hCYP19 expression, cell fusion, and differentiation. o(2) 25-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-128 16199862-11 2005 In the second trimester, increased O(2) tension promotes proteasomal degradation of USF1/2, resulting in syncytiotrophoblast differentiation and induction of hCYP19 expression. o(2) 35-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 158-164 16142378-3 2005 Among the key enzymes metabolizing estrogens are two activating enzymes: cytochrome P450 (CYP)19 (aromatase), which converts androgens to estrogens, and CYP1B1, which converts estrogens predominantly to the 4-catechol estrogens that are further oxidized to catechol estrogen-3,4-quinones. 4-catechol 207-217 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-96 16142378-3 2005 Among the key enzymes metabolizing estrogens are two activating enzymes: cytochrome P450 (CYP)19 (aromatase), which converts androgens to estrogens, and CYP1B1, which converts estrogens predominantly to the 4-catechol estrogens that are further oxidized to catechol estrogen-3,4-quinones. catechol 209-217 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-96 16142378-3 2005 Among the key enzymes metabolizing estrogens are two activating enzymes: cytochrome P450 (CYP)19 (aromatase), which converts androgens to estrogens, and CYP1B1, which converts estrogens predominantly to the 4-catechol estrogens that are further oxidized to catechol estrogen-3,4-quinones. -3,4-quinones 274-287 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-96 16232327-10 2005 There was also significant difference of the (TTTA) 10 allele of CYP19 which was 12.4% in cases and 8.2% in controls (P < 0.05). ttta 46-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-70 16172195-2 2005 Previously, we have reported that lovastatin treatment induced the occurrence of apoptosis and differentiation in ARO anaplastic thyroid cancer cells. Lovastatin 34-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 114-117 16172195-3 2005 Here, we demonstrated that lovastatin inhibited the ARO cell invasiveness and delineated the underlying molecular mechanism. Lovastatin 27-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-55 16172195-10 2005 Taken together, our results suggested that lovastatin suppressed EGF-induced ARO cell invasiveness through the reduction of Rho geranylgeranylation, which in turn suppressed the membrane translocation, and subsequent suppression of Rho/ROCK and FAK/paxillin signaling. Lovastatin 43-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-80 16086586-1 2005 Aromatase is a cytochrome P-450 enzyme complex that catalyzes the conversion of androst-4-ene-3,17-dione (AD) to estrone and formic acid through three sequential oxygenations of the 19-methyl group. Androstenedione 80-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 16086586-1 2005 Aromatase is a cytochrome P-450 enzyme complex that catalyzes the conversion of androst-4-ene-3,17-dione (AD) to estrone and formic acid through three sequential oxygenations of the 19-methyl group. Estrone 113-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 16086586-1 2005 Aromatase is a cytochrome P-450 enzyme complex that catalyzes the conversion of androst-4-ene-3,17-dione (AD) to estrone and formic acid through three sequential oxygenations of the 19-methyl group. formic acid 125-136 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 16086586-4 2005 The productions of (3)H(2)O and (3)HCOOH were blocked by the substrate AD or the inhibitor 4-hydroxy-AD, indicating that these productions are due to a catalytic function of aromatase. (3)h(2)o 19-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 174-183 16086586-4 2005 The productions of (3)H(2)O and (3)HCOOH were blocked by the substrate AD or the inhibitor 4-hydroxy-AD, indicating that these productions are due to a catalytic function of aromatase. (3)hcooh 32-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 174-183 16086586-4 2005 The productions of (3)H(2)O and (3)HCOOH were blocked by the substrate AD or the inhibitor 4-hydroxy-AD, indicating that these productions are due to a catalytic function of aromatase. formestane 91-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 174-183 16086586-7 2005 It is implied that the aromatase-catalyzed 19-oxygenation of 5-ene steroid 4 but not the 4-ene isomer 1 would proceed in the same steric mechanism as that involved in the AD aromatization. 5-ene steroid 61-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 23-32 16086586-7 2005 It is implied that the aromatase-catalyzed 19-oxygenation of 5-ene steroid 4 but not the 4-ene isomer 1 would proceed in the same steric mechanism as that involved in the AD aromatization. 4-ene 89-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 23-32 16507393-5 2005 Beside chemotherapy, celecoxib may promulgate the effect of aromatase inhibitor in breast cancer cells. Celecoxib 21-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-69 16128984-1 2005 The mammalian testis serves two main functions: production of spermatozoa and synthesis of steroids, among them oestrogens are the end products obtained from the irreversible transformation of androgens by aromatase (P450arom). Steroids 91-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 217-225 15800924-2 2005 Aromatase, encoded by CYP19, catalyzes the aromatization of androstenedione and testosterone to estrone and estradiol, respectively. Androstenedione 60-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 22-27 15800924-2 2005 Aromatase, encoded by CYP19, catalyzes the aromatization of androstenedione and testosterone to estrone and estradiol, respectively. Testosterone 80-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 22-27 15800924-2 2005 Aromatase, encoded by CYP19, catalyzes the aromatization of androstenedione and testosterone to estrone and estradiol, respectively. Estrone 96-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 22-27 15800924-2 2005 Aromatase, encoded by CYP19, catalyzes the aromatization of androstenedione and testosterone to estrone and estradiol, respectively. Estradiol 108-117 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 22-27 15800924-11 2005 We evaluated associations between CYP19 haplotypes and plasma steroid hormone levels. Steroids 62-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-39 15800924-12 2005 The haplotype associated with endometrial cancer risk is also significantly associated with the ratios of estrone to androstenedione and estradiol to testosterone, the products and substrates of the enzyme aromatase, encoded by CYP19. Estrone 106-113 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 228-233 15800924-12 2005 The haplotype associated with endometrial cancer risk is also significantly associated with the ratios of estrone to androstenedione and estradiol to testosterone, the products and substrates of the enzyme aromatase, encoded by CYP19. Androstenedione 117-132 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 228-233 15800924-12 2005 The haplotype associated with endometrial cancer risk is also significantly associated with the ratios of estrone to androstenedione and estradiol to testosterone, the products and substrates of the enzyme aromatase, encoded by CYP19. Estradiol 137-146 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 228-233 15800924-12 2005 The haplotype associated with endometrial cancer risk is also significantly associated with the ratios of estrone to androstenedione and estradiol to testosterone, the products and substrates of the enzyme aromatase, encoded by CYP19. Testosterone 150-162 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 228-233 16077024-5 2005 Continuous purinergic triggering by extracellular ATP released by ARO cells is responsible for the control of APE1/Ref-1 intracellular level. Adenosine Triphosphate 50-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-69 16077024-8 2005 The biological relevance of our data is reinforced by the observation that APE1/Ref-1 stimulation by ATP protects ARO cells by H2O2-induced cell death. Adenosine Triphosphate 101-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 114-117 16077024-8 2005 The biological relevance of our data is reinforced by the observation that APE1/Ref-1 stimulation by ATP protects ARO cells by H2O2-induced cell death. Hydrogen Peroxide 127-131 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 114-117 16100519-2 2005 Aromatase inhibitors markedly suppress endogenous oestrogens without directly interacting with oestrogen receptors, and thus have a different mechanism of action to the antioestrogen, tamoxifen. Tamoxifen 184-193 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 16100519-4 2005 In addition, with increased efficacy and favourable safety and tolerability profiles, the aromatase inhibitors are starting to challenge tamoxifen as first choice endocrine treatment in a number of settings. Tamoxifen 137-146 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-99 16103457-2 2005 Four such genes, CYP17, CYP19, CYP11A1, and LH-beta, are involved in the synthesis and conversion of testosterone to dihydrotestosterone and estradiol. Testosterone 101-113 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-29 16009161-0 2005 The aromatase inhibitor letrozole increases the concentration of intraovarian androgens and improves in vitro fertilization outcome in low responder patients: a pilot study. Letrozole 24-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-13 16103457-2 2005 Four such genes, CYP17, CYP19, CYP11A1, and LH-beta, are involved in the synthesis and conversion of testosterone to dihydrotestosterone and estradiol. Dihydrotestosterone 117-136 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-29 16103457-2 2005 Four such genes, CYP17, CYP19, CYP11A1, and LH-beta, are involved in the synthesis and conversion of testosterone to dihydrotestosterone and estradiol. Estradiol 141-150 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-29 16054421-7 2005 In recent clinical studies, anastrozole, letrozole and exemestane, inhibitors of the estrogen synthase, aromatase, have shown advantages over tamoxifen as treatment for advanced disease. Anastrozole 28-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 85-102 16054421-7 2005 In recent clinical studies, anastrozole, letrozole and exemestane, inhibitors of the estrogen synthase, aromatase, have shown advantages over tamoxifen as treatment for advanced disease. Letrozole 41-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 85-102 16054421-7 2005 In recent clinical studies, anastrozole, letrozole and exemestane, inhibitors of the estrogen synthase, aromatase, have shown advantages over tamoxifen as treatment for advanced disease. exemestane 55-65 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 85-102 16033851-7 2005 RESULTS: Treatment of MCF7/Aro cells with estradiol or androstenedione caused modulation of the mTOR pathway, a phenomenon reversed by letrozole or RAD001. Estradiol 42-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-30 16033851-7 2005 RESULTS: Treatment of MCF7/Aro cells with estradiol or androstenedione caused modulation of the mTOR pathway, a phenomenon reversed by letrozole or RAD001. Androstenedione 55-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-30 16033851-7 2005 RESULTS: Treatment of MCF7/Aro cells with estradiol or androstenedione caused modulation of the mTOR pathway, a phenomenon reversed by letrozole or RAD001. Letrozole 135-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-30 16033851-8 2005 In MCF7/Aro and T47D/Aro cells, both agents inhibited androstenedione-induced proliferation; however, in combination, this was significantly augmented (P < 0.001, two-way ANOVA, synergy by isobologram analysis). Androstenedione 54-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 8-11 16033851-8 2005 In MCF7/Aro and T47D/Aro cells, both agents inhibited androstenedione-induced proliferation; however, in combination, this was significantly augmented (P < 0.001, two-way ANOVA, synergy by isobologram analysis). Androstenedione 54-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-24 16023337-6 2005 Apigenin and quercetin only inhibited aromatase/17beta-HSD at high doses as did genistein, biochanin A and daidzein. Quercetin 13-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-47 15925223-3 2005 The first step of the assay consists of a P450arom reaction, which converts a testosterone to a 17beta-estradiol using a recombinant human P450arom and a NADPH regenerating system. Testosterone 78-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-50 15925223-3 2005 The first step of the assay consists of a P450arom reaction, which converts a testosterone to a 17beta-estradiol using a recombinant human P450arom and a NADPH regenerating system. Testosterone 78-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-147 15925223-3 2005 The first step of the assay consists of a P450arom reaction, which converts a testosterone to a 17beta-estradiol using a recombinant human P450arom and a NADPH regenerating system. Estradiol 96-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-50 15925223-3 2005 The first step of the assay consists of a P450arom reaction, which converts a testosterone to a 17beta-estradiol using a recombinant human P450arom and a NADPH regenerating system. Estradiol 96-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-147 15925223-3 2005 The first step of the assay consists of a P450arom reaction, which converts a testosterone to a 17beta-estradiol using a recombinant human P450arom and a NADPH regenerating system. NADP 154-159 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-50 16039845-4 2005 Introduction of halogeno and methyl functions at C-2 of estrone as well as that of a phenalkyl or methyl function at the C-6alpha or C-6beta position markedly increased affinity to aromatase (apparent K(i) value=0.10-0.66 microM for the inhibitors versus 2.5 microM for estrone). Estrone 56-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 181-190 16039845-4 2005 Introduction of halogeno and methyl functions at C-2 of estrone as well as that of a phenalkyl or methyl function at the C-6alpha or C-6beta position markedly increased affinity to aromatase (apparent K(i) value=0.10-0.66 microM for the inhibitors versus 2.5 microM for estrone). c-6alpha 121-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 181-190 16039845-4 2005 Introduction of halogeno and methyl functions at C-2 of estrone as well as that of a phenalkyl or methyl function at the C-6alpha or C-6beta position markedly increased affinity to aromatase (apparent K(i) value=0.10-0.66 microM for the inhibitors versus 2.5 microM for estrone). c-6beta 133-140 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 181-190 16039845-4 2005 Introduction of halogeno and methyl functions at C-2 of estrone as well as that of a phenalkyl or methyl function at the C-6alpha or C-6beta position markedly increased affinity to aromatase (apparent K(i) value=0.10-0.66 microM for the inhibitors versus 2.5 microM for estrone). Estrone 270-277 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 181-190 16002280-7 2005 Differences between aromatase inhibitors and tamoxifen are also apparent in changes in steroid hormone expression. Steroids 87-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-29 15814851-1 2005 Estradiol, the most potent endogenous estrogen, is biosynthesized from androgens by the cytochrome P450 enzyme complex called aromatase. Estradiol 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 126-135 15814851-5 2005 Nonsteroidal aromatase inhibitors can be divided into three classes: aminoglutethimide-like molecules, imidazole/triazole derivatives, and flavonoid analogs. Aminoglutethimide 69-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 15814851-5 2005 Nonsteroidal aromatase inhibitors can be divided into three classes: aminoglutethimide-like molecules, imidazole/triazole derivatives, and flavonoid analogs. imidazole 103-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 15814851-5 2005 Nonsteroidal aromatase inhibitors can be divided into three classes: aminoglutethimide-like molecules, imidazole/triazole derivatives, and flavonoid analogs. Triazoles 113-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 15814851-5 2005 Nonsteroidal aromatase inhibitors can be divided into three classes: aminoglutethimide-like molecules, imidazole/triazole derivatives, and flavonoid analogs. Flavonoids 139-148 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 15814851-8 2005 The potent and selective third-generation aromatase inhibitors, anastrozole, letrozole, and exemestane, were introduced into the market as endocrine therapy in postmenopausal patients failing antiestrogen therapy alone or multiple hormonal therapies. Anastrozole 64-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 15814851-8 2005 The potent and selective third-generation aromatase inhibitors, anastrozole, letrozole, and exemestane, were introduced into the market as endocrine therapy in postmenopausal patients failing antiestrogen therapy alone or multiple hormonal therapies. Letrozole 77-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 15814851-8 2005 The potent and selective third-generation aromatase inhibitors, anastrozole, letrozole, and exemestane, were introduced into the market as endocrine therapy in postmenopausal patients failing antiestrogen therapy alone or multiple hormonal therapies. exemestane 92-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 15814851-11 2005 Use of an aromatase inhibitor as initial therapy or after treatment with tamoxifen is now recommended as adjuvant hormonal therapy for a postmenopausal woman with hormone-dependent breast cancer. Tamoxifen 73-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-19 15964185-4 2005 Our hypothesis is that higher levels of COX expression result in higher levels of prostaglandin E2 (PGE2), which in turn increases CYP19 expression through increases in intracellular cyclic AMP levels. Dinoprostone 82-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 131-136 15964185-4 2005 Our hypothesis is that higher levels of COX expression result in higher levels of prostaglandin E2 (PGE2), which in turn increases CYP19 expression through increases in intracellular cyclic AMP levels. Dinoprostone 100-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 131-136 15687328-3 2005 Our hypothesis is that higher levels of COX-2 expression result in higher levels of prostaglandin E2, which, in turn, increases CYP19 expression through increases in intracellular cAMP levels. Dinoprostone 84-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 128-133 15687328-3 2005 Our hypothesis is that higher levels of COX-2 expression result in higher levels of prostaglandin E2, which, in turn, increases CYP19 expression through increases in intracellular cAMP levels. Cyclic AMP 180-184 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 128-133 15964185-4 2005 Our hypothesis is that higher levels of COX expression result in higher levels of prostaglandin E2 (PGE2), which in turn increases CYP19 expression through increases in intracellular cyclic AMP levels. Cyclic AMP 183-193 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 131-136 16019206-1 2005 The mammalian testis serves two main functions: production of spermatozoa and synthesis of steroids, among them estrogens are the end products obtained from the irreversible transformation of androgens by aromatase (P450arom). Steroids 91-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 216-225 16019206-8 2005 It is noteworthy that dexamethasone alone exerts a positive effect on Cyp19 expression in PS and a negative one in RS. Dexamethasone 22-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 70-75 16019206-9 2005 Cyclic AMP is also a positive regulator of P450arom gene expression in germ cells. Cyclic AMP 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-51 16024248-3 2005 PGE2 stimulates both aromatase expression and activity in endometriotic stromal cells via promoter II region of the aromatase gene. Dinoprostone 0-4 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 16024248-3 2005 PGE2 stimulates both aromatase expression and activity in endometriotic stromal cells via promoter II region of the aromatase gene. Dinoprostone 0-4 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-125 15837728-4 2005 Whereas first- and second-generation aromatase inhibitors inhibit estrogen synthesis in vivo by up to 90%, the third-generation compounds anastrozole, exemestane, and letrozole were found to cause > or =98% aromatase inhibition. Anastrozole 138-149 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 210-219 15837728-4 2005 Whereas first- and second-generation aromatase inhibitors inhibit estrogen synthesis in vivo by up to 90%, the third-generation compounds anastrozole, exemestane, and letrozole were found to cause > or =98% aromatase inhibition. Letrozole 167-176 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 210-219 15817919-0 2005 Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Colforsin 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-67 15817919-0 2005 Forskolin and dexamethasone synergistically induce aromatase (CYP19) expression in the human osteoblastic cell line SV-HFO. Dexamethasone 14-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-67 15817919-2 2005 In a previous study, we reported that the expression of aromatase (CYP19) is dexamethasone (Dex)-dependent and oncostatin M (OSM) increases the expression synergistically with Dex. Dexamethasone 77-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-72 15817919-2 2005 In a previous study, we reported that the expression of aromatase (CYP19) is dexamethasone (Dex)-dependent and oncostatin M (OSM) increases the expression synergistically with Dex. Dexamethasone 92-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-72 15817919-2 2005 In a previous study, we reported that the expression of aromatase (CYP19) is dexamethasone (Dex)-dependent and oncostatin M (OSM) increases the expression synergistically with Dex. Dexamethasone 176-179 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-72 15817919-5 2005 The results of reverse transcriptase (RT)-PCR suggest that the amount of CYP19 gene transcript was also up-regulated by FSK synergistically with Dex, and that promoter I.4, which is not activated by FSK alone, is activated by FSK synergistically with Dex. Colforsin 120-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-78 15817919-5 2005 The results of reverse transcriptase (RT)-PCR suggest that the amount of CYP19 gene transcript was also up-regulated by FSK synergistically with Dex, and that promoter I.4, which is not activated by FSK alone, is activated by FSK synergistically with Dex. Dexamethasone 145-148 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-78 15817919-5 2005 The results of reverse transcriptase (RT)-PCR suggest that the amount of CYP19 gene transcript was also up-regulated by FSK synergistically with Dex, and that promoter I.4, which is not activated by FSK alone, is activated by FSK synergistically with Dex. Dexamethasone 251-254 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-78 15817919-8 2005 CONCLUSIONS: Synergistic up-regulation of aromatase activity, CYP19 gene transcript, and promoter I.4 activity were Dex-dependent and not up-regulated by FSK alone. Dexamethasone 116-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-67 15653211-0 2005 Aromatase inhibitor anastrozole for treating endometrial hyperplasia in obese postmenopausal women. Anastrozole 20-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 15640252-0 2005 Association between endometriosis and genetic polymorphisms of the estradiol-synthesizing enzyme genes HSD17B1 and CYP19. Estradiol 67-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 115-120 15835730-2 2005 Those analogues of fadrozole constitute new potent nonsteroidal inhibitors of aromatase (P450 arom). Fadrozole 19-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 89-98 15968818-3 2005 The pyridylmethanol derivatives were all evaluated in vitro for inhibitory activity against aromatase (P450(AROM), CYP19), using human placental microsomes. Nicotinyl Alcohol 4-19 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 115-120 15876151-6 2005 We find out that SA-beta-Gal staining is detectable in irradiated ARO xenotransplants, but not in control tumors. 2-chloro-10-(4'(N-beta-hydroxyethyl)piperazinyl-1')acetylphenothiazine 17-19 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-69 15876151-6 2005 We find out that SA-beta-Gal staining is detectable in irradiated ARO xenotransplants, but not in control tumors. beta-D-galactose 20-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-69 15653211-1 2005 This is a preliminary study investigating the efficacy of aromatase inhibitor anastrozole in treating endometrial hyperplasia in obese postmenopausal women. Anastrozole 78-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-67 15582747-9 2005 In conclusion, brain P-450arom-ir and the relationship of its regulation with plasma sex steroid levels, estrogen and androgen receptors in the human hypothalamus and basal forebrain are region-specific. Steroids 89-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 16045239-0 2005 Are the T/C polymorphism of the CYP17 gene and the tetranucleotide repeat (TTTA) polymorphism of the CYP19 gene genetic markers for premature coronary artery disease in Caucasians? tetranucleotide 51-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-106 15642171-2 2005 We report findings from the first family-based association study examining associations between female breast cancer risk and polymorphisms in two key estrogen-biosynthesis genes CYP17 (T-->C promoter polymorphism) and CYP19 (TTTA repeat polymorphism). ttta 229-233 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 222-227 15853482-4 2005 Aromatase is the enzyme for the conversion of testosterone to 17beta-estradiol. Testosterone 46-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 15853482-4 2005 Aromatase is the enzyme for the conversion of testosterone to 17beta-estradiol. Estradiol 62-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 15853482-5 2005 Aromatase inhibitors could decrease brain excitability by decreasing local estradiol levels and therefore, could be beneficial for the treatment of epilepsy. Estradiol 75-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 16045239-0 2005 Are the T/C polymorphism of the CYP17 gene and the tetranucleotide repeat (TTTA) polymorphism of the CYP19 gene genetic markers for premature coronary artery disease in Caucasians? ttta 75-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-106 16045239-3 2005 In this cross-sectional association study we tested the hypothesis whether the T/C polymorphism of the CYP17 gene and the tetranucleotide repeat (TTTA) polymorphism of the CYP19 gene are genetic markers for CAD in Caucasians. tetranucleotide 122-137 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 172-177 16045239-3 2005 In this cross-sectional association study we tested the hypothesis whether the T/C polymorphism of the CYP17 gene and the tetranucleotide repeat (TTTA) polymorphism of the CYP19 gene are genetic markers for CAD in Caucasians. ttta 146-150 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 172-177 15743158-5 2004 Also, P450Arom activity was assessed measuring testosterone conversion to estradiol and the concentration of this last hormone in cultured endometrial explants. Testosterone 47-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 6-14 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 19 subfamily A member 1 Homo sapiens 52-57 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. Tretinoin 144-148 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-57 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. Tretinoin 150-173 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-57 15748828-10 2005 Aromatase activity was evaluated in ovarian epithelial cancer (OEC) cell lines by the tritiated water assay and the effects of third-generation aromatase inhibitors (AIs) on aromatase activity and growth were studied. Water 96-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 15748828-11 2005 Letrozole and exemestane were able to completely inhibit aromatase activity in BG1 and PEO14 cell lines. Letrozole 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-66 15748828-11 2005 Letrozole and exemestane were able to completely inhibit aromatase activity in BG1 and PEO14 cell lines. exemestane 14-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-66 15659783-2 2004 We have recently shown that the exposure of MCF-7 breast cancer cells to 0.1% ethanol enhanced their proliferation and increased their content in both estrogen receptor-alpha (ERalpha) and aromatase. Ethanol 78-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 189-198 15659783-6 2004 Neither of these two alcohols induced an increase in aromatase mRNA level. Alcohols 21-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 15659783-8 2004 Taken together, these results suggest that the increase in aromatase expression might be a key event required for the enhanced proliferation observed in the presence of ethanol. Ethanol 169-176 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-68 15743158-5 2004 Also, P450Arom activity was assessed measuring testosterone conversion to estradiol and the concentration of this last hormone in cultured endometrial explants. Estradiol 74-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 6-14 15743158-10 2004 P450Arom activity, in the presence of testosterone, was 7-fold higher in endometrial cultures from women with endometriosis, when compare with the basal culture (p <0.01). Testosterone 38-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 15719595-11 2004 To date, letrozole appears to be the most effective aromatase inhibitor in the first-line advanced breast cancer setting. Letrozole 9-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-61 15719596-1 2004 Third-generation aromatase inhibitors, letrozole, anastrozole, and exemestane, are active and well tolerated in postmenopausal patients with hormone-sensitive advanced or metastatic breast cancer, as either first- or second-line therapy. Anastrozole 50-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 15719596-1 2004 Third-generation aromatase inhibitors, letrozole, anastrozole, and exemestane, are active and well tolerated in postmenopausal patients with hormone-sensitive advanced or metastatic breast cancer, as either first- or second-line therapy. exemestane 67-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 15358790-0 2004 Biochemical and biological characterization of a novel anti-aromatase coumarin derivative. coumarin 70-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-69 15382081-12 2004 ARO and DRO90-1 tumors showed a significant response following either single injection (54 +/- 22 and 292 +/- 138 mm3, respectively) or 3 serial injections (33 +/- 14 and 241 +/- 68 mm3, respectively) compared to saline injections (472 +/- 193 and 1,257 +/- 204 mm3, respectively) at day 20. Sodium Chloride 213-219 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-3 15456920-5 2004 Therefore, aromatase activity can be assayed simply by measuring the production of estrone in the culture medium after addition of the substrate, androstenedione. Estrone 83-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-20 15456920-5 2004 Therefore, aromatase activity can be assayed simply by measuring the production of estrone in the culture medium after addition of the substrate, androstenedione. Androstenedione 146-161 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-20 15456920-6 2004 Furthermore, by making a slight change in the commercial ELISA kit and optimizing the experimental conditions, we developed a sensitive aromatase assay that could measure a wide range of estrone concentrations with very low interference by androgens. Estrone 187-194 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-145 15358790-3 2004 In a recent study, we have found that 4-benzyl-3-(4"-chlorophenyl)-7-methoxycoumarin is a potent competitive inhibitor of aromatase with respect to the androgen substrate. 4-benzyl-3-(4"-chlorophenyl)-7-methoxycoumarin 38-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-131 15358790-2 2004 Aromatase is the enzyme responsible for the conversion of androgens into estrogens, and synthetic aromatase inhibitors such as letrozole, anastrozole, and exemestane have proven to be effective endocrine regimens for ER-positive breast cancer. Letrozole 127-136 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-107 15358790-6 2004 The structure-activity studies have revealed that three functional groups (i.e. 3-(4"-chlorophenyl), 4-benzyl, and 7-methoxyl) of this coumarin are important in its inhibition of aromatase. 3-(4"-chlorophenyl), 4-benzyl, and 7-methoxyl 80-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 179-188 15358790-2 2004 Aromatase is the enzyme responsible for the conversion of androgens into estrogens, and synthetic aromatase inhibitors such as letrozole, anastrozole, and exemestane have proven to be effective endocrine regimens for ER-positive breast cancer. Anastrozole 138-149 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-107 15358790-6 2004 The structure-activity studies have revealed that three functional groups (i.e. 3-(4"-chlorophenyl), 4-benzyl, and 7-methoxyl) of this coumarin are important in its inhibition of aromatase. coumarin 135-143 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 179-188 15358790-2 2004 Aromatase is the enzyme responsible for the conversion of androgens into estrogens, and synthetic aromatase inhibitors such as letrozole, anastrozole, and exemestane have proven to be effective endocrine regimens for ER-positive breast cancer. exemestane 155-165 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 15358790-10 2004 These results demonstrate that coumarins (a common type of phytochemical) or their derivatives can be potent inhibitors of aromatase and may be useful in suppressing aromataseand ER-positive breast tumors. Coumarins 31-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 123-132 15358790-2 2004 Aromatase is the enzyme responsible for the conversion of androgens into estrogens, and synthetic aromatase inhibitors such as letrozole, anastrozole, and exemestane have proven to be effective endocrine regimens for ER-positive breast cancer. exemestane 155-165 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-107 15308167-2 2004 The UTC human cell line ARO has a selective uptake of BPA in vitro and after transplanting into nude mice. bisphenol A 54-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-27 15671766-3 2004 Iodide uptake by ARO-N was 109 times higher than by ARO, and 99mTc and 188Re uptake by ARO-N were 21 and 47 times higher than by ARO, respectively. Iodides 0-6 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-20 15319488-0 2004 Induction and inhibition of aromatase (CYP19) activity by natural and synthetic flavonoid compounds in H295R human adrenocortical carcinoma cells. Flavonoids 80-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-44 15380230-0 2004 Synthesis and evaluation of 4-triazolylflavans as new aromatase inhibitors. 4-triazolylflavans 28-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 54-63 15380230-2 2004 Azole derivatives such as letrozole or anastrozole have been developed for aromatase inhibition and are used for the treatment of breast tumors. Letrozole 26-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-84 15380230-2 2004 Azole derivatives such as letrozole or anastrozole have been developed for aromatase inhibition and are used for the treatment of breast tumors. Azoles 0-5 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-84 15380230-2 2004 Azole derivatives such as letrozole or anastrozole have been developed for aromatase inhibition and are used for the treatment of breast tumors. Anastrozole 39-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-84 15364411-1 2004 The biosynthesis of estradiol and related estrogens is catalyzed by the enzyme aromatase. Estradiol 20-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-88 15380230-3 2004 In this paper, four 4-triazolylflavans were synthesized and were found to exhibit moderate to high inhibitory activity against aromatase. 4-triazolylflavans 20-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-136 15298966-0 2004 The CYP19 gene codon 39 Trp/Arg polymorphism increases breast cancer risk in subsets of premenopausal Japanese. Tryptophan 24-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-9 15242824-7 2004 Aminoglutethimide and 4-hydroxyandrostenedione inhibited EDDP formation by 88 and 70%, respectively, suggesting that CYP19/aromatase is the enzyme catalyzing the reaction. Aminoglutethimide 0-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 117-122 15242824-7 2004 Aminoglutethimide and 4-hydroxyandrostenedione inhibited EDDP formation by 88 and 70%, respectively, suggesting that CYP19/aromatase is the enzyme catalyzing the reaction. formestane 22-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 117-122 15242824-7 2004 Aminoglutethimide and 4-hydroxyandrostenedione inhibited EDDP formation by 88 and 70%, respectively, suggesting that CYP19/aromatase is the enzyme catalyzing the reaction. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 57-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 117-122 15242824-9 2004 The apparent K(m) and V(max) values for the CYP19 catalyzed metabolism of methadone to EDDP were 424 +/- 92 microM and 420 +/- 89 pmol(mgprotein)(-1)min(-1), respectively. Methadone 74-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-49 15242824-9 2004 The apparent K(m) and V(max) values for the CYP19 catalyzed metabolism of methadone to EDDP were 424 +/- 92 microM and 420 +/- 89 pmol(mgprotein)(-1)min(-1), respectively. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 87-91 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-49 15242824-10 2004 Kinetic analysis of a cDNA-expressed CYP19 for the metabolism of methadone to EDDP was identical to that by placental microsomes. Methadone 65-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-42 15242824-10 2004 Kinetic analysis of a cDNA-expressed CYP19 for the metabolism of methadone to EDDP was identical to that by placental microsomes. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 78-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-42 15242824-11 2004 Taken together, these data indicate that CYP19/aromatase is the major enzyme responsible for the metabolism of methadone to EDDP in term human placentas obtained from healthy pregnancies. Methadone 111-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-46 15242824-11 2004 Taken together, these data indicate that CYP19/aromatase is the major enzyme responsible for the metabolism of methadone to EDDP in term human placentas obtained from healthy pregnancies. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 124-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-46 15255840-2 2004 Aromatase is a cytochrome P450 hemoprotein-containing enzyme complex that catalyzes the rate-limiting step in the conversion of androstenedione and testosterone to estrone and estradiol (E2). Androstenedione 128-143 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 15255840-2 2004 Aromatase is a cytochrome P450 hemoprotein-containing enzyme complex that catalyzes the rate-limiting step in the conversion of androstenedione and testosterone to estrone and estradiol (E2). Testosterone 148-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 15255840-2 2004 Aromatase is a cytochrome P450 hemoprotein-containing enzyme complex that catalyzes the rate-limiting step in the conversion of androstenedione and testosterone to estrone and estradiol (E2). Estrone 164-171 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 15255840-2 2004 Aromatase is a cytochrome P450 hemoprotein-containing enzyme complex that catalyzes the rate-limiting step in the conversion of androstenedione and testosterone to estrone and estradiol (E2). Estradiol 176-185 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 15298966-0 2004 The CYP19 gene codon 39 Trp/Arg polymorphism increases breast cancer risk in subsets of premenopausal Japanese. Arginine 28-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-9 15554355-0 2004 Comparative assessment of the inhibition of recombinant human CYP19 (aromatase) by azoles used in agriculture and as drugs for humans. Azoles 83-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-67 15554355-2 2004 Antifungal activity is based on inhibition of fungal CYP51 (lanosterol 14alpha-demethylase), and estrogen biosynthesis reduction is due to azole inhibition of CYP19 (aromatase). Azoles 139-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 159-164 15554355-4 2004 A fluorimetric assay based on human recombinant CYP19 enzyme with dibenzylfluorescein as a substrate was used to compare the inhibitory potency of 22 azole compounds. dibenzylfluorescein 66-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-53 15554355-4 2004 A fluorimetric assay based on human recombinant CYP19 enzyme with dibenzylfluorescein as a substrate was used to compare the inhibitory potency of 22 azole compounds. Azoles 150-155 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-53 15150604-6 2004 The third-generation AIs (anastrozole, letrozole and exemestane) are more potent than the earlier AIs (aminoglutethimide, formestane and fadrozole) with respect to both aromatase inhibition and oestrogen suppression. Anastrozole 26-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-178 15267241-0 2004 Synthesis and aromatase inhibitory activity of novel pyridine-containing isoflavones. pyridine 53-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 15267241-0 2004 Synthesis and aromatase inhibitory activity of novel pyridine-containing isoflavones. Isoflavones 73-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 15267241-3 2004 In addition, there are several classes of natural products that exert potent activities in aromatase inhibition, with the flavonoids being most prominent. Flavonoids 122-132 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-100 15267241-4 2004 Previous studies have exploited flavone and flavanone scaffolds for the development of new aromatase inhibitors. flavone 32-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-100 15267241-4 2004 Previous studies have exploited flavone and flavanone scaffolds for the development of new aromatase inhibitors. flavanone 44-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-100 15267241-5 2004 In this paper, we describe the design, synthesis, and biological evaluation of a novel series of 2-(4"-pyridylmethyl)thioisoflavones as the first example of synthetic isoflavone-based aromatase inhibitors. 2-(4"-pyridylmethyl)thioisoflavones 97-132 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 184-193 15293395-3 2004 The collision cell option of the ICP-MS instrument method was used to improve the performance of the instrument for iron measurements since ArO and ArN interferences could be reduced using this analytical method. Iron 116-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 140-143 15199113-6 2004 RESULTS: CYP19 SNPs (rs10046 and [TCT]+/-) were associated with differences in estradiol level (P =.0006 and P =.0003, respectively) and the estradiol : testosterone ratio (P =.000001() and P =.002). Estradiol 79-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-14 15178053-3 2004 This study sought to develop an in vitro model for assessing EAC effects in sea turtles by examining their ability to alter cytochrome P450 aromatase (CYP19) activity. ethyl acetoacetate 61-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 151-156 15177662-0 2004 A tetranucleotide repeat polymorphism in the CYP19 gene and breast cancer susceptibility in a Greek population exposed and not exposed to pesticides. tetranucleotide 2-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-50 15150604-6 2004 The third-generation AIs (anastrozole, letrozole and exemestane) are more potent than the earlier AIs (aminoglutethimide, formestane and fadrozole) with respect to both aromatase inhibition and oestrogen suppression. Letrozole 39-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-178 15150604-6 2004 The third-generation AIs (anastrozole, letrozole and exemestane) are more potent than the earlier AIs (aminoglutethimide, formestane and fadrozole) with respect to both aromatase inhibition and oestrogen suppression. exemestane 53-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-178 15150604-6 2004 The third-generation AIs (anastrozole, letrozole and exemestane) are more potent than the earlier AIs (aminoglutethimide, formestane and fadrozole) with respect to both aromatase inhibition and oestrogen suppression. Fadrozole 137-146 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-178 15150604-8 2004 Comparison of aromatase inhibition and oestrogen suppression between the third-generation AIs anastrozole and letrozole showed a small but significantly greater difference in the degree of suppression of oestrone and oestrone sulphate (but not oestradiol), with letrozole. Anastrozole 94-105 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 14962994-2 2004 Recombinant gonadal P450arom catalyzed the formation of a novel metabolite from testosterone, identified by gas chromatography/mass spectrometry and biochemical analyses as 1 beta-hydroxytestosterone (1 beta OH-T), in almost equal proportion to 17beta-estradiol (E(2)). Testosterone 80-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-28 15274331-4 2004 In this study, the effects of clodronate, including growth inhibition and cytosolic Ca2+ signaling, were examined and analyzed on ARO, SW579, WRO and TT thyroid cancer cell lines. Clodronic Acid 30-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-133 14962994-2 2004 Recombinant gonadal P450arom catalyzed the formation of a novel metabolite from testosterone, identified by gas chromatography/mass spectrometry and biochemical analyses as 1 beta-hydroxytestosterone (1 beta OH-T), in almost equal proportion to 17beta-estradiol (E(2)). beta-hydroxytestosterone 175-199 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-28 14962994-2 2004 Recombinant gonadal P450arom catalyzed the formation of a novel metabolite from testosterone, identified by gas chromatography/mass spectrometry and biochemical analyses as 1 beta-hydroxytestosterone (1 beta OH-T), in almost equal proportion to 17beta-estradiol (E(2)). Estradiol 245-261 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-28 14962994-6 2004 These results constitute the first of identification of a novel, perhaps potent, nonaromatizable metabolite of testosterone, whose synthesis (paradoxically) can be definitively ascribed to the activity of the gonadal paralogue of porcine P450arom. Testosterone 111-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 238-246 15123030-2 2004 The excitatory aspect of testosterone"s action in the brain may be due to its conversion to estrogen via aromatase. Testosterone 25-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 105-114 15072827-2 2004 We evaluated the association between breast cancer and Val432Leu polymorphism in the CYP1B1 gene and the tetranucleotide repeats in intron 4 of the CYP19 gene. tetranucleotide 105-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 148-153 14691014-0 2004 A benzimidazole fungicide, benomyl, and its metabolite, carbendazim, induce aromatase activity in a human ovarian granulose-like tumor cell line (KGN). benzimidazole 2-15 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 14691014-0 2004 A benzimidazole fungicide, benomyl, and its metabolite, carbendazim, induce aromatase activity in a human ovarian granulose-like tumor cell line (KGN). Benomyl 27-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 14691014-0 2004 A benzimidazole fungicide, benomyl, and its metabolite, carbendazim, induce aromatase activity in a human ovarian granulose-like tumor cell line (KGN). carbendazim 56-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 14691014-6 2004 Only benomyl, known as both a benzimidazole fungicide and a microtubule-interfering agent, was found to induce aromatase activity in association with increased levels of aromatase mRNA in KGN cells. Benomyl 5-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-120 14691014-6 2004 Only benomyl, known as both a benzimidazole fungicide and a microtubule-interfering agent, was found to induce aromatase activity in association with increased levels of aromatase mRNA in KGN cells. Benomyl 5-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 170-179 14691014-9 2004 Treatment with taxol, another class of microtubule-interfering agents, also caused induction of aromatase in KGN cells. Paclitaxel 15-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-105 15132727-10 2004 The association between CYP19 genotypes and BMD was independent of other variables, such as age, height, body weight, calcium intake or years since menopause. Calcium 118-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-29 15123030-4 2004 Use of an aromatase inhibitor, letrozole, normalized his testosterone level and improved his sexual functioning. Letrozole 31-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-19 15123030-4 2004 Use of an aromatase inhibitor, letrozole, normalized his testosterone level and improved his sexual functioning. Testosterone 57-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-19 14982857-1 2004 The present study was undertaken to analyze the effect of a P450 aromatase inhibitor (finrozole) on 4-month-old transgenic mice expressing human P450 aromatase (P450arom) under the human ubiquitin C promoter (AROM+). finrozole 86-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 161-170 15449729-7 2004 Compared with the nonfluorinated parent compounds 1i-1, fluorinated 1-(naphth-2-yl)methylimidazoles 1a-h turned out to be potent inhibitors of CYP17 and CYP19 enzymes. 1-(naphth-2-yl)methylimidazoles 68-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 153-158 15072557-5 2004 Aromatase activity and gene transcript expression were stimulated by dexamethasone. Dexamethasone 69-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 15183689-5 2004 The recombinant P450arom NmA264R was expressed in Escherichia coli (350-400 nmol/L culture) primarily by coexpression with molecular chaperones GroES/GroEL while NmA264C was expressed (240 nmol/L culture) only in the presence of chloramphenicol. Chloramphenicol 229-244 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-24 15183689-8 2004 Purified P450arom NmA264R converted androstenedione to estrone with Vmax 12.4 nmol/(min nmol) and Km) 0.26 microM, and testosterone to estradiol with Vmax 52.2 nmol/(min nmol) and Km 10.9 microM. nma264r 18-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-17 15183689-8 2004 Purified P450arom NmA264R converted androstenedione to estrone with Vmax 12.4 nmol/(min nmol) and Km) 0.26 microM, and testosterone to estradiol with Vmax 52.2 nmol/(min nmol) and Km 10.9 microM. Androstenedione 36-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-17 15183689-8 2004 Purified P450arom NmA264R converted androstenedione to estrone with Vmax 12.4 nmol/(min nmol) and Km) 0.26 microM, and testosterone to estradiol with Vmax 52.2 nmol/(min nmol) and Km 10.9 microM. Testosterone 119-131 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-17 15183689-8 2004 Purified P450arom NmA264R converted androstenedione to estrone with Vmax 12.4 nmol/(min nmol) and Km) 0.26 microM, and testosterone to estradiol with Vmax 52.2 nmol/(min nmol) and Km 10.9 microM. Estradiol 135-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-17 15287512-17 2004 In addition, 16alpha-hydroxylase and P-450arom convert DHEA-S to estriol. Dehydroepiandrosterone Sulfate 55-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-46 15287512-17 2004 In addition, 16alpha-hydroxylase and P-450arom convert DHEA-S to estriol. Estriol 65-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-46 15287512-18 2004 Since it is recently reported that P-450arom is considerably expressed in muscle tissues as well as fat tissues and that muscle tissue may be a major organ to produce estrogens in men and postmenopausal women, estriol production may be increased in the present patient"s muscle. Estriol 210-217 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 35-44 15026173-4 2004 Breast tumor-derived factors such as prostaglandin E(2) (PGE(2)) strongly stimulate aromatase expression via an alternative promoter, promoter II, leading to increased estrogenic drive and tumor growth. Dinoprostone 37-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-93 15104241-6 2004 Evidence obtained from the effects of cytochrome P450 (CYP) inhibitors on the demethylation of LAAM to norLAAM by placental microsomes suggested that CYP 19/aromatase is the major enzyme involved. Methadyl Acetate 95-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 150-156 15104241-6 2004 Evidence obtained from the effects of cytochrome P450 (CYP) inhibitors on the demethylation of LAAM to norLAAM by placental microsomes suggested that CYP 19/aromatase is the major enzyme involved. paracymethadol 103-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 150-156 15104241-9 2004 The kinetic profile determined for a cDNA-expressed CYP 19 metabolism of LAAM to norLAAM was similar to that determined for placental microsomes. Methadyl Acetate 73-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-58 15104241-9 2004 The kinetic profile determined for a cDNA-expressed CYP 19 metabolism of LAAM to norLAAM was similar to that determined for placental microsomes. paracymethadol 81-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-58 15104241-10 2004 Taken together, the above data indicate that CYP 19/aromatase is the enzyme responsible for the N-demethylation of LAAM to norLAAM in term human placentas obtained from healthy pregnant women. Nitrogen 96-97 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-51 15104241-10 2004 Taken together, the above data indicate that CYP 19/aromatase is the enzyme responsible for the N-demethylation of LAAM to norLAAM in term human placentas obtained from healthy pregnant women. Methadyl Acetate 115-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-51 15104241-10 2004 Taken together, the above data indicate that CYP 19/aromatase is the enzyme responsible for the N-demethylation of LAAM to norLAAM in term human placentas obtained from healthy pregnant women. paracymethadol 123-130 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-51 15026173-4 2004 Breast tumor-derived factors such as prostaglandin E(2) (PGE(2)) strongly stimulate aromatase expression via an alternative promoter, promoter II, leading to increased estrogenic drive and tumor growth. Dinoprostone 57-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-93 15185531-1 2004 Aromatase P-450 is a key enzyme in the production of estrogens, that is, the conversion of androstenedione and testosterone to estrone and estradiol. Estrone 127-134 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 15083379-2 2004 Estradiol, the most potent endogenous estrogen, is biosynthesized from androgens by the cytochrome P450 enzyme complex called aromatase. Estradiol 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 126-135 15083379-5 2004 Competitive aromatase inhibitors are molecules that compete with the substrate androstenedione for noncovalent binding to the active site of the enzyme to decrease the amount of product formed. Androstenedione 79-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-21 15083379-7 2004 Nonsteroidal aromatase inhibitors can be divided into three classes: aminoglutethimide-like molecules, imidazole/triazole derivatives, and flavonoid analogs. Aminoglutethimide 69-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 15083379-7 2004 Nonsteroidal aromatase inhibitors can be divided into three classes: aminoglutethimide-like molecules, imidazole/triazole derivatives, and flavonoid analogs. imidazole 103-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 15083379-7 2004 Nonsteroidal aromatase inhibitors can be divided into three classes: aminoglutethimide-like molecules, imidazole/triazole derivatives, and flavonoid analogs. Triazoles 113-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 15083379-7 2004 Nonsteroidal aromatase inhibitors can be divided into three classes: aminoglutethimide-like molecules, imidazole/triazole derivatives, and flavonoid analogs. Flavonoids 139-148 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 15083379-11 2004 Mechanism-based steroidal inhibitors such as 4-hydroxyandrostenedione and exemestane produce potent aromatase inhibition in patients. formestane 45-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-109 15083379-11 2004 Mechanism-based steroidal inhibitors such as 4-hydroxyandrostenedione and exemestane produce potent aromatase inhibition in patients. exemestane 74-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-109 15083379-12 2004 The potent and selective third-generation aromatase inhibitors, anastrozole, letrozole, and exemestane, are approved for clinical use in postmenopausal patients with advanced hormone-dependent breast cancer or in patients failing antiestrogen therapies. Anastrozole 64-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 15083379-12 2004 The potent and selective third-generation aromatase inhibitors, anastrozole, letrozole, and exemestane, are approved for clinical use in postmenopausal patients with advanced hormone-dependent breast cancer or in patients failing antiestrogen therapies. Letrozole 77-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 15083379-12 2004 The potent and selective third-generation aromatase inhibitors, anastrozole, letrozole, and exemestane, are approved for clinical use in postmenopausal patients with advanced hormone-dependent breast cancer or in patients failing antiestrogen therapies. exemestane 92-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 15083381-2 2004 This suggests that leiomyoma cells convert circulating androstenedione into estrone (via aromatase), then into the active form of estrogen, estradiol (via 17beta-HSD type I). Androstenedione 55-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 89-98 14764827-4 2004 N-PA (poorly differentiated) and ARO (anaplastic) cells were treated with increasing valproic acid concentrations. Valproic Acid 85-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-36 14968547-4 2004 Similarly, because aromatase is productive for a multifunctional physiological factor, estrogens, impaired metabolisms of bone, carbohydrate, and fat etc. Carbohydrates 128-140 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 19-28 15185531-1 2004 Aromatase P-450 is a key enzyme in the production of estrogens, that is, the conversion of androstenedione and testosterone to estrone and estradiol. Androstenedione 91-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 15185531-1 2004 Aromatase P-450 is a key enzyme in the production of estrogens, that is, the conversion of androstenedione and testosterone to estrone and estradiol. Testosterone 111-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 15185531-1 2004 Aromatase P-450 is a key enzyme in the production of estrogens, that is, the conversion of androstenedione and testosterone to estrone and estradiol. Estradiol 139-148 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 14979917-14 2004 CONCLUSION: Our results indicate the importance of CYP19 and the enzymes regulating the oestrone sulfate metabolism as factors of disease-free survival in breast cancer, in addition to the well-known factors ER and ERBB2. oestrone sulfate 88-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 51-56 14517714-1 2004 Polymorphisms in the androgen receptor ( AR) gene and genes encoding enzymes involved in synthesis of sex steroids (e.g., the CYP19 gene encoding aromatase) have recently received attention in osteoporosis research. Steroids 106-114 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 126-131 14744739-5 2004 Compared with noncarriers, women carrying two CYP19 7r(-3) alleles had 26% lower estrone (P < 0.001), 19% lower estradiol (P = 0.01), 23% lower free estradiol (P = 0.01), and 22% higher sex hormone-binding globulin concentrations (P = 0.06). Estrone 81-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-51 14517714-3 2004 We genotyped 1792 women for the CYP19 (TTTA)(n) repeat [short (TTTA)(n <or= 7) or long (TTTA)(n > 7)] the CYP19 C(1558)-T, and the AR (CAG)(n) repeat polymorphism [short (CAG)(n < 22), long (CAG)(n >or= 22)], and investigated associations with bone mineral density (BMD) and 5-year change in BMD. ttta 39-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-37 14517714-3 2004 We genotyped 1792 women for the CYP19 (TTTA)(n) repeat [short (TTTA)(n <or= 7) or long (TTTA)(n > 7)] the CYP19 C(1558)-T, and the AR (CAG)(n) repeat polymorphism [short (CAG)(n < 22), long (CAG)(n >or= 22)], and investigated associations with bone mineral density (BMD) and 5-year change in BMD. ttta 39-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 112-117 14517714-3 2004 We genotyped 1792 women for the CYP19 (TTTA)(n) repeat [short (TTTA)(n <or= 7) or long (TTTA)(n > 7)] the CYP19 C(1558)-T, and the AR (CAG)(n) repeat polymorphism [short (CAG)(n < 22), long (CAG)(n >or= 22)], and investigated associations with bone mineral density (BMD) and 5-year change in BMD. ttta 63-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-37 14744739-5 2004 Compared with noncarriers, women carrying two CYP19 7r(-3) alleles had 26% lower estrone (P < 0.001), 19% lower estradiol (P = 0.01), 23% lower free estradiol (P = 0.01), and 22% higher sex hormone-binding globulin concentrations (P = 0.06). Estradiol 115-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-51 14744739-5 2004 Compared with noncarriers, women carrying two CYP19 7r(-3) alleles had 26% lower estrone (P < 0.001), 19% lower estradiol (P = 0.01), 23% lower free estradiol (P = 0.01), and 22% higher sex hormone-binding globulin concentrations (P = 0.06). Estradiol 152-161 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-51 14744739-6 2004 Compared with noncarriers, women carrying at least one CYP19 8r allele had 20% higher estrone (P = 0.003), 18% higher estradiol (P = 0.02), and 21% higher free estradiol concentrations (P = 0.01). Estrone 86-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 55-60 14744739-6 2004 Compared with noncarriers, women carrying at least one CYP19 8r allele had 20% higher estrone (P = 0.003), 18% higher estradiol (P = 0.02), and 21% higher free estradiol concentrations (P = 0.01). Estradiol 118-127 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 55-60 14744739-6 2004 Compared with noncarriers, women carrying at least one CYP19 8r allele had 20% higher estrone (P = 0.003), 18% higher estradiol (P = 0.02), and 21% higher free estradiol concentrations (P = 0.01). Estradiol 160-169 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 55-60 14734494-8 2004 Elevated prostaglandin E2 levels from cyclooxygenase-2 induction by preinvasive and invasive breast lesions increase a number of tumor-promoting pathways, including aromatase, as well as angiogenetic, antiapoptotic, and others. Dinoprostone 9-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 165-174 14715870-4 2004 The CYP 19 genotype was significantly associated with serum estradiol (P = 0.002). Estradiol 60-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-10 14993062-1 2003 The enzyme aromatase catalyzes the conversion of testosterone and other C19 steroids to estradiol. Testosterone 49-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-20 14709151-0 2004 Forskolin up-regulates aromatase (CYP19) activity and gene transcripts in the human adrenocortical carcinoma cell line H295R. Colforsin 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-39 14709151-4 2004 Aromatase activity and the transcript of the CYP19 gene were highly up-regulated by forskolin, but not by dexamethasone. Colforsin 84-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-50 14709151-4 2004 Aromatase activity and the transcript of the CYP19 gene were highly up-regulated by forskolin, but not by dexamethasone. Dexamethasone 106-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-50 14670125-7 2003 High levels of functional IL-12 (26.78 +/- 4.11 ng/ml per 10(6) cells per 48 hr) were produced by scIL-12-transfected ARO cells (ARO/IL-12). scil 98-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 118-121 14670125-7 2003 High levels of functional IL-12 (26.78 +/- 4.11 ng/ml per 10(6) cells per 48 hr) were produced by scIL-12-transfected ARO cells (ARO/IL-12). scil 98-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 14993062-1 2003 The enzyme aromatase catalyzes the conversion of testosterone and other C19 steroids to estradiol. Steroids 76-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-20 14993062-1 2003 The enzyme aromatase catalyzes the conversion of testosterone and other C19 steroids to estradiol. Estradiol 88-97 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-20 14679019-5 2003 Using UV absorbance analysis, high-performance liquid chromatography profiling, accurate mass-mass spectrometry, and nanospray tandem mass spectrometry, most of the compounds in our red wine fraction were identified as procyanidin B dimers that were shown to be aromatase inhibitors. procyanidin B 219-232 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 262-271 14679019-7 2003 Inhibition kinetic analysis on the most potent procyanidin B dimer has revealed that it competes with the binding of the androgen substrate with a K(i) value of 6 micro M. Because mutations at Asp-309, Ser-378, and His-480 of aromatase significantly affected the binding of the procyanidin B dimer, these active site residues are thought to be important residues that interact with this phytochemical. procyanidin B 47-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 226-235 14679019-8 2003 The in vivo efficacy of procyanidin B dimers was evaluated in an aromatase-transfected MCF-7 breast cancer xenograft model. procyanidin B 24-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 14575840-4 2003 Mice transplanted with the human cell line of UTC ARO have a selective uptake of (10)B-borophenylalanine (BPA). (10)b-borophenylalanine 81-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-53 14698204-3 2003 As far as 17beta-estradiol (E(2)) production is concerned, it has been well demonstrated that its biosynthesis in the placenta involves the action of P450 aromatase (P450arom) and 17beta-hydroxysteroid dehydrogenase type 1 (17HSD1). Estradiol 10-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 150-164 14698204-3 2003 As far as 17beta-estradiol (E(2)) production is concerned, it has been well demonstrated that its biosynthesis in the placenta involves the action of P450 aromatase (P450arom) and 17beta-hydroxysteroid dehydrogenase type 1 (17HSD1). Estradiol 10-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 166-174 14575840-4 2003 Mice transplanted with the human cell line of UTC ARO have a selective uptake of (10)B-borophenylalanine (BPA). bisphenol A 106-109 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-53 14981949-3 2003 The genetic polymorphism of the CYP19 intron 4 [TTTA]n tetranucleotide has been studied in relation to breast cancer susceptibility. tetranucleotide 55-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-37 14981949-4 2003 MATERIALS AND METHODS: We examined the association of the tetranucleotide repeat polymorphism of the CYP19 gene with familial prostate cancer risk in a Japanese population by performing a case-control study consisting of 99 familial prostate cancer cases and 116 normal controls. tetranucleotide 58-73 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-106 14602139-8 2003 The combination of CYP19 (TTTA)7(-3bp) and CYP1A1 6235C/T polymorphisms is associated with an ER-positive, but not ER-negative, breast cancer risk, and, thus, would be useful in the selection of candidates for chemoprevention with tamoxifen. Tamoxifen 231-240 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 19-24 14584882-4 2003 Two genes, Atp6a3 (TCIRG1) and ClCN7, have been shown to be associated with human ARO, the latter of which is also thought to be responsible for ADO-II. ado-ii 145-151 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-85 12972182-5 2003 Transfection studies using granulosa cells demonstrated that LRH-1 is a potent regulator of both basal and forskolin-induced transcription of the ovary-specific hCYP19 promoter. Colforsin 107-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 161-167 12808001-0 2003 Aromatase is the major enzyme metabolizing buprenorphine in human placenta. Buprenorphine 43-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 12808001-6 2003 Compounds with selective affinity to the enzyme aromatase (CYP 19), namely 4-hydroxyandrostenedione and aminoglutethimide, caused >70% inhibition of norBUP formation. formestane 75-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-57 12808001-6 2003 Compounds with selective affinity to the enzyme aromatase (CYP 19), namely 4-hydroxyandrostenedione and aminoglutethimide, caused >70% inhibition of norBUP formation. formestane 75-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-65 12808001-6 2003 Compounds with selective affinity to the enzyme aromatase (CYP 19), namely 4-hydroxyandrostenedione and aminoglutethimide, caused >70% inhibition of norBUP formation. Aminoglutethimide 104-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-57 12808001-6 2003 Compounds with selective affinity to the enzyme aromatase (CYP 19), namely 4-hydroxyandrostenedione and aminoglutethimide, caused >70% inhibition of norBUP formation. Aminoglutethimide 104-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-65 12917334-1 2003 In the human placental syncytiotrophoblast, C(19) steroids are converted to estrogens by aromatase P450, product of the CYP19 gene. Steroids 50-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 120-125 14623539-7 2003 Whilst drugs such as anastrozole, exemestane, formestane and letrozole are all effective and specific inhibitors of aromatase, they differ in structure, potency and mechanism of action. Anastrozole 21-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-125 14623539-7 2003 Whilst drugs such as anastrozole, exemestane, formestane and letrozole are all effective and specific inhibitors of aromatase, they differ in structure, potency and mechanism of action. exemestane 34-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-125 14623539-7 2003 Whilst drugs such as anastrozole, exemestane, formestane and letrozole are all effective and specific inhibitors of aromatase, they differ in structure, potency and mechanism of action. formestane 46-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-125 14623539-7 2003 Whilst drugs such as anastrozole, exemestane, formestane and letrozole are all effective and specific inhibitors of aromatase, they differ in structure, potency and mechanism of action. Letrozole 61-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-125 14623550-4 2003 The prostaglandin PGE2 increases intracellular cAMP levels and stimulates estrogen biosynthesis, and previous studies in our laboratories have shown a strong linear association between aromatase (CYP19) expression and expression of the cyclooxygenases (COX-1 and COX-2) in breast cancer specimens. Prostaglandins 4-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 196-201 14623550-4 2003 The prostaglandin PGE2 increases intracellular cAMP levels and stimulates estrogen biosynthesis, and previous studies in our laboratories have shown a strong linear association between aromatase (CYP19) expression and expression of the cyclooxygenases (COX-1 and COX-2) in breast cancer specimens. Dinoprostone 18-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 196-201 14623550-4 2003 The prostaglandin PGE2 increases intracellular cAMP levels and stimulates estrogen biosynthesis, and previous studies in our laboratories have shown a strong linear association between aromatase (CYP19) expression and expression of the cyclooxygenases (COX-1 and COX-2) in breast cancer specimens. Cyclic AMP 47-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 196-201 14623515-9 2003 Thus, in the ovary, CYP19 expression is regulated by FSH which acts through cyclic AMP via the proximal promoter II, whereas in placenta the distal promoter I.1 regulates CYP19 expression in response to retinoids. Cyclic AMP 76-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-25 14623515-9 2003 Thus, in the ovary, CYP19 expression is regulated by FSH which acts through cyclic AMP via the proximal promoter II, whereas in placenta the distal promoter I.1 regulates CYP19 expression in response to retinoids. Retinoids 203-212 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 171-176 14623515-15 2003 This increased CYP19 expression is associated with a switch in promoter usage from the normal adipose-specific promoter I.4 to the cyclic AMP responsive promoter, promoter II. Cyclic AMP 131-141 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-20 12917334-3 2003 On the other hand, when cytotrophoblasts are cultured in 2% O(2), syncytiotrophoblast differentiation and induction of CYP19 expression are prevented. o(2) 60-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 119-124 12957661-7 2003 The mechanism responsible for the superiority of the aromatase inhibitors relates to the estrogen agonistic effects of tamoxifen. Tamoxifen 119-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 12849724-0 2003 Aromatase activity modulation by lindane and bisphenol-A in human placental JEG-3 and transfected kidney E293 cells. Hexachlorocyclohexane 33-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 14611865-5 2003 Furthermore, aromatase mediates neuroprotective effects of precursors of estradiol such as pregnenolone, dehydroepiandrosterone (DHEA) and testosterone. Estradiol 73-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 14611865-5 2003 Furthermore, aromatase mediates neuroprotective effects of precursors of estradiol such as pregnenolone, dehydroepiandrosterone (DHEA) and testosterone. Pregnenolone 91-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 14611865-5 2003 Furthermore, aromatase mediates neuroprotective effects of precursors of estradiol such as pregnenolone, dehydroepiandrosterone (DHEA) and testosterone. Dehydroepiandrosterone 105-127 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 14611865-5 2003 Furthermore, aromatase mediates neuroprotective effects of precursors of estradiol such as pregnenolone, dehydroepiandrosterone (DHEA) and testosterone. Dehydroepiandrosterone 129-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 14611865-5 2003 Furthermore, aromatase mediates neuroprotective effects of precursors of estradiol such as pregnenolone, dehydroepiandrosterone (DHEA) and testosterone. Testosterone 139-151 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 12871867-12 2003 CONCLUSIONS: Similar to CC, aromatase inhibition with letrozole reduces FSH dose required for COS without the undesirable antiestrogenic effects sometimes seen with CC. Letrozole 54-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-37 12871867-12 2003 CONCLUSIONS: Similar to CC, aromatase inhibition with letrozole reduces FSH dose required for COS without the undesirable antiestrogenic effects sometimes seen with CC. carbonyl sulfide 94-97 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-37 12672661-6 2003 Reductase expression did not change with stage of follicular development, but cytochrome b5, P450c17, and P450arom were markedly lower in post-LH tissues. Luteinizing Hormone 143-145 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 106-114 12849724-0 2003 Aromatase activity modulation by lindane and bisphenol-A in human placental JEG-3 and transfected kidney E293 cells. bisphenol A 45-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 12849724-3 2003 Since lindane and bisphenol-A (BPA) are two well-characterized endocrine disruptors that have been detected in animals and humans, it was important to learn whether they could affect aromatase activity and consequently estrogen biosynthesis. Hexachlorocyclohexane 6-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 183-192 12849724-3 2003 Since lindane and bisphenol-A (BPA) are two well-characterized endocrine disruptors that have been detected in animals and humans, it was important to learn whether they could affect aromatase activity and consequently estrogen biosynthesis. bisphenol A 18-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 183-192 12849724-3 2003 Since lindane and bisphenol-A (BPA) are two well-characterized endocrine disruptors that have been detected in animals and humans, it was important to learn whether they could affect aromatase activity and consequently estrogen biosynthesis. bisphenol A 31-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 183-192 12849724-10 2003 Therefore, lindane and BPA modulate aromatase activity suggesting an interaction with the cytochrome P-450 aromatase. Hexachlorocyclohexane 11-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-45 12849724-10 2003 Therefore, lindane and BPA modulate aromatase activity suggesting an interaction with the cytochrome P-450 aromatase. Hexachlorocyclohexane 11-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-116 12849724-10 2003 Therefore, lindane and BPA modulate aromatase activity suggesting an interaction with the cytochrome P-450 aromatase. bisphenol A 23-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-45 12849724-10 2003 Therefore, lindane and BPA modulate aromatase activity suggesting an interaction with the cytochrome P-450 aromatase. bisphenol A 23-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-116 12843186-7 2003 After 6 d of treatment with 50 nM geldanamycin, the percent inhibition of growth was 29.4% in TPC-1, 97.5% in FTC-133, 96.7% in FTC-236, 10.8% in FTC-238, 70.9% in XTC-1, and 45.5% in ARO cell lines. geldanamycin 34-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 184-187 14513436-3 2003 Promising results with the third-generation aromatase inhibitors anastrozole, letrozole, and exemestane in first- and second-line treatment of metastatic breast cancer has prompted their evaluation as adjuvant therapy in patients progressing on tamoxifen or as alternative first-line treatment. Anastrozole 65-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-53 14513436-3 2003 Promising results with the third-generation aromatase inhibitors anastrozole, letrozole, and exemestane in first- and second-line treatment of metastatic breast cancer has prompted their evaluation as adjuvant therapy in patients progressing on tamoxifen or as alternative first-line treatment. Letrozole 78-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-53 14513436-3 2003 Promising results with the third-generation aromatase inhibitors anastrozole, letrozole, and exemestane in first- and second-line treatment of metastatic breast cancer has prompted their evaluation as adjuvant therapy in patients progressing on tamoxifen or as alternative first-line treatment. exemestane 93-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-53 14513436-3 2003 Promising results with the third-generation aromatase inhibitors anastrozole, letrozole, and exemestane in first- and second-line treatment of metastatic breast cancer has prompted their evaluation as adjuvant therapy in patients progressing on tamoxifen or as alternative first-line treatment. Tamoxifen 245-254 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-53 12945785-4 2003 A special catalytic function is proposed for decomposition of the tetrahedral addition intermediate (T+/-) via k3 whereby the catalytic imidazole interacts electrophilically with the leaving phenolate ion and removes a proton from the nitrogen in the rate limiting step with subsequent non-rate limiting ArO-C bond fission. imidazole 136-145 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 304-307 12843139-6 2003 The woman was treated with anastrazole, a potent aromatase inhibitor, with normalization of menstrual cycles. Anastrozole 27-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-58 12792733-5 2003 These hNIS-transduced cells actively transported iodide into the cytoplasm at the level of 11635.3, 61571.6, and 19367.5 pmoles/10(6) cells in ARO, FRO, and NPA, respectively. Iodides 49-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 143-146 12945785-4 2003 A special catalytic function is proposed for decomposition of the tetrahedral addition intermediate (T+/-) via k3 whereby the catalytic imidazole interacts electrophilically with the leaving phenolate ion and removes a proton from the nitrogen in the rate limiting step with subsequent non-rate limiting ArO-C bond fission. Nitrogen 235-243 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 304-307 12846424-1 2003 Estrogen biosynthesis from C19 steroids is catalyzed by aromatase cytochrome P450. Steroids 31-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-65 12846424-3 2003 Breast tumors, however, secrete soluble factors that over-stimulate aromatase expression through an alternative proximal cAMP-responsive promoter, promoter II. Cyclic AMP 121-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-77 12846424-4 2003 We have mapped the cAMP-responsive regions of promoter II by transient transfection of 3T3-L1 preadipocytes with aromatase promoter II reporter genes. Cyclic AMP 19-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-122 12618873-2 2003 The results suggest that the CYP19 Arg(264)Cys polymorphism modifies breast cancer risk (OR=1.5, 95% CI=1.1-2.2), especially in association with alcohol consumption (P for interaction=0.04), whereas the CYP1B1 Leu(432)Val polymorphism appears to play no role here. Alcohols 145-152 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-34 12938779-4 2003 METHODS: Ten male patients with a diagnosis of IHH/PE were treated with the aromatase inhibitor anastrazole (1 mg/d orally). Anastrozole 96-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 12938779-8 2003 CONCLUSION: These results suggest that aromatase inhibition with anastrazole may provide a practical and efficacious alternative for the treatment of IHH but is not effective in preventing premature ejaculation. Anastrozole 65-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-48 12699753-0 2003 Preparation and pharmacological profile of 7-(alpha-azolylbenzyl)-1H-indoles and indolines as new aromatase inhibitors. 7-(alpha-azolylbenzyl)-1h-indoles 43-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-107 12699753-0 2003 Preparation and pharmacological profile of 7-(alpha-azolylbenzyl)-1H-indoles and indolines as new aromatase inhibitors. indoline 81-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-107 12699753-2 2003 New series of 7-(alpha-azolylbenzyl)-1H-indoles and indolines were synthesized as non-steroidal inhibitors of P450 arom. 7-(alpha-azolylbenzyl)-1h-indoles 14-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 110-119 12699753-2 2003 New series of 7-(alpha-azolylbenzyl)-1H-indoles and indolines were synthesized as non-steroidal inhibitors of P450 arom. indoline 52-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 110-119 12699753-4 2003 The most active compound, 1-ethyl-7-[(imidazol-1-yl)(4-chlorophenyl)methyl]-1H-indole 12c exhibited promising relative potency (rp) of 336 (rp of aminoglutethimide=1) and most of the described azoles were active and selective towards P450 arom. 1-ethyl-7-[(imidazol-1-yl)(4-chlorophenyl)methyl]-1h-indole 12c 26-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 234-243 12720534-7 2003 Vitamin D, dexamethasone, 17beta-estradiol and testosterone increased transcript levels of CYP19, whereas interleukin-1beta or tumor necrosis factor alpha decreased them. Estradiol 26-42 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-96 12720534-7 2003 Vitamin D, dexamethasone, 17beta-estradiol and testosterone increased transcript levels of CYP19, whereas interleukin-1beta or tumor necrosis factor alpha decreased them. Testosterone 47-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-96 12720534-10 2003 CONCLUSIONS: Our results suggested that vitamin D, testosterone, estrogens and glucocorticoids regulate CYP19 gene expression in human primary osteoblasts and the main promoter used appears to be promoter I.4. Vitamin D 40-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 104-109 12720534-10 2003 CONCLUSIONS: Our results suggested that vitamin D, testosterone, estrogens and glucocorticoids regulate CYP19 gene expression in human primary osteoblasts and the main promoter used appears to be promoter I.4. Testosterone 51-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 104-109 12722879-1 2003 The third generation of aromatase inhibitors and inactivators, such as anastrozole (Arimidex), letrozole (Femara) and exemestane (Aromasin), have become available for treatment of postmenopausal breast cancer patients. Anastrozole 71-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-33 12722879-1 2003 The third generation of aromatase inhibitors and inactivators, such as anastrozole (Arimidex), letrozole (Femara) and exemestane (Aromasin), have become available for treatment of postmenopausal breast cancer patients. Anastrozole 84-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-33 12722879-1 2003 The third generation of aromatase inhibitors and inactivators, such as anastrozole (Arimidex), letrozole (Femara) and exemestane (Aromasin), have become available for treatment of postmenopausal breast cancer patients. Letrozole 95-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-33 12722879-1 2003 The third generation of aromatase inhibitors and inactivators, such as anastrozole (Arimidex), letrozole (Femara) and exemestane (Aromasin), have become available for treatment of postmenopausal breast cancer patients. Letrozole 106-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-33 12722879-1 2003 The third generation of aromatase inhibitors and inactivators, such as anastrozole (Arimidex), letrozole (Femara) and exemestane (Aromasin), have become available for treatment of postmenopausal breast cancer patients. exemestane 118-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-33 12722879-1 2003 The third generation of aromatase inhibitors and inactivators, such as anastrozole (Arimidex), letrozole (Femara) and exemestane (Aromasin), have become available for treatment of postmenopausal breast cancer patients. exemestane 130-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-33 12690088-2 2003 Polymorphisms of a tetranucleotide repeat [TTTA](n) in the fourth intron of the CYP19 gene are associated with endocrine-dependent diseases and were examined in relation to hormone levels and disease risk factors in premenopausal women. tetranucleotide 19-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 80-85 12720534-4 2003 The effects of vitamin D and other factors on CYP19 expression were analysed by semiquantitative RT-PCR. Vitamin D 15-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-51 12720534-7 2003 Vitamin D, dexamethasone, 17beta-estradiol and testosterone increased transcript levels of CYP19, whereas interleukin-1beta or tumor necrosis factor alpha decreased them. Vitamin D 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-96 12720534-7 2003 Vitamin D, dexamethasone, 17beta-estradiol and testosterone increased transcript levels of CYP19, whereas interleukin-1beta or tumor necrosis factor alpha decreased them. Dexamethasone 11-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-96 12759877-3 2003 Because vitamin D regulates CYP19 gene expression, we also tested for an interaction with a translation start site polymorphism in the vitamin D receptor (VDR) gene. Vitamin D 8-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-33 12683828-1 2003 The synthesis and spectroscopic characterization of the mononuclear uranium complex [((ArO)(3)tacn)U(III)(NCCH(3))] is reported. Uranium 68-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-90 12683828-2 2003 The uranium(III) complex reacts with organic azides to yield uranium(IV) azido as well as uranium(V) imido complexes, [((ArO)(3)tacn)U(IV)(N(3))] and [((ArO)(3)tacn)U(V)(NSi(CH(3))(3))]. Azides 45-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 121-124 12683828-2 2003 The uranium(III) complex reacts with organic azides to yield uranium(IV) azido as well as uranium(V) imido complexes, [((ArO)(3)tacn)U(IV)(N(3))] and [((ArO)(3)tacn)U(V)(NSi(CH(3))(3))]. Azides 45-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 153-156 12606012-1 2003 CYP19 (P450arom) catalyzes the aromatization reaction of C19 steroids leading to estrogens. Steroids 61-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-5 12732294-5 2003 Aromatase activity was assessed using two different methodologies: in 19 cases by definitive product isolation (DPI) and in 42 cases by tritium-release assay (TRA). 3-aminodiphenyleneiodium 112-115 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 12732294-5 2003 Aromatase activity was assessed using two different methodologies: in 19 cases by definitive product isolation (DPI) and in 42 cases by tritium-release assay (TRA). Tritium 136-143 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 12732294-13 2003 The nature of mRNA transcripts in muscle suggests that estrogen formation may be controlled by glucocorticoid- as well as cAMP-dependent promoters of the aromatase gene. Cyclic AMP 122-126 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-163 12606012-1 2003 CYP19 (P450arom) catalyzes the aromatization reaction of C19 steroids leading to estrogens. Steroids 61-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 7-15 12606012-4 2003 Although the N-terminal replacement alone was not sufficient for the expression, human P450arom was successfully expressed up to the level of 240nmol/l culture by the combination of the N-terminal replacement and the induction of cold stress response by 1 microg/ml chloramphenicol. Chloramphenicol 266-281 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-95 12606012-5 2003 Membrane fractions containing the expressed P450arom catalyzed aromatization of androstenedione with a specific activity of 4.9 nmol/min/nmol P450. Androstenedione 80-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-52 12444780-10 2002 Reaction of Al(2)(O-t-Bu)(6) with [Sm(OAr)(3)](2) (Ar = 2,6-i-Pr(2)C(6)H(3)) yields the adduct (ArO)(3)Sm[(mu-O-t-Bu)(2)Al(2)(O-t-Bu)(4)] (5), which crystallizes in the space group P2(1)/n. al(2)(o-t-bu 12-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-99 12444780-10 2002 Reaction of Al(2)(O-t-Bu)(6) with [Sm(OAr)(3)](2) (Ar = 2,6-i-Pr(2)C(6)H(3)) yields the adduct (ArO)(3)Sm[(mu-O-t-Bu)(2)Al(2)(O-t-Bu)(4)] (5), which crystallizes in the space group P2(1)/n. Aluminum 12-14 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-99 12444780-10 2002 Reaction of Al(2)(O-t-Bu)(6) with [Sm(OAr)(3)](2) (Ar = 2,6-i-Pr(2)C(6)H(3)) yields the adduct (ArO)(3)Sm[(mu-O-t-Bu)(2)Al(2)(O-t-Bu)(4)] (5), which crystallizes in the space group P2(1)/n. )(o-t-bu 16-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-99 12589939-1 2002 Aromatase, the product of the CYP19 gene, plays a key role in androgenic steroids transformation into estrogens from various hormonal sensitive tissues. Steroids 73-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 12429043-7 2002 A point mutation creating a cAMP response element (CRE) in the reporter pTRCAT5"-146 CRE led to T3-induced suppression of the reporter gene in ARO cells without changing the basal or T3-induced activities in ARO1 and ARO2 cells. Cyclic AMP 28-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 143-146 12429043-7 2002 A point mutation creating a cAMP response element (CRE) in the reporter pTRCAT5"-146 CRE led to T3-induced suppression of the reporter gene in ARO cells without changing the basal or T3-induced activities in ARO1 and ARO2 cells. Cyclic AMP 28-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 208-212 12499641-0 2002 Aromatase inhibition by 4 beta,5 beta-epoxides of 16 alpha-hydroxyandrostenedione and its 19-oxygenated analogs, potential precursors of estriol production in the feto-placental unit. 4 beta,5 beta-epoxides 24-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 12499641-0 2002 Aromatase inhibition by 4 beta,5 beta-epoxides of 16 alpha-hydroxyandrostenedione and its 19-oxygenated analogs, potential precursors of estriol production in the feto-placental unit. 16-hydroxyandrost-4-en-3,17-dione 50-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 12499641-0 2002 Aromatase inhibition by 4 beta,5 beta-epoxides of 16 alpha-hydroxyandrostenedione and its 19-oxygenated analogs, potential precursors of estriol production in the feto-placental unit. Estriol 137-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 12499641-1 2002 To gain insight into the nature of the substrate binding site and the catalytic function of aromatase, we studied the inhibition of androstenedione aromatization by 4beta,5beta-epoxy-16alpha-hydroxyandrostenedione (4) and its 19-hydroxy and 19-oxo derivatives, 5 and 6, as well as the biochemical aromatization of these steroids in human placental microsomes. Androstenedione 132-147 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 12499641-1 2002 To gain insight into the nature of the substrate binding site and the catalytic function of aromatase, we studied the inhibition of androstenedione aromatization by 4beta,5beta-epoxy-16alpha-hydroxyandrostenedione (4) and its 19-hydroxy and 19-oxo derivatives, 5 and 6, as well as the biochemical aromatization of these steroids in human placental microsomes. 4beta 165-170 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 12499641-1 2002 To gain insight into the nature of the substrate binding site and the catalytic function of aromatase, we studied the inhibition of androstenedione aromatization by 4beta,5beta-epoxy-16alpha-hydroxyandrostenedione (4) and its 19-hydroxy and 19-oxo derivatives, 5 and 6, as well as the biochemical aromatization of these steroids in human placental microsomes. -oxo 243-247 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 12499641-1 2002 To gain insight into the nature of the substrate binding site and the catalytic function of aromatase, we studied the inhibition of androstenedione aromatization by 4beta,5beta-epoxy-16alpha-hydroxyandrostenedione (4) and its 19-hydroxy and 19-oxo derivatives, 5 and 6, as well as the biochemical aromatization of these steroids in human placental microsomes. Steroids 320-328 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 12499641-2 2002 The 19-methyl and 19-oxo compounds, 4 and 6, were weak competitive inhibitors of aromatase, with apparent K(i) values of 246 microM and 270 microM, respectively, whereas the 19-hydroxy compound 5 inhibited aromatase in a non-competitive manner with the K(i) of 135 microM. methyl radical 7-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-90 12499641-2 2002 The 19-methyl and 19-oxo compounds, 4 and 6, were weak competitive inhibitors of aromatase, with apparent K(i) values of 246 microM and 270 microM, respectively, whereas the 19-hydroxy compound 5 inhibited aromatase in a non-competitive manner with the K(i) of 135 microM. 19-oxo 18-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-90 12499641-2 2002 The 19-methyl and 19-oxo compounds, 4 and 6, were weak competitive inhibitors of aromatase, with apparent K(i) values of 246 microM and 270 microM, respectively, whereas the 19-hydroxy compound 5 inhibited aromatase in a non-competitive manner with the K(i) of 135 microM. 19-oxo 18-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 206-215 12589939-1 2002 Aromatase, the product of the CYP19 gene, plays a key role in androgenic steroids transformation into estrogens from various hormonal sensitive tissues. Steroids 73-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-35 12221600-0 2002 Aromatase inhibitory activities of standishinal and the diterpenoids from the bark of Thuja standishii. Diterpenes 56-68 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 12204674-5 2002 However, a 3-bp deletion/insertion CYP19 polymorphism, which is located in the same intron approximately 50 bp upstream to the (TTTA)n repeat, was evidently associated with the menopausal status in both the BC and biBC cohorts. ttta 128-132 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 35-40 12204674-5 2002 However, a 3-bp deletion/insertion CYP19 polymorphism, which is located in the same intron approximately 50 bp upstream to the (TTTA)n repeat, was evidently associated with the menopausal status in both the BC and biBC cohorts. bibc 214-218 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 35-40 12130549-7 2002 Reporter gene assays showed that troglitazone and LG101305 inhibit transcription from promoter II of the CYP19 gene. Troglitazone 33-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 105-110 12489563-1 2002 UNLABELLED: Human aromatase (CYP19) converts C19 androgens to aromatic C18 estrogenic steroids. Steroids 86-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-34 12489563-4 2002 We compared data from human placental assays with BD Gentest"s high throughput recombinant CYP19 enzyme assay using the fluorometric substrate dibenzylfluorescein. dibenzylfluorescein 143-162 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-96 12489563-15 2002 Recombinant CYP19 assay using the fluorometric substrate dibenzylfluorescein, demonstrates rapid screening potential for chemicals that may affect pregnancy outcome as a result of CYP19 inhibition. dibenzylfluorescein 57-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-17 12489563-15 2002 Recombinant CYP19 assay using the fluorometric substrate dibenzylfluorescein, demonstrates rapid screening potential for chemicals that may affect pregnancy outcome as a result of CYP19 inhibition. dibenzylfluorescein 57-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 180-185 12213901-5 2002 Prostaglandin E(2) stimulates cAMP formation, SF-1 binding activity, P450arom mRNA levels, and estrogen synthesis in endometriotic stromal cells. Dinoprostone 0-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-77 12213901-7 2002 Thus, we evaluated herein the possible roles of WT1 and DAX-1 in cAMP/SF-1-mediated regulation of P450arom expression in endometriotic and endometrial stromal cells. Cyclic AMP 65-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-106 12213901-10 2002 Site-directed disruption of the SF-1 binding site (-136/-124 bp) in the P450arom promoter abolished basal or cAMP/SF-1-induced promoter activity in the presence or absence of WT1 or DAX-1. Cyclic AMP 109-113 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 72-80 12213901-14 2002 In summary, WT1 or DAX-1 inhibits cAMP-SF-1 pathway-dependent P450arom expression in cultured human endometriotic and endometrial stromal cells. Cyclic AMP 34-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-70 12089342-1 2002 The porcine gonadal form of aromatase cytochrome P450 (P450arom) exhibits higher sensitivity to inhibition by the imidazole, etomidate, than the placental isozyme. imidazole 114-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-53 12107254-4 2002 Using RT-PCR and specific primers, we amplified the highly conserved helical, aromatic, and heme-binding sequences of the conventional human P450arom from RNA isolated from human spermatozoa. Heme 92-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 141-149 12428207-1 2002 Aromatase (P450arom) catalyzes the conversion of testosterone to estradiol, androstenedione to estrone, and 16a-hydroxylated dehydroepiandrosterone to estriol. Testosterone 49-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-19 12428207-1 2002 Aromatase (P450arom) catalyzes the conversion of testosterone to estradiol, androstenedione to estrone, and 16a-hydroxylated dehydroepiandrosterone to estriol. Estradiol 65-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-19 12428207-1 2002 Aromatase (P450arom) catalyzes the conversion of testosterone to estradiol, androstenedione to estrone, and 16a-hydroxylated dehydroepiandrosterone to estriol. Androstenedione 76-91 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-19 12428207-1 2002 Aromatase (P450arom) catalyzes the conversion of testosterone to estradiol, androstenedione to estrone, and 16a-hydroxylated dehydroepiandrosterone to estriol. Estrone 95-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-19 12428207-1 2002 Aromatase (P450arom) catalyzes the conversion of testosterone to estradiol, androstenedione to estrone, and 16a-hydroxylated dehydroepiandrosterone to estriol. 16a-hydroxylated dehydroepiandrosterone 108-147 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-19 12428207-1 2002 Aromatase (P450arom) catalyzes the conversion of testosterone to estradiol, androstenedione to estrone, and 16a-hydroxylated dehydroepiandrosterone to estriol. Estriol 151-158 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-19 12089342-1 2002 The porcine gonadal form of aromatase cytochrome P450 (P450arom) exhibits higher sensitivity to inhibition by the imidazole, etomidate, than the placental isozyme. imidazole 114-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 55-63 12089342-1 2002 The porcine gonadal form of aromatase cytochrome P450 (P450arom) exhibits higher sensitivity to inhibition by the imidazole, etomidate, than the placental isozyme. Etomidate 125-134 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-53 12089342-1 2002 The porcine gonadal form of aromatase cytochrome P450 (P450arom) exhibits higher sensitivity to inhibition by the imidazole, etomidate, than the placental isozyme. Etomidate 125-134 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 55-63 12089342-7 2002 The same mutation (I(133)M) introduced into human P450arom also markedly increased etomidate sensitivity. Etomidate 83-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-58 12089342-10 2002 These data suggest that Ile(133) is a contact residue in SRS-1 of P450arom, emphasize the functional conservation that exists in SRS-1 of a number of steroid-hydroxylating P450 enzymes, and suggest that substrate and inhibitor binding are dependent on different contact points to varying degrees. Isoleucine 24-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-74 12089342-10 2002 These data suggest that Ile(133) is a contact residue in SRS-1 of P450arom, emphasize the functional conservation that exists in SRS-1 of a number of steroid-hydroxylating P450 enzymes, and suggest that substrate and inhibitor binding are dependent on different contact points to varying degrees. Isoleucine 24-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-70 12089342-10 2002 These data suggest that Ile(133) is a contact residue in SRS-1 of P450arom, emphasize the functional conservation that exists in SRS-1 of a number of steroid-hydroxylating P450 enzymes, and suggest that substrate and inhibitor binding are dependent on different contact points to varying degrees. Steroids 150-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-74 12089342-10 2002 These data suggest that Ile(133) is a contact residue in SRS-1 of P450arom, emphasize the functional conservation that exists in SRS-1 of a number of steroid-hydroxylating P450 enzymes, and suggest that substrate and inhibitor binding are dependent on different contact points to varying degrees. Steroids 150-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-70 11927588-1 2002 Estrogen biosynthesis from C(19) steroids is catalyzed by aromatase cytochrome P450. Steroids 33-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-67 12127262-10 2002 Vinclozolin, as was shown previously for atrazine, induced aromatase activity and CYP19 mRNA levels about 2.5- and 1.5-fold, respectively. vinclozolin 0-11 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-87 12050258-8 2002 TBT caused dose-related increases in steady-state mRNA levels of both hCGbeta and CYP19 in JAR cells following 24- or 48-h exposure to nontoxic concentrations of TBT. tributyltin 0-3 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-87 12051723-0 2002 Novel oxazolidinone based compounds as inhibitors of aromatase and the use of the substrate-heme complex approach in the rationalisation of these compounds. Oxazolidinones 6-19 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 12050209-6 2002 The ONYX-015 virus worked synergistically with two antineoplastic drugs (doxorubicin and paclitaxel) in inducing ARO and KAT-4 cell death. Doxorubicin 73-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-116 12095950-8 2002 On the other hand, the potent effect of anti-aromatase chemicals in red wine overcomes the weak inductive effect of alcohol in wine. Alcohols 116-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-54 12095950-11 2002 An active Pinot Noir red wine SPE C18 column fraction (20% acetonitrile:water) was more effective than complete Pinot Noir wine in suppressing aromatase assay. acetonitrile 59-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 143-152 12095950-11 2002 An active Pinot Noir red wine SPE C18 column fraction (20% acetonitrile:water) was more effective than complete Pinot Noir wine in suppressing aromatase assay. Water 72-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 143-152 12050209-6 2002 The ONYX-015 virus worked synergistically with two antineoplastic drugs (doxorubicin and paclitaxel) in inducing ARO and KAT-4 cell death. Paclitaxel 89-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-116 12050258-8 2002 TBT caused dose-related increases in steady-state mRNA levels of both hCGbeta and CYP19 in JAR cells following 24- or 48-h exposure to nontoxic concentrations of TBT. tributyltin 162-165 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-87 12036037-1 2002 4Beta,19-dihydroxyandrost-5-en-17-one (6) is an excellent competitive inhibitor of estrogen synthetase (aromatase). 4beta 0-5 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-102 12137807-1 2002 Estrogen synthase (aromatase) catalyzes the aromatization of androstenedione (AD) as well as 16alpha-hydroxyandrostenedione (16alpha-OHAD) leading to estrone and estriol, respectively. Androstenedione 61-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-17 12137807-1 2002 Estrogen synthase (aromatase) catalyzes the aromatization of androstenedione (AD) as well as 16alpha-hydroxyandrostenedione (16alpha-OHAD) leading to estrone and estriol, respectively. Androstenedione 78-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-17 12137807-1 2002 Estrogen synthase (aromatase) catalyzes the aromatization of androstenedione (AD) as well as 16alpha-hydroxyandrostenedione (16alpha-OHAD) leading to estrone and estriol, respectively. 16-hydroxyandrost-4-en-3,17-dione 93-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-17 12137807-1 2002 Estrogen synthase (aromatase) catalyzes the aromatization of androstenedione (AD) as well as 16alpha-hydroxyandrostenedione (16alpha-OHAD) leading to estrone and estriol, respectively. 16-hydroxyandrost-4-en-3,17-dione 125-137 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-17 12137807-1 2002 Estrogen synthase (aromatase) catalyzes the aromatization of androstenedione (AD) as well as 16alpha-hydroxyandrostenedione (16alpha-OHAD) leading to estrone and estriol, respectively. Estrone 150-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-17 12137807-1 2002 Estrogen synthase (aromatase) catalyzes the aromatization of androstenedione (AD) as well as 16alpha-hydroxyandrostenedione (16alpha-OHAD) leading to estrone and estriol, respectively. Estriol 162-169 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-17 12036037-1 2002 4Beta,19-dihydroxyandrost-5-en-17-one (6) is an excellent competitive inhibitor of estrogen synthetase (aromatase). 19-dihydroxyandrost-5-en-17-one 6-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-102 11994385-1 2002 In human endometriotic stromal cells, markedly high levels of aromatase P450 (P450arom) mRNA and promoter II activity are present and can be vigorously stimulated by PGE(2) via a cAMP-dependent pathway to give rise to physiologically significant estrogen biosynthesis. Prostaglandins E 166-169 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-86 11994385-1 2002 In human endometriotic stromal cells, markedly high levels of aromatase P450 (P450arom) mRNA and promoter II activity are present and can be vigorously stimulated by PGE(2) via a cAMP-dependent pathway to give rise to physiologically significant estrogen biosynthesis. Cyclic AMP 179-183 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-86 12044960-5 2002 1,25-Dihydroxyvitamin D3 (1,25-(OH)(2)D(3)) alone did not induce the aromatase activity, but enhanced and maintained the glucocorticoid-induced P450AROM gene expression. Calcitriol 0-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 144-152 11933021-2 2002 Nonsteroidal aromatase inhibitors (NSAIs) are competitive inhibitors of aromatase, which bind to the enzyme active site by coordinating the iron atom present in the heme group of the P450 protein. Iron 140-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 11933021-2 2002 Nonsteroidal aromatase inhibitors (NSAIs) are competitive inhibitors of aromatase, which bind to the enzyme active site by coordinating the iron atom present in the heme group of the P450 protein. Iron 140-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 72-81 11933021-2 2002 Nonsteroidal aromatase inhibitors (NSAIs) are competitive inhibitors of aromatase, which bind to the enzyme active site by coordinating the iron atom present in the heme group of the P450 protein. Heme 165-169 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 11933021-2 2002 Nonsteroidal aromatase inhibitors (NSAIs) are competitive inhibitors of aromatase, which bind to the enzyme active site by coordinating the iron atom present in the heme group of the P450 protein. Heme 165-169 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 72-81 12044960-7 2002 These results may suggest that adrenal androgen, DHEA, is converted to estrone in osteoblast by P450AROM, which is positively regulated by glucocorticoid and 1,25-(OH)(2)D(3), and is important in maintaining BMD in the sixth to the seventh decade, after menopause. Dehydroepiandrosterone 49-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-104 12044960-7 2002 These results may suggest that adrenal androgen, DHEA, is converted to estrone in osteoblast by P450AROM, which is positively regulated by glucocorticoid and 1,25-(OH)(2)D(3), and is important in maintaining BMD in the sixth to the seventh decade, after menopause. Estrone 71-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-104 12030609-8 2002 EP-80874 was also able to antagonize the inhibitory activity of CST-14 on the growth of cells (ARO) expressing GHS-R but not sst. ep-80874 0-8 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-98 12113240-2 2002 Estradiol, the most potent endogenous estrogen, is biosynthesized from androgens by the cytochrome P450 enzyme complex called aromatase. Estradiol 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 126-135 12113240-7 2002 Mechanism-based steroidal inhibitors, such as 4-hydroxyandrostenedione and exemestane produce potent aromatase inhibition in patients. formestane 46-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-110 12113240-7 2002 Mechanism-based steroidal inhibitors, such as 4-hydroxyandrostenedione and exemestane produce potent aromatase inhibition in patients. exemestane 75-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-110 12113240-8 2002 The potent and selective third-generation aromatase inhibitors, anastrozole, letrozole and exemestane, are approved for clinical use as first-line endocrine therapy in postmenopausal women with metastatic hormone-dependent breast cancer and as second-line endocrine therapy in postmenopausal patients failing antiestrogen therapy alone or multiple hormonal therapies. Anastrozole 64-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 12113240-8 2002 The potent and selective third-generation aromatase inhibitors, anastrozole, letrozole and exemestane, are approved for clinical use as first-line endocrine therapy in postmenopausal women with metastatic hormone-dependent breast cancer and as second-line endocrine therapy in postmenopausal patients failing antiestrogen therapy alone or multiple hormonal therapies. Letrozole 77-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 12113240-8 2002 The potent and selective third-generation aromatase inhibitors, anastrozole, letrozole and exemestane, are approved for clinical use as first-line endocrine therapy in postmenopausal women with metastatic hormone-dependent breast cancer and as second-line endocrine therapy in postmenopausal patients failing antiestrogen therapy alone or multiple hormonal therapies. exemestane 91-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 11937133-1 2002 OBJECTIVE: To examine the use of the aromatase inhibitor letrozole with FSH for ovarian stimulation in poor responders undergoing ovarian superovulation and IUI. Letrozole 57-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-46 11925378-3 2002 RESULTS: The distribution of the genotypes of CYP17 and alleles of the TTTA repeat polymorphism of CYP19 were not significantly different between the groups. ttta 71-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-104 11904449-0 2002 Testosterone attenuates expression of vascular cell adhesion molecule-1 by conversion to estradiol by aromatase in endothelial cells: implications in atherosclerosis. Testosterone 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 102-111 11893526-1 2002 In humans, aromatase P450, which catalyses conversion of C(19)-steroids to estrogens, is expressed in several tissues, including gonads, brain, adipose tissue, skin and placenta, and is encoded by a single-copy gene (CYP19); however, this does not hold true for all species. c(19)-steroids 57-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 217-222 11904449-4 2002 We evaluated whether testosterone inhibited TNFalpha-induced VCAM-1 expression via its conversion to estradiol by the enzyme aromatase in human umbilical vein endothelial cells (HUVEC). Testosterone 21-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-134 11904449-4 2002 We evaluated whether testosterone inhibited TNFalpha-induced VCAM-1 expression via its conversion to estradiol by the enzyme aromatase in human umbilical vein endothelial cells (HUVEC). Estradiol 101-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-134 11904449-8 2002 Testosterone was less effective in attenuating VCAM-1 expression in the presence of anastrozole, an inhibitor of aromatase, indicating that testosterone inhibited VCAM-1 via conversion to estradiol. Testosterone 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-122 11904449-8 2002 Testosterone was less effective in attenuating VCAM-1 expression in the presence of anastrozole, an inhibitor of aromatase, indicating that testosterone inhibited VCAM-1 via conversion to estradiol. Anastrozole 84-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-122 11904449-12 2002 In conclusion, testosterone inhibited VCAM-1 mRNA and protein expression in HUVEC by its conversion to estradiol via the enzyme aromatase present in the endothelial cells. Testosterone 15-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 128-137 11904449-12 2002 In conclusion, testosterone inhibited VCAM-1 mRNA and protein expression in HUVEC by its conversion to estradiol via the enzyme aromatase present in the endothelial cells. Estradiol 103-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 128-137 11826265-5 2002 Within these sites, aromatase action can generate high levels of estradiol locally without significantly affecting circulating levels. Estradiol 65-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-29 11897504-4 2002 The prostaglandin PGE(2) increases intracellular cAMP levels and stimulates estrogen biosynthesis, and our recent studies have shown a strong linear association between CYP19 expression and the sum of cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) expression in breast cancer specimens. Prostaglandins 4-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-174 11897504-4 2002 The prostaglandin PGE(2) increases intracellular cAMP levels and stimulates estrogen biosynthesis, and our recent studies have shown a strong linear association between CYP19 expression and the sum of cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) expression in breast cancer specimens. Dinoprostone 18-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-174 11897504-4 2002 The prostaglandin PGE(2) increases intracellular cAMP levels and stimulates estrogen biosynthesis, and our recent studies have shown a strong linear association between CYP19 expression and the sum of cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) expression in breast cancer specimens. Cyclic AMP 49-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-174 11929341-0 2002 Tamoxifen resistant and refractory breast cancer: the value of aromatase inhibitors. Tamoxifen 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-72 12053085-5 2002 P450arom catalyzes the conversion of testosterone to estradiol, of androstenedione to estrone, and of 16alpha-hydroxylated dehydroepiandrosterone to estriol. Testosterone 37-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 12053085-5 2002 P450arom catalyzes the conversion of testosterone to estradiol, of androstenedione to estrone, and of 16alpha-hydroxylated dehydroepiandrosterone to estriol. Estradiol 53-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 12053085-5 2002 P450arom catalyzes the conversion of testosterone to estradiol, of androstenedione to estrone, and of 16alpha-hydroxylated dehydroepiandrosterone to estriol. Androstenedione 67-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 12053085-5 2002 P450arom catalyzes the conversion of testosterone to estradiol, of androstenedione to estrone, and of 16alpha-hydroxylated dehydroepiandrosterone to estriol. Estrone 86-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 12053085-5 2002 P450arom catalyzes the conversion of testosterone to estradiol, of androstenedione to estrone, and of 16alpha-hydroxylated dehydroepiandrosterone to estriol. 16alpha-hydroxylated dehydroepiandrosterone 102-145 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 12053085-5 2002 P450arom catalyzes the conversion of testosterone to estradiol, of androstenedione to estrone, and of 16alpha-hydroxylated dehydroepiandrosterone to estriol. Estriol 149-156 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 11786372-2 2002 Aromatase, an enzyme complex comprised of P450aromatase (P450arom) and NADH-cytochrome P450 reductase, converts androgens to estrogens, whereas 17HSD1 catalyzes the reduction of estrone to E(2). Estrone 178-185 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-55 11786372-2 2002 Aromatase, an enzyme complex comprised of P450aromatase (P450arom) and NADH-cytochrome P450 reductase, converts androgens to estrogens, whereas 17HSD1 catalyzes the reduction of estrone to E(2). Estrone 178-185 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-66 11786372-3 2002 In the present study, the effects of retinoic acids (RAs) on P450arom and 17HSD1 expression in placental cells were investigated. Tretinoin 37-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-69 11838734-5 2002 In in vitro studies, the uptake of p-10borophenylalanine (BPA) by the UTC cell line ARO, primary cultures of normal bovine thyroid cells (BT), and human follicular adenoma (FA) thyroid was studied. p-10borophenylalanine 35-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-87 11927281-0 2002 Aromatase inhibitors continue their ATAC on tamoxifen. Tamoxifen 44-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 11838734-5 2002 In in vitro studies, the uptake of p-10borophenylalanine (BPA) by the UTC cell line ARO, primary cultures of normal bovine thyroid cells (BT), and human follicular adenoma (FA) thyroid was studied. bisphenol A 58-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-87 11838734-7 2002 The uptake by quiescent ARO, BT, and FA showed that the ARO/BT and ARO/FA ratios were 4 and 5, respectively (p < 0.001). benzothiazole 60-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-27 11850235-4 2001 Constructs encoding the native and chimeric porcine and human P450arom enzymes were transiently expressed and activity was assessed using the tritiated water assay. Water 152-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-70 11916226-3 2001 The first clinically available aromatase inhibitor, aminoglutethimide, was introduced for the second-line treatment of advanced breast cancer in the late 1970s. Aminoglutethimide 52-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 11916226-6 2001 As a result, several aromatase inhibitors with a high degree of selectivity for aromatase and improved tolerability have become clinically available for the treatment of postmenopausal women with advanced breast cancer: (a) anastrozole; (b) letrozole; (c) fadrozole; (d) formestane; and (e) exemestane. Anastrozole 224-235 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 11916226-6 2001 As a result, several aromatase inhibitors with a high degree of selectivity for aromatase and improved tolerability have become clinically available for the treatment of postmenopausal women with advanced breast cancer: (a) anastrozole; (b) letrozole; (c) fadrozole; (d) formestane; and (e) exemestane. Letrozole 241-250 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 11916226-6 2001 As a result, several aromatase inhibitors with a high degree of selectivity for aromatase and improved tolerability have become clinically available for the treatment of postmenopausal women with advanced breast cancer: (a) anastrozole; (b) letrozole; (c) fadrozole; (d) formestane; and (e) exemestane. Fadrozole 256-265 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 11916226-6 2001 As a result, several aromatase inhibitors with a high degree of selectivity for aromatase and improved tolerability have become clinically available for the treatment of postmenopausal women with advanced breast cancer: (a) anastrozole; (b) letrozole; (c) fadrozole; (d) formestane; and (e) exemestane. formestane 271-281 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 11916226-6 2001 As a result, several aromatase inhibitors with a high degree of selectivity for aromatase and improved tolerability have become clinically available for the treatment of postmenopausal women with advanced breast cancer: (a) anastrozole; (b) letrozole; (c) fadrozole; (d) formestane; and (e) exemestane. exemestane 291-301 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 11916226-7 2001 Of these, formestane and exemestane are steroidal nonreversible aromatase inhibitors, also known as aromatase inactivators, whereas fadrozole, anastrozole, and letrozole are nonsteroidal reversible aromatase inhibitors. formestane 10-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-73 11916226-7 2001 Of these, formestane and exemestane are steroidal nonreversible aromatase inhibitors, also known as aromatase inactivators, whereas fadrozole, anastrozole, and letrozole are nonsteroidal reversible aromatase inhibitors. formestane 10-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-109 11916226-7 2001 Of these, formestane and exemestane are steroidal nonreversible aromatase inhibitors, also known as aromatase inactivators, whereas fadrozole, anastrozole, and letrozole are nonsteroidal reversible aromatase inhibitors. formestane 10-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-109 11916226-7 2001 Of these, formestane and exemestane are steroidal nonreversible aromatase inhibitors, also known as aromatase inactivators, whereas fadrozole, anastrozole, and letrozole are nonsteroidal reversible aromatase inhibitors. exemestane 25-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-73 11916226-7 2001 Of these, formestane and exemestane are steroidal nonreversible aromatase inhibitors, also known as aromatase inactivators, whereas fadrozole, anastrozole, and letrozole are nonsteroidal reversible aromatase inhibitors. exemestane 25-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-109 11916226-7 2001 Of these, formestane and exemestane are steroidal nonreversible aromatase inhibitors, also known as aromatase inactivators, whereas fadrozole, anastrozole, and letrozole are nonsteroidal reversible aromatase inhibitors. exemestane 25-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-109 11916223-1 2001 Aromatase is the key enzyme in the synthesis of estrogens and mediates the conversion of androstenedione and testosterone to estrone and estradiol. Androstenedione 89-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 11916223-1 2001 Aromatase is the key enzyme in the synthesis of estrogens and mediates the conversion of androstenedione and testosterone to estrone and estradiol. Testosterone 109-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 11916223-1 2001 Aromatase is the key enzyme in the synthesis of estrogens and mediates the conversion of androstenedione and testosterone to estrone and estradiol. Estrone 125-132 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 11916223-1 2001 Aromatase is the key enzyme in the synthesis of estrogens and mediates the conversion of androstenedione and testosterone to estrone and estradiol. Estradiol 137-146 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 11916223-3 2001 Aromatase is an excellent target for inhibition, because it is the last step in steroid biosynthesis, and, therefore, there are no important downstream enzymes to be affected. Steroids 80-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 11916223-5 2001 The approach we took to develop the first aromatase inhibitors was to design substrate analogues based on the structure of androstenedione. Androstenedione 123-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 11850224-9 2001 As a result, the inhibitory effect of aromatase activity by troglitazone plus LG100268 was accompanied by the decrease of P450arom mRNA level. Troglitazone 60-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-130 11850224-9 2001 As a result, the inhibitory effect of aromatase activity by troglitazone plus LG100268 was accompanied by the decrease of P450arom mRNA level. LG 100268 78-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-130 11850235-9 2001 Analysis of chimeric constructs indicated that the sensitivity to etomidate was associated with residues in the B, B" and C helices of the gonadal P450arom encompassing only one of six putative substrate recognition sites. Etomidate 66-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 147-155 11850235-11 2001 Therefore, it appears that residues of the porcine gonadal P450arom that are responsible for etomidate binding may be distinct from those involved in substrate recognition and metabolism. Etomidate 93-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-67 11600533-3 2001 In ARO and KAT-4 cells, manumycin- plus paclitaxel-induced DNA fragmentation was blocked by the inhibitors of caspase-9, caspase-8, and caspase-3. manumycin 24-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 3-6 11850206-5 2001 Since prostaglandin E2 is known to regulate aromatase gene expression and is the product of COX-2, an enzyme frequently overexpressed in tumors, immunocytochemistry was performed on the tissue sections using a polyclonal antibody to COX-2. Dinoprostone 6-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-53 11850206-16 2001 In addition, the duration of response was significantly longer with the aromatase inhibitor than with tamoxifen, suggesting that aromatase inhibitors may offer better control of tumor growth than this antiestrogen. Tamoxifen 102-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-138 11850209-10 2001 These results suggest that estrone and estradiol are produced in SMC of the human aortic wall and that their production is mediated by aromatase and 17beta-HSD I, respectively. Estradiol 39-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 11850210-4 2001 The prostaglandin E2 (PGE2) increases intracellular cAMP levels and stimulates estrogen biosynthesis, and our recent studies have shown a strong linear association between CYP19 expression and the sum of COX-1 and COX-2 expression in breast cancer specimens. Dinoprostone 4-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 172-177 11850210-4 2001 The prostaglandin E2 (PGE2) increases intracellular cAMP levels and stimulates estrogen biosynthesis, and our recent studies have shown a strong linear association between CYP19 expression and the sum of COX-1 and COX-2 expression in breast cancer specimens. Dinoprostone 22-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 172-177 11850210-4 2001 The prostaglandin E2 (PGE2) increases intracellular cAMP levels and stimulates estrogen biosynthesis, and our recent studies have shown a strong linear association between CYP19 expression and the sum of COX-1 and COX-2 expression in breast cancer specimens. Cyclic AMP 52-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 172-177 11708799-7 2001 The changes in the aromatase activity by TBT compounds were associated with comparable changes in P450arom mRNA assessed by RT-PCR. tributyltin 41-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-106 11708799-8 2001 The luciferase activity of the P450arom promoter II (1 kb) decreased after the addition of 20 ng/ml TBT compounds in transfected KGN cells either in a basic state or in states stimulated by db-cAMP. tributyltin 100-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-39 11708799-8 2001 The luciferase activity of the P450arom promoter II (1 kb) decreased after the addition of 20 ng/ml TBT compounds in transfected KGN cells either in a basic state or in states stimulated by db-cAMP. Bucladesine 190-197 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-39 11708799-10 2001 These results indicate that TBT compounds inhibited the aromatase activity and also decreased the P450arom mRNA level at the transcriptional level in KGN cells. tributyltin 28-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-106 11916146-0 2001 Enantioselectivity of some 1-[(benzofuran-2-yl) phenylmethyl] imidazoles as aromatase (P450AROM) inhibitors. 1-[(benzofuran-2-yl) phenylmethyl] imidazoles 27-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-95 11916146-1 2001 The enantioselectivity ratio ((+)-:(-)-forms) of three substituted 1-[(benzofuran-2-yl) phenylmethyl] imidazoles as inhibitors of aromatase (P450AROM) was 2.16, 12.3 and 1.0 for the 4-methyl-, 4-fluoro- and 4-chloro-substituted compounds, respectively. 1-[(benzofuran-2-yl) phenylmethyl] imidazoles 67-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 141-149 11916147-2 2001 The flavones were all evaluated in vitro for inhibitory activity against aromatase (P450AROM, CYP19), using human placental microsomes, and for inhibitory activity against 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD-1) using human placental cytosol. Flavones 4-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-92 11916147-2 2001 The flavones were all evaluated in vitro for inhibitory activity against aromatase (P450AROM, CYP19), using human placental microsomes, and for inhibitory activity against 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD-1) using human placental cytosol. Flavones 4-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-99 11916147-3 2001 The phenyl, 4-fluoro-phenyl and 4-bromo-phenyl derivatives displayed moderate inhibitory activity against P450AROM (IC50 17.2, 13.5 and 10.1 microM, respectively), none of the flavones, including the standard genistein, displayed any inhibitory activity against 17beta-HSD type 1 at 100 microM concentration. Genistein 209-218 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 106-114 11600533-3 2001 In ARO and KAT-4 cells, manumycin- plus paclitaxel-induced DNA fragmentation was blocked by the inhibitors of caspase-9, caspase-8, and caspase-3. Paclitaxel 40-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 3-6 11520936-7 2001 We illustrate here how comparative analysis of the distribution of the steroid-metabolizing enzyme estrogen synthetase (aromatase) may help to understand the differences and similarities in the limbic system and hypothalamus of birds and mammals. Steroids 71-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-118 11559181-4 2001 A semiempirical approach based on intersecting parabolas suggests that the observed reactivity is mainly related to the enthalpy of the reaction and allowed to estimate activation energies for the reaction of (4-X-C(6)H(4))(2)N* and ArO* radicals with a variety of silicon hydrides. 4-x-c 210-215 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 233-237 11559181-4 2001 A semiempirical approach based on intersecting parabolas suggests that the observed reactivity is mainly related to the enthalpy of the reaction and allowed to estimate activation energies for the reaction of (4-X-C(6)H(4))(2)N* and ArO* radicals with a variety of silicon hydrides. monosilane 265-281 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 233-237 11550075-2 2001 Estradiol, the most potent endogenous estrogen, is biosynthesized from androgens by the cytochrome P-450 enzyme complex called aromatase. Estradiol 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-136 11550075-7 2001 Competitive aromatase inhibitors are molecules that compete with the substrate androstenedione for noncovalent binding to the active site of the enzyme to decrease the amount of product formed. Androstenedione 79-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-21 11550075-9 2001 The structure-activity relationships for steroidal inhibitors have become more refined in the past decade, and only some modifications can be made to the steroid and still keep its affinity for aromatase. Steroids 41-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 194-203 11550075-10 2001 Nonsteroidal aromatase inhibitors can be divided into three classes: aminoglutethimide-like molecules, imidazole/triazole derivatives, and flavonoid analogs. Aminoglutethimide 69-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 11550075-10 2001 Nonsteroidal aromatase inhibitors can be divided into three classes: aminoglutethimide-like molecules, imidazole/triazole derivatives, and flavonoid analogs. imidazole 103-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 11550075-10 2001 Nonsteroidal aromatase inhibitors can be divided into three classes: aminoglutethimide-like molecules, imidazole/triazole derivatives, and flavonoid analogs. Triazoles 113-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 11550075-10 2001 Nonsteroidal aromatase inhibitors can be divided into three classes: aminoglutethimide-like molecules, imidazole/triazole derivatives, and flavonoid analogs. Flavonoids 139-148 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 11550075-14 2001 Mechanism-based steroidal inhibitors such as 4-hydroxyandrostenedione and exemestane produce prolonged aromatase inhibition in patients. formestane 45-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 103-112 11550075-14 2001 Mechanism-based steroidal inhibitors such as 4-hydroxyandrostenedione and exemestane produce prolonged aromatase inhibition in patients. exemestane 74-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 103-112 11550075-15 2001 The potent and selective third-generation aromatase inhibitors anastrozole, letrozole, and exemestane are approved for clinical use as second-line endocrine therapy in postmenopausal patients failing antiestrogen therapy alone or multiple hormonal therapies. Anastrozole 63-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 11550075-15 2001 The potent and selective third-generation aromatase inhibitors anastrozole, letrozole, and exemestane are approved for clinical use as second-line endocrine therapy in postmenopausal patients failing antiestrogen therapy alone or multiple hormonal therapies. Letrozole 76-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 11550075-15 2001 The potent and selective third-generation aromatase inhibitors anastrozole, letrozole, and exemestane are approved for clinical use as second-line endocrine therapy in postmenopausal patients failing antiestrogen therapy alone or multiple hormonal therapies. exemestane 91-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 11571725-6 2001 RESULTS: The distribution of the tetranucleotide simple tandem repeat polymorphism (STRP) in intron 4 of CYP19 was significantly different in control and cancer patients (P = 0.012). tetranucleotide 33-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 105-110 11403889-3 2001 This immature stage persists concomitantly with a dramatic enhancement of aromatase activity reversed by triiodothyronine (T3) either in vivo or in vitro administration. Triiodothyronine 105-121 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 74-83 12049480-0 2001 1-[(Benzofuran-2-yl)phenylmethyl]triazoles as steroidogenic inhibitors: synthesis and in vitro inhibition of human placental CYP19 aromatase. 1-[(benzofuran-2-yl)phenylmethyl]triazoles 0-42 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-130 12049480-1 2001 Hormone-dependent breast cancer is stimulated by the female hormones oestrone and oestradiol, therefore compounds which inhibit the specific enzymes involved in the formation of the nitogenic hormones, namely CYP19 aromatase (P450arom) and 17beta-hydroxysteroid dehydrogenase (17beta-HSD) type 1, are targets of therapeutic interest for the treatment of breast cancer. Estrone 69-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 209-214 12049480-1 2001 Hormone-dependent breast cancer is stimulated by the female hormones oestrone and oestradiol, therefore compounds which inhibit the specific enzymes involved in the formation of the nitogenic hormones, namely CYP19 aromatase (P450arom) and 17beta-hydroxysteroid dehydrogenase (17beta-HSD) type 1, are targets of therapeutic interest for the treatment of breast cancer. Estrone 69-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 226-234 12049480-1 2001 Hormone-dependent breast cancer is stimulated by the female hormones oestrone and oestradiol, therefore compounds which inhibit the specific enzymes involved in the formation of the nitogenic hormones, namely CYP19 aromatase (P450arom) and 17beta-hydroxysteroid dehydrogenase (17beta-HSD) type 1, are targets of therapeutic interest for the treatment of breast cancer. Estradiol 82-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 209-214 12049480-1 2001 Hormone-dependent breast cancer is stimulated by the female hormones oestrone and oestradiol, therefore compounds which inhibit the specific enzymes involved in the formation of the nitogenic hormones, namely CYP19 aromatase (P450arom) and 17beta-hydroxysteroid dehydrogenase (17beta-HSD) type 1, are targets of therapeutic interest for the treatment of breast cancer. Estradiol 82-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 226-234 11403889-3 2001 This immature stage persists concomitantly with a dramatic enhancement of aromatase activity reversed by triiodothyronine (T3) either in vivo or in vitro administration. Triiodothyronine 123-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 74-83 11403889-7 2001 The presence of a functional P450 aromatase protein in purified Sertoli cells was confirmed by the measurement [3H]H(2)O released after incubation with [1beta-(3)H]androst-4-3,17-dione. Tritium 112-114 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-43 11403889-7 2001 The presence of a functional P450 aromatase protein in purified Sertoli cells was confirmed by the measurement [3H]H(2)O released after incubation with [1beta-(3)H]androst-4-3,17-dione. 1beta-(3)h 153-163 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-43 11403889-7 2001 The presence of a functional P450 aromatase protein in purified Sertoli cells was confirmed by the measurement [3H]H(2)O released after incubation with [1beta-(3)H]androst-4-3,17-dione. androst-4-3,17-dione 164-184 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-43 11403889-10 2001 The detection of mRNA altered transcript coding for putative protein lacking both aromatic and heme-binding regions upon T3 treatment and unable to convert androgens into estrogens, provides a reasonable explanation for the observed discrepancies between aromatase protein pattern, P450arom mRNA levels and aromatase activity. Heme 95-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 255-264 11403889-10 2001 The detection of mRNA altered transcript coding for putative protein lacking both aromatic and heme-binding regions upon T3 treatment and unable to convert androgens into estrogens, provides a reasonable explanation for the observed discrepancies between aromatase protein pattern, P450arom mRNA levels and aromatase activity. Heme 95-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 282-290 11403889-10 2001 The detection of mRNA altered transcript coding for putative protein lacking both aromatic and heme-binding regions upon T3 treatment and unable to convert androgens into estrogens, provides a reasonable explanation for the observed discrepancies between aromatase protein pattern, P450arom mRNA levels and aromatase activity. Heme 95-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 307-316 11354475-2 2001 In basic media, these monomers self-polymerize via a series of nucleophilic aromatic substitution reactions (SNAr), in which aromatic enolates (ArO- nucleophiles) attack the electrophilic carbons bearing F leaving groups to effect fluoride displacement. aromatic enolates 125-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 144-147 11356934-3 2001 To synthesize sesquiterpene lactones with greater specificity for aromatase inhibition and lower cytotoxicity, we chemically reduced the alpha-methylene-gamma-lactone group in the active aromatase inhibitor 10-epi-8-deoxycumambrin B (compound 1), to obtain the new compound 11betaH,13-dihydro-10-epi-8-deoxycumambrin B (compound 2). 13-dihydro-10-epi-8-deoxycumambrin b 282-318 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 187-196 11356934-8 2001 We conclude that the anti-aromatase activity of sesquiterpene lactones does not depend on the presence of the highly reactive alpha-methylene-gamma-lactone group, whereas their cytotoxicity does. sesquiterpene lactones 48-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-35 11399122-7 2001 The increase in adipose tissue is associated with an increase in the enzyme aromatase that converts testosterone to estradiol and leads to diminished testosterone levels that favor the preferential deposition of visceral fat. Testosterone 100-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 11399122-7 2001 The increase in adipose tissue is associated with an increase in the enzyme aromatase that converts testosterone to estradiol and leads to diminished testosterone levels that favor the preferential deposition of visceral fat. Estradiol 116-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 11399122-7 2001 The increase in adipose tissue is associated with an increase in the enzyme aromatase that converts testosterone to estradiol and leads to diminished testosterone levels that favor the preferential deposition of visceral fat. Testosterone 150-162 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 11356934-0 2001 Aromatase inhibition by an 11,13-dihydroderivative of a sesquiterpene lactone. 11,13-dihydroderivative 27-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 11356934-0 2001 Aromatase inhibition by an 11,13-dihydroderivative of a sesquiterpene lactone. sesquiterpene lactone 56-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 11356934-2 2001 A group of sesquiterpene lactones inhibit aromatase activity and also exert cytotoxicity through their reactive alpha-methylene-gamma-lactone group. sesquiterpene lactones 11-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 11356934-3 2001 To synthesize sesquiterpene lactones with greater specificity for aromatase inhibition and lower cytotoxicity, we chemically reduced the alpha-methylene-gamma-lactone group in the active aromatase inhibitor 10-epi-8-deoxycumambrin B (compound 1), to obtain the new compound 11betaH,13-dihydro-10-epi-8-deoxycumambrin B (compound 2). sesquiterpene lactones 14-36 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-75 11356934-3 2001 To synthesize sesquiterpene lactones with greater specificity for aromatase inhibition and lower cytotoxicity, we chemically reduced the alpha-methylene-gamma-lactone group in the active aromatase inhibitor 10-epi-8-deoxycumambrin B (compound 1), to obtain the new compound 11betaH,13-dihydro-10-epi-8-deoxycumambrin B (compound 2). sesquiterpene lactones 14-36 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 187-196 11356934-3 2001 To synthesize sesquiterpene lactones with greater specificity for aromatase inhibition and lower cytotoxicity, we chemically reduced the alpha-methylene-gamma-lactone group in the active aromatase inhibitor 10-epi-8-deoxycumambrin B (compound 1), to obtain the new compound 11betaH,13-dihydro-10-epi-8-deoxycumambrin B (compound 2). 10-epi-8-deoxycumambrin b 207-232 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-75 11356934-3 2001 To synthesize sesquiterpene lactones with greater specificity for aromatase inhibition and lower cytotoxicity, we chemically reduced the alpha-methylene-gamma-lactone group in the active aromatase inhibitor 10-epi-8-deoxycumambrin B (compound 1), to obtain the new compound 11betaH,13-dihydro-10-epi-8-deoxycumambrin B (compound 2). 10-epi-8-deoxycumambrin b 207-232 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 187-196 11356934-3 2001 To synthesize sesquiterpene lactones with greater specificity for aromatase inhibition and lower cytotoxicity, we chemically reduced the alpha-methylene-gamma-lactone group in the active aromatase inhibitor 10-epi-8-deoxycumambrin B (compound 1), to obtain the new compound 11betaH,13-dihydro-10-epi-8-deoxycumambrin B (compound 2). 11betah 274-281 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 187-196 11354475-2 2001 In basic media, these monomers self-polymerize via a series of nucleophilic aromatic substitution reactions (SNAr), in which aromatic enolates (ArO- nucleophiles) attack the electrophilic carbons bearing F leaving groups to effect fluoride displacement. Carbon 188-195 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 144-147 11354475-2 2001 In basic media, these monomers self-polymerize via a series of nucleophilic aromatic substitution reactions (SNAr), in which aromatic enolates (ArO- nucleophiles) attack the electrophilic carbons bearing F leaving groups to effect fluoride displacement. Fluorides 231-239 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 144-147 11472114-3 2001 The one-electron reduction potentials of the aroxyl radicals corresponding to compounds 2a-2d, E degrees (ArO(*)/ArO(-)) were 0.49, 0.49, 0.49, and 0.52 V vs NHE, respectively, as determined by pulse radiolysis. aroxyl radicals 45-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 106-109 11519862-8 2001 The 20% ACN active fraction was heat stable and inhibited aromatase in a non-competitive manner. acetonitrile 8-11 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-67 11519862-10 2001 It was found that the intake of the 20% ACN fraction by gavage completely abrogated aromatase-induced hyperplasia and other changes in the mammary tissue. acetonitrile 40-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-93 11396362-2 2001 The third-generation aromatase inhibitors have replaced megestrol acetate as second-line hormonal therapy in advanced breast cancer, and large clinical trials are maturing to establish their efficacy relative to tamoxifen (Nolvadex) in the first-line metastatic setting. Megestrol Acetate 56-73 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 11427156-1 2001 Aromatase is the enzyme complex that catalyses the synthesis of oestrogens from androgens, and therefore it has unique potential to influence the physiological balance between the sex steroid hormones. Steroids 184-200 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 11472114-3 2001 The one-electron reduction potentials of the aroxyl radicals corresponding to compounds 2a-2d, E degrees (ArO(*)/ArO(-)) were 0.49, 0.49, 0.49, and 0.52 V vs NHE, respectively, as determined by pulse radiolysis. aroxyl radicals 45-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-116 11472114-6 2001 The reduction potentials for the proton coupled oxidation of compounds 2a-2d (ArOH --> ArO(*) + H(+)) as determined by cyclic voltammetry in acetonitrile were 1.35 (irreversible), 1.35 (quasireversible) 1.13 (reversible), and 0.74 (reversible) V vs NHE, respectively. acetonitrile 144-156 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-81 11225118-1 2001 A superoxochromium(III) ion, CraqOO2+, abstracts the hydrogen atom from the hydroxylic group of a substituted, cationic phenol (ArOH), kCrOO = 1.24 M-1 s-1 in acidic aqueous solution at 25 degrees C. The reaction has a large kinetic isotope effect, kArOH/kArOD approximately 12 and produces ArO., which also reacts with CraqOO2+ in a rapid second step, kArO = 1.26 x 10(4) M-1 s-1. craqoo2+ 29-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 128-131 11305590-0 2001 19-Oxygenation of C19-steroids with an A, B-ring enone structure, competitive inhibitors of estrogen biosynthesis, with human placental aromatase. c19-steroids 18-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-145 11305590-0 2001 19-Oxygenation of C19-steroids with an A, B-ring enone structure, competitive inhibitors of estrogen biosynthesis, with human placental aromatase. a, b-ring enone 39-54 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-145 11305590-1 2001 Aromatase is a cytochrome P-450 enzyme complex that catalyzes the conversion of androst-4-ene-3,17-dione (AD) to estrone through three sequential oxygenations of the 19-methyl group. Androstenedione 80-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 11305590-1 2001 Aromatase is a cytochrome P-450 enzyme complex that catalyzes the conversion of androst-4-ene-3,17-dione (AD) to estrone through three sequential oxygenations of the 19-methyl group. Estrone 113-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 11305590-2 2001 To gain insight into the ability of AD isomers, 4-en-6-one 1a, 5-en-4-one 2a, and 5-en-7-one 3a, competitive inhibitors of aromatase with an A, B-ring enone structure to serve as a substrate, we incubated the three inhibitors separately with human placental aromatase in the presence of NADPH in air. 4-en-6-one 48-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 123-132 11305590-2 2001 To gain insight into the ability of AD isomers, 4-en-6-one 1a, 5-en-4-one 2a, and 5-en-7-one 3a, competitive inhibitors of aromatase with an A, B-ring enone structure to serve as a substrate, we incubated the three inhibitors separately with human placental aromatase in the presence of NADPH in air. 5-en-4-one 63-73 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 123-132 11305590-2 2001 To gain insight into the ability of AD isomers, 4-en-6-one 1a, 5-en-4-one 2a, and 5-en-7-one 3a, competitive inhibitors of aromatase with an A, B-ring enone structure to serve as a substrate, we incubated the three inhibitors separately with human placental aromatase in the presence of NADPH in air. 5-en-7-one 82-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 123-132 11305590-2 2001 To gain insight into the ability of AD isomers, 4-en-6-one 1a, 5-en-4-one 2a, and 5-en-7-one 3a, competitive inhibitors of aromatase with an A, B-ring enone structure to serve as a substrate, we incubated the three inhibitors separately with human placental aromatase in the presence of NADPH in air. b-ring enone 144-156 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 123-132 11305590-6 2001 The 19-oxygenation of steroid 2a, the best substrate for aromatase among the three, was kinetically determined to give the Vmax value of 40 pmol/min/mg protein and the Km value of 1.43 microM, respectively. Steroids 22-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-66 11305590-7 2001 The results reveal that a good inhibitor of aromatase is not essentially a good substrate for the enzyme in a series of the A, B-ring enone steroids. a, b-ring enone steroids 124-148 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-53 11225118-1 2001 A superoxochromium(III) ion, CraqOO2+, abstracts the hydrogen atom from the hydroxylic group of a substituted, cationic phenol (ArOH), kCrOO = 1.24 M-1 s-1 in acidic aqueous solution at 25 degrees C. The reaction has a large kinetic isotope effect, kArOH/kArOD approximately 12 and produces ArO., which also reacts with CraqOO2+ in a rapid second step, kArO = 1.26 x 10(4) M-1 s-1. Hydrogen 53-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 128-131 11225118-1 2001 A superoxochromium(III) ion, CraqOO2+, abstracts the hydrogen atom from the hydroxylic group of a substituted, cationic phenol (ArOH), kCrOO = 1.24 M-1 s-1 in acidic aqueous solution at 25 degrees C. The reaction has a large kinetic isotope effect, kArOH/kArOD approximately 12 and produces ArO., which also reacts with CraqOO2+ in a rapid second step, kArO = 1.26 x 10(4) M-1 s-1. Phenol 120-126 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 128-131 11225118-1 2001 A superoxochromium(III) ion, CraqOO2+, abstracts the hydrogen atom from the hydroxylic group of a substituted, cationic phenol (ArOH), kCrOO = 1.24 M-1 s-1 in acidic aqueous solution at 25 degrees C. The reaction has a large kinetic isotope effect, kArOH/kArOD approximately 12 and produces ArO., which also reacts with CraqOO2+ in a rapid second step, kArO = 1.26 x 10(4) M-1 s-1. craqoo2+ 320-328 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 128-131 11384878-7 2001 Although letrozole and anastrozole are approximately equipotent in a cell-free aromatase system (human placental microsomes), letrozole is consistently 10-30 times more potent than anastrozole in inhibiting intracellular aromatase in intact rodent cells, normal human adipose fibroblasts and human cancer cell lines. Anastrozole 23-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-88 11181459-6 2001 The CYP19 intron 4 [TTTA]n repeat is unlikely to have a functional effect on aromatase activity and it is more likely that the [TTTA](8) and [TTTA](10) variants are in linkage disequilibrium with other functional CYP19 variants. ttta 20-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-9 11791126-3 2001 In the 2nd-line treatment for metastatic breast cancer patients, aromatase inhibitors achieved results antitumor activity at least equal to and sometimes better than that of tamoxifen. Tamoxifen 174-183 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 11168357-2 2001 Aromatase (CYP19) catalyzes three consecutive hydroxylation reactions converting C19 androgens to aromatic C18 estrogenic steroids. Steroids 122-130 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-16 11384878-7 2001 Although letrozole and anastrozole are approximately equipotent in a cell-free aromatase system (human placental microsomes), letrozole is consistently 10-30 times more potent than anastrozole in inhibiting intracellular aromatase in intact rodent cells, normal human adipose fibroblasts and human cancer cell lines. Letrozole 126-135 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 221-230 11383455-7 2001 With sufficient amounts of aromatase in breast tissue, enough estradiol as substrate should be available to allow formation of substantial amounts of genotoxic metabolites. Estradiol 62-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-36 11043580-7 2000 The objective of the present study was to utilize cytotrophoblasts isolated from midterm human placenta to analyze the effects of O2 on CYP19 gene expression and the molecular mechanisms that mediate these effects. Oxygen 130-132 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-141 11013343-0 2000 Role of aromatase in sex steroid action. Steroids 25-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 8-17 11043580-9 2000 However, when cytotrophoblasts that had been maintained in 2% O2 for 3 days were placed in a 20% O2 environment, there was a rapid onset of cell fusion and induction of P450arom mRNA and aromatase activity. Oxygen 62-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-177 11043580-9 2000 However, when cytotrophoblasts that had been maintained in 2% O2 for 3 days were placed in a 20% O2 environment, there was a rapid onset of cell fusion and induction of P450arom mRNA and aromatase activity. Oxygen 97-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-177 10999781-4 2000 To reconcile these conflicting data, we used a selective aromatase inhibitor, anastrozole, to examine the impact of E2 suppression on the hypothalamic-pituitary axis in the male. Anastrozole 78-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-66 10936614-0 2000 Synthesis of enterolactone and enterodiol precursors as potential inhibitors of human estrogen synthetase (aromatase). 2,3-bis(3'-hydroxybenzyl)butyrolactone 13-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-105 10901692-8 2000 Because CYP2C18 and CYP19 are expressed at low concentrations in the human liver, these observations indicate that at clinical DDS concentrations, CYP2C9 is a major and CYP2C8 is a likely minor contributor to DDS-NHY in human liver microsomes. Fumigant 93 127-130 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-25 10901692-8 2000 Because CYP2C18 and CYP19 are expressed at low concentrations in the human liver, these observations indicate that at clinical DDS concentrations, CYP2C9 is a major and CYP2C8 is a likely minor contributor to DDS-NHY in human liver microsomes. Fumigant 93 209-212 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-25 10956405-1 2000 Screening of the entire coding and major promoter regions of the CYP19 gene identified two novel polymorphisms at codon 39 (Trp to Arg) and codon 408 (silent) in addition to those reported previously at codon 264 (Arg to Cys) and intron 4 [tetranucleotide (TTTA) simple tandem repeat]. tetranucleotide 240-255 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-70 10956405-1 2000 Screening of the entire coding and major promoter regions of the CYP19 gene identified two novel polymorphisms at codon 39 (Trp to Arg) and codon 408 (silent) in addition to those reported previously at codon 264 (Arg to Cys) and intron 4 [tetranucleotide (TTTA) simple tandem repeat]. ttta 257-261 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-70 10861475-0 2000 A tetranucleotide repeat polymorphism in CYP19 and breast cancer risk. tetranucleotide 2-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-46 10755395-0 2000 A polymorphic tetranucleotide repeat in the CYP19 gene and male breast cancer. tetranucleotide 14-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-49 10861475-2 2000 A tetranucleotide (TTTA) repeat polymorphism is present in intron 4 of CYP19; 2 out of 4 breast cancer case-control studies have reported a greater frequency of 2 specific alleles among affected women. tetranucleotide 2-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-76 10861475-2 2000 A tetranucleotide (TTTA) repeat polymorphism is present in intron 4 of CYP19; 2 out of 4 breast cancer case-control studies have reported a greater frequency of 2 specific alleles among affected women. ttta 19-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-76 10755395-2 2000 This case-control study examines the relationship between a tetranucleotide repeat sequence in the CYP19 gene and the development of male breast cancer. tetranucleotide 60-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-104 10746939-0 2000 2-Chloro-s-triazine herbicides induce aromatase (CYP19) activity in H295R human adrenocortical carcinoma cells: a novel mechanism for estrogenicity? 2-chloro-1,3,5-triazine 0-19 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-54 10842913-1 2000 Aromatase (P450AROM) is the enzyme complex with converts testosterone to estradiol and androstendione to estrone. Testosterone 57-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10842913-1 2000 Aromatase (P450AROM) is the enzyme complex with converts testosterone to estradiol and androstendione to estrone. Testosterone 57-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-19 10842913-1 2000 Aromatase (P450AROM) is the enzyme complex with converts testosterone to estradiol and androstendione to estrone. Estradiol 73-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10842913-1 2000 Aromatase (P450AROM) is the enzyme complex with converts testosterone to estradiol and androstendione to estrone. Estradiol 73-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-19 10842913-1 2000 Aromatase (P450AROM) is the enzyme complex with converts testosterone to estradiol and androstendione to estrone. Androstenedione 87-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10842913-1 2000 Aromatase (P450AROM) is the enzyme complex with converts testosterone to estradiol and androstendione to estrone. Androstenedione 87-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-19 10842913-1 2000 Aromatase (P450AROM) is the enzyme complex with converts testosterone to estradiol and androstendione to estrone. Estrone 105-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10842913-1 2000 Aromatase (P450AROM) is the enzyme complex with converts testosterone to estradiol and androstendione to estrone. Estrone 105-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-19 10842913-3 2000 The aim of the study was to estimate expression of P450AROM messenger ribonucleic acid (mRNA) in normal, hyperplastic and malignant endometrium, and the ability to convert androstenedione to estrone by endometrial cancer tissue. Androstenedione 172-187 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 51-59 10842913-3 2000 The aim of the study was to estimate expression of P450AROM messenger ribonucleic acid (mRNA) in normal, hyperplastic and malignant endometrium, and the ability to convert androstenedione to estrone by endometrial cancer tissue. Estrone 191-198 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 51-59 10746939-5 2000 In this report we demonstrated that the commonly used 2-chloro-s-triazine herbicides atrazine, simazine, and propazine dose-dependently (0-30 microM) induced aromatase (CYP19) activity to an apparent maximum of about 2.5-fold in H295R cells. 2-chloro-1,3,5-triazine 54-73 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-174 10746939-5 2000 In this report we demonstrated that the commonly used 2-chloro-s-triazine herbicides atrazine, simazine, and propazine dose-dependently (0-30 microM) induced aromatase (CYP19) activity to an apparent maximum of about 2.5-fold in H295R cells. Atrazine 85-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-174 10746939-5 2000 In this report we demonstrated that the commonly used 2-chloro-s-triazine herbicides atrazine, simazine, and propazine dose-dependently (0-30 microM) induced aromatase (CYP19) activity to an apparent maximum of about 2.5-fold in H295R cells. Simazine 95-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-174 10746939-5 2000 In this report we demonstrated that the commonly used 2-chloro-s-triazine herbicides atrazine, simazine, and propazine dose-dependently (0-30 microM) induced aromatase (CYP19) activity to an apparent maximum of about 2.5-fold in H295R cells. propazine 109-118 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-174 10746939-7 2000 The triazines increased levels of CYP19 messenger ribonucleic acid (mRNA) between 1.5- and 2-fold. Triazines 4-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-39 10746939-8 2000 The time-response profile of the induction of aromatase activity and CYP19 mRNA by the triazines was similar to that by 8-bromo-cyclic adenosine monophosphate, a known stimulant of the protein kinase-A pathway that mediates the induction of aromatase in these cells. Triazines 87-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-74 11272534-5 2000 The hindered arene oxide ligands ArO- afford the square pyramidal complexes [Mo(OAr)(S2C2R2)2]1- (5, 6). arene 13-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-36 10676649-4 2000 By means of a formazan dye-based spectrophotometric assay of cell viability and light microscopy, manumycin was shown to decrease the number of viable cells in all six of the cell lines though to a lesser degree in DRO and C643 cells than in ARO, Hth-74, KAT-4, and KAT-18 cells. manumycin 98-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 242-245 10676649-13 2000 Combined manumycin and paclitaxel treatments seemed as effective as manumycin against ARO cells and more effective than either manumycin or paclitaxel alone against KAT-4 cells. manumycin 9-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-89 10676649-13 2000 Combined manumycin and paclitaxel treatments seemed as effective as manumycin against ARO cells and more effective than either manumycin or paclitaxel alone against KAT-4 cells. Paclitaxel 23-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-89 10676649-13 2000 Combined manumycin and paclitaxel treatments seemed as effective as manumycin against ARO cells and more effective than either manumycin or paclitaxel alone against KAT-4 cells. manumycin 68-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-89 10676649-13 2000 Combined manumycin and paclitaxel treatments seemed as effective as manumycin against ARO cells and more effective than either manumycin or paclitaxel alone against KAT-4 cells. manumycin 68-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-89 10652212-5 2000 [(3)H] water method was employed to measure Arom activity. Water 7-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-48 10652212-15 2000 Cell proliferation stimulated by IL-1beta was reduced by the addition of the Arom inhibitor fadrozole-HCL (CGS-16949A). Fadrozole 92-105 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-81 10652212-15 2000 Cell proliferation stimulated by IL-1beta was reduced by the addition of the Arom inhibitor fadrozole-HCL (CGS-16949A). cysteinylglycine 107-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-81 10611255-7 2000 Whole ovarian 17alpha-hydroxylase (P450c17) and granulosa cell P450 aromatase (P450arom) mRNA declined in a time-dependent fashion; by 36 h after HCG administration, steroid depletion increased P450arom mRNA, although progestin replacement did not return aromatase to control values (P < 0.05). Steroids 166-173 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-87 10623624-1 2000 A novel molecular modeling study, involving inhibitors bound to the iron of cytochrome P450 heme, is described for nonsteroidal inhibitors of aromatase (AR). Iron 68-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 142-151 10623624-1 2000 A novel molecular modeling study, involving inhibitors bound to the iron of cytochrome P450 heme, is described for nonsteroidal inhibitors of aromatase (AR). Iron 68-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 153-155 10623624-1 2000 A novel molecular modeling study, involving inhibitors bound to the iron of cytochrome P450 heme, is described for nonsteroidal inhibitors of aromatase (AR). Heme 92-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 142-151 10623624-1 2000 A novel molecular modeling study, involving inhibitors bound to the iron of cytochrome P450 heme, is described for nonsteroidal inhibitors of aromatase (AR). Heme 92-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 153-155 10623624-4 2000 Possible reasons for the difference in activity of enantiomers of alternative inhibitors is also suggested, as well as the mode of action of the new AR inhibitor, Arimidex-whose inhibitory activity previously has not been rationalized. Anastrozole 163-171 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 149-151 10963624-6 2000 A number of molecular epidemiologic studies have been conducted to evaluate associations between polymorphic genes involved in steroid hormone metabolism (i.e., CYP17, COMT, CYP1A1, CYP19, GST, and MnSOD) that may account for a proportion of enzymatic variability, and results are discussed in this review. Steroids 127-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 182-187 10536374-7 1999 Aromatase activity decreased during differentiation, as was demonstrated by the conversion of androstenedione (A) and testosterone (T) into estrone (E(1)) and estradiol (E(2)), respectively. Androstenedione 94-109 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10931541-4 2000 Our data for phenol (I) and 4-chlorophenol (II) demonstrate a higher stabilization of ArO(-) anions than was previously accepted. Phenol 13-19 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-89 10931541-4 2000 Our data for phenol (I) and 4-chlorophenol (II) demonstrate a higher stabilization of ArO(-) anions than was previously accepted. 4-chlorophenol 28-42 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-89 10704906-0 1999 19-oxygenations of 3-deoxy androgens, potent competitive inhibitors of estrogen biosynthesis, with human placental aromatase. 3-deoxy androgens 19-36 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 115-124 10704906-1 1999 Aromatase is a cytochrome P450 enzyme complex that catalyzes the conversion of androst-4-ene-3,17-dione (AD) to estrone through three sequential oxygenations of the 19-methyl group. Androstenedione 79-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10704906-1 1999 Aromatase is a cytochrome P450 enzyme complex that catalyzes the conversion of androst-4-ene-3,17-dione (AD) to estrone through three sequential oxygenations of the 19-methyl group. Androstenedione 105-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10704906-1 1999 Aromatase is a cytochrome P450 enzyme complex that catalyzes the conversion of androst-4-ene-3,17-dione (AD) to estrone through three sequential oxygenations of the 19-methyl group. Estrone 112-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10704906-4 1999 Kinetic studies indicated that the 5-ene steroid 4 was surprisingly a good substrate for the aromatase-catalyzing 19-oxygenation with the V(max) value of 45 pmol/min per mg prot which was approx. 5-ene steroid 35-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-102 10704906-7 1999 The results reveal that there is a difference between a binding suitable for serving as an inhibitor of aromatase and a binding suitable for serving as a substrate of the enzyme in the 3-deoxy steroid series and the C-3 carbonyl group of AD is essential for a proper binding as a substrate to the active site of aromatase. Steroids 193-200 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 104-113 10704906-7 1999 The results reveal that there is a difference between a binding suitable for serving as an inhibitor of aromatase and a binding suitable for serving as a substrate of the enzyme in the 3-deoxy steroid series and the C-3 carbonyl group of AD is essential for a proper binding as a substrate to the active site of aromatase. Steroids 193-200 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 312-321 10593363-4 1999 Endometriotic stromal cells aberrantly express aromatase, which converts C19, steroids to estrogens. Steroids 78-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 10593363-5 1999 Aromatase activity in these cells is stimulated by prostaglandin (PG)E2. Dinoprostone 51-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10652502-1 2000 In contrast to normal endometrium, the expression of aromatase is aberrant in endometriosis and is stimulated by prostaglandin E2 (PGE2). Dinoprostone 113-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 10652502-1 2000 In contrast to normal endometrium, the expression of aromatase is aberrant in endometriosis and is stimulated by prostaglandin E2 (PGE2). Dinoprostone 131-135 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 10536374-7 1999 Aromatase activity decreased during differentiation, as was demonstrated by the conversion of androstenedione (A) and testosterone (T) into estrone (E(1)) and estradiol (E(2)), respectively. Testosterone 118-130 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10536374-7 1999 Aromatase activity decreased during differentiation, as was demonstrated by the conversion of androstenedione (A) and testosterone (T) into estrone (E(1)) and estradiol (E(2)), respectively. Estrone 140-147 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10536374-7 1999 Aromatase activity decreased during differentiation, as was demonstrated by the conversion of androstenedione (A) and testosterone (T) into estrone (E(1)) and estradiol (E(2)), respectively. Estradiol 159-168 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10545204-0 1999 Identification and characterization of a cAMP-responsive element in the region upstream from promoter 1.3 of the human aromatase gene. Cyclic AMP 41-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 119-128 10583321-1 1999 OBJECTIVE: In order to evaluate the role of oestrogens on human male sexual behaviour, the gender-identity, psychosexual orientation and sexual activity of a man with a congenital lack of oestradiol resulting from an inactivating mutation of the aromatase P450 gene was investigated. Estradiol 188-198 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 246-255 10537150-5 1999 The P450arom inhibitor fadrazole (1 microM) inhibited more than 97% of this activity, whereas another imidazole, etomidate (1 microM), selectively inhibited gonadal P450arom activity by 92%. FADROZOLE 23-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-12 10537150-5 1999 The P450arom inhibitor fadrazole (1 microM) inhibited more than 97% of this activity, whereas another imidazole, etomidate (1 microM), selectively inhibited gonadal P450arom activity by 92%. FADROZOLE 23-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 165-173 10537150-5 1999 The P450arom inhibitor fadrazole (1 microM) inhibited more than 97% of this activity, whereas another imidazole, etomidate (1 microM), selectively inhibited gonadal P450arom activity by 92%. imidazole 102-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 165-173 10537150-5 1999 The P450arom inhibitor fadrazole (1 microM) inhibited more than 97% of this activity, whereas another imidazole, etomidate (1 microM), selectively inhibited gonadal P450arom activity by 92%. Etomidate 113-122 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 165-173 10537150-9 1999 Testosterone showed a higher affinity for the porcine placental P450arom than the gonadal P450arom, but both isozymes had similar affinities for androstenedione. Testosterone 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-72 10537150-9 1999 Testosterone showed a higher affinity for the porcine placental P450arom than the gonadal P450arom, but both isozymes had similar affinities for androstenedione. Testosterone 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-98 10537150-10 1999 Testosterone was also aromatized more slowly than androstenedione by the porcine gonadal P450arom. Testosterone 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 89-97 10537150-10 1999 Testosterone was also aromatized more slowly than androstenedione by the porcine gonadal P450arom. Androstenedione 50-65 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 89-97 10537150-11 1999 These data suggest that catalytic differences have arisen in the substrate binding pocket during the evolution of isozymes of porcine P450arom that affect androgen metabolism, particularly the aromatization of testosterone. Testosterone 210-222 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 134-142 10502498-0 1999 Cytotoxicity and aromatase (CYP19) activity modulation by organochlorines in human placental JEG-3 and JAR choriocarcinoma cells. Hydrocarbons, Chlorinated 58-73 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-33 10446859-2 1999 METHODS: Two data sets of RA ASPs (225 ASPs and 107 ASPs) were genotyped for a polymorphic tetranucleotide marker at the CYP19 locus using fluorescence-based semiautomated genotyping technology. tetranucleotide 91-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 121-126 10628390-3 1999 In the present study, we investigated the signal transduction mechanism associated with the growth inhibitory effect of apigenin, using a human anaplastic thyroid carcinoma cell line, ARO (UCLA RO-81-A-1). Apigenin 120-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 184-187 10628390-4 1999 Using Western blot method, it was shown that the inhibitory effect of apigenin on ARO cell proliferation is associated with an inhibition of both EGFR tyrosine autophosphorylation and phosphorylation of its downstream effector mitogen activated protein (MAP) kinase. Tyrosine 151-159 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-85 10385215-0 1999 Inhibition of aromatase (P450Arom) by some 1-(benzofuran-2-ylmethyl)imidazoles. 1-(benzofuran-2-ylmethyl)imidazoles 43-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-33 10364566-9 1999 Aromatase was found to be expressed only in cultured SMCs and not in cultured endothelial cells of human aorta and pulmonary artery and to be regulated through dexamethasone and the signaling pathways of protein kinase A and C. Study results revealed the localized expression of aromatase in vascular SMCs, which indicated a possible direct action of locally produced estrogen in an autocrine or paracrine manner, with possible cross talk between smooth muscle and endothelial cells. Dexamethasone 160-173 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10731110-3 1999 In addition, significant aromatase activity, which was stimulated 7-fold by dexamethasone, was measured in metastatic cells isolated from a breast cancer patient. Dexamethasone 76-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-34 10731110-13 1999 When the aromatase inhibitors were combined with the antiestrogen, tamoxifen, tumor growth was suppressed to about the same extent as with the aromatase inhibitors alone. Tamoxifen 67-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-18 10731110-13 1999 When the aromatase inhibitors were combined with the antiestrogen, tamoxifen, tumor growth was suppressed to about the same extent as with the aromatase inhibitors alone. Tamoxifen 67-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 143-152 10403538-2 1999 The increased expression of the aromatase CYP19 gene in breast cancer tissues was recently associated with a promoter region regulated through cAMP-mediated pathways. Cyclic AMP 143-147 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-41 10403538-2 1999 The increased expression of the aromatase CYP19 gene in breast cancer tissues was recently associated with a promoter region regulated through cAMP-mediated pathways. Cyclic AMP 143-147 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-47 10470147-0 1999 Induction of aromatase expression by aminoglutethimide, an aromatase inhibitor that is used to treat breast cancer in postmenopausal women. Aminoglutethimide 37-54 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 10470147-0 1999 Induction of aromatase expression by aminoglutethimide, an aromatase inhibitor that is used to treat breast cancer in postmenopausal women. Aminoglutethimide 37-54 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-68 10470147-1 1999 Aromatase, a cytochrome P450, catalyzes three consecutive hydroxylation reactions converting C19 androgens to aromatic C18 estrogenic steroids. Steroids 134-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10470147-2 1999 Aminoglutethimide (AG) is an aromatase inhibitor used to treat estrogen-dependent breast cancer. Aminoglutethimide 0-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-38 10470147-3 1999 While AG is effective in inhibiting aromatase, it was found that aromatase activity in tumors of some breast cancer patients elevated after AG treatment (Miller and O"Neill, Steroids, 50: 245-252, 1987). Steroids 174-182 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 10200304-1 1999 In humans, aromatase P450 (product of CYP19 gene), which catalyzes conversion of C19 steroids to estrogens, is expressed in a number of tissues, including ovary, adipose, and syncytiotrophoblast of the placenta. Steroids 85-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-20 10200304-1 1999 In humans, aromatase P450 (product of CYP19 gene), which catalyzes conversion of C19 steroids to estrogens, is expressed in a number of tissues, including ovary, adipose, and syncytiotrophoblast of the placenta. Steroids 85-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-43 10418994-3 1999 4-hydroxyandrostenedione (4-OHA), the first selective aromatase inhibitor, has been shown to reduce serum estrogen concentrations and cause complete and partial responses in approximately 25% of patients with hormone responsive disease who have relapsed from previous endocrine treatment. formestane 0-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 54-63 10418994-3 1999 4-hydroxyandrostenedione (4-OHA), the first selective aromatase inhibitor, has been shown to reduce serum estrogen concentrations and cause complete and partial responses in approximately 25% of patients with hormone responsive disease who have relapsed from previous endocrine treatment. formestane 26-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 54-63 10418994-11 1999 This effect could be inhibited by 4-OHA and implies that intratumoral aromatase has functional significance. formestane 34-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 70-79 10418994-17 1999 When the aromatase inhibitors were combined with tamoxifen, tumor growth was suppressed to about the same extent as with the aromatase inhibitors alone. Tamoxifen 49-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-134 10091679-1 1999 Six azolyl substituted indoles were synthesized and tested for their activity to inhibit two P450 enzymes: P450 arom and P450 17a. azolyl substituted indoles 4-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-129 10399883-7 1999 Quantitation of P450AROM, based on binding of [11C]vorozole is an accurate and sensitive in vitro method, which might be extended to the measurement of aromatase expression by a noninvasive technique in the intact ovary in vivo using positron emission tomography. vorozole 51-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-24 10399883-1 1999 An in vitro method for measuring aromatase cytochrome P450 enzyme (P450AROM) in human granulosa cells (GC) has been developed, based on binding of the 11C-labeled aromatase inhibitor vorozole. Carbon-11 151-154 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-75 10399883-1 1999 An in vitro method for measuring aromatase cytochrome P450 enzyme (P450AROM) in human granulosa cells (GC) has been developed, based on binding of the 11C-labeled aromatase inhibitor vorozole. vorozole 183-191 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-75 10399883-5 1999 P450AROM concentrations measured by [11C]vorozole binding correlated positively with aromatisation of [1beta-3H]androst-4-ene-3,17-dione measured as [3H]water release, and a positive association was also found with the ovarian in vivo response to follicle-stimulating hormone (FSH) stimulation expressed as 1000 times the ratio of the number of oocytes recovered from a patient and the total dose of recombinant FSH administered. Carbon-11 37-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 10399883-5 1999 P450AROM concentrations measured by [11C]vorozole binding correlated positively with aromatisation of [1beta-3H]androst-4-ene-3,17-dione measured as [3H]water release, and a positive association was also found with the ovarian in vivo response to follicle-stimulating hormone (FSH) stimulation expressed as 1000 times the ratio of the number of oocytes recovered from a patient and the total dose of recombinant FSH administered. vorozole 41-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 10399883-5 1999 P450AROM concentrations measured by [11C]vorozole binding correlated positively with aromatisation of [1beta-3H]androst-4-ene-3,17-dione measured as [3H]water release, and a positive association was also found with the ovarian in vivo response to follicle-stimulating hormone (FSH) stimulation expressed as 1000 times the ratio of the number of oocytes recovered from a patient and the total dose of recombinant FSH administered. 1beta-3h 103-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 10399883-5 1999 P450AROM concentrations measured by [11C]vorozole binding correlated positively with aromatisation of [1beta-3H]androst-4-ene-3,17-dione measured as [3H]water release, and a positive association was also found with the ovarian in vivo response to follicle-stimulating hormone (FSH) stimulation expressed as 1000 times the ratio of the number of oocytes recovered from a patient and the total dose of recombinant FSH administered. Androstenedione 112-136 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 10399883-5 1999 P450AROM concentrations measured by [11C]vorozole binding correlated positively with aromatisation of [1beta-3H]androst-4-ene-3,17-dione measured as [3H]water release, and a positive association was also found with the ovarian in vivo response to follicle-stimulating hormone (FSH) stimulation expressed as 1000 times the ratio of the number of oocytes recovered from a patient and the total dose of recombinant FSH administered. Tritium 109-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 10399883-5 1999 P450AROM concentrations measured by [11C]vorozole binding correlated positively with aromatisation of [1beta-3H]androst-4-ene-3,17-dione measured as [3H]water release, and a positive association was also found with the ovarian in vivo response to follicle-stimulating hormone (FSH) stimulation expressed as 1000 times the ratio of the number of oocytes recovered from a patient and the total dose of recombinant FSH administered. Water 153-158 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 10732791-4 1999 Aminoglutethimide was the first widely used aromatase inhibitor but had several clinical drawbacks. Aminoglutethimide 0-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-53 10732791-6 1999 This article reviews the clinical data supporting the use of the potent, oral competitive aromatase inhibitors anastrozole, letrozole and vorozole and the irreversible inhibitors 4-OH androstenedione and exemestane. Anastrozole 111-122 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-99 10732791-6 1999 This article reviews the clinical data supporting the use of the potent, oral competitive aromatase inhibitors anastrozole, letrozole and vorozole and the irreversible inhibitors 4-OH androstenedione and exemestane. Letrozole 124-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-99 10732791-6 1999 This article reviews the clinical data supporting the use of the potent, oral competitive aromatase inhibitors anastrozole, letrozole and vorozole and the irreversible inhibitors 4-OH androstenedione and exemestane. vorozole 138-146 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-99 9801824-0 1998 Synthesis of new steroidal isoxazoles: inhibitors of estrogen synthase. Isoxazoles 27-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-70 10027313-3 1999 Our genotyping of 348 cases with breast cancer and 145 controls (all Caucasian women) for a published tetranucleotide repeat polymorphism at intron 4 of the Cyp19 gene revealed the presence of six common and two rare alleles. tetranucleotide 102-117 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 157-162 10851574-2 1999 An enzyme termed aromatase in a number of human tissues and cells, including ovarian granulosa cells, the placental syncytiotrophoblast, adipose and skin fibroblasts, bone, and the brain, catalyzes the conversion of C19 steroids to estrogens. Steroids 220-228 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 9929167-0 1998 Aromatase and 17beta-hydroxysteroid dehydrogenase inhibition by flavonoids. Flavonoids 64-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9929167-1 1998 A method for estimating in the same assay both aromatase and 17beta-hydroxysteroid dehydrogenase activities in human placental microsomes using radiolabelled [1,2,6,7-3H]4-androstene-3,17-dione was proposed. [1,2,6,7-3h]4-androstene-3,17-dione 158-193 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 9929167-3 1998 Using this method, the inhibitory effect of various flavonoids, including flavone, flavanone and isoflavone, on the human placental aromatase and 17beta-hydroxysteroid dehydrogenase was studied. Flavonoids 52-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 132-141 9929167-3 1998 Using this method, the inhibitory effect of various flavonoids, including flavone, flavanone and isoflavone, on the human placental aromatase and 17beta-hydroxysteroid dehydrogenase was studied. flavone 74-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 132-141 9929167-3 1998 Using this method, the inhibitory effect of various flavonoids, including flavone, flavanone and isoflavone, on the human placental aromatase and 17beta-hydroxysteroid dehydrogenase was studied. flavanone 83-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 132-141 9929167-3 1998 Using this method, the inhibitory effect of various flavonoids, including flavone, flavanone and isoflavone, on the human placental aromatase and 17beta-hydroxysteroid dehydrogenase was studied. Isoflavones 97-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 132-141 9929167-4 1998 Flavonoids were shown to be potent inhibitors of both aromatase and 17beta-hydroxysteroid dehydrogenase activities. Flavonoids 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 54-63 9929167-5 1998 We found that 7-hydroxyflavone and apigenin are the most effective aromatase and 17beta-hydroxysteroid dehydrogenase inhibitors, respectively. 7-hydroxyflavone 14-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-76 9929167-7 1998 However, flavonoids with 7-methoxy or 8-hydroxyl groups on the A ring showed only anti-aromatase activity. Flavonoids 9-19 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-96 9817601-1 1998 Aromatase P450 (P450arom), a product of the CYP19 gene, catalyzes the conversion of C19-steroids to estrogens. c19-steroids 84-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-25 9817601-1 1998 Aromatase P450 (P450arom), a product of the CYP19 gene, catalyzes the conversion of C19-steroids to estrogens. c19-steroids 84-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-49 10204168-4 1998 Aromatase inhibitor fadrosol displayed a tendency towards prevention of the activation of androstenedione conversion in cultivated lymphocytes. fadrosol 20-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10204168-4 1998 Aromatase inhibitor fadrosol displayed a tendency towards prevention of the activation of androstenedione conversion in cultivated lymphocytes. Androstenedione 90-105 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9877321-0 1998 Enantioselectivity of some 1-(benzofuran-2-yl)-1-(1-H-imidazol-1-yl) alkanes as inhibitors of P450Arom. 1-(benzofuran-2-yl)-1-(1-h-imidazol-1-yl) alkanes 27-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-102 9719444-3 1998 Southern blot of RT-PCR products with a 32P-labeled cDNA probe for the human aromatase demonstrated that FLG 29.1 cells express aromatase mRNA. Phosphorus-32 40-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-86 9696881-6 1998 RT-PCR and gene transcriptional studies have revealed that aromatase promoter switches from a glucocorticoid-stimulated promoter, I.4, in normal tissue to cAMP-stimulated promoters, I.3 and II, in cancereous tissue. Cyclic AMP 155-159 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-68 9696881-13 1998 The model is used to evaluate the interaction of phytoestrogens such as flavones and isoflavones with aromatase. Flavones 72-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 102-111 9696881-13 1998 The model is used to evaluate the interaction of phytoestrogens such as flavones and isoflavones with aromatase. Isoflavones 85-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 102-111 9696881-14 1998 The study provides a molecular basis as to why isoflavones are significantly poorer inhibitors of aromatase than flavones. Isoflavones 47-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-107 9696881-14 1998 The study provides a molecular basis as to why isoflavones are significantly poorer inhibitors of aromatase than flavones. Flavones 50-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-107 9662830-6 1998 Using a sensitive (attomole sensitivity) competitive RT-PCR technique, aromatase transcript abundance was quantified during embryonic development for embryos treated with and without estrogen. attomole 19-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-80 9662830-10 1998 Aromatase mRNA levels are generally reduced in the brain by estradiol application. Estradiol 60-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9500452-16 1998 A+ cells also grew under these conditions, and the aromatase inhibitor 4-hydroxyandrostenedione reduced both tumor E2 level and growth rate, providing additional evidence of the importance of in situ synthesis. formestane 71-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 51-60 9589695-3 1998 Conversion of C19 steroids to estrogens occurs in a number of human tissues and is catalyzed by aromatase P450 (P450arom), the product of the CYP19 gene in a number of human tissues. Steroids 18-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 112-120 9589695-3 1998 Conversion of C19 steroids to estrogens occurs in a number of human tissues and is catalyzed by aromatase P450 (P450arom), the product of the CYP19 gene in a number of human tissues. Steroids 18-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 142-147 9597182-3 1998 One subset of compounds consists of fadrozole analogues and was studied in a previous work, from which a "local" 3-D quantitative structure-activity relationship (QSAR) model for the inhibition of aromatase was obtained. Fadrozole 36-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 197-206 9528950-0 1998 Characterization of a region upstream of exon I.1 of the human CYP19 (aromatase) gene that mediates regulation by retinoids in human choriocarcinoma cells. Retinoids 114-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-68 9528950-0 1998 Characterization of a region upstream of exon I.1 of the human CYP19 (aromatase) gene that mediates regulation by retinoids in human choriocarcinoma cells. Retinoids 114-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 70-79 9541188-2 1998 Postmenopausal women with hormone dependent breast cancer respond to first generation aromatase inhibitors such as aminoglutethimide with a marked suppression of circulating estradiol levels. Aminoglutethimide 115-132 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-95 9541188-2 1998 Postmenopausal women with hormone dependent breast cancer respond to first generation aromatase inhibitors such as aminoglutethimide with a marked suppression of circulating estradiol levels. Estradiol 174-183 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-95 9641536-1 1998 In the brain, conversion of androgens into estrogens by the enzyme aromatase (estrogen synthase) is a key mechanism by which testosterone regulates many physiological and behavioral processes, including the activation of male sexual behavior, brain sexual differentiation and negative feedback effects of steroid hormones on gonadotropin secretion. Testosterone 125-137 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-95 9641536-1 1998 In the brain, conversion of androgens into estrogens by the enzyme aromatase (estrogen synthase) is a key mechanism by which testosterone regulates many physiological and behavioral processes, including the activation of male sexual behavior, brain sexual differentiation and negative feedback effects of steroid hormones on gonadotropin secretion. Steroids 305-321 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-95 9510005-5 1998 P450AROM mRNA was measured in individual follicles from 16 PCOS and 48 regularly cycling control women by quantitative polymerase chain reaction (PCR) and correlated with follicular fluid oestradiol concentrations and aromatase stimulating bioactivity measured by the rat granulosa cells aromatase bioassay. Estradiol 188-198 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 9435150-0 1998 Molecular basis of the inhibition of human aromatase (estrogen synthetase) by flavone and isoflavone phytoestrogens: A site-directed mutagenesis study. flavone 78-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-52 9435150-0 1998 Molecular basis of the inhibition of human aromatase (estrogen synthetase) by flavone and isoflavone phytoestrogens: A site-directed mutagenesis study. flavone 78-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 54-73 9435150-0 1998 Molecular basis of the inhibition of human aromatase (estrogen synthetase) by flavone and isoflavone phytoestrogens: A site-directed mutagenesis study. Isoflavones 90-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-52 9435150-0 1998 Molecular basis of the inhibition of human aromatase (estrogen synthetase) by flavone and isoflavone phytoestrogens: A site-directed mutagenesis study. Isoflavones 90-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 54-73 9435150-1 1998 Flavone and isoflavone phytoestrogens are plant chemicals and are known to be competitive inhibitors of cytochrome P450 aromatase with respect to the androgen substrate. flavone 0-7 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 120-129 9435150-1 1998 Flavone and isoflavone phytoestrogens are plant chemicals and are known to be competitive inhibitors of cytochrome P450 aromatase with respect to the androgen substrate. Isoflavones 12-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 120-129 9797013-4 1998 Removal of fetal calf serum (FCS) from the culture medium or addition of forskolin or phorbol ester (TPA) also induced rapid elevation of aromatase mRNA and switching to exon 1c, whereas TGFbeta almost abolished the expression, suggesting that cancer cells might secret forskolin- or TPA-like stimulatory factors, or consume TGFbeta-like inhibitory factors in serum for expression of aromatase mRNA. Colforsin 73-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-147 9797013-4 1998 Removal of fetal calf serum (FCS) from the culture medium or addition of forskolin or phorbol ester (TPA) also induced rapid elevation of aromatase mRNA and switching to exon 1c, whereas TGFbeta almost abolished the expression, suggesting that cancer cells might secret forskolin- or TPA-like stimulatory factors, or consume TGFbeta-like inhibitory factors in serum for expression of aromatase mRNA. Colforsin 73-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 384-393 9797013-4 1998 Removal of fetal calf serum (FCS) from the culture medium or addition of forskolin or phorbol ester (TPA) also induced rapid elevation of aromatase mRNA and switching to exon 1c, whereas TGFbeta almost abolished the expression, suggesting that cancer cells might secret forskolin- or TPA-like stimulatory factors, or consume TGFbeta-like inhibitory factors in serum for expression of aromatase mRNA. Phorbol Esters 86-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-147 9797013-4 1998 Removal of fetal calf serum (FCS) from the culture medium or addition of forskolin or phorbol ester (TPA) also induced rapid elevation of aromatase mRNA and switching to exon 1c, whereas TGFbeta almost abolished the expression, suggesting that cancer cells might secret forskolin- or TPA-like stimulatory factors, or consume TGFbeta-like inhibitory factors in serum for expression of aromatase mRNA. Phorbol Esters 86-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 384-393 9797013-4 1998 Removal of fetal calf serum (FCS) from the culture medium or addition of forskolin or phorbol ester (TPA) also induced rapid elevation of aromatase mRNA and switching to exon 1c, whereas TGFbeta almost abolished the expression, suggesting that cancer cells might secret forskolin- or TPA-like stimulatory factors, or consume TGFbeta-like inhibitory factors in serum for expression of aromatase mRNA. Tetradecanoylphorbol Acetate 101-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-147 9797013-4 1998 Removal of fetal calf serum (FCS) from the culture medium or addition of forskolin or phorbol ester (TPA) also induced rapid elevation of aromatase mRNA and switching to exon 1c, whereas TGFbeta almost abolished the expression, suggesting that cancer cells might secret forskolin- or TPA-like stimulatory factors, or consume TGFbeta-like inhibitory factors in serum for expression of aromatase mRNA. Tetradecanoylphorbol Acetate 101-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 384-393 9797022-10 1998 In addition, evidence for the functional significance of tumor aromatase was indicated by a correlation between aromatase activity and expression of proliferating cell nuclear antigen (PCNA) in the tumor, and by increased thymidine incorporation into DNA in response to testosterone in tumors in histoculture which had high aromatase activity but not in those with low activity. Thymidine 222-231 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-72 9797022-10 1998 In addition, evidence for the functional significance of tumor aromatase was indicated by a correlation between aromatase activity and expression of proliferating cell nuclear antigen (PCNA) in the tumor, and by increased thymidine incorporation into DNA in response to testosterone in tumors in histoculture which had high aromatase activity but not in those with low activity. Testosterone 270-282 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-72 9619844-4 1998 The testosterone action could depend on its local conversion to 17beta-estradiol by aromatase which is present in frog liver tissue. Testosterone 4-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-93 9619844-4 1998 The testosterone action could depend on its local conversion to 17beta-estradiol by aromatase which is present in frog liver tissue. Estradiol 64-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-93 9619844-6 1998 Aromatase activity seems depend on testosterone since it decreases after ovariectomy and is restored by testosterone injection in ovariectomized frogs. Testosterone 35-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9619844-6 1998 Aromatase activity seems depend on testosterone since it decreases after ovariectomy and is restored by testosterone injection in ovariectomized frogs. Testosterone 104-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9619844-9 1998 In liver nuclei, testosterone receptor level undergoes modification throughout the sexual cycle which almost coincides with that of plasma testosterone level and liver aromatase activity. Testosterone 17-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 168-177 9619844-10 1998 This could indicate that the testosterone induction of liver aromatase in frogs is via the testosterone receptor, as reported for aromatase of mammalian brain tissues. Testosterone 29-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-70 9619844-10 1998 This could indicate that the testosterone induction of liver aromatase in frogs is via the testosterone receptor, as reported for aromatase of mammalian brain tissues. Testosterone 29-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-139 9435150-4 1998 In combination with computer modeling, the binding characteristics and the structure requirement for flavone and isoflavone phytoestrogens to inhibit human aromatase were obtained. flavone 101-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 156-165 9435150-4 1998 In combination with computer modeling, the binding characteristics and the structure requirement for flavone and isoflavone phytoestrogens to inhibit human aromatase were obtained. Isoflavones 113-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 156-165 9435150-6 1998 This study also provides a molecular basis as to why isoflavones are significantly poorer inhibitors of aromatase than flavones. Isoflavones 53-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 104-113 9435150-6 1998 This study also provides a molecular basis as to why isoflavones are significantly poorer inhibitors of aromatase than flavones. Flavones 56-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 104-113 9511180-1 1998 The aromatase P450 (coded by the CYP19 gene) is responsible for the rate limiting step in the metabolism of C19 steroids to estrogens and is expressed in most breast carcinomas. Steroids 112-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-38 9511181-4 1998 Two-point linkage analysis was performed between this tetra-allelic polymorphism and the chromosome 15 microsatellite markers of Genethon as well as the tetranucleotide polymorphism of the CYP19 gene and a MspI RFLP of the CYP1A1 gene. tetranucleotide 153-168 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 189-194 9475196-6 1998 That only rare stromal cells were immunoreactive for P450arom suggests that most of the testosterone synthesized in the tumor is extragonadally converted to estrogen, resulting in tumor-associated estrinism. Testosterone 88-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-61 9578729-3 1998 Direct correlation was established between aromatase concentration, on the one hand, and tumor size, cell differentiation status and blood-testosterone on the other. Testosterone 139-151 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-52 10388095-27 1998 STRUCTURE OF AROMATASE REVEALS SECRETS OF SELECTIVE INHIBITION: Aromatase is a cytochrome P450 enzyme, with both an iron-containing and a steroid-binding site. Iron 116-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 10388095-27 1998 STRUCTURE OF AROMATASE REVEALS SECRETS OF SELECTIVE INHIBITION: Aromatase is a cytochrome P450 enzyme, with both an iron-containing and a steroid-binding site. Iron 116-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-73 10388095-27 1998 STRUCTURE OF AROMATASE REVEALS SECRETS OF SELECTIVE INHIBITION: Aromatase is a cytochrome P450 enzyme, with both an iron-containing and a steroid-binding site. Steroids 138-145 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 10388095-27 1998 STRUCTURE OF AROMATASE REVEALS SECRETS OF SELECTIVE INHIBITION: Aromatase is a cytochrome P450 enzyme, with both an iron-containing and a steroid-binding site. Steroids 138-145 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-73 10388095-29 1998 From this structural relationship, there are, therefore, two reasonable ways to inhibit aromatase: * by occupying the steroid-binding site of the enzyme with a compound such as formestane (Lentaron&reg;), or * by binding the iron with nitrogen-containing compounds such as aminoglutethimide (Orimeten&reg;), the oldest aromatase inhibitor. Steroids 118-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 10388095-29 1998 From this structural relationship, there are, therefore, two reasonable ways to inhibit aromatase: * by occupying the steroid-binding site of the enzyme with a compound such as formestane (Lentaron&reg;), or * by binding the iron with nitrogen-containing compounds such as aminoglutethimide (Orimeten&reg;), the oldest aromatase inhibitor. Steroids 118-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 327-336 10388095-29 1998 From this structural relationship, there are, therefore, two reasonable ways to inhibit aromatase: * by occupying the steroid-binding site of the enzyme with a compound such as formestane (Lentaron&reg;), or * by binding the iron with nitrogen-containing compounds such as aminoglutethimide (Orimeten&reg;), the oldest aromatase inhibitor. formestane 177-187 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 10388095-29 1998 From this structural relationship, there are, therefore, two reasonable ways to inhibit aromatase: * by occupying the steroid-binding site of the enzyme with a compound such as formestane (Lentaron&reg;), or * by binding the iron with nitrogen-containing compounds such as aminoglutethimide (Orimeten&reg;), the oldest aromatase inhibitor. formestane 189-197 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 10388095-29 1998 From this structural relationship, there are, therefore, two reasonable ways to inhibit aromatase: * by occupying the steroid-binding site of the enzyme with a compound such as formestane (Lentaron&reg;), or * by binding the iron with nitrogen-containing compounds such as aminoglutethimide (Orimeten&reg;), the oldest aromatase inhibitor. Adenosine Monophosphate 198-201 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 10388095-29 1998 From this structural relationship, there are, therefore, two reasonable ways to inhibit aromatase: * by occupying the steroid-binding site of the enzyme with a compound such as formestane (Lentaron&reg;), or * by binding the iron with nitrogen-containing compounds such as aminoglutethimide (Orimeten&reg;), the oldest aromatase inhibitor. Iron 229-233 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 10388095-29 1998 From this structural relationship, there are, therefore, two reasonable ways to inhibit aromatase: * by occupying the steroid-binding site of the enzyme with a compound such as formestane (Lentaron&reg;), or * by binding the iron with nitrogen-containing compounds such as aminoglutethimide (Orimeten&reg;), the oldest aromatase inhibitor. Nitrogen 239-247 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 10388095-29 1998 From this structural relationship, there are, therefore, two reasonable ways to inhibit aromatase: * by occupying the steroid-binding site of the enzyme with a compound such as formestane (Lentaron&reg;), or * by binding the iron with nitrogen-containing compounds such as aminoglutethimide (Orimeten&reg;), the oldest aromatase inhibitor. Aminoglutethimide 277-294 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 10388095-29 1998 From this structural relationship, there are, therefore, two reasonable ways to inhibit aromatase: * by occupying the steroid-binding site of the enzyme with a compound such as formestane (Lentaron&reg;), or * by binding the iron with nitrogen-containing compounds such as aminoglutethimide (Orimeten&reg;), the oldest aromatase inhibitor. Aminoglutethimide 296-304 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 10388095-29 1998 From this structural relationship, there are, therefore, two reasonable ways to inhibit aromatase: * by occupying the steroid-binding site of the enzyme with a compound such as formestane (Lentaron&reg;), or * by binding the iron with nitrogen-containing compounds such as aminoglutethimide (Orimeten&reg;), the oldest aromatase inhibitor. Adenosine Monophosphate 305-308 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 10388095-30 1998 AROMATASE INHIBITORS: STEROIDAL AND NON-STEROIDAL: Formestane (Lentaron&reg;) is the only commercially available steroidal compound which inhibits aromatase and must be administered parenterally. formestane 51-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 10388095-30 1998 AROMATASE INHIBITORS: STEROIDAL AND NON-STEROIDAL: Formestane (Lentaron&reg;) is the only commercially available steroidal compound which inhibits aromatase and must be administered parenterally. formestane 51-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 151-160 10388095-30 1998 AROMATASE INHIBITORS: STEROIDAL AND NON-STEROIDAL: Formestane (Lentaron&reg;) is the only commercially available steroidal compound which inhibits aromatase and must be administered parenterally. formestane 63-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 151-160 10388095-31 1998 Other new aromatase inhibitors such as fadrozole (Afema&reg;) and letrozole (Femara&reg;) are orally active nitrogen-containing compounds that bind the heme iron of aromatase. Fadrozole 39-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-19 10388095-31 1998 Other new aromatase inhibitors such as fadrozole (Afema&reg;) and letrozole (Femara&reg;) are orally active nitrogen-containing compounds that bind the heme iron of aromatase. Fadrozole 39-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 173-182 10388095-31 1998 Other new aromatase inhibitors such as fadrozole (Afema&reg;) and letrozole (Femara&reg;) are orally active nitrogen-containing compounds that bind the heme iron of aromatase. Fadrozole 50-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-19 10388095-31 1998 Other new aromatase inhibitors such as fadrozole (Afema&reg;) and letrozole (Femara&reg;) are orally active nitrogen-containing compounds that bind the heme iron of aromatase. Fadrozole 50-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 173-182 10388095-31 1998 Other new aromatase inhibitors such as fadrozole (Afema&reg;) and letrozole (Femara&reg;) are orally active nitrogen-containing compounds that bind the heme iron of aromatase. Adenosine Monophosphate 56-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-19 10388095-31 1998 Other new aromatase inhibitors such as fadrozole (Afema&reg;) and letrozole (Femara&reg;) are orally active nitrogen-containing compounds that bind the heme iron of aromatase. Letrozole 70-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-19 10388095-31 1998 Other new aromatase inhibitors such as fadrozole (Afema&reg;) and letrozole (Femara&reg;) are orally active nitrogen-containing compounds that bind the heme iron of aromatase. Letrozole 70-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 173-182 10388095-31 1998 Other new aromatase inhibitors such as fadrozole (Afema&reg;) and letrozole (Femara&reg;) are orally active nitrogen-containing compounds that bind the heme iron of aromatase. Adenosine Monophosphate 88-91 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-19 10388095-31 1998 Other new aromatase inhibitors such as fadrozole (Afema&reg;) and letrozole (Femara&reg;) are orally active nitrogen-containing compounds that bind the heme iron of aromatase. Nitrogen 116-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-19 10388095-32 1998 AMINOGLUTETHIMIDE VERSUS LETROZOLE: OLD VERSUS NEW: Although aminoglutethimide has long been used to treat advanced breast cancer, its aromatase inhibition is not selective. Aminoglutethimide 0-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 10388095-32 1998 AMINOGLUTETHIMIDE VERSUS LETROZOLE: OLD VERSUS NEW: Although aminoglutethimide has long been used to treat advanced breast cancer, its aromatase inhibition is not selective. Letrozole 25-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 10388095-32 1998 AMINOGLUTETHIMIDE VERSUS LETROZOLE: OLD VERSUS NEW: Although aminoglutethimide has long been used to treat advanced breast cancer, its aromatase inhibition is not selective. Aminoglutethimide 61-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 10388095-35 1998 The affinity of letrozole (Femara&reg;) for the heme group of aromatase makes it a selective and potent inhibitor (Fig. Letrozole 16-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-75 10388095-35 1998 The affinity of letrozole (Femara&reg;) for the heme group of aromatase makes it a selective and potent inhibitor (Fig. Letrozole 27-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-75 10388095-35 1998 The affinity of letrozole (Femara&reg;) for the heme group of aromatase makes it a selective and potent inhibitor (Fig. Adenosine Monophosphate 34-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-75 10388095-37 1998 In fact, studies show that Femara&reg; has little effect on the other adrenal steroids, and is the most selective aromatase inhibitor available today. Letrozole 27-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 118-127 10388095-37 1998 In fact, studies show that Femara&reg; has little effect on the other adrenal steroids, and is the most selective aromatase inhibitor available today. Adenosine Monophosphate 34-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 118-127 9396278-4 1997 In the corpus luteum, P450scc and 3 beta-HSD catalyze the pathway from cholesterol to progesterone and in the granulosa cells, P450arom (aromatase) and two HSDs catalyze the biosynthesis of estrogen from C19 steroid synthesized in the theca cells having P450(17) alpha. Cholesterol 71-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-147 9396278-4 1997 In the corpus luteum, P450scc and 3 beta-HSD catalyze the pathway from cholesterol to progesterone and in the granulosa cells, P450arom (aromatase) and two HSDs catalyze the biosynthesis of estrogen from C19 steroid synthesized in the theca cells having P450(17) alpha. Steroids 208-215 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-147 9394367-0 1997 Anastrozole: a new selective nonsteroidal aromatase inhibitor. Anastrozole 0-11 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-51 9195917-7 1997 Unexpectedly, however, when the C-terminal domain of the gris ARO/CYC was expressed in a context where aromatase activity was absent, it could modulate the chain length specificity of the tetracenomycin (tcm) minimal PKS, leading to the formation of a novel 18-carbon product in addition to the expected 20-carbon one. 5838 DNI 188-202 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-65 9394367-1 1997 Aromatase (estrogen synthetase) is the enzyme complex responsible for the final step in estrogen synthesis--the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Androstenedione 126-141 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9394367-1 1997 Aromatase (estrogen synthetase) is the enzyme complex responsible for the final step in estrogen synthesis--the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Androstenedione 126-141 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-30 9394367-1 1997 Aromatase (estrogen synthetase) is the enzyme complex responsible for the final step in estrogen synthesis--the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Testosterone 146-158 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9394367-1 1997 Aromatase (estrogen synthetase) is the enzyme complex responsible for the final step in estrogen synthesis--the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Testosterone 146-158 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-30 9394367-1 1997 Aromatase (estrogen synthetase) is the enzyme complex responsible for the final step in estrogen synthesis--the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Estrone 162-169 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9394367-1 1997 Aromatase (estrogen synthetase) is the enzyme complex responsible for the final step in estrogen synthesis--the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Estrone 162-169 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-30 9394367-1 1997 Aromatase (estrogen synthetase) is the enzyme complex responsible for the final step in estrogen synthesis--the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Estradiol 174-183 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9394367-1 1997 Aromatase (estrogen synthetase) is the enzyme complex responsible for the final step in estrogen synthesis--the conversion of androstenedione and testosterone to estrone and estradiol, respectively. Estradiol 174-183 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-30 9394367-3 1997 The most widely used aromatase inhibitor has been aminoglutethimide; however, it is nonselective and also inhibits adrenocorticosteroid synthesis, necessitating hydrocortisone supplementation. Aminoglutethimide 50-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 9394367-3 1997 The most widely used aromatase inhibitor has been aminoglutethimide; however, it is nonselective and also inhibits adrenocorticosteroid synthesis, necessitating hydrocortisone supplementation. Hydrocortisone 161-175 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-30 9394367-5 1997 Formestane, the first selective aromatase inhibitor to be developed, has an improved safety profile and selectivity, but its use has been limited somewhat by its inconvenient administration via intramuscular injection. formestane 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-41 9394367-6 1997 In this article, the preclinical and clinical data published to date on the new third-generation aromatase inhibitor anastrozole (Arimidex) are presented in the context of current endocrine therapies. Anastrozole 117-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 97-106 9394367-6 1997 In this article, the preclinical and clinical data published to date on the new third-generation aromatase inhibitor anastrozole (Arimidex) are presented in the context of current endocrine therapies. Anastrozole 130-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 97-106 9352581-0 1997 Arginine-cysteine polymorphism at codon 264 of the human CYP19 gene does not affect aromatase activity. arginine-cysteine 0-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-62 9314579-11 1997 P450arom activity, as measured by the 3H2O assay, correlated well with the distribution of P450arom mRNA (the 455-nt protected fragment; r = 0.9) in the same tissues. 3h2o 38-42 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-8 9275055-11 1997 One possible explanation for this variability may be that the bCYP19ov gene has a 1-bp deletion in a cAMP-response element-like sequence (CLS) present at -208 to -201 bp in the hCYP19ov gene that we have shown to be critical for cAMP-stimulated transcription of hCYP19ov in the ovary. Cyclic AMP 101-105 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 177-183 9275055-11 1997 One possible explanation for this variability may be that the bCYP19ov gene has a 1-bp deletion in a cAMP-response element-like sequence (CLS) present at -208 to -201 bp in the hCYP19ov gene that we have shown to be critical for cAMP-stimulated transcription of hCYP19ov in the ovary. Cyclic AMP 229-233 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 177-183 9195917-7 1997 Unexpectedly, however, when the C-terminal domain of the gris ARO/CYC was expressed in a context where aromatase activity was absent, it could modulate the chain length specificity of the tetracenomycin (tcm) minimal PKS, leading to the formation of a novel 18-carbon product in addition to the expected 20-carbon one. 5838 DNI 204-207 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-65 9195917-7 1997 Unexpectedly, however, when the C-terminal domain of the gris ARO/CYC was expressed in a context where aromatase activity was absent, it could modulate the chain length specificity of the tetracenomycin (tcm) minimal PKS, leading to the formation of a novel 18-carbon product in addition to the expected 20-carbon one. Carbon 261-267 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-65 9195917-7 1997 Unexpectedly, however, when the C-terminal domain of the gris ARO/CYC was expressed in a context where aromatase activity was absent, it could modulate the chain length specificity of the tetracenomycin (tcm) minimal PKS, leading to the formation of a novel 18-carbon product in addition to the expected 20-carbon one. 20-carbon one 304-317 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-65 9224697-5 1997 Compounds 1 and 2 acted as type II ligands to the heme iron present in the active site of aromatase cytochrome P450 (P450arom). Heme 50-54 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 117-125 9224697-5 1997 Compounds 1 and 2 acted as type II ligands to the heme iron present in the active site of aromatase cytochrome P450 (P450arom). Iron 55-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 117-125 9140125-5 1997 Quantitation of aromatase activity by the [3H] water assay yielded values of 27.23 +/- 6.87 and 26.52 +/- 9.12 fmol/hr/mg of protein for BPH (nine patients) and PC (nine patients), respectively. Tritium 43-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-25 9178927-0 1997 Aromatase inactivation by a suicide substrate, androst-5-ene-4,7,17-trione: the 5beta,6beta-epoxy-19-oxo derivative, as a possible reactive electrophile irreversibly binding to the active site. Androst-5-en-4,7,17-trione 47-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9178927-0 1997 Aromatase inactivation by a suicide substrate, androst-5-ene-4,7,17-trione: the 5beta,6beta-epoxy-19-oxo derivative, as a possible reactive electrophile irreversibly binding to the active site. 5beta 80-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9178927-0 1997 Aromatase inactivation by a suicide substrate, androst-5-ene-4,7,17-trione: the 5beta,6beta-epoxy-19-oxo derivative, as a possible reactive electrophile irreversibly binding to the active site. 6beta-epoxy-19-oxo 86-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. Androst-5-en-4,7,17-trione 79-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. Androst-5-en-4,7,17-trione 79-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. Androst-5-en-4,7,17-trione 79-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. 5beta 146-151 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. epoxyandrosta-4,7,17,19-tetraone 158-190 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. epoxyandrosta-4,7,17,19-tetraone 158-190 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. epoxyandrosta-4,7,17,19-tetraone 158-190 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. 19-oxo-5-ene steroid 337-357 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. 19-oxo-5-ene steroid 337-357 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. 19-oxo-5-ene steroid 337-357 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. Hydrogen Peroxide 365-382 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. Hydrogen Peroxide 365-382 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. Hydrogen Peroxide 365-382 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. Sodium Bicarbonate 402-408 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. Sodium Bicarbonate 402-408 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 9178927-1 1997 In order to understand the mechanism involved in the aromatase inactivation by androst-5-ene-4,7,17-trione (4), a suicide substrate of aromatase, 5beta,6beta-epoxyandrosta-4,7,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase upon treatment of 19-oxo-5-ene steroid 5 with hydrogen peroxide in the presence of NaHCO3. Sodium Bicarbonate 402-408 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-144 9178927-2 1997 The epoxide 6 was a competitive inhibitor of human placental aromatase (Ki = 34 microM); moreover, it inactivated the enzyme in an active-site-directed manner in the absence of NADPH (Ki = 36 microM, a rate constant for inactivation (k(inact)) = 0.027 min(-1)). epoxide 6 4-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-70 9140125-5 1997 Quantitation of aromatase activity by the [3H] water assay yielded values of 27.23 +/- 6.87 and 26.52 +/- 9.12 fmol/hr/mg of protein for BPH (nine patients) and PC (nine patients), respectively. Water 47-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-25 9140125-6 1997 Reverse transcriptase and polymerase chain reaction analysis revealed that the mean aromatase mRNA content was 1.671 +/- 0.82 and 1.11 +/- 0.51 attomole/ng of total RNA (tRNA) for BPH (seven patients) and PC (four patients), respectively. attomole 144-152 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-93 9140222-0 1997 Inhibition of 5 alpha-reductase and aromatase by the ellagitannins oenothein A and oenothein B from Epilobium species. oenothein B 83-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-45 9125517-4 1997 A chemical catalytic mechanism of proteolysis consistent with the kinetic data is proposed, in which Tyr216-ArO-, in the course of being released from the active-site metal ion, deprotonates a water molecule attacking the Zn2+-activated substrate linkage, leading to a metal-coordinated tetrahedral oxyanion adduct that subsequently fragments. Metals 167-172 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-111 9125517-4 1997 A chemical catalytic mechanism of proteolysis consistent with the kinetic data is proposed, in which Tyr216-ArO-, in the course of being released from the active-site metal ion, deprotonates a water molecule attacking the Zn2+-activated substrate linkage, leading to a metal-coordinated tetrahedral oxyanion adduct that subsequently fragments. Water 193-198 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-111 9125517-4 1997 A chemical catalytic mechanism of proteolysis consistent with the kinetic data is proposed, in which Tyr216-ArO-, in the course of being released from the active-site metal ion, deprotonates a water molecule attacking the Zn2+-activated substrate linkage, leading to a metal-coordinated tetrahedral oxyanion adduct that subsequently fragments. Zinc 222-226 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-111 9125517-4 1997 A chemical catalytic mechanism of proteolysis consistent with the kinetic data is proposed, in which Tyr216-ArO-, in the course of being released from the active-site metal ion, deprotonates a water molecule attacking the Zn2+-activated substrate linkage, leading to a metal-coordinated tetrahedral oxyanion adduct that subsequently fragments. Metals 269-274 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-111 9328212-6 1997 Both series of synthetic compounds demonstrated good to excellent aromatase inhibition, and the most effective inhibitors in both series were those compounds with a phenylpropyl substituent at the 7alpha-position of the steroid nucleus. Steroids 220-227 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-75 9328212-7 1997 The 7alpha-arylaliphatic androst-4-ene-3,17-diones exhibited inhibition of JAr aromatase activity with IC50 values from 300 to 434 nM. 7alpha-arylaliphatic androst-4-ene-3,17-diones 4-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-88 9328212-8 1997 More potent aromatase inhibition was observed with the 7alpha-arylaliphatic androsta-1,4-diene-3,17-diones, which exhibited IC50 values from 64 to 232 nM. 7alpha-arylaliphatic androsta-1,4-diene-3,17-diones 55-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-21 9328212-11 1997 In summary, the 7alpha-phenylpropyl androsta-1,4-diene-3,17-dione analogs, which are enzyme-activated irreversible inhibitors, demonstrated the most effective inhibition of aromatase activity present in the JAr cell cultures among the various 7alpha-arylaliphatic androgens. 7alpha-phenylpropyl androsta-1,4-diene-3,17-dione 16-65 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 173-182 9365182-1 1997 Aromatase P450 (P450arom) is responsible for conversion of C19 steroids to estrogens in a number of human tissues, such as the placenta, gonads, adipose tissue, skin and the brain. Steroids 63-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-24 9365187-5 1997 Aromatase mRNA was increasingly expressed in myeloid THP 1 cells differentiated along the monocyte/phagocyte pathway exploiting vitamin D and either granulocyte-macrophage-stimulating factor (GMCSF) or macrophage-stimulating factor (MCSF). Vitamin D 128-137 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9365187-6 1997 In long-term cultures, when sequentially exposed to vitamin D (days 0-21) and GMCSF (days 5-10) and plated on collagen, the amount of expression of aromatase mRNA steadily increased along with the increasing expression of osteopontin mRNA, alpha(v) integrin mRNA, c-fms (MCSF-receptor) mRNA and multinucleated cells developing. Vitamin D 52-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 148-157 9365187-7 1997 The conversion of estradiol from testosterone (10(-7) M/l) in the supernatants of dishes mirrored changes in aromatase mRNA expression and by day 21 rose to 30,000 ng/10(7) cells/24 h. 17Beta-HSD IV mRNA expression was abundant in undifferentiated THP 1 cells and was decreased to approximately 50% by day 21. Estradiol 18-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 109-118 9365187-7 1997 The conversion of estradiol from testosterone (10(-7) M/l) in the supernatants of dishes mirrored changes in aromatase mRNA expression and by day 21 rose to 30,000 ng/10(7) cells/24 h. 17Beta-HSD IV mRNA expression was abundant in undifferentiated THP 1 cells and was decreased to approximately 50% by day 21. Testosterone 33-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 109-118 9365188-11 1997 However, removal of the serum or the addition of forskolin or phorbol ester (TPA) induced a rapid elevation of aromatase mRNA and the switching of aromatase transcripts to exon 1c in the cells, whereas TGFbeta almost abolished the expression of aromatase mRNA. Colforsin 49-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-120 9365188-11 1997 However, removal of the serum or the addition of forskolin or phorbol ester (TPA) induced a rapid elevation of aromatase mRNA and the switching of aromatase transcripts to exon 1c in the cells, whereas TGFbeta almost abolished the expression of aromatase mRNA. Colforsin 49-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 147-156 9365188-11 1997 However, removal of the serum or the addition of forskolin or phorbol ester (TPA) induced a rapid elevation of aromatase mRNA and the switching of aromatase transcripts to exon 1c in the cells, whereas TGFbeta almost abolished the expression of aromatase mRNA. Colforsin 49-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 147-156 9365188-11 1997 However, removal of the serum or the addition of forskolin or phorbol ester (TPA) induced a rapid elevation of aromatase mRNA and the switching of aromatase transcripts to exon 1c in the cells, whereas TGFbeta almost abolished the expression of aromatase mRNA. Phorbol Esters 62-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-120 9365188-11 1997 However, removal of the serum or the addition of forskolin or phorbol ester (TPA) induced a rapid elevation of aromatase mRNA and the switching of aromatase transcripts to exon 1c in the cells, whereas TGFbeta almost abolished the expression of aromatase mRNA. Phorbol Esters 62-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 147-156 9365188-11 1997 However, removal of the serum or the addition of forskolin or phorbol ester (TPA) induced a rapid elevation of aromatase mRNA and the switching of aromatase transcripts to exon 1c in the cells, whereas TGFbeta almost abolished the expression of aromatase mRNA. Phorbol Esters 62-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 147-156 9365188-11 1997 However, removal of the serum or the addition of forskolin or phorbol ester (TPA) induced a rapid elevation of aromatase mRNA and the switching of aromatase transcripts to exon 1c in the cells, whereas TGFbeta almost abolished the expression of aromatase mRNA. Tetradecanoylphorbol Acetate 77-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-120 9365188-11 1997 However, removal of the serum or the addition of forskolin or phorbol ester (TPA) induced a rapid elevation of aromatase mRNA and the switching of aromatase transcripts to exon 1c in the cells, whereas TGFbeta almost abolished the expression of aromatase mRNA. Tetradecanoylphorbol Acetate 77-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 147-156 9365188-11 1997 However, removal of the serum or the addition of forskolin or phorbol ester (TPA) induced a rapid elevation of aromatase mRNA and the switching of aromatase transcripts to exon 1c in the cells, whereas TGFbeta almost abolished the expression of aromatase mRNA. Tetradecanoylphorbol Acetate 77-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 147-156 9365188-12 1997 Because co-culture of cancer cells such as MCF-7 increased aromatase mRNA of the cells cultured in the serum-containing medium, it is possible that cancer cells secret stimulatory factors acting like forskolin or TPA, or consume serum inhibitory factors acting like TGFbeta, consequently causing levels of aromatase mRNA to increase. Colforsin 200-209 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-68 9365191-5 1997 In the present report, we show that prostaglandin (PG) E2 is the most potent factor which stimulates aromatase expression via cyclic AMP and promoter II. Dinoprostone 36-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-110 9365191-5 1997 In the present report, we show that prostaglandin (PG) E2 is the most potent factor which stimulates aromatase expression via cyclic AMP and promoter II. Cyclic AMP 126-136 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-110 9365202-10 1997 In addition, proliferation ([3H]-thymidine incorporation into DNA) during histoculture, was increased by both estradiol and testosterone in tumors with high aromatase activity. Tritium 29-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 157-166 9365202-10 1997 In addition, proliferation ([3H]-thymidine incorporation into DNA) during histoculture, was increased by both estradiol and testosterone in tumors with high aromatase activity. Thymidine 33-42 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 157-166 9365202-10 1997 In addition, proliferation ([3H]-thymidine incorporation into DNA) during histoculture, was increased by both estradiol and testosterone in tumors with high aromatase activity. Estradiol 110-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 157-166 9365202-10 1997 In addition, proliferation ([3H]-thymidine incorporation into DNA) during histoculture, was increased by both estradiol and testosterone in tumors with high aromatase activity. Testosterone 124-136 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 157-166 9365204-7 1997 Aromatase overexpression is not necessarily correlated with expression of 17beta-hydroxysteroid dehydrogenase type 1, which converts estrone to estradiol and estrogen receptor. Estrone 133-140 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9365204-7 1997 Aromatase overexpression is not necessarily correlated with expression of 17beta-hydroxysteroid dehydrogenase type 1, which converts estrone to estradiol and estrogen receptor. Estradiol 144-153 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 9140222-0 1997 Inhibition of 5 alpha-reductase and aromatase by the ellagitannins oenothein A and oenothein B from Epilobium species. Hydrolyzable Tannins 53-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-45 9140222-0 1997 Inhibition of 5 alpha-reductase and aromatase by the ellagitannins oenothein A and oenothein B from Epilobium species. oenothein A 67-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-45 8989235-2 1997 The biosynthesis of estrogens from C19 steroids is catalyzed by a specific form of cytochrome P450, namely aromatase cytochrome P450 (P450arom; the product of the CYP19 gene). Steroids 39-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 134-142 9024261-1 1997 C19 steroids are converted to estrogens by aromatase P450 (P450arom). Steroids 4-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-67 9061607-7 1997 These observations indicated that the capacity of tilapia ovarian follicles to synthesize estradiol-17 beta is closely related to the contents of P450arom mRNA and protein within them. Estradiol 90-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 146-154 8989235-2 1997 The biosynthesis of estrogens from C19 steroids is catalyzed by a specific form of cytochrome P450, namely aromatase cytochrome P450 (P450arom; the product of the CYP19 gene). Steroids 39-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 163-168 11666906-3 1996 1 reacts with 1 equiv of SmI(2) in THF to give Sm(II) mixed aryloxide/iodide [(ArO)Sm(&mgr;-I)(THF)(3)](2) (2), which adopts a dimeric structure via very weak Sm.I (3.534(2) A) interactions. samarium diiodide 25-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-82 9238853-10 1997 PGE2 is also an important regulator of aromatase expression in adipose mesenchymal cells via cAMP and PGE2 appears to be a major factor produced by breast tumors that stimulates estrogen biosynthesis in local mesenchymal sites. Dinoprostone 0-4 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-48 9238853-10 1997 PGE2 is also an important regulator of aromatase expression in adipose mesenchymal cells via cAMP and PGE2 appears to be a major factor produced by breast tumors that stimulates estrogen biosynthesis in local mesenchymal sites. Cyclic AMP 93-97 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-48 9238853-10 1997 PGE2 is also an important regulator of aromatase expression in adipose mesenchymal cells via cAMP and PGE2 appears to be a major factor produced by breast tumors that stimulates estrogen biosynthesis in local mesenchymal sites. Dinoprostone 102-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-48 11666906-3 1996 1 reacts with 1 equiv of SmI(2) in THF to give Sm(II) mixed aryloxide/iodide [(ArO)Sm(&mgr;-I)(THF)(3)](2) (2), which adopts a dimeric structure via very weak Sm.I (3.534(2) A) interactions. tetrahydrofuran 35-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-82 11666906-3 1996 1 reacts with 1 equiv of SmI(2) in THF to give Sm(II) mixed aryloxide/iodide [(ArO)Sm(&mgr;-I)(THF)(3)](2) (2), which adopts a dimeric structure via very weak Sm.I (3.534(2) A) interactions. aryloxide/iodide 60-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-82 11666906-3 1996 1 reacts with 1 equiv of SmI(2) in THF to give Sm(II) mixed aryloxide/iodide [(ArO)Sm(&mgr;-I)(THF)(3)](2) (2), which adopts a dimeric structure via very weak Sm.I (3.534(2) A) interactions. Adenosine Monophosphate 87-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-82 11666906-3 1996 1 reacts with 1 equiv of SmI(2) in THF to give Sm(II) mixed aryloxide/iodide [(ArO)Sm(&mgr;-I)(THF)(3)](2) (2), which adopts a dimeric structure via very weak Sm.I (3.534(2) A) interactions. tetrahydrofuran 99-102 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-82 11666906-5 1996 Oxidation of 1 with 0.5 equiv of I(2) in THF gives monomeric samarium(III) aryloxide/iodide (ArO)(2)SmI(THF)(2) (4), while the similar reaction of 1 with ClCH(2)CH(2)Cl or (t)BuCl in THF affords dimeric samarium(III) aryloxide/chloride [(ArO)(2)Sm(&mgr;-Cl)(THF)](2) (5). tetrahydrofuran 41-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 8940410-0 1996 Estrogen biosynthesis proximal to a breast tumor is stimulated by PGE2 via cyclic AMP, leading to activation of promoter II of the CYP19 (aromatase) gene. Dinoprostone 66-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 131-136 8940410-0 1996 Estrogen biosynthesis proximal to a breast tumor is stimulated by PGE2 via cyclic AMP, leading to activation of promoter II of the CYP19 (aromatase) gene. Dinoprostone 66-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-147 8940410-0 1996 Estrogen biosynthesis proximal to a breast tumor is stimulated by PGE2 via cyclic AMP, leading to activation of promoter II of the CYP19 (aromatase) gene. Cyclic AMP 75-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 131-136 8940410-0 1996 Estrogen biosynthesis proximal to a breast tumor is stimulated by PGE2 via cyclic AMP, leading to activation of promoter II of the CYP19 (aromatase) gene. Cyclic AMP 75-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-147 8940410-1 1996 In the present report, we show that prostaglandin (PG) E2 is the most potent factor which stimulates aromatase expression via cyclic AMP and promoter II. Dinoprostone 36-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-110 8940410-1 1996 In the present report, we show that prostaglandin (PG) E2 is the most potent factor which stimulates aromatase expression via cyclic AMP and promoter II. Cyclic AMP 126-136 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-110 11666906-5 1996 Oxidation of 1 with 0.5 equiv of I(2) in THF gives monomeric samarium(III) aryloxide/iodide (ArO)(2)SmI(THF)(2) (4), while the similar reaction of 1 with ClCH(2)CH(2)Cl or (t)BuCl in THF affords dimeric samarium(III) aryloxide/chloride [(ArO)(2)Sm(&mgr;-Cl)(THF)](2) (5). aryloxide 75-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 11666906-5 1996 Oxidation of 1 with 0.5 equiv of I(2) in THF gives monomeric samarium(III) aryloxide/iodide (ArO)(2)SmI(THF)(2) (4), while the similar reaction of 1 with ClCH(2)CH(2)Cl or (t)BuCl in THF affords dimeric samarium(III) aryloxide/chloride [(ArO)(2)Sm(&mgr;-Cl)(THF)](2) (5). aryloxide 75-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 238-241 11666906-5 1996 Oxidation of 1 with 0.5 equiv of I(2) in THF gives monomeric samarium(III) aryloxide/iodide (ArO)(2)SmI(THF)(2) (4), while the similar reaction of 1 with ClCH(2)CH(2)Cl or (t)BuCl in THF affords dimeric samarium(III) aryloxide/chloride [(ArO)(2)Sm(&mgr;-Cl)(THF)](2) (5). tetrahydrofuran 104-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 11666906-5 1996 Oxidation of 1 with 0.5 equiv of I(2) in THF gives monomeric samarium(III) aryloxide/iodide (ArO)(2)SmI(THF)(2) (4), while the similar reaction of 1 with ClCH(2)CH(2)Cl or (t)BuCl in THF affords dimeric samarium(III) aryloxide/chloride [(ArO)(2)Sm(&mgr;-Cl)(THF)](2) (5). clch(2)ch(2)cl 154-168 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 11666906-5 1996 Oxidation of 1 with 0.5 equiv of I(2) in THF gives monomeric samarium(III) aryloxide/iodide (ArO)(2)SmI(THF)(2) (4), while the similar reaction of 1 with ClCH(2)CH(2)Cl or (t)BuCl in THF affords dimeric samarium(III) aryloxide/chloride [(ArO)(2)Sm(&mgr;-Cl)(THF)](2) (5). bucl 175-179 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 11666906-5 1996 Oxidation of 1 with 0.5 equiv of I(2) in THF gives monomeric samarium(III) aryloxide/iodide (ArO)(2)SmI(THF)(2) (4), while the similar reaction of 1 with ClCH(2)CH(2)Cl or (t)BuCl in THF affords dimeric samarium(III) aryloxide/chloride [(ArO)(2)Sm(&mgr;-Cl)(THF)](2) (5). tetrahydrofuran 104-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 11666906-5 1996 Oxidation of 1 with 0.5 equiv of I(2) in THF gives monomeric samarium(III) aryloxide/iodide (ArO)(2)SmI(THF)(2) (4), while the similar reaction of 1 with ClCH(2)CH(2)Cl or (t)BuCl in THF affords dimeric samarium(III) aryloxide/chloride [(ArO)(2)Sm(&mgr;-Cl)(THF)](2) (5). aryloxide 217-226 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 11666906-5 1996 Oxidation of 1 with 0.5 equiv of I(2) in THF gives monomeric samarium(III) aryloxide/iodide (ArO)(2)SmI(THF)(2) (4), while the similar reaction of 1 with ClCH(2)CH(2)Cl or (t)BuCl in THF affords dimeric samarium(III) aryloxide/chloride [(ArO)(2)Sm(&mgr;-Cl)(THF)](2) (5). Chlorides 227-235 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 11666906-5 1996 Oxidation of 1 with 0.5 equiv of I(2) in THF gives monomeric samarium(III) aryloxide/iodide (ArO)(2)SmI(THF)(2) (4), while the similar reaction of 1 with ClCH(2)CH(2)Cl or (t)BuCl in THF affords dimeric samarium(III) aryloxide/chloride [(ArO)(2)Sm(&mgr;-Cl)(THF)](2) (5). Adenosine Monophosphate 249-252 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 11666906-5 1996 Oxidation of 1 with 0.5 equiv of I(2) in THF gives monomeric samarium(III) aryloxide/iodide (ArO)(2)SmI(THF)(2) (4), while the similar reaction of 1 with ClCH(2)CH(2)Cl or (t)BuCl in THF affords dimeric samarium(III) aryloxide/chloride [(ArO)(2)Sm(&mgr;-Cl)(THF)](2) (5). tetrahydrofuran 104-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 8923858-1 1996 The in situ formation of estradiol plays an important role in the development and biological behavior of human breast cancer Aromatase and 17 beta-hydroxysteroid dehydrogenase type 1 (17 beta-HSD type 1) are two principal enzymes involved in in situ estradiol production. Estradiol 25-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-134 8923858-1 1996 The in situ formation of estradiol plays an important role in the development and biological behavior of human breast cancer Aromatase and 17 beta-hydroxysteroid dehydrogenase type 1 (17 beta-HSD type 1) are two principal enzymes involved in in situ estradiol production. Estradiol 250-259 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 125-134 8923826-3 1996 The biosynthesis of estrogen from C19 steroids is catalyzed by a specific form of cytochrome P450, namely aromatase cytochrome P450 (P450arom; the product of the CYP19 gene). Steroids 38-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 133-141 8923461-1 1996 Expression of aromatase P450 (P450arom; the product of the CYP19 gene) in human adipose stromal cells in primary culture is markedly stimulated by serum in the presence of dexamethasone (DEX). Dexamethasone 172-185 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-38 8923826-3 1996 The biosynthesis of estrogen from C19 steroids is catalyzed by a specific form of cytochrome P450, namely aromatase cytochrome P450 (P450arom; the product of the CYP19 gene). Steroids 38-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 162-167 8923461-1 1996 Expression of aromatase P450 (P450arom; the product of the CYP19 gene) in human adipose stromal cells in primary culture is markedly stimulated by serum in the presence of dexamethasone (DEX). Dexamethasone 172-185 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-64 8923461-1 1996 Expression of aromatase P450 (P450arom; the product of the CYP19 gene) in human adipose stromal cells in primary culture is markedly stimulated by serum in the presence of dexamethasone (DEX). Dexamethasone 187-190 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-38 8923461-1 1996 Expression of aromatase P450 (P450arom; the product of the CYP19 gene) in human adipose stromal cells in primary culture is markedly stimulated by serum in the presence of dexamethasone (DEX). Dexamethasone 187-190 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-64 8632407-0 1996 Tetrahydronaphthalenes: influence of heterocyclic substituents on inhibition of steroid enzymes P450 arom and P450 17. Tetrahydronaphthalenes 0-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-117 8937433-0 1996 Mechanism for aromatase inactivation by a suicide substrate, androst-4-ene-3,6,17-trione. androst-4-ene-3,6,17-trione 61-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 8937433-2 1996 Aromatase is a cytochrome P450 enzyme complex that catalyzes the conversion of androst-4-ene-3,17-dione to estrone through three sequential oxygenations of the 19-methyl group. Androstenedione 79-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 8937433-2 1996 Aromatase is a cytochrome P450 enzyme complex that catalyzes the conversion of androst-4-ene-3,17-dione to estrone through three sequential oxygenations of the 19-methyl group. Estrone 107-114 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 8937433-3 1996 Androst-4-ene-3,6,17-trione (1) is a suicide substrate of aromatase. androst-4-ene-3,6,17-trione 0-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-67 8937433-5 1996 To further clarify the mechanism, 4 beta, 5 beta-epoxyandrosta-3,6,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase, upon treatment of compound 3 with hydrogen peroxide in the presence of NaHCO3. 4 beta, 5 beta-epoxyandrosta-3,6,17,19-tetraone 34-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 200-209 8937433-5 1996 To further clarify the mechanism, 4 beta, 5 beta-epoxyandrosta-3,6,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase, upon treatment of compound 3 with hydrogen peroxide in the presence of NaHCO3. Hydrogen Peroxide 245-262 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 200-209 8937433-5 1996 To further clarify the mechanism, 4 beta, 5 beta-epoxyandrosta-3,6,17,19-tetraone (6) was synthesized as a candidate for a reactive electrophile involved in irreversible binding to the active site of aromatase, upon treatment of compound 3 with hydrogen peroxide in the presence of NaHCO3. Sodium Bicarbonate 282-288 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 200-209 8937433-6 1996 The epoxide 6 inhibited human placental aromatase in a competitive manner (Ki = 30 microM); moreover, it inactivated the enzyme in an active-site-directed manner in the absence of NADPH (K1 = 88 microM, kinact = 0.071 min-1). Epoxy Compounds 4-11 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 40-49 8937433-10 1996 The results clearly indicate that the 4 beta, 5 beta-epoxy-19-oxo compound 6, which is possibly produced from 19-oxo-4-ene steroid 3 through the 19-hydroxy-19-hydroperoxide intermediate, is a reactive electrophile that irreversibly binds to the active site of aromatase. 4 beta, 5 beta-epoxy-19-oxo 38-65 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 260-269 8937433-10 1996 The results clearly indicate that the 4 beta, 5 beta-epoxy-19-oxo compound 6, which is possibly produced from 19-oxo-4-ene steroid 3 through the 19-hydroxy-19-hydroperoxide intermediate, is a reactive electrophile that irreversibly binds to the active site of aromatase. 19-oxo-4-ene steroid 110-130 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 260-269 8937433-10 1996 The results clearly indicate that the 4 beta, 5 beta-epoxy-19-oxo compound 6, which is possibly produced from 19-oxo-4-ene steroid 3 through the 19-hydroxy-19-hydroperoxide intermediate, is a reactive electrophile that irreversibly binds to the active site of aromatase. 19-hydroxy-19-hydroperoxide 145-172 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 260-269 9010616-1 1996 A novel molecular modelling study, involving inhibitors bound to a "substrate-heme complex", is described for steroidal and non-steroidal inhibitors of Aromatase (AR). Heme 78-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 152-161 9010616-1 1996 A novel molecular modelling study, involving inhibitors bound to a "substrate-heme complex", is described for steroidal and non-steroidal inhibitors of Aromatase (AR). Heme 78-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 163-165 8758911-1 1996 Aromatase, a cytochrome P450, catalyzes three consecutive hydroxylation reactions converting C19 androgens to aromatic C18 estrogenic steroids. Steroids 134-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 8686761-5 1996 Quantitation of aromatase activity with the tritiated water method demonstrated 41.62 +/- 9.15 pmol/hour/mg protein in 11 ovarian carcinomas. Water 54-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-25 8686761-11 1996 Increased aromatase expression in stromal cells of human ovarian carcinoma is, therefore, considered to play an important role in the biological behavior of these tumors by producing estrogens in situ as in other female sex-steroid-dependent neoplasms. Steroids 224-231 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-19 8620462-4 1996 The steroidal aromatase inhibitor 4-hydroxy-4-androstene-3,17-dione (4-OHA) and its derivative, 4-methoxy-4-androstene-3,17-dione (4-OMA), inhibited aromatization of testosterone in a competitive pattern of inhibition. formestane 34-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 8620462-4 1996 The steroidal aromatase inhibitor 4-hydroxy-4-androstene-3,17-dione (4-OHA) and its derivative, 4-methoxy-4-androstene-3,17-dione (4-OMA), inhibited aromatization of testosterone in a competitive pattern of inhibition. formestane 69-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 8620462-4 1996 The steroidal aromatase inhibitor 4-hydroxy-4-androstene-3,17-dione (4-OHA) and its derivative, 4-methoxy-4-androstene-3,17-dione (4-OMA), inhibited aromatization of testosterone in a competitive pattern of inhibition. 4-methoxy-4-androstene-3,17-dione 96-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 8620462-4 1996 The steroidal aromatase inhibitor 4-hydroxy-4-androstene-3,17-dione (4-OHA) and its derivative, 4-methoxy-4-androstene-3,17-dione (4-OMA), inhibited aromatization of testosterone in a competitive pattern of inhibition. Testosterone 166-178 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 8854046-0 1996 Molecular modelling study of pyrrolidine-2,5-dione based aromatase inhibitors and other known inhibitors. succinimide 29-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-66 8854046-2 1996 The study suggests that the "larger" inhibitors (such as the [2"(4"-aminophenyl)alkyl] pyrrolidine-2,5-dione based compounds), after an initial binding of the phenylamine nitrogen lone pair electrons with the Fe3+ haem of the cytochrome P-450, preferentially utilise the region of the AR active site which would normally bind C(17) = O of the substrate. [2"(4"-aminophenyl)alkyl] pyrrolidine-2,5-dione 61-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 285-287 8854046-2 1996 The study suggests that the "larger" inhibitors (such as the [2"(4"-aminophenyl)alkyl] pyrrolidine-2,5-dione based compounds), after an initial binding of the phenylamine nitrogen lone pair electrons with the Fe3+ haem of the cytochrome P-450, preferentially utilise the region of the AR active site which would normally bind C(17) = O of the substrate. Aniline Compounds 159-170 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 285-287 8854046-2 1996 The study suggests that the "larger" inhibitors (such as the [2"(4"-aminophenyl)alkyl] pyrrolidine-2,5-dione based compounds), after an initial binding of the phenylamine nitrogen lone pair electrons with the Fe3+ haem of the cytochrome P-450, preferentially utilise the region of the AR active site which would normally bind C(17) = O of the substrate. Nitrogen 171-179 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 285-287 8854046-2 1996 The study suggests that the "larger" inhibitors (such as the [2"(4"-aminophenyl)alkyl] pyrrolidine-2,5-dione based compounds), after an initial binding of the phenylamine nitrogen lone pair electrons with the Fe3+ haem of the cytochrome P-450, preferentially utilise the region of the AR active site which would normally bind C(17) = O of the substrate. fe3+ haem 209-218 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 285-287 8793855-1 1996 Using the membrane impermeant probe NHS-LC-biotin, we show in this report that a fraction of aromatase P450 (P450arom), the enzyme that catalyzes estrogen biosynthesis, is present at the surface of cells in which it is expressed, either endogenously or as a consequence of transfection. sulfosuccinimidyl 6-(biotinamido)hexanoate 36-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 103-107 8793855-1 1996 Using the membrane impermeant probe NHS-LC-biotin, we show in this report that a fraction of aromatase P450 (P450arom), the enzyme that catalyzes estrogen biosynthesis, is present at the surface of cells in which it is expressed, either endogenously or as a consequence of transfection. sulfosuccinimidyl 6-(biotinamido)hexanoate 36-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 109-117 8612525-4 1996 Glucocorticoids at 10(-9)-10(-7) M transiently induced the expression and enzymatic activity of aromatase cytochrome P450 (P450AROM) in primary cultured osteoblasts, and the Km value for androstenedione (4.7 +/- 2.9 nM) was lower than that in adipose tissue and skin. Androstenedione 187-202 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 123-131 8612525-6 1996 1,25-Dihydroxyvitamin D3 [1,25-(OH)2D3] alone did not induce aromatase activity, but enhanced and maintained glucocorticoid-induced P450AROM gene expression. 1,25-dihydroxyvitamin d3 [1,25-(oh)2d3 0-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 132-140 8612525-8 1996 The enhancement of P450AROM activity by 1,25-(OH)2D3 varied from 0.94-fold (no enhancement) to 2.40-fold (maximal enhancement) among the individual human osteoblasts examined, but the magnitude of the enhancement was significantly correlated with the level of vitamin D receptor messenger RNA (P < 0.05). Calcitriol 40-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 19-27 8772611-1 1996 The fibroblast component of adipose tissue is the primary extraglandular site of aromatase P450 (P450arom) expression, which is responsible for the conversion of C19 steroids to estrogens. Steroids 166-174 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 97-105 8632407-9 1996 (P450 arom: K(m) testosterone = 42 nM, K(i)16 = 33 nM, K(i)20 = 3 microM. Testosterone 17-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 1-10 8808317-1 1995 C19 steroids are converted to oestrogens in a number of tissues by a specific form of cytochrome P450, namely aromatase P450 (P450arom; the product of the CYP19 gene). Steroids 4-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 126-134 8793642-2 1996 After electrophoresis in PAG, the new alleles 7, 10, 11, 12, and 13 were detected at the CYP19 locus in Japanese. phenylacetylglycine 25-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 89-94 8550748-1 1996 The conversion of C19 steroids to estrogens occurs in a number of tissues, such as the ovary and placenta, and is catalyzed by aromatase P450 (P450arom; the product of the CYP19 gene). Steroids 22-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-136 8550748-1 1996 The conversion of C19 steroids to estrogens occurs in a number of tissues, such as the ovary and placenta, and is catalyzed by aromatase P450 (P450arom; the product of the CYP19 gene). Steroids 22-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 143-151 8550748-1 1996 The conversion of C19 steroids to estrogens occurs in a number of tissues, such as the ovary and placenta, and is catalyzed by aromatase P450 (P450arom; the product of the CYP19 gene). Steroids 22-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 172-177 8562679-7 1995 The immunoreactive P450arom found in epididymal sperm was shown to be active through use of a 3H2O assay. 3h2o 94-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 19-27 8817247-10 1996 The observed lack of aromatase inhibitor influence upon basal secretion of estradiol by SC separated from mature corpora lutea, and the decrease of testosterone-stimulated estradiol secretion by SC isolated from regressing corpora lutea, indicate that cytodifferentiation during luteinization could cause expression of P450arom in theca-derived luteal cells. Testosterone 148-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 319-327 8808317-1 1995 C19 steroids are converted to oestrogens in a number of tissues by a specific form of cytochrome P450, namely aromatase P450 (P450arom; the product of the CYP19 gene). Steroids 4-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 155-160 8674811-6 1995 In vitro expression indicated that the cDNA encoding porcine placental P-450arom was almost 10-fold more active in the synthesis of estrone from androstenedione than was the ovarian isoform which synthesized more 19OH-androstenedione than estrone. Estrone 132-139 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-80 7501447-3 1995 Despite this, we have detected triplex-directed photoadduct formation at pH 7.0 between the psoralen-linked oligonucleotide and a 30mer duplex representing the aromatase target. psoralen-linked oligonucleotide 92-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 160-169 7579495-0 1995 Aromatase inhibition with 4-OHAndrostenedione after prior aromatase inhibition with aminoglutethimide in women with advanced breast cancer. 4-ohandrostenedione 26-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 7579495-0 1995 Aromatase inhibition with 4-OHAndrostenedione after prior aromatase inhibition with aminoglutethimide in women with advanced breast cancer. 4-ohandrostenedione 26-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-67 7579495-0 1995 Aromatase inhibition with 4-OHAndrostenedione after prior aromatase inhibition with aminoglutethimide in women with advanced breast cancer. Aminoglutethimide 84-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 7579495-0 1995 Aromatase inhibition with 4-OHAndrostenedione after prior aromatase inhibition with aminoglutethimide in women with advanced breast cancer. Aminoglutethimide 84-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-67 7579495-1 1995 One hundred and twelve post menopausal or post oophorectomy women with advanced breast cancer (BC) who had all previously had aminoglutethimide (AG) were treated with the potent aromatase inhibitor 4-hydroxy androstenedione (4-OHA). formestane 198-223 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 178-187 7579495-5 1995 This study shows that potent aromatase inhibition with 4-OHA is effective in women with advanced BC who have already been treated with a less potent aromatase inhibitor, and suggests that relative changes in oestrogen levels may be more important than absolute levels. formestane 55-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-38 7579495-5 1995 This study shows that potent aromatase inhibition with 4-OHA is effective in women with advanced BC who have already been treated with a less potent aromatase inhibitor, and suggests that relative changes in oestrogen levels may be more important than absolute levels. formestane 55-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 149-158 7556673-2 1995 We have designed a 20-base pyrimidine oligodeoxynucleotide (ODN) which forms a sequence-specific triple helix (triplex) with a purine-rich tract in the P450arom coding sequence. 20-base pyrimidine oligodeoxynucleotide 19-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 152-160 7556673-2 1995 We have designed a 20-base pyrimidine oligodeoxynucleotide (ODN) which forms a sequence-specific triple helix (triplex) with a purine-rich tract in the P450arom coding sequence. Oligodeoxyribonucleotides 60-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 152-160 7556673-2 1995 We have designed a 20-base pyrimidine oligodeoxynucleotide (ODN) which forms a sequence-specific triple helix (triplex) with a purine-rich tract in the P450arom coding sequence. purine 127-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 152-160 7556673-4 1995 Cross-linked adducts formed in vitro between ODNs and P450arom expression constructs were used to transfect COS and human MCF-7 breast cancer cells. carbonyl sulfide 108-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 54-62 8674811-6 1995 In vitro expression indicated that the cDNA encoding porcine placental P-450arom was almost 10-fold more active in the synthesis of estrone from androstenedione than was the ovarian isoform which synthesized more 19OH-androstenedione than estrone. Androstenedione 145-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-80 8674811-6 1995 In vitro expression indicated that the cDNA encoding porcine placental P-450arom was almost 10-fold more active in the synthesis of estrone from androstenedione than was the ovarian isoform which synthesized more 19OH-androstenedione than estrone. 19oh-androstenedione 213-233 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-80 8674811-6 1995 In vitro expression indicated that the cDNA encoding porcine placental P-450arom was almost 10-fold more active in the synthesis of estrone from androstenedione than was the ovarian isoform which synthesized more 19OH-androstenedione than estrone. Estrone 239-246 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-80 8674811-9 1995 In addition, activity of the enzyme encoded by the ovarian P-450arom cDNA was suppressed by etomidate, an inhibitor of cytochrome P-450 11beta-hydroxylase, but the placental P-450arom isoform was not. Etomidate 92-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-68 8674811-9 1995 In addition, activity of the enzyme encoded by the ovarian P-450arom cDNA was suppressed by etomidate, an inhibitor of cytochrome P-450 11beta-hydroxylase, but the placental P-450arom isoform was not. Etomidate 92-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 174-183 7783141-0 1995 Pyridyl-substituted tetrahydrocyclopropa[a]naphthalenes: highly active and selective inhibitors of P450 arom. pyridyl-substituted tetrahydrocyclopropa[a]naphthalenes 0-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-108 7662584-1 1995 The synthesis and biological evaluation of 4-amino-, 4-alkoxy-, 4-aryloxy-, 4-alkyl- and 4-aryl-4-androstenedione derivatives as inhibitors of estrogen synthetase (aromatase) are described. 4-amino-, 4-alkoxy-, 4-aryloxy-, 4-alkyl- and 4-aryl-4-androstenedione 43-113 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 143-162 7783141-3 1995 The racemic compounds 1-10 showed an inhibition of human placental aromatase (P450 arom) exhibiting relative potencies (rp) from 3.7 to 303 (compounds 8 and 4, respectively; rp of aminoglutethimide (AG) identical to 1, fadrozole = 359). Aminoglutethimide 180-197 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-87 7783141-3 1995 The racemic compounds 1-10 showed an inhibition of human placental aromatase (P450 arom) exhibiting relative potencies (rp) from 3.7 to 303 (compounds 8 and 4, respectively; rp of aminoglutethimide (AG) identical to 1, fadrozole = 359). Fadrozole 219-228 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-87 7874687-4 1995 Using a cell line derived from the D2 tumor, we have demonstrated the effect of the aromatase substrate, androstenedione, on the proliferation of tumor cells. Androstenedione 105-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-93 7626449-9 1995 Reverse transcription-polymerase chain reaction analysis revealed that dexamethasone increased the transcript of P450AROM gene. Dexamethasone 71-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-121 7626449-11 1995 Dexamethasone and 1 alpha,25-dihydroxyvitamin D3 synergistically enhanced aromatase activity and P450AROM mRNA expression. Dexamethasone 0-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 97-105 7626449-11 1995 Dexamethasone and 1 alpha,25-dihydroxyvitamin D3 synergistically enhanced aromatase activity and P450AROM mRNA expression. Calcitriol 18-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 97-105 7626449-12 1995 These results demonstrate that adrenal androgen, DHEA, is converted to E1 in osteoblast by P450AROM which is positively regulated by glucocorticoid and 1 alpha,25-dihydroxyvitamin D3 and important to maintain BMD in the 6 to 7th decade, after menopause. Dehydroepiandrosterone 49-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-99 7626449-12 1995 These results demonstrate that adrenal androgen, DHEA, is converted to E1 in osteoblast by P450AROM which is positively regulated by glucocorticoid and 1 alpha,25-dihydroxyvitamin D3 and important to maintain BMD in the 6 to 7th decade, after menopause. alpha,25-dihydroxyvitamin d3 154-182 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-99 7626451-9 1995 These results indicate that NF-IL6 is one of the nuclear factors which participate in TPA-mediated transcriptional enhancement of CYP 19 gene expression. Tetradecanoylphorbol Acetate 86-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-136 7720683-3 1995 In FSH-primed cells, TNF alpha inhibited P450-AROM activity in a dose-dependent manner, an effect that was also observed in cells treated with bacterial sphingomyelinase (SMase 0.003-0.3 U/ml) or increasing concentrations (0.1-10 microM) of N-acetylsphingosine (C2-cer) a membrane-permeable analogue of ceramide. N-acetylsphingosine 241-260 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-50 7720683-3 1995 In FSH-primed cells, TNF alpha inhibited P450-AROM activity in a dose-dependent manner, an effect that was also observed in cells treated with bacterial sphingomyelinase (SMase 0.003-0.3 U/ml) or increasing concentrations (0.1-10 microM) of N-acetylsphingosine (C2-cer) a membrane-permeable analogue of ceramide. Ceramides 303-311 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-50 7874687-5 1995 Proliferative effects of androstenedione were blocked by an aromatase inhibitor, providing evidence for the role of int-5/aromatase in this process. Androstenedione 25-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-69 7874687-5 1995 Proliferative effects of androstenedione were blocked by an aromatase inhibitor, providing evidence for the role of int-5/aromatase in this process. Androstenedione 25-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-131 7874687-6 1995 Furthermore, the androstenedione-mediated proliferation was inhibited by the addition of anti-estrogen ICI 164,384, suggesting that the estrogen formed from the conversion of androstenedione by int-5/aromatase acts like a mitogen to stimulate the growth of D2 tumor cells. Androstenedione 17-32 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 200-209 7874687-6 1995 Furthermore, the androstenedione-mediated proliferation was inhibited by the addition of anti-estrogen ICI 164,384, suggesting that the estrogen formed from the conversion of androstenedione by int-5/aromatase acts like a mitogen to stimulate the growth of D2 tumor cells. Androstenedione 175-190 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 200-209 7962323-9 1994 The anaplastic thyroid carcinoma cell line (ARO) had 1 APC allele with an adenine insertion at codon 1556 (ACTA to AACTA), leading to a premature stop codon at 1558. Adenine 74-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-47 8847599-0 1995 Evaluation of 6,7-aziridinyl steroids and related compounds as inhibitors of aromatase (P-450arom). 6,7-aziridinyl steroids 14-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-98 7989490-1 1994 C19 steroids are converted to estrogens in a number of human tissues by the aromatase enzyme complex, which consists of aromatase cytochrome P450 (P450arom; product of the CYP19 gene) and NADPH-cytochrome P450 reductase. Steroids 4-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 147-155 7989490-1 1994 C19 steroids are converted to estrogens in a number of human tissues by the aromatase enzyme complex, which consists of aromatase cytochrome P450 (P450arom; product of the CYP19 gene) and NADPH-cytochrome P450 reductase. Steroids 4-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 172-177 7562025-5 1994 All microsatellites are dinucleotide CA, repeats except for D11S956,P23 (CTAT) and CYP 19, which are tetranucleotide repeats and P23 (GTTTT) which is a pentanucleotide repeat. tetranucleotide 101-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-89 7749129-4 1995 These include enzymatic synthesis of estradiol via sulfatase, aromatase, and 17 beta-hydroxysteroid dehydrogenase in the tumor itself. Estradiol 37-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-71 7749129-7 1995 To effectively suppress tissue concentrations of estrogens and circulating estradiol in postmenopausal patients, various aromatase inhibitors have been developed recently. Estradiol 75-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 121-130 9420848-9 1995 The majority of P450arom 5"-termini expressed by ASC incubated with CM plus DEX contained the promoter I.4-specific sequence. Dexamethasone 76-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-24 8034720-1 1994 Aromatase, a cytochrome P450, catalyzes three consecutive hydroxylation reactions converting C19 androgens to aromatic C18 estrogenic steroids. Steroids 134-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 7819446-8 1994 Thecal P450c17, thecal P450arom, and granulosa P450arom expression decreased coincidentally as serum estradiol-17 beta and follicular fluid estradiol-17 beta, testosterone, and androstenedione levels declined after the presumed gonadotropin surge. Estradiol 101-118 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-55 7819446-8 1994 Thecal P450c17, thecal P450arom, and granulosa P450arom expression decreased coincidentally as serum estradiol-17 beta and follicular fluid estradiol-17 beta, testosterone, and androstenedione levels declined after the presumed gonadotropin surge. Estradiol 140-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-55 7819446-8 1994 Thecal P450c17, thecal P450arom, and granulosa P450arom expression decreased coincidentally as serum estradiol-17 beta and follicular fluid estradiol-17 beta, testosterone, and androstenedione levels declined after the presumed gonadotropin surge. Testosterone 159-171 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-55 7819446-8 1994 Thecal P450c17, thecal P450arom, and granulosa P450arom expression decreased coincidentally as serum estradiol-17 beta and follicular fluid estradiol-17 beta, testosterone, and androstenedione levels declined after the presumed gonadotropin surge. Androstenedione 177-192 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-55 8034720-2 1994 On the basis of a recent computer modeling of the active site of aromatase, a hydrophobic pocket, thought to be important for the binding of some aromatase inhibitors, was predicted to extend roughly in the plane of the steroid substrate, from the position that would be occupied by its C4 and C7 atoms. Steroids 220-227 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 8034720-2 1994 On the basis of a recent computer modeling of the active site of aromatase, a hydrophobic pocket, thought to be important for the binding of some aromatase inhibitors, was predicted to extend roughly in the plane of the steroid substrate, from the position that would be occupied by its C4 and C7 atoms. Steroids 220-227 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 146-155 8034720-7 1994 In addition, this mutation reduces the binding affinity of an aromatase inhibitor, 4-hydroxyandrostenedione, and increases the binding affinity of two aromatase inhibitors, aminoglutethimide and CGS 16949. formestane 83-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-71 8034720-7 1994 In addition, this mutation reduces the binding affinity of an aromatase inhibitor, 4-hydroxyandrostenedione, and increases the binding affinity of two aromatase inhibitors, aminoglutethimide and CGS 16949. Aminoglutethimide 173-190 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-71 8034720-7 1994 In addition, this mutation reduces the binding affinity of an aromatase inhibitor, 4-hydroxyandrostenedione, and increases the binding affinity of two aromatase inhibitors, aminoglutethimide and CGS 16949. Aminoglutethimide 173-190 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 151-160 8034720-7 1994 In addition, this mutation reduces the binding affinity of an aromatase inhibitor, 4-hydroxyandrostenedione, and increases the binding affinity of two aromatase inhibitors, aminoglutethimide and CGS 16949. cysteinylglycine 195-198 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-71 8034720-7 1994 In addition, this mutation reduces the binding affinity of an aromatase inhibitor, 4-hydroxyandrostenedione, and increases the binding affinity of two aromatase inhibitors, aminoglutethimide and CGS 16949. cysteinylglycine 195-198 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 151-160 8182306-6 1994 12-O-tetradecanoyl phorbol 13-acetate (TPA) (0.1-100ng/ml), a protein-kinase C activator, also stimulated aromatase activity and increased the P-450arom concentration. Tetradecanoylphorbol Acetate 0-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 143-152 8174717-7 1994 RESULTS: In vitro, ketoconazole selectively inhibited the key steroidogenic cytochromes, namely P450scc, P45017 alpha, and P450arom (IC50 = 0.5 to 1.0 microgram/mL). Ketoconazole 19-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 123-131 8182306-6 1994 12-O-tetradecanoyl phorbol 13-acetate (TPA) (0.1-100ng/ml), a protein-kinase C activator, also stimulated aromatase activity and increased the P-450arom concentration. Tetradecanoylphorbol Acetate 39-42 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 143-152 8182306-7 1994 On the other hand, Ca2+ ionophore A23187, an agent increasing intracellular Ca2+ accumulation, inhibited aromatase activity and reduced the P-450arom concentration. Calcimycin 34-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 140-149 8132535-4 1994 Transient expression analysis in human BeWo choriocarcinoma cells, in which CYP19 is expressed, shows that hATRE-1 represses the expression of the bacterial chloramphenicol acetyltransferase reporter gene driven by the promoter of CYP19, whereas hATRE-2 enhances the reporter gene expression in response to 12-O-tetradecanoylphorbol 13-acetate. Tetradecanoylphorbol Acetate 307-343 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-81 8060487-0 1994 Differential regulation of cholesterol side-chain cleavage (P450scc) and aromatase (P450arom) enzyme mRNA expression by gonadotrophins and cyclic AMP in human granulosa cells. Cholesterol 27-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-82 8060487-0 1994 Differential regulation of cholesterol side-chain cleavage (P450scc) and aromatase (P450arom) enzyme mRNA expression by gonadotrophins and cyclic AMP in human granulosa cells. Cyclic AMP 139-149 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-82 8060487-0 1994 Differential regulation of cholesterol side-chain cleavage (P450scc) and aromatase (P450arom) enzyme mRNA expression by gonadotrophins and cyclic AMP in human granulosa cells. Cyclic AMP 139-149 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-92 8060487-2 1994 The crucial regulatory enzymes for progesterone and oestradiol biosynthesis in granulosa cells are the cholesterol side-chain cleavage (P450scc) and aromatase (P450arom) enzymes respectively. Progesterone 35-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-158 8060487-2 1994 The crucial regulatory enzymes for progesterone and oestradiol biosynthesis in granulosa cells are the cholesterol side-chain cleavage (P450scc) and aromatase (P450arom) enzymes respectively. Progesterone 35-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 160-168 8060487-2 1994 The crucial regulatory enzymes for progesterone and oestradiol biosynthesis in granulosa cells are the cholesterol side-chain cleavage (P450scc) and aromatase (P450arom) enzymes respectively. Estradiol 52-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-158 8060487-2 1994 The crucial regulatory enzymes for progesterone and oestradiol biosynthesis in granulosa cells are the cholesterol side-chain cleavage (P450scc) and aromatase (P450arom) enzymes respectively. Estradiol 52-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 160-168 8060487-2 1994 The crucial regulatory enzymes for progesterone and oestradiol biosynthesis in granulosa cells are the cholesterol side-chain cleavage (P450scc) and aromatase (P450arom) enzymes respectively. Cholesterol 103-114 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-158 8060487-2 1994 The crucial regulatory enzymes for progesterone and oestradiol biosynthesis in granulosa cells are the cholesterol side-chain cleavage (P450scc) and aromatase (P450arom) enzymes respectively. Cholesterol 103-114 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 160-168 8060487-5 1994 After the period of culture, total RNA was extracted from granulosa cells and Northern analyses were performed utilizing 32P-labelled cDNAs encoding P450arom and P450scc. Phosphorus-32 121-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 149-157 8060487-13 1994 Therefore, FSH/low cAMP levels stimulated P450arom gene expression and oestradiol production, while LH/high cAMP levels maximally induced P450scc gene expression and function, in a development-related manner consistent with steroid production in vivo. Cyclic AMP 19-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-50 8060487-14 1994 These findings support the hypothesis that one set of genes (like P450arom) in human granulosa cells is regulated by FSH/low cAMP levels and another (like P450scc) by LH/high cAMP levels. Cyclic AMP 125-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-75 8060487-14 1994 These findings support the hypothesis that one set of genes (like P450arom) in human granulosa cells is regulated by FSH/low cAMP levels and another (like P450scc) by LH/high cAMP levels. Luteinizing Hormone 167-169 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-75 8060487-14 1994 These findings support the hypothesis that one set of genes (like P450arom) in human granulosa cells is regulated by FSH/low cAMP levels and another (like P450scc) by LH/high cAMP levels. Cyclic AMP 175-179 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-75 18407196-1 1994 Extraglandular conversion of C19 steroids to estrogens takes place primarily in the stromal cell component of adipose tissue and is catalyzed by an enzyme complex comprising aromatase cytochrome P450 (P450arom; the product of the CYP19 gene) together with the flavoprotein NADPH-cytochrome P450 reductase. Steroids 33-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 174-183 18407196-1 1994 Extraglandular conversion of C19 steroids to estrogens takes place primarily in the stromal cell component of adipose tissue and is catalyzed by an enzyme complex comprising aromatase cytochrome P450 (P450arom; the product of the CYP19 gene) together with the flavoprotein NADPH-cytochrome P450 reductase. Steroids 33-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 201-209 18407196-1 1994 Extraglandular conversion of C19 steroids to estrogens takes place primarily in the stromal cell component of adipose tissue and is catalyzed by an enzyme complex comprising aromatase cytochrome P450 (P450arom; the product of the CYP19 gene) together with the flavoprotein NADPH-cytochrome P450 reductase. Steroids 33-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 230-235 8137936-1 1994 The regulation of aromatase (estrogen synthase) activity of cultured stromal vascular cells from human breast adipose tissue by cortisol, db-cAMP and growth factors was studied in a serum-free culture system. Bucladesine 138-145 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-46 8132535-4 1994 Transient expression analysis in human BeWo choriocarcinoma cells, in which CYP19 is expressed, shows that hATRE-1 represses the expression of the bacterial chloramphenicol acetyltransferase reporter gene driven by the promoter of CYP19, whereas hATRE-2 enhances the reporter gene expression in response to 12-O-tetradecanoylphorbol 13-acetate. Tetradecanoylphorbol Acetate 307-343 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 231-236 8170477-1 1994 The formation of estrogens from C19 steroids is catalyzed by a specific form of cytochrome P450, aromatase cytochrome P450 (P450arom; the product of the CYP19 gene). Steroids 36-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 124-132 8129748-6 1994 After incubation of the three double-labeled ATs, the solubilized proteins were subjected to SDS-PAGE and the 3H/14C ratio of the aromatase-bound metabolite in a 46-69 kDa fraction was analyzed. Tritium 110-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-139 8129748-6 1994 After incubation of the three double-labeled ATs, the solubilized proteins were subjected to SDS-PAGE and the 3H/14C ratio of the aromatase-bound metabolite in a 46-69 kDa fraction was analyzed. Carbon-14 113-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-139 8129748-8 1994 Thus, we conclude that further oxygenation of 19-oxo-AT produced by the two initial hydroxylations of AT at C-19 yields not only 6-oxoestrogen (by a mechanism similar to that involved in the aromatization of the natural substrate) but also a reactive electrophile that immediately binds to the active site in an irreversible manner, resulting in inactivation of aromatase. 19-oxo-at 46-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 362-371 7803322-1 1994 A new programme for the treatment of alcohol and drug dependence at the Neuropsychiatric Hospital, Aro, Abeokuta, Nigeria is based on the modified Minnesota/Therapeutic Community model. Alcohols 37-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-102 8126151-1 1994 The conversion of C19 steroids to estrogens occurs in a number of tissues and is catalyzed by a specific form of cytochrome P450, namely aromatase cytochrome P450 (P450arom). Steroids 22-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 164-172 8129748-1 1994 Aromatase catalyzes the conversion of androst-4-ene-3,17-dione to estrogen through sequential oxygenations at the 19-methyl group. Androstenedione 38-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 8129748-2 1994 Androst-4-ene-3,6,17-trione (AT) is a suicide substrate of aromatase, and the mechanism of inactivation of aromatase has been postulated to involve enzymatic oxygenation at the 19-position. androst-4-ene-3,6,17-trione 0-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-68 8129748-2 1994 Androst-4-ene-3,6,17-trione (AT) is a suicide substrate of aromatase, and the mechanism of inactivation of aromatase has been postulated to involve enzymatic oxygenation at the 19-position. androst-4-ene-3,6,17-trione 0-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-116 8129748-2 1994 Androst-4-ene-3,6,17-trione (AT) is a suicide substrate of aromatase, and the mechanism of inactivation of aromatase has been postulated to involve enzymatic oxygenation at the 19-position. androst-4-ene-3,6,17-trione 29-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-68 8129748-2 1994 Androst-4-ene-3,6,17-trione (AT) is a suicide substrate of aromatase, and the mechanism of inactivation of aromatase has been postulated to involve enzymatic oxygenation at the 19-position. androst-4-ene-3,6,17-trione 29-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-116 8170477-1 1994 The formation of estrogens from C19 steroids is catalyzed by a specific form of cytochrome P450, aromatase cytochrome P450 (P450arom; the product of the CYP19 gene). Steroids 36-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 153-158 8286214-1 1994 Aromatase is a key enzyme in the conversion of androstenedione and testosterone to oestrone and oestradiol. Androstenedione 47-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 7873460-0 1994 Aromatase inhibition: basic concepts, and the pharmacodynamics of formestane. formestane 66-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 8286214-1 1994 Aromatase is a key enzyme in the conversion of androstenedione and testosterone to oestrone and oestradiol. Testosterone 67-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 8286214-1 1994 Aromatase is a key enzyme in the conversion of androstenedione and testosterone to oestrone and oestradiol. Estrone 83-91 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 8286214-1 1994 Aromatase is a key enzyme in the conversion of androstenedione and testosterone to oestrone and oestradiol. Estradiol 96-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 8195346-1 1994 This study first examined the relative activities of 17 alpha-hydroxylase, 17,20-lyase and aromatase in human granulosa-lutein cells by challenging the cells with steroid precursors in the oestradiol biosynthetic pathway. Steroids 163-170 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-100 7949202-1 1994 Extraglandular conversion of C19 steroids to estrogens takes place primarily in the stromal cell compartments of adipose tissue and is catalyzed by aromatase cytochrome P450 (P450arom, the product of the CYP19 gene). c19 steroids 29-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 175-183 7949202-1 1994 Extraglandular conversion of C19 steroids to estrogens takes place primarily in the stromal cell compartments of adipose tissue and is catalyzed by aromatase cytochrome P450 (P450arom, the product of the CYP19 gene). c19 steroids 29-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 204-209 8195346-1 1994 This study first examined the relative activities of 17 alpha-hydroxylase, 17,20-lyase and aromatase in human granulosa-lutein cells by challenging the cells with steroid precursors in the oestradiol biosynthetic pathway. Estradiol 189-199 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-100 8117355-1 1993 C19 steroids are converted to estrogens in a number of tissues by a specific form of cytochrome P450, namely aromatase cytochrome P450 (P450arom). Steroids 4-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-144 7874190-1 1994 Extraglandular conversion of C19 steroids to estrogens takes place primarily in the stromal cell component of adipose tissue and is catalyzed by aromatase cytochrome P450 (P450arom; the product of the CYP19 gene). c19 steroids 29-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 145-154 7874190-1 1994 Extraglandular conversion of C19 steroids to estrogens takes place primarily in the stromal cell component of adipose tissue and is catalyzed by aromatase cytochrome P450 (P450arom; the product of the CYP19 gene). c19 steroids 29-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 172-180 7874190-1 1994 Extraglandular conversion of C19 steroids to estrogens takes place primarily in the stromal cell component of adipose tissue and is catalyzed by aromatase cytochrome P450 (P450arom; the product of the CYP19 gene). c19 steroids 29-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 201-206 7874190-10 1994 Our results underscore the importance of aromatase inhibitors as effective agents in treatment of hormone-responsive breast cancer, since aromatase inhibitors reduce local aromatase activity as well as blood estradiol levels. Estradiol 208-217 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-50 7874190-10 1994 Our results underscore the importance of aromatase inhibitors as effective agents in treatment of hormone-responsive breast cancer, since aromatase inhibitors reduce local aromatase activity as well as blood estradiol levels. Estradiol 208-217 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-147 7874190-10 1994 Our results underscore the importance of aromatase inhibitors as effective agents in treatment of hormone-responsive breast cancer, since aromatase inhibitors reduce local aromatase activity as well as blood estradiol levels. Estradiol 208-217 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-147 1331779-1 1992 Aromatase, a cytochrome P-450, catalyzes the formation of aromatic C18 estrogenic steroids from C19 androgens. Steroids 82-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 8145767-4 1993 With this in mind, we have generated a detailed alignment of P450arom, including the definition of putative alpha-helices and beta-sheets based on comparison of the alignments of P450BM3 and P450cam, generated from their three-dimensional structure, and have made mutations in regions we believe to be involved in substrate recognition at the solvent surface and orientation in the heme pocket. Heme 382-386 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-69 8143890-1 1993 Estrogens are synthesized from C19 steroids by a unique form of cytochrome P450, aromatase cytochrome P-450 (P-450AROM; the product of the CYP19 gene). Steroids 35-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 109-118 8143890-1 1993 Estrogens are synthesized from C19 steroids by a unique form of cytochrome P450, aromatase cytochrome P-450 (P-450AROM; the product of the CYP19 gene). Steroids 35-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-144 7684741-8 1993 A single stranded cDNA was synthesized from total RNA using Moloney murine leukemia virus reverse transcriptase and a P450arom-specific oligonucleotide. Oligonucleotides 136-151 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 118-126 8384878-1 1993 It has recently been demonstrated that strong illumination under anaerobic conditions leads to the double reduction of the primary quinone acceptor, QA, which in turn promotes the light-induced formation of triplet reaction center chlorophyll, 3P680, a potentially dangerous species to its protein surroundings in the presence of oxygen [Vass, I., Styring, S., Hundal, T., Koivuniemi, A., Aro, E.-M., & Anderson, B. Oxygen 330-336 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 389-392 8476749-14 1993 In summary, these inhibitors produced enzyme-catalyzed inactivation of reconstituted aromatase activity, and radioiodinated 7 alpha-IP-TADD binds covalently to the cytochrome P450arom. alpha-ip 126-134 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 164-183 8476755-2 1993 However, the sole commercially available aromatase inhibitor, aminoglutethimide, is not very selective. Aminoglutethimide 62-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-50 1472013-1 1992 Aromatase mRNA in human skin fibroblasts was greatly increased in the presence of dexamethasone. Dexamethasone 82-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 1472013-2 1992 Aromatase cDNA was prepared depending on mRNA of dexamethasone-treated fibroblasts by the PCR method including the rapid amplification of cDNA ends (RACE) protocol. Dexamethasone 49-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 8384878-1 1993 It has recently been demonstrated that strong illumination under anaerobic conditions leads to the double reduction of the primary quinone acceptor, QA, which in turn promotes the light-induced formation of triplet reaction center chlorophyll, 3P680, a potentially dangerous species to its protein surroundings in the presence of oxygen [Vass, I., Styring, S., Hundal, T., Koivuniemi, A., Aro, E.-M., & Anderson, B. quinone 131-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 389-392 8384878-1 1993 It has recently been demonstrated that strong illumination under anaerobic conditions leads to the double reduction of the primary quinone acceptor, QA, which in turn promotes the light-induced formation of triplet reaction center chlorophyll, 3P680, a potentially dangerous species to its protein surroundings in the presence of oxygen [Vass, I., Styring, S., Hundal, T., Koivuniemi, A., Aro, E.-M., & Anderson, B. Chlorophyll 231-242 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 389-392 8476772-0 1993 Effect of androstenedione on growth of untransfected and aromatase-transfected MCF-7 cells in culture. Androstenedione 10-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 57-66 8476772-1 1993 Aromatase is present in human breast tumors and in breast cancer cell lines suggesting the possibility of in-situ estrogen production via the androstenedione to estrone and estradiol pathway. Androstenedione 142-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 8476772-1 1993 Aromatase is present in human breast tumors and in breast cancer cell lines suggesting the possibility of in-situ estrogen production via the androstenedione to estrone and estradiol pathway. Estrone 161-168 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 8476772-1 1993 Aromatase is present in human breast tumors and in breast cancer cell lines suggesting the possibility of in-situ estrogen production via the androstenedione to estrone and estradiol pathway. Estradiol 173-182 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 8476772-3 1993 Accordingly, we studied the effects of the aromatase substrate, androstenedione, on the rate of proliferation of wild-type and aromatase-transfected MCF-7 breast cancer cells. Androstenedione 64-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 127-136 8476772-5 1993 In contrast, aromatase-transfected cells contained higher amounts of aromatase, produced predominantly estradiol, and responded to androstenedione with enhanced growth. Estradiol 103-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 8476772-5 1993 In contrast, aromatase-transfected cells contained higher amounts of aromatase, produced predominantly estradiol, and responded to androstenedione with enhanced growth. Androstenedione 131-146 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 8476772-6 1993 An aromatase inhibitor fadrozole hydrochloride, blocked the proliferative effects of androstenedione providing evidence for the role of aromatase in this process. Fadrozole 23-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 3-12 8476772-6 1993 An aromatase inhibitor fadrozole hydrochloride, blocked the proliferative effects of androstenedione providing evidence for the role of aromatase in this process. Fadrozole 23-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-145 8476772-6 1993 An aromatase inhibitor fadrozole hydrochloride, blocked the proliferative effects of androstenedione providing evidence for the role of aromatase in this process. Androstenedione 85-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 3-12 8476772-6 1993 An aromatase inhibitor fadrozole hydrochloride, blocked the proliferative effects of androstenedione providing evidence for the role of aromatase in this process. Androstenedione 85-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 136-145 8476772-7 1993 As further evidence of the requirement for aromatase, cells transfected with the neomycin resistance expression plasmid but lacking the aromatase cDNA did not respond to androstenedione. Neomycin 81-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-52 8476775-1 1993 Aromatase plays a crucial role in the mechanism of action of testosterone in the central nervous system. Testosterone 61-73 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 8476775-3 1993 In the experiments described here the presence of aromatase (assayed by the tritiated water method) has been evaluated in the two main cellular components of the brain: neurons and glia. Water 86-91 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-59 8476778-11 1993 Aminoglutethimide inhibited aromatase activity of benign and malignant ovarian tumors, uterine myoma, choriocarcinoma cells and purified human placental P-450arom in a similar manner. Aminoglutethimide 0-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 153-162 8382517-3 1993 The present study demonstrates that the main mammalian lignan enterolactone (trans-2,3-bis[(3-hydroxyphenyl)methyl]-butyrolactone) and some other diphenols are moderate or weak inhibitors of human estrogen synthetase (aromatase) and that this lignan binds to or near the substrate region of the active site of the P-450 enzyme. lignan enterolactone 55-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 197-216 8382517-3 1993 The present study demonstrates that the main mammalian lignan enterolactone (trans-2,3-bis[(3-hydroxyphenyl)methyl]-butyrolactone) and some other diphenols are moderate or weak inhibitors of human estrogen synthetase (aromatase) and that this lignan binds to or near the substrate region of the active site of the P-450 enzyme. trans-2,3-bis[(3-hydroxyphenyl)methyl]-butyrolactone 77-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 197-216 8382517-3 1993 The present study demonstrates that the main mammalian lignan enterolactone (trans-2,3-bis[(3-hydroxyphenyl)methyl]-butyrolactone) and some other diphenols are moderate or weak inhibitors of human estrogen synthetase (aromatase) and that this lignan binds to or near the substrate region of the active site of the P-450 enzyme. 4,4'-dihydroxybiphenyl 146-155 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 197-216 8382517-3 1993 The present study demonstrates that the main mammalian lignan enterolactone (trans-2,3-bis[(3-hydroxyphenyl)methyl]-butyrolactone) and some other diphenols are moderate or weak inhibitors of human estrogen synthetase (aromatase) and that this lignan binds to or near the substrate region of the active site of the P-450 enzyme. Lignans 55-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 197-216 1394219-0 1992 Phase I and endocrine study of exemestane (FCE 24304), a new aromatase inhibitor, in postmenopausal women. exemestane 31-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-70 1394219-0 1992 Phase I and endocrine study of exemestane (FCE 24304), a new aromatase inhibitor, in postmenopausal women. FCE 43-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-70 1455454-0 1992 Aromatase and testosterone fatty acid esters: the search for a cryptic biosynthetic pathway to estradiol esters. estradiol esters 95-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 1455454-4 1992 This led us to determine whether a cryptic aromatase pathway exists in which testosterone esters could be converted directly into LE2. testosterone esters 77-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-52 1455454-4 1992 This led us to determine whether a cryptic aromatase pathway exists in which testosterone esters could be converted directly into LE2. LE(2) 130-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-52 1455454-5 1992 We tested a representative fatty acid ester, testosterone stearate, both as an inhibitor and as a substrate for the aromatase enzyme from human placental microsomes. Fatty Acids 27-43 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-125 1455454-5 1992 We tested a representative fatty acid ester, testosterone stearate, both as an inhibitor and as a substrate for the aromatase enzyme from human placental microsomes. testosterone stearate 45-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-125 1455454-11 1992 Thus, we conclude that an aromatase pathway involving testosterone esters does not exist and that the sole source of LE2 is through direct esterification of estradiol. testosterone esters 54-73 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-35 1613432-1 1992 Aromatase cytochrome P-450 (P-450AROM) is the enzyme in the steroidogenic pathway controlling the formation of oestrogens from 19 carbon steroid precursors. carbon steroid 130-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 21-26 1339246-7 1992 Maximal P450 Arom mRNA expression was associated with less cAMP generation than for P450 SCC. Cyclic AMP 59-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 8-17 1339246-9 1992 These results indicate that: 1) FSH and LH have markedly different effects on mature granulosa cell function; 2) the differential action of FSH and LH are dose dependently mediated through intracellular cAMP; 3) mRNA expression of P450 SCC and P450 Arom are differentially regulated by gonadotrophins and intracellular cAMP; and 4) LH was the most potent inducer of its own receptor mRNA. Cyclic AMP 203-207 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 244-253 1592866-2 1992 Peripheral conversion of C19 steroids to estrogens is catalyzed by the aromatase enzyme complex which is comprised of a specific form of cytochrome P450, aromatase cytochrome P450 (P450AROM) and the flavoprotein, NADPH-cytochrome P450 reductase. Steroids 29-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 181-189 1637664-0 1992 Aromatase inhibition: 4-hydroxyandrostenedione (4-OHA, CGP 32349) in advanced prostatic cancer. formestane 22-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 1637664-1 1992 We report the use of the steroidal aromatase inhibitor, 4-hydroxyandrostenedione (4-OHA, CGP 32349), in the management of patients with advanced, hormone resistant, prostatic cancer. formestane 56-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 35-44 1637664-1 1992 We report the use of the steroidal aromatase inhibitor, 4-hydroxyandrostenedione (4-OHA, CGP 32349), in the management of patients with advanced, hormone resistant, prostatic cancer. formestane 82-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 35-44 1613432-1 1992 Aromatase cytochrome P-450 (P-450AROM) is the enzyme in the steroidogenic pathway controlling the formation of oestrogens from 19 carbon steroid precursors. carbon steroid 130-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-37 1613432-13 1992 Bu2cAMP increased mRNA levels of P-450AROM by 2.5-fold, while TGF beta inhibited this induction by 35%. bu2camp 0-7 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-42 1740498-1 1992 Aromatase cytochrome P-450 (cytochrome P-450AROM) catalyzes the formation of aromatic C18 estrogenic steroids from C19 androgens. 1-octadecene 86-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-48 1740498-1 1992 Aromatase cytochrome P-450 (cytochrome P-450AROM) catalyzes the formation of aromatic C18 estrogenic steroids from C19 androgens. Steroids 101-109 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-48 1740498-2 1992 Several studies indicate that cytochrome P-450AROM activity is induced by glucocorticoids such as dexamethasone (DEX) and superinduced by DEX plus cycloheximide (CHX). Dexamethasone 98-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-50 1740498-2 1992 Several studies indicate that cytochrome P-450AROM activity is induced by glucocorticoids such as dexamethasone (DEX) and superinduced by DEX plus cycloheximide (CHX). Dexamethasone 113-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-50 1740498-2 1992 Several studies indicate that cytochrome P-450AROM activity is induced by glucocorticoids such as dexamethasone (DEX) and superinduced by DEX plus cycloheximide (CHX). Dexamethasone 138-141 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-50 1740498-2 1992 Several studies indicate that cytochrome P-450AROM activity is induced by glucocorticoids such as dexamethasone (DEX) and superinduced by DEX plus cycloheximide (CHX). Cycloheximide 147-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-50 1740498-2 1992 Several studies indicate that cytochrome P-450AROM activity is induced by glucocorticoids such as dexamethasone (DEX) and superinduced by DEX plus cycloheximide (CHX). Cycloheximide 162-165 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-50 1740498-5 1992 Incubation of cells with DEX plus CHX resulted in a further increase in levels of cytochrome P-450AROM messenger RNA (mRNA) when compared to cells treated with DEX alone, suggesting that inhibition of protein synthesis superinduces transcription of the cytochrome P-450AROM gene. Dexamethasone 25-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-102 1740498-5 1992 Incubation of cells with DEX plus CHX resulted in a further increase in levels of cytochrome P-450AROM messenger RNA (mRNA) when compared to cells treated with DEX alone, suggesting that inhibition of protein synthesis superinduces transcription of the cytochrome P-450AROM gene. Dexamethasone 25-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-102 1740498-5 1992 Incubation of cells with DEX plus CHX resulted in a further increase in levels of cytochrome P-450AROM messenger RNA (mRNA) when compared to cells treated with DEX alone, suggesting that inhibition of protein synthesis superinduces transcription of the cytochrome P-450AROM gene. Cycloheximide 34-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-102 1740498-5 1992 Incubation of cells with DEX plus CHX resulted in a further increase in levels of cytochrome P-450AROM messenger RNA (mRNA) when compared to cells treated with DEX alone, suggesting that inhibition of protein synthesis superinduces transcription of the cytochrome P-450AROM gene. Cycloheximide 34-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-102 1730667-1 1992 Aromatase, a cytochrome P-450, catalyzes the formation of aromatic C18 estrogenic steroids from C19 androgens. Steroids 82-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 1314086-7 1992 It is tentatively concluded that breast tumour aromatase activity is more important for the maintenance of tumour oestradiol levels than aromatase in breast fatty tissue. Estradiol 114-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 1734938-6 1992 The 7-substituted 4,6-androstadiene-3,17-diones produced dose dependent inhibition of aromatase activity in the cell cultures, with IC50 values ranging from 490 nM to 4.5 microM. 7-substituted 4,6-androstadiene-3,17-diones 4-47 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-95 1734938-0 1992 Aromatase inhibition by 7-substituted steroids in human choriocarcinoma cell culture. 7-substituted steroids 24-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 1309352-3 1992 With the use of oligonucleotide probes designed according to known complementary DNA sequences, hybridizable mRNA transcripts of 3.0, 2.4, and 1.6 kilobases for P-450AROM were found in Northern blot analysis of JEG-3 cell RNA. Oligonucleotides 16-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 161-170 1309352-5 1992 Time-dependent increases in P-450AROM mRNA levels in JEG-3 cells were observed for both CT and TPA with maximal effects at 24-48 h. CT and TPA increased P-450AROM mRNA levels in a concentration-dependent manner. Tetradecanoylphorbol Acetate 95-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-37 1309352-5 1992 Time-dependent increases in P-450AROM mRNA levels in JEG-3 cells were observed for both CT and TPA with maximal effects at 24-48 h. CT and TPA increased P-450AROM mRNA levels in a concentration-dependent manner. Tetradecanoylphorbol Acetate 95-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 153-162 1309352-5 1992 Time-dependent increases in P-450AROM mRNA levels in JEG-3 cells were observed for both CT and TPA with maximal effects at 24-48 h. CT and TPA increased P-450AROM mRNA levels in a concentration-dependent manner. Tetradecanoylphorbol Acetate 139-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-37 1309352-5 1992 Time-dependent increases in P-450AROM mRNA levels in JEG-3 cells were observed for both CT and TPA with maximal effects at 24-48 h. CT and TPA increased P-450AROM mRNA levels in a concentration-dependent manner. Tetradecanoylphorbol Acetate 139-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 153-162 1309352-10 1992 Inhibition of protein synthesis by cycloheximide decreased basal, CT and TPA stimulated P-450AROM mRNA levels but increased the expression of 17 beta-HSD mRNA. Cycloheximide 35-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 1309352-10 1992 Inhibition of protein synthesis by cycloheximide decreased basal, CT and TPA stimulated P-450AROM mRNA levels but increased the expression of 17 beta-HSD mRNA. Tetradecanoylphorbol Acetate 73-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 1309352-12 1992 The present results suggest that: 1) induction of P-450AROM mRNA may at least partly be responsible for our previously reported increases in the rate of conversion of androgens to estrogens by CT and TPA in JEG-3 cells; 2) 17 beta-HSD mRNA expression is mainly controlled through a cAMP-dependent mechanism in contrast to the multifactorial control of P-450AROM mRNA; and 3) protein synthesis inhibition by cycloheximide has opposite effects on the mRNA levels of these two key enzymes in placental estrogen metabolism. Tetradecanoylphorbol Acetate 200-203 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-59 1309352-12 1992 The present results suggest that: 1) induction of P-450AROM mRNA may at least partly be responsible for our previously reported increases in the rate of conversion of androgens to estrogens by CT and TPA in JEG-3 cells; 2) 17 beta-HSD mRNA expression is mainly controlled through a cAMP-dependent mechanism in contrast to the multifactorial control of P-450AROM mRNA; and 3) protein synthesis inhibition by cycloheximide has opposite effects on the mRNA levels of these two key enzymes in placental estrogen metabolism. Cyclic AMP 282-286 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-59 1309352-12 1992 The present results suggest that: 1) induction of P-450AROM mRNA may at least partly be responsible for our previously reported increases in the rate of conversion of androgens to estrogens by CT and TPA in JEG-3 cells; 2) 17 beta-HSD mRNA expression is mainly controlled through a cAMP-dependent mechanism in contrast to the multifactorial control of P-450AROM mRNA; and 3) protein synthesis inhibition by cycloheximide has opposite effects on the mRNA levels of these two key enzymes in placental estrogen metabolism. Cycloheximide 407-420 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-59 1734938-1 1992 Androstenedione analogs containing 7 alpha-substituents have proven to be potent inhibitors of aromatase both in vitro and in vivo. Androstenedione 0-15 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-104 1734938-3 1992 In order to examine further the interaction(s) of 7-substituted steroids with aromatase, 7-substituted 4,6-androstadiene-3,17-diones were synthesized and demonstrated competitive inhibition of aromatase activity in human placental microsomes. 7-substituted steroids 50-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-87 1734938-3 1992 In order to examine further the interaction(s) of 7-substituted steroids with aromatase, 7-substituted 4,6-androstadiene-3,17-diones were synthesized and demonstrated competitive inhibition of aromatase activity in human placental microsomes. 7-substituted steroids 50-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 193-202 1734938-3 1992 In order to examine further the interaction(s) of 7-substituted steroids with aromatase, 7-substituted 4,6-androstadiene-3,17-diones were synthesized and demonstrated competitive inhibition of aromatase activity in human placental microsomes. 7-substituted 4,6-androstadiene-3,17-diones 89-132 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 78-87 1734938-3 1992 In order to examine further the interaction(s) of 7-substituted steroids with aromatase, 7-substituted 4,6-androstadiene-3,17-diones were synthesized and demonstrated competitive inhibition of aromatase activity in human placental microsomes. 7-substituted 4,6-androstadiene-3,17-diones 89-132 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 193-202 1734938-4 1992 7-Substituted 1,4,6-androstatriene-3,17-diones demonstrated mechanism-based inhibition of placental aromatase activity. 7-substituted 1,4,6-androstatriene-3,17-diones 0-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-109 1734938-8 1992 These results on the 7-substituted 4,6-androstadiene-3,17-diones are consistent with the data from biochemical enzyme inhibition studies using human placental aromatase. 7-substituted 4,6-androstadiene-3,17-diones 21-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 159-168 1734938-10 1992 Other mechanism-based inhibitors, 7 alpha-(4"-amino)phenylthio-1,4-androstadiene-3,17-dione and 4-hydroxyandrostenedione, also exhibited potent inhibition of aromatase activity in JAr cells. 7-((4'-aminophenyl)thio)-1,4-androstadiene-3,17-dione 34-91 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 158-167 1734938-10 1992 Other mechanism-based inhibitors, 7 alpha-(4"-amino)phenylthio-1,4-androstadiene-3,17-dione and 4-hydroxyandrostenedione, also exhibited potent inhibition of aromatase activity in JAr cells. formestane 96-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 158-167 1724289-1 1991 The formation of estrogens from C19 steroids is catalyzed by a specific form of cytochrome P450, aromatase cytochrome P450 (P450AROM; the product of the CYP19 gene). Steroids 36-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-95 1724289-1 1991 The formation of estrogens from C19 steroids is catalyzed by a specific form of cytochrome P450, aromatase cytochrome P450 (P450AROM; the product of the CYP19 gene). Steroids 36-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 118-122 1724289-1 1991 The formation of estrogens from C19 steroids is catalyzed by a specific form of cytochrome P450, aromatase cytochrome P450 (P450AROM; the product of the CYP19 gene). Steroids 36-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 124-132 1724289-1 1991 The formation of estrogens from C19 steroids is catalyzed by a specific form of cytochrome P450, aromatase cytochrome P450 (P450AROM; the product of the CYP19 gene). Steroids 36-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 153-158 2011509-0 1991 Tetranucleotide repeat polymorphism at the human aromatase cytochrome P-450 gene (CYP19). tetranucleotide 0-15 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-87 2050688-2 1991 As a first step toward investigation of the structure-function relationships of cytochrome P-450AROM, we have used computer modeling to align the amino acid sequence of cytochrome P-450AROM with that of cytochrome P-450CAM from Pseudomonas putida and thus create a substrate pocket using the heme-binding region and the I-helix of cytochrome P-450CAM as the template. Heme 292-296 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 80-100 2050688-2 1991 As a first step toward investigation of the structure-function relationships of cytochrome P-450AROM, we have used computer modeling to align the amino acid sequence of cytochrome P-450AROM with that of cytochrome P-450CAM from Pseudomonas putida and thus create a substrate pocket using the heme-binding region and the I-helix of cytochrome P-450CAM as the template. Heme 292-296 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-189 2050688-11 1991 These results support the role of a carboxylic acid residue at position 302 in the catalytic activity of cytochrome P-450AROM. Carboxylic Acids 36-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 105-125 2004042-0 1991 Effect of ketoconazole on human ovarian C17,20-desmolase and aromatase. Ketoconazole 10-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 40-70 2004042-9 1991 In conclusion, ketoconazole was shown to inhibit human ovarian C17,20-desmolase and aromatase in vitro. Ketoconazole 15-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-93 2031856-3 1991 Under these conditions, in vitro JEG-3 aromatase was inhibited by R 76 713 with IC50-values of 7.6 +/- 0.5 nM and 2.7 +/- 1.1 nM using 500 nM of androstenedione and testosterone as substrate respectively. Androstenedione 145-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-48 2031856-3 1991 Under these conditions, in vitro JEG-3 aromatase was inhibited by R 76 713 with IC50-values of 7.6 +/- 0.5 nM and 2.7 +/- 1.1 nM using 500 nM of androstenedione and testosterone as substrate respectively. Testosterone 165-177 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-48 2031856-4 1991 The Km-value of the aromatase enzyme with androstenedione as substrate was 62 +/- 19 nM; with testosterone as substrate, a value of 166 +/- 27 nM was found. Androstenedione 42-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-29 2031856-4 1991 The Km-value of the aromatase enzyme with androstenedione as substrate was 62 +/- 19 nM; with testosterone as substrate, a value of 166 +/- 27 nM was found. Testosterone 94-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-29 1988941-1 1991 Aromatase, a cytochrome P450, catalyzes the formation of aromatic C-18 estrogenic steroids from C-19 androgens. Steroids 82-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 1988941-7 1991 These results, along with those obtained from inhibition studies with aromatase inhibitors, 4-hydroxyandrostenedione and aminoglutethimide, suggest that Pro-308 is probably situated in the active site of the enzyme and may be interacting with the D ring of the steroids. Steroids 261-269 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 70-79 2285580-7 1990 We attempted to measure aromatase activity in tissue incubation by quantitating 3H2O released during aromatization of androstenedione or testosterone labeled at the C-1 position. 3h2o 80-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-33 1958529-4 1991 The most potent of these, 4-hydroxyandrostenedione (4-OHA) inhibits aromatase competitively but also causes inactivation of the enzyme. formestane 26-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-77 1958529-4 1991 The most potent of these, 4-hydroxyandrostenedione (4-OHA) inhibits aromatase competitively but also causes inactivation of the enzyme. formestane 52-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-77 2285580-7 1990 We attempted to measure aromatase activity in tissue incubation by quantitating 3H2O released during aromatization of androstenedione or testosterone labeled at the C-1 position. Androstenedione 118-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-33 2285580-7 1990 We attempted to measure aromatase activity in tissue incubation by quantitating 3H2O released during aromatization of androstenedione or testosterone labeled at the C-1 position. Testosterone 137-149 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-33 2201397-1 1990 We report the results of the first use of a steroidal aromatase inhibitor, 4-hydroxyandrostenedione (4-OHA, CGP 32349), in the palliation of patients with advanced, hormone resistant, prostatic cancer. formestane 75-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 54-63 1965238-4 1990 Aromatase activity was determined by measuring the tritiated water (3H2O) in the medium after incubating stromal cells with [1 beta-3H]androstenedione. Water 61-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 1965238-4 1990 Aromatase activity was determined by measuring the tritiated water (3H2O) in the medium after incubating stromal cells with [1 beta-3H]androstenedione. 3h2o 68-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 1965238-4 1990 Aromatase activity was determined by measuring the tritiated water (3H2O) in the medium after incubating stromal cells with [1 beta-3H]androstenedione. 1 beta-3h]androstenedione 125-150 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 2268557-0 1990 Aromatase inhibition by flavonoids. Flavonoids 24-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 2201397-1 1990 We report the results of the first use of a steroidal aromatase inhibitor, 4-hydroxyandrostenedione (4-OHA, CGP 32349), in the palliation of patients with advanced, hormone resistant, prostatic cancer. formestane 101-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 54-63 2201397-1 1990 We report the results of the first use of a steroidal aromatase inhibitor, 4-hydroxyandrostenedione (4-OHA, CGP 32349), in the palliation of patients with advanced, hormone resistant, prostatic cancer. cgp 108-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 54-63 34933195-5 2022 Activation of NR1D1 enhances its ability to inhibit the transcriptional activity of CYP19A1 by binding to the RORE on the CYP19A1 promoter, resulting in a decrease in estradiol content. Estradiol 167-176 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-91 2391958-1 1990 To procure an affinity gel capable of purifying antibody against the cytochrome P450 component of estrogen synthetase (P450ES), we coupled purified P450ES to agarose supports. Sepharose 158-165 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-117 2363692-0 1990 Conversion of 19-oxo[2 beta-2H]androgens into oestrogens by human placental aromatase. 19-oxo[2 beta-2h 14-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-85 2180973-1 1990 The levels of expression of mRNA species encoding cholesterol side-chain cleavage cytochrome P-450 (P450scc), 17 alpha-hydroxylase cytochrome P450 (P450(17 alpha], aromatase cytochrome P-450 (P-450AROM), and 3 beta-hydroxysteroid dehydrogenase (3 beta HSD) were examined in human follicles and corpora lutea (CL) throughout the menstrual cycle. Cholesterol 50-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-98 2293555-1 1990 The effect of treatment with the aromatase inhibitor, 4-hydroxyandrostenedione (4-OHA) on the peripheral conversion of androstenedione to estrone has been examined in eight postmenopausal women with advanced breast cancer. Estrone 138-145 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-42 2308324-0 1990 Aromatase inhibition by 4-thiosubstituted-4-androstene-3,17-dione derivatives. 4-thiosubstituted-4-androstene-3,17-dione 24-65 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 2308324-1 1990 The synthesis and evaluation of 4-thiosubstituted-4-androstenedione analogs as inhibitors of estrogen synthetase (aromatase) is described. 4-thiosubstituted-4-androstenedione 32-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-112 2308324-1 1990 The synthesis and evaluation of 4-thiosubstituted-4-androstenedione analogs as inhibitors of estrogen synthetase (aromatase) is described. 4-thiosubstituted-4-androstenedione 32-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 114-123 33808393-0 2021 Celastrol Prevents Oxidative Stress Effects on FSHR, PAPP, and CYP19A1 Gene Expression in Cultured Human Granulosa-Lutein Cells. celastrol 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-70 33776924-7 2021 TLR4 activation also inhibited forskolin-induced secretion of E2 by inhibiting CYP19A1, with no effect on progesterone. Colforsin 31-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-86 33776924-7 2021 TLR4 activation also inhibited forskolin-induced secretion of E2 by inhibiting CYP19A1, with no effect on progesterone. Estradiol 62-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-86 33800928-10 2021 qRT-PCR and linear regression analysis confirmed the expression and correlation of its potential targeted gene, CYP19A1, a key gene involved in estradiol synthesis. Estradiol 144-153 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 112-119 29285776-0 2018 Association of the CYP17 MSP AI (T-34C) and CYP19 codon 39 (Trp/Arg) polymorphisms with susceptibility to acne vulgaris. Tryptophan 60-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-49 34871762-0 2022 Copper exposure disrupts ovarian steroidogenesis in human ovarian granulosa cells via the FSHR/CYP19A1 pathway and alters methylation patterns on the SF-1 gene promoter. Copper 0-6 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-102 34871762-6 2022 Moreover, copper partially reversed the FSH-induced increase in FSHR, CYP19A1 and 17-estradiol levels, and the decreased effect of the FSH receptor binding inhibitor fragment on FSHR, CYP19A1, and 17-estradiol became more apparent after adding copper. Copper 10-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 70-77 34871762-6 2022 Moreover, copper partially reversed the FSH-induced increase in FSHR, CYP19A1 and 17-estradiol levels, and the decreased effect of the FSH receptor binding inhibitor fragment on FSHR, CYP19A1, and 17-estradiol became more apparent after adding copper. Copper 10-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 184-191 2370296-12 1990 Northern analysis performed on total RNA obtained from forskolin- or hCG-stimulated granulosa-lutein cells confirmed that the increase in aromatase activity was associated with a corresponding increase in levels of mRNA specific for aromatase cytochrome P-450. Colforsin 55-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 138-147 2370296-12 1990 Northern analysis performed on total RNA obtained from forskolin- or hCG-stimulated granulosa-lutein cells confirmed that the increase in aromatase activity was associated with a corresponding increase in levels of mRNA specific for aromatase cytochrome P-450. Colforsin 55-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 233-242 2185341-0 1990 Aromatase inhibition with 4-hydroxyandrostenedione in the treatment of postmenopausal patients with advanced breast cancer: a phase II study. formestane 26-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 2185341-2 1990 Between October 1986 and March 1988, 91 postmenopausal patients with advanced breast cancer entered a phase II study performed jointly in three center to investigate the new aromatase inhibitor 4-hydroxyandrostenedione. formestane 194-218 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 174-183 2185341-11 1990 We conclude that aromatase inhibition with 4-hydroxyandrostenedione is efficacious in the treatment of postmenopausal breast cancer. formestane 43-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 2278473-1 1990 Aromatase inhibitor activity of androstenediones]. Androstenedione 32-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 2278473-3 1990 The purpose of this study is the contribution of the parachor in the QSAR of androstenediones derivatives and their inhibition of aromatase enzyme. Androstenedione 77-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-139 1690030-3 1990 The cDNA sequence encodes a polypeptide of 503 amino acids and contains--near the carboxy-terminus, a region of high homology with the putative heme-binding regions of other P-450 cytochromes. Heme 144-148 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 174-179 1690030-4 1990 COS1 cells transfected with an expression plasmid containing the cytochrome P-450AROM cDNA had the capacity to aromatize testosterone, androstenedione and 16 alpha-hydroxyandrostenedione, suggesting that a single polypeptide catalyzes all steps of the aromatization reaction using either of the three major C19-substrates. Testosterone 121-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-85 1690030-4 1990 COS1 cells transfected with an expression plasmid containing the cytochrome P-450AROM cDNA had the capacity to aromatize testosterone, androstenedione and 16 alpha-hydroxyandrostenedione, suggesting that a single polypeptide catalyzes all steps of the aromatization reaction using either of the three major C19-substrates. Androstenedione 135-150 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-85 1690030-4 1990 COS1 cells transfected with an expression plasmid containing the cytochrome P-450AROM cDNA had the capacity to aromatize testosterone, androstenedione and 16 alpha-hydroxyandrostenedione, suggesting that a single polypeptide catalyzes all steps of the aromatization reaction using either of the three major C19-substrates. alpha-hydroxyandrostenedione 158-186 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-85 2293555-0 1990 Effect of treatment with 4-hydroxyandrostenedione on the peripheral conversion of androstenedione to estrone and in vitro tumor aromatase activity in postmenopausal women with breast cancer. formestane 25-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 128-137 2293555-0 1990 Effect of treatment with 4-hydroxyandrostenedione on the peripheral conversion of androstenedione to estrone and in vitro tumor aromatase activity in postmenopausal women with breast cancer. Androstenedione 34-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 128-137 2293555-1 1990 The effect of treatment with the aromatase inhibitor, 4-hydroxyandrostenedione (4-OHA) on the peripheral conversion of androstenedione to estrone has been examined in eight postmenopausal women with advanced breast cancer. formestane 80-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-42 33232722-3 2021 A number steroids show a high level affinity (micro- and submicromole) for the enzyme-ligand complexes of the tested compounds with human CYP51, CYP19 and CYP51 of C. glabrata. Steroids 9-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 145-150 21289517-0 2011 Aromatase inhibitor exemestane has antiproliferative effects on human mesothelioma cells. exemestane 20-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 21289517-4 2011 Treatment of MM cells with exemestane, a CYP19A1 inhibitor, was assessed by cell proliferation kit, cell cycle analysis, and Western blot for caspase, poly(ADP-ribose)polymerase, Bcl-xL, and v-akt murin thymoma viral oncogene homolog (Akt). exemestane 27-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-48 21289517-5 2011 RESULTS: Biological activity of CYP19A1, already present in basal condition, was increased in MPP89 and Ist-Mes1 cells after treatment with cytokine, in all MM cells on prostaglandin-E2 treatment, and in MPP89, Ist-Mes2, and Ist-Mes1 after addition of epidermal growth factor. Dinoprostone 169-185 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-39 21289517-5 2011 RESULTS: Biological activity of CYP19A1, already present in basal condition, was increased in MPP89 and Ist-Mes1 cells after treatment with cytokine, in all MM cells on prostaglandin-E2 treatment, and in MPP89, Ist-Mes2, and Ist-Mes1 after addition of epidermal growth factor. mes2 215-219 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-39 21289517-5 2011 RESULTS: Biological activity of CYP19A1, already present in basal condition, was increased in MPP89 and Ist-Mes1 cells after treatment with cytokine, in all MM cells on prostaglandin-E2 treatment, and in MPP89, Ist-Mes2, and Ist-Mes1 after addition of epidermal growth factor. mes1 108-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-39 21289517-9 2011 CONCLUSION: These findings show that CYP19A1 is present in MM and that cell growth can be down-regulated by exemestane. exemestane 108-118 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-44 34871762-6 2022 Moreover, copper partially reversed the FSH-induced increase in FSHR, CYP19A1 and 17-estradiol levels, and the decreased effect of the FSH receptor binding inhibitor fragment on FSHR, CYP19A1, and 17-estradiol became more apparent after adding copper. Copper 244-250 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 184-191 34871762-8 2022 Overall, our results indicate that copper exposure induces steroidogenesis disorders via the FSHR/CYP19A1 pathway and changes DNA methylation on the SF-1 promoter in human ovarian granulosa cells. Copper 35-41 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-105 34933195-5 2022 Activation of NR1D1 enhances its ability to inhibit the transcriptional activity of CYP19A1 by binding to the RORE on the CYP19A1 promoter, resulting in a decrease in estradiol content. Estradiol 167-176 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-129 34688002-4 2022 Four triazole fungicides regulated eight major genes involved in estrogen synthesis (StAR, CYP11A1, 3betaHSD2, CYP17, CYP19, CYP3A4, CYP1A2 and SCP2) and stimulated the secretion of 17beta-estradiol (E2). Triazoles 5-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 118-123 34906060-9 2022 The o-tolylhydrazone 3b, exhibiting promising ARO inhibition and weak EGFR inhibition, produced a noticeable high overexpression of caspase-9 and showed pre-G1 apoptosis and cell cycle arrest at G2/M phase for MCF-7 cells and at S-phase for MDA-MB-231 cells. o-tolylhydrazone 4-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-49 34935973-8 2022 E2 production was suppressed by the CYP19 inhibitor, letrozole, and induced by CYP19 activators, bisphenol A at 100 microM, and genistein at 100 microM. bisphenol A 97-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-41 34935973-8 2022 E2 production was suppressed by the CYP19 inhibitor, letrozole, and induced by CYP19 activators, bisphenol A at 100 microM, and genistein at 100 microM. Genistein 128-137 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-41 34871401-15 2021 Polymorphisms of the aromatase gene CYP19A1, including single nucleotide polymorphisms and tetranucleotide TTTA repeats polymorphism (TTTAn), also influence hormone profiles, semen quality and treatment efficacy of AIs in male hypogonadotropic hypogonadism and infertility. tetranucleotide 91-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-43 34935973-8 2022 E2 production was suppressed by the CYP19 inhibitor, letrozole, and induced by CYP19 activators, bisphenol A at 100 microM, and genistein at 100 microM. Estradiol 0-2 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-41 34935973-8 2022 E2 production was suppressed by the CYP19 inhibitor, letrozole, and induced by CYP19 activators, bisphenol A at 100 microM, and genistein at 100 microM. Estradiol 0-2 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-84 34935973-8 2022 E2 production was suppressed by the CYP19 inhibitor, letrozole, and induced by CYP19 activators, bisphenol A at 100 microM, and genistein at 100 microM. Letrozole 53-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 36-41 34906060-10 2022 Docking results revealed that 3b, elicited binding affinities to ARO comparable to those of letrozole. Letrozole 92-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-68 34906060-12 2022 Their design offered a way for the optimization and development of apoptotic quinazolinone-based ARO and EGFR inhibitors. Quinazolinones 77-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 97-100 34554372-8 2021 CONCLUSIONS: Our findings suggest an additional and direct effect of exemestane on CYP-19 (aromatase) synthesis presumably due to effects on the CYP19 promoter use that is not present during therapy with the non-steroidal aromatase inhibitor letrozole. exemestane 69-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-89 34554372-8 2021 CONCLUSIONS: Our findings suggest an additional and direct effect of exemestane on CYP-19 (aromatase) synthesis presumably due to effects on the CYP19 promoter use that is not present during therapy with the non-steroidal aromatase inhibitor letrozole. exemestane 69-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 145-150 34688840-0 2021 Organochlorine pesticide dieldrin upregulate proximal promoter (PII) driven CYP19A1 gene expression and increases estrogen production in granulosa cells. Hydrocarbons, Chlorinated 0-14 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-83 34688840-5 2021 In the present study, the attempt has been made to elucidate the effect of dieldrin on the promoter-specific CYP19A1 gene expression and estrogen hormone production in buffalo granulosa cells. Dieldrin 75-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 109-116 34688840-0 2021 Organochlorine pesticide dieldrin upregulate proximal promoter (PII) driven CYP19A1 gene expression and increases estrogen production in granulosa cells. Dieldrin 25-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 76-83 34563259-5 2021 Studies with ovarian theca cells taken from PCOS women have demonstrated increased androgen production due to augmented ovarian steroidogenesis attributed to mainly altered expression of critical enzymes (Cytochrome P450 enzymes: CYP17, CYP21, CYP19, CYP11A) in the steroid hormone biosynthesis pathway. Steroids 266-273 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 244-249 34688840-7 2021 Results showed that dieldrin significantly increased the expression of the CYP19A1 gene after 6 and 12 h while its expression was decreased after 24 h. To understand the upregulation of CYP19A1 gene, promoters" specific CYP19A1 transcript analysis was done. Dieldrin 20-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 75-82 34688840-7 2021 Results showed that dieldrin significantly increased the expression of the CYP19A1 gene after 6 and 12 h while its expression was decreased after 24 h. To understand the upregulation of CYP19A1 gene, promoters" specific CYP19A1 transcript analysis was done. Dieldrin 20-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 186-193 34688840-12 2021 Overall, the present study demonstrated the stimulatory effect of dieldrin on the CYP19A1 gene and will help to understand the toxicological role of dieldrin on the reproductive system. Dieldrin 66-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-89 34688840-12 2021 Overall, the present study demonstrated the stimulatory effect of dieldrin on the CYP19A1 gene and will help to understand the toxicological role of dieldrin on the reproductive system. Dieldrin 149-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-89 34365057-0 2021 New thieno(3,2-d)pyrimidine-based derivatives: Design, synthesis and biological evaluation as antiproliferative agents, EGFR and ARO inhibitors inducing apoptosis in breast cancer cells. thienopyrimidine 4-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-132 34365057-5 2021 Further mechanistic evidences for their anticancer activities were provided by screening the most potent compounds against MCF-7 and/or MDA-MB-231 cells for EGFR and ARO inhibitory activities using erlotinib and letrozole as reference standards respectively. Erlotinib Hydrochloride 198-207 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 166-169 34365057-13 2021 Thus, the thienotriazolopyrimidines 11b and 12 showing good EGFR inhibition and the thieno(3,2-d)-pyrimidine derivatives 3b and 9d, eliciting the best ARO inhibition activity, can be considered as new candidates as anti-breast cancer agents that necessitate further development. thieno(3,2-d)-pyrimidine 84-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 151-154 34749236-0 2021 A low dose of bisphenol A stimulates estradiol production by regulating beta-catenin-FOXL2-CYP19A1 pathway in human ovarian granulosa cells. bis(4-hydroxyphenyl)sulfone 14-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-98 34749236-0 2021 A low dose of bisphenol A stimulates estradiol production by regulating beta-catenin-FOXL2-CYP19A1 pathway in human ovarian granulosa cells. Estradiol 37-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-98 34749236-4 2021 Furthermore, BPA enhanced CYP19A1 (aromatase) expression levels and estradiol (E2) production, but these effects were not observed in FOXL2 knockout (KO) cells. bisphenol A 13-16 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-33 34749236-7 2021 Thus, we reveal a comprehensive molecular signaling cascade encompassing BPA-CTNNB1-FOXL2-CYP19A1-E2 that contributes to the endocrine-disrupting activities of BPA in human ovarian granulosa cells. bisphenol A 160-163 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-97 34523345-3 2021 Two-thirds of the DHOPA are from the oxidation of ARO1 (lumped less-reactive aromatic species; mostly toluene), with the rest from ARO2 (lumped more-reactive aromatic species; mostly xylenes). DHOPA 18-23 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-54 34523345-3 2021 Two-thirds of the DHOPA are from the oxidation of ARO1 (lumped less-reactive aromatic species; mostly toluene), with the rest from ARO2 (lumped more-reactive aromatic species; mostly xylenes). Toluene 102-109 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-54 34404424-6 2021 RESULTS: PGE2 or rofecoxib treatment caused in MPM cells an increased or decreased, respectively, CYP19A1 expression at mRNA and protein levels. Dinoprostone 9-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-105 34455966-0 2022 In-vitro Cytotoxicity and Aromatase Inhibitory Activity of Flavonoids: Synthesis, Molecular Docking and In-silico ADME Prediction. Flavonoids 59-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-35 34455966-1 2022 BACKGROUND: Many natural and synthetic flavonoids have been studied and documented by inhibiting aromatase enzymes for their anti-cancer activity against breast carcinoma. Flavonoids 39-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 97-106 34455966-3 2022 OBJECTIVE: Hence, a series of flavonoids have been synthesized and assessed for their in vitro cytotoxicity and aromatase inhibitory activity. Flavonoids 30-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 112-121 34455966-10 2022 CONCLUSION: Therefore, it was concluded that compounds 2b and 24b had a potent inhibitory effect of aromatase compared with letrozole with an IC50 value of 0.86 muM. Letrozole 124-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 100-109 34404424-2 2021 METHODS: The interaction between COX-2 and CYP19A1 was first investigated on different MPM lines upon PGE2, and COX-2 inhibitor (rofecoxib) treatment by western blot, RT-PCR. Dinoprostone 102-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-50 34538723-2 2022 Estradiol, the major estrogen in humans, is synthesized from testosterone by the action of aromatase and exerts its effects though binding to estrogen receptors. Estradiol 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-100 34538723-2 2022 Estradiol, the major estrogen in humans, is synthesized from testosterone by the action of aromatase and exerts its effects though binding to estrogen receptors. Testosterone 61-73 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-100 34733654-11 2021 The result of the GSEA showed that different outcomes in female and male patients with NMIBC were related to the interaction of CYP19A1 and the cell-cycle-related pathways. gsea 18-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 128-135 34404424-2 2021 METHODS: The interaction between COX-2 and CYP19A1 was first investigated on different MPM lines upon PGE2, and COX-2 inhibitor (rofecoxib) treatment by western blot, RT-PCR. rofecoxib 129-138 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-50 34404424-6 2021 RESULTS: PGE2 or rofecoxib treatment caused in MPM cells an increased or decreased, respectively, CYP19A1 expression at mRNA and protein levels. rofecoxib 17-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-105 34404424-10 2021 The antagonistic effect of rofecoxib and exemestane combination found only in one cell line, was reverted by pretreatment with MK2206, a pAKT inhibitor, indicating pAKT as an actionable mediator in the COX-2-CYP19A1 axis. rofecoxib 27-36 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 208-215 34404424-10 2021 The antagonistic effect of rofecoxib and exemestane combination found only in one cell line, was reverted by pretreatment with MK2206, a pAKT inhibitor, indicating pAKT as an actionable mediator in the COX-2-CYP19A1 axis. exemestane 41-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 208-215 34404424-10 2021 The antagonistic effect of rofecoxib and exemestane combination found only in one cell line, was reverted by pretreatment with MK2206, a pAKT inhibitor, indicating pAKT as an actionable mediator in the COX-2-CYP19A1 axis. MK 2206 127-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 208-215 34096286-3 2021 We found that isorhamnetin promoted the secretion of estrogen and inhibited the secretion of progesterone and testosterone by modulating steroidogenesis-associated proteins and mRNA such as CYP19A1, StAR, and 3beta-HSD in ovarian GCs. 3-methylquercetin 14-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 190-197 34374964-9 2022 The expression of genes and proteins involved in cholesterol transport (ABCA1, LDLR, and SCARB1) and estradiol production (SULT2B1 and CYP19A1) was significantly higher in CCs, and the expression of genes and proteins involved in antiapoptosis (CRLS1, LPCAT3, and PLA2G4A) was significantly higher in MGCs. Estradiol 101-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-142 34096286-3 2021 We found that isorhamnetin promoted the secretion of estrogen and inhibited the secretion of progesterone and testosterone by modulating steroidogenesis-associated proteins and mRNA such as CYP19A1, StAR, and 3beta-HSD in ovarian GCs. Progesterone 93-105 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 190-197 34096286-3 2021 We found that isorhamnetin promoted the secretion of estrogen and inhibited the secretion of progesterone and testosterone by modulating steroidogenesis-associated proteins and mRNA such as CYP19A1, StAR, and 3beta-HSD in ovarian GCs. Testosterone 110-122 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 190-197 34095690-0 2021 19-hydroxy Steroids in the Aromatase Reaction: Review on Expression and Potential Functions. 19-hydroxy steroids 0-19 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-36 34239360-0 2021 High ovarian GDF-8 levels contribute to elevated estradiol production in ovarian hyperstimulation syndrome by stimulating aromatase expression. Estradiol 49-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-131 34239360-4 2021 Aromatase, encoded by the CYP19A1 gene, is the enzyme that catalyzes the final step in estradiol (E2) biosynthesis. Estradiol 87-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 34239360-4 2021 Aromatase, encoded by the CYP19A1 gene, is the enzyme that catalyzes the final step in estradiol (E2) biosynthesis. Estradiol 87-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-33 34239360-4 2021 Aromatase, encoded by the CYP19A1 gene, is the enzyme that catalyzes the final step in estradiol (E2) biosynthesis. Estradiol 98-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 34239360-4 2021 Aromatase, encoded by the CYP19A1 gene, is the enzyme that catalyzes the final step in estradiol (E2) biosynthesis. Estradiol 98-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-33 34239360-12 2021 Blocking the function of ALK5 by the administration of its inhibitor, SB431542, alleviates OHSS symptoms and the upregulation of aromatase. 4-(5-benzo(1,3)dioxol-5-yl-4-pyridin-2-yl-1H-imidazol-2-yl)benzamide 70-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-138 34095690-2 2021 Here, we review an irreversible aromatase reaction from the substrate androstenedione. Androstenedione 70-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-41 34095690-8 2021 We focus primarily on 19-hydroxy steroids, and in particular on the 19-hydroxyandrostenedione produced by the incomplete aromatase reaction. 19-hydroxy-4-androstene-3,17-dione 68-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 121-130 34095690-9 2021 Using a PubMed database and the search term "aromatase reaction," 19-hydroxylation of androgens and steroid measurements, we detail the chemistry of the aromatase reaction and list previous and current methods used to measure 19-hydroxy steroids. Steroids 100-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 153-162 34095690-9 2021 Using a PubMed database and the search term "aromatase reaction," 19-hydroxylation of androgens and steroid measurements, we detail the chemistry of the aromatase reaction and list previous and current methods used to measure 19-hydroxy steroids. Steroids 237-245 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-54 35625301-6 2022 Docking studies and molecular dynamics simulations indicated that these three isoflavones, generated via biotransformation, are potent inhibitors of the target protein aromatase (CYP19A1); consequently, they can be used to prevent breast cancer risk in postmenopausal women. Isoflavones 78-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 179-186 35300988-4 2022 In our study, PITX2 significantly facilitated the secretion level of estrogen and progesterone through increasing CYP11A1, CYP19A1, and STAR mRNA and protein expressions in GCs. Progesterone 82-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 123-130 35621695-3 2022 Competitive inhibition experiments in vitro and molecular docking showed that BIBD-069 and BIBD-071 have high affinity for aromatase. bibd-069 78-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 123-132 35621695-3 2022 Competitive inhibition experiments in vitro and molecular docking showed that BIBD-069 and BIBD-071 have high affinity for aromatase. bibd-071 91-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 123-132 35621695-5 2022 Biodistribution and in vivo inhibition experiments confirmed that (18F)BIBD-069 and (18F)BIBD-071 specifically bind to aromatase. bibd-069 71-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 119-128 35621695-5 2022 Biodistribution and in vivo inhibition experiments confirmed that (18F)BIBD-069 and (18F)BIBD-071 specifically bind to aromatase. bibd-071 89-97 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 119-128 35176449-0 2022 Exploration of structural requirements for azole chemicals towards human aromatase CYP19A1 activity: Classification modeling, structure-activity relationships and read-across study. Azoles 43-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-90 35176449-4 2022 A toxicological evaluation of commonly used azole-based drugs and agrochemicals with respect to CYP19A1is currently requested by the European Union- Registration, Evaluation, Authorization and Restriction of Chemicals (EU-REACH) regulations due to their potential as endocrine disruptors. Azoles 44-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 96-103 35199272-6 2022 In this review, we analyzed certain researched effects of BPA, while focusing on its ability to alter the expression of various significant genes like GnRH, AdipoQ, ESR1, StAR, CYP11A1, CYP19A1, and many more involved in the pathways and endocrine regulation, whose disruption is commonly associated with the clinical manifestations of PCOS. bisphenol A 58-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 186-193 35538794-4 2022 The IC50 concentration of SIRT1 activator (SRT1720), SIRT1 inhibitor (EX527) and aromatase inhibitors (letrozole and fadrozole) was determined by XTT method. Letrozole 103-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-90 35538794-4 2022 The IC50 concentration of SIRT1 activator (SRT1720), SIRT1 inhibitor (EX527) and aromatase inhibitors (letrozole and fadrozole) was determined by XTT method. Fadrozole 117-126 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-90 35511873-10 2022 RESULTS: VAT aromatase activity was positively associated with VAT adipocyte hypertrophy (p-valueadj < 0.01) and negatively with plasma HDL-cholesterol (p-valueadj < 0.01), while SAT aromatase activity predicted dyslipidemia in women even after adjustment for waist circumference, age and hormonal contraceptive use. Cholesterol 140-151 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-22 35457011-4 2022 Recently, it has been demonstrated that tadalafil may display novel actions on androgen receptor (AR) expression and activity and cytochrome P19a1 (Cyp19a1) and estrogen receptor beta (ERbeta) expression in different in vitro systems, such as adipose, bone and prostate cancer cells, where it can act as a selective modulator of steroid hormone production. Tadalafil 40-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 130-146 35473535-4 2022 METHODS: The anti-proliferative effects of aromatase inhibitors were evaluated by MTT assay. monooxyethylene trimethylolpropane tristearate 82-85 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-52 35473535-15 2022 CONCLUSION: Aromatase inhibition by exemestane or curcumin had significantly inhibited the growth of lung cancer cell lines, synergized with cisplatin, raloxifene and celecoxib, suppressed lung cancer cell migratory potential, induced apoptosis, and reduced colony formation of lung cancer cells. exemestane 36-46 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-21 35473535-15 2022 CONCLUSION: Aromatase inhibition by exemestane or curcumin had significantly inhibited the growth of lung cancer cell lines, synergized with cisplatin, raloxifene and celecoxib, suppressed lung cancer cell migratory potential, induced apoptosis, and reduced colony formation of lung cancer cells. Curcumin 50-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-21 35473535-15 2022 CONCLUSION: Aromatase inhibition by exemestane or curcumin had significantly inhibited the growth of lung cancer cell lines, synergized with cisplatin, raloxifene and celecoxib, suppressed lung cancer cell migratory potential, induced apoptosis, and reduced colony formation of lung cancer cells. Cisplatin 141-150 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-21 35473535-15 2022 CONCLUSION: Aromatase inhibition by exemestane or curcumin had significantly inhibited the growth of lung cancer cell lines, synergized with cisplatin, raloxifene and celecoxib, suppressed lung cancer cell migratory potential, induced apoptosis, and reduced colony formation of lung cancer cells. Raloxifene Hydrochloride 152-162 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-21 35473535-15 2022 CONCLUSION: Aromatase inhibition by exemestane or curcumin had significantly inhibited the growth of lung cancer cell lines, synergized with cisplatin, raloxifene and celecoxib, suppressed lung cancer cell migratory potential, induced apoptosis, and reduced colony formation of lung cancer cells. Celecoxib 167-176 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-21 35457011-4 2022 Recently, it has been demonstrated that tadalafil may display novel actions on androgen receptor (AR) expression and activity and cytochrome P19a1 (Cyp19a1) and estrogen receptor beta (ERbeta) expression in different in vitro systems, such as adipose, bone and prostate cancer cells, where it can act as a selective modulator of steroid hormone production. Tadalafil 40-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 148-155 35424860-11 2022 Methyl gallate linked with the EGFR receptor through hydrogen bonding with a pose score of -4.5287 kcal mol-1 and RMSD value of 1.69 A. Clitorin showed the strongest interaction with aromatase (CYP19A1) for the breast cancer receptor with a posing score of -14.2074 and RMSD value of 1.56 A. methyl gallate 0-14 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 194-201 35373882-5 2022 The observed chemoselectivity for Ar - OAc versus ArO - Ac bond cleavage was rationalized based on mechanistic experiments and DFT calculations. Argon 34-36 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-53 35373882-5 2022 The observed chemoselectivity for Ar - OAc versus ArO - Ac bond cleavage was rationalized based on mechanistic experiments and DFT calculations. Actinium 56-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 50-53 35456610-7 2022 Several SNPs located in genes FDPS and FNTA were associated with the bisphosphonate-induced changes in hip bone mineral density (BMD), whereas polymorphisms of the PDSS1, CYP19A1, CYP1A1, and CYP1A2 genes were associated with SERM-induced changes in spine BMD. Diphosphonates 69-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 171-178 35175101-1 2022 This study aimed to clarify 1) the influence of genetic polymorphisms in the cytochrome P450 aromatase gene (CYP19A1) on circulating estradiol levels in men and 2) whether estrogen-related genetic polymorphisms, such as the CYP19A1 rs936306 and estrogen receptor-alpha (ESR1) rs2234693 polymorphisms, predict exercise-induced muscle damage. Estradiol 133-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 109-116 35175101-1 2022 This study aimed to clarify 1) the influence of genetic polymorphisms in the cytochrome P450 aromatase gene (CYP19A1) on circulating estradiol levels in men and 2) whether estrogen-related genetic polymorphisms, such as the CYP19A1 rs936306 and estrogen receptor-alpha (ESR1) rs2234693 polymorphisms, predict exercise-induced muscle damage. Estradiol 133-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 224-231 35175101-5 2022 Male subjects with the TT genotype of the CYP19A1 polymorphism exhibited significantly higher serum estradiol levels than the C allele carriers. Estradiol 100-109 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-49 35175101-9 2022 The results of this study suggest that genetic polymorphisms in CYP19A1 rs936306 influence serum estradiol levels in men, and genetic polymorphisms in CYP19A1 and ESR1 are associated with serum CK activity in men. Estradiol 97-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-71 35449546-4 2022 Methods/Design: The ACO/ARO/AIO-21 is an investigator-driven, prospective, open-labeled phase I drug-repurposing trial assessing the maximum tolerated dose (MTD) of capecitabine administered concurrently to standard preoperative radiotherapy (45 Gy in 25 fractions followed by 9 Gy boost in 5 fractions) in combination with fixed doses of the IL1-RA anakinra (100 mg, days -10 to 30). Capecitabine 165-177 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-27 35449546-10 2022 Discussion: Based on extensive preclinical research, the ACO/ARO/AIO-21 phase I trial will assess whether the IL-1RA anakinra can be safely combined with fluoropyrimidine-based CRT in rectal cancer. 2-fluoropyrimidine 154-170 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-64 35424731-3 2022 This effect was explained as dependent on the ratio (ArO-)/(ArOH) and for diluted ArOH corresponds to an increased contribution of much faster electron transfer (ET, ArO-/dpph ) over the Hydrogen Atom Transfer (HAT, ArOH/dpph ). aroh 60-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 166-169 35424731-3 2022 This effect was explained as dependent on the ratio (ArO-)/(ArOH) and for diluted ArOH corresponds to an increased contribution of much faster electron transfer (ET, ArO-/dpph ) over the Hydrogen Atom Transfer (HAT, ArOH/dpph ). aroh 82-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-56 35424731-3 2022 This effect was explained as dependent on the ratio (ArO-)/(ArOH) and for diluted ArOH corresponds to an increased contribution of much faster electron transfer (ET, ArO-/dpph ) over the Hydrogen Atom Transfer (HAT, ArOH/dpph ). Hydrogen 187-195 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-56 35424731-3 2022 This effect was explained as dependent on the ratio (ArO-)/(ArOH) and for diluted ArOH corresponds to an increased contribution of much faster electron transfer (ET, ArO-/dpph ) over the Hydrogen Atom Transfer (HAT, ArOH/dpph ). aroh 82-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 166-169 35424731-3 2022 This effect was explained as dependent on the ratio (ArO-)/(ArOH) and for diluted ArOH corresponds to an increased contribution of much faster electron transfer (ET, ArO-/dpph ) over the Hydrogen Atom Transfer (HAT, ArOH/dpph ). aroh 216-220 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-56 35424731-3 2022 This effect was explained as dependent on the ratio (ArO-)/(ArOH) and for diluted ArOH corresponds to an increased contribution of much faster electron transfer (ET, ArO-/dpph ) over the Hydrogen Atom Transfer (HAT, ArOH/dpph ). 1,1-diphenyl-2-picrylhydrazyl 171-175 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-56 35424731-3 2022 This effect was explained as dependent on the ratio (ArO-)/(ArOH) and for diluted ArOH corresponds to an increased contribution of much faster electron transfer (ET, ArO-/dpph ) over the Hydrogen Atom Transfer (HAT, ArOH/dpph ). aroh 216-220 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 166-169 35424731-3 2022 This effect was explained as dependent on the ratio (ArO-)/(ArOH) and for diluted ArOH corresponds to an increased contribution of much faster electron transfer (ET, ArO-/dpph ) over the Hydrogen Atom Transfer (HAT, ArOH/dpph ). 1,1-diphenyl-2-picrylhydrazyl 221-225 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-56 35181438-0 2022 Endocrine disruptor hexachlorobenzene induces cell migration and invasion, and enhances aromatase expression levels in human endometrial stromal cells. Hexachlorobenzene 20-37 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 35424731-3 2022 This effect was explained as dependent on the ratio (ArO-)/(ArOH) and for diluted ArOH corresponds to an increased contribution of much faster electron transfer (ET, ArO-/dpph ) over the Hydrogen Atom Transfer (HAT, ArOH/dpph ). 1,1-diphenyl-2-picrylhydrazyl 221-225 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 166-169 35181438-5 2022 Results show that HCB increases ERalpha and aromatase protein levels and reduces PR content in both T-HESC and ESC. Hexachlorobenzene 18-21 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-53 35123662-4 2022 METHODS: We did a meta-analysis of individual patient data from randomised trials comparing aromatase inhibitors (anastrozole, exemestane, or letrozole) versus tamoxifen for 3 or 5 years in premenopausal women with ER-positive breast cancer receiving ovarian suppression (goserelin or triptorelin) or ablation. Anastrozole 114-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 35123662-4 2022 METHODS: We did a meta-analysis of individual patient data from randomised trials comparing aromatase inhibitors (anastrozole, exemestane, or letrozole) versus tamoxifen for 3 or 5 years in premenopausal women with ER-positive breast cancer receiving ovarian suppression (goserelin or triptorelin) or ablation. exemestane 127-137 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 35123662-4 2022 METHODS: We did a meta-analysis of individual patient data from randomised trials comparing aromatase inhibitors (anastrozole, exemestane, or letrozole) versus tamoxifen for 3 or 5 years in premenopausal women with ER-positive breast cancer receiving ovarian suppression (goserelin or triptorelin) or ablation. Letrozole 142-151 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 2533308-3 1989 Analysis of the radio-labeled protein, isolated by antibody to the cytochrome P-450arom from different preparations (P45FBIII or R-8-2) showed a major band at molecular weight 54k on SDS polyacrylamide gel electrophoresis (SDS-PAGE). Sodium Dodecyl Sulfate 183-186 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-87 35096586-5 2021 HeyL overexpression increased endogenous estradiol levels and estrogen receptor-alpha (ERalpha) transcriptional activity by upregulating CYP19A1 expression, which encodes aromatase, enhancing prostate cancer stem cell (PCSC) properties in PC3 cells. Estradiol 41-50 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 137-144 34978244-2 2022 CYP19A1 (aromatase) is the enzyme responsible for metabolizing methadone and buprenorphine in the human placenta. Methadone 63-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-7 34978244-2 2022 CYP19A1 (aromatase) is the enzyme responsible for metabolizing methadone and buprenorphine in the human placenta. Buprenorphine 77-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-7 2603928-4 1989 The amount of aromatase P-450 (aromatase cytochrome P-450) in tissue homogenates was determined after sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotting by use of a polyclonal antibody directed against aromatase cytochrome P-450. Sodium Dodecyl Sulfate 102-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 2603928-4 1989 The amount of aromatase P-450 (aromatase cytochrome P-450) in tissue homogenates was determined after sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotting by use of a polyclonal antibody directed against aromatase cytochrome P-450. Sodium Dodecyl Sulfate 102-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 2603928-4 1989 The amount of aromatase P-450 (aromatase cytochrome P-450) in tissue homogenates was determined after sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotting by use of a polyclonal antibody directed against aromatase cytochrome P-450. Sodium Dodecyl Sulfate 102-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 35014677-11 2022 Additionally, sitagliptin reversed the inhibitory effects caused by GOA-BSA treatment on the cell cycle progression and on the activation of the CREB/aromatase pathway in KGN cells, as determined by the increased expression levels of the cell cycle-associated proteins as well as those of the CREB protein and the CYP19A1 and CYP11A1 enzymes. Sitagliptin Phosphate 14-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 150-159 35014677-11 2022 Additionally, sitagliptin reversed the inhibitory effects caused by GOA-BSA treatment on the cell cycle progression and on the activation of the CREB/aromatase pathway in KGN cells, as determined by the increased expression levels of the cell cycle-associated proteins as well as those of the CREB protein and the CYP19A1 and CYP11A1 enzymes. Sitagliptin Phosphate 14-25 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 314-321 34601599-3 2022 METHODS: As primary genetic instrument, we used a genetic variant in the CYP19A1 gene that is strongly associated with serum E2 levels. Estradiol 125-127 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 73-80 2603928-4 1989 The amount of aromatase P-450 (aromatase cytochrome P-450) in tissue homogenates was determined after sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotting by use of a polyclonal antibody directed against aromatase cytochrome P-450. polyacrylamide 125-139 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 2603928-4 1989 The amount of aromatase P-450 (aromatase cytochrome P-450) in tissue homogenates was determined after sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotting by use of a polyclonal antibody directed against aromatase cytochrome P-450. polyacrylamide 125-139 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 2603928-4 1989 The amount of aromatase P-450 (aromatase cytochrome P-450) in tissue homogenates was determined after sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotting by use of a polyclonal antibody directed against aromatase cytochrome P-450. polyacrylamide 125-139 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-40 2603928-5 1989 The activity of aromatase in microsomal preparations was assayed by determining the rate of incorporation of tritium from 1-[3H]androstenedione into [3H]water. Tritium 109-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-25 2603928-5 1989 The activity of aromatase in microsomal preparations was assayed by determining the rate of incorporation of tritium from 1-[3H]androstenedione into [3H]water. 1-[3h]androstenedione 122-143 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-25 2603928-5 1989 The activity of aromatase in microsomal preparations was assayed by determining the rate of incorporation of tritium from 1-[3H]androstenedione into [3H]water. Tritium 125-127 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-25 2603928-5 1989 The activity of aromatase in microsomal preparations was assayed by determining the rate of incorporation of tritium from 1-[3H]androstenedione into [3H]water. Water 153-158 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-25 2603928-12 1989 In conclusion, the placenta is a major site of conversion of C19 steroid precursors to estrogens because of the amount of enzyme and the high rate of activity of aromatase compared with those of other fetal tissues. Steroids 65-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 162-171 2533308-3 1989 Analysis of the radio-labeled protein, isolated by antibody to the cytochrome P-450arom from different preparations (P45FBIII or R-8-2) showed a major band at molecular weight 54k on SDS polyacrylamide gel electrophoresis (SDS-PAGE). polyacrylamide 187-201 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-87 2533308-3 1989 Analysis of the radio-labeled protein, isolated by antibody to the cytochrome P-450arom from different preparations (P45FBIII or R-8-2) showed a major band at molecular weight 54k on SDS polyacrylamide gel electrophoresis (SDS-PAGE). Sodium Dodecyl Sulfate 223-226 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 67-87 2533308-5 1989 The radio-labeled protein isolated by anti-NADPH cytochrome P-450 reductase, REDFBIV, showed a major band at the molecular weight 73k on SDS-PAGE with comparable intensity in control and hormone treated samples. Sodium Dodecyl Sulfate 137-140 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-65 2533308-7 1989 When a secretory endometrium and a decidua were labeled with [35S]methionine, the cytochrome P-450arom was detected only in the decidua. Sulfur-35 62-65 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-102 2533308-7 1989 When a secretory endometrium and a decidua were labeled with [35S]methionine, the cytochrome P-450arom was detected only in the decidua. Methionine 66-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-102 2550923-4 1989 The aromatase specific activity, low in freshly isolated cells, increased fourfold in attached cells by 3 h, and achieved a 10- to 15-fold increase by 40 to 48 h. In attached cells grown with cAMP, aromatase activity was further stimulated by about fourfold, relative to the control. Cyclic AMP 192-196 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 4-13 2550508-0 1989 Estrogen synthetase (aromatase) activity in primary culture of human term placental cells: effects of cell preparation, growth medium, and serum on adenosine 3",5"-monophosphate response. Cyclic AMP 148-177 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-19 2550923-4 1989 The aromatase specific activity, low in freshly isolated cells, increased fourfold in attached cells by 3 h, and achieved a 10- to 15-fold increase by 40 to 48 h. In attached cells grown with cAMP, aromatase activity was further stimulated by about fourfold, relative to the control. Cyclic AMP 192-196 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 198-207 2550923-6 1989 The effect of cAMP on aromatase in these unattached cells was manifested by a two-fold stimulation of activity by 18 h, relative to control unattached cells. Cyclic AMP 14-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 22-31 3497895-6 1987 These peroxyl radicals cannot be observed directly, but their reactions with the two flavonols, kaempferol and quercetin, acting as radical-scavenging antioxidants, produced strongly absorbing aroxyl radicals (ArO.). perhydroxyl radical 6-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 210-213 3419162-4 1988 An injection of PMSG resulted in a specific stimulation of aromatase activity 12 times the increase in ovarian weight in 48 h. Kinetic studies demonstrated that, although the PMSG-stimulated ovarian microsomes had one-tenth the specific activity of the human placenta, the Km values were similar (about 33 and 44 nM, respectively). pmsg 16-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-68 3419162-4 1988 An injection of PMSG resulted in a specific stimulation of aromatase activity 12 times the increase in ovarian weight in 48 h. Kinetic studies demonstrated that, although the PMSG-stimulated ovarian microsomes had one-tenth the specific activity of the human placenta, the Km values were similar (about 33 and 44 nM, respectively). pmsg 175-179 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 59-68 3419162-5 1988 The potent inhibitor of placenta aromatase, 10-propargylestr-4-ene-3,17-dione, was used to further characterize the enzyme. plomestane 44-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-42 3126038-2 1988 A deoxyoligonucleotide (62 mer) probe derived from an amino acid sequence of purified human placental P450arom was used to screen a rat granulosa cell lambda gt11 cDNA expression library. deoxyoligonucleotide 2-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 102-110 3126038-4 1988 In vitro translation using mRNA that had been selected by hybridization to a 1.2-kb rat P450arom cDNA insert yielded an 35S-labeled translation product that bound antihuman aromatase immunoglobulin and comigrated with purified human placental aromatase on sodium dodecyl sulfate-polyacrylamide gel electrophoresis, thus verifying that the clones do encode for P450arom. Sulfur-35 120-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 360-368 3347045-4 1988 In contrast to human placental estrogen synthetase, this enzyme aromatized testosterone and 19-nortestosterone with similar efficiency. Testosterone 75-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-50 3347045-4 1988 In contrast to human placental estrogen synthetase, this enzyme aromatized testosterone and 19-nortestosterone with similar efficiency. Nandrolone 92-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-50 2541021-0 1989 Alternative usage of different poly(A) addition signals for two major species of mRNA encoding human aromatase P-450. Poly A 31-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-116 2653438-0 1989 Tritium release from [19-3H]-19,19-difluoroandrost-4-ene-3,17-dione during inactivation of aromatase. Tritium 0-7 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-100 2653438-0 1989 Tritium release from [19-3H]-19,19-difluoroandrost-4-ene-3,17-dione during inactivation of aromatase. [19-3h]-19,19-difluoroandrost-4-ene-3,17-dione 21-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-100 2653438-1 1989 Aromatase is a cytochrome P-450 enzyme involved in the conversion of androst-4-ene-3,17-dione to estrogen via sequential oxidations at the 19-methyl group. Androstenedione 69-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 2653438-2 1989 Previous studies from this laboratory showed that 19,19-difluoroandrost-4-ene-3,17-dione (5) is a mechanism-based inactivator of aromatase. 19,19-difluoroandrost-4-ene-3,17-dione 50-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-138 2653438-7 1989 Incubation of [19-3H]19,19-difluoroandrost-4-ene-3,17-dione with human placental microsomal aromatase at differing protein concentrations resulted in time-dependent NADPH-dependent, and protein-dependent release of tritium. [19-3h]19,19-difluoroandrost-4-ene-3,17-dione 14-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 2653438-7 1989 Incubation of [19-3H]19,19-difluoroandrost-4-ene-3,17-dione with human placental microsomal aromatase at differing protein concentrations resulted in time-dependent NADPH-dependent, and protein-dependent release of tritium. NADP 165-170 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 2653438-7 1989 Incubation of [19-3H]19,19-difluoroandrost-4-ene-3,17-dione with human placental microsomal aromatase at differing protein concentrations resulted in time-dependent NADPH-dependent, and protein-dependent release of tritium. Tritium 215-222 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 2653438-8 1989 This tritium release is not observed in the presence of (19R)-10 beta-oxiranylestr-4-ene-3,17-dione, a powerful competitive inhibitor of aromatase. Tritium 5-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 137-146 2653438-8 1989 This tritium release is not observed in the presence of (19R)-10 beta-oxiranylestr-4-ene-3,17-dione, a powerful competitive inhibitor of aromatase. (19r)-10 beta-oxiranylestr-4-ene-3,17-dione 56-99 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 137-146 2653438-9 1989 We conclude that aromatase attacks the 19-carbon of 19,19-difluoroandrost-4-ene-3,17-dione, as originally postulated. Carbon 42-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 2653438-9 1989 We conclude that aromatase attacks the 19-carbon of 19,19-difluoroandrost-4-ene-3,17-dione, as originally postulated. 19,19-difluoroandrost-4-ene-3,17-dione 52-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 2606936-3 1989 Nicotine and its metabolite cotinine are strong inhibitors of the aromatase. Nicotine 0-8 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-75 2606936-3 1989 Nicotine and its metabolite cotinine are strong inhibitors of the aromatase. Cotinine 28-36 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 66-75 3144064-1 1987 To determine the molecular basis for changes in aromatase (P450arom) activity in rat ovarian follicles and corpora lutea, seven clones for rat P450arom cDNA have been identified and isolated from a rat granulosa cell lambda gt11 cDNA expression library using a 62 mer deoxyoligonucleotide probe (derived from an amino acid sequence of purified human placental aromatase) and a human placental P450arom cDNA probe. deoxyoligonucleotide 268-288 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 143-151 3504613-2 1987 In addition, 22 of the tumors were studied for their response to the aromatase inhibitor, 4-OH-androstenedione, in a soft agar clonogenic cell assay system. 4-oh-androstenedione 90-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-78 3504613-5 1987 Of 12 tumors grown successfully in the soft agar culture system, only 1 showed a substantial (greater than 50%) reduction in colony-forming efficiency after exposure to the aromatase inhibitor. Agar 44-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 173-182 3504615-1 1987 Tumor aromatase has been correlated with clinical response to treatment with aminoglutethimide in patients with estrogen receptor-positive advanced breast cancer. Aminoglutethimide 77-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 6-15 3504615-3 1987 Measurement of in vitro aromatase in sequential biopsies of large primary tumors before and during treatment with aminoglutethimide-hydrocortisone showed a marked but paradoxical rise in activity following therapy. aminoglutethimide-hydrocortisone 114-146 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-33 3497895-6 1987 These peroxyl radicals cannot be observed directly, but their reactions with the two flavonols, kaempferol and quercetin, acting as radical-scavenging antioxidants, produced strongly absorbing aroxyl radicals (ArO.). Quercetin 111-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 210-213 3497895-6 1987 These peroxyl radicals cannot be observed directly, but their reactions with the two flavonols, kaempferol and quercetin, acting as radical-scavenging antioxidants, produced strongly absorbing aroxyl radicals (ArO.). aroxyl radicals 193-208 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 210-213 3497895-6 1987 These peroxyl radicals cannot be observed directly, but their reactions with the two flavonols, kaempferol and quercetin, acting as radical-scavenging antioxidants, produced strongly absorbing aroxyl radicals (ArO.). Flavonols 85-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 210-213 3497895-6 1987 These peroxyl radicals cannot be observed directly, but their reactions with the two flavonols, kaempferol and quercetin, acting as radical-scavenging antioxidants, produced strongly absorbing aroxyl radicals (ArO.). kaempferol 96-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 210-213 3558412-2 1987 The mechanism of inhibition of estrogen synthetase (P-450arom) by 19R- and 19S-isomers of 10-oxiranyl-and 10-thiiranyl-4-estrene-3,17-dione was investigated using human placental microsomes and purified enzyme preparations. 10-oxiranyl-and 10-thiiranyl-4-estrene-3,17-dione 90-139 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-50 3504063-5 1987 For example, they provided a system for the study of the potency and specificity of the aromatase inhibitors 4-OHA and MDL 18,962. formestane 109-114 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-97 3504063-6 1987 Finally, the influence of DEX on aromatase in genital skin fibroblasts differs in some important respects from the pattern of control observed in adipose tissue stromal-vascular cells. Dextromethorphan 26-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-42 3032980-1 1987 Effects of dibutyryl cyclic AMP, epidermal growth factor, and phorbol esters on the synthesis of aromatase cytochrome P-450. Bucladesine 11-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 118-123 3032980-1 1987 Effects of dibutyryl cyclic AMP, epidermal growth factor, and phorbol esters on the synthesis of aromatase cytochrome P-450. Phorbol Esters 62-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 118-123 3558412-2 1987 The mechanism of inhibition of estrogen synthetase (P-450arom) by 19R- and 19S-isomers of 10-oxiranyl-and 10-thiiranyl-4-estrene-3,17-dione was investigated using human placental microsomes and purified enzyme preparations. 10-oxiranyl-and 10-thiiranyl-4-estrene-3,17-dione 90-139 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 52-61 3558412-6 1987 Spectral titrations of microsomal preparations and purified P-450arom showed that binding of the 19R-isomers shifts the Soret maximum of the ferric enzyme to 411 nm (10-oxirane) or 425 nm (10-thiirane); addition of excess androstenedione reversed the spectral changes, producing the high spin form of the enzyme with a Soret peak at 393 nm. 10-oxirane 166-176 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-69 3558412-6 1987 Spectral titrations of microsomal preparations and purified P-450arom showed that binding of the 19R-isomers shifts the Soret maximum of the ferric enzyme to 411 nm (10-oxirane) or 425 nm (10-thiirane); addition of excess androstenedione reversed the spectral changes, producing the high spin form of the enzyme with a Soret peak at 393 nm. 10-thiirane 189-200 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-69 3558412-6 1987 Spectral titrations of microsomal preparations and purified P-450arom showed that binding of the 19R-isomers shifts the Soret maximum of the ferric enzyme to 411 nm (10-oxirane) or 425 nm (10-thiirane); addition of excess androstenedione reversed the spectral changes, producing the high spin form of the enzyme with a Soret peak at 393 nm. Androstenedione 222-237 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-69 3558412-10 1987 Stereoselective binding of the 19R-isomers by P-450arom further suggests that the heme is likely to be positioned above C1 and C2 of the A ring. Heme 82-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-55 2950193-2 1987 In randomized clinical trials involving postmenopausal patients, tamoxifen has been found to be as effective as high-dose estrogens, androgens, progestins, and the aromatase inhibitor, aminoglutethimide. Aminoglutethimide 185-202 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 164-173 2936597-0 1986 Modulation of aromatase activity in human endometrial stromal cells by steroids, tamoxifen and RU 486. Steroids 71-79 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 3546561-2 1987 The major source of estrogen in postmenopausal women is the peripheral conversion of androstenedione to estrone through the enzyme aromatase. Androstenedione 85-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 131-140 3546561-2 1987 The major source of estrogen in postmenopausal women is the peripheral conversion of androstenedione to estrone through the enzyme aromatase. Estrone 104-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 131-140 3546561-8 1987 One inhibitor of aromatase, aminoglutethimide, has been extensively studied in patients with breast cancer. Aminoglutethimide 28-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 17-26 3546561-11 1987 In randomized clinical trials with this regimen, aromatase inhibition with aminoglutethimide produced tumor regression with similar frequency as did surgical hypophysectomy, surgical adrenalectomy, or tamoxifen administration. Aminoglutethimide 75-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 49-58 3546561-14 1987 Low-dose aminoglutethimide appears to block aromatase effectively and to have limited side effects, and is undergoing extensive clinical trial. Aminoglutethimide 9-26 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-53 3546561-15 1987 A more specific aromatase inhibitor, 4-hydroxyandrostenedione, is now also being tested clinically, whereas MDL 18962, another new selective inhibitor, is undergoing study in animals. formestane 37-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 16-25 3818600-1 1987 Microsomal estrogen synthetase (cytochrome P-450ES), also known as aromatase, was purified from fresh human placenta microsomes by DEAE-Trisacryl and testosterone-agarose chromatography. DEAE-Trisacryl 131-145 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-30 3818600-1 1987 Microsomal estrogen synthetase (cytochrome P-450ES), also known as aromatase, was purified from fresh human placenta microsomes by DEAE-Trisacryl and testosterone-agarose chromatography. Testosterone 150-162 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-30 3818600-1 1987 Microsomal estrogen synthetase (cytochrome P-450ES), also known as aromatase, was purified from fresh human placenta microsomes by DEAE-Trisacryl and testosterone-agarose chromatography. Sepharose 163-170 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-30 3818600-2 1987 Estrogen synthetase assays were done with androstenedione as substrate, NADPH as electron donor, and a partially purified P-450 reductase from human placenta as the electron carrier. Androstenedione 42-57 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-19 2447395-4 1987 Some aspects of the activity of known aromatase inhibitors as substrates for enzymes of steroid metabolism and their potential relevance to the pharmacology of the compounds are discussed. Steroids 88-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-47 3098102-0 1986 Human chorionic gonadotropin reduces aromatizable androgens and aromatase activity in women stimulated by clomiphene citrate and human menopausal gonadotropin. Clomiphene 106-124 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 64-73 3098102-6 1986 Aromatase activity appeared to be inhibited because of a significant reduction in the estradiol/testosterone ratio 34 to 36 hours after human chorionic gonadotropin administration. Estradiol 86-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 3098102-6 1986 Aromatase activity appeared to be inhibited because of a significant reduction in the estradiol/testosterone ratio 34 to 36 hours after human chorionic gonadotropin administration. Testosterone 96-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 3098102-7 1986 Thus human chorionic gonadotropin, which triggers ovulation in women treated by clomiphene citrate-human menopausal gonadotropin, appears to partially reduce aromatizable substrate as well as inhibit aromatase activity. Clomiphene 80-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 200-209 3741479-0 1986 Inhibition of aromatase cytochrome P-450 (estrogen synthetase) by derivatives of alpha-naphthoflavone. alpha-naphthoflavone 81-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-61 3557327-2 1987 Groups matched on their initial levels of behavior received either continued treatment with TP alone, or TP together with 6 mg/day injections of the aromatase inhibitor 1,4,6-androstatriene-3,17-dione (ATD). androsta-1,4,6-triene-3,17-dione 169-200 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 149-158 3320558-1 1987 Aromatase catalyzes the conversion of androgens to estrogens through a series of monooxygenations to achieve the 19-desmolation and aromatization of the neutral steroid ring-A structure. Steroids 161-168 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 3320558-3 1987 Partially purified aromatase in each form was immunoaffinity chromatographed to give a single band (SDS-PAGE) cytochrome P-450 of 55 kDa, utilizing a mouse monoclonal anti-human placental aromatase cytochrome P-450 IgGi (MAb3-2C2) which is capable of suppressing placental aromatase activity. Sodium Dodecyl Sulfate 100-103 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 19-28 3320558-4 1987 The purified cytochrome P-450 showed specific aromatase activity of 25-30 nmol/min per mg with Km of 20-30 nM for androstenedione on reconstitution with NADPH-cyt P-450 reductase and dilauroyl L-alpha-phosphatidylcholine. Androstenedione 114-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-55 3320558-4 1987 The purified cytochrome P-450 showed specific aromatase activity of 25-30 nmol/min per mg with Km of 20-30 nM for androstenedione on reconstitution with NADPH-cyt P-450 reductase and dilauroyl L-alpha-phosphatidylcholine. dilauroyl l-alpha-phosphatidylcholine 183-220 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-55 3320558-7 1987 Use of purified aromatase and [19-3H3, 4-14C]androstenedione led us to discover a metabolic switching by aromatase to 2 beta-hydroxylation of androgen. [19-3h3, 4-14c]androstenedione 30-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 105-114 3747512-2 1986 Aromatase activity was expressed as pmol (estrone + estradiol) formed in the medium per mg cell protein per day. Estrone 42-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 3747512-2 1986 Aromatase activity was expressed as pmol (estrone + estradiol) formed in the medium per mg cell protein per day. Estradiol 52-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 3747512-3 1986 Using this method we were able to investigate the kinetic properties of aromatase in different cell strains and its stimulation by dexamethasone. Dexamethasone 131-144 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 72-81 3747512-9 1986 The stimulatory effect of dexamethasone on aromatase activity in cultured fibroblasts was measured after preincubation of the cells for 48 h with dexamethasone, by determining estrogen formation after 24 h incubation of the cells with androstenedione 10(-6) M using this enzymatic method. Dexamethasone 26-39 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-52 3747512-9 1986 The stimulatory effect of dexamethasone on aromatase activity in cultured fibroblasts was measured after preincubation of the cells for 48 h with dexamethasone, by determining estrogen formation after 24 h incubation of the cells with androstenedione 10(-6) M using this enzymatic method. Dexamethasone 146-159 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-52 3747512-9 1986 The stimulatory effect of dexamethasone on aromatase activity in cultured fibroblasts was measured after preincubation of the cells for 48 h with dexamethasone, by determining estrogen formation after 24 h incubation of the cells with androstenedione 10(-6) M using this enzymatic method. Androstenedione 235-250 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 43-52 2936597-0 1986 Modulation of aromatase activity in human endometrial stromal cells by steroids, tamoxifen and RU 486. Tamoxifen 81-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 2936597-0 1986 Modulation of aromatase activity in human endometrial stromal cells by steroids, tamoxifen and RU 486. Mifepristone 95-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-23 2936597-1 1986 The regulation of aromatase activity (AA) in human endometrial stromal cells by various steroids was studied in primary cell culture. Steroids 88-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 18-27 3527304-7 1986 Aromatase inhibitors can be used to suppress the levels of circulating estrone, estrone sulfate, and estradiol in postmenopausal women. Estrone 71-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 3959033-1 1986 The synthesis and evaluation of 4-(substituted thio)-4-androstene-3,17-dione derivatives as inhibitors of estrogen synthetase (aromatase) are described. 4-(substituted thio)-4-androstene-3,17-dione 32-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 106-125 3527304-7 1986 Aromatase inhibitors can be used to suppress the levels of circulating estrone, estrone sulfate, and estradiol in postmenopausal women. estrone sulfate 80-95 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 3527304-7 1986 Aromatase inhibitors can be used to suppress the levels of circulating estrone, estrone sulfate, and estradiol in postmenopausal women. Estradiol 101-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 3527304-8 1986 Aminoglutethimide, the major inhibitor currently used clinically, acts in a competitive fashion and blocks cholesterol side chain cleavage and 11 beta-hydroxylase as well as aromatase. Aminoglutethimide 0-17 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 174-183 3527304-13 1986 Further development of active aromatase inhibitors should allow precise control of estradiol levels in women with breast cancer. Estradiol 83-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 30-39 6474163-0 1984 Inhibition of human estrogen synthetase (aromatase) by flavones. Flavones 55-63 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-39 3094971-5 1986 The earliest available aromatase antagonist, aminoglutethimide, suppresses estrogen production to the same extent as surgical ablation and is an effective treatment for breast cancer. Aminoglutethimide 45-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 23-32 3994715-9 1985 We suggest that these 6-hydroperoxyandrogens may function as active-site directed inhibitors and inactivators of estrogen synthetase through oxidation of cysteine residues. Cysteine 154-162 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-132 4083898-1 1985 Aromatase cytochrome P-450 (P-450AROM) was partially purified from human placental microsomes by hydrophobic affinity chromatography using Phenyl-Sepharose and ion-exchange chromatography on DEAE-cellulose. phenyl-sepharose 139-155 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-37 4083898-1 1985 Aromatase cytochrome P-450 (P-450AROM) was partially purified from human placental microsomes by hydrophobic affinity chromatography using Phenyl-Sepharose and ion-exchange chromatography on DEAE-cellulose. DEAE-Cellulose 191-205 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 28-37 4083898-2 1985 The resulting preparation had a specific activity of 2 nmol/mg protein with respect to cytochrome P-450 content and displayed a type I difference spectrum upon addition of the substrate androstenedione. Androstenedione 186-201 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 98-103 4083898-9 1985 The monoclonal IgG was covalently linked to a Sepharose 4B column and was used for immunoaffinity chromatography of cytochrome P-450AROM. Sepharose 46-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-136 2983707-0 1985 Estrogen synthetase stimulation by hemin in human choriocarcinoma cell culture. Hemin 35-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-19 6547488-1 1984 Derivatives of 19- azaandrostenedione (10 beta-amino-4- estrene -3,17-dione, 2) and 19-amino-4-androstene-3,17-dione (3) were synthesized as potential inhibitors of aromtase (estrogen synthetase). 19-amino-4-androstene-3,17-dione 84-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 175-194 6723577-0 1984 Inhibition of estrogen synthetase (aromatase) by 4-cyclohexylaniline. 4-cyclohexylaniline 49-68 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 14-33 6288342-0 1982 Estrogen synthetase stimulation by dibutyryl cyclic adenosine 3",5"-monophosphate in human choriocarcinoma cell culture: the relative biosynthetic rate of the nicotinamide adenine dinucleotide phosphate (reduced form)-cytochrome c reductase component. Bucladesine 35-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-19 6288342-0 1982 Estrogen synthetase stimulation by dibutyryl cyclic adenosine 3",5"-monophosphate in human choriocarcinoma cell culture: the relative biosynthetic rate of the nicotinamide adenine dinucleotide phosphate (reduced form)-cytochrome c reductase component. NADP 159-202 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-19 7093221-1 1982 19,19-Difluoroandrost-4-ene-3,17-dione (1) and 19-fluorcandrost-4-ene-3,17-dione (2) have been synthesized, and the interaction of these compounds with the estrogen synthetase (aromatase) activity of human placental microsomes has been studied. 19,19-difluoroandrost-4-ene-3,17-dione 0-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 156-175 6282920-0 1982 Effects of dibutyryl adenosine 3",5"-monophosphate, luteinizing hormone-releasing hormone, and aromatase inhibitor on simultaneous outputs of progesterone, 17 beta-estradiol, and human chorionic gonadotropin by term placental explants. Progesterone 142-154 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-104 6282920-7 1982 Aromatase inhibitor decreased both 17 beta-estradiol and hCG outputs in a dose-dependent fashion, but it was without effect on the output of progesterone. Estradiol 35-52 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 6282920-10 1982 Aromatase inhibitor decreased the output of both hCG and 17 beta-estradiol, but the effect on hCG was direct and not due to the decrease in 17 beta-estradiol. Estradiol 57-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 7093221-1 1982 19,19-Difluoroandrost-4-ene-3,17-dione (1) and 19-fluorcandrost-4-ene-3,17-dione (2) have been synthesized, and the interaction of these compounds with the estrogen synthetase (aromatase) activity of human placental microsomes has been studied. 19-fluorcandrost-4-ene-3,17-dione 47-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 156-175 7058343-1 1982 Norethisterone (17 alpha-ethynyl-19-nortestosterone) is an effective irreversible inhibitor of estrogen synthetase (aromatase), the enzyme responsible for the conversion of androgens to estrogens, even at a 2 X 10(-6) molar concentration. Norethindrone 0-14 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-114 7058343-1 1982 Norethisterone (17 alpha-ethynyl-19-nortestosterone) is an effective irreversible inhibitor of estrogen synthetase (aromatase), the enzyme responsible for the conversion of androgens to estrogens, even at a 2 X 10(-6) molar concentration. Norethindrone 16-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-114 7305387-0 1981 Cooperative interaction of reduced pyridine nucleotides with estrogen synthetase of human placental microsomes. pyridine nucleotides 35-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-80 6261425-0 1981 Macromolecular synthesis is required for stimulation of estrogen synthetase activity by dibutyryl cyclic AMP plus theophylline in choriocarcinoma cell culture. Bucladesine 88-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-75 6261425-0 1981 Macromolecular synthesis is required for stimulation of estrogen synthetase activity by dibutyryl cyclic AMP plus theophylline in choriocarcinoma cell culture. Theophylline 114-126 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-75 6261425-1 1981 Estrogen secretion and estrogen synthetase (aromatase) activity are stimulated in human trophoblast cells (JAr line) after addition of 1 mM dibutyryl cyclic AMP plus 1 mM theophylline (dbT) to the growth medium. Theophylline 171-183 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 23-42 6261425-1 1981 Estrogen secretion and estrogen synthetase (aromatase) activity are stimulated in human trophoblast cells (JAr line) after addition of 1 mM dibutyryl cyclic AMP plus 1 mM theophylline (dbT) to the growth medium. Bucladesine 140-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 23-42 6261425-1 1981 Estrogen secretion and estrogen synthetase (aromatase) activity are stimulated in human trophoblast cells (JAr line) after addition of 1 mM dibutyryl cyclic AMP plus 1 mM theophylline (dbT) to the growth medium. di-n-butyltin 185-188 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 23-42 212849-2 1978 The stimulation of estrogen biosynthesis by N6,O2"--dibutyryl adenosine 3":5"-cyclic monophosphate and theophylline dbT) in cultures of the JAr line of choriocarcinoma cells was investigated by measuring the specific activity and kinetic constants of estrogen synthetase (aromatase) in the various subcellular fractions after differential centrifugation of homogenized cells in isotonic sucrose. Bucladesine 44-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 251-270 7392614-1 1980 The molecular conformation of 19-hydroxy-4-androstene-3,17-dione, as an intermediate for estrogen synthetase. 19-hydroxy-4-androstene-3,17-dione 30-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 89-108 494371-0 1979 Synthesis and some reactions of 6-bromoandrogens: potential affinity ligand and inactivator of estrogen synthetase. 6-bromoandrogens 32-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-114 217389-0 1978 Trophoblast estrogen synthetase stimulation by dibutyryl cyclic AMP and theophylline: increase in cytochrome P-450 content. Bucladesine 47-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-31 217389-0 1978 Trophoblast estrogen synthetase stimulation by dibutyryl cyclic AMP and theophylline: increase in cytochrome P-450 content. Theophylline 72-84 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 12-31 34003583-4 2022 As such, ERRalpha deregulated the calcitriol-VDR transcription by enhancing the expression of CYP24A1 as well as of both ERalpha and aromatase (CYP19A1) in calcitriol-treated cells. Calcitriol 34-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 144-151 33677314-7 2021 Both alpha-CYP and beta-CYP upregulated genes encoding estrogen- and aldosterone-forming enzymes including 17-betaHSD, CYP19, STAR, and CYP11B2. cypermethrin 10-14 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 119-124 33677314-7 2021 Both alpha-CYP and beta-CYP upregulated genes encoding estrogen- and aldosterone-forming enzymes including 17-betaHSD, CYP19, STAR, and CYP11B2. Aldosterone 69-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 119-124 849980-1 1977 In previous investigations of human placental estrogen synthetase (aromatase), activity was found in the microsomal and mitochondrial fractions after homogenization and subcellular fractionation of placental tissue in isotonic sucrose. Sucrose 227-234 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 46-65 4342056-7 1972 When the CPV-1 in fraction 5 were isolated from cells briefly incubated with (3)H-uridine and (3)H-adenosine prior to cell disruption, a large proportion was found to be labeled by high-resolution autoradiography. (3)h-uridine 77-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-14 4342056-7 1972 When the CPV-1 in fraction 5 were isolated from cells briefly incubated with (3)H-uridine and (3)H-adenosine prior to cell disruption, a large proportion was found to be labeled by high-resolution autoradiography. h-adenosine 97-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-14 4672392-7 1972 Inhibition of cellular ribonucleic acid (RNA) and protein synthesis by actinomycin D during SFV infection did not decrease the counts of CPV-1; however, biogenesis of CPV-1 was decreased when viral replication was limited by inhibitors of viral RNA synthesis (guanidine) or of viral protein synthesis (cycloheximide). Guanidine 260-269 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 167-172 4672392-7 1972 Inhibition of cellular ribonucleic acid (RNA) and protein synthesis by actinomycin D during SFV infection did not decrease the counts of CPV-1; however, biogenesis of CPV-1 was decreased when viral replication was limited by inhibitors of viral RNA synthesis (guanidine) or of viral protein synthesis (cycloheximide). Cycloheximide 302-315 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 167-172 34003583-4 2022 As such, ERRalpha deregulated the calcitriol-VDR transcription by enhancing the expression of CYP24A1 as well as of both ERalpha and aromatase (CYP19A1) in calcitriol-treated cells. Calcitriol 156-166 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 144-151 34001150-9 2021 CONCLUSION: Our results suggest that increased levels of estrogen and pregnenolone in follicular fluid may affect follicle development in PCOS patients, and the mechanism is partially related to HSD17B1, CYP19A1 and CYP11A1 expression change in FF exosomes. Pregnenolone 70-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 204-211 33485996-7 2021 It showed that dexamethasone decreased miRNA320a-3p and P450arom expression, as well as E2 synthesis, and increased RUNX2 expression. Dexamethasone 15-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 56-64 33948439-4 2021 The perchlorate"s effects were detected by histopathological examination, aromatase/CYP19 A1 activity, reactive oxygen species production (ROS), and Caspase-3 expression. perchlorate 4-15 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 74-83 33948439-4 2021 The perchlorate"s effects were detected by histopathological examination, aromatase/CYP19 A1 activity, reactive oxygen species production (ROS), and Caspase-3 expression. perchlorate 4-15 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 84-92 33881728-0 2021 Copper and Selenium stimulates CYP19A1 expression in caprine ovarian granulosa cells: possible involvement of AKT and WNT signalling pathways. Selenium 11-19 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 31-38 33881728-12 2021 The mRNA expression of AKT1, CYP19A1, WNT2 & 4, FZD6 and APC2 were significantly (p < 0.05) higher in Cu and Cu + Se groups compared to control. Copper 102-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-36 33881728-12 2021 The mRNA expression of AKT1, CYP19A1, WNT2 & 4, FZD6 and APC2 were significantly (p < 0.05) higher in Cu and Cu + Se groups compared to control. Copper 109-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-36 33881728-15 2021 Our study support a key role of copper and selenium in activation of AKT and WNT signalling pathway that further lead to increase in the mRNA expression of CYP19A1. Copper 32-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 156-163 33881728-15 2021 Our study support a key role of copper and selenium in activation of AKT and WNT signalling pathway that further lead to increase in the mRNA expression of CYP19A1. Selenium 43-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 156-163 33948439-7 2021 Despite the slight induction effect of the lowest perchlorate concentration (5 mug/L) on caspase 3 expression, CYP 19 activity, and ROS generation, it did not affect placental cellular viability. perchlorate 50-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 111-117 33948439-8 2021 Conclusion: This study suggested that perchlorate could modulate aromatase activity and placental cytotoxicity. perchlorate 38-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 33113109-7 2021 RESULTS: Incubating TMSCs in differentiating media induced the gene expression of cytochrome P450 19A1 (CYP19A1), which plays a key role in estrogen biosynthesis, and increased 17beta-estradiol secretion upon testosterone addition. Testosterone 209-221 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 82-102 33113109-7 2021 RESULTS: Incubating TMSCs in differentiating media induced the gene expression of cytochrome P450 19A1 (CYP19A1), which plays a key role in estrogen biosynthesis, and increased 17beta-estradiol secretion upon testosterone addition. Testosterone 209-221 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 104-111 33705547-5 2021 PARTICIPANTS: We assessed the association between endogenous estradiol genetically predicted by 22 variants in the CYP19A1 gene region and risk of thromboembolism (5815 cases) in 170,593 unrelated men of white ancestry in the UK Biobank. Estradiol 61-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 115-122 33705547-7 2021 RESULTS: Endogenous estradiol genetically predicted by variants in the CYP19A1 gene region was inversely associated with risk of thromboembolism (odds ratio per SD increase in estradiol 0.74, 95% confidence interval 0.62-0.90). Estradiol 20-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-78 33705547-7 2021 RESULTS: Endogenous estradiol genetically predicted by variants in the CYP19A1 gene region was inversely associated with risk of thromboembolism (odds ratio per SD increase in estradiol 0.74, 95% confidence interval 0.62-0.90). Estradiol 176-185 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-78 33705547-9 2021 Subsequent explorative analyses evaluating potential repercussions of thromboembolism revealed that endogenous estradiol genetically predicted by variants in the CYP19A1 gene region was inversely associated with risk of ischemic stroke (0.68, 0.49-0.95) but not myocardial infarction (0.97, 0.84-1.13). Estradiol 111-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 162-169 33705547-11 2021 The ratio between testosterone and estradiol, determined by aromatase (CYP19A1) activity, may contribute to the overall impact of sex steroids on thromboembolism in men. Testosterone 18-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-78 33705547-11 2021 The ratio between testosterone and estradiol, determined by aromatase (CYP19A1) activity, may contribute to the overall impact of sex steroids on thromboembolism in men. Estradiol 35-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-78 33705547-11 2021 The ratio between testosterone and estradiol, determined by aromatase (CYP19A1) activity, may contribute to the overall impact of sex steroids on thromboembolism in men. Steroids 134-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 71-78 33485996-9 2021 The overexpression of miRNA320a-3p in vitro could also reverse the effects of dexamethasone on RUNX2, P450arom, and E2 levels. Dexamethasone 78-91 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 102-110 33302164-11 2021 Low-dose NAC upregulated CYP19A1 mRNA expression, and high-dose NAC downregulated CYP11A1 mRNA abundance (P < 0.05). Acetylcysteine 9-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 25-32 32407592-10 2021 The risk allele rs7175922 in CYP19A1 was significantly associated with higher levels of estradiol (p = 0.02) and an increased risk of POPH (OR 2.36, 95% CI 1.12-4.91, p = 0.02) whereas other SNPs were not. Estradiol 88-97 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 29-36 33383348-1 2021 BACKGROUND: The CAO/ARO/AIO trial has shown that oxaliplatin added to preoperative chemoradiotherapy and postoperative chemotherapy significantly improved disease-free survival in locally advanced rectal cancer (LARC). Oxaliplatin 49-60 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 20-23 33410845-0 2021 Bismuth(III)-catalyzed regioselective alkylation of tetrahydroquinolines and indolines towards the synthesis of bioactive core-biaryl oxindoles and CYP19 inhibitors. Bismuth(III) 0-12 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 148-153 33410845-0 2021 Bismuth(III)-catalyzed regioselective alkylation of tetrahydroquinolines and indolines towards the synthesis of bioactive core-biaryl oxindoles and CYP19 inhibitors. 1,2,3,4-tetrahydroquinoline 52-72 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 148-153 33410845-0 2021 Bismuth(III)-catalyzed regioselective alkylation of tetrahydroquinolines and indolines towards the synthesis of bioactive core-biaryl oxindoles and CYP19 inhibitors. indoline 77-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 148-153 33451122-5 2021 Interestingly, TAD counteracted Cyp19a1 stimulation induced by BCT (p < 0.05) but did not alter the effect induced by BCT on the AR protein expression. bicalutamide 63-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-39 33512728-2 2021 Ovarian granulosa cells are the main source of E2 production because these cells express aromatase, which is encoded by the CYP19A1 gene and catalyzes the final step in E2 biosynthesis from androgens. Estradiol 47-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 89-98 33512728-2 2021 Ovarian granulosa cells are the main source of E2 production because these cells express aromatase, which is encoded by the CYP19A1 gene and catalyzes the final step in E2 biosynthesis from androgens. Estradiol 47-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 124-131 33512728-2 2021 Ovarian granulosa cells are the main source of E2 production because these cells express aromatase, which is encoded by the CYP19A1 gene and catalyzes the final step in E2 biosynthesis from androgens. Estradiol 169-171 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 89-98 33512728-2 2021 Ovarian granulosa cells are the main source of E2 production because these cells express aromatase, which is encoded by the CYP19A1 gene and catalyzes the final step in E2 biosynthesis from androgens. Estradiol 169-171 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 124-131 33130394-7 2021 Based on molecular mechanics/Generalized Born Surface Area (GBSA) free energies and ligand stability inside the active site cavity during its 120 ns MD run, it can be concluded that mycoleptodiscin B is a potent aromatase inhibitor and could be subjected to further in vitro and in vivo experiments in the drug development pipeline. mycoleptodiscin B 182-199 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 212-221 32653739-4 2021 MicroRNA-mRNA network analysis and luciferase reporter assays showed that CYP19A1, the pivotal enzyme for E2 synthesis signaling, was directly targeted by miR-146b. Estradiol 106-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 74-81 33098425-7 2021 The results highlight that at high concentrations, acetaminophen reduced the gene expression of aromatase (CYP19A1) and type 1 3beta-hydroxysteroid dehydrogenase (HSD3B1), and increased the expression of 17beta-hydroxysteroid dehydrogenase (HSD17B1). Acetaminophen 51-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 107-114 33098425-8 2021 Additionally, acetaminophen at high concentrations also reduced the protein expression of aromatase (CYP19A1). Acetaminophen 14-27 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-108 33290750-6 2020 Moreover, the expression of steroid metabolism-related genes (3betaHSD, Cyp11a1, StAR and Cyp19a1) in these two groups was down-regulated, with lower levels of estradiol (E2) and progesterone (P). Steroids 28-35 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-97 32239131-8 2020 Significant predictors for ARO colonization on NF admission included lower functional status (adjusted odds ratio [aOR]>1 for all four AROs) and recent exposure to glycopeptides (aOR>2 for VREfm, VREfc and MRSA) or 3rd/4th-generation cephalosporins (aOR>2 for MRSA and VREfm). Glycopeptides 164-177 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-30 32239131-8 2020 Significant predictors for ARO colonization on NF admission included lower functional status (adjusted odds ratio [aOR]>1 for all four AROs) and recent exposure to glycopeptides (aOR>2 for VREfm, VREfc and MRSA) or 3rd/4th-generation cephalosporins (aOR>2 for MRSA and VREfm). Cephalosporins 234-248 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-30 33290750-6 2020 Moreover, the expression of steroid metabolism-related genes (3betaHSD, Cyp11a1, StAR and Cyp19a1) in these two groups was down-regulated, with lower levels of estradiol (E2) and progesterone (P). Estradiol 160-169 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-97 33290750-6 2020 Moreover, the expression of steroid metabolism-related genes (3betaHSD, Cyp11a1, StAR and Cyp19a1) in these two groups was down-regulated, with lower levels of estradiol (E2) and progesterone (P). Estradiol 171-173 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-97 33290750-6 2020 Moreover, the expression of steroid metabolism-related genes (3betaHSD, Cyp11a1, StAR and Cyp19a1) in these two groups was down-regulated, with lower levels of estradiol (E2) and progesterone (P). Progesterone 179-191 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-97 33290750-6 2020 Moreover, the expression of steroid metabolism-related genes (3betaHSD, Cyp11a1, StAR and Cyp19a1) in these two groups was down-regulated, with lower levels of estradiol (E2) and progesterone (P). Progesterone 193-194 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-97 33053908-0 2020 Chrysoeriol Prevents TNFalpha-Induced CYP19 Gene Expression via EGR-1 Downregulation in MCF7 Breast Cancer Cells. chrysoeriol 0-11 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 38-43 33002342-6 2020 Hu-17 reduced the expression of CYP19A1 mRNA, responsible for producing aromatase, by suppressing the phosphorylation of cAMP response element binding-1. hu-17 0-5 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-39 33002342-6 2020 Hu-17 reduced the expression of CYP19A1 mRNA, responsible for producing aromatase, by suppressing the phosphorylation of cAMP response element binding-1. Cyclic AMP 121-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-39 32698066-0 2020 Effector role of cytochrome P450 reductase for androstenedione binding to human aromatase. Androstenedione 47-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 80-89 32761874-0 2020 Quantification of aromatase binding in the female human brain using [11 C]cetrozole positron emission tomography. cetrozole 74-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 18-27 32761874-1 2020 Aromatase, the enzyme that in the brain converts testosterone and androstenedione to estradiol and estrone, respectively, is a putative key factor in psychoneuroendocrinology. Testosterone 49-61 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 32761874-1 2020 Aromatase, the enzyme that in the brain converts testosterone and androstenedione to estradiol and estrone, respectively, is a putative key factor in psychoneuroendocrinology. Androstenedione 66-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 32761874-1 2020 Aromatase, the enzyme that in the brain converts testosterone and androstenedione to estradiol and estrone, respectively, is a putative key factor in psychoneuroendocrinology. Estradiol 85-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 32761874-1 2020 Aromatase, the enzyme that in the brain converts testosterone and androstenedione to estradiol and estrone, respectively, is a putative key factor in psychoneuroendocrinology. Estrone 99-106 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 32761874-12 2020 As PET tracer, [11 C]cetrozole can be employed for relatively short brain scans to measure aromatase binding using a reference tissue-based approach. cetrozole 21-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-100 33053908-3 2020 This study aimed to identify active flavones that inhibit CYP19 expression and to explore the underlying mechanisms. Flavones 36-44 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 58-63 33053908-7 2020 We further characterized and demonstrated that chrysoeriol inhibits TNFalpha-induced CYP19 expression through inhibition of extracellular signal-regulated kinase 1/2 (ERK1/2)-mediated EGR-1 expression. chrysoeriol 47-58 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 85-90 33102564-6 2020 Moreover, CYP19A1 knockdown increased (P < 0.05) the concentrations of progesterone secretion (P4) in BFGCs through increasing the mRNA expression levels of three steroidogenic genes (HSD17B1, HSD17B7, and CYP17A1). Progesterone 71-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-17 32645328-1 2020 The interactions of pharmacologically active 3-keto-Delta4-metabolite of anticancer drug abiraterone (D4A) with steroid-metabolizing cytochromes P450 (CYP51A1, CYP11A1, CYP19A1) was studied by absorption spectroscopy and molecular docking. abiraterone 89-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-176 32645328-1 2020 The interactions of pharmacologically active 3-keto-Delta4-metabolite of anticancer drug abiraterone (D4A) with steroid-metabolizing cytochromes P450 (CYP51A1, CYP11A1, CYP19A1) was studied by absorption spectroscopy and molecular docking. Steroids 112-119 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 169-176 32645328-4 2020 On the contrary, D4A interacts with the active site of CYP19A1, the key enzyme of estrogen biosynthesis, inducing type II spectral changes, while abiraterone does not. abiraterone 146-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 55-62 33102564-0 2020 Knockdown of CYP19A1 in Buffalo Follicular Granulosa Cells Results in Increased Progesterone Secretion and Promotes Cell Proliferation. Progesterone 80-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 13-20 32805116-0 2020 Small Phosphine Ligands Enable Selective Oxidative Addition of Ar-O over Ar-Cl Bonds at Nickel(0). phosphine 6-15 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-67 32681791-0 2020 Synthesis, investigation of biological effects and in silico studies of new benzimidazole derivatives as aromatase inhibitors. benzimidazole 76-89 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 105-114 32805116-0 2020 Small Phosphine Ligands Enable Selective Oxidative Addition of Ar-O over Ar-Cl Bonds at Nickel(0). Nickel 88-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 63-67 32464592-2 2020 Herein we evaluate binding of androstenedione, testosterone, and 7-hydroxyflavone to CYP19A1, also known as aromatase, in phospholipid nanodiscs by stopped-flow UV-vis spectroscopy. Androstenedione 30-45 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 85-92 32405760-0 2020 Aromatase inhibitors attenuate the effect of alendronate in women with breast cancer. Alendronate 45-56 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 32544536-7 2020 Evaluating E2 biosynthesis at high altitude, we report higher plasma levels of CYP11A1, CYP19A1, E2, lower levels of testosterone (T) and T/E2 ratio as compared to sea level. Estradiol 11-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 88-95 32862830-11 2020 Whole genome sequencing of AROs cultured from RBX2660 product and patient samples indicate ARO eradication in patients via RBX2660 administration, but also, to a lesser extent, introduction of RBX2660-derived AROs. rbx2660 46-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-30 32862830-13 2020 Antibiotic discontinuation alone resulted in significant recovery of gut microbial diversity and reduced ARG relative abundance, but RBX2660 administration more rapidly and completely changed the composition of patients" microbiome, resistome, and ARO colonization by transplanting RBX2660 microbiota into the recipients. rbx2660 133-140 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 248-251 32854459-0 2020 miR-4463 regulates aromatase expression and activity for 17beta-estradiol synthesis in response to follicle-stimulating hormone. Estradiol 57-73 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 19-28 32854459-9 2020 In particular, upregulation of miR-4463 expression by FSHR deficiency in human granulosa cells impaired 17beta-estradiol synthesis by targeting CYP19A1 and ESR1. Estradiol 104-120 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 144-151 32786398-3 2020 A complementary approach employing molecular dynamics simulations and hydrogen-deuterium exchange mass spectrometry (HDX-MS) was employed to interrogate the changes in CYP19A1 dynamics coupled to binding androstenedione (ASD). Hydrogen 70-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 168-175 32786398-3 2020 A complementary approach employing molecular dynamics simulations and hydrogen-deuterium exchange mass spectrometry (HDX-MS) was employed to interrogate the changes in CYP19A1 dynamics coupled to binding androstenedione (ASD). Androstenedione 204-219 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 168-175 32701512-3 2020 Mechanistically, CSMD1 regulates CYP19 expression in a SNP-, and drug-dependent fashion and this regulation is different among three AIs, anastrozole, exemestane, and letrozole. Anastrozole 138-149 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-38 32701512-3 2020 Mechanistically, CSMD1 regulates CYP19 expression in a SNP-, and drug-dependent fashion and this regulation is different among three AIs, anastrozole, exemestane, and letrozole. exemestane 151-161 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 33-38 32701512-5 2020 The SNP in CSMD1 that was associated with increased CSMD1 and CYP19 expression levels increased anastrozole sensitivity, but not letrozole or exemestane sensitivity. Anastrozole 96-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 62-67 32449548-3 2020 There are many aromatase inhibitors available in clinical practice like exemestane, formestane, anastrozole, letrozole, fadrozole, vorozole, and so forth, but the major challenge in anti-breast cancer therapy is the toxicity associated with aromatase inhibitors, especially the steroidal class of drugs. exemestane 72-82 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-24 32449548-3 2020 There are many aromatase inhibitors available in clinical practice like exemestane, formestane, anastrozole, letrozole, fadrozole, vorozole, and so forth, but the major challenge in anti-breast cancer therapy is the toxicity associated with aromatase inhibitors, especially the steroidal class of drugs. formestane 84-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-24 32449548-3 2020 There are many aromatase inhibitors available in clinical practice like exemestane, formestane, anastrozole, letrozole, fadrozole, vorozole, and so forth, but the major challenge in anti-breast cancer therapy is the toxicity associated with aromatase inhibitors, especially the steroidal class of drugs. Anastrozole 96-107 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-24 32449548-3 2020 There are many aromatase inhibitors available in clinical practice like exemestane, formestane, anastrozole, letrozole, fadrozole, vorozole, and so forth, but the major challenge in anti-breast cancer therapy is the toxicity associated with aromatase inhibitors, especially the steroidal class of drugs. Letrozole 109-118 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-24 32449548-3 2020 There are many aromatase inhibitors available in clinical practice like exemestane, formestane, anastrozole, letrozole, fadrozole, vorozole, and so forth, but the major challenge in anti-breast cancer therapy is the toxicity associated with aromatase inhibitors, especially the steroidal class of drugs. Fadrozole 120-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-24 32449548-3 2020 There are many aromatase inhibitors available in clinical practice like exemestane, formestane, anastrozole, letrozole, fadrozole, vorozole, and so forth, but the major challenge in anti-breast cancer therapy is the toxicity associated with aromatase inhibitors, especially the steroidal class of drugs. vorozole 131-139 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-24 32464592-2 2020 Herein we evaluate binding of androstenedione, testosterone, and 7-hydroxyflavone to CYP19A1, also known as aromatase, in phospholipid nanodiscs by stopped-flow UV-vis spectroscopy. Testosterone 47-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 85-92 32464592-2 2020 Herein we evaluate binding of androstenedione, testosterone, and 7-hydroxyflavone to CYP19A1, also known as aromatase, in phospholipid nanodiscs by stopped-flow UV-vis spectroscopy. 7-hydroxyflavone 65-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 85-92 32464592-6 2020 In addition, 7-hydroxyflavone binds to CYP19A1 nanodiscs with comparable affinity to the substrates and induces an unusual spectral response likely attributable to hydrogen bonding to, rather than displacement of the heme-coordinated water molecule. 7-hydroxyflavone 13-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 39-46 32360836-0 2020 Luteolin-7-methylether from Leonurus japonicus inhibits estrogen biosynthesis in human ovarian granulosa cells by suppression of aromatase (CYP19). luteolin-7-methylether 0-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 140-145 32334352-5 2020 According to the qRT-PCR data, 0.25 to 25 muM mono(2-ethylhexyl) phthalate (MEHP) significantly upregulated the expression levels of 17beta-HSD1 and CYP19A1, and downregulated those of CYP17A1, CYP11A1 and StAR. mono-(2-ethylhexyl)phthalate 46-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 149-156 32385792-0 2020 Association of CYP19A1 gene variations with adjuvant letrozole-induced adverse events in South Indian postmenopausal breast cancer cohort expressing hormone-receptor positivity. Letrozole 53-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 15-22 32334352-5 2020 According to the qRT-PCR data, 0.25 to 25 muM mono(2-ethylhexyl) phthalate (MEHP) significantly upregulated the expression levels of 17beta-HSD1 and CYP19A1, and downregulated those of CYP17A1, CYP11A1 and StAR. mono-(2-ethylhexyl)phthalate 76-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 149-156 32385792-3 2020 Therefore, we aimed to determine whether SNPs in the CYP19A1 gene are associated with adjuvant letrozole-induced "specific" AEs in postmenopausal hormone receptor-positive (HR+) breast cancer patients. Letrozole 95-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-60 32385792-10 2020 CONCLUSIONS: To the best of our knowledge, this is the first study assessing the impact of CYP19A1 genetic variations with adjuvant letrozole treatment-associated AEs in Indian women. Letrozole 132-141 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 91-98 32385792-11 2020 Genetic variations in the CYP19A1 gene is associated with letrozole-induced AEs and warrants further investigation in larger cohorts to validate this finding. Letrozole 58-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 26-33 32511037-10 2020 Multiple ANGPLT3 inhibitors (evinacumab, IONIS-ANGPTL3-LRx, ARO-ANG3) effectuate TG reductions up to 70% with concomitant potent reduction in all other apoB containing lipoprotein fractions. Triglycerides 81-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 60-63 32098767-5 2020 Preclinical laboratory studies examined mechanisms of action in addition to aromatase inhibition and showed that only anastrozole could directly bind to ERalpha, activate estrogen response element-dependent transcription, and stimulate growth of an aromatase-deficient CYP19A1-/- T47D breast cancer cell line. Anastrozole 118-129 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 269-276 32219443-6 2020 Furthermore, we identified aromatase, an indispensable enzyme for estradiol biosynthesis, as a novel neosubstrate of cereblon. Estradiol 66-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 27-36 32219443-8 2020 Finally, aromatase was significantly degraded in the bone marrow of patients with multiple myeloma who developed thrombocytopenia with IMiD treatment. imid 135-139 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-18 32219443-9 2020 These data suggest that aromatase is a neosubstrate of cereblon that is responsible for IMiD-induced thrombocytopenia. imid 88-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-33 32505695-8 2020 Treatment with DOX changes the expression of the aromatase gene (CYP19A1) and the secretion of 17beta-estradiol (E2) in a cell-specific manner. Doxorubicin 15-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-72 32113805-7 2020 Melamine and nicotine increased CYP19A1, melamine increased UGT and GST, PhIP with ethanol decreased CYP19A1 and increased GST, and PhIP with buprenorphine decreased CAT. melamine 0-8 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-39 32113805-7 2020 Melamine and nicotine increased CYP19A1, melamine increased UGT and GST, PhIP with ethanol decreased CYP19A1 and increased GST, and PhIP with buprenorphine decreased CAT. Nicotine 13-21 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 32-39 32113805-7 2020 Melamine and nicotine increased CYP19A1, melamine increased UGT and GST, PhIP with ethanol decreased CYP19A1 and increased GST, and PhIP with buprenorphine decreased CAT. Ethanol 83-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-108 32521813-7 2020 Sesquiterpene-mediated dysregulation of both cholesterol and lipid metabolism was observed, during which these compounds influenced the protein expression of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) and sterol regulatory element-binding protein 2 (SREBP-2), as well as the mRNA expression of HMGCR, CYP19A1, PLIN2, FASN, SCD, ACACB, and GPAM genes. Sesquiterpenes 0-13 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 307-314 32460723-11 2020 However, in women with recurrence, there was increased expression of CYP19A1 mRNA in those who had the luminal hybrid subtype and locoregional relapse and decreased expression in those negative for HER2. Phenobarbital 103-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 69-76 31757843-2 2020 The goal of the present study was to validate and optimize PET with 11C-vorozole for measuring aromatase expression in postmenopausal breast cancer in vivo. 11c-vorozole 68-80 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-104 31757843-12 2020 Conclusion: PET with 11C-vorozole is a useful technique for measuring aromatase expression in individual breast lesions, enabling noninvasive quantitative measurement of baseline and posttreatment aromatase availability in primary tumors and metastatic lesions. 11c-vorozole 21-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 70-79 31757843-12 2020 Conclusion: PET with 11C-vorozole is a useful technique for measuring aromatase expression in individual breast lesions, enabling noninvasive quantitative measurement of baseline and posttreatment aromatase availability in primary tumors and metastatic lesions. 11c-vorozole 21-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 197-206 32049373-0 2020 Mechanistic Insights into the Regio- and Stereoselectivities of Testosterone and Dihydrotestosterone Hydroxylation Catalyzed by CYP3A4 and CYP19A1. Testosterone 64-76 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-146 32049373-0 2020 Mechanistic Insights into the Regio- and Stereoselectivities of Testosterone and Dihydrotestosterone Hydroxylation Catalyzed by CYP3A4 and CYP19A1. Dihydrotestosterone 81-100 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-146 32049373-4 2020 Herein, we use a multi-scale modeling approach to investigate the selectivity of testosterone (TES) and dihydrotestosterone (DHT) hydroxylation catalyzed by two important P450s, CYP3A4 and CYP19A1. Testosterone 81-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 189-196 32049373-4 2020 Herein, we use a multi-scale modeling approach to investigate the selectivity of testosterone (TES) and dihydrotestosterone (DHT) hydroxylation catalyzed by two important P450s, CYP3A4 and CYP19A1. Testosterone 95-98 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 189-196 32049373-4 2020 Herein, we use a multi-scale modeling approach to investigate the selectivity of testosterone (TES) and dihydrotestosterone (DHT) hydroxylation catalyzed by two important P450s, CYP3A4 and CYP19A1. Dihydrotestosterone 104-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 189-196 32049373-4 2020 Herein, we use a multi-scale modeling approach to investigate the selectivity of testosterone (TES) and dihydrotestosterone (DHT) hydroxylation catalyzed by two important P450s, CYP3A4 and CYP19A1. Dihydrotestosterone 125-128 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 189-196 32049373-9 2020 Our study unravels the mechanism underlying the selectivity of TES/DHT hydroxylation mediated by CYP3A4 and CYP19A1 and is helpful for understanding the selectivity of other substrates that are hydroxylated by P450s. Testosterone 63-66 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-115 32049373-9 2020 Our study unravels the mechanism underlying the selectivity of TES/DHT hydroxylation mediated by CYP3A4 and CYP19A1 and is helpful for understanding the selectivity of other substrates that are hydroxylated by P450s. Dihydrotestosterone 67-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 108-115 31680156-6 2020 By using mouse primary granulosa cell and KGN cell line, we demonstrated that declined FOXL2 and CYP19A1 in ovarian granulosa cell partially may contributes to disturbed sex hormone synthesis in female offspring of testosterone treated mice during late gestational stage. Testosterone 215-227 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 97-104 32328497-17 2020 The effect of the heme moiety as prosthetic group of aromatase further depends on the absorption of iron as a function of pH and redox state. Heme 18-22 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 32270588-11 2020 ASA treatment reduced E2 production, Cyp19a1 expression, glutathione peroxidase (GPx) activity, and estradiol receptor expression in CGCs. Aspirin 0-3 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 37-44 32270588-12 2020 The addition of AA prevented the ASA-induced E2 reduction (p < .05) and expression of Cyp19a1. Aspirin 33-36 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-93 32267265-9 2020 The formation of O+ and ArO+ involves direct abstraction of O- from O2 by Ar2+. Oxygen 68-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-27 32267265-9 2020 The formation of O+ and ArO+ involves direct abstraction of O- from O2 by Ar2+. ar2+ 74-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 24-27 31996328-14 2020 In summary, our results suggest that miR-326 upregulate CREB and CREB may activate C/EBP-beta and later inhibited the transcription of CYP19A1 and decreased estradiol-17b production. Estradiol 157-166 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 135-142 32328497-7 2020 Articles retrieved showed that traditional antioxidation diet is one of the most approved explanations of inhibition of aromatase by phytonutrients of flavonoid derivatives. Flavonoids 151-160 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 120-129 32328497-8 2020 Flavonoids compete for the oxygen provided by the heme moiety of aromatase in the course of aromatase-catalyzed conversion of steroid precursors to estrogens. Flavonoids 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 32328497-17 2020 The effect of the heme moiety as prosthetic group of aromatase further depends on the absorption of iron as a function of pH and redox state. Iron 100-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-62 32328497-8 2020 Flavonoids compete for the oxygen provided by the heme moiety of aromatase in the course of aromatase-catalyzed conversion of steroid precursors to estrogens. Flavonoids 0-10 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 32046297-5 2020 This theoretical study investigated the active agonist and antagonist properties of "chemical classes of azoles" to determine the relationships of azole interaction with CYP19A1, using stereochemical and electronic properties of the molecules through classification and multilinear regression (MLR) modeling. Azoles 105-111 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 170-177 32328497-8 2020 Flavonoids compete for the oxygen provided by the heme moiety of aromatase in the course of aromatase-catalyzed conversion of steroid precursors to estrogens. Oxygen 27-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 32328497-8 2020 Flavonoids compete for the oxygen provided by the heme moiety of aromatase in the course of aromatase-catalyzed conversion of steroid precursors to estrogens. Oxygen 27-33 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 32328497-8 2020 Flavonoids compete for the oxygen provided by the heme moiety of aromatase in the course of aromatase-catalyzed conversion of steroid precursors to estrogens. Heme 50-54 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 32328497-8 2020 Flavonoids compete for the oxygen provided by the heme moiety of aromatase in the course of aromatase-catalyzed conversion of steroid precursors to estrogens. Heme 50-54 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 32328497-8 2020 Flavonoids compete for the oxygen provided by the heme moiety of aromatase in the course of aromatase-catalyzed conversion of steroid precursors to estrogens. Steroids 126-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 32328497-8 2020 Flavonoids compete for the oxygen provided by the heme moiety of aromatase in the course of aromatase-catalyzed conversion of steroid precursors to estrogens. Steroids 126-133 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 92-101 32328497-15 2020 Aromatase itself depends on the expression of the heme moiety encoded in the genotype. Heme 50-54 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 32004675-5 2020 These cells also showed increased 17beta-hydroxysteroid dehydrogenases types 2 (HSD17B2) relative to control group and increased expression of aromatase (CYP19) after exposure to progesterone. Progesterone 179-191 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-159 31698960-7 2020 This study showed that both classic ESR1 and GPER1 were involved in the inhibitory effect of both PAH mixtures on E2 secretion and confirmed that expression of P450arom could be downregulated through the aryl hydrocarbon receptor and additionally through the ESR2. Estradiol 114-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 160-168 32252458-3 2020 In this study, sixteen of benzimidazole-triazolothiadiazine derivatives have been synthesized and studied as potent aromatase inhibitors. benzimidazole-triazolothiadiazine 26-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 116-125 32252458-7 2020 Furthermore, compound 5e and reference drug letrozole were docked into human placental aromatase enzyme to predict their possible binding modes with the enzyme. Letrozole 44-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 87-96 31471170-10 2020 CONCLUSIONS: The transition from primary treatments to adjuvant AHT therapy with receiving a prescription for an aromatase inhibitor caught many participants off guard. Anhydrothymidine 64-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-122 31953017-6 2020 We also show that CBD prevents the increase on transcript levels of CYP19A1 gene and the elevation of E2 levels that are observed in differentiating ESCs. Cannabidiol 18-21 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 68-75 32435378-0 2020 Targeting Orthosteric and Allosteric Pockets of Aromatase via Dual-Mode Novel Azole Inhibitors. Azoles 78-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-57 32435378-5 2020 In this context, here we designed, synthesized, and performed in vitro inhibitory tests on the aromatase enzyme and distinct ER+/ER- BC cell line types of novel azole bridged xanthones. Azoles 161-166 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-104 32435378-5 2020 In this context, here we designed, synthesized, and performed in vitro inhibitory tests on the aromatase enzyme and distinct ER+/ER- BC cell line types of novel azole bridged xanthones. Xanthones 175-184 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 95-104 32073264-1 2020 An unprecedented photocatalytic system consisting of benzimidazolium aryloxide betaines (BI+-ArO-) and stoichiometric hydride reducing reagents was developed for carrying out desulfonylation reactions of N-sulfonyl-indoles, -amides and -amines, and -sulfonylketones. Betaine 53-87 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 32073264-1 2020 An unprecedented photocatalytic system consisting of benzimidazolium aryloxide betaines (BI+-ArO-) and stoichiometric hydride reducing reagents was developed for carrying out desulfonylation reactions of N-sulfonyl-indoles, -amides and -amines, and -sulfonylketones. sodium hydride 118-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 32073264-1 2020 An unprecedented photocatalytic system consisting of benzimidazolium aryloxide betaines (BI+-ArO-) and stoichiometric hydride reducing reagents was developed for carrying out desulfonylation reactions of N-sulfonyl-indoles, -amides and -amines, and -sulfonylketones. indole 204-222 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 32073264-1 2020 An unprecedented photocatalytic system consisting of benzimidazolium aryloxide betaines (BI+-ArO-) and stoichiometric hydride reducing reagents was developed for carrying out desulfonylation reactions of N-sulfonyl-indoles, -amides and -amines, and -sulfonylketones. Amides 225-231 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 32073264-1 2020 An unprecedented photocatalytic system consisting of benzimidazolium aryloxide betaines (BI+-ArO-) and stoichiometric hydride reducing reagents was developed for carrying out desulfonylation reactions of N-sulfonyl-indoles, -amides and -amines, and -sulfonylketones. Amines 237-243 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 32073264-1 2020 An unprecedented photocatalytic system consisting of benzimidazolium aryloxide betaines (BI+-ArO-) and stoichiometric hydride reducing reagents was developed for carrying out desulfonylation reactions of N-sulfonyl-indoles, -amides and -amines, and -sulfonylketones. sulfonylketones 251-266 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-96 32073264-3 2020 In the catalytic system, visible light activated benzimidazoline aryloxides (BIH-ArO-), generated in-situ by hydride reduction of the corresponding betaines BI+-ArO-, donate both an electron and a hydrogen atom to the substrates. sodium hydride 109-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-84 32073264-3 2020 In the catalytic system, visible light activated benzimidazoline aryloxides (BIH-ArO-), generated in-situ by hydride reduction of the corresponding betaines BI+-ArO-, donate both an electron and a hydrogen atom to the substrates. sodium hydride 109-116 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 161-164 32073264-3 2020 In the catalytic system, visible light activated benzimidazoline aryloxides (BIH-ArO-), generated in-situ by hydride reduction of the corresponding betaines BI+-ArO-, donate both an electron and a hydrogen atom to the substrates. Hydrogen 197-205 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 81-84 32073264-3 2020 In the catalytic system, visible light activated benzimidazoline aryloxides (BIH-ArO-), generated in-situ by hydride reduction of the corresponding betaines BI+-ArO-, donate both an electron and a hydrogen atom to the substrates. Hydrogen 197-205 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 161-164 32309755-8 2020 In functional experiments, we assessed the effects of FSH, estradiol (E2), and the aromatase inhibitor letrozole on AGCT cell viability using 2 in vitro models: KGN cells and primary cultures of AGCT cells. Letrozole 103-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 83-92 32046297-5 2020 This theoretical study investigated the active agonist and antagonist properties of "chemical classes of azoles" to determine the relationships of azole interaction with CYP19A1, using stereochemical and electronic properties of the molecules through classification and multilinear regression (MLR) modeling. Azoles 105-110 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 170-177 32046297-10 2020 This study proposes that the electron penetration of azoles toward heme group decides the binding behavior and stereochemistry requirement for antagonist activity against CYP19A1 enzyme. Azoles 53-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 171-178 32046297-10 2020 This study proposes that the electron penetration of azoles toward heme group decides the binding behavior and stereochemistry requirement for antagonist activity against CYP19A1 enzyme. Heme 67-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 171-178 31535401-0 2020 Effect of Aromatase Inhibition (Exemestane) on Urine Concentration of Osteoprotegerin in Healthy Postmenopausal Women. exemestane 32-42 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 10-19 31579960-6 2020 Supplementation of Genistein promoted the secretion of E2 and increased the expression of Star and Cyp19a1 mRNA, whereas suppressed the level of progesterone (P4 ) accompanied with a decline in the level of Hsd3b1 mRNA expression. Genistein 19-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 99-106 31579960-7 2020 H89 blocked the regulation of Genistein on the secretion of E2 and P4 , and alleviated the ascending of Star and Cyp19a1 elicited by Genistein. N-(2-(4-bromocinnamylamino)ethyl)-5-isoquinolinesulfonamide 0-3 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-120 31579960-7 2020 H89 blocked the regulation of Genistein on the secretion of E2 and P4 , and alleviated the ascending of Star and Cyp19a1 elicited by Genistein. Genistein 133-142 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 113-120 31669572-1 2020 Aromatase (CYP19A1) converts androgens into estrogens and is required for female sexual development and growth and development in both sexes. Androgens 29-38 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-18 31669572-1 2020 Aromatase (CYP19A1) converts androgens into estrogens and is required for female sexual development and growth and development in both sexes. Estrogens 44-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 11-18 31669572-2 2020 CYP19A1 is a member of cytochrome P450 family of heme-thiolate monooxygenases located in the endoplasmic reticulum and depends on reducing equivalents from the reduced nicotinamide adenine dinucleotide phosphate via the cytochrome P450 oxidoreductase coded by POR. Heme 49-62 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-7 31669572-2 2020 CYP19A1 is a member of cytochrome P450 family of heme-thiolate monooxygenases located in the endoplasmic reticulum and depends on reducing equivalents from the reduced nicotinamide adenine dinucleotide phosphate via the cytochrome P450 oxidoreductase coded by POR. NADP 168-211 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-7 31669572-3 2020 Both the CYP19A1 and POR genes are highly polymorphic, and mutations in both these genes are linked to disorders of steroid biosynthesis. Steroids 116-123 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 9-16 31535401-1 2020 This pilot study examined how exemestane (an aromatase inhibitor [AI]) affected osteoprotegerin (OPG) urine concentrations in postmenopausal women. exemestane 30-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-54 31734841-1 2020 Aromatase is the enzyme responsible for conversion of C19 androgenic steroids to the corresponding estrogens: a reaction known as aromatization. Steroids 69-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31629071-2 2020 Hs578t cells expressed aromatase (CYP19) mainly via the healthy stromal CYP19 promoter I.4, but also to a lesser extent via the breast cancer-relevant promoters PII, I.3 and I.7, and produced estrogens from androgen precursors. Estrogens 192-201 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-39 31629071-2 2020 Hs578t cells expressed aromatase (CYP19) mainly via the healthy stromal CYP19 promoter I.4, but also to a lesser extent via the breast cancer-relevant promoters PII, I.3 and I.7, and produced estrogens from androgen precursors. Androgens 207-215 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 34-39 31850443-3 2020 This methodology has also been applied to the synthesis of 2-aroyl indoles and the potent CYP19 inhibitor 1-(benzofuran-2-yl(phenyl)methyl)-1H-1,2,4-triazole. indole 59-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-95 31850443-3 2020 This methodology has also been applied to the synthesis of 2-aroyl indoles and the potent CYP19 inhibitor 1-(benzofuran-2-yl(phenyl)methyl)-1H-1,2,4-triazole. MEN 11066 106-157 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 90-95 31734841-1 2020 Aromatase is the enzyme responsible for conversion of C19 androgenic steroids to the corresponding estrogens: a reaction known as aromatization. Estrogens 99-108 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31929813-1 2019 Aromatase, a cytochrome P450 enzyme that converts androgens into estrogens, is an important drug target for hormone-dependent diseases. Androgens 50-59 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 32741938-0 2020 Evaluation of Synthesized Ester or Amide Coumarin Derivatives on Aromatase Inhibitory Activity. Esters 26-31 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 32741938-0 2020 Evaluation of Synthesized Ester or Amide Coumarin Derivatives on Aromatase Inhibitory Activity. Amides 35-40 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 32741938-0 2020 Evaluation of Synthesized Ester or Amide Coumarin Derivatives on Aromatase Inhibitory Activity. coumarin 41-49 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-74 32741938-5 2020 In this regard, we previously reported that diethylaminocoumarin dimers and thiazolyl coumarin derivatives possess strong aromatase inhibiting capabilities. diethylaminocoumarin 44-64 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-131 32741938-5 2020 In this regard, we previously reported that diethylaminocoumarin dimers and thiazolyl coumarin derivatives possess strong aromatase inhibiting capabilities. thiazolyl coumarin 76-94 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 122-131 32741938-6 2020 In this study, we further examined the structure-activity relationships of coumarin derivatives synthesized from thiazolyl coumarin derivatives and their aromatase inhibiting capabilities. coumarin 75-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-163 32741938-6 2020 In this study, we further examined the structure-activity relationships of coumarin derivatives synthesized from thiazolyl coumarin derivatives and their aromatase inhibiting capabilities. thiazolyl 113-122 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-163 32741938-6 2020 In this study, we further examined the structure-activity relationships of coumarin derivatives synthesized from thiazolyl coumarin derivatives and their aromatase inhibiting capabilities. coumarin 123-131 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 154-163 32741938-7 2020 Consequently, amide coumarin N-benzhydryl-7-(diethylamino)-2-oxo-2H-chromene-3-carboxamide (IC50 values 4.5 microM) is inhibitor of aromatase. amide coumarin n-benzhydryl-7-(diethylamino)-2-oxo-2h-chromene-3-carboxamide 14-90 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 132-141 32741938-8 2020 This inhibitor was found to be comparable aromatase inhibitory activity to the 1st generation aromatase inhibitor aminoglutethimide (3.2 microM). Aminoglutethimide 114-131 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 94-103 32741938-9 2020 Substitution of the amide group on the amide coumarin derivative affects the aromatase inhibiting activity. coumarin 45-53 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 77-86 31645096-3 2019 Chemoselectivity of various O,S-nucleophiles toward difluorocarbene was systematically studied, suggesting the reactivity order ArS- > RS-, ArO- > ROH > RO-, ArSH, ArOH, RSH. difluorocarbene 52-67 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 140-143 31936362-8 2020 Interestingly, the expression of StAR and CYP19A1 in the CIPGCs (-Dox) was significantly increased by adding porcine follicular fluid (PFF) to mimic an ovary follicle environment. 1-deoxysphinganine 66-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 42-49 31786088-1 2020 Aromatase is the requisite and limiting enzyme in the production of estrogens from androgens. Estrogens 68-77 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31786088-1 2020 Aromatase is the requisite and limiting enzyme in the production of estrogens from androgens. Androgens 83-92 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31929813-1 2019 Aromatase, a cytochrome P450 enzyme that converts androgens into estrogens, is an important drug target for hormone-dependent diseases. Estrogens 65-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31929813-3 2019 In this study, the inhibitory effect of MHT on aromatase activity was observed using dibenzylfluorescein (DBF) and KGN cells, and the dose-dependent effect of MHT was verified (IC50 values of 251 mug/mL and 246 mug/mL as determined by the two methods, respectively). dibenzylfluorescein 85-104 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 31929813-3 2019 In this study, the inhibitory effect of MHT on aromatase activity was observed using dibenzylfluorescein (DBF) and KGN cells, and the dose-dependent effect of MHT was verified (IC50 values of 251 mug/mL and 246 mug/mL as determined by the two methods, respectively). dibenzylfluorescein 106-109 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 47-56 31374317-0 2019 Bisphenol A affects estradiol metabolism by targeting CYP1A1 and CYP19A1 in human placental JEG-3 cells. bis(4-hydroxyphenyl)sulfone 0-9 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-72 31536780-5 2019 Furthermore, we provided mechanistic insight into the ability of estragole and trans-anethole to stimulate promoter-specific expression of CYP19 through activation of the PKA pathway in H295R cells and the PKC pathway in BeWo cells, in both cases associated with increased cAMP levels. estragole 65-74 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-144 31536780-5 2019 Furthermore, we provided mechanistic insight into the ability of estragole and trans-anethole to stimulate promoter-specific expression of CYP19 through activation of the PKA pathway in H295R cells and the PKC pathway in BeWo cells, in both cases associated with increased cAMP levels. anethole 79-93 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-144 31536780-5 2019 Furthermore, we provided mechanistic insight into the ability of estragole and trans-anethole to stimulate promoter-specific expression of CYP19 through activation of the PKA pathway in H295R cells and the PKC pathway in BeWo cells, in both cases associated with increased cAMP levels. Cyclic AMP 273-277 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 139-144 31454668-7 2019 In cells exposed to a differentiation stimulus, AEA-treatment prevents the increase of the expression of CYP19A1 gene encoding aromatase, E2 levels and of estradiol receptor expression, that are observed in differentiating cells. anandamide 48-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 105-112 31509772-3 2019 More specifically, serotonin (5-HT) regulates the estrogen biosynthetic enzyme aromatase (cytochrome P450 19; CYP19), which is disrupted by fluoxetine and its active metabolite norfluoxetine in BeWo choriocarcinoma cells. Serotonin 19-28 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 110-115 31509772-3 2019 More specifically, serotonin (5-HT) regulates the estrogen biosynthetic enzyme aromatase (cytochrome P450 19; CYP19), which is disrupted by fluoxetine and its active metabolite norfluoxetine in BeWo choriocarcinoma cells. Serotonin 30-34 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 110-115 31509772-3 2019 More specifically, serotonin (5-HT) regulates the estrogen biosynthetic enzyme aromatase (cytochrome P450 19; CYP19), which is disrupted by fluoxetine and its active metabolite norfluoxetine in BeWo choriocarcinoma cells. Fluoxetine 140-150 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 110-115 31509772-3 2019 More specifically, serotonin (5-HT) regulates the estrogen biosynthetic enzyme aromatase (cytochrome P450 19; CYP19), which is disrupted by fluoxetine and its active metabolite norfluoxetine in BeWo choriocarcinoma cells. norfluoxetine 177-190 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 110-115 31509772-4 2019 Based on molecular simulations, the present study illustrates that the SRIs fluoxetine, norfluoxetine, paroxetine, sertraline, citalopram and venlafaxine exhibit binding affinity for the active-site pocket of CYP19, suggesting potential competitive inhibition. Fluoxetine 76-86 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 209-214 31509772-4 2019 Based on molecular simulations, the present study illustrates that the SRIs fluoxetine, norfluoxetine, paroxetine, sertraline, citalopram and venlafaxine exhibit binding affinity for the active-site pocket of CYP19, suggesting potential competitive inhibition. norfluoxetine 88-101 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 209-214 31509772-4 2019 Based on molecular simulations, the present study illustrates that the SRIs fluoxetine, norfluoxetine, paroxetine, sertraline, citalopram and venlafaxine exhibit binding affinity for the active-site pocket of CYP19, suggesting potential competitive inhibition. Paroxetine 103-113 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 209-214 31509772-4 2019 Based on molecular simulations, the present study illustrates that the SRIs fluoxetine, norfluoxetine, paroxetine, sertraline, citalopram and venlafaxine exhibit binding affinity for the active-site pocket of CYP19, suggesting potential competitive inhibition. Sertraline 115-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 209-214 31509772-4 2019 Based on molecular simulations, the present study illustrates that the SRIs fluoxetine, norfluoxetine, paroxetine, sertraline, citalopram and venlafaxine exhibit binding affinity for the active-site pocket of CYP19, suggesting potential competitive inhibition. Citalopram 127-137 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 209-214 31509772-4 2019 Based on molecular simulations, the present study illustrates that the SRIs fluoxetine, norfluoxetine, paroxetine, sertraline, citalopram and venlafaxine exhibit binding affinity for the active-site pocket of CYP19, suggesting potential competitive inhibition. Venlafaxine Hydrochloride 142-153 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 209-214 31509772-8 2019 We also showed that 5-HT and the 5-HT2A receptor agonist 2,5-dimethoxy-4-iodoamphetamine (DOI) induced CYP19 activity. Serotonin 20-24 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 103-108 31509772-8 2019 We also showed that 5-HT and the 5-HT2A receptor agonist 2,5-dimethoxy-4-iodoamphetamine (DOI) induced CYP19 activity. 4-iodo-2,5-dimethoxyphenylisopropylamine 57-88 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 103-108 31509772-9 2019 An increase in phosphorylation of serine and tyrosine and a decrease in threonine phosphorylation of CYP19 was also associated with DOI treatment. Threonine 72-81 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 101-106 31509772-10 2019 Our results contribute to better understanding how 5-HT and SRIs interact with CYP19 and may affect estrogen production. Serotonin 51-55 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 79-84 31374317-0 2019 Bisphenol A affects estradiol metabolism by targeting CYP1A1 and CYP19A1 in human placental JEG-3 cells. Estradiol 20-29 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 65-72 31374317-4 2019 We observed that BPA significantly reduced estradiol level of JEG-3 cells in a dose-dependent manner, which was accompanied by an increase in CYP1A1 protein level and an inhibition of CYP19A1 protein level. bisphenol A 17-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 184-191 31374317-6 2019 Meanwhile, estradiol was almost undetectable in CYP19A1 knockdown group. Estradiol 11-20 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-55 31374317-7 2019 On the contrary, the group with over-expression of CYP19A1 gene increased estradiol level by 8.6 fold while the CYP1A1 knockdown group increased by 5.6 fold. Estradiol 74-83 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 51-58 31374317-8 2019 In summary, our research clearly showed that BPA alters JEG-3 estradiol synthesis and catabolism due to its action on CYP1A1 and CYP19A1 protein levels and may interfere with the normal process of placenta formation and embryonic development during early pregnancy. bisphenol A 45-48 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-136 31374317-8 2019 In summary, our research clearly showed that BPA alters JEG-3 estradiol synthesis and catabolism due to its action on CYP1A1 and CYP19A1 protein levels and may interfere with the normal process of placenta formation and embryonic development during early pregnancy. Estradiol 62-71 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 129-136 31741086-1 2019 Aberrant expression or hyperactivation of aromatase (CYP19A1)-estrogen receptor (ESR) axis is well identified as one of the major causes of breast cancer. Estrogens 62-70 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 53-60 31741086-2 2019 Lots of drugs have been developed for targeting CYP19A1 or ESR respectively, such as anastrozole and fulvestrant. anastrozole 85-96 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-55 31741086-2 2019 Lots of drugs have been developed for targeting CYP19A1 or ESR respectively, such as anastrozole and fulvestrant. ici 182,780 101-112 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 48-55 31737354-1 2019 Aromatase and seladin-1 are enzymes that have major roles in estrogen synthesis and are important in both brain physiology and pathology. Estrogens 61-69 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31737354-2 2019 Aromatase is the key enzyme that catalyzes estrogen biosynthesis from androgen precursors and regulates the brain"s neurosteroidogenic activity. Estrogens 43-51 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31737354-2 2019 Aromatase is the key enzyme that catalyzes estrogen biosynthesis from androgen precursors and regulates the brain"s neurosteroidogenic activity. Androgens 70-78 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31737354-6 2019 Therefore, the expression of local brain aromatase and seladin-1 is important, as they produce neuroactive steroids in the brain for the protection of neuronal damage. Steroids 107-115 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 41-50 31683864-5 2019 In this study, serum estradiol (E2) concentration and the expressions of Bmal1, Lepr, Cyp19a1, and Cyp11a1 genes were found to display well-synchronized circadian rhythms. Estradiol 21-30 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 86-93 31772710-5 2019 Results: Melatonin triggered upregulation of cytochrome P450 family 19 subfamily A member 1 (CYP19A1) expression via the extracellular signal-regulated kinase pathway in luteinized granulosa cells. Melatonin 9-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 45-91 31772710-5 2019 Results: Melatonin triggered upregulation of cytochrome P450 family 19 subfamily A member 1 (CYP19A1) expression via the extracellular signal-regulated kinase pathway in luteinized granulosa cells. Melatonin 9-18 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 93-100 31553790-0 2019 Human chorionic gonadotropin-induced amphiregulin stimulates aromatase expression in human granulosa-lutein cells: a mechanism for estradiol production in the luteal phase. Estradiol 131-140 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 61-70 31553790-6 2019 Aromatase is an enzyme responsible for a key step in the biosynthesis of E2. Estradiol 73-75 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 0-9 31641693-1 2019 Background: The effect of extended adjuvant aromatase inhibition in hormone-positive breast cancer after sequential tamoxifen, aromatase inhibitor treatment of 5 years was recently investigated by the DATA study. Tamoxifen 116-125 cytochrome P450 family 19 subfamily A member 1 Homo sapiens 44-53