1
|
Hassan AA, Abdelgayed SS, Mansour SZ. Liver and ovarian toxicities boosted by bisphenol and gamma radiation in female albino rats. Hum Exp Toxicol 2024; 43:9603271231219264. [PMID: 38263794 DOI: 10.1177/09603271231219264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Bisphenol A (BPA), a carbon-based synthetic polymer compound, was newly classified as an environmental toxicant and an endocrine-disrupting chemical leading to abnormalities in cell proliferation, apoptosis, or migration that contributes to cancer development and progression. This study aims to evaluate the effect of the elevation of γ- radiation dose and BPA on the liver and ovaries of female rats. In this study, eighty female albino rats (130-150 g) were used in this work. Rats in this experiment received BPA in ethanol (50 mg/kg b. wt.) for 30 days, day after day, and in the irradiated groups, animals were administered BPA and then exposed to γ- radiation in doses (2, 4, and 6 Gy) one shot dose. Several members of the cytochrome family were examined. Exposure to γ-radiation and BPA showed an increase in cytochrome P450 and b5 fold change. Further, BPA and γ-radiation activate α and β estrogen receptors and also downregulate aromatase (CYT19) fold change. The current results also revealed that BPA and/or γ-radiation regulate the protein expression of the PI3K/Akt signaling pathway. The steroidogenic acute regulatory protein (StAR) appeared to be targeted by BPA and γ-radiation and its relative expression was elevated significantly by raising the γ-radiation dose. In conclusion, exposure to BPA, an endocrine-disrupting chemical, leads to marked toxicity. Additionally, toxicity is heightened by increasing the γ-radiation dose, either alone or in combination with BPA.
Collapse
Affiliation(s)
- Asmaa A Hassan
- Radiation Biology Department, National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Sherein S Abdelgayed
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Somaya Z Mansour
- Radiation Biology Department, National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
2
|
Sato N, Itakura Y, Yoshida Y, Akahira J, Fujishima F, Nakamura Y. Post-menopausal ovarian fibroma associated with steroid hormone synthesis: A case report. Taiwan J Obstet Gynecol 2023; 62:566-570. [PMID: 37407196 DOI: 10.1016/j.tjog.2023.01.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2023] [Indexed: 07/07/2023] Open
Abstract
OBJECTIVE Ovarian fibromas are benign, sex cord-stromal tumors occurring in both peri- and post-menopausal women. Generally, these tumors are non-functional and do not produce hormones. However, this case report proves the first case of steroid hormone synthesis in an ovarian fibroma by immunohistochemistry. CASE REPORT A 77-year-old post-menopausal woman presented with a left ovarian tumor, abnormal endometrial thickness, and high levels of estradiol (E2). The tumor was found to be a fibroma, which was positive for alpha-inhibin. We examined estrogen-producing enzymes using immunohistochemistry. The tumor was positive for estrogen receptor, progesterone receptor, 17β-hydroxysteroid dehydrogenase (HSD)-1, adrenal 4 binding protein/steroidogenic factor 1, 17β-HSD-5, steroid sulfatase, and P450c17. CONCLUSION This case study shows that E2 can be locally produced from circulating inactive steroids, by estrogen-producing enzymes. This is the first report of steroid hormone synthesis in an ovarian fibroma.
Collapse
Affiliation(s)
- Naomi Sato
- Iwate Prefectural Central Hospital, Department of Pathology, Morioka, Iwate, Japan; Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
| | - Yuko Itakura
- Japanese Red Cross Ishinomaki Hospital, Department of Pathology, Ishinomaki, Miyagi, Japan
| | - Yuji Yoshida
- Japanese Red Cross Ishinomaki Hospital, Department of Gynecology, Ishinomaki, Miyagi, Japan
| | - Junichi Akahira
- Sendai Kousei Hospital, Department of Pathology, Sendai, Miyagi, Japan
| | - Fumiyoshi Fujishima
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| |
Collapse
|
3
|
Abedal-Majed MA, Springman SA, Jafar HD, Bell BE, Kurz SG, Wilson KE, Cupp AS. Naturally occurring androgen excess cows are present in dairy and beef herds and have similar characteristics to women with PCOS. J Anim Sci 2022; 100:6596684. [PMID: 35648128 DOI: 10.1093/jas/skac151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023] Open
Abstract
Beef cows with excess androstenedione (A4; High A4) in follicular fluid (FF) and secreted by the ovarian cortex have been reported from the University of Nebraska-Lincoln physiology herd displaying characteristics reminiscent of polycystic ovary syndrome (PCOS). Thus, we hypothesized that naturally occurring High A4 cows were present in other dairy and beef herds. Fourteen Jordan (Amman, Jordon) dairy heifers and 16 U.S. Meat Animal Research Center beef heifers were classified by FF (High A4: A4 > 40 ng/mL and Control: A4 < 20 ng/mL) and/or cortex culture media (High A4 > 1 ng/mL/d or Control < 1 ng/mL/d). High A4 dairy heifers (n = 6) had greater A4 concentrations (7.6-fold) in FF and (98-fold) greater in ovarian cortex culture media with greater numbers of primordial and fewer later-stage follicles than Controls (n = 8) even after 7 d of culture. Also, the ovarian cortex had greater staining for Picro Sirius red in High A4 dairy heifers compared with Controls indicating increased fibrosis. Thecal cells from High A4 dairy heifers had greater STAR, LHCGR, CYP17A, CD68, and PECAM mRNA expression with increased mRNA abundance of CYP17A1 and CD68 in the ovarian cortex cultures compared with Control dairy heifers. Similarly, cortex culture media from High A4 beef heifers (n = 10) had increased A4 (290-fold; P ≤ 0.001), testosterone (1,427-fold; P ≤ 0.001), and progesterone (9-fold; P ≤ 0.01) compared with Control heifers with increased primordial follicles and decreased later-stage follicles even after 7 d of culture, indicating abnormal follicular development. High A4 ovarian cortex cultures from beef heifers also had increased fibrosis markers and greater expression of PECAM (P = 0.01) with a tendency for increased vascular endothelial cadherin compared with Controls (n = 6). These two trials support our hypothesis that naturally occurring androgen excess cows are present in other dairy and beef herds. The ability to identify these females that have excess A4 ovarian microenvironments may allow for their use in understanding factors causing abnormal follicle development linked to androgen excess and inflammation.
Collapse
Affiliation(s)
- Mohamed A Abedal-Majed
- Department of Animal Production, School of Agriculture, The University of Jordan, Amman, Jordan
| | - Shelby A Springman
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Hanan D Jafar
- Cell Therapy Center, The University of Jordan, Amman, Jordan.,Department of Anatomy and Histology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Brooke E Bell
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Scott G Kurz
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Kyle E Wilson
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Andrea S Cupp
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| |
Collapse
|
4
|
Abedal-Majed MA, Springman SA, Sutton CM, Snider AP, Bell BE, Hart M, Kurz SG, Bergman J, Summers AF, McFee RM, Davis JS, Wood JR, Cupp AS. VEGFA165 can rescue excess steroid secretion, inflammatory markers, and follicle arrest in the ovarian cortex of High A4 cows†. Biol Reprod 2022; 106:118-131. [PMID: 34726240 PMCID: PMC9630404 DOI: 10.1093/biolre/ioab201] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
A population of cows with excess androstenedione (A4; High A4) in follicular fluid, with follicular arrest, granulosa cell dysfunction, and a 17% reduction in calving rate was previously identified. We hypothesized that excess A4 in the ovarian microenvironment caused the follicular arrest in High A4 cows and that vascular endothelial growth factor A would rescue the High A4 phenotype. In trial 1, prior to culture, High A4 ovarian cortex (n = 9) had greater numbers of early stage follicles (primordial) and fewer later-stage follicles compared to controls (n = 11). Culture for 7 days did not relieve this follicular arrest; instead, High A4 ovarian cortex had increased indicators of inflammation, anti-Mullerian hormone, and A4 secretion compared to controls. In trial 2, we tested if vascular endothelial growth factor A isoforms could rescue the High A4 phenotype. High A4 (n = 5) and control (n = 5) ovarian cortex was cultured with (1) PBS, (2) VEGFA165 (50 ng/mL), (3) VEGFA165B (50 ng/mL), or (4) VEGFA165 + VEGFA165B (50 ng/mL each) for 7 days. Follicular progression increased with VEGFA165 in High A4 cows with greater early primary, primary, and secondary follicles than controls. Similar to trial 1, High A4 ovarian cortex secreted greater concentrations of A4 and other steroids and had greater indicators of inflammation compared to controls. However, VEGFA165 rescued steroidogenesis, oxidative stress, and fibrosis. The VEGFA165 and VEGFA165b both reduced IL-13, INFα, and INFβ secretion in High A4 cows to control levels. Thus, VEGFA165 may be a potential therapeutic to restore the ovarian steroidogenic microenvironment and may promote folliculogenesis.
Collapse
Affiliation(s)
- Mohamed A Abedal-Majed
- Department of Animal Production, School of Agriculture, University of Jordan, Amman-Jordan, Jordan
| | - Shelby A Springman
- Department of Animal Science, Animal Science Building, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Courtney M Sutton
- Department of Animal Science, Animal Science Building, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Alexandria P Snider
- Department of Animal Science, Animal Science Building, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Brooke E Bell
- Department of Animal Science, Animal Science Building, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | | - Scott G Kurz
- Department of Animal Science, Animal Science Building, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Jeff Bergman
- Department of Animal Science, Animal Science Building, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Adam F Summers
- Department of Animal and Range Sciences, New Mexico State University, Las Cruces, NM, USA
| | - Renee M McFee
- School of Veterinary and Biomedical Sciences, Veterinary Medicine and Biomedical Sciences Hall (VBS), University of Nebraska-Lincoln, Lincoln, NE, USA
| | - John S Davis
- Olson Center for Women’s Health, University of Nebraska Medical Center, Omaha, NE, USA
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Jennifer R Wood
- Department of Animal Science, Animal Science Building, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Andrea S Cupp
- Department of Animal Science, Animal Science Building, University of Nebraska-Lincoln, Lincoln, NE, USA
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| |
Collapse
|
5
|
Saini S, Bhat RA, Waiz HA, Waiz SA. A study on steroidogenic elaborations of stroma and their regulation in response to ovarian hormones in goats. Anim Reprod Sci 2021; 228:106748. [PMID: 33845412 DOI: 10.1016/j.anireprosci.2021.106748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 11/24/2022]
Abstract
Stromal tissue is an essential componenlt of the ovary not only for providing structural support but also for contributing to the early follicular growth with their bi-directional paracrine signaling. Estradiol is a major female hormone mainly secreted by the follicular cells in the ovary. To examine the relationship between 17β-estradiol and the factors involved in androgen production in stromal cells, ovarian stromal cells were cultured in the graded concentrations (50 and 100 ng/mL) of 17β-estradiol for varying time periods (24 and 48 h). The cells were processed for transmission electron microscopy to study the changes in steroidogenic functions of the cells. The effect of estradiol treatment was also evaluated on the quantity of androgen production and abundance of steroidogenic enzymes and proteins. The results indicated 17β-estradiol increased androgen production in ovarian stromal cells. In addition to enhanced androstenedione and testosterone production, estradiol stimulation was also based on the marked increase in abundance of mRNA transcript of steroidogenic enzymes [Star (Steroidogenic Acute Regulatory Protein), Cyp11a1, Cyp17a1, and hsd3b1 (3β-hydroxysteroid dehydrogenase)], as well as abundances of StAR and CYP11A1 protein. Thus, 17β-estradiol enhanced steroidogenesis in ovarian stromal cells. This study provided a basis for further exploration of regulation of steroidogenesis in ovarian stromal cells and the feedback mechanisms in association with estradiol.
Collapse
Affiliation(s)
- Sudha Saini
- Department of Zoology, Kurukshetra University, Kurukshetra, 136119, India
| | - Rayees Ahmad Bhat
- Department of Zoology, Kurukshetra University, Kurukshetra, 136119, India.
| | - Hina Ashraf Waiz
- Assistant Professor Livestock Production and Management CVAS, Navania, Udaipur, Rajasthan University of Veterinary and Animal Sciences Bikaner, India
| | | |
Collapse
|
6
|
New insights into the GDF9-Hedgehog-GLI signaling pathway in human ovaries: from fetus to postmenopause. J Assist Reprod Genet 2021; 38:1387-1403. [PMID: 33772413 DOI: 10.1007/s10815-021-02161-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/18/2021] [Indexed: 10/21/2022] Open
Abstract
RESEARCH QUESTION Are glioma-associated oncogene homolog 1, 2, and 3 (GLI1, 2, and 3) and protein patched homolog 1 (PTCH1) specific markers for precursor theca cells in human ovaries as in mouse ovaries? DESIGN To study the GDF9-HH-GLI pathway and assess whether GLI1 and 3 and PTCH1 are specific markers for precursor theca cells in the human ovary, growth differentiation factor 9 (GDF9), Indian Hedgehog (IHH), Desert Hedgehog (DHH), Sonic Hedgehog (SHH), PTCH1 and GLI1, 2 and 3 were investigated in fetal (n=9), prepubertal (n=9), reproductive-age (n=15), and postmenopausal (n=8) human ovarian tissue. Immunohistochemistry against GDF9, IHH, DHH, SHH, PTCH1, GLI1, GLI2, and GLI3 was performed on human ovarian tissue sections fixed in 4% formaldehyde and embedded in paraffin. Western blotting was carried out on extracted proteins from the same samples used in the previous step to prove the antibodies' specificity. The quantitative real-time polymerase chain reaction was performed to identify mRNA levels for Gdf9, Ihh, Gli1, Gli2, and Gli3 in menopausal ovaries. RESULTS Our results showed that, in contrast to mice, all studied proteins were expressed in primordial follicles of fetal, prepubertal, and reproductive-age human ovaries and stromal cells of reproductive-age and postmenopausal ovaries. Intriguingly, Gdf9, Ihh, and Gli3 mRNA, but not Gli1 and 2, was detected in postmenopausal ovaries. Moreover, GLI1, GLI3, and PTCH1 are not limited to a specific population of cells. They were spread throughout the organ, which means they are not specific markers for precursor theca cells in human ovaries. CONCLUSION These results could provide a basis for understanding how this pathway modulates follicle development and ovarian cell steroidogenesis in human ovaries.
Collapse
|
7
|
Prenatal and pubertal exposure to 17α-ethinylestradiol disrupts folliculogenesis and promotes morphophysiological changes in ovaries of old gerbils ( Meriones unguiculatus). J Dev Orig Health Dis 2021; 13:49-60. [PMID: 33650479 DOI: 10.1017/s2040174421000040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
17α-Ethinylestradiol is an endocrine-disrupting chemical that make up most contraceptive pills and can be found in the environment. Exposure to ethinylestradiol in different development periods may promote changes in morphophysiological parameters of reproductive and endocrine organs. Considering that the effects of low doses (15 µg/kg/day) of ethinylestradiol in ovaries from 12-month-old female gerbils (Meriones unguiculatus) were investigated. Four experimental groups used were control (without treatment), EE/PRE (treated from the 18th to the 22nd gestational day), EE/PUB (treated from the 42nd to the 49th day of life), and EE/PRE-PUB (treated in the both periods). The animals were euthanized at 12 months. Testosterone and 17β-estradiol levels were measured. The ovaries were stained with Hematoxylin and Eosin, Periodic Acid Schiff, and Gomori's Trichome. The follicles, corpus luteum, interstitial gland, lipofuscin, ovarian epithelium, and tunica albuginea were analyzed. Estradiol was higher in EE/PRE and EE/PUB groups, while testosterone was higher only in EE/PUB group. The main changes in follicle count occurred in EE/PUB and EE/PRE-PUB groups, with higher primordial follicle count and lower maturation of follicles. The corpus luteum was more evident in EE/PRE group. No differences were found in atretic follicles count. A higher area occupied by interstitial gland cells and lipofuscin deposit in these cells was noted in EE/PUB and EE/PRE-PUB groups. Higher epithelium height and thicker tunic albuginea were showed in treated groups. These results suggest that exposure to doses of EE2 in prenatal and pubertal periods of the development leads to morphological changes in senile ovaries.
Collapse
|
8
|
Creevey L, Bleach R, Madden SF, Toomey S, Bane FT, Varešlija D, Hill AD, Young LS, McIlroy M. Altered Steroid Milieu in AI-Resistant Breast Cancer Facilitates AR Mediated Gene-Expression Associated with Poor Response to Therapy. Mol Cancer Ther 2019; 18:1731-1743. [PMID: 31289138 DOI: 10.1158/1535-7163.mct-18-0791] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/23/2018] [Accepted: 07/03/2019] [Indexed: 11/16/2022]
Abstract
Divergent roles for androgen receptor (AR) in breast cancer have been reported. Following aromatase inhibitor (AI) treatment, the conversion of circulating androgens into estrogens can be diminished by >99%. We wished to establish whether the steroid environment can dictate the role of AR and the implications of this for subsequent therapy. This study utilizes models of AI resistance to explore responsiveness to PI3K/mTOR and anti-AR therapy when cells are exposed to unconverted weak androgens. Transcriptomic alterations driven by androstenedione (4AD) were assessed by RNA-sequencing. AR and estrogen receptor (ER) recruitment to target gene promoters was evaluated using ChIP, and relevance to patient profiles was performed using publicly available data sets. Although BEZ235 showed decreased viability across AI-sensitive and -resistant cell lines, anti-AR treatment elicited a decrease in cell viability only in the AI-resistant model. Serum and glucocorticoid-regulated kinase 3 (SGK3) and cAMP-dependent protein kinase inhibitor β (PKIB) were confirmed to be regulated by 4AD and shown to be mediated by AR; crucially, reexposure to estradiol suppressed expression of these genes. Meta-analysis of transcript levels showed high expression of SGK3 and PKIB to be associated with poor response to endocrine therapy (HR = 2.551, P = 0.003). Furthermore, this study found levels of SGK3 to be sustained in patients who do not respond to AI therapy. This study highlights the importance of the tumor steroid environment. SGK3 and PKIB are associated with poor response to endocrine therapy and could have utility in tailoring therapeutic approaches.
Collapse
Affiliation(s)
- Laura Creevey
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Rachel Bleach
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Stephen F Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Sinead Toomey
- Department of Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Fiona T Bane
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Damir Varešlija
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Arnold D Hill
- Department of Surgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Leonie S Young
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Marie McIlroy
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.
| |
Collapse
|
9
|
Nanba AT, Rege J, Ren J, Auchus RJ, Rainey WE, Turcu AF. 11-Oxygenated C19 Steroids Do Not Decline With Age in Women. J Clin Endocrinol Metab 2019; 104:2615-2622. [PMID: 30753518 PMCID: PMC6525564 DOI: 10.1210/jc.2018-02527] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/05/2019] [Indexed: 01/21/2023]
Abstract
CONTEXT The ovaries and adrenals are sources of androgens in women. Although dehydroepiandrosterone (DHEA), DHEA sulfate (DHEAS), and testosterone (T) all decline with age, these C19 steroids correlate poorly with parameters of androgen action in postmenopausal women. OBJECTIVE To comprehensively compare the androgen profiles of pre- and postmenopausal women. METHODS We quantified 19 steroids-including DHEA; DHEAS; T; androstenedione (A4); and the following adrenal-specific 11-oxygenated C19 steroids (11oxyandrogens): 11β-hydroxytestosterone (11OHT), 11-ketotestosterone (11KT), 11β-hydroxyandrostenedione (11OHA4), and 11-ketoandrostenedione (11KA4)-using liquid chromatography-tandem mass spectrometry in morning serum obtained from 100 premenopausal (age 20 to 40 years) and 100 postmenopausal (age ≥ 60 years) women. Double immunofluorescence of 3β-hydroxysteroid dehydrogenase type 2 (HSD3B2) with cytochrome b5 (CYB5A) or sulfotransferase 2A1 (SULT2A1) was performed in normal adrenal glands obtained from eight premenopausal and eight postmenopausal women. RESULTS DHEA, DHEAS, A4, and T were significantly higher in pre- than in postmenopausal women (2.9, 2.8, 2.9, and 1.6-fold, respectively; P < 0.0001). In contrast, the 11-oxyandrogens did not decrease with aging, and the 11OHT/T and 11OHA4/A4 ratios showed strong positive correlations with age (r = 0.5 and 0.8, respectively; P < 0.0001). Double immunofluorescence analysis showed that with the involution of the zona reticularis in the old adrenals, the sharp zonal segregation of HSD3B2 and CYB5A becomes less distinct, and areas of HSD3B2 and CYB5A overlap are observed. CONCLUSIONS Unlike DHEA, DHEAS, A4, and T, the 11oxyandrogens do not decline in aging women. Structural changes within the adrenal cortex might explain the evolution of androgen profiles in aging women.
Collapse
Affiliation(s)
- Aya T Nanba
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - Juilee Rege
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Jianwei Ren
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - Richard J Auchus
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | - William E Rainey
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Adina F Turcu
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan
- Correspondence and Reprint Requests: Adina F. Turcu, MD, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, 1150 W Medical Center Drive, MSRB II, 5570B, Ann Arbor, Michigan 48109. E-mail:
| |
Collapse
|
10
|
Bleach R, McIlroy M. The Divergent Function of Androgen Receptor in Breast Cancer; Analysis of Steroid Mediators and Tumor Intracrinology. Front Endocrinol (Lausanne) 2018; 9:594. [PMID: 30416486 PMCID: PMC6213369 DOI: 10.3389/fendo.2018.00594] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/19/2018] [Indexed: 12/16/2022] Open
Abstract
Androgen receptor (AR) is the most widely expressed steroid receptor protein in normal breast tissue and is detectable in approximately 90% of primary breast cancers and 75% of metastatic lesions. However, the role of AR in breast cancer development and progression is mired in controversy with evidence suggesting it can either inhibit or promote breast tumorigenesis. Studies have shown it to antagonize estrogen receptor alpha (ERα) DNA binding, thereby preventing pro-proliferative gene transcription; whilst others have demonstrated AR to take on the mantle of a pseudo ERα particularly in the setting of triple negative breast cancer. Evidence for a potentiating role of AR in the development of endocrine resistant breast cancer has also been mounting with reports associating high AR expression with poor response to endocrine treatment. The resurgence of interest into the function of AR in breast cancer has resulted in various emergent clinical trials evaluating anti-AR therapy and selective androgen receptor modulators in the treatment of advanced breast cancer. Trials have reported varied response rates dependent upon subtype with overall clinical benefit rates of ~19-29% for anti-androgen monotherapy, suggesting that with enhanced patient stratification AR could prove efficacious as a breast cancer therapy. Androgens and AR have been reported to facilitate tumor stemness in some cancers; a process which may be mediated through genomic or non-genomic actions of the AR, with the latter mechanism being relatively unexplored in breast cancer. Steroidogenic ligands of the AR are produced in females by the gonads and as sex-steroid precursors secreted from the adrenal glands. These androgens provide an abundant reservoir from which all estrogens are subsequently synthesized and their levels are undiminished in the event of standard hormonal therapeutic intervention in breast cancer. Steroid levels are known to be altered by lifestyle factors such as diet and exercise; understanding their potential role in dictating the function of AR in breast cancer development could therefore have wide-ranging effects in prevention and treatment of this disease. This review will outline the endogenous biochemical drivers of both genomic and non-genomic AR activation and how these may be modulated by current hormonal therapies.
Collapse
Affiliation(s)
| | - Marie McIlroy
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
11
|
McKinnon B, Mueller M, Montgomery G. Progesterone Resistance in Endometriosis: an Acquired Property? Trends Endocrinol Metab 2018; 29:535-548. [PMID: 29934050 DOI: 10.1016/j.tem.2018.05.006] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/25/2018] [Accepted: 05/29/2018] [Indexed: 12/21/2022]
Abstract
Endometriosis is the growth of endometrial tissue outside the uterus and is characterized by progesterone resistance and changes in global and progesterone target gene expression. However, the mechanism behind this and whether it is innate, acquired, or present in both the eutopic and ectopic tissue in not always clear. We find large-scale gene expression studies in eutopic tissue, indicative of progesterone resistance, are often contradictory, potentially due to the dynamic nature of this tissue, whereas suppressed progesterone receptor expression is supported in ectopic but not eutopic tissue. This suggests more studies are required in eutopic tissue particularly, and that potentially the suppressed progesterone receptor (PR) expression is a consequence of the pathogenic process and exposure to the peritoneal environment.
Collapse
Affiliation(s)
- Brett McKinnon
- Department of Gynecology and Obstetrics, Frauenklinik, Inselspital Bern, Switzerland.
| | - Michael Mueller
- Department of Gynecology and Obstetrics, Frauenklinik, Inselspital Bern, Switzerland
| | - Grant Montgomery
- Genomics of Reproductive Disorders, Institute for Molecular Bioscience, University of Queensland, Australia
| |
Collapse
|
12
|
Tenkorang MA, Snyder B, Cunningham RL. Sex-related differences in oxidative stress and neurodegeneration. Steroids 2018; 133:21-27. [PMID: 29274405 PMCID: PMC5864532 DOI: 10.1016/j.steroids.2017.12.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/13/2017] [Accepted: 12/18/2017] [Indexed: 02/07/2023]
Abstract
Oxidative stress has been implicated in a number of neurodegenerative diseases spanning various fields of research. Reactive oxygen species can be beneficial or harmful, depending on their concentration. High levels of reactive oxygen species can lead to oxidative stress, which is an imbalance between free radicals and antioxidants. Increased oxidative stress can result in cell loss. Interestingly, sex differences have been observed in oxidative stress generation, which may underlie sex differences observed in neurodegenerative disorders. An enhanced knowledge of the role of sex hormones on oxidative stress signaling and cell loss can yield valuable information, leading to sex-based mechanistic approaches to neurodegeneration.
Collapse
Affiliation(s)
- Mavis A Tenkorang
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Brina Snyder
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Rebecca L Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States.
| |
Collapse
|
13
|
Parity-Dependent Hemosiderin and Lipofuscin Accumulation in the Reproductively Aged Mouse Ovary. Anal Cell Pathol (Amst) 2018; 2018:1289103. [PMID: 29736365 PMCID: PMC5874974 DOI: 10.1155/2018/1289103] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/08/2018] [Indexed: 01/10/2023] Open
Abstract
The progressive decline of the ovarian follicle pool leads to reproductive ageing. The latter is accompanied by age-related disorders, including various types of cancer. In fact, the highest rates of ovarian cancer (OC) occur at postmenopause while OC risk is significantly modulated by parity records during previous fertile life. We approached the age-parity relationship in the C57BL/6 mouse model and herein describe the presence of nonheme iron (hemosiderin) and deposits of the "age pigment" lipofuscin in reproductively aged mouse ovaries by applying conventional histochemical methods and autofluorescence. In addition, the 8-OHdG adduct was evaluated in ovarian genomic DNA. Both hemosiderin and lipofuscin were significantly higher in virgin compared to multiparous ovaries. The same pattern was observed for 8-OHdG. We conclude that nulliparity induces a long-term accumulation of iron and lipofuscin with concomitant oxidative damage to DNA in the mouse ovary. Since lipofuscin is a widely accepted senescence marker and given the recently postulated role of lipofuscin-associated iron as a source of reactive oxygen species (ROS) in senescent cells, these findings suggest a possible pathogenic mechanism by which nulliparity contributes to an increased OC risk in the postmenopausal ovary.
Collapse
|
14
|
Atwood CS, Hayashi K, Meethal SV, Gonzales T, Bowen RL. Does the degree of endocrine dyscrasia post-reproduction dictate post-reproductive lifespan? Lessons from semelparous and iteroparous species. GeroScience 2017; 39:103-116. [PMID: 28271270 PMCID: PMC5352586 DOI: 10.1007/s11357-016-9955-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 12/23/2016] [Indexed: 12/16/2022] Open
Abstract
Post-reproductive lifespan varies greatly among species; human post-reproductive lifespan comprises ~30-50% of their total longevity, while semelparous salmon and dasyurid marsupials post-reproductive lifespan comprises <4% of their total longevity. To examine if the magnitude of hypothalamic-pituitary-gonadal (HPG) axis dyscrasia at the time of reproductive senescence determines post-reproductive lifespan, we examined the difference between pre- and post-reproductive (1) circulating sex hormones and (2) the ratio of sex steroids to gonadotropins (e.g., 17β-estradiol/follicle-stimulating hormone (FSH)), an index of the dysregulation of the HPG axis and the level of dyotic (death) signaling post-reproduction. Animals with a shorter post-reproductive lifespan (<4% total longevity) had a more marked decline in circulating sex steroids and corresponding elevation in gonadotropins compared to animals with a longer post-reproductive lifespan (30-60% total longevity). In semelparous female salmon of short post-reproductive lifespan (1%), these divergent changes in circulating hormone concentration post-reproduction equated to a 711-fold decrease in the ratio of 17β-estradiol/FSH between the reproductive and post-reproductive periods. In contrast, the decrease in the ratio of 17β-estradiol/FSH in iteroparous female mammals with long post-reproductive lifespan was significantly less (1.7-34-fold) post-reproduction. Likewise, in male semelparous salmon, the decrease in the ratio of testosterone/FSH (82-fold) was considerably larger than for iteroparous species (1.3-11-fold). These results suggest that (1) organisms with greater reproductive endocrine dyscrasia more rapidly undergo senescence and die, and (2) the contribution post-reproduction by non-gonadal (and perhaps gonadal) tissues to circulating sex hormones dictates post-reproductive tissue health and longevity. In this way, reproduction and longevity are coupled, with the degree of non-gonadal tissue hormone production dictating the rate of somatic tissue demise post-reproduction and the differences in post-reproductive lifespans between species.
Collapse
Affiliation(s)
- Craig S Atwood
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, William S. Middleton Memorial VA (GRECC 11G), 2500 Overlook Terrace, Madison, WI, 53705, USA.
- Geriatric Research, Education and Clinical Center, Veterans Administration Hospital, Madison, WI, 53705, USA.
- School of Exercise, Biomedical and Health Sciences, Edith Cowan University, Joondalup, WA, 6027, Australia.
| | - Kentaro Hayashi
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, William S. Middleton Memorial VA (GRECC 11G), 2500 Overlook Terrace, Madison, WI, 53705, USA
| | - Sivan Vadakkadath Meethal
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, William S. Middleton Memorial VA (GRECC 11G), 2500 Overlook Terrace, Madison, WI, 53705, USA
| | - Tina Gonzales
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, William S. Middleton Memorial VA (GRECC 11G), 2500 Overlook Terrace, Madison, WI, 53705, USA
| | - Richard L Bowen
- Department of Psychiatry, Medical University of South Carolina, Charleston, SC, 29425, USA
| |
Collapse
|
15
|
Liu W, Wang LY, Xing XX, Fan GW. Conditions and possible mechanisms of VCD-induced ovarian failure. Altern Lab Anim 2016; 43:385-92. [PMID: 26753941 DOI: 10.1177/026119291504300606] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Perimenopause is an important period in women's lives, in which they experience a series of physiological changes. Current animal models of perimenopause fail to adequately replicate this particular stage in female life, while current in vitro models are too simplistic and cannot account for systemic effects. Neither the naturally-ageing animal model, nor the ovariectomised animal model, mimic the natural transitional process that is the menopause. In vivo and in vitro studies have confirmed that the occupational chemical, 4-vinylcyclohexene diepoxide (VCD), can cause selective destruction of the ovarian primordial and primary follicles of rats and mice by accelerating the apoptotic process, which successfully mimics the perimenopausal state in women. However, it is the in vivo VCD-induced rodent perimenopausal models that are currently the most widely used in research, rather than any of the available in vitro models. Studies on the mechanisms involved have found that VCD induces ovotoxicity via interference with the c-kit/kit ligand and apoptotic signalling pathways, among others. Overall, the VCD-induced perimenopausal animal models have provided some insight into female perimenopause, but they are far from ideal models of the human situation.
Collapse
Affiliation(s)
- Wei Liu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ling-Yan Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiao-Xue Xing
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guan-Wei Fan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
16
|
Labrie F. All sex steroids are made intracellularly in peripheral tissues by the mechanisms of intracrinology after menopause. J Steroid Biochem Mol Biol 2015; 145:133-8. [PMID: 24923731 DOI: 10.1016/j.jsbmb.2014.06.001] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 05/21/2014] [Accepted: 06/06/2014] [Indexed: 11/22/2022]
Abstract
Following the arrest of estradiol secretion by the ovaries at menopause, all estrogens and all androgens in postmenopausal women are made locally in peripheral target tissues according to the physiological mechanisms of intracrinology. The locally made sex steroids exert their action and are inactivated intracellularly without biologically significant release of the active sex steroids in the circulation. The level of expression of the steroid-forming and steroid-inactivating enzymes is specific to each cell type in each tissue, thus permitting to each cell/tissue to synthesize a small amount of androgens and/or estrogens in order to meet the local physiological needs without affecting the other tissues of the organism. Achieved after 500 million years of evolution, combination of the arrest of ovarian estrogen secretion, the availability of high circulating levels of DHEA and the expression of the peripheral sex steroid-forming enzymes have permitted the appearance of menopause with a continuing access to intratissular sex steroids for the individual cells/tissues without systemic exposure to circulating estradiol. In fact, one essential condition of menopause is to maintain serum estradiol at biologically inactive (substhreshold) concentrations, thus avoiding stimulation of the endometrium and risk of endometrial cancer. Measurement of the low levels of serum estrogens and androgens in postmenopausal women absolutely requires the use of MS/MS-based technology in order to obtain reliable accurate, specific and precise assays. While the activity of the series of steroidogenic enzymes can vary, the serum levels of DHEA show large individual variations going from barely detectable to practically normal "premenopausal" values, thus explaining the absence of menopausal symptoms in about 25% of women. It should be added that the intracrine system has no feedback elements to adjust the serum levels of DHEA, thus meaning that women with low DHEA activity will not be improved without external supplementation. Exogenous DHEA, however, follows the same intracrine rules as described for endogenous DHEA, thus maintaining serum estrogen levels at substhreshold or biologically inactive concentrations. Such blood concentrations are not different from those observed in normal postmenopausal women having high serum DHEA concentrations. Androgens, on the other hand, are practically all made intracellularly from DHEA by the mechanisms of intracrinology and are always maintained at very low levels in the blood in both pre- and postmenopausal women. Proof of the importance of intracrinology is also provided, among others, by the well-recognized benefits of aromatase inhibitors and antiestrogens used successfully for the treatment of breast cancer in postmenopausal women where all estrogens are made locally. Each medical indication for the use of DHEA, however, requires clinical trials performed according to the FDA guidelines and the best rules of clinical medicine.
Collapse
|
17
|
Panoulis K, Christantoni E, Pliatsika P, Anagnostis P, Goulis DG, Kondi-Pafiti A, Armeni E, Augoulea A, Triantafyllou N, Creatsa M, Lambrinoudaki I. Expression of gonadal steroid receptors in the ovaries of post-menopausal women with malignant or benign endometrial pathology: a pilot study. Gynecol Endocrinol 2015; 31:613-7. [PMID: 26036714 DOI: 10.3109/09513590.2015.1021324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
This pilot study aimed to investigate the expression of estrogen (ER) and progesterone receptors (PR), as well as their subtypes [alpha (ERα), beta (ERβ)], in the ovaries of postmenopausal women with benign or malignant endometrial pathology. Twenty postmenopausal women (age 66.2 ± 7.4 years) were included, diagnosed with benign (n = 10) or malignant [(serous/papillary (n = 4), endometrioid (n = 6)] endometrial lesions. Higher ERβ and PR ovarian expressions were observed comparing women with endometrioid versus non-endometrioid endometrial carcinoma (p = 0.022 and p = 0.029, respectively). Age, age at menarche and presence of hypertension were negatively associated with ERs and PR expression. The expression of ERα and ERβ was inversely correlated with menopausal age, which was not verified for PR. No significant association was observed between ERs or PR expression and benign or malignant endometrial pathology. Higher expression of ERβ and PR in the postmenopausal ovary is associated with the presence of a less aggressive type of endometrial cancer, comparing women with endometrioid versus non-endometrioid lesions. The expression pattern of ovarian receptors did not differ regarding the development of benign or malignant endometrial lesions. Larger observational studies are necessary to confirm the significance of our findings.
Collapse
Affiliation(s)
- Konstantinos Panoulis
- a Second Department of Obstetrics and Gynecology , Aretaieio Hospital, National and Kapodistrian University of Athens , Athens , Greece
| | - Evanthia Christantoni
- a Second Department of Obstetrics and Gynecology , Aretaieio Hospital, National and Kapodistrian University of Athens , Athens , Greece
| | - Paraskevi Pliatsika
- a Second Department of Obstetrics and Gynecology , Aretaieio Hospital, National and Kapodistrian University of Athens , Athens , Greece
| | - Panagiotis Anagnostis
- b Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology , Medical School, Aristotle University of Thessaloniki , Thessaloniki , Greece , and
| | - Dimitrios G Goulis
- b Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology , Medical School, Aristotle University of Thessaloniki , Thessaloniki , Greece , and
| | - Agathi Kondi-Pafiti
- a Second Department of Obstetrics and Gynecology , Aretaieio Hospital, National and Kapodistrian University of Athens , Athens , Greece
| | - Eleni Armeni
- a Second Department of Obstetrics and Gynecology , Aretaieio Hospital, National and Kapodistrian University of Athens , Athens , Greece
| | - Areti Augoulea
- a Second Department of Obstetrics and Gynecology , Aretaieio Hospital, National and Kapodistrian University of Athens , Athens , Greece
| | - Nikolaos Triantafyllou
- c First Neurology Department , Aiginiteion Hospital, National and Kapodistrian University of Athens , Athens , Greece
| | - Maria Creatsa
- a Second Department of Obstetrics and Gynecology , Aretaieio Hospital, National and Kapodistrian University of Athens , Athens , Greece
| | - Irene Lambrinoudaki
- a Second Department of Obstetrics and Gynecology , Aretaieio Hospital, National and Kapodistrian University of Athens , Athens , Greece
| |
Collapse
|
18
|
Brodowska A, Brodowski J, Laszczyńska M, Słuczanowska-Głąbowska S, Rumianowski B, Rotter I, Starczewski A, Ratajczak MZ. Immunoexpression of aromatase cytochrome P450 and 17β-hydroxysteroid dehydrogenase in women's ovaries after menopause. J Ovarian Res 2014; 7:52. [PMID: 24855493 PMCID: PMC4030461 DOI: 10.1186/1757-2215-7-52] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 05/05/2014] [Indexed: 11/25/2022] Open
Abstract
Background Menopause results in a lack of regular menstrual cycles, leading to the reduction of estrogen production. On the other hand, ovarian androgen synthesis is still present at reduced levels and requires expression of several steroidogenic enzymes. Methods This study was performed on 104 postmenopausal women hospitalized due to uterine leiomyomas, endometriosis, and/or a prolapsed uterus. Patients were divided into three groups depending on the time from menopause. Group A patients experienced menopause 1–5 years before enrollment in the study (42 women). Group B included women who had their last menstruation 5–10 years before the study (40 women). Group C consisted of 22 women who were more than 10 years past menopause. Hysterectomy or removal of the uterine corpus with adnexa was performed during laparotomy. We evaluated the expression of aromatase cytochrome P450 (CYP 19) and 17β-hydroxysteroid dehydrogenase (17β HSD) by employing immunohistochemistry. Results Activity of 17β-HSD and CYP19 was demonstrated in the cytoplasm of stromal cells of postmenopausal ovaries, epithelium cells coating the ovaries, vascular endothelial cells, and epithelial inclusion cysts. However, overall expression of both 17β-HSD and CYP 19 decreased with time after menopause. Conclusion Demonstration of the activity of the key enzymes of ovarian steroidogenesis, CYP 19 and 17β-HSD, confirms steroidogenic activity in the ovaries of postmenopausal women. Nevertheless, ovarian steroidogenic activity decreases with time, and its significant decrease occurs 10 years after menopause.
Collapse
Affiliation(s)
- Agnieszka Brodowska
- Department of Gynaecology and Urogynaecology, Pomeranian Medical University, Siedlecka 2, 72 - 010 Police, Poland
| | - Jacek Brodowski
- Laboratory of Primary Health Care, Pomeranian Medical University, Żołnierska 48, 71-210 Szczecin, Poland
| | - Maria Laszczyńska
- Department of Histology and Developmental Biology, Pomeranian Medical University, Żołnierska 48, 71-210 Szczecin, Poland
| | | | - Bogdan Rumianowski
- Department of Histology and Developmental Biology, Pomeranian Medical University, Żołnierska 48, 71-210 Szczecin, Poland
| | - Iwona Rotter
- Laboratory of Medical Rehabilitation, Pomeranian Medical University, Grudziądzka 31, 70-103 Szczecin, Poland
| | - Andrzej Starczewski
- Department of Gynaecology and Urogynaecology, Pomeranian Medical University, Siedlecka 2, 72 - 010 Police, Poland
| | - Mariusz Z Ratajczak
- Department of Physiology Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland ; Stem Cell Biology Program, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| |
Collapse
|
19
|
Craig ZR, Hannon PR, Flaws JA. Pregnenolone co-treatment partially restores steroidogenesis, but does not prevent growth inhibition and increased atresia in mouse ovarian antral follicles treated with mono-hydroxy methoxychlor. Toxicol Appl Pharmacol 2013; 272:780-6. [PMID: 23948739 PMCID: PMC3805676 DOI: 10.1016/j.taap.2013.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 07/09/2013] [Accepted: 08/02/2013] [Indexed: 11/26/2022]
Abstract
Mono-hydroxy methoxychlor (mono-OH MXC) is a metabolite of the pesticide, methoxychlor (MXC). Although MXC is known to decrease antral follicle numbers, and increase follicle death in rodents, not much is known about the ovarian effects of mono-OH MXC. Previous studies indicate that mono-OH MXC inhibits mouse antral follicle growth, increases follicle death, and inhibits steroidogenesis in vitro. Further, previous studies indicate that CYP11A1 expression and production of progesterone (P4) may be the early targets of mono-OH MXC in the steroidogenic pathway. Thus, this study tested whether supplementing pregnenolone, the precursor of progesterone and the substrate for HSD3B, would prevent decreased steroidogenesis, inhibited follicle growth, and increased follicle atresia in mono-OH MXC-treated follicles. Mouse antral follicles were exposed to vehicle (dimethylsulfoxide), mono-OH MXC (10 μg/mL), pregnenolone (1 μg/mL), or mono-OH MXC and pregnenolone together for 96 h. Levels of P4, androstenedione (A), testosterone (T), estrone (E1), and 17β-estradiol (E2) in media were determined, and follicles were processed for histological evaluation of atresia. Pregnenolone treatment alone stimulated production of all steroid hormones except E2. Mono-OH MXC-treated follicles had decreased sex steroids, but when given pregnenolone, produced levels of P4, A, T, and E1 that were comparable to those in vehicle-treated follicles. Pregnenolone treatment did not prevent growth inhibition and increased atresia in mono-OH MXC-treated follicles. Collectively, these data support the idea that the most upstream effect of mono-OH MXC on steroidogenesis is by reducing the availability of pregnenolone, and that adding pregnenolone may not be sufficient to prevent inhibited follicle growth and survival.
Collapse
Affiliation(s)
- Zelieann R. Craig
- Department of Comparative Biosciences, University of Illinois, 2001 S. Lincoln Ave, Urbana, IL, USA
| | - Patrick R. Hannon
- Department of Comparative Biosciences, University of Illinois, 2001 S. Lincoln Ave, Urbana, IL, USA
| | - Jodi A. Flaws
- Department of Comparative Biosciences, University of Illinois, 2001 S. Lincoln Ave, Urbana, IL, USA
| |
Collapse
|
20
|
Dehydroepiandrosterone sulfate levels reflect endogenous luteinizing hormone production and response to human chorionic gonadotropin challenge in older female macaque (Macaca fascicularis). Menopause 2013; 20:329-35. [PMID: 23435031 DOI: 10.1097/gme.0b013e3182698f80] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We propose that the adrenal gland of an older higher primate female animal model will respond to human chorionic gonadotropin (hCG) hormone challenge by secreting additional dehydroepiandrosterone sulfate (DHEAS). Such a response in surgically and chemically castrated animals will provide proof of concept and a validated animal model for future studies to explore the rise in DHEAS during the menopausal transition of women. METHODS Twenty-four 18- to 26-year-old female cynomolgus monkeys were screened for ovarian function and then either ovariectomized (n = 4) or treated with a gonadotropin-releasing hormone agonist (GnRHa; n = 20) to block ovarian steroid production. After a recovery period from surgical procedure or down-regulation, a single-dose challenge (1,000 IU/animal, IM) of hCG was then administered to determine if luteinizing hormone (LH)/chorionic gonadotropin could accelerate circulating DHEAS production. Serum DHEAS, bioactive LH, and urinary metabolites of ovarian sex steroids were monitored before, during, and after these treatments. RESULTS Circulating LH bioactivity and immunoreactive DHEAS concentrations were suppressed in all animals 14 days postadministration of GnRHa. Urinary metabolites of estradiol and progesterone remained low after the surgical procedure or a flare reaction to GnRHa. Circulating DHEAS levels were increased after hCG administration, and the increase in individual animals was proportional to the pretreatment DHEAS at baseline. Circulating DHEAS concentrations were positively correlated to endogenous LH bioactive concentrations prior to hCG challenge and were subsequently further elevated by the hCG challenge while no concomitant change in ovarian steroid hormone excretion was observed. CONCLUSIONS These data demonstrate a positive adrenal androgen response to LH/chorionic gonadotropin in older female higher primates and suggest a mechanism for the rise in adrenal androgen production during the menopausal transition in women. These results also illustrate that the nonhuman primate animal model can be effectively used to investigate this phenomenon.
Collapse
|
21
|
Karman BN, Basavarajappa MS, Hannon P, Flaws JA. Dioxin exposure reduces the steroidogenic capacity of mouse antral follicles mainly at the level of HSD17B1 without altering atresia. Toxicol Appl Pharmacol 2012; 264:1-12. [PMID: 22889882 DOI: 10.1016/j.taap.2012.07.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 07/19/2012] [Accepted: 07/30/2012] [Indexed: 01/18/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent ovarian toxicant. Previously, we demonstrated that in vitro TCDD (1nM) exposure decreases production/secretion of the sex steroid hormones progesterone (P4), androstenedione (A4), testosterone (T), and 17β-estradiol (E2) in mouse antral follicles. The purpose of this study was to determine the mechanism by which TCDD inhibits steroidogenesis. Specifically, we examined the effects of TCDD on the steroidogenic enzymes, atresia, and the aryl hydrocarbon receptor (AHR) protein. TCDD exposure for 48h increased levels of A4, without changing HSD3B1 protein, HSD17B1 protein, estrone (E1), T or E2 levels. Further, TCDD did not alter atresia ratings compared to vehicle at 48h. TCDD, however, did down regulate the AHR protein at 48h. TCDD exposure for 96h decreased transcript levels for Cyp11a1, Cyp17a1, Hsd17b1, and Cyp19a1, but increased Hsd3b1 transcript. TCDD exposure particularly lowered both Hsd17b1 transcript and HSD17B1 protein. However, TCDD exposure did not affect levels of E1 in the media nor atresia ratings at 96h. TCDD, however, decreased levels of the proapoptotic factor Bax. Collectively, these data suggest that TCDD exposure causes a major block in the steroidogenic enzyme conversion of A4 to T and E1 to E2 and that it regulates apoptotic pathways, favoring survival over death in antral follicles. Finally, the down-regulation of the AHR protein in TCDD exposed follicles persisted at 96h, indicating that the activation and proteasomal degradation of this receptor likely plays a central role in the impaired steroidogenic capacity and altered apoptotic pathway of exposed antral follicles.
Collapse
Affiliation(s)
- Bethany N Karman
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA.
| | | | | | | |
Collapse
|
22
|
Ethun KF, Wood CE, Parker CR, Kaplan JR, Chen H, Appt SE. Effect of ovarian aging on androgen biosynthesis in a cynomolgus macaque model. Climacteric 2011; 15:82-92. [PMID: 21864136 DOI: 10.3109/13697137.2011.571321] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE The role of androgens in chronic disease pathogenesis, cognitive function and libido during menopause is of increasing interest. The aim of this study was to characterize the distribution and expression of androgenic proteins in the macaque ovary and to investigate the relationship between serum androgen concentrations, follicle number, and the persistence of androgenesis in the aging macaque ovary. METHODS The subjects were 26 adult female cynomolgus macaques. Ovaries were immunostained for cytochrome P450 17α-hydroxylase/17-20 lyase (P450c17), 3β-hydroxysteroid dehydrogenase (3βHSD), and cytochrome b5 (cytb5). Based on primordial follicle counts, animals were divided into tertiles (low (≤200), intermediate (226-1232), and high (2372-4356)) to evaluate differences in androgen staining and changes in serum androgen concentrations following ovariectomy. RESULTS Positive immunostaining for P450c17 and cytb5 within the theca interna layer of growing follicles persisted in advanced atretic follicles and secondary interstitial cells (residual stromal cells). Ovaries with low follicle numbers had less staining for all androgenic proteins compared to ovaries with higher numbers of growing follicles. Immunostaining for cytb5 was the most reliable marker for persistent androgenesis in ovaries with minimal primordial follicle numbers (<100) and residual stromal cells. Following ovariectomy, a significant decrease in testosterone (-27.7%, -30.8%, -27.5%; p < 0.01) and androstenedione (-33.4%, -35.7%, -46.0%; p < 0.01) was observed in monkeys with low, intermediate, and high primordial follicle counts, respectively. CONCLUSIONS Despite low follicle numbers, the aging macaque ovary retains the necessary proteins for androgenesis within residual stromal cells and contributes to peripheral androgen concentrations.
Collapse
Affiliation(s)
- K F Ethun
- Department of Pathology/Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA
| | | | | | | | | | | |
Collapse
|
23
|
Van Kempen TA, Milner TA, Waters EM. Accelerated ovarian failure: a novel, chemically induced animal model of menopause. Brain Res 2011; 1379:176-87. [PMID: 21211517 PMCID: PMC3078694 DOI: 10.1016/j.brainres.2010.12.064] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 12/17/2010] [Accepted: 12/20/2010] [Indexed: 10/18/2022]
Abstract
Current rodent models of menopause fail to adequately recapitulate the menopause transition. The intact aging model fails to achieve very low estrogen levels, and the ovariectomy model lacks a perimenopause phase. A new rodent model of accelerated ovarian failure (AOF) successfully replicates human perimenopause and postmenopause, including estrous acyclicity and fluctuating, followed by undetectable, estrogen levels, and allows for the dissociation of the effects of hormone levels from the effects of aging. In this model, an ovotoxic chemical, 4-vinylcyclohexene diepoxide (VCD), selective for primary and primordial follicles, is injected intraperitonelly in animals for 15 days. As the mature follicle population is depleted through natural cycling, ovarian failure follows increasing periods of acyclity. Administered at low doses, VCD specifically causes apoptotic cell death of primordial follicles but does not affect other peripheral tissues, including the liver and spleen, nor does it affect brain inflammation markers. In addition to reducing confounds associated with genetic and surgical manipulations, the AOF model maintains the presence of ovarian tissue which importantly parallels to the menopause transition in humans. The VCD injection procedure can be applied to studies using transgenic or knockout mice strains, or in other disease-state models (e.g., ischemia, atherosclerosis, or diabetes). This AOF model of menopause will generate new insights into women's health particularly in determining the critical periods (i.e., a window of opportunity) during perimenopause for restoring ovarian hormones for the most efficacious effect on memory and mood disorders as well as other menopausal symptoms.
Collapse
Affiliation(s)
- Tracey A Van Kempen
- Division of Neurobiology, Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA.
| | | | | |
Collapse
|
24
|
Noël JC, Anaf V, Borghese B, Vaiman D, Fayt I, Chapron C. The steroidogenic factor-1 protein is not expressed in various forms of endometriosis but is strongly present in ovarian cortical or medullary mesenchymatous cells adjacent to endometriotic foci. Fertil Steril 2011; 95:2655-7. [PMID: 21324456 DOI: 10.1016/j.fertnstert.2011.01.131] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Revised: 12/28/2010] [Accepted: 01/10/2011] [Indexed: 10/18/2022]
Abstract
Steroidogenic factor-1 (SF-1) protein expression was not observed in any form of endometriosis (peritoneal, ovarian, or deep infiltrating endometriosis), which suggests that SF-1 locally produced by endometrial or stromal cells may not play a major role in the development of endometriosis. However, the strong expression of SF-1 in cortical and medullary ovarian mesenchymatous cells may be capable of creating a favorable steroidogenic environment and the development of the disease.
Collapse
Affiliation(s)
- Jean-Christophe Noël
- Department of Gynecopathology, Erasme University Hospital, Free University of Brussels, Brussels, Belgium.
| | | | | | | | | | | |
Collapse
|
25
|
Wide distribution of the serum dehydroepiandrosterone and sex steroid levels in postmenopausal women. Menopause 2011; 18:30-43. [DOI: 10.1097/gme.0b013e3181e195a6] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
26
|
Toloubeydokhti T, Pan Q, Luo X, Bukulmez O, Chegini N. The expression and ovarian steroid regulation of endometrial micro-RNAs. Reprod Sci 2010; 15:993-1001. [PMID: 19088369 DOI: 10.1177/1933719108324132] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
MicroRNAs (miRNAs) which regulate gene expression stability displayed an aberrant expression profile in ectopic endometrium (ECE) as compared to eutopic (EUE) and normal endometrium (NE). We assessed the expression of miR-17-5p, miR-23a, miR-23b and miR-542-3p, their predicted target genes, steroidogenic acute regulatory protein, aromatase and cyclooxygenase-2, and influence of ovarian steroids on their expression in endometrial stromal (ESC) and glandular epithelial cells (GEC). The results indicated a lower expression of miR-23b and miR-542-3p and higher level of miR-17-5p in paired ECE and EUE as compared with NE. These levels were elevated and inversely correlated with the level of expression of their respective target genes in ECE. The expression of these miRNAs and genes was differentially regulated by 17beta- estradiol, medroxyprogesterone acetate, ICI-182780 and RU-486, or their respective combinations in ESC and GEC. We concluded that altered expression of specific miRNAs in ECE, affecting the stability of their target genes expression, has direct implications in pathogenesis of endometriosis.
Collapse
Affiliation(s)
- Tannaz Toloubeydokhti
- Division of Reproductive Endocrinology & Infertility, Department of Obstetrics and Gynecology, University of Florida College of Medicine, Gainesville, Florida 32610, USA
| | | | | | | | | |
Collapse
|
27
|
Craig ZR, Leslie TC, Hatfield KP, Gupta RK, Flaws JA. Mono-hydroxy methoxychlor alters levels of key sex steroids and steroidogenic enzymes in cultured mouse antral follicles. Toxicol Appl Pharmacol 2010; 249:107-13. [PMID: 20840852 DOI: 10.1016/j.taap.2010.09.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 08/31/2010] [Accepted: 09/02/2010] [Indexed: 01/24/2023]
Abstract
Methoxychlor (MXC) is an organochlorine pesticide that reduces fertility in female rodents by decreasing antral follicle numbers and increasing follicular death. MXC is metabolized in the body to mono-hydroxy MXC (mono-OH). Little is known about the effects of mono-OH on the ovary. Thus, this work tested the hypothesis that mono-OH exposure decreases production of 17β-estradiol (E₂) by cultured mouse antral follicles. Antral follicles were isolated from CD-1 mice (age 35-39 days) and exposed to dimethylsulfoxide (DMSO), or mono-OH (0.1-10 μg/mL) for 96 h. Media and follicles were collected for analysis of sex steroid levels and mRNA expression, respectively. Mono-OH treatment (10 μg/mL) decreased E(2) (DMSO: 3009.72±744.99 ng/mL; mono-OH 0.1 μg/mL: 1679.66±461.99 ng/mL; 1 μg/mL: 1752.72±532.41 ng/mL; 10 μg/mL: 45.89±33.83 ng/mL), testosterone (DMSO: 15.43±2.86 ng/mL; mono-OH 0.1μg/mL: 17.17±4.71 ng/mL; 1 μg/mL: 13.64±3.53 ng/mL; 10 μg/mL: 1.29±0.23 ng/mL), androstenedione (DMSO: 1.92±0.34 ng/mL; mono-OH 0.1 μg/mL: 1.49±0.43ng/mL; 1 μg/mL: 0.64±0.31 ng/mL; 10 μg/mL: 0.12±0.06 ng/mL) and progesterone (DMSO: 24.11±4.21 ng/mL; mono-OH 0.1μg/mL: 26.77±4.41 ng/mL; 1 μg/mL: 20.90±3.75 ng/mL; 10 μg/mL: 9.44±2.97 ng/mL) levels. Mono-OH did not alter expression of Star, Hsd3b1, Hsd17b1 and Cyp1b1, but it did reduce levels of Cyp11a1, Cyp17a1 and Cyp19a1 mRNA. Collectively, these data suggest that mono-OH significantly decreases levels of key sex steroid hormones and the expression of enzymes required for steroidogenesis.
Collapse
Affiliation(s)
- Zelieann R Craig
- Department of Comparative Biosciences, University of Illinois, Urbana, IL, USA.
| | | | | | | | | |
Collapse
|
28
|
Craig ZR, Marion SL, Funk JL, Bouxsein ML, Hoyer PB. Retaining Residual Ovarian Tissue following Ovarian Failure Has Limited Influence on Bone Loss in Aged Mice. J Osteoporos 2010; 2010:157323. [PMID: 20948577 PMCID: PMC2951122 DOI: 10.4061/2010/157323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 05/28/2010] [Indexed: 11/20/2022] Open
Abstract
Previous work showed that retaining residual ovarian tissue protects young mice from accelerated bone loss following ovarian failure. The present study was designed to determine whether this protection is also present in aged animals. Aged (9-12 months) C57BL/6Hsd female mice were divided into: CON (vehicle), VCD (160 mg/kg; 15d), or OVX (ovariectomized). Lumbar BMD was monitored by DXA and μCT used to assess vertebral microarchitecture. BMD was not different between VCD and CON at any time point but was lower (P < .05) than baseline, starting 1 month after ovarian failure in VCD and OVX mice. Following μCT analysis there were no differences between CON and VCD, but OVX mice had lower bone volume fraction, trabecular thickness, and a trend for decreased connectivity density. These findings provide evidence that retention of residual ovarian tissue may protect aged follicle-depleted mice from accelerated bone loss to a lesser extent than that observed in young mice.
Collapse
Affiliation(s)
- Zelieann R. Craig
- Department of Veterinary Biosciences, University of Illinois, 2001 S. Lincoln Avenue, Urbana, IL 61802, USA
| | - Samuel L. Marion
- Department of Physiology, University of Arizona, 1501 N Campbell Avenue, Tucson, AZ 85724, USA
| | - Janet L. Funk
- Department of Medicine, University of Arizona, P.O. Box 245218, Tucson, AZ 85724, USA
| | - Mary L. Bouxsein
- Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Patricia B. Hoyer
- Department of Physiology, University of Arizona, 1501 N Campbell Avenue, Tucson, AZ 85724, USA,*Patricia B. Hoyer:
| |
Collapse
|
29
|
Sutton-Tyrrell K, Zhao X, Santoro N, Lasley B, Sowers M, Johnston J, Mackey R, Matthews K. Reproductive hormones and obesity: 9 years of observation from the Study of Women's Health Across the Nation. Am J Epidemiol 2010; 171:1203-13. [PMID: 20427327 PMCID: PMC2915490 DOI: 10.1093/aje/kwq049] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 02/18/2010] [Indexed: 12/27/2022] Open
Abstract
The effect of change in reproductive hormones and menopause on incident obesity (body mass index > or =30 kg/m(2)) and severe obesity (body mass index > or =35 kg/m(2)) was evaluated over 9 years in 3,260 US women recruited in the multiethnic Study of Women's Health Across the Nation in 1996-1997. After 9 years, cumulative incidences of obesity and severe obesity reached 21.8% and 12.3%, respectively. In multivariate analysis, hormone changes, chronic health conditions, lower physical activity, race/ethnicity, and age were significantly associated with incident obesity and/or severe obesity. The odds of incident severe obesity increased with surgical menopause (odds ratio (OR) = 5.07, 95% confidence interval (CI): 2.29, 11.20; P < 0.001) and initiation of hormone therapy prior to 12 months of amenorrhea (OR = 2.94, 95% CI: 1.14, 7.58; P = 0.03). Predictors of obesity included an increase in free androgen index (OR = 1.37, 95% CI: 1.12, 1.68; P = 0.002) and a decrease in sex hormone-binding globulin (OR = 0.60, 95% CI: 0.45, 0.80; P = 0.0005). Similar results were found for severe obesity. Obesity rates varied by race, but no hormone-by-race interactions were observed. These longitudinal data demonstrate that higher androgens, lower sex hormone-binding globulin, surgical menopause, and early hormone therapy use predict incident obesity and/or severe obesity in a multiracial cohort of women transitioning into menopause.
Collapse
Affiliation(s)
- Kim Sutton-Tyrrell
- Department of Epidemiology, University of Pittsburgh, Pennsylvania 15261, USA.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
|
31
|
Labrie F. Intravaginal DHEA, by a strictly local action, exerts beneficial effects on both vaginal atrophy symptoms and sexual dysfunction. Horm Mol Biol Clin Investig 2010; 4:499-507. [DOI: 10.1515/hmbci.2010.064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 11/04/2010] [Indexed: 11/15/2022]
Abstract
Abstract: Following complete cessation of estrogen secretion by the ovaries at menopause, all estrogens and practically all androgens are made from dehydroepiandro-sterone (DHEA) of adrenal/ovarian origin. Although being an inactive molecule itself, DHEA is transformed at various levels and ratios into estrogens and/or androgens only in the tissues that possess the required cell-specific steroidogenic enzymes with minimal or no release of the active hormones in the blood according to the mechanisms of intracrinology. Vaginal atrophy affects 50% of postmenopausal women from 50 to 60 years of age and 72% of women 70 years and older.: At the standard 12-week time interval, 0.5% DHEA caused a 45.9±5.31 (p<0.0001 vs. placebo) decrease in the percentage of parabasal cells, a 6.8%±1.29% (p<0.0001 vs. placebo) increase in superficial cells, a 1.3±0.13 unit (p<0.0001 vs. placebo) decrease in vaginal pH and a 1.5±0.14 score unit (p<0.0001 vs. placebo) decrease in the severity of the most bothersome symptom. Similar changes were observed on vaginal secretions, color, epithelial surface thickness and epithelial integrity. In addition to the effects of intravaginal DHEA on the symptoms and signs of vaginal atrophy, a time- and dose-dependent improvement in the four domains of sexual function was observed, namely desire, arousal, orgasm and pain at sexual intercourse.: The present data indicate that combined androgenic/estrogenic stimulation in the three layers of the vagina exerts important beneficial effects on sexual function in women without systemic action on the brain and other extravaginal tissues.
Collapse
|
32
|
Thomas JL, Bucholtz KM, Sun J, Mack VL, Kacsoh B. Structural basis for the selective inhibition of human 3beta-hydroxysteroid dehydrogenase 1 in human breast tumor MCF-7 cells. Mol Cell Endocrinol 2009; 301:174-82. [PMID: 18955108 PMCID: PMC2667100 DOI: 10.1016/j.mce.2008.09.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2008] [Revised: 09/03/2008] [Accepted: 09/05/2008] [Indexed: 11/23/2022]
Abstract
Human 3beta-hydroxysteroid dehydrogenase/isomerase type 1 (3beta-HSD1) is a critical enzyme in the conversion of DHEA to estradiol in breast tumors and may be a target enzyme for inhibition in the treatment of breast cancer in postmenopausal women. Human 3beta-HSD2 participates in the production of cortisol and aldosterone in the human adrenal gland in this population. In our recombinant human breast tumor MCF-7 Tet-off cells that express either 3beta-HSD1 or 3beta-HSD2, trilostane and epostane inhibit the DHEA-induced proliferation of MCF-7 3beta-HSD1 cells with 12- to 16-fold lower IC(50) values compared to the MCF-7 3beta-HSD2 cells. The compounds also competitively inhibit purified human 3beta-HSD1 with 12- to 16-fold lower K(i) values compared to the noncompetitive K(i) values measured for human 3beta-HSD2. Using our structural model of 3beta-HSD1, trilostane or 17beta-acetoxy-trilostane was docked in the active site of 3beta-HSD1, and Arg195 in 3beta-HSD1 or Pro195 in 3beta-HSD2 was identified as a potentially critical residue (one of 23 non-identical residues in the two isoenzymes). The P195R mutant of 3beta-HSD2 were created, expressed and purified. Kinetic analyses of enzyme inhibition suggest that the high affinity, competitive inhibition of 3beta-HSD1 by trilostane and epostane may be related to the presence of Arg195 in 3beta-HSD1 vs. Pro195 in 3beta-HSD2.
Collapse
Affiliation(s)
- James L Thomas
- Division of Basic Medical Sciences, Mercer University School of Medicine, Mercer University, 1550 College Street, Macon, GA 31207, USA.
| | | | | | | | | |
Collapse
|
33
|
Rivera Z, Christian PJ, Marion SL, Brooks HL, Hoyer PB. Steroidogenic capacity of residual ovarian tissue in 4-vinylcyclohexene diepoxide-treated mice. Biol Reprod 2008; 80:328-36. [PMID: 18829706 DOI: 10.1095/biolreprod.108.070359] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Menopause is an important public health issue because of its association with a number of disorders. Androgens produced by residual ovarian tissue after menopause could impact the development of these disorders. It has been unclear, however, whether the postmenopausal ovary retains steroidogenic capacity. Thus, an ovary-intact mouse model for menopause that uses the occupational chemical 4-vinylcyclohexene diepoxide (VCD) was used to characterize the expression of steroidogenic genes in residual ovarian tissue of follicle-depleted mice. Female B6C3F1 mice (age, 28 days) were dosed daily for 20 days with either vehicle or VCD (160 mg kg(-1) day(-1)) to induce ovarian failure. Ovaries were collected on Day 181 and analyzed for mRNA and protein. Acyclic aged mice were used as controls for natural ovarian senescence. Relative to cycling controls, expression of mRNA encoding steroidogenic acute regulatory protein (Star); cholesterol side-chain cleavage (Cyp11a1); 3beta-hydroxysteroid dehydrogenase (Hsd3b); 17alpha-hydroxylase (Cyp17a1); scavenger receptor class B, type 1 (Scarb1); low-density lipoprotein receptor (Ldlr); and luteinizing hormone receptor (Lhcgr) was enriched in VCD-treated ovaries. In acyclic aged ovaries, mRNA expression for only Cyp17a1 and Lhcgr was greater than that in controls. Compared to cycling controls, ovaries from VCD-treated and aged mice had similar levels of HSD3B, CYP17A1, and LHCGR protein. The pattern of protein immunofluorescence staining for HSD3B in follicle-depleted (VCD-treated) ovaries was homogeneous, whereas that for CYP17A1 was only seen in residual interstitial cells. Circulating levels of FSH and LH were increased, and androstenedione levels were detectable following follicle depletion in VCD-treated mice. These findings support the idea that residual ovarian tissue in VCD-treated mice retains androgenic capacity.
Collapse
Affiliation(s)
- Zelieann Rivera
- Department of Physiology, University of Arizona, Tucson, Arizona 85724-5051, USA
| | | | | | | | | |
Collapse
|
34
|
Abstract
CONTEXT Several studies previously reported that the postmenopausal ovary produces androgens. However, these findings have recently been questioned in a group of women with adrenal insufficiency. OBJECTIVE We sought to use contemporary assay methodologies to investigate whether the postmenopausal ovary is hormonally active and contributes to the circulating pool of androgens. DESIGN AND PATIENTS Serum was collected from the ovarian veins of 13 postmenopausal women undergoing total abdominal hysterectomy and bilateral oophorectomy, with sufficient quantities obtained to allow for measurement of several hormones. Serum was also analyzed from peripheral blood collected preoperatively, intraoperatively, and postoperatively. SETTING The study took place at the Los Angeles County Women's and Children's Hospital, University of Southern California Keck School of Medicine. MAIN OUTCOME MEASURES Testosterone (T), androstenedione (A), dehydroepiandrosterone (DHEA), estrone (E1), and estradiol (E2) were measured by RIA with preceding organic solvent extraction and Celite column chromatography. RESULTS Statistically significant gradients were seen between the ovarian venous and peripheral samples for T, A, DHEA, E1, and E2. Postoperative levels of T and E1, but not A, DHEA, or E2, were statistically significantly lower than preoperative levels. A gradient for T between the ovarian venous and peripheral blood was present in four of five women who were menopausal for more than 10 yr. CONCLUSIONS The postmenopausal ovary is hormonally active, contributing significantly to the circulating pool of T. Furthermore, this contribution appears to persist in women as long as 10 yr beyond the menopause.
Collapse
Affiliation(s)
- Robin H Fogle
- Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA 90033, USA.
| | | | | | | |
Collapse
|
35
|
Kamenicky P, Houdoin L, Ferlicot S, Salenave S, Brailly S, Droupy S, Meduri G, Sasano H, Suzuki T, Young J, Chanson P. Benign cortisol-secreting adrenocortical adenomas produce small amounts of androgens. Clin Endocrinol (Oxf) 2007; 66:778-88. [PMID: 17408424 DOI: 10.1111/j.1365-2265.2007.02810.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Serum androgen levels are below normal in patients with benign cortisol-secreting adrenocortical adenomas, owing to ACTH suppression. Associated androgen secretion is usually considered as indicative of malignancy. The objective of the study was to analyse the androgen-producing ability of cortisol-secreting adrenocortical adenomas. DESIGN Retrospective data collection in a single referral hospital centre. METHODS Dehydroepiandrosterone sulfate (DHEAS), Delta4androstenedione and testosterone concentrations were measured before and after adrenalectomy and then at 6-month intervals in 20 women (eight cortisol-secreting adrenocortical adenomas, six subclinical cortisol-secreting adrenocortical adenomas, and six nonfunctional adenomas). RESULTS Before adrenalectomy, serum androgen concentrations were measurable in all women with clinically apparent and subclinical cortisol-secreting adrenocortical adenomas. DHEAS levels were either at the lower end of the normal range or below normal, but were always clearly detectable. Postoperatively, during adrenocortical insufficiency, DHEAS, Delta4androstenedione and testosterone concentrations fell to near the detection limit in all patients with cortisol-secreting adrenocortical adenomas (P = 0.008 for each marker) and showed a similar tendency to fall in all patients with subclinical cortisol-secreting adrenocortical adenomas. Pre- and post-treatment androgen concentrations did not differ in patients with nonfunctional adenomas. Immunohistochemical analysis confirmed CYP17, HSD3B2, SULT2A1 and CYB5 expression by all cortisol-producing tumours. The intensity of CYP17 and SULT2A1 expression was stronger in cortisol-secreting adenomas than in their adjacent normal adrenal tissue. CONCLUSION Both clinically apparent and subclinical cortisol-secreting adrenocortical adenomas appear to show moderate autonomous androgen production. Thus, weak androgen secretion in patients with adrenocortical tumours should not necessarily be considered as a sign of malignancy.
Collapse
Affiliation(s)
- Peter Kamenicky
- Department of Endocrinology and Reproductive Diseases, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, and Inserm, U693, Le Kremlin-Bicêtre, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Schumacher M, Guennoun R, Ghoumari A, Massaad C, Robert F, El-Etr M, Akwa Y, Rajkowski K, Baulieu EE. Novel perspectives for progesterone in hormone replacement therapy, with special reference to the nervous system. Endocr Rev 2007; 28:387-439. [PMID: 17431228 DOI: 10.1210/er.2006-0050] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The utility and safety of postmenopausal hormone replacement therapy has recently been put into question by large clinical trials. Their outcome has been extensively commented upon, but discussions have mainly been limited to the effects of estrogens. In fact, progestagens are generally only considered with respect to their usefulness in preventing estrogen stimulation of uterine hyperplasia and malignancy. In addition, various risks have been attributed to progestagens and their omission from hormone replacement therapy has been considered, but this may underestimate their potential benefits and therapeutic promises. A major reason for the controversial reputation of progestagens is that they are generally considered as a single class. Moreover, the term progesterone is often used as a generic one for the different types of both natural and synthetic progestagens. This is not appropriate because natural progesterone has properties very distinct from the synthetic progestins. Within the nervous system, the neuroprotective and promyelinating effects of progesterone are promising, not only for preventing but also for reversing age-dependent changes and dysfunctions. There is indeed strong evidence that the aging nervous system remains at least to some extent sensitive to these beneficial effects of progesterone. The actions of progesterone in peripheral target tissues including breast, blood vessels, and bones are less well understood, but there is evidence for the beneficial effects of progesterone. The variety of signaling mechanisms of progesterone offers exciting possibilities for the development of more selective, efficient, and safe progestagens. The recognition that progesterone is synthesized by neurons and glial cells requires a reevaluation of hormonal aging.
Collapse
Affiliation(s)
- Michael Schumacher
- INSERM UMR 788, 80, rue du Général Leclerc, 94276 Kremlin-Bicêtre, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Webber KM, Stocco DM, Casadesus G, Bowen RL, Atwood CS, Previll LA, Harris PLR, Zhu X, Perry G, Smith MA. Steroidogenic acute regulatory protein (StAR): evidence of gonadotropin-induced steroidogenesis in Alzheimer disease. Mol Neurodegener 2006; 1:14. [PMID: 17018137 PMCID: PMC1592538 DOI: 10.1186/1750-1326-1-14] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2006] [Accepted: 10/03/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer disease (AD) is clinically characterized by progressive memory loss, impairments in behavior, language and visual-spatial skills and ultimately, death. Epidemiological data reporting the predisposition of women to AD has led to a number of lines of evidence suggesting that age-related changes in hormones of the hypothalamic-pituitary-gonadal (HPG) axis following reproductive senescence, may contribute to the etiology of AD. Recent studies from our group and others have reported not only increases in circulating gonadotropins, namely luteinizing hormone (LH) in individuals with AD compared with control individuals, but also significant elevations of LH in vulnerable neuronal populations in individuals with AD compared to control cases as well as the highest density of gonadotropin receptors in the brain are found within the hippocampus, a region devastated in AD. However, while LH is higher in AD patients, the downstream consequences of this are incompletely understood. To begin to examine this issue, here, we examined the expression levels of steroidogenic acute regulatory (StAR) protein, which regulates the first key event in steroidogenesis, namely, the transport of cholesterol into the mitochondria, and is regulated by LH through the cyclic AMP second messenger pathway, in AD and control brain tissue. RESULTS Our data revealed that StAR protein was markedly increased in both the cytoplasm of hippocampal pyramidal neurons as well as in the cytoplasm of other non-neuronal cell types from AD brains when compared with age-matched controls. Importantly, and suggestive of a direct mechanistic link, StAR protein expression in AD brains colocalized with LH receptor expression. CONCLUSION Therefore, our findings suggest that LH is not only able to bind to its receptor and induce potentially pathogenic signaling in AD, but also that steroidogenic pathways regulated by LH may play a role in AD.
Collapse
Affiliation(s)
- Kate M Webber
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Douglas M Stocco
- Department of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Gemma Casadesus
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Richard L Bowen
- Voyager Pharmaceutical Corporation, Raleigh, North Carolina, USA
- Raleigh, North Carolina, USA
| | - Craig S Atwood
- School of Medicine, University of Wisconsin and William S. Middleton Memorial Veterans Administration, Madison, Wisconsin, USA
| | - Laura A Previll
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Peggy LR Harris
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - George Perry
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- College of Sciences, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Mark A Smith
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|