1
|
Luo S, Zhang X, Liu Z, Wang C, Pei J, Yu Y, Liu H, Gu W. Low-dose aspirin for the prevention of preeclampsia in women with polycystic ovary syndrome: a retrospective cohort study. BMC Pregnancy Childbirth 2025; 25:98. [PMID: 39885419 DOI: 10.1186/s12884-025-07183-1] [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: 05/03/2024] [Accepted: 01/15/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND The objective of this study was to investigate the efficacy of low-dose aspirin (LDA) in preventing preeclampsia among pregnant women with polycystic ovary syndrome (PCOS), given the increased susceptibility of this population to preeclampsia development. METHODS A retrospective cohort study was conducted on pregnant women with PCOS who delivered between January 1, 2018 and February 10, 2024 at our institution. Clinical characteristics and obstetric data were extracted from medical records. Propensity score matching (PSM) was employed to analyze the association between LDA use and PE incidence. RESULTS The study cohort comprised 1522 pregnant women with PCOS. Among 395 pregnant women identified as high-risk for preeclampsia, 98 were administered LDA for preeclampsia prevention, while 297 did not receive LDA. Following PSM, no statistically significant difference was observed in preeclampsia risk between the LDA and non-LDA groups. Additionally, maternal and neonatal outcomes were comparable between the two groups. CONCLUSIONS This cohort analysis did not provide sufficient evidence to support the efficacy of LDA in preventing preeclampsia among PCOS patients at high risk for preeclampsia.
Collapse
Affiliation(s)
- Shouling Luo
- The Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Huangpu Area, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Xiaoyue Zhang
- The Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Huangpu Area, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Zhenzhen Liu
- The Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Huangpu Area, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Chengjie Wang
- The Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Huangpu Area, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Jiangnan Pei
- The Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Huangpu Area, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yi Yu
- The Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Huangpu Area, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Haiyan Liu
- The Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Huangpu Area, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Weirong Gu
- The Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Huangpu Area, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.
| |
Collapse
|
2
|
Jin Q, Xu G, Ying Y, Liu L, Zheng H, Xu S, Yin P, Chen Y. Effects of non-pharmacological interventions on biochemical hyperandrogenism in women with polycystic ovary syndrome: a systematic review and network meta-analysis. J Ovarian Res 2025; 18:8. [PMID: 39833948 PMCID: PMC11744805 DOI: 10.1186/s13048-025-01595-5] [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: 09/19/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE To systematically evaluate the effectiveness of non-pharmacological interventions (NPIs), including electroacupuncture, exercise, diet, and lifestyle changes, in reducing androgen levels in women with polycystic ovary syndrome (PCOS) through a systematic review and network meta-analysis. METHODS Comprehensive searches were conducted in PubMed, Embase, Cochrane Library, Web of Science, CNKI, and Wanfang up to June 2024. Randomized controlled trials (RCTs) comparing NPIs with other NPIs or placebo treatments in adult women with PCOS were included. Study selection was independently performed by three authors. Quality assessment followed PRISMA guidelines using the Cochrane RoB2 tool. The confidence of evidence was examined using Confidence in Network Meta-Analysis (CINeMA). Traditional meta-analysis of continuous variables was conducted using Stata 17.0 software with a random-effects model, reporting effect sizes as standardized mean differences (SMD) and weighted mean differences (WMD). Network meta-analysis (NMA) was used to synthesize data, with network diagrams illustrating comparisons between NPIs. We assessed the consistency of the results, performed sensitivity analyses, and examined publication bias to evaluate the influence of individual studies. Furthermore, subgroup analysis and network meta-regression analysis were conducted to explore potential sources of heterogeneity. RESULTS The review included 21 studies with 1,196 participants, with meta-analysis focusing on 17 studies involving 1,013 participants. NPIs significantly reduced serum testosterone (SMD = -0.57; 95% CI: -0.86 to -0.29, p < 0.01), A4 (SMD = -1.37; 95% CI: -2.63 to -0.12, p = 0.03), and mFG score (WMD = -0.81; 95% CI: -1.26 to -0.37, p < 0.01). Notably, the reduction in testosterone levels achieved with NPIs met the Minimum Clinically Important Difference (MCID) of 12.47 ng/dL (WMD = -12.57; 95% CI: -18.92 to -6.23; p < 0.01), affirming the clinical relevance of these reductions. No significant effects were observed on Free Androgen Index (FAI), Sex Hormone-Binding Globulin (SHBG), Dehydroepiandrosterone (DHEA), DHEA Sulfate (DHEAS), Free Testosterone (FT), or Dihydrotestosterone (DHT) levels (all p > 0.05). The NMA (18 studies, 1,067 participants) identified electroacupuncture combined with diet and exercise as the most effective intervention for reducing serum testosterone (WMD = -21.75; 95% CI: -49.58 to 6.07; SUCRA 72.3%). Evidence certainty for many interventions was low, highlighting the need for higher-quality studies. Sensitivity analysis confirmed the robustness of the findings, and no publication bias was detected. CONCLUSIONS NPIs, particularly electroacupuncture combined with exercise and dietary management, effectively reduce androgen levels in PCOS patients. These findings provide valuable guidance for clinicians and women with PCOS, with multi-component approaches recommended for more substantial clinical benefit. TRIAL REGISTRATION PROSPERO CRD42023426226.
Collapse
Affiliation(s)
- Qi Jin
- LongHua Hospital Shanghai University of Traditional Chinese Medicine, No.725 Wanping South Road, Xuhui District, Shanghai, 200032, China
- Shanghai Municipal Hospital of Traditional Chinese Medicine, China. No. 274 Zhijiang Middle Road, Jing'an District, Shanghai, 200071, China
| | - Ge Xu
- Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Pudong New Area, Shanghai, 201203, China
| | - Yuchen Ying
- LongHua Hospital Shanghai University of Traditional Chinese Medicine, No.725 Wanping South Road, Xuhui District, Shanghai, 200032, China
| | - Lumin Liu
- LongHua Hospital Shanghai University of Traditional Chinese Medicine, No.725 Wanping South Road, Xuhui District, Shanghai, 200032, China
| | - Huimin Zheng
- Shanghai Municipal Hospital of Traditional Chinese Medicine, China. No. 274 Zhijiang Middle Road, Jing'an District, Shanghai, 200071, China
| | - Shifen Xu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, China. No. 274 Zhijiang Middle Road, Jing'an District, Shanghai, 200071, China
| | - Ping Yin
- LongHua Hospital Shanghai University of Traditional Chinese Medicine, No.725 Wanping South Road, Xuhui District, Shanghai, 200032, China.
| | - Yuelai Chen
- LongHua Hospital Shanghai University of Traditional Chinese Medicine, No.725 Wanping South Road, Xuhui District, Shanghai, 200032, China.
| |
Collapse
|
3
|
Lakshmanan M, Saini M, Nune M. Exploring the innovative application of cerium oxide nanoparticles for addressing oxidative stress in ovarian tissue regeneration. J Ovarian Res 2024; 17:241. [PMID: 39633503 PMCID: PMC11619646 DOI: 10.1186/s13048-024-01566-2] [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: 10/02/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024] Open
Abstract
The female reproductive system dysfunction considerably affects the overall health of women and children on a global scale. Over the decade, the incidence of reproductive disorders has become a significant source of suffering for women. Infertility in women may be caused by a range of acquired and congenital abnormalities. Ovaries play a central role in the female reproductive function. Any defect in the normal functioning of these endocrine organs causes health issues and reproductive challenges extending beyond infertility, as the hormones interact with other tissues and biological processes in the body. The complex pathophysiology of ovarian disorders makes it a multifactorial disease. The key etiological factors associated with the diseases include genetic factors, hormonal imbalance, environmental and lifestyle factors, inflammatory conditions, oxidative stress, autoimmune diseases, metabolic factors, and age. Oxidative stress is a major contributor to disease development and progression affecting the oocyte quality, fertilization, embryo development, and implantation. The choice of treatment for ovarian disorders varies among individuals and has associated complications. Reproductive tissue engineering holds great promise for overcoming the challenges associated with the current therapeutic approach to tissue regeneration. Furthermore, incorporating nanotechnology into tissue engineering could offer an efficient treatment strategy. This review provides an overview of incorporating antioxidant nanomaterials for engineering ovarian tissue to address the disease recurrence and associated pathophysiology. Cerium oxide nanoparticles (CeO2 NPs) are prioritized for evaluation primarily due to their antioxidant properties. In conclusion, the review explores the potential applications of CeO2 NPs for effective and clinically significant ovarian tissue regeneration.
Collapse
Affiliation(s)
- Maya Lakshmanan
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Monika Saini
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi, 110029, India
| | - Manasa Nune
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
4
|
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
|
5
|
Pratama G, Wiweko B, Asmarinah, Widyahening IS, Andraini T, Bayuaji H, Hestiantoro A. Mechanism of elevated LH/FSH ratio in lean PCOS revisited: a path analysis. Sci Rep 2024; 14:8229. [PMID: 38589425 PMCID: PMC11002031 DOI: 10.1038/s41598-024-58064-0] [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: 11/19/2023] [Accepted: 03/25/2024] [Indexed: 04/10/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder affecting 5-20% of reproductive-age women. However, the treatment of PCOS is mainly based on symptoms and not on its pathophysiology. Neuroendocrine disturbance, as shown by an elevated LH/FSH ratio in PCOS patients, was thought to be the central mechanism of the syndrome, especially in lean PCOS. LH and FSH secretion are influenced by GnRH pulsatility of GnRH neurons in the hypothalamus. Kisspeptin is the main regulator of GnRH secretion, whereas neurokinin B (NKB) and dynorphin regulate kisspeptin secretion in KNDy neurons. This study aims to deepen the understanding of the neuroendocrine disorder in lean PCOS patients and its potential pathophysiology-based therapy. A cross-sectional study was performed at Dr. Cipto Mangunkusumo Kencana Hospital and the IMERI UI HRIFP cluster with 110 lean PCOS patients as subjects. LH, FSH, LH/FSH ratio, kisspeptin, NKB, dynorphin, leptin, adiponectin, AMH, fasting blood glucose, fasting insulin, HOMA-IR, testosterone, and SHBG were measured. Bivariate and path analyses were performed to determine the relationship between variables. There was a negative association between dynorphin and kisspeptin, while NKB levels were not associated with kisspeptin. There was no direct association between kisspeptin and the LH/FSH ratio; interestingly, dynorphin was positively associated with the LH/FSH ratio in both bivariate and pathway analyses. AMH was positively correlated with the LH/FSH ratio in both analyses. Path analysis showed an association between dynorphin and kisspeptin levels in lean PCOS, while NKB was not correlated with kisspeptin. Furthermore, there was a correlation between AMH and the LH/FSH ratio, but kisspeptin levels did not show a direct significant relationship with the LH/FSH ratio. HOMA-IR was negatively associated with adiponectin levels and positively associated with leptin and FAI levels. In conclusion, AMH positively correlates with FAI levels and is directly associated with the LH/FSH ratio, showing its important role in neuroendocrinology in lean PCOS. From the path analysis, AMH was also an intermediary variable between HOMA-IR and FAI with the LH/FSH ratio. Interestingly, this study found a direct positive correlation between dynorphin and the LH/FSH ratio, while no association between kisspeptin and the LH/FSH ratio was found. Further research is needed to investigate AMH and dynorphin as potential therapeutic targets in the management of lean PCOS patients.
Collapse
Affiliation(s)
- Gita Pratama
- Department of Obstetrics and Gynecology, Faculty of Medicine Universitas Indonesia, Dr. Cipto Mangunkusumo General Hospital, Jakarta, Indonesia.
- Cluster of Human Reproduction, Infertility and Family Planning, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.
- Yasmin IVF Clinic, Dr. Cipto Mangunkusumo General Hospital, Jakarta, Indonesia.
| | - Budi Wiweko
- Department of Obstetrics and Gynecology, Faculty of Medicine Universitas Indonesia, Dr. Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
- Cluster of Human Reproduction, Infertility and Family Planning, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
- Yasmin IVF Clinic, Dr. Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Asmarinah
- Department of Medical Biology, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Indah S Widyahening
- Department of Community Medicine, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Trinovita Andraini
- Department of Physiology, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Hartanto Bayuaji
- Department of Obstetrics and Gynecology, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Andon Hestiantoro
- Department of Obstetrics and Gynecology, Faculty of Medicine Universitas Indonesia, Dr. Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
- Cluster of Human Reproduction, Infertility and Family Planning, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
- Yasmin IVF Clinic, Dr. Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| |
Collapse
|
6
|
Kolnikaj TS, Herman R, Janež A, Jensterle M. The Current and Emerging Role of Statins in the Treatment of PCOS: The Evidence to Date. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:244. [PMID: 38399531 PMCID: PMC10890374 DOI: 10.3390/medicina60020244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024]
Abstract
Polycystic ovary syndrome (PCOS) manifests a multifactorial pathology characterized by polycystic ovaries, menstrual cycle disorders, varying degrees of hyperandrogenism, and an ad-verse metabolic risk profile. The position of hyperandrogenism in this syndrome has been extensively studied. A multitude of mechanisms place it in the position of cause but also of consequence; therefore, ongoing research efforts are focused on identifying medications that can effectively reduce levels of androgens in women with PCOS. Moreover, lipid abnormalities are common in this population, with up to 70% of patients having dyslipidemia. Statins may have potential therapeutic benefits for women with PCOS, as they have been shown to improve insulin resistance and reduce the risk of cardiovascular disease. In addition, their role in accelerated steroidogenesis by limiting one source of cholesterol, influencing enzymatic activity, and providing several other beneficial mechanisms is widely investigated. This review aimed to provide a comprehensive overview of the pathogenesis of androgen excess and dyslipidemia in PCOS, as well as the therapeutic potential of statins.
Collapse
Affiliation(s)
- Tea Shehu Kolnikaj
- Department of Endocrinology, Diabetes and Metabolic Diseases, University of Medicine Tirana, 1000 Tirana, Albania;
| | - Rok Herman
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia; (R.H.); (A.J.)
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Andrej Janež
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia; (R.H.); (A.J.)
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Mojca Jensterle
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia; (R.H.); (A.J.)
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
7
|
Абсатарова ЮС, Андреева ЕН, Евсеева ЮС, Зеленкова-Захарчук ТА, Шереметьева ЕВ, Григорян ОР, Михеев РК. [Endocrine and psychosomatic disorders in patients with amenorrhea]. PROBLEMY ENDOKRINOLOGII 2024; 69:121-131. [PMID: 38312002 PMCID: PMC10848186 DOI: 10.14341/probl13366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 09/22/2023] [Indexed: 02/06/2024]
Abstract
The article presents data on the relationship of pathogenetic mechanisms for the development of menstrual disorders of functional and organic origin in connection with mental disturbances from the point of view of the psychosomatic concept. According to the latter, functional disorders of the menstrual cycle are considered as psychosomatic, in which gynecological pathology develops as a result of psychopathological illness. A striking example of such a disorder is functional hypothalamic amenorrhea. At the same time, endocrinopathies, such as polycystic ovary syndrome and premature ovarian insufficiency, can also be considered in the paradigm of psychosomatic illnesses of ovarian function due to the high prevalence of anxiety and depressive disorders in this cohort of patients. This review highlights the importance of interdisciplinary collaboration between a gynecologist and a psychiatrist for the most effective reproductive rehabilitation of patients with amenorrhea. Literature search was carried out in national (eLibrary, CyberLeninka.ru) and international (PubMed, Cochrane Library) databases in Russian and English. The priority was free access to the full text of articles. The choice of sources was prioritized for the period from 2018 to 2023.However, taking into account the insufficient knowledge of the chosen topic, the choice of sources dates back to 1985.
Collapse
Affiliation(s)
| | - Е. Н. Андреева
- Национальный медицинский исследовательский центр эндокринологии; Московский государственный медико-стоматологический университет им. А.И. Евдокимова Министерства здравоохранения Российской Федерации
| | - Ю. С. Евсеева
- Национальный медицинский исследовательский центр эндокринологии
| | | | | | - О. Р. Григорян
- Национальный медицинский исследовательский центр эндокринологии
| | - Р. К. Михеев
- Национальный медицинский исследовательский центр эндокринологии
| |
Collapse
|
8
|
Sarkisian KI, Ho L, Yang J, Mandelbaum R, Stanczyk FZ. Neuroendocrine, neurotransmitter, and gut microbiota imbalance contributing to potential psychiatric disorder prevalence in polycystic ovarian syndrome. F S Rep 2023; 4:337-342. [PMID: 38204948 PMCID: PMC10774902 DOI: 10.1016/j.xfre.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 01/12/2024] Open
Abstract
Polycystic ovarian syndrome (PCOS) is the most common endocrine disorder in women, affecting up to 15% of reproductive-aged women. Polycystic ovarian syndrome is a heterogeneous disorder, both in the sense that many different factors may play a role in its manifestation and that multiple systems throughout the body can be affected. Polycystic ovarian syndrome has been linked to an increased prevalence of various psychiatric disorders, including depression and anxiety. Despite the socioeconomic effect that these disorders may have on patients with PCOS and society as a whole, this association is largely lacking in research. There are currently several theories regarding the link between PCOS and mental health. Some suggest that the overactive hypothalamic-pituitary-ovarian and hypothalamic-pituitary-adrenal axes in PCOS patients may alter the hormonal profile and contribute to the development of psychiatric disorders. Other studies speculate that abnormal levels of neurotransmitters and neuronal signaling may play a role. Recently, more research has begun to focus on the gut-brain axis, addressing the nutritional needs of PCOS patients. Studies show that dietary factors such as probiotics and micronutrient supplementation may significantly improve psychiatric symptoms in PCOS patients while helping regulate neurotransmitter levels in the body. In this review, we examine different theories regarding the association between PCOS and psychiatric disorders and point out different areas of research that are needed to broaden our understanding of this association.
Collapse
Affiliation(s)
- Karis I. Sarkisian
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Lananh Ho
- Department of Biomedical Engineering, University of Rochester, Rochester, New York
| | - Jane Yang
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Rachel Mandelbaum
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Frank Z. Stanczyk
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
9
|
Chen WH, Shi YC, Huang QY, Chen JM, Wang ZY, Lin S, Shi QY. Potential for NPY receptor-related therapies for polycystic ovary syndrome: an updated review. Hormones (Athens) 2023; 22:441-451. [PMID: 37452264 PMCID: PMC10449684 DOI: 10.1007/s42000-023-00460-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 06/13/2023] [Indexed: 07/18/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a complex endocrine disease that can cause female infertility and bring economic burden to families and to society. The clinical and/or biochemical manifestations include hyperandrogenism, persistent anovulation, and polycystic ovarian changes, often accompanied by insulin resistance and obesity. Although its pathogenesis is unclear, PCOS involves the abnormal regulation of the hypothalamic-pituitary-ovarian axis and the abnormal activation of GnRH neurons. Neuropeptide Y (NPY) is widely distributed in the arcuate nucleus of the hypothalamus and functions as the physiological integrator of two neuroendocrine systems, one governing feeding and the other controlling reproduction. In recent years, an increasing number of studies have focused on the improvement of the reproductive and metabolic status of PCOS through the therapeutic application of NPY and its receptors. In this review, we summarize the central and peripheral regulation of NPY and its receptors in the development of PCOS and discuss the potential for NPY receptor-related therapies for PCOS.
Collapse
Affiliation(s)
- Wei-Hong Chen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Yan-Chuan Shi
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Qiao-Yi Huang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Jia-Ming Chen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Zhi-Yi Wang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China.
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia.
| | - Qi-Yang Shi
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China.
| |
Collapse
|
10
|
Liang J, Gao Y, Feng Z, Zhang B, Na Z, Li D. Reactive oxygen species and ovarian diseases: Antioxidant strategies. Redox Biol 2023; 62:102659. [PMID: 36917900 PMCID: PMC10023995 DOI: 10.1016/j.redox.2023.102659] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/02/2023] [Accepted: 03/05/2023] [Indexed: 03/09/2023] Open
Abstract
Reactive oxygen species (ROS) are mainly produced in mitochondria and are involved in various physiological activities of the ovary through signaling and are critical for regulating the ovarian cycle. Notably, the imbalance between ROS generation and the antioxidant defense system contributes to the development of ovarian diseases. These contradictory effects have critical implications for potential antioxidant strategies that aim to scavenge excessive ROS. However, much remains to be learned about how ROS causes various ovarian diseases to the application of antioxidant therapy for ovarian diseases. Here, we review the mechanisms of ROS generation and maintenance of homeostasis in the ovary and its associated physiological effects. Additionally, we have highlighted the pathological mechanisms of ROS in ovarian diseases and potential antioxidant strategies for treatment.
Collapse
Affiliation(s)
- Junzhi Liang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yingzhuo Gao
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Ziyi Feng
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Bowen Zhang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Zhijing Na
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, 110004, China.
| | - Da Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, 110004, China; Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang, 110004, China.
| |
Collapse
|
11
|
Gurule S, Sustaita-Monroe J, Padmanabhan V, Cardoso R. Developmental programming of the neuroendocrine axis by steroid hormones: Insights from the sheep model of PCOS. Front Endocrinol (Lausanne) 2023; 14:1096187. [PMID: 36755919 PMCID: PMC9899912 DOI: 10.3389/fendo.2023.1096187] [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: 11/11/2022] [Accepted: 01/06/2023] [Indexed: 01/25/2023] Open
Abstract
The reproductive neuroendocrine system is a key target for the developmental programming effects of steroid hormones during early life. While gonadal steroids play an important role in controlling the physiological development of the neuroendocrine axis, human fetuses are susceptible to adverse programming due to exposure to endocrine disrupting chemicals with steroidal activity, inadvertent use of contraceptive pills during pregnancy, as well as from disease states that result in abnormal steroid production. Animal models provide an unparalleled resource to understand the effects of steroid hormones on the development of the neuroendocrine axis and their role on the developmental origins of health and disease. In female sheep, exposure to testosterone (T) excess during fetal development results in an array of reproductive disorders that recapitulate those seen in women with polycystic ovary syndrome (PCOS), including disrupted neuroendocrine feedback mechanisms, increased pituitary responsiveness to gonadotropin-releasing hormone (GnRH), luteinizing hormone (LH) hypersecretion, functional hyperandrogenism, multifollicular ovarian morphology, and premature reproductive failure. Similar to a large proportion of women with PCOS, these prenatally T-treated sheep also manifest insulin resistance and cardiovascular alterations, including hypertension. This review article focuses on the effects of prenatal androgens on the developmental programming of hypothalamic and pituitary alterations in the sheep model of PCOS phenotype, centering specifically on key neurons, neuropeptides, and regulatory pathways controlling GnRH and LH secretion. Insights obtained from the sheep model as well as other animal models of perinatal androgen excess can have important translational relevance to treat and prevent neuroendocrine dysfunction in women with PCOS and other fertility disorders.
Collapse
Affiliation(s)
- Sara Gurule
- Department of Animal Science, Texas A&M University, College Station, TX, United States
| | | | | | - Rodolfo Cardoso
- Department of Animal Science, Texas A&M University, College Station, TX, United States
| |
Collapse
|
12
|
Liu Y, Lin J, Shen X, Zhu Q, Kuang Y. Letrozole cotreatment improves the follicular output rate in high-body-mass-index women with polycystic ovary syndrome undergoing IVF treatment. Front Endocrinol (Lausanne) 2023; 14:1072170. [PMID: 36936138 PMCID: PMC10020617 DOI: 10.3389/fendo.2023.1072170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Women who have polycystic ovary syndrome (PCOS) with high body mass index (BMI) typically have an attenuated ovarian response and decreased follicular size, which are linked to unfavourable clinical outcomes following in vitro fertilization (IVF) therapy. The follicular output rate (FORT), a qualitative indicator of follicular response, seems to be positively linked to the clinical outcomes of IVF. Progestin-primed ovarian stimulation (PPOS) has become an alternative to gonadotropin-releasing hormone (GnRH) analogues to inhibit the premature luteinizing hormone (LH) surge. As letrozole (LE) shows promise in enhancing ovarian response, we compared PPOS with and without LE for PCOS in high BMI women with a focus on the FORT and associated clinical and pregnancy outcomes. METHODS For the recruited 1508 women, ten variables including AFC; age; basal sex hormone level; BMI; infertility type; period of infertility and number of previous IVF attempts were chosen in the propensity score matching (PSM) model to match 1374 women who taken the MPA+ hMG protocol with 134 women who received the MPA+ hMG+ LE treatment at a 1:1 ratio. FORT was selected as the primary outcome measure. The number of oocytes retrieved, viable embryos, hMG dosage, duration, oocyte maturity rate, fertilization rate, and implantation rate were established as secondary outcomes. RESULTS FORT was substantially elevated in the MPA+hMG+LE group compared with the MPA+hMG group (61% [35%, 86%] vs. 40% [25%, 60%], P <.001). Interestingly, the LE cotreatment group had a considerably lower mature oocyte rate despite having a similar number of mature oocytes and embryos recovered. The average hMG dosages and durations in the study group were similar to those in the control group. The implantation rate in the study group was numerically higher but without statistic significant than that in the control groups (43.15% (107/248) vs. 38.59% (115/298), OR 1.008, 95% CI 0.901-1.127; P >.05). CONCLUSION The effect of LE combined with PPOS on FORT is better than the effect of the standard PPOS treatment in women with PCOS and a high BMI, but there is no substantially beneficial impact on pregnancy outcomes or the cycle features of COS, including consumption of hMG.
Collapse
|
13
|
Donaldson NM, Prescott M, Ruddenklau A, Campbell RE, Desroziers E. Maternal androgen excess significantly impairs sexual behavior in male and female mouse offspring: Perspective for a biological origin of sexual dysfunction in PCOS. Front Endocrinol (Lausanne) 2023; 14:1116482. [PMID: 36875467 PMCID: PMC9975579 DOI: 10.3389/fendo.2023.1116482] [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: 12/05/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) is the most common infertility disorder worldwide, typically characterised by high circulating androgen levels, oligo- or anovulation, and polycystic ovarian morphology. Sexual dysfunction, including decreased sexual desire and increased sexual dissatisfaction, is also reported by women with PCOS. The origins of these sexual difficulties remain largely unidentified. To investigate potential biological origins of sexual dysfunction in PCOS patients, we asked whether the well-characterized, prenatally androgenized (PNA) mouse model of PCOS exhibits modified sex behaviours and whether central brain circuits associated with female sex behaviour are differentially regulated. As a male equivalent of PCOS is reported in the brothers of women with PCOS, we also investigated the impact of maternal androgen excess on the sex behaviour of male siblings. METHODS Adult male and female offspring of dams exposed to dihydrotestosterone (PNAM/PNAF) or an oil vehicle (VEH) from gestational days 16 to 18 were tested for a suite of sex-specific behaviours. RESULTS PNAM showed a reduction in their mounting capabilities, however, most of PNAM where able to reach ejaculation by the end of the test similar to the VEH control males. In contrast, PNAF exhibited a significant impairment in the female-typical sexual behaviour, lordosis. Interestingly, while neuronal activation was largely similar between PNAF and VEH females, impaired lordosis behaviour in PNAF was unexpectedly associated with decreased neuronal activation in the dorsomedial hypothalamic nucleus (DMH). CONCLUSION Taken together, these data link prenatal androgen exposure that drives a PCOS-like phenotype with altered sexual behaviours in both sexes.
Collapse
|
14
|
Huang C, Liu H, Yang W, Li Y, Wu B, Chen J, Yang Z, Liao C, Liu L, Zhang X. Distinct Gut Microbiota Structure and Function of Children with Idiopathic Central and Peripheral Precocious Puberty. Int J Endocrinol 2022; 2022:7175250. [PMID: 36405779 PMCID: PMC9668478 DOI: 10.1155/2022/7175250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 08/18/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022] Open
Abstract
Precocious puberty (PP) is one of the most common endocrine diseases in children, and the pathogenesis is currently unknown. Recent studies on the gut-brain axis have shown that there is a correlation between childhood endocrine diseases and the gut microbiota (GM). To explore the GM characteristics of children with different types of PP, we recruited 27 idiopathic central precocious puberty children (ICPP group), 18 peripheral precocious puberty children (PPP group), and 23 healthy children of the same age (HC group). Their stool samples were subjected to 16S rDNA sequencing. In this study, we found that the OTUs numbers, the annotated genera, and α-diversity of GM of the ICPP and PPP group were all significantly higher than that in the HC group (P < 0.05). The abundance of butyrate-producing bacteria Prevotella, Lachnospiracea incertae sedis, Roseburia, Ruminococcus, and Alistipes was significantly higher in the ICPP group and the PPP group, and Bacteroides and Faecalibacterium showed significantly higher abundance in the HC group. The GM symbiosis network showed that both Bacteroides and Faecalibacterium were negatively correlated with these butyrate-producing bacteria. The abundances of most significantly changed genera were gradually increased from HC to PPP, and to the ICPP group, while only Bacteroides was gradually decreased. After the prediction of the metabolic pathways of the GM, the cell motility, signal transduction, and environmental adaptation were significantly enriched in the ICPP and the PPP groups (P < 0.05), while the carbohydrate metabolism pathway was significantly lower (P < 0.001). Overall, this study showed that the GM composition and predicted functional pattern of children with ICPP and PPP are different from healthy children, and PPP may be a transitional stage between ICPP and HC children, which provide a theoretical basis for clinical intervention based on GM in the treatment of PP.
Collapse
Affiliation(s)
- Congfu Huang
- Department of Pediatrics, Longgang District Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Haiying Liu
- Department of Pediatrics, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Wei Yang
- Department of Pediatrics, The People's Hospital of Shenzhen Baoan District, Shenzhen, China
| | - Yinhu Li
- Department of Pediatrics, Longgang District Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Bin Wu
- Department of Pediatrics, Longgang District Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Junru Chen
- Department of Pediatrics, Longgang District Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Zhenyu Yang
- School of Statistics and Data Science, NanKai University, Tianjin, China
| | - Cuifang Liao
- Department of Computer Science, City University of Hong Kong, Hong Kong, China
| | - Limei Liu
- Department of Pediatrics, Longgang District Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Xiaowei Zhang
- Department of Obstetrics and Gynaecology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
15
|
Review of the effects of polycystic ovary syndrome on Cognition: Looking beyond the androgen hypothesis. Front Neuroendocrinol 2022; 67:101038. [PMID: 36154816 DOI: 10.1016/j.yfrne.2022.101038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/12/2022] [Accepted: 09/16/2022] [Indexed: 11/21/2022]
Abstract
Polycystic-ovary syndrome (PCOS) is the most common endocrine disorder affecting women of reproductive age, and many features associated with PCOS - such as elevated androgens, insulin resistance and inflammation - are known to affect cognition. However, effects of PCOS on cognition are not well-understood. Here we review the current literature on PCOS and cognition, note the extent of PCOS symptomatology studied in relation to cognitive outcomes, and identify key research gaps and common methodological concerns. Findings indicate a pattern of worse performance across cognitive domains and brain measures in women with PCOS relative to non-PCOS controls, as well as a lack of evidence for the common assumption that women with PCOS will have higher performance on tasks with a demonstrated male-advantage due to high testosterone levels. We suggest strategies for moving beyond the focus on elevated androgens, in favor of research practices that account for the nuances and heterogeneity of PCOS symptoms.
Collapse
|
16
|
Sivasankari R, Usha B. Reshaping the Gut Microbiota Through Lifestyle Interventions in Women with PCOS: A Review. Indian J Microbiol 2022; 62:351-363. [PMID: 35974920 PMCID: PMC9375820 DOI: 10.1007/s12088-022-01019-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/30/2022] [Indexed: 11/05/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine disorder evolving as a global threat to women's health. However, its multifactorial etiology causes difficulty in eliminating it. The interrelation between the gut microbiota and metabolic disorders has been trending recently, giving rise to new opportunities on the etiology and pathogenesis of PCOS. Lifestyle interventions such as healthy diet, physical exercises, and behavioral interventions such as regulation of stress and sleep cycles have been identified to improve the symptoms of PCOS across the endocrinological, metabolic and psychological scales and are recommended as the first line of treatment for PCOS. The impact of the unhealthy lifestyle factors on intestinal dysbiosis that cause PCOS is summarized in this review. This review also provides an insight on the therapeutic approaches that primarily target the gut microbiota and offers novel gut microflora-associated treatment strategies for PCOS. Further, this survey also highlights the need for the implementation of lifestyle management strategies and strongly recommends a healthy and stress-free lifestyle to promote gut health and manage PCOS.
Collapse
Affiliation(s)
- Ramadurai Sivasankari
- Department of Genetic Engineering, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Tamil Nadu 603 203 India
| | - Balasundaram Usha
- Department of Genetic Engineering, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Tamil Nadu 603 203 India
| |
Collapse
|
17
|
Sánchez-Garrido MA, García-Galiano D, Tena-Sempere M. Early programming of reproductive health and fertility: novel neuroendocrine mechanisms and implications in reproductive medicine. Hum Reprod Update 2022; 28:346-375. [PMID: 35187579 PMCID: PMC9071071 DOI: 10.1093/humupd/dmac005] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/29/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND According to the Developmental Origins of Health and Disease (DOHaD) hypothesis, environmental changes taking place during early maturational periods may alter normal development and predispose to the occurrence of diverse pathologies later in life. Indeed, adverse conditions during these critical developmental windows of high plasticity have been reported to alter the offspring developmental trajectory, causing permanent functional and structural perturbations that in the long term may enhance disease susceptibility. However, while solid evidence has documented that fluctuations in environmental factors, ranging from nutrient availability to chemicals, in early developmental stages (including the peri-conceptional period) have discernible programming effects that increase vulnerability to develop metabolic perturbations, the impact and eventual mechanisms involved, of such developmental alterations on the reproductive phenotype of offspring have received less attention. OBJECTIVE AND RATIONALE This review will summarize recent advances in basic and clinical research that support the concept of DOHaD in the context of the impact of nutritional and hormonal perturbations, occurring during the periconceptional, fetal and early postnatal stages, on different aspects of reproductive function in both sexes. Special emphasis will be given to the effects of early nutritional stress on the timing of puberty and adult gonadotropic function, and to address the underlying neuroendocrine pathways, with particular attention to involvement of the Kiss1 system in these reproductive perturbations. The implications of such phenomena in terms of reproductive medicine will also be considered. SEARCH METHODS A comprehensive MEDLINE search, using PubMed as main interface, of research articles and reviews, published mainly between 2006 and 2021, has been carried out. Search was implemented using multiple terms, focusing on clinical and preclinical data from DOHaD studies, addressing periconceptional, gestational and perinatal programming of reproduction. Selected studies addressing early programming of metabolic function have also been considered, when relevant. OUTCOMES A solid body of evidence, from clinical and preclinical studies, has documented the impact of nutritional and hormonal fluctuations during the periconceptional, prenatal and early postnatal periods on pubertal maturation, as well as adult gonadotropic function and fertility. Furthermore, exposure to environmental chemicals, such as bisphenol A, and maternal stress has been shown to negatively influence pubertal development and gonadotropic function in adulthood. The underlying neuroendocrine pathways and mechanisms involved have been also addressed, mainly by preclinical studies, which have identified an, as yet incomplete, array of molecular and neurohormonal effectors. These include, prominently, epigenetic regulatory mechanisms and the hypothalamic Kiss1 system, which likely contribute to the generation of reproductive alterations in conditions of early nutritional and/or metabolic stress. In addition to the Kiss1 system, other major hypothalamic regulators of GnRH neurosecretion, such as γ-aminobutyric acid and glutamate, may be targets of developmental programming. WIDER IMPLICATIONS This review addresses an underdeveloped area of reproductive biology and medicine that may help to improve our understanding of human reproductive disorders and stresses the importance, and eventual pathogenic impact, of early determinants of puberty, adult reproductive function and fertility.
Collapse
Affiliation(s)
- Miguel Angel Sánchez-Garrido
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
| | - David García-Galiano
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
18
|
Perdices-Lopez C, Avendaño MS, Barroso A, Gaytán F, Ruiz-Pino F, Vázquez MJ, Leon S, Song YB, Sobrino V, Heras V, Romero-Ruiz A, Roa J, Mayor F, Murga C, Pinilla L, Kaiser UB, Tena-Sempere M. Connecting nutritional deprivation and pubertal inhibition via GRK2-mediated repression of kisspeptin actions in GnRH neurons. Metabolism 2022; 129:155141. [PMID: 35074314 PMCID: PMC10283027 DOI: 10.1016/j.metabol.2022.155141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/31/2021] [Accepted: 01/14/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND Perturbations in the timing of puberty, with potential adverse consequences in later health, are increasingly common. The underlying neurohormonal mechanisms are unfolded, but nutritional alterations are key contributors. Efforts to unveil the basis of normal puberty and its metabolic control have focused on mechanisms controlling expression of Kiss1, the gene encoding the puberty-activating neuropeptide, kisspeptin. However, other regulatory phenomena remain ill-defined. Here, we address the putative role of the G protein-coupled-receptor kinase-2, GRK2, in GnRH neurons, as modulator of pubertal timing via repression of the actions of kisspeptin, in normal maturation and conditions of nutritional deficiency. METHODS Hypothalamic RNA and protein expression analyses were conducted in maturing female rats. Pharmacological studies involved central administration of GRK2 inhibitor, βARK1-I, and assessment of gonadotropin responses to kisspeptin or phenotypic and hormonal markers of puberty, under normal nutrition or early subnutrition in female rats. In addition, a mouse line with selective ablation of GRK2 in GnRH neurons, aka G-GRKO, was generated, in which hormonal responses to kisspeptin and puberty onset were monitored, in normal conditions and after nutritional deprivation. RESULTS Hypothalamic GRK2 expression increased along postnatal maturation in female rats, especially in the preoptic area, where most GnRH neurons reside, but decreased during the juvenile-to-pubertal transition. Blockade of GRK2 activity enhanced Ca+2 responses to kisspeptin in vitro, while central inhibition of GRK2 in vivo augmented gonadotropin responses to kisspeptin and advanced puberty onset. Postnatal undernutrition increased hypothalamic GRK2 expression and delayed puberty onset, the latter being partially reversed by central GRK2 inhibition. Conditional ablation of GRK2 in GnRH neurons enhanced gonadotropin responses to kisspeptin, accelerated puberty onset, and increased LH pulse frequency, while partially prevented the negative impact of subnutrition on pubertal timing and LH pulsatility in mice. CONCLUSIONS Our data disclose a novel pathway whereby GRK2 negatively regulates kisspeptin actions in GnRH neurons, as major regulatory mechanism for tuning pubertal timing in nutritionally-compromised conditions.
Collapse
Affiliation(s)
- Cecilia Perdices-Lopez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain; Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBER-OBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - María S Avendaño
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain; Hospital Universitario Reina Sofía, 14004 Córdoba, Spain.
| | - Alexia Barroso
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain; Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBER-OBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - Francisco Gaytán
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain; Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBER-OBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - Francisco Ruiz-Pino
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain; Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBER-OBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - Maria J Vázquez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain; Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBER-OBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - Silvia Leon
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain; Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Yong Bhum Song
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Veronica Sobrino
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain
| | - Violeta Heras
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain
| | - Antonio Romero-Ruiz
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain; Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Juan Roa
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain; Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBER-OBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - Federico Mayor
- Department of Molecular Biology, Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Universidad Autónoma de Madrid, 28029 Madrid, Spain; CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Cristina Murga
- Department of Molecular Biology, Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Universidad Autónoma de Madrid, 28029 Madrid, Spain; CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Leonor Pinilla
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain; Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBER-OBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain; Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBER-OBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain; CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain; Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine and Turku Center for Disease Modeling, University of Turku, Turku, Finland.
| |
Collapse
|
19
|
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: 1.0] [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
|
20
|
Chen C, Yu S, Yu W, Yan Z, Jin W, Si J, Li M, Cai R, Li D, Wang L, Chen Q, Kuang Y, Lyu Q, Long H. Luteinizing Hormone Suppression by Progestin-Primed Ovarian Stimulation Is Associated With Higher Implantation Rate for Patients With Polycystic Ovary Syndrome Who Underwent in vitro Fertilization/Intracytoplasmic Sperm Injection Cycles: Comparing With Short Protocol. Front Physiol 2022; 12:744968. [PMID: 35222055 PMCID: PMC8874211 DOI: 10.3389/fphys.2021.744968] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/30/2021] [Indexed: 12/18/2022] Open
Abstract
Background Many studies have demonstrated the positive clinical value of progestin-primed ovarian stimulation (PPOS) in patients with polycystic ovary syndrome (PCOS) who underwent assisted reproductive technology. However, the underlying factors contributing to this phenomenon remain unclear. We conducted a retrospective observational study to compare the clinical outcomes of women with PCOS who underwent PPOS or the short protocol to identify possible factors that influence the outcome. Methods This study included 304 patients who underwent PPOS and 152 patients who underwent short protocol from April 2014 to July 2019 after propensity-score matching. Human menopausal gonadotropin (hMG) dose, hormone profile, embryo development, and clinical outcomes of frozen-thawed embryo transfer (FET) cycles were compared. The primary outcome measure was the implantation rate. Logistic regression was performed to identify contributing factors, and receiver operating characteristic curve analysis was used to calculate the cutoff of luteinizing hormone (LH) difference ratio in clinical outcomes. Results Compared with the short protocol, PPOS resulted in a higher implantation rate (43.4% vs. 31.9%, P < 0.05), clinical pregnancy rate (61.8% vs. 47.4%, P < 0.05), and live birth rate (48.4% vs. 36.8%, P < 0.05). Similar fertilization, cleavage, and valid embryo rate per oocyte retrieved between groups were observed. The LH difference ratio was positively associated with implantation rate [P = 0.027, odds ratio (OR) = 1.861, 95% CI: 1.074–3.226]. The relationship between the LH difference ratio with clinical outcomes was confirmed by receiver operating characteristic curve analysis and comparisons among patients grouped by the LH difference ratio. Conclusion The implantation rate was associated with the LH difference ratio during ovary stimulation in patients with PCOS. Our results provide the explanation why PPOS shows the positive clinical outcomes for patients with PCOS.
Collapse
|
21
|
Sadeghi HM, Adeli I, Calina D, Docea AO, Mousavi T, Daniali M, Nikfar S, Tsatsakis A, Abdollahi M. Polycystic Ovary Syndrome: A Comprehensive Review of Pathogenesis, Management, and Drug Repurposing. Int J Mol Sci 2022; 23:ijms23020583. [PMID: 35054768 PMCID: PMC8775814 DOI: 10.3390/ijms23020583] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine-gynecology disorder affecting many women of childbearing age. Although a part of the involved mechanism in PCOS occurrence is discovered, the exact etiology and pathophysiology are not comprehensively understood yet. We searched PubMed for PCOS pathogenesis and management in this article and ClinicalTrials.gov for information on repurposed medications. All responsible factors behind PCOS were thoroughly evaluated. Furthermore, the complete information on PCOS commonly prescribed and repurposed medications is summarized through tables. Epigenetics, environmental toxicants, stress, diet as external factors, insulin resistance, hyperandrogenism, inflammation, oxidative stress, and obesity as internal factors were investigated. Lifestyle modifications and complementary and alternative medicines are preferred first-line therapy in many cases. Medications, including 3-hydroxy-3-methyl-3-glutaryl-coenzyme A (HMG-CoA) reductase inhibitors, thiazolidinediones, sodium-glucose cotransporter-2 inhibitors, dipeptidyl peptidase-4 inhibitors, glucose-like peptide-1 receptor agonists, mucolytic agents, and some supplements have supporting data for being repurposed in PCOS. Since there are few completed clinical trials with a low population and mostly without results on PCOS repurposed medications, it would be helpful to do further research and run well-designed clinical trials on this subject. Moreover, understanding more about PCOS would be beneficial to find new medications implying the effect via the novel discovered routes.
Collapse
Affiliation(s)
- Hosna Mohammad Sadeghi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 11369, Iran; (H.M.S.); (I.A.); (T.M.); (M.D.)
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 11369, Iran
| | - Ida Adeli
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 11369, Iran; (H.M.S.); (I.A.); (T.M.); (M.D.)
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 11369, Iran
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Correspondence: (D.C.); (M.A.)
| | - Anca Oana Docea
- Department of Toxicology, Faculty of Pharmacy, University of Medicine and Pharmacy, Petru Rares, 200349 Craiova, Romania;
| | - Taraneh Mousavi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 11369, Iran; (H.M.S.); (I.A.); (T.M.); (M.D.)
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 11369, Iran
| | - Marzieh Daniali
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 11369, Iran; (H.M.S.); (I.A.); (T.M.); (M.D.)
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 11369, Iran
| | - Shekoufeh Nikfar
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 11369, Iran;
- Personalized Medicine Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran 11369, Iran
- Evidence-Based Evaluation of Cost-Effectiveness and Clinical Outcomes Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 11369, Iran
| | - Aristidis Tsatsakis
- Department of Analytical and Forensic Medical Toxicology, Sechenov University, 119991 Moscow, Russia;
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
- Laboratory of Toxicology, Medical School, University of Crete, 70013 Heraklion, Greece
| | - Mohammad Abdollahi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 11369, Iran; (H.M.S.); (I.A.); (T.M.); (M.D.)
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 11369, Iran
- Correspondence: (D.C.); (M.A.)
| |
Collapse
|
22
|
Ma H, Ishida K, Xu C, Takahashi K, Li Y, Zhang C, Kang Q, Jia Y, Hu W, Matsumaru D, Nakanishi T, Hu J. Triphenyl phosphate delayed pubertal timing and induced decline of ovarian reserve in mice as an estrogen receptor antagonist. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 290:118096. [PMID: 34488164 DOI: 10.1016/j.envpol.2021.118096] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 06/13/2023]
Abstract
Although concerns have been raised about the adverse effects of triphenyl phosphate (TPhP) on female fertility, its risk to ovarian functioning remains unknown. In this study, female C57BL/6 mice at postnatal day 21 were exposed on a daily basis to TPhP dose of 2, 10, and 50 mg/kg for 40 days. A significant delay in pubertal timing was observed in the mice exposed to 50 mg/kg of TPhP. An estrogen-responsive reporter transgenic mice assay demonstrated that TPhP significantly downregulated the estrogen receptor (ER) signaling by 45.1% in the whole body in the 50 mg/kg group, and by 14.7-43.7% in the uterus for all exposure groups compared with the control. This strong antagonistic activity of TPhP toward ER explained the delay in pubertal timing. A significant reduction in the number of follicles in all stages was observed in mice after being exposed to TPhP for 40 days at concentrations of 10 and 50 mg/kg, resulting in a decline of the ovarian reserve. The elevation of the follicle-stimulating hormone concentration may have contributed to this phenomenon, as controlled by the antagonistic activity of TPhP toward ER in the brain. The toxic effects of TPhP on ovarian functioning highlight this chemical as a potential risk factor for female fertility.
Collapse
Affiliation(s)
- Haojia Ma
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Keishi Ishida
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Chenke Xu
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Kyosuke Takahashi
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Yu Li
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Chenhao Zhang
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Qiyue Kang
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Yingting Jia
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Wenxin Hu
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China
| | - Daisuke Matsumaru
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Tsuyoshi Nakanishi
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, Gifu, 501-1196, Japan
| | - Jianying Hu
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
23
|
Watanabe Y, Prescott M, Campbell RE, Jasoni CL. Prenatal androgenization causes expression changes of progesterone and androgen receptor mRNAs in the arcuate nucleus of female mice across development. J Neuroendocrinol 2021; 33:e13058. [PMID: 34748236 DOI: 10.1111/jne.13058] [Citation(s) in RCA: 3] [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: 02/26/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 11/27/2022]
Abstract
Prenatal exposure to excess androgens is associated with the development of polycystic ovary syndrome (PCOS). In prenatally androgenised (PNA) mice, a model of PCOS, progesterone receptor (PR) protein expression is reduced in arcuate nucleus (ARC) GABA neurons. This suggests a mechanism for PCOS-related impaired steroid hormone feedback and implicates androgen excess with respect to inducing transcriptional repression of the PR-encoding gene Pgr in the ARC. However, the androgen sensitivity of ARC neurons and the relative gene expression of PRs over development and following prenatal androgen exposure remain unknown. Here, we used a quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR) of microdissected ARC to determine the relative androgen receptor (Ar) and progesterone receptor (Pgr) gene expression in PNA and control mice at five developmental timepoints. In a two-way analysis of variance, none of the genes examined showed expression changes with a statistically significant interaction between treatment and age, although PgrA showed a borderline interaction. For all genes, there was a statistically significant main effect of age on expression levels, reflecting a general increase in expression with increasing age, regardless of treatment. For PgrB and Ar, there was a statistically significant main effect of treatment, indicating a change in expression following PNA (increased for PgrB and decreased for Ar), regardless of age. For PgrA, there was a borderline main effect of treatment, suggesting a possible change in expression following PNA, regardless of age. PgrAB gene expression changes showed no significant main effect of treatment. We additionally examined androgen and progesterone responsiveness specifically in P60 ARC GABA neurons using RNAScope® (Advanced Cell Diagnostics, Inc.) in situ hybridization. This analysis revealed that Pgr and Ar were expressed in the majority of ARC GABA neurons in normal adult females. However, our RNAScope® analysis did not show significant changes in Pgr or Ar expression within ARC GABA neurons following PNA. Lastly, because GABA drive to gonadotropin-releasing hormone neurons is increased in PNA, we hypothesised that PNA mice would show increased expression of glutamic acid decarboxylase (GAD), the rate-limiting enzyme in GABA production. However, the RT-qPCR showed that the expression of GAD encoding genes (Gad1 and Gad2) was unchanged in adult PNA mice compared to controls. Our findings indicate that PNA treatment can impact Pgr and Ar mRNA expression in adulthood. This may reflect altered circulating steroid hormones in PNA mice or PNA-induced epigenetic changes in the regulation of Pgr and Ar gene expression in ARC neurons.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Arcuate Nucleus of Hypothalamus/growth & development
- Arcuate Nucleus of Hypothalamus/metabolism
- Embryo, Mammalian
- Female
- Gene Expression Regulation, Developmental
- Growth and Development/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Pregnancy
- Prenatal Exposure Delayed Effects/genetics
- Prenatal Exposure Delayed Effects/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Virilism/embryology
- Virilism/genetics
- Virilism/metabolism
Collapse
Affiliation(s)
- Yugo Watanabe
- Department of Anatomy, Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Melanie Prescott
- Department of Physiology, Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Rebecca E Campbell
- Department of Physiology, Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Christine L Jasoni
- Department of Anatomy, Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| |
Collapse
|
24
|
Jin L, Yu J, Chen Y, Pang H, Sheng J, Huang H. Polycystic Ovary Syndrome and Risk of Five Common Psychiatric Disorders Among European Women: A Two-Sample Mendelian Randomization Study. Front Genet 2021; 12:689897. [PMID: 34211505 PMCID: PMC8239353 DOI: 10.3389/fgene.2021.689897] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/25/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Observational studies have implied an association between polycystic ovary syndrome (PCOS) and psychiatric disorders. Here we examined whether PCOS might contribute causally to such disorders, focusing on anxiety disorder (AD), bipolar disorder (BIP), major depression disorder (MDD), obsessive compulsive disorder (OCD), and schizophrenia (SCZ). Methods: Causality was explored using two-sample Mendelian randomization (MR) with genetic variants as instrumental variables. The genetic variants were from summary data of genome-wide association studies in European populations. First, potential causal effects of PCOS on each psychiatric disorder were evaluated, and then potential reverse causality was also assessed once PCOS was found to be causally associated with any psychiatric disorder. Causal effects were explored using inverse variance weighting, MR-Egger analysis, simulation extrapolation, and weighted median analysis. Results: Genetically predicted PCOS was positively associated with OCD based on inverse variance weighting (OR 1.339, 95% CI 1.083–1.657, p = 0.007), simulation extrapolation (OR 1.382, 95% CI 1.149–1.662, p = 0.009) and weighted median analysis (OR 1.493, 95% CI 1.145–1.946, p = 0.003). However, genetically predicted OCD was not associated with PCOS. Genetically predicted PCOS did not exert causal effects on AD, BIP, MDD, or SCZ. Conclusions: In European populations, PCOS may be a causal factor in OCD, but not AD, BIP, MDD, or SCZ.
Collapse
Affiliation(s)
- Luyang Jin
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Reproductive Genetics, Ministry of Education, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jia'en Yu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Reproductive Genetics, Ministry of Education, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuxiao Chen
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haiyan Pang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Reproductive Genetics, Ministry of Education, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianzhong Sheng
- Key Laboratory of Reproductive Genetics, Ministry of Education, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Hefeng Huang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Reproductive Genetics, Ministry of Education, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
25
|
Evaluation of the relationship between serum ferritin and insulin resistance and visceral adiposity index (VAI) in women with polycystic ovary syndrome. Eat Weight Disord 2021; 26:1581-1593. [PMID: 32772321 DOI: 10.1007/s40519-020-00980-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 07/30/2020] [Indexed: 01/08/2023] Open
Abstract
PURPOSE There is a relationship between polycystic ovary syndrome (PCOS) and adipose tissue dysfunction (ADD), but this relationship is not clear. It has been recently shown that iron accumulation in adipose tissue is among the causes of adipose tissue dysfunction. Data on adipose tissue dysfunction in women with PCOS are insufficient. In this study, we aimed to evaluate the relationship between serum ferritin levels (iron accumulation biomarker) and visceral adiposity index (an indicator of adipose tissue dysfunction). METHODS The study is a case-control study. Women with diagnosed PCOS with 2003 Rotterdam Diagnostic Criteria (n = 40) were compared with non-PCOS group (n = 40). In this study, the cholesterol ratios, the homeostatic model evaluation index for insulin resistance (HOMA-IR) and the quantitative insulin sensitivity control index were calculated using biochemical parameters, and the visceral adiposity index (VAI) and the lipid accumulation product (LAP) were calculated using both anthropometric and biochemical parameters. In this study, insulin resistance was evaluated by HOMA-IR and adipose tissue dysfunction was evaluated by VAI index. RESULTS According to the results of this study, women with PCOS have a worse metabolic status than women without PCOS. However, this has been shown only in overweight and obese women, not in women with normal weight. CONCLUSION As a result, the presence of obesity in women with PCOS exacerbates metabolic status. LEVEL OF EVIDENCE Level V, cross-sectional descriptive study.
Collapse
|
26
|
Zhou Y, Lan H, Dong Z, Cao W, Zeng Z, Song JL. Dietary proanthocyanidins alleviated ovarian fibrosis in letrozole-induced polycystic ovary syndrome in rats. J Food Biochem 2021; 45:e13723. [PMID: 33818798 DOI: 10.1111/jfbc.13723] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/12/2021] [Accepted: 03/26/2021] [Indexed: 11/30/2022]
Abstract
This study investigated the effects of proanthocyanidins (PCs) on ovarian fibrosis in letrozole-induced polycystic ovary syndrome (PCOS) in rats. The administration of PCs effectively reduced the body weight (BW) and relative ovarian weight in PCOS rats. ELISA results revealed that PCs significantly reduced the level of serum T, LH, LH/FSH in the PCOS group. In addition, qRT-PCR results revealed that treatment with PCs significantly increased the main antioxidant enzymes (Cat, Sod2, Gpx3, Mgst1, Gsta4, Sod1 and Prdx3) in PCOS rats. Also, the expression analysis of proteins by Western blotting revealed that PCs significantly decreased the level of TGF-βR1, p-Smad3, p-Smad2 and Smad4 and reversed the downregulation of Smad7 in PCOS rats. The study suggested that PCs improved ovarian fibrosis in PCOS rats by regulating the serum hormone level, inhibiting oxidative stress and suppressing the activation of the TGF-β1/Smads signaling pathway. PRACTICAL APPLICATIONS: Currently, plant extracts are being widely used to treat female reproductive and metabolic disorders. Particularly, proanthocyanidins (PCs), the well-known natural polyphenolic compounds, which are a significant source of antioxidants present in many colored fruits, are consumed as fruits as well as a dietary supplement to prevent many disorders. Recent pharmacological studies have reported that PCs have many health beneficial properties, such as antioxidant activity, improving cholesterol homeostasis, blood lipid regulatory properties, microcirculation improvement effect, antitumor activity and anti-aging activity. Despite these properties of PCs, the antifibrosis effect of PCs has not been studied to date. The main purpose of this study was to research the role and the mechanisms of PCs in ovarian fibrosis in PCOS rats.
Collapse
Affiliation(s)
- Yanyuan Zhou
- Department of Analytical Chemistry & Drug Analysis, School of Pharmacy, Guilin Medical University, Guilin, China
| | - Huan Lan
- Department of Analytical Chemistry & Drug Analysis, School of Pharmacy, Guilin Medical University, Guilin, China
| | - Zhewen Dong
- Department of Analytical Chemistry & Drug Analysis, School of Pharmacy, Guilin Medical University, Guilin, China
| | - Wenjing Cao
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, China
| | - Zhen Zeng
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, China.,Department of Maternal and Child Health, XiangYa School of Public Health, Central South University, Changsha, China
| | - Jia-Le Song
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, China.,Department of Clinical Nutrition, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
27
|
Landers RSM, Padmanabhan V, Cardoso RC. Developmental programming: gestational testosterone excess disrupts LH secretion in the female sheep fetus. Reprod Biol Endocrinol 2020; 18:106. [PMID: 33158439 PMCID: PMC7648305 DOI: 10.1186/s12958-020-00667-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/30/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Prenatal testosterone (T) excess results in reproductive and metabolic perturbations in female sheep that closely recapitulate those seen in women with polycystic ovary syndrome (PCOS). At the neuroendocrine level, prenatal T-treated sheep manifest increased pituitary sensitivity to GnRH and subsequent LH hypersecretion. In this study, we investigated the early effects of gestational T-treatment on LH secretion and pituitary function in the female sheep fetus. Additionally, because prenatal T effects can be mediated via the androgen receptor or due to changes in insulin homeostasis, prenatal co-treatment with an androgen antagonist (flutamide) or an insulin sensitizer (rosiglitazone) were tested. METHODS Pregnant sheep were treated from gestational day (GD) 30 to 90 with either: 1) vehicle (control); 2) T-propionate (~ 1.2 mg/kg); 3) T-propionate and flutamide (15 mg/kg/day); and 4) T-propionate and rosiglitazone (8 mg/day). At GD 90, LH concentrations were determined in the uterine artery (maternal) and umbilical artery (fetal), and female fetuses were euthanized. Pituitary glands were collected, weighed, and protein level of several key regulators of LH secretion was determined. RESULTS Fetal pituitary weight was significantly reduced by prenatal T-treatment. Flutamide completely prevented the reduction in pituitary weight, while rosiglitazone only partially prevented this reduction. Prenatal T markedly reduced fetal LH concentrations and flutamide co-treatment partially restored LH to control levels. Prenatal T resulted in a marked reduction in LH-β protein level, which was associated with a reduction in GnRH receptor and estrogen receptor-α levels and an increase in androgen receptor. With the exception of androgen receptor, flutamide co-treatment completely prevented these alterations in the fetal pituitary, while rosiglitazone largely failed to prevent these changes. Prenatal T-treatment did not alter the protein levels of insulin receptor-β and activation (phosphorylation) of the insulin signaling pathways. CONCLUSIONS These findings demonstrate that prenatal T-treatment results in reduced fetal LH secretion, reduced fetal pituitary weight, and altered protein levels of several regulators of gonadotropin secretion. The observations that flutamide co-treatment prevented these changes suggest that programming during fetal development likely occurs via direct androgen actions.
Collapse
Affiliation(s)
- Renata S M Landers
- Department of Animal Science, Texas A&M University, 2471 TAMU, College Station, TX, 77843-2471, USA
| | | | - Rodolfo C Cardoso
- Department of Animal Science, Texas A&M University, 2471 TAMU, College Station, TX, 77843-2471, USA.
| |
Collapse
|
28
|
Saad MA, Eltarzy MA, Abdel Salam RM, Ahmed MAE. Liraglutide mends cognitive impairment by averting Notch signaling pathway overexpression in a rat model of polycystic ovary syndrome. Life Sci 2020; 265:118731. [PMID: 33160995 DOI: 10.1016/j.lfs.2020.118731] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/23/2020] [Accepted: 11/04/2020] [Indexed: 01/06/2023]
Abstract
AIMS Polycystic ovary syndrome (PCOS), the rifest endocrine disorder in women, is involved in disrupting many metabolic processes. However, the impact of PCOS on cognitive deficits is still uncertain. Recently, Notch signaling pathway was identified as a key modifier in regulating the pathological process in the ovary and various neurodegenerative disorders. Liraglutide has favourable neuroprotective effects that may protect against the possible cognitive dysfunction in PCOS. MAIN METHODS PCOS was induced in rats by administrating Letrozole orally for 21 successive days. Then, Liraglutide (LIR) was administered intraperitoneally for 30 days. Memory was examined using Y-maze, novel object recognition (NOR), and Morris water maze (MWM) tests. Western blotting, enzyme immunoassay, and quantitative real-time PCR were used to examine Notch signaling downstream targets, as well as assessing the expression of the components of various pathways cross talked with Notch signaling in memory impairment. Furthermore, histopathological examination was performed to examine neuronal changes. KEY FINDINGS Notch signaling was overexpressed in PCOS rats, which increased Aβ aggregation, apoptosis, and neuroinflammation. Additionally, histopathological examination showed neuronal degeneration, which was marked by diminished acetylcholine levels in the PCOS rats' hippocampi. Finally, serum levels of insulin and testosterone were elevated while estradiol was reduced. Treatment with LIR repaired Notch signaling-attributed changes and improved the PCOS-induced memory impairment in rats. SIGNIFICANCE The obtained findings confirm that Notch signaling activation in the hippocampus of rats impairs cognitive functions in PCOS, which is mitigated by LIR. Therefore, LIR may offer a novel therapeutic intervention to impede PCOS-induced dementia.
Collapse
Affiliation(s)
- Muhammed A Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt; School of Pharmacy, NewGiza University, Giza, Egypt
| | - Muhammad A Eltarzy
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), 6th of October City, Giza, Egypt
| | - Rania M Abdel Salam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt; School of Pharmacy, NewGiza University, Giza, Egypt
| | - Maha A E Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), 6th of October City, Giza, Egypt.
| |
Collapse
|
29
|
Gao Z, Ma X, Liu J, Ge Y, Wang L, Fu P, Liu Z, Yao R, Yan X. Troxerutin protects against DHT-induced polycystic ovary syndrome in rats. J Ovarian Res 2020; 13:106. [PMID: 32921318 PMCID: PMC7489018 DOI: 10.1186/s13048-020-00701-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 08/04/2020] [Indexed: 01/17/2023] Open
Abstract
The exact pathogenesis of polycystic ovary syndrome (PCOS), the most common neuroendocrine disorder in women of reproductive age, has not been fully elucidated. Recent studies suggested that chronic inflammation and neurotransmitter disorder involved in the progress of PCOS. Troxerutin, a natural flavonoid, was reported to possess neuroprotective effect in several disease models by inhibiting inflammation or enhancing neurotrophic factor. In this study, we investigated the possible protective effect and mechanism of troxerutin in a dihydrotestosterone (DHT)-induced rat model of PCOS. The PCOS rat models were treated with troxerutin at a dose of 150 mg/kg or 300 mg/kg for up to 4 weeks. Results showed that 300 mg/kg troxerutin significantly decreased the body weight gain and improved the pathological changes of ovary induced by DHT. Meanwhile, the elevated gonadotrophin-releasing hormone (GnRH), gonadotrophin and testosterone in the serum of PCOS rats were reduced with the treatment of troxerutin. The expression of kisspeptin and NKB in arcuate nucleus and their receptors kiss1r and NK3r in GnRH positive neurons of median eminence were markedly decreased in troxerutin-treated rats. Of note, the GnRH inhibitory regulator GABA and stimulatory regulator glutamate were also restored to the normal level by troxerutin. The present study indicated that troxerutin may exhibit a protective effect in PCOS rat model via regulating neurotransmitter release.
Collapse
Affiliation(s)
- Zixuan Gao
- Department of Histology and Embryology, Xuzhou Medical University, Xuzhou, 221009, PR China.,Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221009, PR China
| | - Xiaochen Ma
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221009, PR China.,Clinical Center of Reproductive Medicine, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221000, PR China
| | - Jing Liu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221009, PR China
| | - Yuhang Ge
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221009, PR China.,Department of Human Anatomy, Xuzhou Medical University, Xuzhou, 221009, PR China
| | - Lei Wang
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221009, PR China
| | - Ping Fu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221009, PR China
| | - Zhian Liu
- Department of Human Anatomy, Xuzhou Medical University, Xuzhou, 221009, PR China
| | - Ruiqin Yao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221009, PR China.
| | - Xiaonan Yan
- Clinical Center of Reproductive Medicine, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221000, PR China.
| |
Collapse
|
30
|
Romero-Ruiz A, Skorupskaite K, Gaytan F, Torres E, Perdices-Lopez C, Mannaerts BM, Qi S, Leon S, Manfredi-Lozano M, Lopez-Rodriguez C, Avendaño MS, Sanchez-Garrido MA, Vazquez MJ, Pinilla L, van Duin M, Kohout TA, Anderson RA, Tena-Sempere M. Kisspeptin treatment induces gonadotropic responses and rescues ovulation in a subset of preclinical models and women with polycystic ovary syndrome. Hum Reprod 2020; 34:2495-2512. [PMID: 31820802 PMCID: PMC6936723 DOI: 10.1093/humrep/dez205] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/25/2019] [Indexed: 12/14/2022] Open
Abstract
STUDY QUESTION Can kisspeptin treatment induce gonadotrophin responses and ovulation in preclinical models and anovulatory women with polycystic ovary syndrome (PCOS)? SUMMARY ANSWER Kisspeptin administration in some anovulatory preclinical models and women with PCOS can stimulate reproductive hormone secretion and ovulation, albeit with incomplete efficacy. WHAT IS KNOWN ALREADY PCOS is a prevalent, heterogeneous endocrine disorder, characterized by ovulatory dysfunction, hyperandrogenism and deregulated gonadotrophin secretion, in need of improved therapeutic options. Kisspeptins (encoded by Kiss1) are master regulators of the reproductive axis, acting mainly at GnRH neurons, with kisspeptins being an essential drive for gonadotrophin-driven ovarian follicular maturation and ovulation. Altered Kiss1 expression has been found in rodent models of PCOS, although the eventual pathophysiological role of kisspeptins in PCOS remains unknown. STUDY DESIGN, SIZE, DURATION Gonadotrophin and ovarian/ovulatory responses to kisspeptin-54 (KP-54) were evaluated in three preclinical models of PCOS, generated by androgen exposures at different developmental windows, and a pilot exploratory cohort of anovulatory women with PCOS. PARTICIPANTS/MATERIALS, SETTING, METHODS Three models of PCOS were generated by exposure of female rats to androgens at different periods of development: PNA (prenatal androgenization; N = 20), NeNA (neonatal androgenization; N = 20) and PWA (post-weaning androgenization; N = 20). At adulthood (postnatal day 100), rats were subjected to daily treatments with a bolus of KP-54 (100 μg/kg, s.c.) or vehicle for 11 days (N = 10 per model and treatment). On Days 1, 4, 7 and 11, LH and FSH responses were assessed at different time-points within 4 h after KP-54 injection, while ovarian responses, in terms of follicular maturation and ovulation, were measured at the end of the treatment. In addition, hormonal (gonadotrophin, estrogen and inhibin B) and ovulatory responses to repeated KP-54 administration, at doses of 6.4-12.8 nmol/kg, s.c. bd for 21 days, were evaluated in a pilot cohort of anovulatory women (N = 12) diagnosed with PCOS, according to the Rotterdam criteria. MAIN RESULTS AND THE ROLE OF CHANCE Deregulated reproductive indices were detected in all PCOS models: PNA, NeNA and PWA. Yet, anovulation was observed only in NeNA and PWA rats. However, while anovulatory NeNA rats displayed significant LH and FSH responses to KP-54 (P < 0.05), which rescued ovulation, PWA rats showed blunted LH secretion after repeated KP-54 injection and failed to ovulate. In women with PCOS, KP-54 resulted in a small rise in LH (P < 0.05), with an equivalent elevation in serum estradiol levels (P < 0.05). Two women showed growth of a dominant follicle with subsequent ovulation, one woman displayed follicle growth but not ovulation and desensitization was observed in another patient. No follicular response was detected in the other women. LIMITATIONS, REASONS FOR CAUTION While three different preclinical PCOS models were used in order to capture the heterogeneity of clinical presentations of the syndrome, it must be noted that rat models recapitulate many but not all the features of this condition. Additionally, our pilot study was intended as proof of principle, and the number of participants is low, but the convergent findings in preclinical and clinical studies reinforce the validity of our conclusions. WIDER IMPLICATIONS OF THE FINDINGS Our first-in-rodent and -human studies demonstrate that KP-54 administration in anovulatory preclinical models and women with PCOS can stimulate reproductive hormone secretion and ovulation, albeit with incomplete efficacy. As our rat models likely reflect the diversity of PCOS phenotypes, our results argue for the need of personalized management of anovulatory dysfunction in women with PCOS, some of whom may benefit from kisspeptin-based treatments. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by research agreements between Ferring Research Institute and the Universities of Cordoba and Edinburgh. K.S. was supported by the Wellcome Trust Scottish Translational Medicine and Therapeutics Initiative (STMTI). Some of this work was undertaken in the MRC Centre for Reproductive Health which is funded by the MRC Centre grant MR/N022556/1. M.T.-S. is a member of CIBER Fisiopatología de la Obesidad y Nutrición, which is an initiative of Instituto de Salud Carlos III. Dr Mannaerts is an employee of Ferring International PharmaScience Center (Copenhagen, Denmark), and Drs Qi, van Duin and Kohout are employees of the Ferring Research Institute (San Diego, USA). Dr Anderson and Dr Tena-Sempere were recipients of a grant support from the Ferring Research Institute, and Dr Anderson has undertaken consultancy work and received speaker fees outside this study from Merck, IBSA, Roche Diagnostics, NeRRe Therapeutics and Sojournix Inc. Dr Skorupskaite was supported by the Wellcome Trust through the Scottish Translational Medicine and Therapeutics Initiative 102419/Z/13/A. The other authors have no competing interest.
Collapse
Affiliation(s)
- A Romero-Ruiz
- Department of Cell Biology, Physiology & Immunology, University of Córdoba, 14004 Córdoba, Spain.,Maimónides Institute of Biomedical Research of Córdoba (IMIBIC)/Reina Sofia University Hospital, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - K Skorupskaite
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - F Gaytan
- Department of Cell Biology, Physiology & Immunology, University of Córdoba, 14004 Córdoba, Spain.,Maimónides Institute of Biomedical Research of Córdoba (IMIBIC)/Reina Sofia University Hospital, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - E Torres
- Department of Cell Biology, Physiology & Immunology, University of Córdoba, 14004 Córdoba, Spain.,Maimónides Institute of Biomedical Research of Córdoba (IMIBIC)/Reina Sofia University Hospital, 14004 Córdoba, Spain
| | - C Perdices-Lopez
- Department of Cell Biology, Physiology & Immunology, University of Córdoba, 14004 Córdoba, Spain.,Maimónides Institute of Biomedical Research of Córdoba (IMIBIC)/Reina Sofia University Hospital, 14004 Córdoba, Spain
| | - B M Mannaerts
- Ferring International PharmaScience Center, Copenhagen, Denmark
| | - S Qi
- Ferring Research Institute, San Diego, CA 92121, USA
| | - S Leon
- Department of Cell Biology, Physiology & Immunology, University of Córdoba, 14004 Córdoba, Spain.,Maimónides Institute of Biomedical Research of Córdoba (IMIBIC)/Reina Sofia University Hospital, 14004 Córdoba, Spain
| | - M Manfredi-Lozano
- Department of Cell Biology, Physiology & Immunology, University of Córdoba, 14004 Córdoba, Spain.,Maimónides Institute of Biomedical Research of Córdoba (IMIBIC)/Reina Sofia University Hospital, 14004 Córdoba, Spain
| | - C Lopez-Rodriguez
- Department of Cell Biology, Physiology & Immunology, University of Córdoba, 14004 Córdoba, Spain.,Maimónides Institute of Biomedical Research of Córdoba (IMIBIC)/Reina Sofia University Hospital, 14004 Córdoba, Spain
| | - M S Avendaño
- Department of Cell Biology, Physiology & Immunology, University of Córdoba, 14004 Córdoba, Spain.,Maimónides Institute of Biomedical Research of Córdoba (IMIBIC)/Reina Sofia University Hospital, 14004 Córdoba, Spain
| | - M A Sanchez-Garrido
- Department of Cell Biology, Physiology & Immunology, University of Córdoba, 14004 Córdoba, Spain.,Maimónides Institute of Biomedical Research of Córdoba (IMIBIC)/Reina Sofia University Hospital, 14004 Córdoba, Spain
| | - M J Vazquez
- Department of Cell Biology, Physiology & Immunology, University of Córdoba, 14004 Córdoba, Spain.,Maimónides Institute of Biomedical Research of Córdoba (IMIBIC)/Reina Sofia University Hospital, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - L Pinilla
- Department of Cell Biology, Physiology & Immunology, University of Córdoba, 14004 Córdoba, Spain.,Maimónides Institute of Biomedical Research of Córdoba (IMIBIC)/Reina Sofia University Hospital, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - M van Duin
- Ferring Research Institute, San Diego, CA 92121, USA
| | - T A Kohout
- Ferring Research Institute, San Diego, CA 92121, USA
| | - R A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - M Tena-Sempere
- Department of Cell Biology, Physiology & Immunology, University of Córdoba, 14004 Córdoba, Spain.,Maimónides Institute of Biomedical Research of Córdoba (IMIBIC)/Reina Sofia University Hospital, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 14004 Córdoba, Spain.,FiDiPro Program, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland
| |
Collapse
|
31
|
Wang Y, Guo X, Xu W, Cai J, Zhang Y, Wu C, Li S, Sun Y, Liu W, Tao T. Role of Androgen in Liver Fat Content in Women: Metabolically Advantageous or Disadvantageous? Endocr Pract 2020; 26:1003-1016. [PMID: 33471689 DOI: 10.4158/ep-2019-0407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 04/29/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Androgens have a controversial effect on liver fat content (LFC) in androgen-excess females and androgen-deficient males. Polycystic ovarian syndrome (PCOS) is often associated with hyperandrogenism and nonalcoholic fatty liver disease. The aim of this study was to explore the association between hyperandrogenemia and increased liver fat content in women with PCOS, independent of other metabolic parameters. METHODS This case series study included 501 women with PCOS and 112 aged-matched controls in the outpatient department of a tertiary hospital. Anthropometric measurements, hepatic and renal function, glucose and lipid metabolism parameters, and sex hormones were examined in these women. LFC was measured by quantitative ultrasonography. RESULTS Women with hyperandrogenism (P<.001), an oligomenorrhoea/anovulation phenotype (P = .0064), and a diagnosis of PCOS (P<.001) had higher LFC. Androgen level is an important factor among the 9 independent risk factors of LFC (P = .0239) and may have a dimorphic impact on LFC. In all women, when the free androgen index (FAI) was less than 41.94, LFC increased with the elevated FAI; when the FAI was greater than 41.94, LFC decreased with the elevated FAI (P<.001). In women with PCOS, receiver operating characteristic curve analysis demonstrated that LFC could at least partially predict impaired glucose regulation, impaired lipid metabolism, and insulin resistance (P<.0001 for all). CONCLUSION Androgen level is associated with LFC in dimorphic directions. LFC may be a predictive factor of insulin resistance, impaired glucose regulation, and impaired lipid metabolism in women with PCOS.
Collapse
Affiliation(s)
- Yuying Wang
- From the Department of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiaojing Guo
- the Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Wendi Xu
- From the Department of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jie Cai
- From the Department of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yi Zhang
- From the Department of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Chunhua Wu
- the Division of Ultrasonography, Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shengxian Li
- From the Department of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yun Sun
- the Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Liu
- From the Department of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China; the Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China..
| | - Tao Tao
- From the Department of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
32
|
Penix J, DeFazio RA, Dulka EA, Schnell S, Moenter SM. Firing patterns of gonadotropin-releasing hormone neurons are sculpted by their biologic state. ROYAL SOCIETY OPEN SCIENCE 2020; 7:201040. [PMID: 32968535 PMCID: PMC7481724 DOI: 10.1098/rsos.201040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/13/2020] [Indexed: 06/11/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons form the final pathway for the central neuronal control of fertility. GnRH is released in pulses that vary in frequency in females, helping drive hormonal changes of the reproductive cycle. In the common fertility disorder polycystic ovary syndrome (PCOS), persistent high-frequency hormone release is associated with disrupted cycles. We investigated long- and short-term action potential patterns of GnRH neurons in brain slices before and after puberty in female control and prenatally androgenized (PNA) mice, which mimic aspects of PCOS. A Monte Carlo (MC) approach was used to randomize action potential interval order. Dataset distributions were analysed to assess (i) if organization persists in GnRH neuron activity in vitro, and (ii) to determine if any organization changes with development and/or PNA treatment. GnRH neurons in adult control, but not PNA, mice produce long-term patterns different from MC distributions. Short-term patterns differ from MC distributions before puberty but become absorbed into the distributions with maturation, and the distributions narrow. These maturational changes are blunted by PNA treatment. Firing patterns of GnRH neurons in brain slices thus maintain organization dictated at least in part by the biologic status of the source and are disrupted in models of disease.
Collapse
Affiliation(s)
- Jonathon Penix
- Departments of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - R. Anthony DeFazio
- Departments of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eden A. Dulka
- Departments of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Santiago Schnell
- Departments of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
- Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Suzanne M. Moenter
- Departments of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
- Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
33
|
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: 10.2] [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
|
34
|
Mintziori G, Nigdelis MP, Mathew H, Mousiolis A, Goulis DG, Mantzoros CS. The effect of excess body fat on female and male reproduction. Metabolism 2020; 107:154193. [PMID: 32119876 DOI: 10.1016/j.metabol.2020.154193] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 02/11/2020] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
The dramatic increase in the prevalence of obesity coincides with a decline in reproductive health indices in both sexes. Energy excess mediates changes to the regulatory mechanisms of the reproductive system. Obese individuals exhibit increased estrogen concentrations, due to the overexpression of aromatase in the adipose tissue; via a negative feedback loop, men present with symptoms of hypogonadotropic hypogonadism. These hormonal changes, along with increased oxidative stress, lipotoxicity and disturbances in the concentrations of adipokines, directly affect the gonads, peripheral reproductive organs and the embryo. Clinical evidence is somewhat contradicting, with only some studies advocating worse semen parameters, increased incidence of erectile dysfunction, increased doses of ovulation induction medications, and worse live birth rates in assisted reproductive technology (ART) cycles in obese individuals compared with those of normal weight. Similar conclusions are drawn about patients with insulin resistance syndromes, namely polycystic ovary syndrome (PCOS). As far as treatment options are concerned, lifestyle changes, medical therapy and bariatric surgery may improve the reproductive outcome, although the evidence remains inconclusive. In this review, we summarize the evidence on the association of obesity and reproductive health on both the molecular and the clinical level, and the effect of weight-loss interventions on reproductive potential.
Collapse
Affiliation(s)
- Gesthimani Mintziori
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, Greece.
| | - Meletios P Nigdelis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, Greece
| | - Hannah Mathew
- Department of Medicine Boston VA Healthcare System and Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Athanasios Mousiolis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, Greece
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, Greece
| | - Christos S Mantzoros
- Department of Medicine Boston VA Healthcare System and Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
35
|
Skorupskaite K, George JT, Veldhuis JD, Millar RP, Anderson RA. Kisspeptin and neurokinin B interactions in modulating gonadotropin secretion in women with polycystic ovary syndrome. Hum Reprod 2020; 35:1421-1431. [PMID: 32510130 PMCID: PMC7316500 DOI: 10.1093/humrep/deaa104] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
STUDY QUESTION What is the role of the hypothalamic neuropeptide neurokinin B (NKB) and its interaction with kisspeptin on GnRH/LH secretion in women with polycystic ovary syndrome (PCOS)? SUMMARY ANSWER Administration of neurokinin 3 receptor antagonist (NK3Ra) for 7 days reduced LH and FSH secretion and LH pulse frequency in women with PCOS, whilst the stimulatory LH response to kisspeptin-10 was maintained. WHAT IS KNOWN ALREADY PCOS is characterized by abnormal GnRH/LH secretion. NKB and kisspeptin are master regulators of GnRH/LH secretion, but their role in PCOS is unclear. STUDY DESIGN, SIZE, DURATION The NK3Ra MLE4901, 40 mg orally twice a day, was administered to women with PCOS for 7 days (n = 8) (vs no treatment, n = 7). On the last day of NK3Ra administration or the equivalent day in those not treated, women were randomized to 7-h kisspeptin-10 (4 µg/kg/h i.v.) or vehicle infusion. This was repeated with the alternate infusion in a subsequent cycle. PARTICIPANTS/MATERIALS, SETTING, METHODS Subjects were women with PCOS, studied in a Clinical Research Facility. Reproductive hormones were measured before and after NK3Ra administration. On the last day of NK3Ra administration (or the equivalent cycle day in untreated women), all women attended for an 8-h frequent blood sampling to allow analysis of the pulsatile LH secretion. MAIN RESULTS AND THE ROLE OF CHANCE NK3Ra reduced LH secretion (4.0 ± 0.4 vs 6.5 ± 0.8 IU/l, P < 0.05) and pulse frequency (0.5 ± 0.1 vs 0.8 ± 0.1 pulses/h, P < 0.05); FSH secretion was also reduced (2.0 ± 0.3 vs 2.5 ± 0.4 IU/l, P < 0.05). Without NK3Ra pre-treatment, kisspeptin-10 increased LH secretion (5.2 ± 0.5 to 7.8 ± 1.0 IU/L, P < 0.05), with a positive relationship to oestradiol concentrations (r2 = 0.59, P < 0.05). After NK3Ra administration, the LH response to kisspeptin-10 was preserved (vehicle 3.5 ± 0.3 vs 9.0 ± 2.2 IU/l with kisspeptin-10, P < 0.05), but the positive correlation with oestradiol concentrations was abolished (r2 = 0.07, ns. after NK3Ra). FSH secretion was increased by kisspeptin-10 after NK3Ra treatment, but not without NK3Ra treatment. LIMITATIONS, REASONS FOR CAUTION The study did not explore the dose relationship of the effect of NK3R antagonism. The impact of obesity or other aspects of the variability of the PCOS phenotype was not studied due to the small number of subjects. WIDER IMPLICATIONS OF THE FINDINGS These data demonstrate the interactive regulation of GnRH/LH secretion by NKB and kisspeptin in PCOS, and that the NKB system mediates aspects of oestrogenic feedback. STUDY FUNDING/COMPETING INTEREST(S) Wellcome Trust through Scottish Translational Medicine and Therapeutics Initiative (102419/Z/13/A) and MRC grants (G0701682 to R.P.M. and R.A.A.) and MR/N022556/1 to the MRC Centre for Reproductive Health. This work was performed within the Edinburgh Clinical Research Facility. J.T.G. has undertaken consultancy work for AstraZeneca and Takeda Pharmaceuticals and is an employee of Boehringer Ingelheim. R.P.M. has consulted for Ogeda and was CEO of Peptocrine. R.A.A. has undertaken consultancy work for Merck, Ferring, NeRRe Therapeutics and Sojournix Inc. J.D.V. and K.S. have nothing to disclose. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Karolina Skorupskaite
- MRC Centre for Reproductive Health, The Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Jyothis T George
- Warwick Medical School, Coventry CV4 7AL, UK
- Boehringer Ingelheim, Bracknell RG12 8YS, UK
| | - Johannes D Veldhuis
- Endocrine Research Unit, Center for Translational Science Activities, Mayo Clinic, Rochester, MN 55905, USA
| | - Robert P Millar
- Centre for Neuroendocrinology and Mammal Research Institute, University of Pretoria, 0028 Pretoria, South Africa
- Institute for Infectious Diseases and Molecular Medicine, University of Cape Town, 7925 Observatory, South Africa
| | - Richard A Anderson
- MRC Centre for Reproductive Health, The Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
36
|
Gourbesville C, Kerlan V, Reznik Y. Le syndrome des ovaires polykystiques : quelles nouveautés en 2019 ? ANNALES D'ENDOCRINOLOGIE 2020; 80 Suppl 1:S29-S37. [PMID: 31606059 DOI: 10.1016/s0003-4266(19)30114-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PolyCystic Ovary Syndrome (PCOS) is the first endocrinopathy of women of child-bearing age and the leading cause of anovulatory infertility. The pathophysiology of this syndrome is complex and involves genetic traits highlighted by GWAS and epigenetic traits with DNA methylation modifications. Initially described as an ovarian disease, works carried out over recent years were turned towards neuroendocrine disorder involving GABAergic pathways, KNDy neurons and a possible role of prenatal androgen exposure determined by animal models. Clinically, PCOS leads to many complications including psychological and emotional disorders demonstrated in large populations of PCOS women. © 2019 Published by Elsevier Masson SAS. All rights reserved. Cet article fait partie du numéro supplément Les Must de l'Endocrinologie 2019 réalisé avec le soutien institutionnel de Ipsen-Pharma.
Collapse
Affiliation(s)
| | | | - Yves Reznik
- Endocrinologie et métabolismes, CHU de Caen, Caen, France
| |
Collapse
|
37
|
Involvement of GJA1 and Gap Junctional Intercellular Communication between Cumulus Cells and Oocytes from Women with PCOS. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5403904. [PMID: 32190671 PMCID: PMC7066426 DOI: 10.1155/2020/5403904] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 02/14/2020] [Indexed: 12/27/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common female endocrine system disease that affects 17.8% of women of reproductive age and leads to infertility, obesity, glucose metabolic disorders, cardiovascular disease, and body-mind problems. However, the etiology of PCOS remains unclear. Follicular growth is disrupted as a result of ovarian hyperandrogenism and distorted intraovarian paracrine signaling in women with PCOS. Microcommunication between oocytes and cumulus cells plays a critical role in folliculogenesis. Gap junction alpha 1 (GJA1) plays a crucial role in the developing follicles by forming communication channels between cumulus cells and oocytes, but this has not yet been reported in women with PCOS. Therefore, we aimed to study the role of GJA1 in the microcommunication between oocytes and cumulus cells in women with PCOS. In our study, cumulus cell-oocyte complexes (COCs) from women were isolated via ultrasound-guided vaginal puncture, and oocytes were selected from COCs and categorized based on 3 oocyte maturation stages. Then, RT-qPCR and immunofluorescence analysis were performed to detect both the gene expression and protein of GJA1 in oocytes from women with and without PCOS. There was no statistically significant difference in age and BMI (body mass index), but patients with PCOS had a higher ratio of basic LH/FSH (luteinizing hormone/follicle-stimulating hormone), androstenedione, and total ovarian volume. The qRT-PCR results showed higher gene expression of GJA1 in oocytes without PCOS at the germinal vesicle (GV) stage compared with that of oocytes from women with PCOS. Immunofluorescence analysis showed that the expression level of GJA1 in oocytes from women with PCOS was very weak compared with that of oocytes from women without PCOS. In conclusion, GJA1 may play a critical role in the development of oogenesis arrest in women with PCOS throughout the oogenesis processes, including oogenesis and oocyte maturation.
Collapse
|
38
|
Lai W, Li X, Zhu H, Zhu X, Tan H, Feng P, Chen L, Luo C. Plasma luteinizing hormone level affects the brain activity of patients with polycystic ovary syndrome. Psychoneuroendocrinology 2020; 112:104535. [PMID: 31841986 DOI: 10.1016/j.psyneuen.2019.104535] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Cognitive function has been reported to be impaired in women with polycystic ovary syndrome (PCOS). This study aimed to investigate the effect of PCOS on brain activity and explore the relationship between brain activity and sex hormone levels in women with PCOS (WPCOS). METHODS Twenty-one women aged 18-45 years old with new-diagnosed PCOS were enrolled. Plasma levels of six sex hormones including luteinizing hormone (LH) and follicle-stimulating hormone (FSH) were tested during the 2-5 days of their menstrual periods. Twenty-seven healthy controls (HC) were recruited. Every subject underwent a resting-state functional magnetic resonance imaging (fMRI). The amplitude of low-frequency fluctuation (ALFF) of the whole brain was evaluated followed by the functional connectivity (FC) analysis. Finally, the correlation between the ALFF, FC of the significant areas and the plasma hormone levels were analyzed. RESULTS The patients showed increased ALFF value in the left inferior temporal gyrus (ITG.L) and decreased ALFF value in the left inferior occipital gyrus (IOG.L) as well as the superior frontal gyrus (SFG.R, P < 0.005). For the FC analysis, patients showed decreased FC in SFG.R with the right middle frontal gyrus (MFG.R, P < 0.05). The FC between SFG.R and MFG.R was negatively correlated with LH level (R=-0.594, P = 0.005) and with the LH/FSH ratio (R=-0.521, P = 0.015). CONCLUSION PCOS can induce changes in activities of brain regions responsible for visuospatial working memory, face processing and episodic memory. The reduced functional connectivity within the right frontal lobe is related with the high LH level in WPCOS.
Collapse
Affiliation(s)
- Wanlin Lai
- Department of Neurology, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, Sichuan 610041, PR China
| | - Xuan Li
- MOE Key Lab for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, PR China
| | - Huili Zhu
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, No. 20, Section 3, Renmin South Road, Chengdu, Sichuan 610041, PR China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, No. 20, Section 3, Renmin South Road, Chengdu, Sichuan 610041, PR China
| | - Xi Zhu
- Department of Neurology, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, Sichuan 610041, PR China
| | - Huiwen Tan
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, Sichuan 610041, PR China
| | - Peimin Feng
- Department of Gastroenterology, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, PR China
| | - Lei Chen
- Department of Neurology, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, Sichuan 610041, PR China.
| | - Cheng Luo
- MOE Key Lab for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, PR China.
| |
Collapse
|
39
|
Ilie IR. Neurotransmitter, neuropeptide and gut peptide profile in PCOS-pathways contributing to the pathophysiology, food intake and psychiatric manifestations of PCOS. Adv Clin Chem 2019; 96:85-135. [PMID: 32362321 DOI: 10.1016/bs.acc.2019.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a major health problem with a heterogeneous hormone-imbalance and clinical presentation across the lifespan of women. Increased androgen production and abnormal gonadotropin-releasing hormone (GnRH) release and gonadotropin secretion, resulting in chronic anovulation are well-known features of the PCOS. The brain is both at the top of the neuroendocrine axis regulating ovarian function and a sensitive target of peripheral gonadal hormones and peptides. Current literature illustrates that neurotransmitters regulate various functions of the body, including reproduction, mood and body weight. Neurotransmitter alteration could be one of the reasons for disturbed GnRH release, consequently directing the ovarian dysfunction in PCOS, since there is plenty evidence for altered catecholamine metabolism and brain serotonin or opioid activity described in PCOS. Further, the dysregulated neurotransmitter and neuropeptide profile in PCOS could also be the reason for low self-esteem, anxiety, mood swings and depression or obesity, features closely associated with PCOS women. Can these altered central brain circuits, or the disrupted gut-brain axis be the tie that would both explain and link the pathogenesis of this disorder, the occurrence of depression, anxiety and other mood disorders as well as of obesity, insulin resistance and abnormal appetite in PCOS? This review intends to provide the reader with a comprehensive overview of what is known about the relatively understudied, but very complex role that neurotransmitters, neuropeptides and gut peptides play in PCOS. The answer to the above question may help the development of drugs to specifically target these central and peripheral circuits, thereby providing a valuable treatment for PCOS patients that present to the clinic with GnRH/LH hypersecretion, obesity or psychiatric manifestations.
Collapse
Affiliation(s)
- Ioana R Ilie
- Department of Endocrinology, University of Medicine and Pharmacy 'Iuliu-Hatieganu', Cluj-Napoca, Romania.
| |
Collapse
|
40
|
Coyle C, Campbell RE. Pathological pulses in PCOS. Mol Cell Endocrinol 2019; 498:110561. [PMID: 31461666 DOI: 10.1016/j.mce.2019.110561] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/22/2019] [Accepted: 08/22/2019] [Indexed: 12/18/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a highly prevalent endocrine disorder associated with hyperandrogenism and anovulation. Although a spectrum disorder, many women with PCOS exhibit elevated luteinizing hormone (LH) pulse frequency and an elevated LH to follicle stimulating hormone ratio. This aberrant pattern of gonadotrophin signalling drives many of the downstream ovarian features of PCOS, including increased androgen synthesis, and indicates neuroendocrine impairments upstream. Decreased responsiveness to gonadal steroid hormone negative feedback in PCOS patients points toward dysfunction within the gonadotropin-releasing hormone (GnRH) neuronal network in the brain. Excessive androgen exposure during development or over pubertal onset can recapitulate the neuroendocrine pathology of PCOS in pre-clinical models, and these models have been fundamental in beginning to pick apart the specific central mechanisms involved. This mini-review will briefly describe the pathology of PCOS associated with high frequency GnRH/LH pulses and then highlight what is currently known, and yet to be discovered, about the central mechanisms involved.
Collapse
Affiliation(s)
- Christopher Coyle
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, 9054, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, 9054, New Zealand.
| |
Collapse
|
41
|
Kelley AS, Smith YR, Padmanabhan V. A Narrative Review of Placental Contribution to Adverse Pregnancy Outcomes in Women With Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2019; 104:5299-5315. [PMID: 31393571 PMCID: PMC6767873 DOI: 10.1210/jc.2019-00383] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 08/01/2019] [Indexed: 12/29/2022]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS) is the most common endocrinopathy of reproductive-aged women. In pregnancy, women with PCOS experience increased risk of miscarriage, gestational diabetes, preeclampsia, and extremes of fetal birth weight, and their offspring are predisposed to reproductive and cardiometabolic dysfunction in adulthood. Pregnancy complications, adverse fetal outcomes, and developmental programming of long-term health risks are known to have placental origins. These findings highlight the plausibility of placental compromise in pregnancies of women with PCOS. EVIDENCE SYNTHESIS A comprehensive PubMed search was performed using terms "polycystic ovary syndrome," "placenta," "developmental programming," "hyperandrogenism," "androgen excess," "insulin resistance," "hyperinsulinemia," "pregnancy," and "pregnancy complications" in both human and animal experimental models. CONCLUSIONS There is limited human placental research specific to pregnancy of women with PCOS. Gestational androgen excess and insulin resistance are two clinical hallmarks of PCOS that may contribute to placental dysfunction and underlie the higher rates of maternal-fetal complications observed in pregnancies of women with PCOS. Additional research is needed to prevent adverse maternal and developmental outcomes in women with PCOS and their offspring.
Collapse
Affiliation(s)
- Angela S Kelley
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | - Yolanda R Smith
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | - Vasantha Padmanabhan
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
- Correspondence and Reprint Requests: Vasantha Padmanabhan, PhD, Department of Pediatrics, University of Michigan, 7510 MSRB 1, 1500 West Medical Center Drive, Ann Arbor, Michigan 48109. E-mail:
| |
Collapse
|
42
|
Ruddenklau A, Campbell RE. Neuroendocrine Impairments of Polycystic Ovary Syndrome. Endocrinology 2019; 160:2230-2242. [PMID: 31265059 DOI: 10.1210/en.2019-00428] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent and distressing disorder of largely unknown etiology. Although PCOS defined by ovarian dysfunction, accumulating evidence supports a critical role for the brain in the ontogeny and pathophysiology of PCOS. A critical pathological feature of PCOS is impaired gonadal steroid hormone negative feedback to the GnRH neuronal network in the brain that regulates fertility. This impairment is associated with androgen excess, a cardinal feature of PCOS. Impaired steroid hormone feedback to GnRH neurons is thought to drive hyperactivity of the neuroendocrine axis controlling fertility, leading to a vicious cycle of androgen excess and reproductive dysfunction. Decades of clinical research have been unable to uncover the mechanisms underlying this impairment, because of the extreme difficulty in studying the brain in humans. It is only recently, with the development of preclinical models of PCOS, that we have begun to unravel the role of the brain in the development and progression of PCOS. Here, we provide a succinct overview of what is known about alterations in the steroid hormone-sensitive GnRH neuronal network that may underlie the neuroendocrine defects in clinical PCOS, with a particular focus on those that may contribute to impaired progesterone negative feedback, and the likely role of androgens in driving this impairment.
Collapse
Affiliation(s)
- Amy Ruddenklau
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
43
|
Rahimi Z, Mohammadi M Sc E. The CYP17 MSP AI (T-34C) and CYP19A1 (Trp39Arg) variants in polycystic ovary syndrome: A case-control study. Int J Reprod Biomed 2019; 17. [PMID: 31435594 PMCID: PMC6661133 DOI: 10.18502/ijrm.v17i3.4519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 06/18/2018] [Accepted: 01/19/2019] [Indexed: 12/20/2022] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is a common and chronic disorder of endocrine glands where genetic factors play a major role in the susceptibility to the disease. The cytochrome (CYP) 17 enzyme is essential for androgens biosynthesis. Also, the CYP19 enzyme converts the androgens to the aromatic estrogens. Objective We aimed to investigate the association of CYP 17 MSP AI (T-34C) and CYP 19A1 (Trp39Arg) variants with the pathogenesis of PCOS in a population from Western Iran with Kurdish ethnic background. Materials and Methods The present case-control study consisted of 50 patients with PCOS and 109 controls. The CYP17 T-34C and CYP19A1 (Trp39Arg) polymorphisms were identified by polymerase chain reaction-restriction fragment length polymorphism. The serum lipid and lipoprotein profile were detected by the Bionic Diagnostic Kits. Estradiol, dehydroepiandrosterone (DHEA), and sex hormone-binding globulin (SHBG) levels were measured using the chemiluminescent method. Results The serum levels of estradiol and SHBG in PCOS patients were lower than controls (p < 0.001 and p = 0.06, respectively). However, the level of DHEA was higher (p = 0.01) in patients compared to controls. The higher frequency of CYP17 TC genotype in patients (30%) compared to controls (15.6%) was associated with 2.31-fold susceptibility to PCOS (p = 0.038). The frequency of CYP19 TC genotype was 6.4% in controls and 10% in patients (p = 0.42).
Conclusion The present study suggests that CYP17 TC genotype could be associated with the risk of PCOS. Also, the study indicated the sex steroid hormones level alteration and the lower level of SHBG in PCOS patients compared to healthy individuals.
Collapse
Affiliation(s)
- Zohreh Rahimi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Clinical Biochemistry, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ehsan Mohammadi M Sc
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Clinical Biochemistry, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
44
|
Wang Z, Feng M, Awe O, Ma Y, Shen M, Xue P, Ahima R, Wolfe A, Segars J, Wu S. Gonadotrope androgen receptor mediates pituitary responsiveness to hormones and androgen-induced subfertility. JCI Insight 2019; 5:127817. [PMID: 31393859 DOI: 10.1172/jci.insight.127817] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Many women with hyperandrogenemia suffer from irregular menses and infertility. However, it is unknown whether androgens directly affect reproduction. Since animal models of hyperandrogenemia-induced infertility are associated with obesity, which may impact reproductive function, we have created a lean mouse model of elevated androgen using implantation of low dose dihydrotestosterone (DHT) pellets to separate the effects of elevated androgen from obesity. The hypothalamic-pituitary-gonadal axis controls reproduction. While we have demonstrated that androgen impairs ovarian function, androgen could also disrupt neuroendocrine function at the level of brain and/or pituitary to cause infertility. To understand how elevated androgens might act on pituitary gonadotropes to influence reproductive function, female mice with disruption of the androgen receptor (Ar) gene specifically in pituitary gonadotropes (PitARKO) were produced. DHT treated control mice with intact pituitary Ar (Con-DHT) exhibit disrupted estrous cyclicity and fertility with reduced pituitary responsiveness to GnRH at the level of both calcium signaling and LH secretion. These effects were ameliorated in DHT treated PitARKO mice. Calcium signaling controls GnRH regulation of LH vesicle exotocysis. Our data implicated upregulation of GEM (a voltage-dependent calcium channel inhibitor) in the pituitary as a potential mechanism for androgen's pathological effects. These results demonstrate that gonadotrope AR, as an extra-ovarian regulator, plays an important role in reproductive pathophysiology.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mingxiao Feng
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Olubusayo Awe
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yaping Ma
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Mingjie Shen
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Gynecology and Obstetrics, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Xue
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Andrew Wolfe
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Molecular and Cellular Physiology, and
| | - James Segars
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sheng Wu
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Molecular and Cellular Physiology, and.,Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
45
|
Silva MSB, Desroziers E, Hessler S, Prescott M, Coyle C, Herbison AE, Campbell RE. Activation of arcuate nucleus GABA neurons promotes luteinizing hormone secretion and reproductive dysfunction: Implications for polycystic ovary syndrome. EBioMedicine 2019; 44:582-596. [PMID: 31178425 PMCID: PMC6606966 DOI: 10.1016/j.ebiom.2019.05.065] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/26/2019] [Accepted: 05/30/2019] [Indexed: 12/29/2022] Open
Abstract
Background Enhanced GABA activity in the brain and a hyperactive hypothalamic-pituitary-gonadal axis are associated with polycystic ovary syndrome (PCOS), the most common form of anovulatory infertility. Women with PCOS exhibit elevated cerebrospinal fluid GABA levels and preclinical models of PCOS exhibit increased GABAergic input to GnRH neurons, the central regulators of reproduction. The arcuate nucleus (ARN) is postulated as the anatomical origin of elevated GABAergic innervation; however, the functional role of this circuit is undefined. Methods We employed a combination of targeted optogenetic and chemogenetic approaches to assess the impact of acute and chronic ARN GABA neuron activation. Selective acute activation of ARN GABA neurons and their fiber projections was coupled with serial blood sampling for luteinizing hormone secretion in anesthetized male, female and prenatally androgenised (PNA) mice modelling PCOS. In addition, GnRH neuron responses to ARN GABA fiber stimulation were recorded in ex vivo brain slices. Chronic activation of ARN GABA neurons in healthy female mice was coupled with reproductive phenotyping for PCOS-like features. Findings Acute stimulation of ARN GABA fibers adjacent to GnRH neurons resulted in a significant and long-lasting increase in LH secretion in male and female mice. The amplitude of this response was blunted in PNA mice, which also exhibited a blunted LH response to GnRH administration. Infrequent and variable GABAA-dependent changes in GnRH neuron firing were observed in brain slices. Chronic activation of ARN GABA neurons in healthy females impaired estrous cyclicity, decreased corpora lutea number and increased circulating testosterone levels. Interpretation ARN GABA neurons can stimulate the hypothalamic-pituitary axis and chronic activation of ARN GABA neurons can mimic the reproductive deficits of PCOS in healthy females. Unexpectedly blunted HPG axis responses in PNA mice may reflect a history of high frequency GnRH/LH secretion and reduced LH stores, but also raise questions about impaired function within the ARN GABA population and the involvement of other circuits.
Collapse
Affiliation(s)
- Mauro S B Silva
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Elodie Desroziers
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Sabine Hessler
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Melanie Prescott
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Chris Coyle
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand.
| |
Collapse
|
46
|
Glueck CJ, Goldenberg N. Characteristics of obesity in polycystic ovary syndrome: Etiology, treatment, and genetics. Metabolism 2019; 92:108-120. [PMID: 30445140 DOI: 10.1016/j.metabol.2018.11.002] [Citation(s) in RCA: 224] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/22/2018] [Accepted: 11/05/2018] [Indexed: 01/01/2023]
Abstract
Polycystic ovary syndrome (PCOS) has multiple etiologies including ovarian and adrenal hyperandrogenism, neuro-endocrine and hypothalamic-pituitary dysfunction, and disorders of peripheral insulin resistance. Obesity is neither necessary nor sufficient for the PCOS phenotype, and the association of PCOS with obesity is not universal, with national, cultural, and ethnic differences. Obesity, particularly visceral adiposity which is common in obese and non-obese women with PCOS, amplifies and worsens all metabolic and reproductive outcomes in PCOS. Obesity increases insulin resistance and compensatory hyperinsulinemia, which in turn increases adipogenesis and decreases lipolysis. Obesity sensitizes thecal cells to LH stimulation and amplifies functional ovarian hyperandrogenism by upregulating ovarian androgen production. Obesity increases inflammatory adipokines which, in turn, increase insulin resistance and adipogenesis. Lifestyle interventions focused on diet-weight loss and concurrent exercise are central to therapy which also commonly subsequently needs to include pharmacologic therapy. PCOS symptoms commonly improve with 5% to 10% weight loss, but 25% to 50% weight loss, usually achievable only through bariatric surgery, may be required for morbid obesity unresponsive to lifestyle-medical treatment. Bariatric surgery is a valuable approach to weight loss in PCOS where BMI is ≥40 kg/m2 when non-surgical treatment and/or induction of pregnancy have failed, and can be an initial treatment when BMI is ≥50 kg/m2. Further research in PCOS is needed to better understand the fundamental basis of the disorder, to ameliorate obesity, to correct hyperandrogenism, ovulation, hyperinsulinemia, and to optimize metabolic homeostasis.
Collapse
Affiliation(s)
- Charles J Glueck
- The Cholesterol, Metabolism, and Thrombosis Research Center, 3906 Middleton Avenue, Cincinnati, OH 45220, United States of America.
| | - Naila Goldenberg
- The Cholesterol, Metabolism, and Thrombosis Research Center, 3906 Middleton Avenue, Cincinnati, OH 45220, United States of America
| |
Collapse
|
47
|
Peecher DL, Binder AK, Gabriel KI. Rodent models of mental illness in polycystic ovary syndrome: the potential role of hypothalamic-pituitary-adrenal dysregulation and lessons for behavioral researchers. Biol Reprod 2019; 100:590-600. [PMID: 30388193 DOI: 10.1093/biolre/ioy233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/04/2018] [Accepted: 10/31/2018] [Indexed: 12/20/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most commonly diagnosed endocrine disorder in women of reproductive age, with phenotypes including ovarian and metabolic dysfunctions. Women with PCOS also show increased rates of mental illness, dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, and altered responsiveness to stressors that may contribute to the higher rates of mental illness, specifically depression and anxiety. Animal models of PCOS have provided insight into the ovarian and metabolic mechanisms that underlie the syndrome, and several models have been used to study the behavioral consequences associated with PCOS in the laboratory. Several studies in rodent models of PCOS demonstrate changes in anxiety-like behavior, but researchers often neglect to report procedural details or behavioral data crucial to interpreting the differences observed in those studies. Additionally, the impact of potential HPA dysregulation in animal models of PCOS may influence behavioral findings, although only three studies to date have examined this. As such, researchers should consider and report stress-associated variables (e.g., time of day, light/dark cycle, light intensity, housing, and procedures to control experimenter and litter effects) that may influence depression- and anxiety-like behaviors in rodents. This review will summarize the behavioral and HPA-related studies in women with PCOS and rodent models of the disease, and provide considerations for future studies.
Collapse
Affiliation(s)
- Danielle L Peecher
- Department of Psychology, Central Washington University, Ellensburg, Washington, USA.,Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - April K Binder
- Center for Reproductive Biology, Washington State University, Pullman, Washington, USA.,Department of Biological Sciences, Central Washington University, Ellensburg, Washington, USA
| | - Kara I Gabriel
- Department of Psychology, Central Washington University, Ellensburg, Washington, USA
| |
Collapse
|
48
|
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: 76] [Impact Index Per Article: 12.7] [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
|
49
|
Decreased Serum Level of Gamma-amino Butyric Acid in Egyptian Infertile Females with Polycystic Ovary Syndrome is Correlated with Dyslipidemia, Total Testosterone and 25(OH) Vitamin D Levels. J Med Biochem 2019; 38:512-518. [PMID: 31496917 PMCID: PMC6708297 DOI: 10.2478/jomb-2018-0051] [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: 10/23/2018] [Accepted: 12/26/2018] [Indexed: 02/06/2023] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is one of the most common female endocrine disorders around the world. Increasing evidence suggests that neurotransmitter Gamma-aminobutyric acid (GABA) is involved in the pathogenesis of PCOS through its central role in the hypothalamus. However, the peripheral role of GABA in PCOS has not been sufficiently investigated in spite of its existence in peripheral organs. First, the aim of this study is to, investigate serum GABA level in Egyptian PCOS patients. Second, to explore the correlation between serum GABA level with Body Mass Index (BMI), dyslipidemia, totaltestosterone and 25 (OH) vitamin D. Methods Eighty PCOS patients and eighty age-matched healthy females were included in this study. All parameters were assessed colourimetrically or with ELISA. Results PCOS patients exhibited significantly decreased serum GABA level compared to controls (p < 0.001). There was a significant positive correlation between serum GABA and 25(OH) vitamin D levels (r = 0.26, p = 0.018), and a significant negative correlation with total testosterone (r = - 0.3, p = 0.02), total cholesterol (TC) (r = - 0.31, p = 0.01) and LDL-Cholesterol (LDL-C) (r = - 0.23, p = 0.045), respectively. Conclusions The findings of this study suggest that disrupted GABA level in the peripheral circulation is an additional contributing factor to PCOS manifestations. GABA deficiency was correlated with 25 (OH) vitamin D deficiency, dyslipidemia, and total testosterone. Further investigations for GABA adjustment might provide a promising means for better management of PCOS symptoms.
Collapse
|
50
|
Crocetin attenuates DHT-induced polycystic ovary syndrome in mice via revising kisspeptin neurons. Biomed Pharmacother 2018; 107:1363-1369. [DOI: 10.1016/j.biopha.2018.08.135] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/15/2018] [Accepted: 08/25/2018] [Indexed: 11/20/2022] Open
|