1
|
Crespi BJ, Bushell A, Dinsdale N. Testosterone mediates life-history trade-offs in female mammals. Biol Rev Camb Philos Soc 2024. [PMID: 39542451 DOI: 10.1111/brv.13166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
Hormones mediate life-history trade-offs. In female mammals, such trade-offs have been studied predominantly in the contexts of oestrogen, progesterone and prolactin. We evaluate the hypothesis that prenatal and postnatal testosterone levels structure and regulate trade-offs in females involving components of reproduction and survival. This hypothesis is predicated on the observation that testosterone confers competition-related and survival-related benefits, but also reproduction-related costs, to female mammals. The hypothesis is supported by field and laboratory data from diverse non-human animals, and data from healthy women. Most broadly, relatively low testosterone level in females has been associated with earlier, faster and higher offspring production, greater attractiveness to males, and reduced dominance or competitiveness, whereas higher testosterone level is associated with delayed and reduced reproduction but increased dominance, status, aggression, and resource accrual. The magnitude of testosterone-mediated trade-offs is expected to depend upon the strength of female-female competition, which represents some function of species-specific ecology, behaviour and mating system. Testosterone-associated trade-offs have, until now, been virtually ignored in studies of female life history, reproductive physiology, evolutionary endocrinology, and female-limited disease, probably due to researcher biases towards conceptualizing androgens as hormones with effects mainly restricted to males.
Collapse
Affiliation(s)
- Bernard J Crespi
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, V5A 1S6, Canada
| | - Aiden Bushell
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, V5A 1S6, Canada
| | - Natalie Dinsdale
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, V5A 1S6, Canada
| |
Collapse
|
2
|
Xu Y, Zhang Z, Wang R, Xue S, Ying Q, Jin L. Roles of estrogen and its receptors in polycystic ovary syndrome. Front Cell Dev Biol 2024; 12:1395331. [PMID: 38961865 PMCID: PMC11219844 DOI: 10.3389/fcell.2024.1395331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/21/2024] [Indexed: 07/05/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine disorder characterized by abnormal steroid hormone levels in peripheral blood and poor-quality oocytes. In the ovary, androgen is produced by theca cells, and estrogen is produced by granulosa cells. Androgen is converted to estrogen in granulosa cells, with cytochrome P450 aromatase as the limiting enzyme during this process. Estrogen receptors (ER) include ER alpha, ER beta, and membrane receptor GPR30. Studies have demonstrated that the abnormal functions of estrogen and its receptors and estradiol synthesis-related enzymes are closely related to PCOS. In recent years, some estrogen-related drugs have made significant progress in clinical application for subfertility with PCOS, such as letrozole and clomiphene. This article will elaborate on the recent advances in PCOS caused by abnormal expression of estrogen and its receptors and the application of related targeted small molecule drugs in clinical research and treatment.
Collapse
Affiliation(s)
- Yao Xu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ziyi Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Rongxiang Wang
- Reproductive Medicine Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Songguo Xue
- Reproductive Medicine Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qian Ying
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Liping Jin
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| |
Collapse
|
3
|
James DW, Quintela M, Lucini L, Al Kafri NAA, Healey GD, Jones N, Younas K, Bunkheila A, Margarit L, Francis LW, Gonzalez D, Conlan RS. Homeobox regulator Wilms Tumour 1 is displaced by androgen receptor at cis-regulatory elements in the endometrium of PCOS patients. Front Endocrinol (Lausanne) 2024; 15:1368494. [PMID: 38745948 PMCID: PMC11091321 DOI: 10.3389/fendo.2024.1368494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/08/2024] [Indexed: 05/16/2024] Open
Abstract
Decidualisation, the process whereby endometrial stromal cells undergo morphological and functional transformation in preparation for trophoblast invasion, is often disrupted in women with polycystic ovary syndrome (PCOS) resulting in complications with pregnancy and/or infertility. The transcription factor Wilms tumour suppressor 1 (WT1) is a key regulator of the decidualization process, which is reduced in patients with PCOS, a complex condition characterized by increased expression of androgen receptor in endometrial cells and high presence of circulating androgens. Using genome-wide chromatin immunoprecipitation approaches on primary human endometrial stromal cells, we identify key genes regulated by WT1 during decidualization, including homeobox transcription factors which are important for regulating cell differentiation. Furthermore, we found that AR in PCOS patients binds to the same DNA regions as WT1 in samples from healthy endometrium, suggesting dysregulation of genes important to decidualisation pathways in PCOS endometrium due to competitive binding between WT1 and AR. Integrating RNA-seq and H3K4me3 and H3K27ac ChIP-seq metadata with our WT1/AR data, we identified a number of key genes involved in immune response and angiogenesis pathways that are dysregulated in PCOS patients. This is likely due to epigenetic alterations at distal enhancer regions allowing AR to recruit cofactors such as MAGEA11, and demonstrates the consequences of AR disruption of WT1 in PCOS endometrium.
Collapse
Affiliation(s)
- David W. James
- Swansea University Medical School, Swansea, United Kingdom
| | | | - Lisa Lucini
- Swansea University Medical School, Swansea, United Kingdom
| | | | | | - Nicholas Jones
- Swansea University Medical School, Swansea, United Kingdom
| | - Kinza Younas
- Swansea University Medical School, Swansea, United Kingdom
- Swansea Bay University Health Board, Swansea, United Kingdom
| | - Adnan Bunkheila
- Swansea University Medical School, Swansea, United Kingdom
- Swansea Bay University Health Board, Swansea, United Kingdom
| | - Lavinia Margarit
- Swansea University Medical School, Swansea, United Kingdom
- Cwm Taf Morgannwg University Health Board, Bridgend, United Kingdom
| | | | | | | |
Collapse
|
4
|
Abbott DH, Hutcherson BA, Dumesic DA. Anti-Müllerian Hormone: A Molecular Key to Unlocking Polycystic Ovary Syndrome? Semin Reprod Med 2024; 42:41-48. [PMID: 38908381 DOI: 10.1055/s-0044-1787525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Anti-Müllerian hormone (AMH) is an important component within androgen receptor (AR)-regulated pathways governing the hyperandrogenic origin of polycystic ovary syndrome (PCOS). In women with PCOS, granulosa cell AMH overexpression in developing ovarian follicles contributes to elevated circulating AMH levels beginning at birth and continuing in adolescent daughters of PCOS women. A 6 to 7% incidence among PCOS women of gene variants coding for AMH or its receptor, AMHR2, suggests genetic contributions to AMH-related pathogenesis. Discrete gestational AMH administration to pregnant mice induces hypergonadotropic hyperandrogenic, PCOS-like female offspring with high circulating AMH levels that persist over three generations, suggesting epigenetic contributions to PCOS through developmental programming. Moreover, adult-onset, selective hyperactivation of hypothalamic neurons expressing gonadotropin-releasing hormone (GnRH) induces hypergonadotropic hyperandrogenism and PCOS-like traits in female mice. Both gestational and adult AMH inductions of PCOS-like traits are prevented by GnRH antagonist coadministration, implicating luteinizing hormone-dependent ovarian theca cell testosterone (T) action, mediated through the AR in AMH-induced pathogenesis. Interestingly, gestational or peripubertal exogenous T or dihydrotestosterone induction of PCOS-like traits in female mice, rats, sheep, and monkeys fails to elicit ovarian AMH hypersecretion; thus, AMH excess per se may lead to a distinct pathogenic contribution to hyperandrogenic PCOS origins.
Collapse
Affiliation(s)
- David H Abbott
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin
- Endocrinology and Reproductive Physiology Training Program, University of Wisconsin, Madison, Wisconsin
| | - Beverly A Hutcherson
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin
- Endocrinology and Reproductive Physiology Training Program, University of Wisconsin, Madison, Wisconsin
- Dean's Office, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, University of California, Los Angeles, California
| |
Collapse
|
5
|
Liu H, Tu M, Yin Z, Zhang D, Ma J, He F. Unraveling the complexity of polycystic ovary syndrome with animal models. J Genet Genomics 2024; 51:144-158. [PMID: 37777062 DOI: 10.1016/j.jgg.2023.09.012] [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] [Received: 07/03/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/02/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a highly familial and heritable endocrine disorder. Over half of the daughters born to women with PCOS may eventually develop their own PCOS-related symptoms. Progress in the treatment of PCOS is currently hindered by the complexity of its clinical manifestations and incomplete knowledge of its etiopathogenesis. Various animal models, including experimentally induced, naturally occurring, and spontaneously arising ones, have been established to emulate a wide range of phenotypical and pathological traits of human PCOS. These studies have led to a paradigm shift in understanding the genetic, developmental, and evolutionary origins of this disorder. Furthermore, emerging evidence suggests that animal models are useful in evaluating state-of-the-art drugs and treatments for PCOS. This review aims to provide a comprehensive summary of recent studies of PCOS in animal models, highlighting the power of these disease models in understanding the biology of PCOS and aiding high-throughput approaches.
Collapse
Affiliation(s)
- Huanju Liu
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Mixue Tu
- Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Zhiyong Yin
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Dan Zhang
- Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Zhejiang Provincial Clinical Research Center for Child Health, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Clinical Research Center on Birth Defect Prevention and Intervention of Zhejiang Province, Hangzhou, Zhejiang 310006, China.
| | - Jun Ma
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorder, Hangzhou, Zhejiang 310058, China.
| | - Feng He
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorder, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
6
|
Hu L, Liu Y, Dong P, Ye P. Protective effect of wuzibushen recipe on follicular development via regulating androgen receptor in polycystic ovary syndrome model rats. Gynecol Endocrinol 2023; 39:2190808. [PMID: 36963422 DOI: 10.1080/09513590.2023.2190808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/26/2023] Open
Abstract
OBJECTIVES The study aimed to explore the role and mechanism of WZBS recipe on PCOS. METHODS PCOS model was established. After modeling, PCOS rats were intragastrically administered with Diane-35 or WZBS recipe (6.93 g/kg/d). Then, the ovarian and uterine morphology were observed, the estrous cycle was assessed. HE and oil red O staining were conducted for ovarian morphological analysis and counting ovarian follicle and corpora lutea number. Furthermore, the serum content of testosterone (T) and sex-hormone-binding globulin (SHBG) were assessed by ELISA kits. The androgen receptor (AR), CX43 mRNA and protein expression were measured by q-PCR and Western blot. RESULTS WZBS recipe increased uterine implanted blastocysts, reduced cystic dilated follicles, and normalized estrous cycle in PCOS rats. Meanwhile, WZBS recipe alleviated ovarian injury, increased mature follicles and corpora lutea number in PCOS rats. Moreover, WZBS recipe decreased serum T content, AR expression and increased serum SHBG content, CX43 expression in PCOS rats. CONCLUSIONS This study reveals that WZBS recipe may attenuate PCOS by protecting follicular development via down-regulating AR.
Collapse
Affiliation(s)
- Lanyawen Hu
- Department of Reproductive Immunology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Yan Liu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Panpan Dong
- Department of Reproductive Immunology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Ping Ye
- Department of Reproductive Immunology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| |
Collapse
|
7
|
Dumesic DA, Abbott DH, Chazenbalk GD. An Evolutionary Model for the Ancient Origins of Polycystic Ovary Syndrome. J Clin Med 2023; 12:6120. [PMID: 37834765 PMCID: PMC10573644 DOI: 10.3390/jcm12196120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrinopathy of reproductive-aged women, characterized by hyperandrogenism, oligo-anovulation and insulin resistance and closely linked with preferential abdominal fat accumulation. As an ancestral primate trait, PCOS was likely further selected in humans when scarcity of food in hunter-gatherers of the late Pleistocene additionally programmed for enhanced fat storage to meet the metabolic demands of reproduction in later life. As an evolutionary model for PCOS, healthy normal-weight women with hyperandrogenic PCOS have subcutaneous (SC) abdominal adipose stem cells that favor fat storage through exaggerated lipid accumulation during development to adipocytes in vitro. In turn, fat storage is counterbalanced by reduced insulin sensitivity and preferential accumulation of highly lipolytic intra-abdominal fat in vivo. This metabolic adaptation in PCOS balances energy storage with glucose availability and fatty acid oxidation for optimal energy use during reproduction; its accompanying oligo-anovulation allowed PCOS women from antiquity sufficient time and strength for childrearing of fewer offspring with a greater likelihood of childhood survival. Heritable PCOS characteristics are affected by today's contemporary environment through epigenetic events that predispose women to lipotoxicity, with excess weight gain and pregnancy complications, calling for an emphasis on preventive healthcare to optimize the long-term, endocrine-metabolic health of PCOS women in today's obesogenic environment.
Collapse
Affiliation(s)
- Daniel A. Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA;
| | - David H. Abbott
- Department of Obstetrics and Gynecology, Wisconsin National Primate Research Center, University of Wisconsin, 1223 Capitol Court, Madison, WI 53715, USA;
| | - Gregorio D. Chazenbalk
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA;
| |
Collapse
|
8
|
Yan X, Zhu A, Li Y, Yang Z, Wang Y, Liu L, Liu W, Liu D, Li F, Du J, Cheng F, Gao X, Zhao J. Systematical assessment of digit ratio in a female masculinization disease: polycystic ovary syndrome. Front Endocrinol (Lausanne) 2023; 14:1146124. [PMID: 37223048 PMCID: PMC10202172 DOI: 10.3389/fendo.2023.1146124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/30/2023] [Indexed: 05/25/2023] Open
Abstract
Background In recent years, the right ratio of 2nd and 4th digit length (2D:4D) is regarded as an anatomical marker of prenatal testosterone exposure. Polycystic ovary syndrome (PCOS) is a female masculinized disease and is determined by prenatal testosterone exposure. Whether the ratio in the right hand of PCOS women is reduced or not compared with non-PCOS women is under debate. To further investigate the relationship between PCOS and digit ratio, we systematically measured all the digit ratios. Methods We recruited 34 non-PCOS women, 116 PCOS women, and 40 men and systematically measured all the ratios of digit length (2D:3D, 2D:4D, 2D:5D, 3D:4D, 3D:5D, and 4D:5D) of right hands and left hands. Results Left 2D:3D, 2D:4D, and 2D:5D in men were significantly lower than those in non-PCOS women. Significantly lower digit ratios of left 2D:3D and 2D:4D were observed in PCOS compared with non-PCOS women. In the subgroup analysis, the left ratio of digit length in 2D:3D and 2D:5D of the hyperandrogenism subgroup was lower than that of the non-hyperandrogenism subgroup without statistical significance. The logistic regression model of PCOS revealed that 2D:3D, 2D:4D, 2D:5D, and 3D:4D of left hands were statistically related to the diagnosis of PCOS among all the digit ratios. Conclusion Not only 2D:4D but also other digit ratios, such as 2D:3D and 2D:5D, are a marker of prenatal testosterone exposure and may be an anatomical marker of PCOS. The majority of these significant differences included left 2D, with the following order: non-PCOS women > PCOS women > men.
Collapse
Affiliation(s)
- Xueqi Yan
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Shandong University, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Aiqing Zhu
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Shandong University, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Yexing Li
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Shandong University, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Ziyi Yang
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Shandong University, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Yuteng Wang
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Shandong University, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Li Liu
- Department of Reproductive Medicine, Yinchuan Maternal and Child Health Hospital, Yinchuan, Ning xia, China
| | - Wei Liu
- Department of Reproductive Surgery, Northwest Women’s and Children’s Hospital, Xi’an, Shanxi, China
| | - Dan Liu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Tang Du Hospital, The Air Force Military Medical University, Xi’an, Shanxi, China
| | - Fenghua Li
- Department of Reproductive Medicine, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China
| | - Juan Du
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
| | - Fang Cheng
- Department of Reproductive Medicine, Yinchuan Maternal and Child Health Hospital, Yinchuan, Ning xia, China
| | - Xueying Gao
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Shandong University, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Junli Zhao
- Reproductive Medicine Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
9
|
Abedal-Majed MA, Abuajamieh M, Al-Qaisi M, Sargent KM, Titi HH, Alnimer MA, Abdelqader A, Shamoun AI, Cupp AS. Sheep with ovarian androgen excess have fibrosis and follicular arrest with increased mRNA abundance for steroidogenic enzymes and gonadotropin receptors. J Anim Sci 2023; 101:skad082. [PMID: 37061806 PMCID: PMC10184696 DOI: 10.1093/jas/skad082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/15/2023] [Indexed: 04/17/2023] Open
Abstract
An androgen excess ovarian micro-environment may limit follicle progression in sheep. Two populations of ewes with divergent follicular fluid androstenedione (A4) were identified in a flock in Jordan: High A4; (A4) ≥ 30 ng/mL, (N = 12) or Control A4 (Control); A4 ≤ 15 ng/mL; (N = 12). We hypothesized High A4 ewes would have increased steroidogenic enzyme mRNA abundance, inflammation, and follicular arrest. Messenger RNA abundance for steroidogenic enzymes StAR, CYP17A1, CYP11A1, and HSD3B1 were increased in theca cells while CYP17A1, CYP19A1, and HSD3B1 were increased in granulosa cells in High A4 ewes compared to Control. Gonadotropin receptor mRNA expression for LHCGR was increased in theca and FSHR in granulosa in High A4 ewes. Messenger RNA expression of FOS when reduced, increases expression of CYP17A1 which was observed in High A4 granulosa cells compared to Control. Furthermore, High A4 ewes had greater numbers of primordial follicles (P < 0.001) and fewer developing follicles compared to Control before, and after 7 d of culture, indicating follicular arrest was not alleviated by cortex culture. Increased fibrosis in the ovarian cortex was detected in High A4 ewes relative to Control (P < 0.001) suggesting increased inflammation and altered extracellular matrix deposition. Thus, this High A4 ewes population has similar characteristics to High A4 cows and women with polycystic ovary syndrome suggesting that naturally occurring androgen excess occurs in multiple species and may be a causative factor in follicular arrest and subsequent female sub- or infertility.
Collapse
Affiliation(s)
- Mohamed A Abedal-Majed
- Department of Animal Production, School of Agriculture, The University of Jordan, Amman 11942, Jordan
| | - Mohannad Abuajamieh
- Department of Animal Production, School of Agriculture, The University of Jordan, Amman 11942, Jordan
| | - Mohmmad Al-Qaisi
- Department of Animal Production, School of Agriculture, The University of Jordan, Amman 11942, Jordan
| | - Kevin M Sargent
- Department of Agriculture, Southeast Missouri State University, Cape Girardeau, MO 63701, USA
| | - Hosam H Titi
- Department of Animal Production, School of Agriculture, The University of Jordan, Amman 11942, Jordan
| | - Mufeed A Alnimer
- Department of Animal Production, School of Agriculture, The University of Jordan, Amman 11942, Jordan
| | - Anas Abdelqader
- Department of Animal Production, School of Agriculture, The University of Jordan, Amman 11942, Jordan
| | - Ahmad I Shamoun
- Department of Animal Production, School of Agriculture, The University of Jordan, Amman 11942, Jordan
| | - Andrea S Cupp
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln (UNL), Nebraska 68583, USA
| |
Collapse
|
10
|
Sudhakaran G, Rajesh R, Murugan R, Velayutham M, Guru A, Boopathi S, Muthupandian S, Gopinath P, Arockiaraj J. Nimbin analog
N2
alleviates high testosterone induced oxidative stress in
CHO
cells and alters the expression of
Tox3
and
Dennd1a
signal transduction pathway involved in the
PCOS
zebrafish. Phytother Res 2022; 37:1449-1461. [PMID: 36450691 DOI: 10.1002/ptr.7685] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/26/2022] [Accepted: 10/16/2022] [Indexed: 12/04/2022]
Abstract
Polycystic ovarian syndrome (PCOS) is a hormonal disorder that causes enlargement of ovaries and follicular maturation arrest, which lacks efficient treatment. N2, a semi-natural triterpenoid from the neem family, was already reported to have antioxidant and antiinflammatory properties in our previous report. This study investigated the anti-androgenic property of N2 on testosterone-induced oxidative stress in Chinese Hamster Ovarian cells (CHO) and PCOS zebrafish model. The testosterone exposure disrupted the antioxidant enzymes and ROS level and enhanced the apoptosis in both CHO cells and PCOS zebrafish. However, N2 significantly protected the CHO cells from ROS and apoptosis. N2 improved the Gonado somatic index (GSI) and upregulated the expression of the SOD enzyme in zebrafish ovaries. Moreover, the testosterone-induced follicular maturation arrest was normalized by N2 treatment in histopathology studies. In addition, the gene expression studies of Tox3 and Denndla in zebrafish demonstrated that N2 could impair PCOS condition. Furthermore, to confirm the N2 activity, the in-silico studies were performed against PCOS susceptible genes Tox3 and Dennd1a using molecular docking and molecular dynamic simulations. The results suggested that N2 alleviated the oxidative stress and apoptosis in-vitro and in-vivo and altered the expression of PCOS key genes.
Collapse
Affiliation(s)
- Gokul Sudhakaran
- Department of Biotechnology College of Science and Humanities, SRM Institute of Science and Technology Chennai India
| | - Ravi Rajesh
- Department of Chemistry College of Engineering and Technology, SRM Institute of Science and Technology Chennai India
| | - Raghul Murugan
- Department of Biotechnology College of Science and Humanities, SRM Institute of Science and Technology Chennai India
| | - Manikandan Velayutham
- Department of Biotechnology College of Science and Humanities, SRM Institute of Science and Technology Chennai India
| | - Ajay Guru
- Department of Conservative Dentistry and Endodontics Saveetha Dental College and Hospitals, SIMATS Chennai India
| | - Seenivasan Boopathi
- Department of Biotechnology College of Science and Humanities, SRM Institute of Science and Technology Chennai India
| | - Saravanan Muthupandian
- AMR and Nanomedicine Lab, Department of Pharmacology, Saveetha Dental College Saveetha Institute of Medical and Technical Sciences (SIMATS) Chennai India
| | - Pushparathinam Gopinath
- Department of Chemistry College of Engineering and Technology, SRM Institute of Science and Technology Chennai India
| | - Jesu Arockiaraj
- Department of Biotechnology College of Science and Humanities, SRM Institute of Science and Technology Chennai India
| |
Collapse
|
11
|
Stener-Victorin E. Update on Animal Models of Polycystic Ovary Syndrome. Endocrinology 2022; 163:bqac164. [PMID: 36201611 PMCID: PMC9631972 DOI: 10.1210/endocr/bqac164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Indexed: 11/19/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a complex disease affecting up to 15% of women of reproductive age. Women with PCOS suffer from reproductive dysfunctions with excessive androgen secretion and irregular ovulation, leading to reduced fertility and pregnancy complications. The syndrome is associated with a wide range of comorbidities including type 2 diabetes, obesity, and psychiatric disorders. Despite the high prevalence of PCOS, its etiology remains unclear. To understand the pathophysiology of PCOS, how it is inherited, and how to predict PCOS, and prevent and treat women with the syndrome, animal models provide an important approach to answering these fundamental questions. This minireview summarizes recent investigative efforts on PCOS-like rodent models aiming to define underlying mechanisms of the disease and provide guidance in model selection. The focus is on new genetic rodent models, on a naturally occurring rodent model, and provides an update on prenatal and peripubertal exposure models.
Collapse
|
12
|
Walters KA, Moreno-Asso A, Stepto NK, Pankhurst MW, Rodriguez Paris V, Rodgers RJ. Key signalling pathways underlying the aetiology of polycystic ovary syndrome. J Endocrinol 2022; 255:R1-R26. [PMID: 35980384 DOI: 10.1530/joe-22-0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/11/2022] [Indexed: 11/08/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine condition characterised by a range of reproductive, endocrine, metabolic and psychological abnormalities. Reports estimate that around 10% of women of reproductive age are affected by PCOS, representing a significant prevalence worldwide, which poses a high economic health burden. As the origin of PCOS remains largely unknown, there is neither a cure nor mechanism-based treatments leaving patient management suboptimal and focused solely on symptomatic treatment. However, if the underlying mechanisms underpinning the development of PCOS were uncovered then this would pave the way for the development of new interventions for PCOS. Recently, there have been significant advances in our understanding of the underlying pathways likely involved in PCOS pathogenesis. Key insights include the potential involvement of androgens, insulin, anti-Müllerian hormone and transforming growth factor beta in the development of PCOS. This review will summarise the significant scientific discoveries on these factors that have enhanced our knowledge of the mechanisms involved in the development of PCOS and discuss the impact these insights may have in shaping the future development of effective strategies for women with PCOS.
Collapse
Affiliation(s)
- Kirsty A Walters
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Alba Moreno-Asso
- Institute for Health and Sport, Victoria University, Footscray, Victoria, Australia
- Australian Institute of Musculoskeletal Science, Victoria University, St. Albans, Victoria, Australia
| | - Nigel K Stepto
- Institute for Health and Sport, Victoria University, Footscray, Victoria, Australia
- Australian Institute of Musculoskeletal Science, Victoria University, St. Albans, Victoria, Australia
- Monash Centre for Health Research and Implementation, Monash University and Monash Health, Clayton, Victoria, Australia
- Medicine at Western Health, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael W Pankhurst
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Valentina Rodriguez Paris
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Raymond J Rodgers
- The Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
13
|
McCartney CR, Campbell RE, Marshall JC, Moenter SM. The role of gonadotropin-releasing hormone neurons in polycystic ovary syndrome. J Neuroendocrinol 2022; 34:e13093. [PMID: 35083794 PMCID: PMC9232905 DOI: 10.1111/jne.13093] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/21/2021] [Accepted: 01/11/2022] [Indexed: 01/28/2023]
Abstract
Given the critical central role of gonadotropin-releasing hormone (GnRH) neurons in fertility, it is not surprising that the GnRH neural network is implicated in the pathology of polycystic ovary syndrome (PCOS), the most common cause of anovulatory infertility. Although many symptoms of PCOS relate most proximately to ovarian dysfunction, the central reproductive neuroendocrine system ultimately drives ovarian function through its regulation of anterior pituitary gonadotropin release. The typical cyclical changes in frequency of GnRH release are often absent in women with PCOS, resulting in a persistent high-frequency drive promoting gonadotropin changes (i.e., relatively high luteinizing hormone and relatively low follicle-stimulating hormone concentrations) that contribute to ovarian hyperandrogenemia and ovulatory dysfunction. However, the specific mechanisms underpinning GnRH neuron dysfunction in PCOS remain unclear. Here, we summarize several preclinical and clinical studies that explore the causes of aberrant GnRH secretion in PCOS and the role of disordered GnRH secretion in PCOS pathophysiology.
Collapse
Affiliation(s)
- Christopher R. McCartney
- Center for Research in Reproduction and Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVAUSA
| | - Rebecca E. Campbell
- Centre for Neuroendocrinology and Department of PhysiologySchool of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| | - John C. Marshall
- Center for Research in Reproduction and Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVAUSA
| | - Suzanne M. Moenter
- Departments of Molecular & Integrative PhysiologyInternal MedicineObstetrics and GynecologyUniversity of MichiganAnn ArborMIUSA
| |
Collapse
|
14
|
Silva MSB, Campbell RE. Polycystic Ovary Syndrome and the Neuroendocrine Consequences of Androgen Excess. Compr Physiol 2022; 12:3347-3369. [PMID: 35578968 DOI: 10.1002/cphy.c210025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a major endocrine disorder strongly associated with androgen excess and frequently leading to female infertility. Although classically considered an ovarian disease, altered neuroendocrine control of gonadotropin-releasing hormone (GnRH) neurons in the brain and abnormal gonadotropin secretion may underpin PCOS presentation. Defective regulation of GnRH pulse generation in PCOS promotes high luteinizing hormone (LH) pulsatile secretion, which in turn overstimulates ovarian androgen production. Early and emerging evidence from preclinical models suggests that maternal androgen excess programs abnormalities in developing neuroendocrine circuits that are associated with PCOS pathology, and that these abnormalities are sustained by postpubertal elevation of endogenous androgen levels. This article will discuss experimental evidence, from the clinic and in preclinical animal models, that has significantly contributed to our understanding of how androgen excess influences the assembly and maintenance of neuroendocrine impairments in the female brain. Abnormal central gamma-aminobutyric acid (GABA) signaling has been identified in both patients and preclinical models as a possible link between androgen excess and elevated GnRH/LH secretion. Enhanced GABAergic innervation and drive to GnRH neurons is suspected to contribute to the pathogenesis and early manifestation of neuroendocrine derangement in PCOS. Accordingly, this article also provides an overview of GABA regulation of GnRH neuron function from prenatal development to adulthood to discuss possible avenues for future discovery research and therapeutic interventions. © 2022 American Physiological Society. Compr Physiol 12:3347-3369, 2022.
Collapse
Affiliation(s)
- Mauro S B Silva
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
15
|
Zhou R, Bruns CM, Bird IM, Kemnitz JW, Dumesic DA, Abbott DH. Experimentally Induced Hyperinsulinemia Fails to Induce Polycystic Ovary Syndrome-like Traits in Female Rhesus Macaques. Int J Mol Sci 2022; 23:ijms23052635. [PMID: 35269778 PMCID: PMC8910161 DOI: 10.3390/ijms23052635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/12/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022] Open
Abstract
As in women with polycystic ovary syndrome (PCOS), hyperinsulinemia is associated with anovulation in PCOS-like female rhesus monkeys. Insulin sensitizers ameliorate hyperinsulinemia and stimulate ovulatory menstrual cycles in PCOS-like monkeys. To determine whether hyperinsulinemia (>694 pmol/L), alone, induces PCOS-like traits, five PCOS-like female rhesus monkeys with minimal PCOS-like traits, and four control females of similar mid-to-late reproductive years and body mass index, received daily subcutaneous injections of recombinant human insulin or diluent for 6−7 months. A cross-over experimental design enabled use of the same monkeys in each treatment phase. Insulin treatment unexpectedly normalized follicular phase duration in PCOS-like, but not control, females. In response to an intramuscular injection of 200 IU hCG, neither prenatally androgenized nor control females demonstrated ovarian hyperandrogenic responses while receiving insulin. An intravenous GnRH (100 ng/kg) injection also did not reveal evidence of hypergonadotropism. Taken together, these results suggest that experimentally induced adult hyperinsulinemia, alone, is insufficient to induce PCOS-like traits in female rhesus monkeys and to amplify intrinsic PCOS-like pathophysiology.
Collapse
Affiliation(s)
- Rao Zhou
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, USA; (R.Z.); (J.W.K.)
- Endocrinology Reproductive Physiology Training Program, University of Wisconsin, Madison, WI 53715, USA;
| | - Cristin M. Bruns
- Departments of Medicine, University of Wisconsin, Madison, WI 53715, USA;
| | - Ian M. Bird
- Endocrinology Reproductive Physiology Training Program, University of Wisconsin, Madison, WI 53715, USA;
- Departments of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53715, USA
| | - Joseph W. Kemnitz
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, USA; (R.Z.); (J.W.K.)
- Departments of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53715, USA
| | - Daniel A. Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
| | - David H. Abbott
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, USA; (R.Z.); (J.W.K.)
- Endocrinology Reproductive Physiology Training Program, University of Wisconsin, Madison, WI 53715, USA;
- Departments of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53715, USA
- Correspondence:
| |
Collapse
|
16
|
Parker J, O’Brien C, Hawrelak J, Gersh FL. Polycystic Ovary Syndrome: An Evolutionary Adaptation to Lifestyle and the Environment. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19031336. [PMID: 35162359 PMCID: PMC8835454 DOI: 10.3390/ijerph19031336] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023]
Abstract
Polycystic ovary syndrome (PCOS) is increasingly recognized as a complex metabolic disorder that manifests in genetically susceptible women following a range of negative exposures to nutritional and environmental factors related to contemporary lifestyle. The hypothesis that PCOS phenotypes are derived from a mismatch between ancient genetic survival mechanisms and modern lifestyle practices is supported by a diversity of research findings. The proposed evolutionary model of the pathogenesis of PCOS incorporates evidence related to evolutionary theory, genetic studies, in utero developmental epigenetic programming, transgenerational inheritance, metabolic features including insulin resistance, obesity and the apparent paradox of lean phenotypes, reproductive effects and subfertility, the impact of the microbiome and dysbiosis, endocrine-disrupting chemical exposure, and the influence of lifestyle factors such as poor-quality diet and physical inactivity. Based on these premises, the diverse lines of research are synthesized into a composite evolutionary model of the pathogenesis of PCOS. It is hoped that this model will assist clinicians and patients to understand the importance of lifestyle interventions in the prevention and management of PCOS and provide a conceptual framework for future research. It is appreciated that this theory represents a synthesis of the current evidence and that it is expected to evolve and change over time.
Collapse
Affiliation(s)
- Jim Parker
- School of Medicine, University of Wollongong, Wollongong 2500, Australia
- Correspondence:
| | - Claire O’Brien
- Faculty of Science and Technology, University of Canberra, Bruce 2617, Australia;
| | - Jason Hawrelak
- College of Health and Medicine, University of Tasmania, Hobart 7005, Australia;
| | - Felice L. Gersh
- College of Medicine, University of Arizona, Tucson, AZ 85004, USA;
| |
Collapse
|
17
|
Dumesic DA, Padmanabhan V, Chazenbalk GD, Abbott DH. Polycystic ovary syndrome as a plausible evolutionary outcome of metabolic adaptation. Reprod Biol Endocrinol 2022; 20:12. [PMID: 35012577 PMCID: PMC8744313 DOI: 10.1186/s12958-021-00878-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/10/2021] [Indexed: 12/22/2022] Open
Abstract
As a common endocrinopathy of reproductive-aged women, polycystic ovary syndrome (PCOS) is characterized by hyperandrogenism, oligo-anovulation and polycystic ovarian morphology. It is linked with insulin resistance through preferential abdominal fat accumulation that is worsened by obesity. Over the past two millennia, menstrual irregularity, male-type habitus and sub-infertility have been described in women and confirm that these clinical features of PCOS were common in antiquity. Recent findings in normal-weight hyperandrogenic PCOS women show that exaggerated lipid accumulation by subcutaneous (SC) abdominal stem cells during development to adipocytes in vitro occurs in combination with reduced insulin sensitivity and preferential accumulation of highly-lipolytic intra-abdominal fat in vivo. This PCOS phenotype may be an evolutionary metabolic adaptation to balance energy storage with glucose availability and fatty acid oxidation for optimal energy use during reproduction. This review integrates fundamental endocrine-metabolic changes in healthy, normal-weight PCOS women with similar PCOS-like traits present in animal models in which tissue differentiation is completed during fetal life as in humans to support the evolutionary concept that PCOS has common ancestral and developmental origins.
Collapse
Affiliation(s)
- Daniel A. Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Room 22-178 CHS, Los Angeles, CA 90095 USA
| | | | - Gregorio D. Chazenbalk
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Room 22-178 CHS, Los Angeles, CA 90095 USA
| | - David H. Abbott
- Department of Obstetrics and Gynecology, University of Wisconsin and Wisconsin National Primate Research Center, 1223 Capitol Court, Madison, WI 53715 USA
| |
Collapse
|
18
|
Evidence-based hormonal, mutational, and endocrine-disrupting chemical-induced zebrafish as an alternative model to study PCOS condition similar to mammalian PCOS model. Life Sci 2022; 291:120276. [PMID: 34990650 DOI: 10.1016/j.lfs.2021.120276] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/15/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022]
Abstract
Polycystic ovarian syndrome (PCOS) causes swollen ovaries in women at reproductive age due to hormonal disorder with small cysts on the outer edges. The cause of the disorder is still yet to be found. Multiple factors have increased PCOS prevalence, hyperandrogenism, oxidative stress, inflammation, and insulin resistance. Various animal PCOS models have been developed to imitate the pathophysiology of PCOS in humans. Zebrafish is one of the most versatile animal experimental models because of the transparency of the embryos, small size, and rapid growth. The zebrafish similarity to higher vertebrates made it a useful non-mammalian model for PCOS drug testing and screening. This review provides an insight into the usage of zebrafish, a non-mammalian model for PCOS, as an opportunity for evaluating future initiatives in such a research domain.
Collapse
|
19
|
Prenatal Androgen Treatment Does Not Alter the Firing Activity of Hypothalamic Arcuate Kisspeptin Neurons in Female Mice. eNeuro 2021; 8:ENEURO.0306-21.2021. [PMID: 34503965 PMCID: PMC8482853 DOI: 10.1523/eneuro.0306-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 11/21/2022] Open
Abstract
Neuroendocrine control of reproduction is disrupted in many individuals with polycystic ovary syndrome (PCOS), who present with increased luteinizing hormone (LH), and presumably gonadotropin-releasing hormone (GnRH), release frequency, and high androgen levels. Prenatal androgenization (PNA) recapitulates these phenotypes in primates and rodents. Female offspring of mice injected with dihydrotestosterone (DHT) on gestational days 16-18 exhibit disrupted estrous cyclicity, increased LH and testosterone, and increased GnRH neuron firing rate as adults. PNA also alters the developmental trajectory of GnRH neuron firing rates, markedly blunting the prepubertal peak in firing that occurs in three-week (3wk)-old controls. GnRH neurons do not express detectable androgen receptors and are thus probably not the direct target of DHT. Rather, PNA likely alters GnRH neuronal activity by modulating upstream neurons, such as hypothalamic arcuate neurons co-expressing kisspeptin, neurokinin B (gene Tac2), and dynorphin, also known as KNDy neurons. We hypothesized PNA treatment changes firing rates of KNDy neurons in a similar age-dependent manner as GnRH neurons. We conducted targeted extracellular recordings (0.5-2 h) of Tac2-identified KNDy neurons from control and PNA mice at 3wks of age and in adulthood. About half of neurons were quiescent (<0.005 Hz). Long-term firing rates of active cells varied, suggestive of episodic activity, but were not different among groups. Short-term burst firing was also similar. We thus reject the hypothesis that PNA alters the firing rate of KNDy neurons. This does not preclude altered neurosecretory output of KNDy neurons, involvement of other neuronal populations, or in vivo networks as critical drivers of altered GnRH firing rates in PNA mice.
Collapse
|
20
|
Phillips S, Timms P, Jelocnik M. Is Chlamydia to Blame for Koala Reproductive Cysts? Pathogens 2021; 10:pathogens10091140. [PMID: 34578173 PMCID: PMC8467779 DOI: 10.3390/pathogens10091140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022] Open
Abstract
A significant threat to koala populations is infection from Chlamydia, which results in disease and death. Wild koalas with Chlamydia infections are admitted to wildlife hospitals and treated with antibiotics; however, up to 50% of koalas that present to wildlife hospitals do not survive. A major contributor to high mortality is the development of reproductive cysts, resulting in female infertility and euthanasia. However, the diagnosis of reproductive disease is limited to ultrasound with no further investigations. This communication highlights reports of histological and microbiological findings, the accuracy of ultrasound to necropsy reports and other possible causes for reproductive cyst development previously reported in other hosts. Our conclusions identify a significant knowledge gap in the aetiology of koala reproductive cysts and highlight the urgent need for future investigations.
Collapse
|
21
|
Parker J, O'Brien C, Gersh FL. Developmental origins and transgenerational inheritance of polycystic ovary syndrome. Aust N Z J Obstet Gynaecol 2021; 61:922-926. [PMID: 34403138 DOI: 10.1111/ajo.13420] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 08/01/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND There has been increasing awareness that polycystic ovary syndrome (PCOS) phenotypes may represent a mismatch between ancient genetically programmed metabolic and reproductive survival mechanisms and modern lifestyle practices. In-utero developmental programming of metabolic and endocrine pathways may play an important role in activating gene variants that predispose the offspring to develop PCOS when exposed to specific postnatal conditions. Postnatal exposure to lifestyle factors such as poor-quality diet and endocrine disrupting chemicals may modulate epigenetically programmed pathways that result in the observed pathophysiological changes and clinical features seen in women with PCOS. AIM To review the developmental origins and transgenerational transmission of PCOS and the impact of lifestyle, androgens and endocrine disrupting chemicals on fetal epigenetic programming. MATERIALS AND METHODS The literature was reviewed using Google, Google Scholar, Medline and PubMed databases. The results are presented as a narrative review. RESULTS Human observational and animal experimental data support the hypothesis that PCOS is an inherited condition that arises as a result of developmental programming of normal gene variants. It is likely that these genes can be amplified by in-utero androgen exposure and activated by a range of postnatal lifestyle and environmental factors. Endocrine disrupting chemicals have the potential to influence developmental programming of PCOS susceptibility genes. CONCLUSIONS The current evidence suggests that developmental epigenetic programming following exposure to an adverse maternal metabolic and endocrine environment contributes to the pathogenesis of PCOS. Lifestyle interventions, as recommended by the International Guidelines, have the potential to reduce both symptoms and transgenerational transmission of PCOS.
Collapse
Affiliation(s)
- Jim Parker
- School of Medicine, University of Wollongong, Wollongong, New South Wales, Australia
| | - Claire O'Brien
- Faculty of Science and Technology, University of Canberra, Canberra, Australian Capital Territory, Australia
| | - Felice L Gersh
- Internal Medicine, University of Arizona College of Medicine, Irvine, CA, USA
| |
Collapse
|
22
|
Lesseur C, Pirrotte P, Pathak KV, Manservisi F, Mandrioli D, Belpoggi F, Panzacchi S, Li Q, Barrett ES, Nguyen RHN, Sathyanarayana S, Swan SH, Chen J. Maternal urinary levels of glyphosate during pregnancy and anogenital distance in newborns in a US multicenter pregnancy cohort. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 280:117002. [PMID: 33812205 PMCID: PMC8165010 DOI: 10.1016/j.envpol.2021.117002] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 05/05/2023]
Abstract
Human exposure to glyphosate has become ubiquitous because of its increasing agricultural use. Recent studies suggest endocrine disrupting effects of glyphosate. Specifically, in our work in rodents, low-dose early-life exposure to Roundup® (glyphosate-based herbicide) lengthened anogenital distance (AGD) in male and female offspring. AGD is a marker of the prenatal hormone milieu in rodents and humans. The relationship between glyphosate exposure and AGD has not been studied in humans. We conducted a pilot study in 94 mother-infant pairs (45 female and 49 male) from The Infant Development and the Environment Study (TIDES). For each infant, two AGD measurements were collected after birth; the anopenile (AGD-AP) and anoscrotal (AGD-AS) distances for males, and anoclitoral (AGD-AC) and anofourchette distances (AGD-AF) for females. We measured levels of glyphosate and its degradation product aminomethylphosphonic acid (AMPA) in 2nd trimester maternal urine samples using ultra-high-performance liquid chromatography-tandem mass spectrometry. We assessed the relationship between exposure and AGD using sex-stratified multivariable linear regression models. Glyphosate and AMPA were detected in 95% and 93% of the samples (median 0.22 ng/mL and 0.14 ng/mL, respectively). Their concentrations were moderately correlated (r = 0.55, p = 5.7 × 10-9). In female infants, high maternal urinary glyphosate (above the median) was associated with longer AGD-AC (β = 1.48, 95%CI (-0.01, 3.0), p = 0.05), but this was not significant after covariate adjustment. Increased AMPA was associated with longer AGD-AF (β = 1.96, 95%CI (0.44, 3.5), p = 0.01) after adjusting for infant size and age at AGD exam. No associations were detected in male offspring. These preliminary findings partially reproduce our previous results in rodents and suggest that glyphosate is a sex-specific endocrine disruptor with androgenic effects in humans. Given the increasing glyphosate exposures in the US population, larger studies should evaluate potential developmental effects on endocrine and reproductive systems.
Collapse
Affiliation(s)
- Corina Lesseur
- Department of Environmental Medicine and Public Heath, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Patrick Pirrotte
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Khyatiben V Pathak
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Fabiana Manservisi
- Cesare Maltoni Cancer Research Center (CMCRC), Ramazzini Institute (RI), Via Saliceto, 3, 40010, Bentivoglio, Bologna, Italy; Department of Veterinary Medical Sciences, University of Bologna, Italy
| | - Daniele Mandrioli
- Cesare Maltoni Cancer Research Center (CMCRC), Ramazzini Institute (RI), Via Saliceto, 3, 40010, Bentivoglio, Bologna, Italy
| | - Fiorella Belpoggi
- Cesare Maltoni Cancer Research Center (CMCRC), Ramazzini Institute (RI), Via Saliceto, 3, 40010, Bentivoglio, Bologna, Italy
| | - Simona Panzacchi
- Cesare Maltoni Cancer Research Center (CMCRC), Ramazzini Institute (RI), Via Saliceto, 3, 40010, Bentivoglio, Bologna, Italy
| | - Qian Li
- Department of Environmental Medicine and Public Heath, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily S Barrett
- Department of Biostatistics & Epidemiology, Rutgers School of Public Health, Piscataway, NJ, USA
| | - Ruby H N Nguyen
- Department of Epidemiology & Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Sheela Sathyanarayana
- Department of Pediatrics, University of Washington and Seattle Children's Research Institute, Seattle, WA, USA
| | - Shanna H Swan
- Department of Environmental Medicine and Public Heath, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Chen
- Department of Environmental Medicine and Public Heath, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
23
|
Corrie L, Gulati M, Singh SK, Kapoor B, Khursheed R, Awasthi A, Vishwas S, Chellappan DK, Gupta G, Jha NK, Anand K, Dua K. Recent updates on animal models for understanding the etiopathogenesis of polycystic ovarian syndrome. Life Sci 2021; 280:119753. [PMID: 34171379 DOI: 10.1016/j.lfs.2021.119753] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 12/17/2022]
Abstract
Polycystic ovarian syndrome (PCOS) is the primary cause of female infertility affecting several women worldwide. Changes in hormonal functions such as hyperandrogenism are considered a significant factor in developing PCOS in women. In addition, many molecular pathways are involved in the pathogenesis of PCOS in women. To have better insights about PCOS, it is data from clinical studies carried on women suffering from PCOS should be collected. However, this approach has several implications, including ethical considerations, cost involved and availability of subject. Moreover, during the early drug development process, it is always advisable to use non-human models mimicking human physiology as they are less expensive, readily available, have a shorter gestation period and less risk involved. Many animal models have been reported that resemble the PCOS pathways in human subjects. However, the models developed on rats and mice are more preferred over other rodent/non-rodent models due to their closer resemblance with human PCOS development mechanism. The most extensively reported PCOS models for rats and mice include those induced by using testosterone, letrozole and estradiol valerate. As the pathophysiology of PCOS is complex, none of the explored models completely surrogates the PCOS related conditions occurring in women. Hence, there is a need to develop an animal model that can resemble the pathophysiology of PCOS in women. The review focuses on various animal models explored to understand the pathophysiology of PCOS. The article also highlights some environmental and food-related models that have been used to induce PCOS.
Collapse
Affiliation(s)
- Leander Corrie
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India.
| | - Bhupinder Kapoor
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Rubiya Khursheed
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Ankit Awasthi
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310, India
| | - Krishnan Anand
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences, National Health Laboratory Service, University of the Free State, Bloemfontein, South Africa
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Australia
| |
Collapse
|
24
|
Bourgneuf C, Bailbé D, Lamazière A, Dupont C, Moldes M, Farabos D, Roblot N, Gauthier C, Mathieu d'Argent E, Cohen-Tannoudji J, Monniaux D, Fève B, Movassat J, di Clemente N, Racine C. The Goto-Kakizaki rat is a spontaneous prototypical rodent model of polycystic ovary syndrome. Nat Commun 2021; 12:1064. [PMID: 33594056 PMCID: PMC7886868 DOI: 10.1038/s41467-021-21308-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 01/18/2021] [Indexed: 12/21/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is characterized by an oligo-anovulation, hyperandrogenism and polycystic ovarian morphology combined with major metabolic disturbances. However, despite the high prevalence and the human and economic consequences of this syndrome, its etiology remains unknown. In this study, we show that female Goto-Kakizaki (GK) rats, a type 2 diabetes mellitus model, encapsulate naturally all the reproductive and metabolic hallmarks of lean women with PCOS at puberty and in adulthood. The analysis of their gestation and of their fetuses demonstrates that this PCOS-like phenotype is developmentally programmed. GK rats also develop features of ovarian hyperstimulation syndrome. Lastly, a comparison between GK rats and a cohort of women with PCOS reveals a similar reproductive signature. Thus, this spontaneous rodent model of PCOS represents an original tool for the identification of the mechanisms involved in its pathogenesis and for the development of novel strategies for its treatment.
Collapse
Affiliation(s)
- Camille Bourgneuf
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Danielle Bailbé
- Université de Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Antonin Lamazière
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Saint-Antoine, Département PM2, Paris, France
| | - Charlotte Dupont
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Service de biologie de la reproduction-CECOS, Paris, France
| | - Marthe Moldes
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Dominique Farabos
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Saint-Antoine, Département PM2, Paris, France
| | - Natacha Roblot
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Camille Gauthier
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Emmanuelle Mathieu d'Argent
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Service de biologie de la reproduction-CECOS, Paris, France
| | | | | | - Bruno Fève
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Saint-Antoine, Service Endocrinologie, CRMR PRISIS, Paris, France
| | - Jamileh Movassat
- Université de Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Nathalie di Clemente
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Chrystèle Racine
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.
- Institut Hospitalo-Universitaire ICAN, Paris, France.
- Université de Paris, Paris, France.
| |
Collapse
|
25
|
Witchel SF, Plant TM. Intertwined reproductive endocrinology: Puberty and polycystic ovary syndrome. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2020; 14:127-136. [PMID: 33102929 PMCID: PMC7583558 DOI: 10.1016/j.coemr.2020.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous familial disorder often emerging during the peri-pubertal years concomitantly with the onset of gonadarche and adrenarche. Both gonadarche and PCOS reflect functional changes in the hypothalamic-pituitary-ovarian axis. During this transition, normal girls manifest features consistent with PCOS such as irregular menses, mild hyperandrogenism, and multi-follicular ovary morphology. Themes common to puberty and PCOS, neuroendocrine features, androgen exposure, and insulin sensitivity, will be considered to address the possibility that PCOS interferes with the normal pubertal transition.
Collapse
Affiliation(s)
- Selma Feldman Witchel
- Division of Pediatric Endocrinology, UPMC Children's Hospital of Pittsburgh/University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224 USA
| | - Tony M Plant
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, PA 15213, USA
| |
Collapse
|
26
|
Santos LCDS, Lapa Neto CJC, Santos AMGD, Marinho KSDN, Nascimento BJD, Alves ER, Teixeir ÁAC, Wanderley-Teixeira V. Immunohistochemical and histophysiological study of prolonged use of nandrolone on reproductive organs and fertility. Biotech Histochem 2020; 96:468-486. [PMID: 32981356 DOI: 10.1080/10520295.2020.1822545] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
We investigated possible changes in morphology and immunohistochemistry of the uterus and ovaries of rats caused by nandrolone (ND); we also investigated effects on fertility. We used 30 rats divided into three experimental groups: control (C), control vehicle (CV) and 5 mg/kg ND. Rats treated with ND exhibited loss of estrous cyclicity with predominance of the estrus phase, increased body weight and an organosomatic index that was decreased for the ovaries, but increased for the uterus. In the ovary, we observed a reduction in primary and secondary follicles and an increase in tertiary follicles; no corpora lutea were observed. Estrogen and progesterone levels were reduced. In the uterus, the endometrium was edematous with hyperplasic glands. The cytokines, TNFα and IL6, and the apoptotic index were increased in rats treated with ND. VEGF-A was increased in the ovaries and decreased in the uterus. We conclude that ND disrupts ovarian and uterine histophysiology by establishing an anovulatory and inflammatory condition, which directly affects reproduction.
Collapse
Affiliation(s)
| | | | | | | | - Bruno José do Nascimento
- Department of Animal Morphology and Physiology, Rural Federal University of Pernambuco, Recife, Brazil
| | - Erique Ricardo Alves
- Department of Animal Morphology and Physiology, Rural Federal University of Pernambuco, Recife, Brazil
| | | | | |
Collapse
|
27
|
Stener-Victorin E, Padmanabhan V, Walters KA, Campbell RE, Benrick A, Giacobini P, Dumesic DA, Abbott DH. Animal Models to Understand the Etiology and Pathophysiology of Polycystic Ovary Syndrome. Endocr Rev 2020; 41:bnaa010. [PMID: 32310267 PMCID: PMC7279705 DOI: 10.1210/endrev/bnaa010] [Citation(s) in RCA: 169] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/14/2020] [Indexed: 12/14/2022]
Abstract
More than 1 out of 10 women worldwide are diagnosed with polycystic ovary syndrome (PCOS), the leading cause of female reproductive and metabolic dysfunction. Despite its high prevalence, PCOS and its accompanying morbidities are likely underdiagnosed, averaging > 2 years and 3 physicians before women are diagnosed. Although it has been intensively researched, the underlying cause(s) of PCOS have yet to be defined. In order to understand PCOS pathophysiology, its developmental origins, and how to predict and prevent PCOS onset, there is an urgent need for safe and effective markers and treatments. In this review, we detail which animal models are more suitable for contributing to our understanding of the etiology and pathophysiology of PCOS. We summarize and highlight advantages and limitations of hormonal or genetic manipulation of animal models, as well as of naturally occurring PCOS-like females.
Collapse
Affiliation(s)
| | - Vasantha Padmanabhan
- Departments of Pediatrics, Obstetrics and Gynecology, and Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan
| | - Kirsty A Walters
- Fertility & Research Centre, School of Women’s and Children’s Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Anna Benrick
- Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- School of Health Sciences and Education, University of Skövde, Skövde, Sweden
| | - Paolo Giacobini
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, California
| | - David H Abbott
- Department of Obstetrics and Gynecology, Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
28
|
New insights into anti-Müllerian hormone role in the hypothalamic-pituitary-gonadal axis and neuroendocrine development. Cell Mol Life Sci 2020; 78:1-16. [PMID: 32564094 PMCID: PMC7867527 DOI: 10.1007/s00018-020-03576-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/08/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022]
Abstract
Research into the physiological actions of anti-Müllerian hormone (AMH) has rapidly expanded from its classical role in male sexual differentiation to the regulation of ovarian function, routine clinical use in reproductive health and potential use as a biomarker in the diagnosis of polycystic ovary syndrome (PCOS). During the past 10 years, the notion that AMH could act exclusively at gonadal levels has undergone another paradigm shift as several exciting studies reported unforeseen AMH actions throughout the Hypothalamic–Pituitary–Gonadal (HPG) axis. In this review, we will focus on these findings reporting novel AMH actions across the HPG axis and we will discuss their potential impact and significance to better understand human reproductive disorders characterized by either developmental alterations of neuroendocrine circuits regulating fertility and/or alterations of their function in adult life. Finally, we will summarize recent preclinical studies suggesting that elevated levels of AMH may potentially be a contributing factor to the central pathophysiology of PCOS and other reproductive diseases.
Collapse
|
29
|
Abstract
Although the fundamental symptoms of polycystic ovary syndrome (PCOS) relate most directly to ovarian dysfunction, central neuroendocrine systems play a prominent role in its pathophysiology. Gonadotropin-releasing hormone (GnRH) pulse generator resistance to negative feedback contributes to rapid GnRH pulse secretion, which promotes gonadotropin abnormalities that foster ovarian hyperandrogenemia and ovulatory dysfunction. The causes of GnRH neuron dysfunction, however, have remained enigmatic. In this review, we highlight a number of recent preclinical and clinical studies pertinent to the neuroendocrine abnormalities of PCOS, including those that have provided important insights into the relevance of animal models with PCOS-like features, the potential roles of kisspeptin and γ-aminobutyric acid (GABA)-ergic neurons, and the potential role of anti-Müllerian hormone.
Collapse
|
30
|
Ferreira SR, Goyeneche AA, Heber MF, Abruzzese GA, Telleria CM, Motta AB. Prenatally androgenized female rats develop uterine hyperplasia when adult. Mol Cell Endocrinol 2020; 499:110610. [PMID: 31589912 DOI: 10.1016/j.mce.2019.110610] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/19/2019] [Accepted: 10/03/2019] [Indexed: 12/20/2022]
Abstract
Prenatal hyperandrogenization (PH) is hypothesized as one of the main factors contributing to the development of polycystic ovary syndrome (PCOS). In this study, we aimed to investigate the impact of prenatal exposure to androgen excess on the uterus when animals reach their adulthood. We found that PH altered the morphology of the uteri that show a hyperplastic morphology with increased total uterine thickness as well as luminal epithelium thickness, with both enhanced and altered distribution of glands as compared with controls. Morphological alterations were associated with an unbalanced homeostasis as assessed by the expression of regulators of cell cycle progression and cell death dynamics. PH also causes disturbances in the cell cycle of the uterine tissue and dysregulates cell death and survival pathways leading to the development of uterine hyperplasia. These findings suggest that PH may have a deleterious effect on the uterus.
Collapse
Affiliation(s)
- Silvana Rocío Ferreira
- Laboratorio de Fisio-Patología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Argentina.
| | - Alicia Alejandra Goyeneche
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC H3A 2B4, Canada
| | - María Florencia Heber
- Laboratorio de Fisio-Patología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Argentina
| | - Giselle Adriana Abruzzese
- Laboratorio de Fisio-Patología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Argentina
| | - Carlos Marcelo Telleria
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC H3A 2B4, Canada
| | - Alicia Beatriz Motta
- Laboratorio de Fisio-Patología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Argentina
| |
Collapse
|
31
|
Abbott DH, Rogers J, Dumesic DA, Levine JE. Naturally Occurring and Experimentally Induced Rhesus Macaque Models for Polycystic Ovary Syndrome: Translational Gateways to Clinical Application. Med Sci (Basel) 2019; 7:medsci7120107. [PMID: 31783681 PMCID: PMC6950671 DOI: 10.3390/medsci7120107] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 11/16/2019] [Accepted: 11/16/2019] [Indexed: 12/19/2022] Open
Abstract
Indian rhesus macaque nonhuman primate models for polycystic ovary syndrome (PCOS) implicate both female hyperandrogenism and developmental molecular origins as core components of PCOS etiopathogenesis. Establishing and exploiting macaque models for translational impact into the clinic, however, has required multi-year, integrated basic-clinical science collaborations. Paradigm shifting insight has accrued from such concerted investment, leading to novel mechanistic understanding of PCOS, including hyperandrogenic fetal and peripubertal origins, epigenetic programming, altered neural function, defective oocytes and embryos, adipogenic constraint enhancing progression to insulin resistance, pancreatic decompensation and type 2 diabetes, together with placental compromise, all contributing to transgenerational transmission of traits likely to manifest in adult PCOS phenotypes. Our recent demonstration of PCOS-related traits in naturally hyperandrogenic (High T) female macaques additionally creates opportunities to employ whole genome sequencing to enable exploration of gene variants within human PCOS candidate genes contributing to PCOS-related traits in macaque models. This review will therefore consider Indian macaque model contributions to various aspects of PCOS-related pathophysiology, as well as the benefits of using macaque models with compellingly close homologies to the human genome, phenotype, development and aging.
Collapse
Affiliation(s)
- David H. Abbott
- Department of Obstetrics and Gynecology, Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, USA
- Correspondence: ; Tel.: +1-608-698-1953
| | - Jeffrey Rogers
- Department of Molecular and Human Genetics and Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Daniel A. Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA;
| | - Jon E. Levine
- Department of Neuroscience, Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, USA;
| |
Collapse
|
32
|
Abbott DH, Kraynak M, Dumesic DA, Levine JE. In utero Androgen Excess: A Developmental Commonality Preceding Polycystic Ovary Syndrome? FRONTIERS OF HORMONE RESEARCH 2019; 53:1-17. [PMID: 31499494 DOI: 10.1159/000494899] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In utero androgen excess reliably induces polycystic ovary syndrome (PCOS)-like reproductive and metabolic traits in female monkeys, sheep, rats, and mice. In humans, however, substantial technical and ethical constraints on fetal sampling have curtailed safe, pathogenic exploration during gestation. Evidence consistent with in utero origins for PCOS in humans has thus been slow to amass, but the balance now leans toward developmental fetal origins. Given that PCOS is familial and highly heritable, difficulties encountered in discerning genetic contributions to PCOS pathogenesis are puzzling and, to date, accounts for <10% of PCOS presentations. Unaccounted heritability notwithstanding, molecular commonality in pathogenic mechanisms is emerging, suggested by co-occurrence at the same gene loci of (1) PCOS genetic variants (PCOS women), (2) epigenetic alterations in DNA methylation (PCOS women), and (3) bioinformatics, gene networks-identified, epigenetic alterations in DNA methylation (female rhesus monkeys exposed to testosterone (T) in utero). In addition, naturally occurring hyperandrogenism in female monkeys singles out individuals with PCOS-like reproductive and metabolic traits accompanied by somatic biomarkers of in utero T exposure. Such phenotypic and molecular convergence between highly related species suggests not only dual genetic and epigenetic contributions to a developmental origin of PCOS but also common molecular pathogenesis extending beyond humans.
Collapse
Affiliation(s)
- David H Abbott
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA, .,Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin, USA, .,Endocrinology-Reproductive Physiology Training Program, University of Wisconsin, Madison, Wisconsin, USA,
| | - Marissa Kraynak
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA.,Endocrinology-Reproductive Physiology Training Program, University of Wisconsin, Madison, Wisconsin, USA
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Jon E Levine
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA.,Department of Neuroscience, University of Wisconsin, Madison, Wisconsin, USA.,Endocrinology-Reproductive Physiology Training Program, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
33
|
Witchel SF, Oberfield SE, Peña AS. Polycystic Ovary Syndrome: Pathophysiology, Presentation, and Treatment With Emphasis on Adolescent Girls. J Endocr Soc 2019; 3:1545-1573. [PMID: 31384717 PMCID: PMC6676075 DOI: 10.1210/js.2019-00078] [Citation(s) in RCA: 234] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/30/2019] [Indexed: 02/06/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous disorder characterized by hyperandrogenism and chronic anovulation. Depending on diagnostic criteria, 6% to 20% of reproductive aged women are affected. Symptoms of PCOS arise during the early pubertal years. Both normal female pubertal development and PCOS are characterized by irregular menstrual cycles, anovulation, and acne. Owing to the complicated interwoven pathophysiology, discerning the inciting causes is challenging. Most available clinical data communicate findings and outcomes in adult women. Whereas the Rotterdam criteria are accepted for adult women, different diagnostic criteria for PCOS in adolescent girls have been delineated. Diagnostic features for adolescent girls are menstrual irregularity, clinical hyperandrogenism, and/or hyperandrogenemia. Pelvic ultrasound findings are not needed for the diagnosis of PCOS in adolescent girls. Even before definitive diagnosis of PCOS, adolescents with clinical signs of androgen excess and oligomenorrhea/amenorrhea, features of PCOS, can be regarded as being "at risk for PCOS." Management of both those at risk for PCOS and those with a confirmed PCOS diagnosis includes education, healthy lifestyle interventions, and therapeutic interventions targeting their symptoms. Interventions can include metformin, combined oral contraceptive pills, spironolactone, and local treatments for hirsutism and acne. In addition to ascertaining for associated comorbidities, management should also include regular follow-up visits and planned transition to adult care providers. Comprehensive knowledge regarding the pathogenesis of PCOS will enable earlier identification of girls with high propensity to develop PCOS. Timely implementation of individualized therapeutic interventions will improve overall management of PCOS during adolescence, prevent associated comorbidities, and improve quality of life.
Collapse
Affiliation(s)
- Selma Feldman Witchel
- UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sharon E Oberfield
- Division of Pediatric Endocrinology, Columbia University Medical Center, New York–Presbyterian Morgan Stanley Children’s Hospital, New York, New York
| | - Alexia S Peña
- Robinson Research Institute, University of Adelaide, North Adelaide, South Australia, Australia
| |
Collapse
|
34
|
Vázquez-Martínez ER, Gómez-Viais YI, García-Gómez E, Reyes-Mayoral C, Reyes-Muñoz E, Camacho-Arroyo I, Cerbón M. DNA methylation in the pathogenesis of polycystic ovary syndrome. Reproduction 2019; 158:R27-R40. [DOI: 10.1530/rep-18-0449] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 04/08/2019] [Indexed: 12/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the leading endocrine and metabolic disorder in premenopausal women characterized by hyperandrogenism and abnormal development of ovarian follicles. To date, the PCOS etiology remains unclear and has been related to insulin resistance, obesity, type 2 diabetes mellitus, cardiovascular disease and infertility, among other morbidities. Substantial evidence illustrates the impact of genetic, intrauterine and environmental factors on the PCOS etiology. Lately, epigenetic factors have garnered considerable attention in the pathogenesis of PCOS considering that changes in the content of DNA methylation, histone acetylation and noncoding RNAs have been reported in various tissues of women with this disease. DNA methylation is changed in the peripheral and umbilical cord blood, as well as in ovarian and adipose tissue of women with PCOS, suggesting the involvement of this epigenetic modification in the pathogenesis of the disease. Perhaps, these defects in DNA methylation promote the deregulation of genes involved in inflammation, hormone synthesis and signaling and glucose and lipid metabolism. Research on the role of DNA methylation in the pathogenesis of PCOS is just beginning, and several issues await investigation. This review aims to provide an overview of current research focused on DNA methylation and PCOS, as well as discuss the perspectives regarding this topic.
Collapse
|
35
|
Affiliation(s)
- Mohamed A Abedal-Majed
- Department of Animal Production, School of Agriculture, University of Jordan, Amman, Jordan
| | - Andrea S Cupp
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE
| |
Collapse
|
36
|
Ryu Y, Kim SW, Kim YY, Ku SY. Animal Models for Human Polycystic Ovary Syndrome (PCOS) Focused on the Use of Indirect Hormonal Perturbations: A Review of the Literature. Int J Mol Sci 2019; 20:2720. [PMID: 31163591 PMCID: PMC6600358 DOI: 10.3390/ijms20112720] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/14/2019] [Accepted: 05/23/2019] [Indexed: 02/07/2023] Open
Abstract
Hormonal disturbances, such as hyperandrogenism, are considered important for developing polycystic ovary syndrome (PCOS) in humans. Accordingly, directly hormone-regulated animal models are widely used for studying PCOS, as they replicate several key PCOS features. However, the pathogenesis and treatment of PCOS are still unclear. In this review, we aimed to investigate animal PCOS models and PCOS-like phenotypes in animal experiments without direct hormonal interventions and determine the underlying mechanisms for a better understanding of PCOS. We summarized animal PCOS models that used indirect hormonal interventions and suggested or discussed pathogenesis of PCOS-like features in animals and PCOS-like phenotypes generated in other animals. We presented integrated physiological insights and shared cellular pathways underlying the pathogenesis of PCOS in reviewed animal models. Our review indicates that the hormonal and metabolic changes could be due to molecular dysregulations, such as upregulated PI3K-Akt and extracellular signal-regulated kinase (ERK) signalling, that potentially cause PCOS-like phenotypes in the animal models. This review will be helpful for considering alternative animal PCOS models to determine the cellular/molecular mechanisms underlying PCOS symptoms. The efforts to determine the specific cellular mechanisms of PCOS will contribute to novel treatments and control methods for this complex syndrome.
Collapse
Affiliation(s)
- Youngjae Ryu
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea; (Y.R.); (Y.Y.K.)
| | - Sung Woo Kim
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul 03080, Korea;
| | - Yoon Young Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea; (Y.R.); (Y.Y.K.)
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul 03080, Korea;
| | - Seung-Yup Ku
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea; (Y.R.); (Y.Y.K.)
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul 03080, Korea;
| |
Collapse
|
37
|
Vanky E, Engen Hanem LG, Abbott DH. Children born to women with polycystic ovary syndrome-short- and long-term impacts on health and development. Fertil Steril 2019; 111:1065-1075. [PMID: 31056313 DOI: 10.1016/j.fertnstert.2019.03.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/11/2019] [Accepted: 03/11/2019] [Indexed: 12/30/2022]
Abstract
Maternal PCOS status may negatively influence offspring infant and childhood growth, cardiometabolic health, reproductive health, and neurodevelopment. Current findings across studies are divergent, often because of small numbers of subjects, as well as heterogeneous selection criteria, ethnicities, and definitions of control groups. Coexisting maternal obesity, pregnancy complications, and comorbidity make it difficult to identify the contribution of maternal PCOS. Large, prospective, international, multiethnic studies with standardized investigation protocols and questionnaires on PCOS offspring health and development are needed.
Collapse
Affiliation(s)
- Eszter Vanky
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Department of Gynecology and Obstetrics, St. Olav's Hospital, Trondheim, Norway.
| | - Liv Guro Engen Hanem
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - David H Abbott
- Department of Obstetrics and Gynecology and Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
38
|
Bimber BN, Yan MY, Peterson SM, Ferguson B. mGAP: the macaque genotype and phenotype resource, a framework for accessing and interpreting macaque variant data, and identifying new models of human disease. BMC Genomics 2019; 20:176. [PMID: 30841849 PMCID: PMC6402181 DOI: 10.1186/s12864-019-5559-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 02/22/2019] [Indexed: 11/17/2022] Open
Abstract
Background Non-human primates (NHPs), particularly macaques, serve as critical and highly relevant pre-clinical models of human disease. The similarity in human and macaque natural disease susceptibility, along with parallel genetic risk alleles, underscores the value of macaques in the development of effective treatment strategies. Nonetheless, there are limited genomic resources available to support the exploration and discovery of macaque models of inherited disease. Notably, there are few public databases tailored to searching NHP sequence variants, and no other database making use of centralized variant calling, or providing genotype-level data and predicted pathogenic effects for each variant. Results The macaque Genotype And Phenotype (mGAP) resource is the first public website providing searchable, annotated macaque variant data. The mGAP resource includes a catalog of high confidence variants, derived from whole genome sequence (WGS). The current mGAP release at time of publication (1.7) contains 17,087,212 variants based on the sequence analysis of 293 rhesus macaques. A custom pipeline was developed to enable annotation of the macaque variants, leveraging human data sources that include regulatory elements (ENCODE, RegulomeDB), known disease- or phenotype-associated variants (GRASP), predicted impact (SIFT, PolyPhen2), and sequence conservation (Phylop, PhastCons). Currently mGAP includes 2767 variants that are identical to alleles listed in the human ClinVar database, of which 276 variants, spanning 258 genes, are identified as pathogenic. An additional 12,472 variants are predicted as high impact (SnpEff) and 13,129 are predicted as damaging (PolyPhen2). In total, these variants are predicted to be associated with more than 2000 human disease or phenotype entries reported in OMIM (Online Mendelian Inheritance in Man). Importantly, mGAP also provides genotype-level data for all subjects, allowing identification of specific individuals harboring alleles of interest. Conclusions The mGAP resource provides variant and genotype data from hundreds of rhesus macaques, processed in a consistent manner across all subjects (https://mgap.ohsu.edu). Together with the extensive variant annotations, mGAP presents unprecedented opportunity to investigate potential genetic associations with currently characterized disease models, and to uncover new macaque models based on parallels with human risk alleles. Electronic supplementary material The online version of this article (10.1186/s12864-019-5559-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Benjamin N Bimber
- Division of Genetics, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR, 97006, USA.,Division of Pathobiology, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR, 97006, USA
| | - Melissa Y Yan
- Division of Genetics, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR, 97006, USA
| | - Samuel M Peterson
- Division of Genetics, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR, 97006, USA
| | - Betsy Ferguson
- Division of Genetics, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR, 97006, USA. .,Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR, 97006, USA. .,Molecular and Medical Genetics Department, Oregon Health and Sciences University, Portland, OR, 97239, USA.
| |
Collapse
|
39
|
Abbott DH, Dumesic DA, Levine JE. Hyperandrogenic origins of polycystic ovary syndrome - implications for pathophysiology and therapy. Expert Rev Endocrinol Metab 2019; 14:131-143. [PMID: 30767580 PMCID: PMC6992448 DOI: 10.1080/17446651.2019.1576522] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 01/28/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) diagnosis comprises combinations of female hyperandrogenism, menstrual irregularity and polycystic ovaries. While it is a familial and highly prevalent endocrine disorder, progress towards a cure is hindered by absence of a definitive pathogenic mechanism and lack of an animal model of naturally occurring PCOS. AREAS COVERED These include an overview of PCOS and its potential etiology, and an examination of insights gained into its pathogenic origins. Animal models derived from experimentally-induced hyperandrogenism during gestation, or from naturally-occurring PCOS-like traits, most reliably demonstrate reproductive, neuroendocrine and metabolic pathogenesis. EXPERT OPINION Genetic studies, while identifying at least 17 PCOS risk genes, account for <10% of women with PCOS. A number of PCOS risk genes involve regulation of gonadotropin secretion or action, suggesting a reproductive neuroendocrine basis for PCOS pathogenesis. Consistent with this notion, a number of animal models employing fetal androgen excess demonstrate epigenetic induction of PCOS-like traits, including reproductive neuroendocrine and metabolic dysfunction. Monkey models are most comprehensive, while mouse models provide molecular insight, including identifying the androgen receptor, particularly in neurons, as mediating androgen-induced PCOS-like programming. Naturally-occurring female hyperandrogenism is also demonstrated in monkeys. Animal models are poised to delineate molecular gateways to PCOS pathogenesis.
Collapse
Affiliation(s)
- David H Abbott
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI, USA
- Department of Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, USA
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jon E Levine
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI, USA
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
40
|
Abbott DH. Metformin use in polycystic ovary syndrome pregnancy impacts on offspring obesity. THE LANCET CHILD & ADOLESCENT HEALTH 2019; 3:132-134. [PMID: 30704878 DOI: 10.1016/s2352-4642(19)30001-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 10/27/2022]
Affiliation(s)
- David H Abbott
- Department of Obstetrics and Gynecology, Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
41
|
Barbotin AL, Peigné M, Malone SA, Giacobini P. Emerging Roles of Anti-Müllerian Hormone in Hypothalamic-Pituitary Function. Neuroendocrinology 2019; 109:218-229. [PMID: 31280262 PMCID: PMC6878735 DOI: 10.1159/000500689] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/01/2019] [Indexed: 12/29/2022]
Abstract
Since its initial discovery in the 1940s, research into the physiological actions of anti-Müllerian hormone (AMH), from its eponymous role in male developmental biology to its routine clinical use in female reproductive health, has undergone a paradigm shifting change. With several exciting studies recently reporting hitherto unforeseen AMH actions at all levels in the hypogonadal-pituitary-gonadal axis, the importance of this hormone for both hypothalamic and pituitary reproductive control is finding increasing support and significance. In this review, we will briefly summarize what is known about the traditional roles and biology of AMH and how this could be integrated with new findings of AMH actions at the level of the hypothalamic-pituitary axis. We also synthesize the important findings from these new studies and discuss their potential impact and significance to our understanding of one of the most common reproductive disorders currently affecting women, polycystic ovary syndrome.
Collapse
Affiliation(s)
- Anne-Laure Barbotin
- Université de Lille, Inserm, CHU Lille, UMR-S 1172, Laboratoire du Développement et Plasticité du Cerveau Neuroendocrine, Centre de Recherche Jean-Pierre Aubert, Lille, France
- Institut de Biologie de la Reproduction-Spermiologie-CECOS, CHU de Lille, Lille, France
| | - Maëliss Peigné
- Université de Lille, Inserm, CHU Lille, UMR-S 1172, Laboratoire du Développement et Plasticité du Cerveau Neuroendocrine, Centre de Recherche Jean-Pierre Aubert, Lille, France
- AP-HP, Unité de Médecine de la Reproduction, Service de Gynécologie-Obstétrique, Hôpital Bichat-Claude Bernard, Paris, France
| | - Samuel Andrew Malone
- Université de Lille, Inserm, CHU Lille, UMR-S 1172, Laboratoire du Développement et Plasticité du Cerveau Neuroendocrine, Centre de Recherche Jean-Pierre Aubert, Lille, France
| | - Paolo Giacobini
- Université de Lille, Inserm, CHU Lille, UMR-S 1172, Laboratoire du Développement et Plasticité du Cerveau Neuroendocrine, Centre de Recherche Jean-Pierre Aubert, Lille, France,
| |
Collapse
|
42
|
Tonellotto Dos Santos J, Escarião da Nóbrega J, Serrano Mujica LK, Dos Santos Amaral C, Machado FA, Manta MW, Rizzetti TM, Zanella R, Fighera R, Antoniazzi AQ, Gonçalves PBD, Comim FV. Prenatal Androgenization of Ewes as a Model of Hirsutism in Polycystic Ovary Syndrome. Endocrinology 2018; 159:4056-4064. [PMID: 30376052 DOI: 10.1210/en.2018-00781] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/24/2018] [Indexed: 02/04/2023]
Abstract
The main clinical feature associated with hyperandrogenism in polycystic ovary syndrome (PCOS) in humans is hirsutism, where hair increases its length, pigmentation, and particularly its diameter. Currently, it is not known whether PCOS animal models also exhibit changes in the hair. Therefore, the aim of this study was to explore the wool characteristics in sheep prenatally androgenized (PA) with testosterone propionate. After 4 and 13 months of life, wool was collected from the top of the shoulder of both females and males (both androgenized and controls). The offspring sheep were followed for up to 19 months of life to evaluate testosterone and androstenedione serum levels by ultra-high-performance liquid chromatography-tandem mass spectrometry, determine insulin and glucose response to intravenous glucose tolerance test, and address estrus cyclicity during the second breeding season. PA male animals showed a reduction in wool fiber diameter at 4 months of age compared with controls (P = 0.02) but not at 13 months, whereas PA females showed increased hair diameter at 13 months (P = 0.002), with no difference at 4 months. No substantial changes in other hair parameters (length, color, and medullation) were identified. In addition, increased levels of serum testosterone were observed in PA female sheep compared with controls at 12 months (P = 0.03). Our results indicate for the first time, to our knowledge, that changes in wool fiber diameter observed in PA ewes replicate, at the translational level, the increase in hair diameter in hirsute women with PCOS.
Collapse
Affiliation(s)
- Joabel Tonellotto Dos Santos
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Jandui Escarião da Nóbrega
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Lady Katerine Serrano Mujica
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Carolina Dos Santos Amaral
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Fabrício Amadori Machado
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Manuela W Manta
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Tiele Medianeira Rizzetti
- Laboratory of Pesticide Analysis (LARP), Department of Chemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | - Renato Zanella
- Laboratory of Pesticide Analysis (LARP), Department of Chemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | - Rafael Fighera
- Department of Pathology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Alfredo Quites Antoniazzi
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Paulo Bayard Dias Gonçalves
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Fabio Vasconcellos Comim
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
- Department of Clinical Medicine, Health Science Center, Federal University of Santa Maria, Santa Maria, Brazil
| |
Collapse
|
43
|
Abruzzese GA, Crisosto N, De Grava Kempinas W, Sotomayor-Zárate R. Developmental programming of the female neuroendocrine system by steroids. J Neuroendocrinol 2018; 30:e12632. [PMID: 29968423 DOI: 10.1111/jne.12632] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 06/03/2018] [Accepted: 07/01/2018] [Indexed: 12/30/2022]
Abstract
Developmental programming refers to processes that occur during early life that may have long-term consequences, modulating adult health and disease. Complex diseases, such as diabetes, cancer and cardiovascular disease, have a high prevalence in different populations, are multifactorial, and may have a strong environmental component. The environment interacts with organisms, affecting their behaviour, morphology and physiology. This interaction may induce permanent or long-term changes, and organisms may be more susceptible to environmental factors during certain developmental stages, such as the prenatal and early postnatal periods. Several factors have been identified as responsible for inducing the reprogramming of various reproductive and nonreproductive tissues. Among them, both natural and synthetic steroids, such as endocrine disruptors, are known to have either detrimental or positive effects on organisms depending on the dose of exposure, stage of development and biological sexual background. The present review focuses on the action of steroids and endocrine disruptors as agents involved in developmental programming and on their modulation and effects on female neuroendocrine functions.
Collapse
Affiliation(s)
- Giselle Adriana Abruzzese
- Laboratorio de Fisio-patología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Nicolás Crisosto
- Endocrinology and Metabolism Laboratory West Division, School of Medicine, University of Chile, Santiago, Chile
- Endocrinology Unit, Clínica Las Condes, Santiago, Chile
| | - Wilma De Grava Kempinas
- Laboratory of Reproductive and Developmental Biology and Toxicology, Department of Morphology, Institute of Biosciences, Universidade Estadual Paulista-UNESP, Botucatu, Sao Paulo, Brazil
| | - Ramón Sotomayor-Zárate
- Laboratorio de Neuroquímica y Neurofarmacología, Centro de Neurobiología y Fisiopatología Integrativa, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
44
|
Min Z, Gao Q, Zhen X, Fan Y, Tan T, Li R, Zhao Y, Yu Y. New insights into the genic and metabolic characteristics of induced pluripotent stem cells from polycystic ovary syndrome women. Stem Cell Res Ther 2018; 9:210. [PMID: 30092830 PMCID: PMC6085636 DOI: 10.1186/s13287-018-0950-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/02/2018] [Accepted: 07/04/2018] [Indexed: 12/19/2022] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder that affects female fertility. However, with the lack of a corresponding research model, the pathology mechanism of PCOS is poorly understood. Induced pluripotent stem cell (iPSC) technology has been recognized as means to generate patient-specific stem cells for disease modeling. Methods The mRNA abundance of iPSCs was analyzed by RNA microarray and real-time polymerase chain reaction (RT-PCR). Karyotyping of iPSCs was performed with cytogenetic analysis. The mitochondrial respiration ability and glycolytic function were measured by the Seahorse Bioscience XF extracellular flux analyzer. The expression of iPSC-associated markers was identified by immunofluorescence and RT-PCR. The teratoma formation of iPSCs was studied using immunochemistry. Results A PCOS patient-derived iPSC model was established from somatic cells of PCOS patients. Through comprehensive transcriptional profiling analysis of the RNA microarray, PCOS patient-derived iPSCs showed metabolic abnormalities and mitochondrial dysfunction compared with non-PCOS patient-derived iPSCs in vitro. Specifically, a total of 2904 genes were differentially expressed between the two iPSC populations, of which 1416 genes were upregulated and 1488 genes were downregulated (fold change > 2, p < 0.01). Gene Ontology (GO) term enrichment results showed that upregulated genes were enriched in metabolic processes and mitochondrial activities which participated in the tricarboxylic acid (TCA) cycle, the respiratory electron transport chain (ETC), and glycogenolysis. On the other hand, the downregulated genes were related to cell communication, glucose transport, and uptake. The differentially expressed genes were verified by RT-PCR in PCOS patient-derived iPSCs and granulosa cells from PCOS patients. The PCOS patient-derived iPSCs demonstrated decreased mitochondrial respiration ability and glycolytic function (p < 0.05) but increased mitochondrial copy numbers and biogenesis (p < 0.05). Subsequently, some genes related to glucose metabolism were rescued by treating with metformin in PCOS patient-derived iPSCs. Meanwhile, the ATP production ability of mitochondria and the glycolysis ability of PCOS patient-derived iPSCs also partially returned to normal levels. However, metformin had little effect on mitochondrial maximal respiration ability and maximal glycolytic capacity. Conclusions We measured differences in iPSCs from women with and without PCOS in gene transcription and mitochondrial respiratory function. PCOS patient-derived iPSCs showed abnormal expression of metabolic genes and mitochondrial dysfunction in vitro. The study provides a novel cell model in vitro for studying the clinical causes and molecular mechanisms of PCOS. Electronic supplementary material The online version of this article (10.1186/s13287-018-0950-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zheying Min
- Department of Obstetrics and Gynecology, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Qian Gao
- Department of Obstetrics and Gynecology, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Xiumei Zhen
- Department of Obstetrics and Gynecology, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Tao Tan
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
| | - Rong Li
- Department of Obstetrics and Gynecology, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Yue Zhao
- Department of Obstetrics and Gynecology, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China.
| | - Yang Yu
- Department of Obstetrics and Gynecology, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
45
|
Walters KA, Bertoldo MJ, Handelsman DJ. Evidence from animal models on the pathogenesis of PCOS. Best Pract Res Clin Endocrinol Metab 2018; 32:271-281. [PMID: 29779581 DOI: 10.1016/j.beem.2018.03.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Polycystic ovarian syndrome (PCOS) is the most common endocrine condition in women, and is characterized by reproductive, endocrine and metabolic features. However, there is no simple unequivocal diagnostic test for PCOS, its etiology remains unknown and there is no cure. Hence, the management of PCOS is suboptimal as it relies on the ad hoc empirical management of its symptoms only. Decisive studies are required to unravel the origins of PCOS, but due to ethical and logistical reasons these are not possible in humans. Experimental animal models for PCOS have been established which have enhanced our understanding of the mechanisms underlying PCOS and propose novel mechanism-based therapies to treat the condition. This review examines the findings from various animal models to reveal the current knowledge of the mechanisms underpinning the development of PCOS, and also provides insights into the implications from these studies for improved clinical management of this disorder.
Collapse
Affiliation(s)
- K A Walters
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, NSW 2052, Australia.
| | - M J Bertoldo
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, NSW 2052, Australia.
| | - D J Handelsman
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales 2139, Australia.
| |
Collapse
|
46
|
Elevated prenatal anti-Müllerian hormone reprograms the fetus and induces polycystic ovary syndrome in adulthood. Nat Med 2018; 24:834-846. [PMID: 29760445 PMCID: PMC6098696 DOI: 10.1038/s41591-018-0035-5] [Citation(s) in RCA: 253] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 03/12/2018] [Indexed: 01/01/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the main cause of female infertility worldwide and corresponds with a high degree of comorbidities and economic burden. How PCOS is passed on from one generation to the next is not clear, but it may be a developmental condition. Most women with PCOS exhibit higher levels of circulating luteinizing hormone, suggestive of heightened gonadotropin-releasing hormone (GnRH) release, and Anti-Müllerian Hormone (AMH) as compared to healthy women. Excess AMH in utero may affect the development of the female fetus. However, as AMH levels drop during pregnancy in women with normal fertility it was unclear if their levels were also elevated in pregnant women with PCOS. Here, we measured AMH in a cohort of pregnant women with PCOS and control women and found that AMH is significantly more elevated in the former group versus the latter. To determine if the elevation of AMH during pregnancy in women with PCOS is a bystander effect or a driver of the condition in the offspring, we modelled our clinical findings by treating pregnant mice with AMH and followed the neuroendocrine phenotype of their female progeny postnatally. This treatment resulted in maternal neuroendocrine-driven testosterone excess and diminished placental metabolism of testosterone to estradiol, resulting in a masculinization of the exposed female fetus and a PCOS-like reproductive and neuroendocrine phenotype in adulthood. We found that the affected females had persistently hyperactivated GnRH neurons and that GnRH antagonist treatment in the adult female offspring restored their neuroendocrine phenotype to a normal state. These findings highlight a critical role for excess prenatal AMH exposure and subsequent aberrant GnRH receptor signaling in the neuroendocrine dysfunctions of PCOS, while offering a new potential therapeutic avenue to treat the condition during adulthood.
Collapse
|
47
|
Wang Z, Shen M, Xue P, DiVall SA, Segars J, Wu S. Female Offspring From Chronic Hyperandrogenemic Dams Exhibit Delayed Puberty and Impaired Ovarian Reserve. Endocrinology 2018; 159:1242-1252. [PMID: 29315373 PMCID: PMC5793796 DOI: 10.1210/en.2017-03078] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/28/2017] [Indexed: 11/19/2022]
Abstract
Female offspring of many species exposed to high doses of androgens in utero experience endocrine dysfunction during adulthood. The phenotype of offspring from females with prepregnancy hyperandrogenemia and impaired ovulation, however, has not been examined. We developed a mouse model of hyperandrogenemia by implanting a low-dose dihydrotestosterone (DHT) pellet 15 days before conception. Female offspring born to dams with hyperandrogenemia (DHT daughters) had delayed puberty (P < 0.05) with first estrus on postnatal day (PND) 41 compared with daughters from dams with physiological levels of DHT (non-DHT daughters, PND37.5). Serum follicle-stimulating hormone (FSH) levels in the DHT daughters were fourfold higher (P < 0.05) on PND21, and anti-Müllerian hormone levels were higher (P < 0.05) on PND26 than in non-DHT daughters (controls). DHT daughters showed an extended time in metestrus/diestrus and a shorter time in the proestrus/estrus phase compared with non-DHT daughters (P < 0.05). To examine ovarian response to gonadotropins, superovulation was induced and in vitro fertilization (IVF) was performed. Fewer numbers of oocytes were retrieved from the DHT daughters compared with non-DHT daughters (P < 0.05). At IVF, there was no difference in rates of fertilization or cleavage of oocytes from either group. There were fewer (P < 0.01) primordial follicles (6.5 ± 0.8 vs 14.5 ± 2.1 per ovary) in the ovaries of DHT daughters compared with non-DHT daughters. Daughters from hyperandrogenemic females exhibited elevated prepubertal FSH levels, diminished ovarian response to superovulation, impaired estrous cyclicity, delayed onset of puberty, and reduced ovarian reserve, suggesting that fetal androgen exposure had lasting effects on female reproductive function.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | - Mingjie Shen
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
- Department of Gynecology/Obstetrics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 21203, People’s Republic of China
| | - Ping Xue
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | - Sara A. DiVall
- Department of Pediatrics, Seattle Children’s Hospital, Seattle, Washington 98105
| | - James Segars
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | - Sheng Wu
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| |
Collapse
|
48
|
Abbott DH, Vepraskas SH, Horton TH, Terasawa E, Levine JE. Accelerated Episodic Luteinizing Hormone Release Accompanies Blunted Progesterone Regulation in PCOS-like Female Rhesus Monkeys (Macaca Mulatta) Exposed to Testosterone during Early-to-Mid Gestation. Neuroendocrinology 2018; 107:133-146. [PMID: 29949806 PMCID: PMC7363207 DOI: 10.1159/000490570] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 06/04/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND/AIMS Ovarian theca cell hyperandrogenism in women with polycystic ovary syndrome (PCOS) is compounded by androgen receptor-mediated impairment of estradiol and progesterone negative feedback regulation of episodic luteinizing hormone (LH) release. The resultant LH hypersecretion, likely the product of accelerated episodic release of gonadotropin-releasing hormone (GnRH) from the median eminence of the hypothalamus, hyperstimulates ovarian theca cell steroidogenesis, enabling testosterone (T) and androstenedione excess. Prenatally androgenized (PA) female monkeys exposed to fetal male levels of T during early-to-mid gestation, when adult, demonstrate PCOS-like traits, including high T and LH levels. This study tests the hypothesis that progesterone resistance-associated acceleration in episodic LH release contributes to PA monkey LH excess. METHODS A total of 4 PA and 3 regularly cycling, healthy control adult female rhesus monkeys of comparable age and body mass index underwent (1) a 10 h, frequent intravenous sampling assessment for LH episodic release, immediately followed by (2) IV infusion of exogenous GnRH to quantify continuing pituitary LH responsiveness, and subsequently (3) an SC injection of a progesterone receptor antagonist, mifepristone, to examine LH responses to blockade of progesterone-mediated action. RESULTS Compared to controls, the relatively hyperandrogenic PA females exhibited ~100% increase (p = 0.037) in LH pulse frequency, positive correlation of LH pulse amplitude (p = 0.017) with androstenedione, ~100% greater increase (p = 0.034) in acute (0-10 min) LH responses to exogenous GnRH, and an absence (p = 0.008) of modest LH elevation following acute progesterone receptor blockade suggestive of diminished progesterone negative feedback. CONCLUSION Such dysregulation of LH release in PCOS-like monkeys implicates impaired feedback control of episodic release of hypothalamic GnRH reminiscent of PCOS neuroendocrinopathy.
Collapse
Affiliation(s)
- David H Abbott
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Sarah H Vepraskas
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin, USA
| | - Teresa H Horton
- Department of Neurobiology and Physiology, Institute for Neuroscience, Center for Reproductive Science, Northwestern University, Evanston, Illinois, USA
| | - Ei Terasawa
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Jon E Levine
- Department of Neuroscience, University of Wisconsin, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
49
|
Abbott DH. Neuronal androgen receptor: Molecular gateway to polycystic ovary syndrome? Proc Natl Acad Sci U S A 2017; 114:4045-4047. [PMID: 28377515 PMCID: PMC5402428 DOI: 10.1073/pnas.1703436114] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023] Open
Affiliation(s)
- David H Abbott
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53715;
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715
| |
Collapse
|