151
|
Lilienkampf A, Karkola S, Alho-Richmond S, Koskimies P, Johansson N, Huhtinen K, Vihko K, Wähälä K. Synthesis and Biological Evaluation of 17β-Hydroxysteroid Dehydrogenase Type 1 (17β-HSD1) Inhibitors Based on a Thieno[2,3-d]pyrimidin-4(3H)-one Core. J Med Chem 2009; 52:6660-71. [DOI: 10.1021/jm900928k] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Annamaria Lilienkampf
- Laboratory of Organic Chemistry, Department of Chemistry, P.O. Box 55, FIN-00014 University of Helsinki, Finland
| | - Sampo Karkola
- Laboratory of Organic Chemistry, Department of Chemistry, P.O. Box 55, FIN-00014 University of Helsinki, Finland
| | - Sari Alho-Richmond
- Laboratory of Organic Chemistry, Department of Chemistry, P.O. Box 55, FIN-00014 University of Helsinki, Finland
| | - Pasi Koskimies
- Hormos Medical Ltd., PharmaCity, FIN-20520 Turku, Finland
| | - Nina Johansson
- Hormos Medical Ltd., PharmaCity, FIN-20520 Turku, Finland
| | - Kaisa Huhtinen
- Hormos Medical Ltd., PharmaCity, FIN-20520 Turku, Finland
| | - Kimmo Vihko
- Hormos Medical Ltd., PharmaCity, FIN-20520 Turku, Finland
| | - Kristiina Wähälä
- Laboratory of Organic Chemistry, Department of Chemistry, P.O. Box 55, FIN-00014 University of Helsinki, Finland
| |
Collapse
|
152
|
Elam MB, Cowan GS, Rooney RJ, Hiler ML, Yellaturu CR, Deng X, Howell GE, Park EA, Gerling IC, Patel D, Corton JC, Cagen LM, Wilcox HG, Gandhi M, Bahr MH, Allan MC, Wodi LA, Cook GA, Hughes TA, Raghow R. Hepatic gene expression in morbidly obese women: implications for disease susceptibility. Obesity (Silver Spring) 2009; 17:1563-73. [PMID: 19265796 DOI: 10.1038/oby.2009.49] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The objective of this study was to determine the molecular bases of disordered hepatic function and disease susceptibility in obesity. We compared global gene expression in liver biopsies from morbidly obese (MO) women undergoing gastric bypass (GBP) surgery with that of women undergoing ventral hernia repair who had experienced massive weight loss (MWL) following prior GBP. Metabolic and hormonal profiles were examined in MO vs. MWL groups. Additionally, we analyzed individual profiles of hepatic gene expression in liver biopsy specimens obtained from MO and MWL subjects. All patients underwent preoperative metabolic profiling. RNAs were extracted from wedge biopsies of livers from MO and MWL subjects, and analysis of mRNA expression was carried out using Affymetrix HG-U133A microarray gene chips. Genes exhibiting greater than twofold differential expression between MO and MWL subjects were organized according to gene ontology and hierarchical clustering, and expression of key genes exhibiting differential regulation was quantified by real-time-polymerase chain reaction (RT-PCR). We discovered 154 genes to be differentially expressed in livers of MWL and MO subjects. A total of 28 candidate disease susceptibility genes were identified that encoded proteins regulating lipid and energy homeostasis (PLIN, ENO3, ELOVL2, APOF, LEPR, IGFBP1, DDIT4), signal transduction (MAP2K6, SOCS-2), postinflammatory tissue repair (HLA-DQB1, SPP1, P4HA1, LUM), bile acid transport (SULT2A, ABCB11), and metabolism of xenobiotics (GSTT2, CYP1A1). Using gene expression profiling, we have identified novel candidate disease susceptibility genes whose expression is altered in livers of MO subjects. The significance of altered expression of these genes to obesity-related disease is discussed.
Collapse
Affiliation(s)
- Marshall B Elam
- Department of Medicine and Research Service, Veterans Affairs Medical Center, Memphis, Tennessee, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Zheng X, Boerboom D, Carrière PD. Transforming growth factor-β1 inhibits luteinization and promotes apoptosis in bovine granulosa cells. Reproduction 2009; 137:969-77. [DOI: 10.1530/rep-08-0365] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We have previously shown that TGFB1 inhibits estradiol (E2) and progesterone (P4) biosynthesis in FSH-stimulated bovine granulosa cells by selective inhibition of steroidogenic enzymes. The objective of this study was to assess the effects of TGFB1 on E2and P4steroidogenesis in bovine granulosa cells cultured in the absence of FSH and to measure the effects of TGFB1 on cell proliferation and apoptosis in the presence and absence of FSH. Bovine granulosa cells from 2 to 5 mm follicles were cultured in serum-free medium for 2–6 days. In the absence of FSH, the secretion of P4increased with time in culture (P<0.05). Addition of TGFB1 for 6 days decreased P4secretion and mRNA levels of the P4synthesis-associated genesSTAR,CYP11A1,HSD3B1, andGSTA(P<0.05). In the absence of FSH, the secretion of E2decreased and addition of TGFB1 for 6 days partially reversed this decline and stimulated E2biosynthesis,CYP19A1andHSD17B1mRNA levels and CYP19A1 activity (P<0.05). Conversely, TGFB1 did not affectHSD17B7expression and HSD17B-reducing activity. TGFB1 decreased the proportion of cells in the G0/G1 and S+G2/M phases in FSH-stimulated and unstimulated granulosa cells (P<0.05). Furthermore, in the presence or absence of FSH, TGFB1 increased the proportion of cells in apoptosis measured by propidium iodide staining and flow cytometry and confirmed by increased levels of cleaved caspase-3 (P<0.05). Our results therefore indicate that TGFB1 inhibits luteinization in cultured bovine granulosa cells while maintaining an estrogenic phenotype, and this effect was associated with increased apoptosis.
Collapse
|
154
|
Lyssimachou A, Ramón M, Porte C. Comparative study on the metabolism of the androgen precursor androstenedione in two gastropod species: in vitro alterations by TBT and TPT. Comp Biochem Physiol C Toxicol Pharmacol 2009; 149:409-13. [PMID: 18849009 DOI: 10.1016/j.cbpc.2008.09.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Revised: 09/26/2008] [Accepted: 09/26/2008] [Indexed: 01/09/2023]
Abstract
A comparative study was performed to assess the metabolism of the androgen precursor androstenedione (AD) in two gastropod species from the Muricidae family: Bolinus brandaris and Hexaplex trunculus. AD was mainly converted to 5alpha-dihydrotestosterone by microsomal fractions isolated from Bolinus brandaris, whereas it was primarily metabolized to testosterone by Hexaplex trunculus. Sex differences in the metabolism of AD were only detected in Bolinus brandaris and attributed to higher 5alpha-reductase activity in males. Thereafter, the effect of the organotin compounds, tributyltin (TBT) and triphenyltin (TPT), on the metabolism of AD was investigated. A significant interference was only detected in females, and differences between the modes of action of the two compounds were observed: TPT was a strong inhibitor of 5alpha-reductase activity in B. brandaris at a concentration as low as 100 nM whereas only TBT (10 microM) altered the metabolism of AD in H. trunculus by increasing the activity 17beta-hydroxysteroid dehydrogenase (17beta-HSD). Thus, this work shows that the metabolism of the androgen precursor AD strongly differs among gastropod species, both in terms of activity and metabolic profile, and further demonstrates the ability of TBT and TPT to interfere with key enzymatic pathways involved in androgen synthesis.
Collapse
Affiliation(s)
- Angeliki Lyssimachou
- Environmental Chemistry Department, IIQAB-CSIC, Jordi Girona 18, 08034 Barcelona, Spain
| | | | | |
Collapse
|
155
|
Nagasaki S, Miki Y, Akahira JI, Suzuki T, Sasano H. 17beta-hydroxysteroid dehydrogenases in human breast cancer. Ann N Y Acad Sci 2009; 1155:25-32. [PMID: 19250189 DOI: 10.1111/j.1749-6632.2008.03682.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Estrogen plays a pivotal role in development and progression of human breast carcinoma. Before menopause the main source of estrogen in women is circulating estrogen secreted from the ovary, but following menopause the source changes to the hormone that is converted from circulating adrenal androgens in peripheral tissues. Therefore, adrenal androgens have to be converted to estrogen to stimulate breast carcinoma cells. In these steps, several enzymes such as aromatase, steroid sulfatase, and 17beta-hydroxysteroid dehydrogenases (17beta-HSDs) are involved in the production of estrogens. The reaction related to 17beta-HSDs activity is one of the last steps of estradiol biosynthesis, and 14 isozymes of 17beta-HSD have been identified at this juncture. The balance of the relative expression levels of 17beta-HSD isozymes in human breast carcinomas is thought to play a pivotal role in supply of estradiol to estrogen receptor positive carcinoma cells. Understanding the character of 17beta-HSD isozymes in human breast carcinoma thus provides important information on the mechanisms of biosynthesis of estradiol in breast carcinoma and for development of a therapeutic agent targeted for inhibition of local estradiol synthesis in breast carcinoma cells. In the present review we summarize the roles played by 17beta-HSDs in human breast carcinoma to obtain a better understanding of the properties of 17beta-HSDs in human breast carcinoma.
Collapse
Affiliation(s)
- Shuji Nagasaki
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | |
Collapse
|
156
|
Smuc T, Hevir N, Ribic-Pucelj M, Husen B, Thole H, Rizner TL. Disturbed estrogen and progesterone action in ovarian endometriosis. Mol Cell Endocrinol 2009; 301:59-64. [PMID: 18762229 DOI: 10.1016/j.mce.2008.07.020] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2008] [Revised: 07/22/2008] [Accepted: 07/22/2008] [Indexed: 12/21/2022]
Abstract
Endometriosis is a very common disease in pre-menopausal women, where defective metabolism of steroid hormones plays an important role in its development and promotion. In the present study, we have examined the expression of 11 estrogen and progesterone metabolizing enzymes and their corresponding receptors in samples of ovarian endometriomas and control endometrium. Expression analysis revealed significant up-regulation of enzymes involved in estradiol formation (aromatase, sulfatase and all reductive 17beta-hydroxysteroid dehydrogenases) and in progesterone inactivation (AKR1C1 and AKR1C3). Among the estrogen and progesterone receptors, ERalpha was down-regulated, ERbeta was up-regulated, and there was no significant difference in expression of progesterone receptors A and B (PRAB). Our data indicate that several enzymes of estrogen and progesterone metabolism are aberrantly expressed in endometriosis, which can lead to increased local levels of mitogenic estradiol and decreased levels of protective progesterone. Changes in estrogen receptor expression suggest that estradiol may also act via non-estrogen receptor-mediated pathways, while expression of progesterone receptors still needs further investigation.
Collapse
Affiliation(s)
- Tina Smuc
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia.
| | | | | | | | | | | |
Collapse
|
157
|
Aka JA, Mazumdar M, Lin SX. Reductive 17beta-hydroxysteroid dehydrogenases in the sulfatase pathway: critical in the cell proliferation of breast cancer. Mol Cell Endocrinol 2009; 301:183-90. [PMID: 19038308 DOI: 10.1016/j.mce.2008.10.042] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 10/10/2008] [Accepted: 10/10/2008] [Indexed: 01/30/2023]
Abstract
Estradiol, the most potent estrogen, plays critical roles in tumor cell proliferation and breast cancer development. It can be synthesized via the aromatase pathway or the sulfatase pathway, and the later has been demonstrated to be more significant. Reductive 17beta-hydroxysteroid dehydrogenases (17beta-HSDs) catalyze the last step in estrogen activation and are thus critical in breast cancer development. 17beta-HSD Type 1 (17beta-HSD1) is of great importance since it efficiently synthesizes the most potent estrogen estradiol, as well as other estrogens as 5-androstene-3beta,17beta-diol and 5alpha-androstane-3beta,17beta-diol, and inactivates the most active androgen dihydrotestosterone (DHT), all contributing to the stimulation and development of breast cancers. Rational inhibitor design based on the new structure information has been developed, yielding interesting compounds and lead chemicals. This was demonstrated by a hybrid inhibitor that interacts with both the substrate and cofactor binding sites and a recently designed inhibitor 3-(3',17'beta-dihydroxyestra-1',3',5'(10')-trien-16'beta-methyl) benzamide which has been crystallized in complex with 17beta-HSD1. Both inhibitors demonstrate nM level K(i)in vitro. New non-steroidal inhibitors have been designed and reported very recently. The Type 7 17beta-HSD, expressed in several tissues including breast and ovary, can also contribute to estrogen synthesis and DHT inactivation in breast cancer cells. The enzyme role in steroid metabolism and cancer cell proliferation needs to be compared to that in cholesterogenesis. Breast cancer cell lines provide an excellent platform for such study. T47D, MCF-7 and MDA-MB-231-luc cells have been used to create xenografts in nude mice as animal models, now with the possibility of bioluminescent imaging to provide rapid, non-invasive, and quantitative analysis of tumor biomass and metastasis. Here we review the roles of the sulfatase and aromatase pathways and the contribution of the reductive 17beta-HSDs for hormone metabolism in breast cancer.
Collapse
Affiliation(s)
- Juliette A Aka
- Laboratory of Molecular Endocrinology and Oncology, CHUL Research Center (CHUQ) and Laval University, Quebec, Canada G1V 4G2
| | | | | |
Collapse
|
158
|
Rasiah KK, Gardiner-Garden M, Padilla EJD, Möller G, Kench JG, Alles MC, Eggleton SA, Stricker PD, Adamski J, Sutherland RL, Henshall SM, Hayes VM. HSD17B4 overexpression, an independent biomarker of poor patient outcome in prostate cancer. Mol Cell Endocrinol 2009; 301:89-96. [PMID: 19100308 DOI: 10.1016/j.mce.2008.11.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2008] [Revised: 11/17/2008] [Accepted: 11/18/2008] [Indexed: 01/27/2023]
Abstract
Steroid hormones and their metabolising enzymes have been studied extensively for their potential role in prostate cancer, with more recent interest in the androgen/estrogen inactivating enzyme 17beta-hydroxysteroid dehydrogenase type 4 (HSD17B4). Gene expression profiling showed HSD17B4 to be significantly overexpressed in prostate cancer compared to matched-benign epithelium. We therefore hypothesized that altered HSD17B4 expression may contribute to prostate cancer progression via altered hormone balance. In this study, HSD17B4 mRNA and protein expression were assessed by in situ hybridisation (ISH) and immunohistochemistry (IHC), respectively, in tissue arrays of prostate tissue from 172 patients treated by radical prostatectomy. Overexpression of HSD17B4 mRNA and protein was associated with prostate cancer (P<0.0001) and multivariate Cox proportional hazards analysis, adjusted for known prognostic indicators, demonstrated HSD17B4 mRNA and high protein expression were significant independent predictors of poor patient outcome as measured by time until PSA relapse (mRNA: hazards ratio [HR]=1.90, 95% confidence interval [CI]=1.15-3.12; P<0.0001; and protein: HR=2.09, 95% CI=1.31-3.33; P=0.0026). Here we provide strong evidence that both mRNA and protein overexpression of HSD17B4 is not only associated with the presence of prostate cancer, but is also a significant independent predictor of poor patient outcome.
Collapse
Affiliation(s)
- Krishan K Rasiah
- Cancer Research Program, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst, Sydney, NSW, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
159
|
Lamminen T, Saloniemi T, Huhtinen K, Koskimies P, Messinger J, Husen B, Thole H, Poutanen M. In vivo mouse model for analysis of hydroxysteroid (17beta) dehydrogenase 1 inhibitors. Mol Cell Endocrinol 2009; 301:158-62. [PMID: 19026716 DOI: 10.1016/j.mce.2008.10.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Revised: 10/22/2008] [Accepted: 10/22/2008] [Indexed: 11/19/2022]
Abstract
Hydroxysteroid (17beta) dehydrogenase 1 (HSD17B1) catalyzes the reaction between the low active 17-ketosteroids and the highly active 17beta-hydroxysteroids. In the present study, we have generated transgenic (TG) mice expressing human (h) HSD17B1 under mouse mammary tumor virus (MMTV) promoter (MMTV-hHSD17B1TG mice). The MMTV-hHSD17B1TG mice were used to characterize HSD17B1 enzyme activity and properties of HSD17B1 inhibitor in vivo. Expression of the transgene was detected by enzyme activity and RT-PCR analysis. Increased HSD17B1 activity in the TG mice was detected in vivo by applying estrone as a substrate via an intravenous injection. The developed enzyme activity measurement was then applied to analyze the efficacy of HSD17B1 inhibitor in vivo. The results indicated that the MMTV-hHSD17B1TG mouse model is a valuable novel tool to test human HSD17B1 inhibition by various compounds in vivo. With the potent hHSD17B1 inhibitor compound tested, at highest an 85% and 33% inhibition of the enzyme activity in males and in females, respectively, was observed.
Collapse
Affiliation(s)
- Tarja Lamminen
- Department of Physiology, Institute of Biomedicine, FIN-20014 University of Turku, Kiinamyllynkatu 10, Turku, Finland
| | | | | | | | | | | | | | | |
Collapse
|
160
|
Tarrant AM, Reitzel AM, Blomquist CH, Haller F, Tokarz J, Adamski J. Steroid metabolism in cnidarians: insights from Nematostella vectensis. Mol Cell Endocrinol 2009; 301:27-36. [PMID: 18984032 DOI: 10.1016/j.mce.2008.09.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2008] [Revised: 09/30/2008] [Accepted: 09/30/2008] [Indexed: 11/17/2022]
Abstract
Cnidarians occupy a key evolutionary position as a sister group to bilaterian animals. While cnidarians contain a diverse complement of steroids, sterols, and other lipid metabolites, relatively little is known of the endogenous steroid metabolism or function in cnidarian tissues. Incubations of cnidarian tissues with steroid substrates have indicated the presence of steroid metabolizing enzymes, particularly enzymes with 17beta-hydroxysteroid dehydrogenase (17beta-HSD) activity. Through analysis of the genome of the starlet sea anemone, Nematostella vectensis, we identified a suite of genes in the short chain dehydrogenase/reductase (SDR) superfamily including homologs of genes that metabolize steroids in other animals. A more detailed analysis of Hsd17b4 revealed complex evolutionary relationships, apparent intron loss in several taxa, and predominantly adult expression in N. vectensis. Due to its ease of culture and available molecular tools N. vectensis is an excellent model for investigation of cnidarian steroid metabolism and gene function.
Collapse
Affiliation(s)
- Ann M Tarrant
- Biology Department, Woods Hole Oceanographic Institution, 45 Water Street, Woods Hole, MA 02543, USA.
| | | | | | | | | | | |
Collapse
|
161
|
Delvoux B, Groothuis P, D'Hooghe T, Kyama C, Dunselman G, Romano A. Increased production of 17beta-estradiol in endometriosis lesions is the result of impaired metabolism. J Clin Endocrinol Metab 2009; 94:876-83. [PMID: 19088158 DOI: 10.1210/jc.2008-2218] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
CONTEXT substantial evidence suggests that the expression of steroid metabolizing enzymes in endometriosis is altered, turning the ectopic endometrium into a source of 17beta-estradiol. However, whether these differences result in a net increase in local 17beta-estradiol production/activity has not been shown. SUBJECTS AND METHODS The activities of the most important steroidogenic enzymes synthesizing and inactivating 17beta-estradiol were determined by HPLC in matched eutopic and ectopic tissue from patients with endometriosis (n = 14) and in endometrium from controls (n = 20). RESULTS Aromatase activity is negligible in the ectopic endometrium, whereas the activity of estrogen sulfatase is high though not different between ectopic, eutopic and control endometrium. The activity of 17beta-hydroxysteroid dehydrogenases (17beta-HSDs) converting estrone into 17beta-estradiol is higher in the ectopic compared to the eutopic endometrium in patients. The activity of 17beta-HSDs converting 17beta-estradiol back to estrone is significantly lower in the ectopic compared to the eutopic endometrium of both patients and controls. To evaluate the net metabolic capacity of tissues to synthesize 17beta-estradiol, we calculated the activity ratio between 17beta-HSDs synthesizing versus 17beta-HSDs inactivating 17beta-estradiol. This ratio is significantly higher in the ectopic compared to the eutopic endometrium of patients and controls, indicating a high synthesis of 17beta-estradiol in the ectopic locations. This is further supported by the elevated mRNA levels of the estrogen-responsive gene TFF1 in all ectopic compared to eutopic endometria. CONCLUSION Endometriotic lesions have higher production of 17beta-estradiol than the eutopic endometrium of patients and controls. This is mostly the result of impaired metabolism.
Collapse
Affiliation(s)
- Bert Delvoux
- GROW, School for Oncology and Developmental Biology, University Maastricht, and Department of Obstetrics and Gynecology, University Hospital Maastricht, 6202 AZ Maastricht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
162
|
Meier M, Möller G, Adamski J. Perspectives in Understanding the Role of Human 17β-Hydroxysteroid Dehydrogenases in Health and Disease. Ann N Y Acad Sci 2009; 1155:15-24. [DOI: 10.1111/j.1749-6632.2009.03702.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
163
|
Middelbos IS, Vester BM, Karr-Lilienthal LK, Schook LB, Swanson KS. Age and diet affect gene expression profile in canine skeletal muscle. PLoS One 2009; 4:e4481. [PMID: 19221602 PMCID: PMC2637985 DOI: 10.1371/journal.pone.0004481] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Accepted: 01/07/2009] [Indexed: 11/19/2022] Open
Abstract
We evaluated gene transcription in canine skeletal muscle (biceps femoris) using microarray analysis to identify effects of age and diet on gene expression. Twelve female beagles were used (six 1-year olds and six 12-year olds) and they were fed one of two experimental diets for 12 months. One diet contained primarily plant-based protein sources (PPB), whereas the second diet contained primarily animal-based protein sources (APB). Affymetrix GeneChip Canine Genome Arrays were used to hybridize extracted RNA. Age had the greatest effect on gene transcription (262 differentially expressed genes), whereas the effect of diet was relatively small (22 differentially expressed genes). Effects of age (regardless of diet) were most notable on genes related to metabolism, cell cycle and cell development, and transcription function. All these genes were predominantly down-regulated in geriatric dogs. Age-affected genes that were differentially expressed on only one of two diets were primarily noted in the PPB diet group (144/165 genes). Again, genes related to cell cycle (22/35) and metabolism (15/19) had predominantly decreased transcription in geriatric dogs, but 6/8 genes related to muscle development had increased expression. Effects of diet on muscle gene expression were mostly noted in geriatric dogs, but no consistent patterns in transcription were observed. The insight these data provide into gene expression profiles of canine skeletal muscle as affected by age, could serve as a foundation for future research pertaining to age-related muscle diseases.
Collapse
Affiliation(s)
- Ingmar S. Middelbos
- Department of Animal Sciences, University of Illinois, Urbana, Illinois, United States of America
| | - Brittany M. Vester
- Department of Animal Sciences, University of Illinois, Urbana, Illinois, United States of America
| | - Lisa K. Karr-Lilienthal
- Department of Animal Sciences, University of Illinois, Urbana, Illinois, United States of America
| | - Lawrence B. Schook
- Department of Animal Sciences, University of Illinois, Urbana, Illinois, United States of America
- Division of Nutritional Sciences, University of Illinois, Urbana, Illinois, United States of America
| | - Kelly S. Swanson
- Department of Animal Sciences, University of Illinois, Urbana, Illinois, United States of America
- Division of Nutritional Sciences, University of Illinois, Urbana, Illinois, United States of America
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana, Illinois, United States of America
| |
Collapse
|
164
|
Putti R, Varricchio E, Gay F, Elena C, Paolucci M. Leptin effects on testis and epididymis in the lizard Podarcis sicula, during summer regression. Gen Comp Endocrinol 2009; 160:168-75. [PMID: 19056389 DOI: 10.1016/j.ygcen.2008.11.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Revised: 11/03/2008] [Accepted: 11/14/2008] [Indexed: 10/21/2022]
Abstract
In this study we assessed the effect of leptin treatment on testicular morphology, spermatogenesis, Peroxisome Proliferator Activated Receptor (PPAR) alpha, 17beta-hydroxysteroide dehydrogenase, 17beta-estradiol and testosterone levels in the testis and blood of the lizard Podarcis sicula at the beginning of summer regression before entering the refractory period, when lizards no longer respond to hormonal and environmental stimuli. Lizards treated with five injections of leptin showed seminiferous tubules with germinal cells at all stages and wider lumina with respect to the controls. After 10 injections, the diameter of the lumina increased compared to the controls and 5 injection-group. After 10 injections plus 20 days before the sacrifice, the seminiferous tubules with open lumina and germinal cells were less abundant than in the 5 and 10 injection-groups. In all groups, the epididymis epithelium was higher than in the controls, with mitosis and binucleated cells. In both the control and treated animals secondary spermatocytes and spermatids were immunoreactive to leptin receptor and PPARalpha. In treated animals the interstitial cells and peritubular fibrocytes were also leptin receptor immunoreactive, while PPARalpha immunoreactivity translocated from the cytoplasm to the nucleus. 17beta-HSD immunoreactivity was present in the spermatids and interstitial cells of control lizards and in secondary spermatocytes and spermatids of treated lizards. Leptin treatment had no statistically significant effect on testicular and circulating 17beta-estradiol and testosterone levels. These observations indicate that leptin brings about a delay in testis summer regression in Podarcis sicula, playing a regulatory role in reproduction in this species as already hypothesized for mammals.
Collapse
Affiliation(s)
- Rosalba Putti
- Department of Comparative and Evolutionary Biology, Faculty of Sciences, University of Naples, Federico II, Via Mezzocannone, 8, 80134 Napoli, Italy
| | | | | | | | | |
Collapse
|
165
|
Sato R, Suzuki T, Katayose Y, Miura K, Shiiba K, Tateno H, Miki Y, Akahira J, Kamogawa Y, Nagasaki S, Yamamoto K, Ii T, Egawa S, Evans DB, Unno M, Sasano H. Steroid sulfatase and estrogen sulfotransferase in colon carcinoma: regulators of intratumoral estrogen concentrations and potent prognostic factors. Cancer Res 2009; 69:914-22. [PMID: 19141651 DOI: 10.1158/0008-5472.can-08-0906] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Previous epidemiologic and in vitro studies have indicated a potential involvement of estrogens in the pathogenesis of human colon carcinoma, but the precise roles of estrogens have remained largely unknown. Therefore, in this study, we first measured intratumoral concentrations of estrogens in 53 colon carcinomas using liquid chromatography-electrospray ionization tandem mass spectrometry (LC-ESI-MS). Tissue concentrations of total estrogen [estrone (E(1)) + estradiol] and E(1) were significantly (2.0- and 2.4-fold, respectively) higher in colon carcinoma tissues than in nonneoplastic colonic mucosa (n = 31), and higher intratumoral concentrations of total estrogen and E(1) were significantly associated with adverse clinical outcome. Intratumoral concentration of total estrogen was significantly associated with the combined status of steroid sulfatase (STS) and estrogen sulfotransferase (EST), but not with that of aromatase. Thus, we subsequently examined the STS/EST status in 328 colon carcinomas using immunohistochemistry. Immunoreactivities for STS and EST were detected in 61% and 44% of the cases, respectively. The -/+ group of the STS/EST status was inversely associated with Dukes' stage, depth of invasion, lymph node metastasis, and distant metastasis and positively correlated with Ki-67 labeling index of the carcinomas. In addition, this -/+ group had significantly longer survival, and a multivariate analysis revealed the STS/EST status as an independent prognostic factor. Results from our present study showed that the STS/EST status of carcinoma tissue determined intratumoral estrogen levels and could be a significant prognostic factor in colon carcinoma, suggesting that estrogens are locally produced mainly through the sulfatase pathway and play important roles in the progression of the disease.
Collapse
Affiliation(s)
- Ryuichiro Sato
- Department of Surgery and Pathology, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Rotinen M, Celay J, Alonso MM, Arrazola A, Encio I, Villar J. Estradiol induces type 8 17beta-hydroxysteroid dehydrogenase expression: crosstalk between estrogen receptor alpha and C/EBPbeta. J Endocrinol 2009; 200:85-92. [PMID: 18852215 DOI: 10.1677/joe-08-0134] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Hydroxysteroid (17-beta) dehydrogenase (HSD17B) are the enzymes responsible for the reversible interconversion of 17-hydroxy and 17-keto steroids. The human and mouse type 8 17beta-HSD (HSD17B8) selectively catalyze the conversion of estradiol (E2) to estrone (E1). We previously described thatHSD17B8 is transcriptionally regulated by C/EBPbeta, and that C/EBPbeta is bound to CCAAT boxes located at -5 and -46 of the transcription start site in basal conditions in HepG2 cells. Furthermore, ectopic expression of C/EBPbeta transactivated the HSD17B8 promoter activity. Here, we show that HSD17B8 expression is up-regulated in response to E2 in the estrogen receptor alpha (ERalpha) positive MCF-7 cells. Results showed that this induction is mediated by ERalpha because i) E2 did not induce HSD17B8 expression in ERalphanegative HepG2 cells, ii) ectopic expression of ERalpha restored E2-induced HSD17B8 expression, and iii) this induction was blocked by the anti-ER ICI 182,780. Additional experiments showed that no estrogen response element was necessary for this regulation. However, the CCAAT boxes located at the HSD17B8 proximal promoter were required for E2-induced transcription. Furthermore, co-immunoprecipitation studies revealed tethering of ERalphatoC/EBPbeta in response to E2 in cells expressing ERalpha. Additionally, chromatin immunoprecipitation assays demonstrated that, in response to E2, ERalpha is recruited to the CCAAT boxes in which C/EBPbeta is already bound. Taken together, our results reveal that ERalpha is involved in the transcriptional regulation of HSD17B8 gene in response to E2 through its interaction with C/EBPbeta.
Collapse
Affiliation(s)
- Mirja Rotinen
- Department of Health Sciences, Universidad Pública de Navarra, Pamplona, Spain
| | | | | | | | | | | |
Collapse
|
167
|
Vingren JL, Kraemer WJ, Hatfield DL, Anderson JM, Volek JS, Ratamess NA, Thomas GA, Ho JY, Fragala MS, Maresh CM. Effect of resistance exercise on muscle steroidogenesis. J Appl Physiol (1985) 2008; 105:1754-60. [DOI: 10.1152/japplphysiol.91235.2008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Circulating testosterone is elevated acutely following resistance exercise (RE) and is an important anabolic hormone for muscle adaptations to resistance training. The purpose of this study was to examine the acute effect of heavy RE on intracrine muscle testosterone production in young resistance-trained men and women. Fifteen young, highly resistance-trained men ( n = 8; 21 ± 1 yr, 175.3 ± 6.7 cm, 90.8 ± 11.6 kg) and women ( n = 7; 24 ± 5 yr, 164.6 ± 6.7 cm, 76.4 ± 15.6 kg) completed 6 sets of 10 repetitions of Smith machine squats with 80% of their 1-repetition maximum. Before RE and 10 and 70 min after RE, muscle biopsies were obtained from the vastus lateralis. Before RE, after 3 and 6 sets of squats, and 5, 15, 30, and 70 min into recovery from RE, blood samples were obtained using venipuncture from an antecubital vein. Muscle samples were analyzed for testosterone, 17β-hydroxysteroid dehydrogenase (HSD) type 3, and 3β-HSD type 1 and 2 content. Blood samples were analyzed for glucose and lactate concentrations. No changes were found for muscle testosterone, 3β-HSD type 1 and 2, and 17β-HSD type 3 concentrations. However, a change in protein migration in the Bis-Tris gel was observed for 17β-HSD type 3 postexercise; this change in migration indicated an ∼2.8 kDa increase in molecular mass. These findings indicate that species differences in muscle testosterone production may exist between rats and humans. In humans, muscle testosterone concentrations do not appear to be affected by RE. This study expands on the current knowledge obtained from animal studies by examining resting and postexercise concentrations of muscle testosterone and steroidogenic enzymes in humans.
Collapse
|
168
|
Expression of 17beta-hydroxysteroid dehydrogenases and other estrogen-metabolizing enzymes in different cancer cell lines. Chem Biol Interact 2008; 178:228-33. [PMID: 19022235 DOI: 10.1016/j.cbi.2008.10.038] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2008] [Revised: 10/03/2008] [Accepted: 10/07/2008] [Indexed: 11/22/2022]
Abstract
Estrogen action is regulated at the receptor level by regulation of expression of estrogen receptors, and at the pre-receptor level by interconversions between the active hormone (estradiol) and its inactive counterparts (estrone, estrone-sulfate). In peripheral tissues, estrogens can be produced via the aromatase or the sulfatase pathways. Aromatase converts androstenedione and testosterone to estrone and estradiol, respectively, and sulfatase releases estrogens from inactive sulfates, while sulfotransferase catalyzes the reverse reaction. In both pathways, 17beta-hydroxysteroid dehydrogenases (17beta-HSDs) are of paramount importance as they catalyze activation of estrone to estradiol and inactivation of estradiol to estrone. These enzymes belong to either the short-chain dehydrogenase/reductase (SDR) or the aldo-keto reductase (AKR) protein superfamilies. Differential expression of these pre-receptor regulatory enzymes can lead to high estradiol concentrations, which have been implicated in the development of different diseases. Here, we have examined gene expression levels of estrogen-metabolizing enzymes, as six SDRs (17beta-HSD types 1, 2, 4, 7, 8, 12) and one AKR (17beta-HSD type 5; AKR1C3), of aromatase, steroid sulfatase (STS) and estrogen sulfotransferase (SULT1E1), and of the alpha and beta estrogen receptors (ERs), in breast cancer (MCF-7), endometrial cancer (Ishikawa), choriocarcinoma (JEG3) and liver cancer (HepG2) cell lines. After RNA isolation and cDNA synthesis, real-time PCR analyses were performed. The expression of AKR1C3 was examined also at the protein level. Our data show that in all four cancer cell lines, estradiol can be synthesized from estrone by the action of 17beta-HSD type 12, or from estrone-sulfate by sulfatase. In JEG3 and HepG2 cells, estradiol can be formed from androgens by aromatase and 17beta-HSD type 1. Also in HepG2 cells, AKR1C3, which converts androstenedione to testosterone, in concert with aromatase might be responsible for estradiol formation. In MCF7 and Ishikawa cells, estradiol exerts its actions through ERalpha, while in JEG3 and HepG2 cells, it may act through non-ER-mediated pathways.
Collapse
|
169
|
Zhang Z, Hu J, Zhen H, Wu X, Huang C. Reproductive inhibition and transgenerational toxicity of triphenyltin on medaka (Oiyzias latipes) at environmentally relevant levels. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2008; 42:8133-8139. [PMID: 19031914 DOI: 10.1021/es801573x] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
An increasing number of studies have reported unexpectedly high body burdens of triphenyltin (TPT) in wild fishes around the world. To assess the effects of TPT on fish, we exposed pairs of medaka (Oryzias latipes) to different levels of TPT for 5 weeks, and the reproduction responses and transgenerational effects were studied. The results demonstrated that TPT exposure markedly suppressed the spawning frequency, spawned egg number, egg quality and gonad development, and induced teratogenesis, such as hemorrhaging, eye defects, morphological malformation and conjoined twins, less hatchability, and swim-up failure in the F1 generation, thereby resulting in a significant decrease in the capacity to produce viable offspring (p < 0.01). The residual TPT levels in the exposure fish are in the range of 6.52 +/- 0.56 to 5595 +/- 1016 ng of TPT/g of wet weight, similar to those reported in wild fish around the world, indicating TPT contamination in the real world would have a significant adverse effect on the health of fish population. Down-regulation of vitellogenin (VTG) genes in the female of the TPT exposure groups was recognized as a cause for the decreased fecundity. Expressions of VEGFs and PAX6 associated with vascular or ocular development respectively, were measured in hemorrhaging and eye defects embryos and showed good correlations with response outcomes.
Collapse
Affiliation(s)
- Zhaobin Zhang
- College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | | | | | | | | |
Collapse
|
170
|
Brozic P, Cesar J, Kovac A, Davies M, Johnson AP, Fishwick CWG, Lanisnik Rizner T, Gobec S. Derivatives of pyrimidine, phthalimide and anthranilic acid as inhibitors of human hydroxysteroid dehydrogenase AKR1C1. Chem Biol Interact 2008; 178:158-64. [PMID: 19007763 DOI: 10.1016/j.cbi.2008.10.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Revised: 10/08/2008] [Accepted: 10/08/2008] [Indexed: 11/19/2022]
Abstract
Human hydroxysteroid dehydrogenase (HSD) AKR1C1 is a member of the aldo-keto reductase superfamily, and it functions mainly as a 20alpha-HSD. It catalyzes the reduction of the potent progesterone to the weak 20alpha-hydroxyprogesterone, and of 3alpha,5alpha-tetrahydroprogesterone (5alpha-THP; allopregnanolone) to 5alpha-pregnane-3alpha,20alpha-diol. AKR1C1 thus decreases the levels of progesterone and 5alpha-THP in peripheral tissue. Progesterone inhibits cell proliferation, stimulates differentiation of endometrial cells, and is also important for maintenance of pregnancy, while 5alpha-THP allosterically modulates the activity of the gamma-aminobutyric acid receptor. Inhibitors of AKR1C1 are thus potential agents for treatment of endometrial cancer and endometriosis, as well as other diseases like premenstrual syndrome, catamenial epilepsy and depressive disorders.We have synthesized a series of pyrimidine, phthalimido and athranilic acid derivatives, and have here examined their inhibitory properties towards AKR1C1. A common aldo-keto reductase substrate, 1-acenaphthenol, was used to monitor the NAD(+)-dependent oxidation catalyzed by AKR1C1. The most potent inhibitors of AKR1C1 were the pyrimidine derivative N-benzyl-2-(2-(4-methoxybenzyl)-6-oxo-1,6-dihydropyrimidin-4-yl)acetamide (K(i)=17 microM) and the anthranilic acid derivative 2-(((2',3-dichlorobiphenyl-4-yl)carbonyl)(methyl)amino)benzoic acid (K(i)=33 microM), both of which are non-competitive inhibitors.
Collapse
Affiliation(s)
- P Brozic
- Faculty of Medicine, Institute of Biochemistry, University of Ljubljana, Ljubljana, Slovenia
| | | | | | | | | | | | | | | |
Collapse
|
171
|
Zivec M, Sova M, Brunskole M, Lenarsic R, Rizner TL, Gobec S. New inhibitors of fungal 17β-hydroxysteroid dehydrogenase based on the [1,5]-benzodiazepine scaffold. J Enzyme Inhib Med Chem 2008; 22:29-36. [PMID: 17373544 DOI: 10.1080/14756360600953819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The synthesis and activity of a new series of non-steroidal inhibitors of 17beta-hydroxysteroid dehydrogenase that are based on a 1,5-benzodiazepine scaffold are presented. Their inhibitory potential was screened against 17beta-hydroxysteroid dehydrogenase from the fungus Cochliobolus lunatus (17beta-HSDcl), a model enzyme of the short-chain dehydrogenase/reductase superfamily. Some of these compounds are potent inhibitors of 17beta-HSDcl activity, with IC50 values in the low micromolar range and represent promising lead compounds that should be further developed and investigated as inhibitors of human 17beta-HSD isoforms, which are the enzymes associated with the development of many hormone-dependent and neuronal diseases.
Collapse
Affiliation(s)
- Matej Zivec
- Faculty of Pharmacy, University of Ljubljana, Askerceva 7, 1000 Ljubljana, Slovenia
| | | | | | | | | | | |
Collapse
|
172
|
Tremblay MR, Boivin RP, Luu-The V, Poirier D. Inhibitors of type 1 17β-hydroxysteroid dehydrogenase with reduced estrogenic activity: Modifications of the positions 3 and 6 of estradiol. J Enzyme Inhib Med Chem 2008; 20:153-63. [PMID: 15968820 DOI: 10.1080/14756360500043307] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Breast cancer is the second most frequent cancer affecting women. Among all endocrine therapies for the treatment of breast cancer, inhibition of estrogen biosynthesis is becoming an interesting complementary approach to the use of antiestrogens. The enzyme type 1 17beta-hydroxysteroid dehydrogenase (17beta-HSD) plays a critical role in the biosynthesis of estradiol catalyzing preferentially the reduction of estrone into estradiol, the most active estrogen. Consequently, this enzyme is an interesting biological target for designing drugs for the treatment of estrogen-sensitive diseases such as breast cancer. Our group has reported the synthesis and the biological evaluation of N-methyl, N-butyl 6beta-(thiaheptamamide)estradiol as a potent reversible inhibitor of type 1 17beta-HSD. Unfortunately, this inhibitor has shown an estrogen effect, thus reducing its possible therapeutic interest. Herein three strategies to modify the biological profile (estrogenicity and inhibitory potency) of the initial lead compound were reported. In a first approach, the thioether bond was replaced with a more stable ether bond. Secondly, the hydroxyl group at position 3, which is responsible for a tight binding with the estrogen receptor, was removed. Finally, the amide group of the side-chain was changed to a methyl group. Moreover, the relationship between the inhibitory potency and the configuration of the side-chain at position 6 was investigated. The present study confirmed that the 6beta-configuration of the side chain led to a much better inhibition than the 6alpha-configuration. The replacement of the 3-OH by a hydrogen atom as well as that of the amide group by a methyl was clearly unfavorable for the inhibition of type 1 17beta-HSD. Changing the thioether for an ether bond decreased by 10-fold the estrogenic profile of the lead compound while the inhibitory potency on type 1 17beta-HSD was only decreased by 5-fold. This study contributes to the knowledge required for the development of compounds with the desired profile, that is, a potent inhibitor of type 1 17beta-HSD without estrogen-like effects.
Collapse
Affiliation(s)
- Martin R Tremblay
- Oncology and Molecular Endocrinology Research Center, CHUL Research Center and Université Laval, CHUQ-Pavillon CHUL, 2705 Boulevard Laurier, Québec, Qc G1V 4G2, Canada
| | | | | | | |
Collapse
|
173
|
Lactational exposure to hexavalent chromium delays puberty by impairing ovarian development, steroidogenesis and pituitary hormone synthesis in developing Wistar rats. Toxicol Appl Pharmacol 2008; 232:180-9. [DOI: 10.1016/j.taap.2008.06.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2008] [Revised: 06/07/2008] [Accepted: 06/10/2008] [Indexed: 01/18/2023]
|
174
|
Zheng X, Price CA, Tremblay Y, Lussier JG, Carrière PD. Role of transforming growth factor-β1 in gene expression and activity of estradiol and progesterone-generating enzymes in FSH-stimulated bovine granulosa cells. Reproduction 2008; 136:447-57. [DOI: 10.1530/rep-07-0316] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Survival and inhibitory factors regulate steroidogenesis and determine the fate of developing follicles. The objective of this study was to determine the role of transforming growth factor-β1 (TGFB1) in the regulation of estradiol-17β (E2) and progesterone (P4) secretion in FSH-stimulated bovine granulosa cells. Granulosa cells were obtained from 2 to 5 mm follicles and cultured in serum-free medium. FSH dose (1 and 10 ng/ml for 6 days) and time in culture (2, 4, and 6 days with 1 ng/ml FSH) increased E2secretion and mRNA expression of E2-related enzymes cytochrome P450 aromatase (CYP19A1) and 17β-hydroxysteroid dehydrogenase type 1 (HSD17B1), but notHSD17B7. TGFB1 in the presence of FSH (1 ng/ml) inhibited E2secretion, and decreased mRNA expression of FSH receptor(FSHR),CYP19A1, andHSD17B1, but notHSD17B7. FSH dose did not affect P4secretion and mRNA expression of 3β-hydroxysteroid dehydrogenase (HSD3B) and α-glutathioneS-transferase (GSTA), but inhibited the amount of steroidogenic acute regulatory protein(STAR)mRNA. Conversely, P4and mRNA expression ofSTAR, cytochrome P450 side-chain cleavage(CYP11A1),HSD3B, andGSTAincreased with time in culture. TGFB1 inhibited P4secretion and decreased mRNA expression ofSTAR,CYP11A1,HSD3B, andGSTA. TGFB1 modified the formation of granulosa cell clumps and reduced total cell protein. Finally, TGFB1 decreased conversion of androgens to E2, but did not decrease the conversion of estrone (E1) to E2and pregnenolone to P4. Overall, these results indicate that TGFB1 counteracts stimulation of E2and P4synthesis in granulosa cells by inhibiting key enzymes involved in the conversion of androgens to E2and cholesterol to P4without shutting down HSD17B reducing activity and HSD3B activity.
Collapse
|
175
|
Schuster D, Nashev LG, Kirchmair J, Laggner C, Wolber G, Langer T, Odermatt A. Discovery of Nonsteroidal 17β-Hydroxysteroid Dehydrogenase 1 Inhibitors by Pharmacophore-Based Screening of Virtual Compound Libraries. J Med Chem 2008; 51:4188-99. [DOI: 10.1021/jm800054h] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Daniela Schuster
- Computer-Aided Molecular Design Group, Department of Pharmaceutical Chemistry, University of Innsbruck, Innrain 52c, A-6020 Innsbruck, Austria, and Center of Molecular Biosciences Innsbruck—CMBI, Peter-Mayr-Strasse 1a, A-6020 Innsbruck, Austria, Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland, Inte:Ligand Software-Entwicklungs and Consulting GmbH, Marihilferstrasse 74B/11, A-1070 Wien, Austria
| | - Lyubomir G. Nashev
- Computer-Aided Molecular Design Group, Department of Pharmaceutical Chemistry, University of Innsbruck, Innrain 52c, A-6020 Innsbruck, Austria, and Center of Molecular Biosciences Innsbruck—CMBI, Peter-Mayr-Strasse 1a, A-6020 Innsbruck, Austria, Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland, Inte:Ligand Software-Entwicklungs and Consulting GmbH, Marihilferstrasse 74B/11, A-1070 Wien, Austria
| | - Johannes Kirchmair
- Computer-Aided Molecular Design Group, Department of Pharmaceutical Chemistry, University of Innsbruck, Innrain 52c, A-6020 Innsbruck, Austria, and Center of Molecular Biosciences Innsbruck—CMBI, Peter-Mayr-Strasse 1a, A-6020 Innsbruck, Austria, Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland, Inte:Ligand Software-Entwicklungs and Consulting GmbH, Marihilferstrasse 74B/11, A-1070 Wien, Austria
| | - Christian Laggner
- Computer-Aided Molecular Design Group, Department of Pharmaceutical Chemistry, University of Innsbruck, Innrain 52c, A-6020 Innsbruck, Austria, and Center of Molecular Biosciences Innsbruck—CMBI, Peter-Mayr-Strasse 1a, A-6020 Innsbruck, Austria, Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland, Inte:Ligand Software-Entwicklungs and Consulting GmbH, Marihilferstrasse 74B/11, A-1070 Wien, Austria
| | - Gerhard Wolber
- Computer-Aided Molecular Design Group, Department of Pharmaceutical Chemistry, University of Innsbruck, Innrain 52c, A-6020 Innsbruck, Austria, and Center of Molecular Biosciences Innsbruck—CMBI, Peter-Mayr-Strasse 1a, A-6020 Innsbruck, Austria, Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland, Inte:Ligand Software-Entwicklungs and Consulting GmbH, Marihilferstrasse 74B/11, A-1070 Wien, Austria
| | - Thierry Langer
- Computer-Aided Molecular Design Group, Department of Pharmaceutical Chemistry, University of Innsbruck, Innrain 52c, A-6020 Innsbruck, Austria, and Center of Molecular Biosciences Innsbruck—CMBI, Peter-Mayr-Strasse 1a, A-6020 Innsbruck, Austria, Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland, Inte:Ligand Software-Entwicklungs and Consulting GmbH, Marihilferstrasse 74B/11, A-1070 Wien, Austria
| | - Alex Odermatt
- Computer-Aided Molecular Design Group, Department of Pharmaceutical Chemistry, University of Innsbruck, Innrain 52c, A-6020 Innsbruck, Austria, and Center of Molecular Biosciences Innsbruck—CMBI, Peter-Mayr-Strasse 1a, A-6020 Innsbruck, Austria, Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland, Inte:Ligand Software-Entwicklungs and Consulting GmbH, Marihilferstrasse 74B/11, A-1070 Wien, Austria
| |
Collapse
|
176
|
Bacallao K, Leon L, Gabler F, Soto E, Romero C, Valladares L, Vega M. In situ estrogen metabolism in proliferative endometria from untreated women with polycystic ovarian syndrome with and without endometrial hyperplasia. J Steroid Biochem Mol Biol 2008; 110:163-9. [PMID: 18467089 DOI: 10.1016/j.jsbmb.2008.03.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Revised: 03/06/2008] [Accepted: 03/25/2008] [Indexed: 01/18/2023]
Abstract
The aim of the present investigation was to study whether the endocrinological status of women bearing polycystic ovarian syndrome (PCOS) affects the endometrial in situ steroid metabolism. For this purpose, we evaluated the mRNA levels (RT-PCR), and the activity of steroid metabolic enzymes: P450 aromatase, steroid sulfatase (STS), estrogen sulfotransferase (EST) and 17beta-hydroxysteroid dehydrogenase (17beta-HSD) in 23 samples of normal endometria (CE), 18 PCOS endometria without treatment (PCOSE), 10 specimens from PCOS women with endometrial hyperplasia (HPCOSE), and 7 endometria from patients with endometrial hyperplasia not associated to PCOS (EH). The data showed lower levels of STS mRNA for PCOSE and HPCOSE (p<0.05, p<0.01, respectively) and of EST for HPCOSE and EH compared to control (p<0.05). However, higher levels for EST mRNA were obtained in PCOSE (p<0.05) versus CE. The mRNA and protein levels for P450 aromatase were undetectable in all analyzed endometria. The relationship between the activities of STS and EST was lower in PCOSE and HPCOSE (p<0.05) versus CE. The ratio between the mRNA from 17beta-HSD type 1/type 2 was higher in PCOSE (p<0.05), whereas, a diminution in the 17beta-HSD type 2 activity was observed in PCOSE (p<0.05). These results indicate that the activity of enzymes related to the steroid metabolism in analyzed PCOSE differ from those found in the CE. Consequently, PCOSE may present an in situ deregulation of the steroid metabolism.
Collapse
Affiliation(s)
- K Bacallao
- Institute of Maternal and Child Research, School of Medicine, University of Chile, Chile; Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont # 999, Santiago, Chile
| | | | | | | | | | | | | |
Collapse
|
177
|
Figueroa JD, Sakoda LC, Graubard BI, Chanock S, Rubertone MV, Erickson RL, McGlynn KA. Genetic variation in hormone metabolizing genes and risk of testicular germ cell tumors. Cancer Causes Control 2008; 19:917-29. [DOI: 10.1007/s10552-008-9153-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2007] [Accepted: 03/23/2008] [Indexed: 10/22/2022]
|
178
|
Allan GM, Vicker N, Lawrence HR, Tutill HJ, Day JM, Huchet M, Ferrandis E, Reed MJ, Purohit A, Potter BVL. Novel inhibitors of 17beta-hydroxysteroid dehydrogenase type 1: templates for design. Bioorg Med Chem 2008; 16:4438-56. [PMID: 18329273 DOI: 10.1016/j.bmc.2008.02.059] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Revised: 02/12/2008] [Accepted: 02/19/2008] [Indexed: 12/31/2022]
Abstract
The 17beta-hydroxysteroid dehydrogenases (17beta-HSDs) catalyze the interconversion between the oxidized and reduced forms of androgens and estrogens at the 17 position. The 17beta-HSD type 1 enzyme (17beta-HSD1) catalyzes the reduction of estrone (E1) to estradiol and is expressed in malignant breast cells. Inhibitors of this enzyme thus have potential as treatments for hormone dependent breast cancer. Syntheses and biological evaluation of novel non-steroidal inhibitors designed to mimic the E1 template are reported using information from potent steroidal inhibitors. Of the templates investigated biphenyl ethanone was promising and led to inhibitors with IC(50) values in the low micromolar range.
Collapse
Affiliation(s)
- Gillian M Allan
- Medicinal Chemistry, Department of Pharmacy and Pharmacology and Sterix Ltd, University of Bath, Claverton Down BA2 7AY, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Abstract
It is well recognized that there are two androgens, namely testosterone (T) and dihydrotestosterone (DHT); T plays an important role in the testis and muscle, and DHT is crucial for the development, function and pathology of the prostate. It is generally thought that DHT is produced from the 5alpha-reduction of circulating T before being inactivated by 3alpha-hydroxysteroid dehydrogenase (3alpha-HSD) that converts DHT into 5alpha-androstane-3alpha,17beta-diol (3alpha-diol). However, the presence of various steroidogenic enzymes in the prostate as well as the availability at high levels of various steroid precursors such as dehydroepiandrosterone sulphate (DHEAS), dehydroepiandrosterone (DHEA) and 4-androstenedione (4-dione) strongly suggest the existence of additional pathways involved in the biosynthesis and metabolism of DHT. Because steroidogenesis could be different in different species, data from the literature obtained from various human, dog, rat and mouse prostate tissues, as well as primary cells and prostatic cancer cell lines, provide a somewhat confusing picture. In the present chapter, we review the data in order to provide a clearer picture of the pathways involved in DHT biosynthesis and metabolism in the human prostate.
Collapse
Affiliation(s)
- Van Luu-The
- Laboratory of Molecular Endocrinology and Oncology, Laval University Hospital Research Center, Laval University, Québec, Canada.
| | | | | |
Collapse
|
180
|
Horiguchi Y, Araki M, Motojima K. 17beta-Hydroxysteroid dehydrogenase type 13 is a liver-specific lipid droplet-associated protein. Biochem Biophys Res Commun 2008; 370:235-8. [PMID: 18359291 DOI: 10.1016/j.bbrc.2008.03.063] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Accepted: 03/14/2008] [Indexed: 10/22/2022]
Abstract
17beta-Hydroxysteroid dehydrogenase (17betaHSD) type 13 is identified as a new lipid droplet-associated protein. 17betaHSD type 13 has an N-terminal sequence similar to that of 17betaHSD type 11, and both sequences function as an endoplasmic reticulum and lipid droplet-targeting signal. Localization of native 17betaHSD type 13 on the lipid droplets was confirmed by subcellular fractionation and Western blotting. In contrast to 17betaHSD type 11, however, expression of 17betaHSD type 13 is largely restricted to the liver and is not enhanced by peroxisome proliferator-activated receptor alpha and its ligand. Instead the expression level of 17betaHSD type 13 in the receptor-null mice was increased several-fold. 17betaHSD type 13 may have a distinct physiological role as a lipid droplet-associated protein in the liver.
Collapse
Affiliation(s)
- Yuka Horiguchi
- Department of Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan
| | | | | |
Collapse
|
181
|
Plourde M, Samson C, Durocher F, Sinilnokova O, Simard J. Characterization of HSD17B1 sequence variants in breast cancer cases from French Canadian families with high risk of breast and ovarian cancer. J Steroid Biochem Mol Biol 2008; 109:115-28. [PMID: 18083510 DOI: 10.1016/j.jsbmb.2007.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2007] [Revised: 10/16/2007] [Accepted: 10/30/2007] [Indexed: 11/24/2022]
Abstract
A family history of disease and estrogen exposure are risk factors for breast cancer. The HSD17B1 gene encodes a key steroidogenic enzyme that catalyses the final step of estradiol biosynthesis, rendering it a good candidate gene for breast cancer susceptibility. The current study was designed to screen for HSD17B1 germline mutations potentially involved in breast cancer susceptibility. DNA samples from 50 individuals affected with breast cancer from non-BRCA1/2 French Canadian families with a high risk of breast and ovarian cancer were screened for sequence variants in HSD17B1. Our study identified 28 sequence variants, including three non-synonymous variants, p.Ala238Val, p.Arg259His, p.Ser313Gly, one of which (p.Arg259His) was not previously reported. Functional assays failed to show changes in either activity or recombinant proteins levels for all three variants. Thus, our resequencing analysis does not support the existence of deleterious, gain-of-function or transcription mutations in HSD17B1, which could explain the clustering of breast cancer cases in non-BRCA1/2 high-risk French Canadian families. However, a haplotype-based approach was used to establish tSNPs, providing a valuable tool for further searches of common disease-associated variants in this gene, using large cohorts.
Collapse
Affiliation(s)
- Marie Plourde
- Cancer Genomics Laboratory, Oncology and Molecular Endocrinology Research Center, Centre Hospitalier Universitaire de Québec and Laval University, Quebec G1V 4G2, Canada
| | | | | | | | | |
Collapse
|
182
|
Laplante Y, Poirier D. Proliferative effect of androst-4-ene-3,17-dione and its metabolites in the androgen-sensitive LNCaP cell line. Steroids 2008; 73:266-71. [PMID: 18082864 DOI: 10.1016/j.steroids.2007.10.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Revised: 06/15/2007] [Accepted: 10/24/2007] [Indexed: 11/16/2022]
Abstract
As a therapeutic approach for the treatment of androgen-sensitive diseases, it would be tempting to lower the level of the potent androgens testosterone (T) and dihydrotestosterone (DHT) by using inhibitors of type 3 and type 5 17beta-hydroxysteroid dehydrogenases (17beta-HSDs). However, the efficiency of such a strategy will be optimal only if androst-4-ene-3,17-dione (Delta4-dione), the precursor of T, does not possess per se agonist activity on the androgen receptor (AR). To determine if the proliferative effect previously observed on AR(+) cells for Delta4-dione originates from its direct (per se) action on AR or from its transformation into a metabolite, we started a series of experimentations using the human prostate cancer LNCaP cell line, which expresses a highly sensitive AR. By real-time RT-PCR analysis, we detected type 1 5alpha-reductase (5alpha-R), a small amount of type 5 17beta-HSD, but not type 2 5alpha-R nor type 3 17beta-HSD. We then studied the transformation of labeled Delta4-dione in LNCaP cells after 1-7 days and the most important metabolite detected was 5alpha-androstane-3,17-dione (A-dione), which is the product of 5alpha-R activity. We measured only low levels of androsterone (ADT) and epi-ADT. This result was next confirmed by using an inhibitor of 5alpha-R that completely inhibited the transformation of Delta4-dione into A-dione, and consequently into ADT and epi-ADT. The proliferative effect of Delta4-dione (carefully purified) on LNCaP (AR(+)) cells was next determined in presence or absence of the 5alpha-R inhibitor. Although the cells proliferate in the presence of Delta4-dione only, no cell proliferation was observed with a combination of Delta4-dione and 5alpha-R inhibitor, suggesting that Delta4-dione is not androgenic per se. We next determined that A-dione and epi-ADT stimulated cell growth with the same pattern and potency as Delta4-dione, whereas ADT had a 3.5-fold lower proliferative activity. In conclusion, Delta4-dione is not in itself an agonist steroid on LNCaP (AR(+)) cells, and its proliferative activity appears to be mediated by its transformation into A-dione and/or into epi-ADT.
Collapse
Affiliation(s)
- Yannick Laplante
- Medicinal Chemistry Division, Oncology and Molecular Endocrinology Research Center, CHUQ-Pavillon CHUL and Université Laval, Québec, Canada
| | | |
Collapse
|
183
|
Leon L, Bacallao K, Gabler F, Romero C, Valladares L, Vega M. Activities of steroid metabolic enzymes in secretory endometria from untreated women with Polycystic Ovary Syndrome. Steroids 2008; 73:88-95. [PMID: 17953976 DOI: 10.1016/j.steroids.2007.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Revised: 09/10/2007] [Accepted: 09/11/2007] [Indexed: 10/22/2022]
Abstract
Polycystic Ovary Syndrome (PCOS) is an endocrine-metabolic pathology related with infertility and recurrent miscarriage. We have previously shown that the endometrium of these patients can exhibit a potentially higher sensitivity to estrogen action, being estrogens important regulators of the cell cycle and tissue homeostasis. The effect of estrogens on tissues depends on their in situ availability, which is in part regulated by the activity of steroid metabolic enzymes within the tissues. Therefore, the objective of the present study was to analyze if the activity and/or expression of steroid metabolic enzymes in endometria from women with PCOS differ from controls. For this purpose, the activity of the enzymes was determined by using radiometric assays and the mRNA levels measured by semi-quantitative RT-PCR. Both assays were assessed in endometria obtained during mid secretory phase from control (CE, n=12) and PCOS women (PCOSE, n=11). For the statistical analyses, Mann-Whitney and Student's t-tests were used to compare CE and PCOSE, considering a p value <0.05 significantly different. The results showed an increase in the sulfatase activity in PCOS respect to control endometria (200+/-28pmol/mg vs. 115+/-13pmol/mgproth; p<0.05), in agreement with the higher mRNA levels found for the enzyme in PCOSE. In addition, a PCOSE exhibited lower activity of sulfotransferase respect to the control group (50+/-21pmol/mg vs. 124+/-10pmol/mgproth; p<0.05), whereas a higher level of 17beta-hydroxysteroid dehydrogenase type 1mRNA was found in PCOSE compared with the control tissues (p<0.05). The activity of 17beta-hydroxysteroid dehydrogenase type 2 and the mRNA levels of sulfotransferase were similar in both groups; meanwhile, the expression of aromatase was undetectable. These data indicate that the sulfatase pathway could play an important role in the local production of estrogens in PCOSE from secretory phase. This potentially higher bioavailability of estrogens in endometria from PCOS women could influence the deregulation of tissue homeostasis that we have previously reported, and could partially explain the poor reproductive performance observed in this group of patients.
Collapse
Affiliation(s)
- Luis Leon
- Institute of Maternal and Child Research, School of Medicine, University of Chile, Chile
| | | | | | | | | | | |
Collapse
|
184
|
The direct action of estrone on vascular tissue involves genomic and non-genomic actions. Life Sci 2008; 82:115-23. [DOI: 10.1016/j.lfs.2007.10.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Revised: 10/11/2007] [Accepted: 10/22/2007] [Indexed: 11/15/2022]
|
185
|
Blum JW, Baumrucker CR. Insulin-like growth factors (IGFs), IGF binding proteins, and other endocrine factors in milk: role in the newborn. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 606:397-422. [PMID: 18183939 DOI: 10.1007/978-0-387-74087-4_16] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The role of colostrum and milk in the neonate has been chiefly recognized as a comprehensive nutrient foodstuff. In addition, the provision of colostrum-the first milk-for early immune capacity has been well documented for several species. Colostrum is additionally a rich and concentrated source of various factors that demonstrate biological activity in vitro. Three hypotheses have been proposed for the phenotypic function of these secreted bioactive components: (1) only mammary disposal, (2) mammary cell regulation, and (3) neonatal function [gastrointestinal tract (GIT) or systemic]. Traditionally, it was assumed that the development of the GIT is preprogrammed and not influenced by events occurring in the intestinal lumen. However, a large volume of research has demonstrated that colostrum (or milk-borne) bioactive components can basically contribute to the regulation of GIT growth and differentiation, while their role in postnatal development at physiological concentrations has remained elusive. Much of our current understanding is derived from cell culture and laboratory animals, but experimentation with agriculturally important species is taking place. This chapter provides an overview of work conducted primarily in neonatal calves and secondarily in other species on the effects on neonates of selected peptide endocrine factors (hormones, growth factors, in part cytokines) in colostrum. The primary focus will be on insulin-like growth factors (IGFs) and IGF binding proteins (IGFBPs) and other bioactive peptides, but new interest and concern about steroids (especially estrogens) in milk are considered as well.
Collapse
Affiliation(s)
- Jürg W Blum
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, CH-3012 Bern, Switzerland.
| | | |
Collapse
|
186
|
Yokoi Y, Horiguchi Y, Araki M, Motojima K. Regulated expression by PPARalpha and unique localization of 17beta-hydroxysteroid dehydrogenase type 11 protein in mouse intestine and liver. FEBS J 2007; 274:4837-47. [PMID: 17714510 DOI: 10.1111/j.1742-4658.2007.06005.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
17beta-Hydroxysteroid dehydrogenase type 11 (17beta-HSD11) is a member of the short-chain dehydrogenase/reductase family involved in the activation and inactivation of sex steroid hormones. We recently identified 17beta-HSD11 as a gene that is efficiently regulated by peroxisome proliferator-activated receptor-alpha PPARalpha in the intestine and the liver [Motojima K (2004) Eur J Biochem271, 4141-4146]. In this study, we characterized 17beta-HSD11 at the protein level to obtain information about its physiologic role in the intestine and liver. For this purpose, specific antibodies against 17beta-HSD11 were obtained. Western blotting analysis showed that administration of a peroxisome proliferator-activated receptor-alpha agonist induced 17beta-HSD11 protein in the jejunum but not in the colon, and to a much higher extent than in the liver of mice. A subcellular localization study using Chinese hamster ovary cells and green fluorescent protein-tagged 17beta-HSD11 showed that it was mostly localized in the endoplasmic reticulum under normal conditions, whereas it was concentrated on lipid droplets when they were induced. A pulse-chase experiment suggested that 17beta-HSD11 was redistributed to the lipid droplets via the endoplasmic reticulum. Immunohistochemical analysis using tissue sections showed that 17beta-HSD11 was induced mostly in intestinal epithelia and hepatocytes, with heterogeneous localization both in the cytoplasm and in vesicular structures. A subcellular fractionation study of liver homogenates confirmed that 17beta-HSD11 was localized mostly in the endoplasmic reticulum when mice were fed a normal diet, but was distributed in both the endoplasmic reticulum and the lipid droplets of which formation was induced by feeding a diet containing a proliferator-activated receptor-alpha agonist. Taken together, these data indicate that 17beta-HSD11 localizes both in the endoplasmic reticulum and in lipid droplets, depending on physiologic conditions, and that lipid droplet 17beta-HSD11 is not merely an endoplasmic reticulum contaminant or a nonphysiologically associated protein in the cultured cells, but a bona fide protein component of the membranes of both intracellular compartments.
Collapse
Affiliation(s)
- Yasuhide Yokoi
- Department of Biochemistry, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | | | | | | |
Collapse
|
187
|
Abstract
The presence of sex steroids and their receptors has been demonstrated in all vertebrate groups from Agnatha to Mammalia but not in invertebrates. In genomic analyses of urochordates, cytochrome P450 (CYP) genes important for biosynthesis of sex steroids are absent. In the present study, we confirmed the presence of estrogen, androgen, and progesterone by using radioimmunoassay in gonads of amphioxus, Branchiostoma belcheri, which is considered to be evolutionarily closer to vertebrates than other invertebrates. Furthermore, CYP genes encoding CYP11A, CYP17, and CYP19 and transcripts for 17beta-hydroxysteroid dehydrogenase were cloned from amphioxus ovaries. Among invertebrates, the presence of hydroxysteroid dehydrogenase enzymes and metabolized steroids was shown in paracytic Taenia and corals. However, CYPs metabolizing sex steroids have not been demonstrated in invertebrates, nor has an attempt been made to consider the entire pathway from cholesterol to estrogen. This study is the first evidence to suggest the presence of CYP enzymes in sex steroid production in invertebrates.
Collapse
Affiliation(s)
- Takanobu Mizuta
- Center for Advanced Marine Research, Ocean Research Institute, University of Tokyo, Nakano, Tokyo 164-8639, Japan
| | | |
Collapse
|
188
|
Bérubé M, Poirier D. Chemical synthesis and in vitro biological evaluation of a phosphorylated bisubstrate inhibitor of type 3 17beta-hydroxysteroid dehydrogenase. J Enzyme Inhib Med Chem 2007; 22:201-11. [PMID: 17518347 DOI: 10.1080/14756360601051423] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Type 3 17beta-hydroxysteroid dehydrogenase (17beta-HSD) catalyzes the last step in the biosynthesis of the potent androgen testosterone (T) by selectively reducing the C17 ketone of 4-androstene-3,17-dione (delta4-dione), with NADPH as cofactor. This enzyme is thus an interesting therapeutic target for androgen-sensitive diseases. Using an efficient convergent chemical approach we synthesized a phosphorylated version of the best delta4-dione/adenosine hybrid inhibitor of type 3 17beta-HSD previously reported. An appropriately protected C2' phosphorylated adenosine was first prepared and linked by esterification to the steroid delta4-dione bearing an alkyl spacer. After three deprotection steps, the phosphorylated bisubstrate inhibitor was obtained. The inhibitory potency of this compound was evaluated on homogenated HEK-293 cells overexpressing type 3 17beta-HSD and compared to the best non-phosphorylated bisubstrate inhibitor. Unexpectedly, the phosphorylated derivative was slightly less potent than the non-phosphorylated bisubstrate inhibitor of type 3 17beta-HSD. Two hypotheses are discussed to explain this result: 1) the phosphorylated adenosine moiety does not interact optimally with the cofactor-binding site and 2) the bisubstrate inhibitors, phosphorylated or not, interact only with the substrate-binding site of type 3 17beta-HSD.
Collapse
Affiliation(s)
- Marie Bérubé
- Medicinal Chemistry Division, Oncology and Molecular Endocrinology Research Center, CHUQ-Pavillon CHUL and Université Laval, Québec G1V 4G2, Canada
| | | |
Collapse
|
189
|
Takeuchi S, Mukai N, Tateishi T, Miyakawa S. Production of sex steroid hormones from DHEA in articular chondrocyte of rats. Am J Physiol Endocrinol Metab 2007; 293:E410-5. [PMID: 17473054 DOI: 10.1152/ajpendo.00042.2007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Dehydroepiandrosterone (DHEA), a precursor of sex steroid hormones, is synthesized by cholesterol side-chain cleavage cytochrome P-450 and 17alpha-hydroxylase cytochrome P-450 mainly from cholesterol and converted to testosterone and estrogen by 3beta-hydroxysteroid dehydrogenase (3beta-HSD), 17beta-HSD, and aromatase cytochrome P-450. Although sex steroid hormones have important effects in the protection of articular cartilage, it is unclear whether articular cartilage has a local steroidogenic enzymatic machinery capable of metabolizing DHEA. This study was aimed to clarify whether steroidogenesis-related enzymes are expressed in articular chondrocytes, whether expression levels are changed by DHEA, and whether articular chondrocytes are capable of synthesizing sex steroid hormones from DHEA. Articular chondrocytes isolated from adult rats were cultured with DHEA for 3 days. All of the mRNA expressions of steroidogenesis-related enzymes were detected in cultured articular chondrocytes of rats, but the mRNA expression levels of testosterone and estradiol in cultured media increased after the addition of DHEA. These findings provided the first evidence that articular chondrocytes expressed steroidogenesis-related enzyme genes and that they are capable of locally synthesizing sex steroid hormones locally from DHEA.
Collapse
Affiliation(s)
- Satsuki Takeuchi
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki 305-8574, Japan
| | | | | | | |
Collapse
|
190
|
Köhrle J. Thyroid hormone transporters in health and disease: advances in thyroid hormone deiodination. Best Pract Res Clin Endocrinol Metab 2007; 21:173-91. [PMID: 17574002 DOI: 10.1016/j.beem.2007.04.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thyroid hormone metabolism by the three deiodinase selenoproteins -- DIO1, DIO2, and DIO3 -- regulates the local availability of various iodothyronine metabolites and thus mediates their effects on gene expression, thermoregulation, energy metabolism, and many key reactions during the development and maintenance of an adult organism. Circulating serum levels of thyroid hormone and thyroid-stimulating hormone, used as a combined indicator of thyroid hormone status, reflect a composite picture of: thyroid secretion; tissue-specific production of T(3) by DIO1 and DIO2 activity, which both contribute to circulating levels of T(3); and degradation of the prohormone T4, of the thyromimetically active T(3), of the inactive rT(3), of other iodothyronines metabolites with a lower iodine content and of thyroid hormone conjugates. Degradation reactions are catalyzed by either DIO1 or DIO3. Aberrant expression of individual deiodinases in disease, single nucleotide polymorphisms in their genes, and novel regulators of DIO gene expression (such as bile acids) provide a more complex picture of the fine tuning and the adaptation of systemic and local bioavailability of thyroid hormones.
Collapse
Affiliation(s)
- Josef Köhrle
- Institut für Experimentelle Endokrinologie und Endokrinologisches Forschungszentrum der Charité EnForCé, Charité Universitätsmedizin Berlin, CCM Charitéplatz 1, Berlin, Germany.
| |
Collapse
|
191
|
Villar J, Celay J, Alonso MM, Rotinen M, de Miguel C, Migliaccio M, Encío I. Transcriptional regulation of the human type 8 17beta-hydroxysteroid dehydrogenase gene by C/EBPbeta. J Steroid Biochem Mol Biol 2007; 105:131-9. [PMID: 17583490 DOI: 10.1016/j.jsbmb.2006.12.106] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Accepted: 12/07/2006] [Indexed: 11/23/2022]
Abstract
17beta-Hydroxysteroid dehydrogenases (17beta-HSD) regulate the intracellular concentration of active sex steroid hormones in target tissues. To date, at least 14 different isozymes have been identified. The type 8 17beta-hydroxysteroid dehydrogenase (17beta-HSD8) selectively catalyzes the conversion of estradiol (E2) to estrone (E1). To map the promoter region and to investigate its regulation, we cloned and fused a 1600 bp DNA fragment upstream of the 17beta-HSD8 transcriptional start site to a luciferase reporter gene. After transient transfection in HepG2 cells, this fragment was shown to possess promoter activity. Deletion constructs of the 5' flanking region of the 17beta-HSD8 gene led to the identification of the minimal promoter region within the first 75 bp upstream of the transcriptional start site. This region included two CCAAT boxes and sequences closely resembling the consensus Sp1 and NF-kappaB motifs. Site directed mutagenesis revealed that the CCAAT boxes were essential for transcription in HepG2. EMSA, supershift and chromatin immunoprecipitation reflected that these sequences were binding sites for C/EBPbeta. Furthermore, promoter activity was increased by the co-transfection of a C/EBPbeta expression vector, and this transactivation was through both CCAAT boxes. Our studies indicate that C/EBPbeta is essential for the transcription of the 17beta-HSD8 gene in the liver.
Collapse
Affiliation(s)
- Joaquín Villar
- Departamento de Ciencias de la Salud, Universidad Pública de Navarra, Avda. Barañain s/n, 31008 Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
192
|
Petry CJ, Ong KK, Wingate DL, de Zegher F, Ibáñez L, Dunger DB. Lack of association between common polymorphisms in the 17beta-hydroxysteroid dehydrogenase type V gene (HSD17B5) and precocious pubarche. J Steroid Biochem Mol Biol 2007; 105:176-80. [PMID: 17583494 DOI: 10.1016/j.jsbmb.2007.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Accepted: 01/29/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND 17beta-Hydroxysteroid dehydrogenase (type V; HSD17B5) is a key enzyme involved in testosterone production in females. A single nucleotide polymorphism (SNP) in the promoter region of its gene was recently found to be associated with polycystic ovary syndrome (PCOS) and its related hyperandrogenaemia. Precocious pubarche (PP) is a clinical entity pointing to adrenal androgen excess from mid-childhood onward and is associated with ovarian androgen excess from puberty onward. It is therefore a strong risk factor for PCOS. METHODS To investigate associations between this promoter SNP along with three exonic SNPs (one non-synonymous and two synonymous) from the same gene, and PP, a case-control study was performed in 190 girls with PP (84 of which were also tested for functional ovarian hyperandrogenism) from Barcelona, Spain and 71 healthy controls. Clinical features and hormone concentrations relevant to hyperandrogenism were compared by HSD17B5 genotype and haplotype. RESULTS Neither HSD17B5 genotypes nor haplotype were associated with PP, or subsequent androgen excess in girls from Barcelona (all P>0.05). CONCLUSIONS HSD17B5 SNPs predicted to have functional effects do not appear to be a risk factor for PP in girls from Barcelona, despite these girls being at high risk of developing androgen excess in adulthood.
Collapse
Affiliation(s)
- Clive J Petry
- Department of Paediatrics, University of Cambridge, Cambridge CB2 2QQ, UK.
| | | | | | | | | | | |
Collapse
|
193
|
Ashibe B, Hirai T, Higashi K, Sekimizu K, Motojima K. Dual subcellular localization in the endoplasmic reticulum and peroxisomes and a vital role in protecting against oxidative stress of fatty aldehyde dehydrogenase are achieved by alternative splicing. J Biol Chem 2007; 282:20763-73. [PMID: 17510064 DOI: 10.1074/jbc.m611853200] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Fatty aldehyde dehydrogenase (FALDH, ALDH3A2) is thought to be involved in the degradation of phytanic acid, a saturated branched chain fatty acid derived from chlorophyll. However, the identity, subcellular distribution, and physiological roles of FALDH are unclear because several variants produced by alternative splicing are present in varying amounts at different subcellular locations. Subcellular fractionation experiments do not provide a clear-cut conclusion because of the incomplete separation of organelles. We established human cell lines heterologously expressing mouse FALDH from each cDNA without tagging under the control of an inducible promoter and detected the variant FALDH proteins using a mouse FALDH-specific antibody. One variant, FALDH-V, was exclusively detected in peroxisomal membranes. Human FALDH-V with an amino-terminal Myc sequence also localized to peroxisomes. The most dominant form, FALDH-N, and other variants examined, however, were distributed in the endoplasmic reticulum. A gas chromatography-mass spectrometry-based analysis of metabolites in FALDH-expressing cells incubated with phytol or phytanic acid showed that FALDH-V, not FALDH-N, is the key aldehyde dehydrogenase in the degradation pathway and that it protects peroxisomes from oxidative stress. In contrast, both FALDHs had a protective effect against oxidative stress induced by a model aldehyde for lipid peroxidation, dodecanal. These results suggest that FALDH variants are produced by alternative splicing and share an important role in protecting against oxidative stress in an organelle-specific manner.
Collapse
Affiliation(s)
- Bunichiro Ashibe
- Department of Biochemistry, Meiji Pharmaceutical University, Noshio 2-522-1, Kiyose, Tokyo 204-8588, Japan
| | | | | | | | | |
Collapse
|
194
|
Mindnich R, Adamski J. Functional aspects of 17beta-hydroxysteroid dehydrogenase 1 determined by comparison to a closely related retinol dehydrogenase. J Steroid Biochem Mol Biol 2007; 104:334-9. [PMID: 17467981 DOI: 10.1016/j.jsbmb.2007.03.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Determining the functional aspects of a gene or protein is a difficult and time-consuming process. De novo analysis is surely the hardest and so it is often quite useful to start with a comparison to functionally or structurally related proteins. Although 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD 1) can hardly be called a new protein but rather the best characterized among the family of 17beta-HSDs some aspects of structure-function relationships remain unclear. We have sought new aspects of 17beta-HSD 1 function through a comparison with its closest homolog, a photoreceptor-associated retinol dehydrogenase (prRDH). Overall amino acid identity and size of the proteins are highly conserved, but major differences occur in the C-termini, where prRDH, but not 17beta-HSD 1, harbors motifs indicative of membrane localization. To gain insight into substrate discrimination by prRDH and 17beta-HSD 1, we constructed 3D-structure models of the corresponding zebrafish enzymes. Investigation of the substrate binding site revealed a few identical amino acids, and suggested a role for G143 in zebrafish 17beta-HSD 1 and M146 and M147 in the two zebrafish paralogs prRDH 1 and prRDH 2, respectively, in substrate specificity. Activity measurements of modified proteins in transiently transfected intact HEK 293 cells hint at a putative role of these amino acids in discrimination between steroid and retinoid substrates.
Collapse
Affiliation(s)
- R Mindnich
- GSF-National Research Center for Environment and Health, Institute of Experimental Genetics, Genome Analysis Center, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | | |
Collapse
|
195
|
Badrinarayanan R, Rengarajan S, Nithya P, Balasubramanian K. Corticosterone impairs the mRNA expression and activity of 3beta- and 17beta-hydroxysteroid dehydrogenases in adult rat Leydig cells. Biochem Cell Biol 2007; 84:745-54. [PMID: 17167538 DOI: 10.1139/o06-074] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Clinical and experimental studies, including our own observations, have shown the adverse effects of excess glucocorticoids on testicular steroid hormone production. The present study was designed to gain insight into the molecular mechanisms by which excess corticosterone impairs Leydig cell steroidogenesis. To achieve this, adult rats were administered with corticosterone-21-acetate (2 mg/100 g body weight) twice daily for 15 days. After the treatment period, rats were killed by decapitation. The testes were removed, decapsulated aseptically and used for the isolation of Leydig cells. Purified Leydig cells were used for assessing the activity of 3beta- and 17beta-hydroxysteroid dehydrogenases (HSDs) and total RNA isolation. For in vitro studies, purified Leydig cells (7.5 x 10(6) cells) of control rats were plated in culture flasks and exposed to different concentrations (50, 100, 200, 400, and 800 nmol/L) of corticosterone for 24 h. At the end of incubation, total RNA was isolated from cultured Leydig cells, and the mRNA of 3beta- and 17beta-HSDs was quantified by RT-PCR. A significant reduction in the activities and levels of 3beta-HSD type-I and 17beta-HSD type-III mRNAs in Leydig cells were observed. In vitro studies demonstrated a dose-dependent significant impairment in both the activity and mRNA expression of these enzymes. These results suggest that corticosterone might have a direct effect on the transcription of the genes of 3beta- and 17beta-HSD. It is inferred from the present in vivo and in vitro studies that one of the molecular mechanisms by which excess corticosterone decreases the steroidogenic potency of Leydig cells is by suppressing the mRNA expression of 3beta-HSD type-I and 17beta-HSD type-III enzymes.
Collapse
Affiliation(s)
- R Badrinarayanan
- Department of Endocrinology, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai 600113, Tamilnadu, India
| | | | | | | |
Collapse
|
196
|
Ings JS, Van Der Kraak GJ. Characterization of the mRNA expression of StAR and steroidogenic enzymes in zebrafish ovarian follicles. Mol Reprod Dev 2007; 73:943-54. [PMID: 16700073 DOI: 10.1002/mrd.20490] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The objective of this study was to investigate the levels of expression of steroid biosynthetic enzymes and steroidogenic acute regulatory protein (StAR) at different stages of ovarian follicular development in zebrafish (Danio rerio), and to investigate the sites within the steroid biosynthetic pathway that may be regulated by gonadotropins. Ovarian follicles of sexually mature fish were separated into primary, previtellogenic, vitellogenic, and mature stages and the expression of StAR, P450 side chain cleavage (P450scc), 3beta-hydroxysteroid dehydrogenase (3beta-HSD), P450 hydroxylase/lyase (P450c17), 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD1), 17beta-hydroxysteroid dehydrogenase type 3 (17beta-HSD3), and P450 aromatase (P450aromA) was determined by Real time RT-PCR. The expression of all genes changed significantly as follicles grew, with a decrease in the expression of StAR, P450scc, 3beta-HSD and P450c17 with maturation, and an increase in the expression of 17beta-HSD3 during vitellogenesis and 17beta-HSD1 and P450aromA during previtellogenesis. In vitro incubation of vitellogenic follicles demonstrated that the expression of StAR, 17beta-HSD3, and P450aromA increased in response to hCG, and decreased in the absence of hCG. In contrast, the expression of P450scc, 3beta-HSD, P450c17, and 17beta-HSD1 remained constant between treatments and over time. Testosterone and estradiol production in the culture medium was stimulated by human chorionic gonadotropin (hCG). These experiments aid in the characterization of the roles and regulation of steroids throughout ovarian development, and suggest that gonadotropins play a key role in the regulation of StAR, 17beta-HSD3, and P450aromA in zebrafish.
Collapse
Affiliation(s)
- Jennifer S Ings
- Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada
| | | |
Collapse
|
197
|
Faucher F, Cantin L, Pereira de Jésus-Tran K, Lemieux M, Luu-The V, Labrie F, Breton R. Mouse 17alpha-hydroxysteroid dehydrogenase (AKR1C21) binds steroids differently from other aldo-keto reductases: identification and characterization of amino acid residues critical for substrate binding. J Mol Biol 2007; 369:525-40. [PMID: 17442338 DOI: 10.1016/j.jmb.2007.03.058] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 03/20/2007] [Accepted: 03/20/2007] [Indexed: 11/27/2022]
Abstract
The mouse 17alpha-hydroxysteroid dehydrogenase (m17alpha-HSD) is the unique known member of the aldo-keto reductase (AKR) superfamily able to catalyze efficiently and in a stereospecific manner the conversion of androstenedione (Delta4) into epi-testosterone (epi-T), the 17alpha-epimer of testosterone. Structural and mutagenic studies had already identified one of the residues delineating the steroid-binding cavity, A24, as the major molecular determinant for the stereospecificity of m17alpha-HSD. We report here a ternary complex crystal structure (m17alpha-HSD:NADP(+):epi-T) determined at 1.85 A resolution that confirms this and reveals a unique steroid-binding mode for an AKR enzyme. Indeed, in addition to the interactions found in all other AKRs (van der Waals contacts stabilizing the core of the steroid and the hydrogen bonds established at the catalytic site by the Y55 and H117 residues with the oxygen atom of the ketone group to be reduced), m17alpha-HSD establishes with the other extremity of the steroid nucleus an additional interaction involving K31. By combining direct mutagenesis and kinetic studies, we found that the elimination of this hydrogen bond did not affect the affinity of the enzyme for its steroid substrate but led to a slight but significant increase of its catalytic efficiency (k(cat)/K(m)), suggesting a role for K31 in the release of the steroidal product at the end of the reaction. This previously unobserved steroid-binding mode for an AKR is similar to that adopted by other steroid-binding proteins, the hydroxysteroid dehydrogenases of the short-chain dehydrogenases/reductases (SDR) family and the steroid hormone nuclear receptors. Mutagenesis and structural studies made on the human type 3 3alpha-HSD, a closely related enzyme that shares 73% amino acids identity with the m17alpha-HSD, also revealed that the residue at position 24 of these two enzymes directly affects the binding and/or the release of NADPH, in addition to its role in their 17alpha/17beta stereospecificity.
Collapse
Affiliation(s)
- Frédérick Faucher
- Oncology and Molecular Endocrinology Research Center, Laval University Medical Center (CHUL), Laval University, Québec, Canada G1V 4G2
| | | | | | | | | | | | | |
Collapse
|
198
|
Lukacik P, Keller B, Bunkoczi G, Kavanagh K, Hwa Lee W, Adamski J, Oppermann U. Structural and biochemical characterization of human orphan DHRS10 reveals a novel cytosolic enzyme with steroid dehydrogenase activity. Biochem J 2007; 402:419-27. [PMID: 17067289 PMCID: PMC1863559 DOI: 10.1042/bj20061319] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
To this day, a significant proportion of the human genome remains devoid of functional characterization. In this study, we present evidence that the previously functionally uncharacterized product of the human DHRS10 gene is endowed with 17beta-HSD (17beta-hydroxysteroid dehydrogenase) activity. 17beta-HSD enzymes are primarily involved in the metabolism of steroids at the C-17 position and also of other substrates such as fatty acids, prostaglandins and xenobiotics. In vitro, DHRS10 converts NAD+ into NADH in the presence of oestradiol, testosterone and 5-androstene-3beta,17beta-diol. Furthermore, the product of oestradiol oxidation, oestrone, was identified in intact cells transfected with a construct plasmid encoding the DHRS10 protein. In situ fluorescence hybridization studies have revealed the cytoplasmic localization of DHRS10. Along with tissue expression data, this suggests a role for DHRS10 in the local inactivation of steroids in the central nervous system and placenta. The crystal structure of the DHRS10 apoenzyme exhibits secondary structure of the SDR (short-chain dehydrogenase/reductase) family: a Rossmann-fold with variable loops surrounding the active site. It also reveals a broad and deep active site cleft into which NAD+ and oestradiol can be docked in a catalytically competent orientation.
Collapse
Key Words
- crystal structure
- dhrs10
- 17β-hydroxysteroid dehydrogenase
- pre-receptor control
- short-chain dehydrogenase/reductase
- steroid metabolism
- dhea, dehydroepiandrosterone
- er, oestrogen receptor
- gfp, green fluorescent protein
- hek-293t cells, hek-293 cells (human embryonic kidney cells) expressing the large t-antigen of sv40 (simian virus 40)
- hsd, hydroxysteroid dehydrogenase
- mdr, medium-chain dehydrogenase/reductase
- ncs, non-crystallographic symmetry
- rmsd, root mean square deviation
- sdr, short-chain dehydrogenase/reductase
- tcep, tris-(2-carboxyethyl)phosphine
Collapse
Affiliation(s)
- Petra Lukacik
- *Structural Genomics Consortium, University of Oxford, Oxford OX3 7LD, U.K
- Correspondence may be addressed to either of these authors (email or )
| | - Brigitte Keller
- †GSF-National Research Center for Environment and Health, Institute for Experimental Genetics, Genome Analysis Center, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Gabor Bunkoczi
- *Structural Genomics Consortium, University of Oxford, Oxford OX3 7LD, U.K
| | - Kathryn Kavanagh
- *Structural Genomics Consortium, University of Oxford, Oxford OX3 7LD, U.K
| | - Wen Hwa Lee
- *Structural Genomics Consortium, University of Oxford, Oxford OX3 7LD, U.K
| | - Jerzy Adamski
- †GSF-National Research Center for Environment and Health, Institute for Experimental Genetics, Genome Analysis Center, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Udo Oppermann
- *Structural Genomics Consortium, University of Oxford, Oxford OX3 7LD, U.K
- Correspondence may be addressed to either of these authors (email or )
| |
Collapse
|
199
|
Kristan K, Stojan J, Adamski J, Lanisnik Rizner T. Rational design of novel mutants of fungal 17β-hydroxysteroid dehydrogenase. J Biotechnol 2007; 129:123-30. [PMID: 17196285 DOI: 10.1016/j.jbiotec.2006.11.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2004] [Revised: 07/26/2006] [Accepted: 08/03/2006] [Indexed: 11/29/2022]
Abstract
Reduction of 17-ketosteroids is a biocatalytic process of economic significance for the production of steroid drugs. This reaction can be catalyzed by different microbial 17beta-hydroxysteroid dehydrogenases (17beta-HSD), like the 17beta-HSD activity of Saccharomyces cerevisiae, Pichia faranosa and Mycobacterium sp., and by purified 3beta,17beta-HSD from Pseudomonas testosteroni. In addition to the bacterial 3beta,17beta-HSD the 17beta-HSD of the filamentous fungus Cochliobolus lunatus is the only microbial 17beta-HSD that has been expressed as a recombinant protein and fully characterized. On the basis of its modeled 3D structure, we selected several positions for the replacement of amino acids by site-directed mutagenesis to change substrate specificity, alter coenzyme requirements, and improve overall catalytic activity. Replacement of Val161 and Tyr212 in the substrate-binding region by Gly and Ala, respectively, increased the initial rates for the conversion of androstenedione to testosterone. Replacement of Tyr49 within the coenzyme binding site by Asp changed the coenzyme specificity of the enzyme. This latter mutant can convert the steroids not only in the presence of NADP(+) and NADPH, but also in the presence of NADH and NAD(+). The replacement of His164, located in the non-flexible part of the 'lid' covering the active center resulted in a conformation of the enzyme that possessed a higher catalytic activity.
Collapse
Affiliation(s)
- Katja Kristan
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | | | | | | |
Collapse
|
200
|
Van der Geize R, Yam K, Heuser T, Wilbrink MH, Hara H, Anderton MC, Sim E, Dijkhuizen L, Davies JE, Mohn WW, Eltis LD. A gene cluster encoding cholesterol catabolism in a soil actinomycete provides insight into Mycobacterium tuberculosis survival in macrophages. Proc Natl Acad Sci U S A 2007; 104:1947-52. [PMID: 17264217 PMCID: PMC1794314 DOI: 10.1073/pnas.0605728104] [Citation(s) in RCA: 400] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2006] [Indexed: 01/21/2023] Open
Abstract
Rhodococcus sp. strain RHA1, a soil bacterium related to Mycobacterium tuberculosis, degrades an exceptionally broad range of organic compounds. Transcriptomic analysis of cholesterol-grown RHA1 revealed a catabolic pathway predicted to proceed via 4-androstene-3,17-dione and 3,4-dihydroxy-9,10-seconandrost-1,3,5(10)-triene-9,17-dione (3,4-DHSA). Inactivation of each of the hsaC, supAB, and mce4 genes in RHA1 substantiated their roles in cholesterol catabolism. Moreover, the hsaC(-) mutant accumulated 3,4-DHSA, indicating that HsaC(RHA1), formerly annotated as a biphenyl-degrading dioxygenase, catalyzes the oxygenolytic cleavage of steroid ring A. Bioinformatic analyses revealed that 51 rhodococcal genes specifically expressed during growth on cholesterol, including all predicted to specify the catabolism of rings A and B, are conserved within an 82-gene cluster in M. tuberculosis H37Rv and Mycobacterium bovis bacillus Calmette-Guérin. M. bovis bacillus Calmette-Guérin grew on cholesterol, and hsaC and kshA were up-regulated under these conditions. Heterologously produced HsaC(H37Rv) and HsaD(H37Rv) transformed 3,4-DHSA and its ring-cleaved product, respectively, with apparent specificities approximately 40-fold higher than for the corresponding biphenyl metabolites. Overall, we annotated 28 RHA1 genes and proposed physiological roles for a similar number of mycobacterial genes. During survival of M. tuberculosis in the macrophage, these genes are specifically expressed, and many appear to be essential. We have delineated a complete suite of genes necessary for microbial steroid degradation, and pathogenic mycobacteria have been shown to catabolize cholesterol. The results suggest that cholesterol metabolism is central to M. tuberculosis's unusual ability to survive in macrophages and provide insights into potential targets for novel therapeutics.
Collapse
Affiliation(s)
- Robert Van der Geize
- *Department of Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9751 NN, Groningen, The Netherlands
| | - Katherine Yam
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada V6T 1Z3; and
| | - Thomas Heuser
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada V6T 1Z3; and
| | - Maarten H. Wilbrink
- *Department of Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9751 NN, Groningen, The Netherlands
| | - Hirofumi Hara
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada V6T 1Z3; and
| | - Matthew C. Anderton
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| | - Edith Sim
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| | - Lubbert Dijkhuizen
- *Department of Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9751 NN, Groningen, The Netherlands
| | - Julian E. Davies
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada V6T 1Z3; and
| | - William W. Mohn
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada V6T 1Z3; and
| | - Lindsay D. Eltis
- *Department of Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9751 NN, Groningen, The Netherlands
| |
Collapse
|