1
|
Cheng R, Zhu Y, Yang LQ, Xiao L, Fang XH, Zhang Q. Dysregulated serum levels of kisspeptin, NKB, GABA in women with polycystic ovary syndrome and their association with hormonal profiles. Gynecol Endocrinol 2025; 41:2463533. [PMID: 39932739 DOI: 10.1080/09513590.2025.2463533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/12/2025] [Accepted: 02/01/2025] [Indexed: 05/08/2025] Open
Abstract
INTRODUCTION The clinical study aimed to evaluate the levels of serum kisspeptin, NKB, and GABA in Chinese patients with polycystic ovary syndrome (PCOS) and explore their association with hormonal profiles, as well as the relationship between these levels in PCOS patients and controls. METHODS From December 2022 to December 2023, medical records of 60 individuals diagnosed with PCOS and 32 healthy subjects were obtained. Serum kisspeptin, NKB and GABA levels were quantified using enzyme-linked immunosorbent assay (ELISA) kits. To assess the correlation, either Pearson's or Spearman's analytical method was employed between serum kisspeptin, NKB, and GABA levels and hormonal profiles. RESULTS The results showed that serum kisspeptin and NKB levels were significantly lower in PCOS patients (p﹤.05), while GABA levels were elevated compared to those in the control group (p﹤.05). In PCOS patients, kisspeptin was notably positive-correlated with LH and LH/FSH (p﹤.05), but no significant correlation was found between NKB, GABA, and hormonal profiles. In PCOS, GABA levels had a strong positive correlation with NKB levels (r = 0.613, p = .000), but this was not observed in the control group. In control patients, kisspeptin levels were positively associated with NKB (r = 0.475, p = .011), strongly negatively correlated with GABA levels (r = -0.773, p = .000), but these were not observed in the PCOS group. CONCLUSION The study concluded that patients with PCOS have dysregulated levels of serum kisspeptin, NKB, and GABA and that they may have paradoxical effects under physiological and pathological situations.
Collapse
Affiliation(s)
- Ran Cheng
- Department of Traditional Chinese Medical Gynecology, Hangzhou TCM Hospital, Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China
| | - Ying Zhu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liu-Qing Yang
- Department of Traditional Chinese Medical Gynecology, Hangzhou TCM Hospital, Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China
| | - Lu Xiao
- Healthcare Office, Guang'anmen Hospital, China Academy of Chinese Medical, Beijing, China
| | - Xiao-Hong Fang
- Department of Traditional Chinese Medical Gynecology, Hangzhou TCM Hospital, Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China
| | - Qin Zhang
- Department of Traditional Chinese Medical Gynecology, Hangzhou TCM Hospital, Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China
| |
Collapse
|
2
|
Moore AM. Neuroendocrine mechanisms responsible for elevated gonadotrophin-releasing hormone and luteinising hormone pulses in polycystic ovary syndrome. J Neuroendocrinol 2025:e70028. [PMID: 40251138 DOI: 10.1111/jne.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/20/2025]
Abstract
Polycystic ovary syndrome (PCOS) is the leading cause of anovulatory infertility in premenopausal individuals with ovaries worldwide. Despite the diagnostic features of anovulation, ovarian cysts, and hyperandrogenemia, which indicate that ovary dysfunction is the cause of the syndrome, changes in central neuroendocrine circuits are a significant cause of PCOS pathology. Specifically, cells in the hypothalamus have a diminished ability to transmit negative feedback signals from gonadal sex steroid hormones to gonadotropin-releasing hormone (GnRH) neurons. This results in an elevated frequency of pulsatile hypothalamic GnRH and pituitary luteinizing hormone (LH) secretion, leading to ovarian hyperandrogenism and ovulatory dysfunction. In recent years, preclinical research in animal models has rapidly advanced our understanding of the neural mechanisms underlying GnRH pulse generation with the identification of KNDy cells-a unique cell population in the hypothalamus expressing the neuropeptides kisspeptin, neurokinin B and dynorphin. As a result, therapeutics targeting KNDy cell signaling have emerged as a promising avenue for treating GnRH/LH hypersecretion in PCOS patients. However, the precise central changes underpinning impaired negative feedback regulation of GnRH pulse generation in PCOS patients are still unclear. Evidence from both the clinic and animal models suggests that changes in the regulation of KNDy cells may be directly responsible for elevated GnRH and LH pulse frequency in PCOS. However, other cell populations regulating GnRH secretion may also be involved. This review provides an overview of our current understanding of the aetiology and contribution of neuroendocrine dysfunction in PCOS pathology. It also examines the evidence for neural mechanisms underlying GnRH/LH hypersecretion, which may serve as central targets in developing novel treatments. Finally, this review highlights key knowledge gaps that are hindering the development of preventive and curative interventions.
Collapse
Affiliation(s)
- Aleisha M Moore
- Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| |
Collapse
|
3
|
Del Río JP, Tsompanidis A, Gaspar PA, Maturana-Hurtado A, Rojas-Costa GM, Dagnino-Subiabre A, Olea A, Maliqueo M, Echiburú B, de Guevara AL, Montiel JF, Baron-Cohen S, Crisosto N. Women with polycystic ovary syndrome (PCOS): Likelihood of cooccurring neuropsychiatric conditions and the dual hit hypothesis. Front Neuroendocrinol 2025; 77:101188. [PMID: 40120958 DOI: 10.1016/j.yfrne.2025.101188] [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: 06/28/2024] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Polycystic Ovary Syndrome (PCOS) is the most common endocrine-metabolic disorder in women of reproductive age. Hyperandrogenism has been proposed as its main pathophysiological feature. PCOS is associated with co-occurring conditions, including psychiatric disorders such as anxiety, depression, and neurodevelopmental conditions such as autism. Exposure to hyperandrogenism during prenatal life and adolescence may explain this association. PCOS women exhibit hyperandrogenism during pregnancy, and up to 70% of their daughters will present a similar phenotype from puberty onwards. The 'dual hit hypothesis' proposes that stressors during prenatal life and adolescence can synergistically lead to co-occurring conditions in adulthood. PCOS has been recently proposed as an independent likelihood factor for the development of neuropsychiatric conditions. However, the specific mechanisms require further research to develop effective interventions. This review discusses how hyperandrogenism can affect neurodevelopment during two key periods of brain development, which may explain the long-term impact of PCOS on mental health.
Collapse
Affiliation(s)
- Juan Pablo Del Río
- Department of Child and Adolescent Psychiatry and Mental Health, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; Millennium Nucleus to Improve the Mental Health of Adolescents and Youths, Imhay. Santiago, Chile
| | - Alexandros Tsompanidis
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, CB2 8AH, UK
| | - Pablo A Gaspar
- Millennium Nucleus to Improve the Mental Health of Adolescents and Youths, Imhay. Santiago, Chile; Psychiatry and Mental Health Department, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Alejandro Maturana-Hurtado
- Department of Child and Adolescent Psychiatry and Mental Health, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Gonzalo M Rojas-Costa
- School of Medicine, Finis Terrae University, 750115, Santiago, Chile; Biomedical Imaging Unit and Artificial Intelligence FISABIO-CIPF, Foundation for the Promotion of Health and Biomedical Research of the Valencia Region, Valencia, Spain
| | - Alexies Dagnino-Subiabre
- Laboratory of Stress Neurobiology, CIESAL, Faculty of Sciences, Institute of Physiology, Universidad de Valparaíso, Valparaíso 2360102, Chile; Millennium Institute for Depression and Personality Research (MIDAP), Santiago, Chile
| | - Arabia Olea
- Laboratory of Stress Neurobiology, CIESAL, Faculty of Sciences, Institute of Physiology, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Manuel Maliqueo
- Laboratory of Endocrinology and Metabolism, Department of Medicine West Division, Universidad de Chile, Santiago 8350499, Chile
| | - Bárbara Echiburú
- Laboratory of Endocrinology and Metabolism, Department of Medicine West Division, Universidad de Chile, Santiago 8350499, Chile
| | - Amanda Ladrón de Guevara
- Laboratory of Endocrinology and Metabolism, Department of Medicine West Division, Universidad de Chile, Santiago 8350499, Chile
| | - Juan F Montiel
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad Diego Portales, Santiago 8370191, Chile
| | - Simon Baron-Cohen
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK; Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, CB2 8AH, UK
| | - Nicolás Crisosto
- Laboratory of Endocrinology and Metabolism, Department of Medicine West Division, Universidad de Chile, Santiago 8350499, Chile; Endocrinology Unit, Department of Medicine, Clínica Alemana de Santiago, Faculty of Medicine, Universidad del Desarrollo, Santiago 7610658, Chile.
| |
Collapse
|
4
|
Li C, Cheng D, Ren H, Zhang T. Unraveling the gut microbiota's role in PCOS: a new frontier in metabolic health. Front Endocrinol (Lausanne) 2025; 16:1529703. [PMID: 40171188 PMCID: PMC11958223 DOI: 10.3389/fendo.2025.1529703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder affecting reproductive-age women, characterized primarily by hyperandrogenism, ovulatory dysfunction, and metabolic abnormalities. In recent years, the gut microbiota has garnered widespread attention for its potential role as a key regulator of host metabolism in the pathogenesis of PCOS. Studies have shown that PCOS patients exhibit dysbiosis in their gut microbiota, characterized by reduced microbial diversity, an imbalance in the ratio of Firmicutes to Bacteroidetes, changes in the abundance of specific taxa, and abnormal levels of metabolic products. These alterations may exacerbate metabolic dysfunction in PCOS through multiple mechanisms, including influencing host energy metabolism, disrupting lipid and bile acid metabolism, and inducing chronic inflammation. Addressing gut dysbiosis through the modulation of patients' microbiomes-such the use of, prebiotics, fecal microbiota transplantation, and optimizing diet lifestyle-may offer strategies for improving metabolic abnormalities and alleviating clinical symptoms in PCOS. Additionally, the gut microbiome promises as a potential marker, aiding in the precise diagnosis and personalization of PCOS. Although our current understanding of how the gut microbiota influences PCOS is still limited, research is needed to explore the causal relationships and mechanisms involved, providing a more reliable theoretical basis for clinical. This review aims summarize the research progress on the relationship between gut microbiota and PCOS, and to suggest future directions to promote the development of prevention and treatment strategies for PCOS.
Collapse
Affiliation(s)
- Caihong Li
- Department of Assisted Reproductive Laboratory, Shenyang Jinghua Hospital, Shenyang, China
| | - Dongkai Cheng
- Department of Assisted Reproductive Laboratory, Shenyang Jinghua Hospital, Shenyang, China
| | - Haiqin Ren
- Department of Assisted Reproductive Laboratory, Shenyang Jinghua Hospital, Shenyang, China
| | - Tao Zhang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, Shenyang, China
| |
Collapse
|
5
|
Motafeghi F, Amiri M, Noroozzadeh M, Tehrani FR. The impact of GABA and GABAergic pathway in polycystic ovary syndrome: a systematic review. Obstet Gynecol Sci 2025; 68:93-108. [PMID: 39935052 PMCID: PMC11976924 DOI: 10.5468/ogs.24255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/03/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Emerging evidence indicates that dysfunction of the gamma-aminobutyric acid (GABA)ergic pathway may contribute to the pathophysiology of polycystic ovary syndrome (PCOS), and GABA demonstrates potential in the management of PCOS symptoms. This systematic review aimed to determine the role of the GABAergic pathway in PCOS and evaluate the impact of GABA on improving the condition. Web of Science, Embase, Scopus, Cochrane, and PubMed databases were systematically searched for experimental studies, clinical trials, animal studies, and cellular investigations. The search was conducted for relevant English-language manuscripts, published up to February 2024, using keywords, such as "polycystic ovary syndrome", PCOS, "gamma-aminobutyric acid" and GABA. Quality assessment of the included studies was performed using the Cochrane Collaboration's tool and the Newcastle-Ottawa scale. The results indicate that GABAergic dysfunction adversely affects gonadotrophin-releasing hormone neuronal activity, leading to hormonal imbalances and reproductive issues. Prenatal androgen exposure and kisspeptin signaling influence GABAergic transmission to GnRH neurons, thereby linking GABA to the pathogenesis of PCOS. Additionally, GABAergic signaling affects peripheral tissues relevant to PCOS, including the immune system, gut-brain axis, and ovaries. GABA supplementation has demonstrated potential benefits in enhancing metabolic and reproductive health, such as reducing insulin resistance and modulating sex hormone levels, as supported by animal models and clinical studies involving females with PCOS. The GABAergic signaling pathway may represent a promising therapeutic target for the management of PCOS. Nevertheless, further studies are required to validate these findings and deepen our understanding of the role of GABA in the pathogenesis and treatment of PCOS.
Collapse
Affiliation(s)
- Farzaneh Motafeghi
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran,
Iran
| | - Mina Amiri
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran,
Iran
- Foundation for Research & Education Excellence, Vestavia Hills, AL,
USA
| | - Mahsa Noroozzadeh
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran,
Iran
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran,
Iran
- Foundation for Research & Education Excellence, Vestavia Hills, AL,
USA
| |
Collapse
|
6
|
Ge H, Huang D, Tan L, Luo D, Zhou L, Liu H, Zhang Y, Liu D, Wu X, Wang L, Xiong L, Yang Y, Han TL, He C, Qi H. Metabolic Profiles of Pregnancy with Polycystic Ovary Syndrome: Insights into Maternal-Fetal Metabolic Communication. J Clin Endocrinol Metab 2025:dgaf057. [PMID: 39880380 DOI: 10.1210/clinem/dgaf057] [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: 10/21/2024] [Revised: 01/04/2025] [Accepted: 01/29/2025] [Indexed: 01/31/2025]
Abstract
CONTEXT PCOS pregnancies are linked to metabolic disorders affecting maternal and fetal outcomes, with maternal metabolites differing from those in normal pregnancies. OBJECTIVE To investigate the metabolic communication at the maternal-fetal interface in PCOS pregnancies. DESIGN Placenta and umbilical cord serum were analyzed using gas chromatography-mass spectrometry. In-depth analysis was performed with clinical characteristics. SETTING Placenta and umbilical cord serum were analyzed using gas chromatography-mass spectrometry, alongside clinical characteristics. PARTICIPANTS 45 uncomplicated PCOS pregnancies and 50 normal pregnancies. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) The metabolic characteristics at the maternal-fetal interface in PCOS pregnancies and the underlying mechanisms. RESULTS A total of 79 metabolites in the placenta and 25 in umbilical cord serum showed significant differences between polycystic ovary syndrome (PCOS) and normal pregnancies. The 10 most significant placental metabolites were identified through receiver operating characteristic (ROC) analysis, 9 of which correlated significantly with maternal serum testosterone levels. Lasso regression analysis identified 4 key placental metabolite combinations: gamma-aminobutyric acid, proline, glycine, and isoleucine, achieving an area under the curve (AUC) of 93.24%. In umbilical cord serum, 6 metabolites differed significantly between PCOS and normal pregnancies, with the highest AUC reaching 76.07%, and 5 of these metabolites showed significant correlations with maternal serum testosterone levels. Nine differential metabolites were shared between the placenta and umbilical cord serum, which also shared metabolic pathways, including ABC transporters and aminoacyl-tRNA biosynthesis, potentially influencing maternal-fetal interactions. CONCLUSION This study identifies the metabolomic profile and key pathways in maternal-fetal communication during PCOS pregnancies.
Collapse
Affiliation(s)
- Huisheng Ge
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dongni Huang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China
| | - Lunbo Tan
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China
| | - Dan Luo
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Liu Zhou
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hong Liu
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yilan Zhang
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dandan Liu
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xixi Wu
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lulu Wang
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Liling Xiong
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Yang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ting-Li Han
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Chengjin He
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hongbo Qi
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Dai R, Sun Y. Altered GnRH neuron-glia networks close to interface of polycystic ovary syndrome: Molecular mechanism and clinical perspectives. Life Sci 2025; 361:123318. [PMID: 39719166 DOI: 10.1016/j.lfs.2024.123318] [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: 08/27/2024] [Revised: 11/24/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024]
Abstract
Polycystic ovary syndrome (PCOS) has been noticed as a neuroendocrine syndrome manifested by reproductive hormone dysregulation involving increased luteinizing hormone (LH) pulse frequency and an increased LH to follicle-stimulating hormone ratio, yet theory is just beginning to be established. Neuroglia located in the arcuate nucleus and median eminence (ARC-ME) that are close to gonadotropin-releasing hormone (GnRH) axon terminals, comprise the blood-brain barrier and fenestrated vessels implying their putative roles in the modulation of the abnormal GnRH pulse in PCOS. This review outlines the disturbances of neuron-glia networks that underlie hypothetically the deregulation of GnRH-LH release and impaired sex hormone negative feedback in PCOS. We then discuss chronic and low-grade inflammatory status together with gut dysbiosis and how the detriments may intrude the hypothalamus by virtue of violating interfaces between the brain and periphery, which might contribute to the etiology of the impaired neural circuits in the ARC-ME to induce PCOS.
Collapse
Affiliation(s)
- Ruoxi Dai
- Hospital & Institute of Obstetrics and Gynecology, Fudan University, Shanghai 200081, China
| | - Yan Sun
- Hospital & Institute of Obstetrics and Gynecology, Fudan University, Shanghai 200081, China; The Academy of Integrative Medicine, Fudan University, Shanghai 200081, China; Shanghai Key Laboratory of Female Reproductive Endocrine-related Disease, Shanghai 200081, China.
| |
Collapse
|
8
|
Sucquart IE, Coyle C, Rodriguez Paris V, Prescott M, Glendining KA, Potapov K, Begg DP, Gilchrist RB, Walters KA, Campbell RE. Investigating GABA Neuron-Specific Androgen Receptor Knockout in two Hyperandrogenic Models of PCOS. Endocrinology 2024; 165:bqae060. [PMID: 38788194 PMCID: PMC11151696 DOI: 10.1210/endocr/bqae060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
Androgen excess is a hallmark feature of polycystic ovary syndrome (PCOS), the most common form of anovulatory infertility. Clinical and preclinical evidence links developmental or chronic exposure to hyperandrogenism with programming and evoking the reproductive and metabolic traits of PCOS. While critical androgen targets remain to be determined, central GABAergic neurons are postulated to be involved. Here, we tested the hypothesis that androgen signaling in GABAergic neurons is critical in PCOS pathogenesis in 2 well-characterized hyperandrogenic mouse models of PCOS. Using cre-lox transgenics, GABA-specific androgen receptor knockout (GABARKO) mice were generated and exposed to either acute prenatal androgen excess (PNA) or chronic peripubertal androgen excess (PPA). Females were phenotyped for reproductive and metabolic features associated with each model and brains of PNA mice were assessed for elevated GABAergic input to gonadotropin-releasing hormone (GnRH) neurons. Reproductive and metabolic dysfunction induced by PPA, including acyclicity, absence of corpora lutea, obesity, adipocyte hypertrophy, and impaired glucose homeostasis, was not different between GABARKO and wild-type (WT) mice. In PNA mice, acyclicity remained in GABARKO mice while ovarian morphology and luteinizing hormone secretion was not significantly impacted by PNA or genotype. However, PNA predictably increased the density of putative GABAergic synapses to GnRH neurons in adult WT mice, and this PNA-induced plasticity was absent in GABARKO mice. Together, these findings suggest that while direct androgen signaling in GABA neurons is largely not required for the development of PCOS-like traits in androgenized models of PCOS, developmental programming of GnRH neuron innervation is dependent upon androgen signaling in GABA neurons.
Collapse
Affiliation(s)
- Irene E Sucquart
- Fertility & Research Centre, School of Clinical Medicine, University of New South Wales Sydney, Randwick, NSW 2031, Australia
| | - Chris Coyle
- Centre of Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand 9054
| | - Valentina Rodriguez Paris
- Fertility & Research Centre, School of Clinical Medicine, University of New South Wales Sydney, Randwick, NSW 2031, Australia
- School of Biomedical Sciences, University of New South Wales Sydney, Randwick, NSW 2052, Australia
| | - Melanie Prescott
- Centre of Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand 9054
| | - Kelly A Glendining
- Centre of Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand 9054
| | - Kyoko Potapov
- Centre of Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand 9054
| | - Denovan P Begg
- Department of Behavioural Neuroscience, School of Psychology, University of New South Wales Sydney, Randwick, NSW, Australia
| | - Robert B Gilchrist
- Fertility & Research Centre, School of Clinical Medicine, University of New South Wales Sydney, Randwick, NSW 2031, Australia
| | - Kirsty A Walters
- Fertility & Research Centre, School of Clinical Medicine, University of New South Wales Sydney, Randwick, NSW 2031, Australia
| | - Rebecca E Campbell
- Centre of Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand 9054
| |
Collapse
|
9
|
Nisa KU, Tarfeen N, Mir SA, Waza AA, Ahmad MB, Ganai BA. Molecular Mechanisms in the Etiology of Polycystic Ovary Syndrome (PCOS): A Multifaceted Hypothesis Towards the Disease with Potential Therapeutics. Indian J Clin Biochem 2024; 39:18-36. [PMID: 38223007 PMCID: PMC10784448 DOI: 10.1007/s12291-023-01130-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/01/2023] [Indexed: 03/28/2023]
Abstract
Among the premenopausal women, Polycystic Ovary Syndrome (PCOS) is the most prevalent endocrinopathy affecting the reproductive system and metabolic rhythms leading to disrupted menstrual cycle. Being heterogeneous in nature it is characterized by complex symptomology of oligomennorhoea, excess of androgens triggering masculine phenotypic appearance and/or multiple follicular ovaries. The etiology of this complex disorder remains somewhat doubtful and the researchers hypothesize multisystem links in the pathogenesis of this disease. In this review, we attempt to present several hypotheses that tend to contribute to the etiology of PCOS. Metabolic inflexibility, aberrant pattern of gonadotropin signaling along with the evolutionary, genetic and environmental factors have been discussed. Considered a lifelong endocrinological implication, no universal treatment is available for PCOS so far however; multiple drug therapy is often advised along with simple life style intervention is mainly advised to manage its cardinal symptoms. Here we aimed to present a summarized view of pathophysiological links of PCOS with potential therapeutic strategies.
Collapse
Affiliation(s)
- Khair Ul Nisa
- Department of Environmental Science, University of Kashmir, Srinagar, 190006 India
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, 190006 India
| | - Najeebul Tarfeen
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, 190006 India
| | - Shahnaz Ahmad Mir
- Department of Endocrinology, Government Medical College, Shireen Bagh, Srinagar, 190010 India
| | - Ajaz Ahmad Waza
- Multidisciplinary Research Unit (MRU), Government Medical Collage (GMC), Srinagar, 190010 India
| | - Mir Bilal Ahmad
- Department of Biochemistry, University of Kashmir, Srinagar, 190006 India
| | - Bashir Ahmad Ganai
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, 190006 India
| |
Collapse
|
10
|
Panda SP, Kesharwani A, Singh GD, Prasanth D, Vatchavai BR, Kumari PVK, Panda SK, Mallick SP. Impose of KNDy/GnRH neural circuit in PCOS, ageing, cancer and Alzheimer's disease: StAR actions in prevention of neuroendocrine dysfunction. Ageing Res Rev 2023; 92:102086. [PMID: 37821047 DOI: 10.1016/j.arr.2023.102086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/06/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
The Kisspeptin1 (KISS1)/neurokinin B (NKB)/Dynorphin (Dyn) [KNDy] neurons in the hypothalamus regulate the reproduction stage in human beings and rodents. KNDy neurons co-expressed all KISS1, NKB, and Dyn peptides, and hence commonly regarded as KISS1 neurons. KNDy neurons contribute to the "GnRH pulse generator" and are implicated in the regulation of pulsatile GnRH release. The estradiol (E2)-estrogen receptor (ER) interactions over GnRH neurons in the hypothalamus cause nitric oxide (NO) discharge, in addition to presynaptic GABA and glutamate discharge from respective neurons. The released GABA and glutamate facilitate the activity of GnRH neurons via GABAA-R and AMPA/kainate-R. The KISS1 stimulates MAPK/ERK1/2 signaling and cause the release of Ca2+ from intracellular store, which contribute to neuroendocrine function, increase apoptosis and decrease cell proliferation and metastasis. The ageing in women deteriorates KISS1/KISS1R interaction in the hypothalamus which causes lower levels of GnRH. Because examining the human brain is so challenging, decades of clinical research have failed to find the causes of KNDy/GnRH dysfunction. The KISS1/KISS1R interactions in the brain have a neuroprotective effect against Alzheimer's disease (AD). These findings modulate the pathophysiological role of the KNDy/GnRH neural network in polycystic ovarian syndrome (PCOS) associated with ageing and, its protective role in cancer and AD. This review concludes with protecting effect of the steroid-derived acute regulatory enzyme (StAR) against neurotoxicity in the hippocampus, and hypothalamus, and these measures are fundamental for delaying ageing with PCOS. StAR could serve as novel diagnostic marker and therapeutic target for the most prevalent hormone-sensitive breast cancers (BCs).
Collapse
Affiliation(s)
- Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| | - Adarsh Kesharwani
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | | | - Dsnbk Prasanth
- KVSR Siddhartha College of Pharmaceutical Sciences, Vijayawada, Andhrapradesh, India
| | - Bhaskara Raju Vatchavai
- Sri Vasavi Institute of Pharmaceutical Sciences, Pedatadepalli, Tadepalligudem, Andhrapradesh, India
| | - P V Kamala Kumari
- Vignan Institute of Pharmaceutical Technology, Duvvada, Visakhapatnam, Andhrapradesh, India
| | | | | |
Collapse
|
11
|
Yiangou A, Mollan SP, Sinclair AJ. Idiopathic intracranial hypertension: a step change in understanding the disease mechanisms. Nat Rev Neurol 2023; 19:769-785. [PMID: 37957260 DOI: 10.1038/s41582-023-00893-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 11/15/2023]
Abstract
The understanding of idiopathic intracranial hypertension (IIH) has evolved over the past few years. Previously, IIH was considered a disease exclusively affecting the neuro-ophthalmic axis, characterized by raised intracranial pressure, headache and papilloedema, and resulting in the risk of severe and permanent visual loss and life-changing disabling headaches. Recent advances have begun to redefine IIH as a probable metabolic disease involving a range of systemic manifestations. More than 95% of individuals affected by the disease are women of reproductive age with obesity. The incidence is rapidly rising and parallels the escalating worldwide obesity rates. Contemporary insights identify associations with insulin resistance, type 2 diabetes and a twofold increased risk of cardiovascular disease in excess of that driven by obesity alone. Adipose distribution in people with IIH, like that in other metabolic diseases, is preferentially centripetal and is associated with changes in intracranial pressure. Evidence now demonstrates adipose tissue dysfunction in people with IIH, involving transcriptional and metabolic priming for lipogenesis and weight gain. Hormonal perturbations are also observed, including a unique phenotype of androgen excess that promotes cerebrospinal fluid secretion. Knowledge of these additional disease features is driving research into novel therapeutic targets and altering the approach to multidisciplinary care.
Collapse
Affiliation(s)
- Andreas Yiangou
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Department of Neurology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Susan P Mollan
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Birmingham Neuro-Ophthalmology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Alexandra J Sinclair
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK.
- Department of Neurology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK.
| |
Collapse
|
12
|
Абсатарова ЮС, Евсеева ЮС, Андреева ЕН. [Neuroendocrine features of the pathogenesis of polycystic ovary syndrome (literature review)]. PROBLEMY ENDOKRINOLOGII 2023; 69:107-114. [PMID: 37968958 PMCID: PMC10680539 DOI: 10.14341/probl13350] [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: 08/22/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 11/17/2023]
Abstract
Polycystic ovary syndrome (PCOS) is one of the most pressing problems in endocrine gynecology. The main signs of the disease are hyperandrogenism, menstrual and/or ovulatory dysfunction, and polycystic ovarian structure according to ultrasound. Women with PCOS are at risk for developing metabolic syndrome, type 2 diabetes, cardiovascular disease, and endometrial cancer. In this connection, the pathogenetic mechanisms of the occurrence of this syndrome are continuously studied and new methods of treatment are being sought. PCOS is characterized by a wide range of various disorders of the neuroendocrine regulation of the reproductive system. The main focus of the review is aimed at summarizing information about the etiological role of neuropeptides and neurotransmitters, such as phoenixin, galanins, orexins, GABA, in the pathophysiology of PCOS and about the possibility of their use for diagnostic and therapeutic purposes. In recent decades, the interest of scientists has been focused on the study of KNDy neurons, because it is the kisspeptin synthesized by them that is one of the main regulators of the hypothalamic-pituitary-ovarian axis. This article discusses data on the significance of KNDy neurons in the pathogenesis of the syndrome. Information is provided on the effect of elevated levels of androgens and anti-Müllerian hormone on GnRH neurons. Also analyzed are studies on functional and structural disorders in the hypothalamus in PCOS. 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 1998.
Collapse
Affiliation(s)
| | - Ю. С. Евсеева
- Национальный медицинский исследовательский центр эндокринологии
| | - Е. Н. Андреева
- Национальный медицинский исследовательский центр эндокринологии; Московский государственный медико-стоматологический университет имени А.И. Евдокимова
| |
Collapse
|
13
|
Dong J, Rees DA. Polycystic ovary syndrome: pathophysiology and therapeutic opportunities. BMJ MEDICINE 2023; 2:e000548. [PMID: 37859784 PMCID: PMC10583117 DOI: 10.1136/bmjmed-2023-000548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023]
Abstract
Polycystic ovary syndrome is characterised by excessive levels of androgens and ovulatory dysfunction, and is a common endocrine disorder in women of reproductive age. Polycystic ovary syndrome arises as a result of polygenic susceptibility in combination with environmental influences that might include epigenetic alterations and in utero programming. In addition to the well recognised clinical manifestations of hyperandrogenism and ovulatory dysfunction, women with polycystic ovary syndrome have an increased risk of adverse mental health outcomes, pregnancy complications, and cardiometabolic disease. Unlicensed treatments have limited efficacy, mostly because drug development has been hampered by an incomplete understanding of the underlying pathophysiological processes. Advances in genetics, metabolomics, and adipocyte biology have improved our understanding of key changes in neuroendocrine, enteroendocrine, and steroidogenic pathways, including increased gonadotrophin releasing hormone pulsatility, androgen excess, insulin resistance, and changes in the gut microbiome. Many patients with polycystic ovary syndrome have high levels of 11-oxygenated androgens, with high androgenic potency, that might mediate metabolic risk. These advances have prompted the development of new treatments, including those that target the neurokinin-kisspeptin axis upstream of gonadotrophin releasing hormone, with the potential to lessen adverse clinical sequelae and improve patient outcomes.
Collapse
Affiliation(s)
- Jiawen Dong
- Neuroscience and Mental Health Innovation Institute, School of Medicine, Cardiff University, Cardiff, UK
| | - D Aled Rees
- Neuroscience and Mental Health Innovation Institute, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
14
|
Liu C, Zhao M, Zhao Y, Hu Y. Association between serum total testosterone levels and metabolic syndrome among adult women in the United States, NHANES 2011-2016. Front Endocrinol (Lausanne) 2023; 14:1053665. [PMID: 36843599 PMCID: PMC9946982 DOI: 10.3389/fendo.2023.1053665] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
OBJECTIVE To investigate the association between serum total testosterone (TT) levels and metabolic syndrome (MetS) or its components among adult women. METHODS 2,678 women from NHANES 2011-2016 were included in this cross-sectional study. MetS was determined according to the National Cholesterol Education Program Adult Treatment Panel III guidelines. The association between serum TT levels and MetS was evaluated by two logistics regression models and the adjusted restricted cubic spline (RCS). Stratified analysis and sensitive analysis were also conducted. RESULTS Continuous TT levels were negatively associated with the occurrence of MetS, and the ORs associated with per SD increase in ln TT were 0.70 (95%CI: 0.58-0.85) in 2011-2014 and 0.56 (95%CI: 0.39-0.79) in 2015-2016 in Model A. High TT group were less likely to have MetS (OR=0.60, 95%CI: 0.45-0.80 in 2011-2014 and OR=0.50, 95%CI: 0.32-0.78 in 2015-2016) when compared to the low TT group. When TT levels were divided into quartiles, TT levels were negatively correlated with the incidence of MetS (p for trend < 0.001). Similar trend was observed in Model B. Multivariate-adjusted logistic regression with RCS exhibited that TT had a L-shaped dose-response association with MetS or its components. Interaction analyses revealed that women who were less than 50 years old (OR=0.37, 95%CI: 0.22, 0.63), with depression (OR=0.50, 95%CI: 0.29, 0.87) or being smokers (OR=0.37, 95%CI: 0.23, 0.54) showed lower ORs than those who were over 50 years old (OR=0.66, 95%CI: 0.40, 1.09), without depression (OR=0.59, 95%CI: 0.41, 0.85) or non-smokers (OR=0.59, 95%CI: 0.39, 0.89) when measure the association between ln TT and the occurrence of MetS. CONCLUSIONS Our study indicated that TT levels are negatively correlated with the occurrence of MetS, with interaction effects of age, smoke behaviors, and depressive status.
Collapse
Affiliation(s)
- Chenning Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao, Macao SAR, China
| | - Meiduo Zhao
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
| | - Yuanjia Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao, Macao SAR, China
- *Correspondence: Yuanjia Hu,
| |
Collapse
|
15
|
Garg A, Patel B, Abbara A, Dhillo WS. Treatments targeting neuroendocrine dysfunction in polycystic ovary syndrome (PCOS). Clin Endocrinol (Oxf) 2022; 97:156-164. [PMID: 35262967 DOI: 10.1111/cen.14704] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/30/2021] [Accepted: 01/04/2022] [Indexed: 01/01/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women of reproductive age and is the leading cause of anovulatory subfertility. Increased gonadotrophin releasing hormone (GnRH) pulsatility in the hypothalamus results in preferential luteinizing hormone (LH) secretion from the pituitary gland, leading to ovarian hyperandrogenism and oligo/anovulation. The resultant hyperandrogenism reduces negative feedback from sex steroids such as oestradiol and progesterone to the hypothalamus, and thus perpetuates the increase in GnRH pulsatility. GnRH neurons do not have receptors for oestrogen, progesterone, or androgens, and thus the disrupted feedback is hypothesized to occur via upstream neurons. Likely candidates for these upstream regulators of GnRH neuronal pulsatility are Kisspeptin, Neurokinin B (NKB), and Dynorphin neurons (termed KNDy neurons). Growing insight into the neuroendocrine dysfunction underpinning the heightened GnRH pulsatility seen in PCOS has led to research on the use of pharmaceutical agents that specifically target the activity of these KNDy neurons to attenuate symptoms of PCOS. This review aims to highlight the neuroendocrine abnormalities that lead to increased GnRH pulsatility in PCOS, and outline data on recent therapeutic advancements that could potentially be used to treat PCOS. Emerging evidence has investigated the use of neurokinin 3 receptor (NK3R) antagonists as a method of reducing GnRH pulsatility and alleviating features of PCOS such as hyperandrogenism. We also consider other potential mechanisms by which increased GnRH pulsatility is controlled, which could form the basis of future avenues of research.
Collapse
Affiliation(s)
- Akanksha Garg
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Bijal Patel
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Ali Abbara
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Waljit S Dhillo
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
16
|
Berga SL. Brain phenotype in PCOS: androgens, anovulation, and gender. Gynecol Endocrinol 2022; 38:615-616. [PMID: 35971943 DOI: 10.1080/09513590.2022.2106475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/23/2022] [Indexed: 10/15/2022] Open
Affiliation(s)
- Sarah L Berga
- Department of Obstetrics and Gynecology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo SUNY, Buffalo, NY, USA
| |
Collapse
|
17
|
Female sexual behavior is disrupted in a preclinical mouse model of PCOS via an attenuated hypothalamic nitric oxide pathway. Proc Natl Acad Sci U S A 2022; 119:e2203503119. [PMID: 35867816 PMCID: PMC9335209 DOI: 10.1073/pnas.2203503119] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a major endocrine disorder leading to female infertility worldwide. Patients with PCOS also often experience sexual dysfunction; however, the developmental and central mechanisms mediating this behavioral derangement are unclear. Here, we show that prenatal excess of anti-Müllerian hormone triggers PCOS-like impairment in female sexual behavior in mice. Sexual dysfunction in PCOS-like mice is associated with decreased expression of progesterone-sensitive neuronal nitric oxide synthase (nNOS) neurons in the hypothalamus. Chemogenetic inhibition of nNOS neuronal activity in the ventromedial nucleus of the hypothalamus recapitulates PCOS-like sexual dysfunction. Of clinical relevance, administration of nitric oxide donor rescues normal sexual behavior in PCOS-like mice. Women with polycystic ovary syndrome (PCOS) frequently experience decreased sexual arousal, desire, and sexual satisfaction. While the hypothalamus is known to regulate sexual behavior, the specific neuronal pathways affected in patients with PCOS are not known. To dissect the underlying neural circuitry, we capitalized on a robust preclinical animal model that reliably recapitulates all cardinal PCOS features. We discovered that female mice prenatally treated with anti-Müllerian hormone (PAMH) display impaired sexual behavior and sexual partner preference over the reproductive age. Blunted female sexual behavior was associated with increased sexual rejection and independent of sex steroid hormone status. Structurally, sexual dysfunction was associated with a substantial loss of neuronal nitric oxide synthase (nNOS)-expressing neurons in the ventromedial nucleus of the hypothalamus (VMH) and other areas of hypothalamic nuclei involved in social behaviors. Using in vivo chemogenetic manipulation, we show that nNOSVMH neurons are required for the display of normal sexual behavior in female mice and that pharmacological replenishment of nitric oxide restores normal sexual performance in PAMH mice. Our data provide a framework to investigate facets of hypothalamic nNOS neuron biology with implications for sexual disturbances in PCOS.
Collapse
|
18
|
Desroziers E. Unusual suspects: Glial cells in fertility regulation and their suspected role in polycystic ovary syndrome. J Neuroendocrinol 2022; 34:e13136. [PMID: 35445462 PMCID: PMC9489003 DOI: 10.1111/jne.13136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 11/28/2022]
Abstract
Gonadotropin-releasing-hormone (GnRH) neurons sitting within the hypothalamus control the production of gametes and sex steroids by the gonads, therefore ensuring survival of species. As orchestrators of reproductive function, GnRH neurons integrate information from external and internal cues. This occurs through an extensively studied neuronal network known as the "GnRH neuronal network." However, the brain is not simply composed of neurons. Evidence suggests a role for glial cells in controlling GnRH neuron activity, secretion and fertility outcomes, although numerous questions remain. Glial cells have historically been seen as support cells for neurons. This idea has been challenged by the discovery that some neurological diseases originate from glial dysfunction. The prevalence of infertility disorders is increasing worldwide, with one in four couples being affected; therefore, it remains essential to understand the mechanisms by which the brain controls fertility. The "GnRH glial network" could be a major player in infertility disorders and represent a potential therapeutic target. In polycystic ovary syndrome (PCOS), the most common infertility disorder of reproductive aged women worldwide, the brain is considered a prime suspect. Recent studies have demonstrated pathological neuronal wiring of the "GnRH neuronal network" in PCOS-like animal models. However, the role of the "GnRH glial network" remains to be elucidated. In this review, I aim to propose glial cells as unusual suspects in infertility disorders such as PCOS. In the first part, I state our current knowledge about the role of glia in the regulation of GnRH neurons and fertility. In the second part, based on our recent findings, I discuss how glial cells could be implicated in PCOS pathology.
Collapse
Affiliation(s)
- Elodie Desroziers
- Department of Physiology, Centre for NeuroendocrinologyUniversity of OtagoDunedinNew Zealand
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine – Institut de Biologie Paris Seine, Neuroplasticity of Reproductive Behaviours TeamParisFrance
| |
Collapse
|
19
|
McCartney CR, Campbell RE, Marshall JC, Moenter SM. The role of gonadotropin-releasing hormone neurons in polycystic ovary syndrome. J Neuroendocrinol 2022; 34:e13093. [PMID: 35083794 PMCID: PMC9232905 DOI: 10.1111/jne.13093] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/21/2021] [Accepted: 01/11/2022] [Indexed: 01/28/2023]
Abstract
Given the critical central role of gonadotropin-releasing hormone (GnRH) neurons in fertility, it is not surprising that the GnRH neural network is implicated in the pathology of polycystic ovary syndrome (PCOS), the most common cause of anovulatory infertility. Although many symptoms of PCOS relate most proximately to ovarian dysfunction, the central reproductive neuroendocrine system ultimately drives ovarian function through its regulation of anterior pituitary gonadotropin release. The typical cyclical changes in frequency of GnRH release are often absent in women with PCOS, resulting in a persistent high-frequency drive promoting gonadotropin changes (i.e., relatively high luteinizing hormone and relatively low follicle-stimulating hormone concentrations) that contribute to ovarian hyperandrogenemia and ovulatory dysfunction. However, the specific mechanisms underpinning GnRH neuron dysfunction in PCOS remain unclear. Here, we summarize several preclinical and clinical studies that explore the causes of aberrant GnRH secretion in PCOS and the role of disordered GnRH secretion in PCOS pathophysiology.
Collapse
Affiliation(s)
- Christopher R. McCartney
- Center for Research in Reproduction and Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVAUSA
| | - Rebecca E. Campbell
- Centre for Neuroendocrinology and Department of PhysiologySchool of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| | - John C. Marshall
- Center for Research in Reproduction and Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVAUSA
| | - Suzanne M. Moenter
- Departments of Molecular & Integrative PhysiologyInternal MedicineObstetrics and GynecologyUniversity of MichiganAnn ArborMIUSA
| |
Collapse
|
20
|
Xu X, Shen HR, Yu M, Du MR, Li XL. MicroRNA let-7i inhibits granulosa-luteal cell proliferation and oestradiol biosynthesis by directly targeting IMP2. Reprod Biomed Online 2022; 44:803-816. [PMID: 35339367 DOI: 10.1016/j.rbmo.2022.01.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 11/30/2022]
Abstract
RESEARCH QUESTION Increased granulosa cell division is associated with abnormal folliculogenesis in polycystic ovary syndrome (PCOS). Lethal-7i microRNA (let-7i) may play an important role in the follicular development and granulosa cell growth; therefore is let-7i involved in PCOS pathogenesis? DESIGN The expression of let-7i was measured in granulosa-luteal cells (GLC) from women with or without PCOS. A human granulosa cell line, KGN, was used for the functional study. Mimics and inhibitors of let-7i, lentiviruses expressing insulin-like growth factor 2 mRNA binding protein (IMP2), and small-interfering RNAs were transfected into KGN cells. KGN cell proliferation was determined by 5-ethynyl-2'-deoxyuridine (EdU) and Cell Counting Kit-8 (CCK-8) assays. The cell cycle and apoptosis were assessed by propidium iodide-annexin V (PI-A) staining and fluorescence-activated cell sorting. Oestradiol concentration was determined by enzyme-linked immunoassay. Bioinformatics analysis and luciferase reporter assay were applied to confirm the let-7i target genes. RESULTS The study showed that let-7i was down-regulated in PCOS GLC (P = 0.001). Mimics of let-7i inhibited KGN proliferation (P = 0.001), and decreased aromatase expression (P = 0.030) and oestradiol production (P = 0.029), whereas let-7i inhibitors had the opposite effect. Bioinformatics analysis and quantitative real-time (qRT) PCR identified IMP2 as a target of let-7i (P = 0.021). qRT-PCR and western blot analysis indicated that IMP2 was up-regulated in GLC in women with PCOS (P = 0.001 and P = 0.044), and IMP2 expression was suppressed by let-7i in KGN cells (P < 0.001). Luciferase reporter assay results (P = 0.002), combined with the rescue assay, confirmed that let-7i inhibited KGN cell proliferation and reduced oestradiol concentration by directly targeting IMP2. CONCLUSIONS let-7i was down-regulated in PCOS GLC. Overexpression of let-7i inhibited KGN cell proliferation and decreased oestradiol production in an IMP2-dependent manner, providing a new molecular mechanism for PCOS.
Collapse
Affiliation(s)
- Xiao Xu
- Obstetrics and Gynecology Hospital, Fudan University Shanghai, People's Republic of China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai, People's Republic of China
| | - Hao-Ran Shen
- Obstetrics and Gynecology Hospital, Fudan University Shanghai, People's Republic of China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai, People's Republic of China
| | - Min Yu
- Obstetrics and Gynecology Hospital, Fudan University Shanghai, People's Republic of China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai, People's Republic of China
| | - Mei-Rong Du
- Obstetrics and Gynecology Hospital, Fudan University Shanghai, People's Republic of China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai, People's Republic of China.
| | - Xue-Lian Li
- Obstetrics and Gynecology Hospital, Fudan University Shanghai, People's Republic of China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai, People's Republic of China.
| |
Collapse
|
21
|
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
|
22
|
Neuroendocrine Determinants of Polycystic Ovary Syndrome. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19053089. [PMID: 35270780 PMCID: PMC8910170 DOI: 10.3390/ijerph19053089] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/11/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women and a major cause of anovulatory infertility. A diagnosis of PCOS is established based the presence of two out of three clinical symptoms, which are criteria accepted by the ESHRE/ASRM (European Society of Human Reproduction and Embryology/American Society for Reproductive Medicine). Gonadotropin-releasing hormone (GnRH) is responsible for the release of luteinizing hormone, and follicle stimulating hormone from the pituitary and contributes a leading role in controlling reproductive function in humans. The goal of this review is to present the current knowledge on neuroendocrine determinations of PCOS. The role of such neurohormones as GnRH, and neuropeptides kisspeptin, neurokinin B, phoenixin-14, and galanin is discussed in this aspect. Additionally, different neurotransmitters (gamma-aminobutyric acid (GABA), glutamate, serotonin, dopamine, and acetylcholine) can also be involved in neuroendocrine etiopathogenesis of PCOS. Studies have shown a persistent rapid GnRH pulse frequency in women with PCOS present during the whole ovulatory cycle. Other studies have proved that patients with PCOS are characterized by higher serum kisspeptin levels. The observations of elevated serum kisspeptin levels in PCOS correspond with the hypothesis that overactivity in the kisspeptin system is responsible for hypothalamic-pituitary-gonadal axis overactivity. In turn, this causes menstrual disorders, hyperandrogenemia and hyperandrogenism. Moreover, abnormal regulation of Neurokinin B (NKB) is also suspected of contributing to PCOS development, while NKB antagonists are used in the treatment of PCOS leading to reduction in Luteinizing hormone (LH) concentration and total testosterone concentration. GnRH secretion is regulated not only by kisspeptin and neurokinin B, but also by other neurohormones, such as phoenixin-14, galanin, and Glucagon-like peptide-1 (GLP-1), that have favorable effects in counteracting the progress of PCOS. A similar process is associated with the neurotransmitters such as GABA, glutamate, serotonin, dopamine, and acetylcholine, as well as the opioid system, which may interfere with secretion of GnRH, and therefore, influence the development and severity of symptoms in PCOS patients. Additional studies are required to explain entire, real mechanisms responsible for PCOS neuroendocrine background.
Collapse
|
23
|
Miniksar DY, Cansız MA, Kılıç M, Göçmen AY. Relationship between sleep problems and chronotypes of children and adolescents with attention deficit and hyperactivity disorder and serum GABA, glutamate and homocysteine levels. Chronobiol Int 2022; 39:386-397. [PMID: 34961406 DOI: 10.1080/07420528.2021.2018452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 02/07/2023]
Abstract
In this study, we aimed to determine the relationship between chronotype, sleep problems and serum levels of GABA (gamma amminobutyric acid), glutamate and homocysteine in children and adolescents diagnosed with attention deficit and hyperactivity disorder (ADHD) as well as factors affecting this relationship. Sleep problems of 46 children and adolescents aged 7-18 years diagnosed with ADHD and 30 healthy volunteers aged 7-18 years were evaluated with Children's Sleep Habits Questionnaire (CSHQ) while chronotypes were evaluated with Children's Chronotype Questionnaire (CCQ). Serum glutamate, GABA and homocysteine levels were measured using immunosorbent test (ELISA) kits. Sleep problems were significantly more common in the ADHD group compared to the control group (p < .001). Serum GABA, glutamate and homocysteine levels were found to be predictor biomarkers for ADHD, independent of total sleep problem score. When the homocysteine levels were above the cut-off point of 9.445 µmol/L, the sensitivity in early diagnosis of ADHD was 84.8% and the specificity was 70.0%. Although ADHD is a disorder in which sleep problems are common, increased serum GABA, glutamate and homocysteine are important in diagnosing ADHD independent of ADHD-related sleep problems. Homocysteine levels may be an important predictor for the presence of ADHD.
Collapse
Affiliation(s)
| | | | - Mahmut Kılıç
- Department of Public Health, Yozgat Bozok University, Yozgat, Turkey
| | | |
Collapse
|
24
|
Emanuel RHK, Roberts J, Docherty PD, Lunt H, Campbell RE, Möller K. A review of the hormones involved in the endocrine dysfunctions of polycystic ovary syndrome and their interactions. Front Endocrinol (Lausanne) 2022; 13:1017468. [PMID: 36457554 PMCID: PMC9705998 DOI: 10.3389/fendo.2022.1017468] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/27/2022] [Indexed: 11/16/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) affects up to 20% of women but remains poorly understood. It is a heterogeneous condition with many potential comorbidities. This review offers an overview of the dysregulation of the reproductive and metabolic systems associated with PCOS. Review of the literature informed the development of a comprehensive summarizing 'wiring' diagram of PCOS-related features. This review provides a justification for each diagram aspect from the relevant academic literature, and explores the interactions between the hypothalamus, ovarian follicles, adipose tissue, reproductive hormones and other organ systems. The diagram will provide an efficient and useful tool for those researching and treating PCOS to understand the current state of knowledge on the complexity and variability of PCOS.
Collapse
Affiliation(s)
- Rebecca H. K. Emanuel
- Department of Mechanical Engineering, University of Canterbury, Christchurch, New Zealand
| | - Josh Roberts
- Department of Mechanical Engineering, University of Canterbury, Christchurch, New Zealand
| | - Paul D. Docherty
- Department of Mechanical Engineering, University of Canterbury, Christchurch, New Zealand
- Institute of Technical Medicine, Furtwangen University, Villingen-Schwenningen, Germany
- *Correspondence: Paul D. Docherty,
| | - Helen Lunt
- Diabetes Services, Te Whatu Ora Waitaha Canterbury, Canterbury, New Zealand
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Rebecca E. Campbell
- School of Biomedical Sciences, Department of Physiology, Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - Knut Möller
- Institute of Technical Medicine, Furtwangen University, Villingen-Schwenningen, Germany
| |
Collapse
|
25
|
Shaaban Z, Tamadon A, Jafarzadeh Shirazi MR, Zamiri MJ, Derakhshanfar A. Maternal aromatase inhibition via letrozole altered RFamide-related peptide-3 and gonadotropin-releasing hormone expression in pubertal female rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:110-120. [PMID: 35656443 PMCID: PMC9118272 DOI: 10.22038/ijbms.2022.60962.13499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/22/2021] [Indexed: 11/21/2022]
Abstract
Objectives Despite prevalence of polycystic ovary syndrome (PCOS) among childbearing women and development of many animal models for this syndrome, information on its etiology is still scarce. The intrauterine hyperandrogenic environment may underlie changes at the level of hypothalamus, pituitary, ovary organization in female offspring, and PCOS later in life. Letrozole has been shown to mimic reproductive and metabolic characteristics of PCOS in adult rodent models. Therefore, this research aimed to assess the condition in a prenatal letrozole-treated rat model. Materials and Methods Twenty-eight female rats dams receiving letrozole at certain doses during late pregnancy were used in the trial. Pregnant Sprague-Dawley rats (n=21) received letrozole treatment on gestation days 16-18 at doses of 1.25, 1.0, 0.75, 0.5, and 0.25 mg/kg body weight (BW). Results Prenatal letrozole treatment delayed parturition time and reduced the litter size in pregnant dams (P<0.0001). Late puberty onset, irregular ovarian cyclicity, increased anogenital distance (AGD), body weight gain, serum testosterone concentration, and reduced estradiol levels (P<0.0001) were observed in the female offspring of dams receiving 1.25 and 1 mg/kg BW letrozole. Furthermore, letrozole at 1.25 and 1 mg/kg BW showed increased RFRP and decreased GnRH mRNA expression (P<0.0001). Letrozole treatment at doses of 1 mg/kg BW and lower was not fetotoxic. Conclusion It was concluded that 1 mg/kg BW letrozole may be suggested for prenatal PCOS induction.
Collapse
Affiliation(s)
- Zahra Shaaban
- Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran, These authors contributed equally to this work
| | - Amin Tamadon
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran, These authors contributed equally to this work
| | - Mohammad Reza Jafarzadeh Shirazi
- Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran,Corresponding author: Mohammad Reza Jafarzadeh Shirazi. Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran. Tel/Fax: +98-71-3228-6073;
| | - Mohammad Javad Zamiri
- Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran
| | - Amin Derakhshanfar
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran,Center of Comparative and Experimental Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
26
|
Zhang W, Tan B, Deng J, Haitao Z. Multiomics analysis of soybean meal induced marine fish enteritis in juvenile pearl gentian grouper, Epinephelus fuscoguttatus ♀ × Epinephelus lanceolatus ♂. Sci Rep 2021; 11:23319. [PMID: 34857775 PMCID: PMC8640039 DOI: 10.1038/s41598-021-02278-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 09/20/2021] [Indexed: 01/02/2023] Open
Abstract
As an important protein source, soybean products can cause intestinal inflammation and injury in many animals including human beings, particularly infants and juvenile individuals. Research in this field has been performed for terrestrial animals and fish, but still lacks integrity and systematicness. In this study, the main biological processes in the intestinal tract of marine fish juvenile pearl gentian grouper in the state of soybean meal-induced enteritis (SBMIE) were analyzed. A total of 720 groupers with an approximate initial weight of 12.5 g were randomly divided into three groups: the fish meal (FM) control group, the 20% SBM group (SBM20), and the SBM40 group (n = 4). Three iso-nitrogenous and iso-lipidic diets were prepared and fed to fish for 10 weeks. Each barrel contained a water volume of about 1 m3 in and was exposed to natural light and temperature. Results indicated that the growth and physiology of groupers fed with SBM were significantly negatively affected, with the gene expressions of intestinal structural protein abnormal. 16SrDNA high-throughput sequencing showed that the intestinal microflora played an important role in the pathogenesis of pearl gentian grouper SBMIE, which may activate a variety of pathogen pattern recognition receptors, such as toll-like receptors (TLRs), RIG-I-like receptors, and nod-like receptors. Transcriptome analysis revealed that changes of the SBMIE signaling pathway in pearl gentian groupers were conservative to some extent than that of terrestrial animals and freshwater fish. Moreover, the TLRs-nuclear factor kappa-B signaling pathway becomes activated, which played an important role in SBMIE. Meanwhile, the signal pathways related to nutrient absorption and metabolism were generally inhibited. Metabolomics analysis showed that isoflavones and saponins accounted for a large proportion in the potential biomarkers of pearl gentian grouper SBMIE, and most of the biomarkers had significantly positive or negative correlations with each other; 56 metabolites were exchanged between intestinal tissues and contents, which may play an important role in the development of enteritis, including unsaturated fatty acids, organic acids, amino acids, vitamins, small peptides, and nucleotides, etc. These results provide a basic theoretical reference for solving the intestinal issues of fish SBMIE and research of inflammatory bowel disease in mammals.
Collapse
Affiliation(s)
- Wei Zhang
- Laboratory of Aquatic Animal Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524025, Guangdong, People's Republic of China
- Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, 524025, Guangdong, People's Republic of China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, 524025, Guangdong, People's Republic of China
| | - Beiping Tan
- Laboratory of Aquatic Animal Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524025, Guangdong, People's Republic of China.
- Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, 524025, Guangdong, People's Republic of China.
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, 524025, Guangdong, People's Republic of China.
- Laboratory of Aquatic Animal Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, People's Republic of China.
| | - Junming Deng
- Laboratory of Aquatic Animal Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524025, Guangdong, People's Republic of China
- Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, 524025, Guangdong, People's Republic of China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, 524025, Guangdong, People's Republic of China
| | - Zhang Haitao
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, 524025, Guangdong, People's Republic of China
| |
Collapse
|
27
|
Ye Z, Zhang C, Wang S, Zhang Y, Li R, Zhao Y, Qiao J. Amino acid signatures in relation to polycystic ovary syndrome and increased risk of different metabolic disturbances. Reprod Biomed Online 2021; 44:737-746. [DOI: 10.1016/j.rbmo.2021.11.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/13/2022]
|
28
|
Liang Z, Di N, Li L, Yang D. Gut microbiota alterations reveal potential gut-brain axis changes in polycystic ovary syndrome. J Endocrinol Invest 2021; 44:1727-1737. [PMID: 33387350 DOI: 10.1007/s40618-020-01481-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Polycystic ovary syndrome (PCOS) is a common heterogeneous endocrine disorder companied with neuroendocrine and metabolic disorders. Gut microbiota has been implicated to play a key role in metabolic diseases and the production of neurotransmitters. Previous studies have reported the alterations in the gut microbiota of PCOS patients and animal models, however, most of the articles did not take the effect of age or diet on gut microbiota into account. The aim of this study was to identify the differential gut microbial species in PCOS patients compared with age and BMI-matched healthy control women. METHODS We performed physical examinations and dietary survey in 20 women with PCOS (lean PCOS, PL, n = 10; overweight PCOS, PO, n = 10) and 20 healthy control women (lean control, CL, n = 10; overweight control, CO, n = 10), and collected the blood on the days 1-3 of the menstrual cycle for the measurement of endocrine and metabolic profiles, and inflammatory factors; and collected the feces in non-menstrual period to investigate the composition of gut microbiota by sequencing the V4 region of the 16S rDNA gene in fecal samples. The correlations between clinical parameters and the differential species were evaluated. RESULTS Dietary analysis showed that the intake of dietary fiber, vitamin D were significantly decreased in PCOS. For the first time, our study found an increase of gamma-aminobutyric acid (GABA)-producing species in PCOS, including Parabacteroides distasonis, Bacteroides fragilis and Escherichia coli, which significantly positively correlated with serum LH levels and LH:FSH ratios. CONCLUSIONS GABA-producing bacteria that were increased in PCOS, including Parabacteroides distasonis, Bacteroides fragilis and Escherichia coli, showed positive relationship with serum LH levels and LH:FSH ratios. In conclusion, gut microbial dysbiosis in women with PCOS is associated with neuroendocrine changes, revealing a potential gut-brain axis in PCOS.
Collapse
Affiliation(s)
- Z Liang
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - N Di
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - L Li
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - D Yang
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
29
|
Dinsdale NL, Crespi BJ. Endometriosis and polycystic ovary syndrome are diametric disorders. Evol Appl 2021; 14:1693-1715. [PMID: 34295358 PMCID: PMC8288001 DOI: 10.1111/eva.13244] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/09/2021] [Accepted: 04/10/2021] [Indexed: 12/15/2022] Open
Abstract
Evolutionary and comparative approaches can yield novel insights into human adaptation and disease. Endometriosis and polycystic ovary syndrome (PCOS) each affect up to 10% of women and significantly reduce the health, fertility, and quality of life of those affected. PCOS and endometriosis have yet to be considered as related to one another, although both conditions involve alterations to prenatal testosterone levels and atypical functioning of the hypothalamic-pituitary-gonadal (HPG) axis. Here, we propose and evaluate the novel hypothesis that endometriosis and PCOS represent extreme and diametric (opposite) outcomes of variation in HPG axis development and activity, with endometriosis mediated in notable part by low prenatal and postnatal testosterone, while PCOS is mediated by high prenatal testosterone. This diametric disorder hypothesis predicts that, for characteristics shaped by the HPG axis, including hormonal profiles, reproductive physiology, life-history traits, and body morphology, women with PCOS and women with endometriosis will manifest opposite phenotypes. To evaluate these predictions, we review and synthesize existing evidence from developmental biology, endocrinology, physiology, life history, and epidemiology. The hypothesis of diametric phenotypes between endometriosis and PCOS is strongly supported across these diverse fields of research. Furthermore, the contrasts between endometriosis and PCOS in humans parallel differences among nonhuman animals in effects of low versus high prenatal testosterone on female reproductive traits. These findings suggest that PCOS and endometriosis represent maladaptive extremes of both female life-history variation and expression of sexually dimorphic female reproductive traits. The diametric disorder hypothesis for endometriosis and PCOS provides novel, unifying, proximate, and evolutionary explanations for endometriosis risk, synthesizes diverse lines of research concerning the two most common female reproductive disorders, and generates future avenues of research for improving the quality of life and health of women.
Collapse
Affiliation(s)
| | - Bernard J. Crespi
- Department of Biological SciencesSimon Fraser UniversityBurnabyBCCanada
| |
Collapse
|
30
|
Concentrations of estradiol, progesterone and testosterone in sefrum and cerebrospinal fluid of patients with aneurysmal subarachnoid hemorrhage correlate weakly with transcranial Doppler flow velocities. BMC Neurosci 2021; 22:29. [PMID: 33892632 PMCID: PMC8067654 DOI: 10.1186/s12868-021-00634-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 04/13/2021] [Indexed: 12/14/2022] Open
Abstract
Background The implication of the steroids estradiol, progesterone and testosterone in cerebral vasospasm after aneurysmal subarachnoid hemorrhage (aSAH) has not been comprehensively assessed. In rodents, studies suggested beneficial effects of steroids on cerebral vasospasm after experimental SAH. Studies in humans are warranted, however, a general dilemma of human studies on neuroactive substances is that the brain is not directly accessible and that concentrations in the periphery may not adequately parallel concentrations in the central compartments. In the present study, concentrations of estradiol, progesterone and testosterone in serum and cerebrospinal fluid (CSF) of patients with aSAH were determined. Blood flow velocities in cerebral arteries were measured by transcranial Doppler sonography (TCD). The aim of this study was to evaluate the correlations between the cerebral blood flow velocities and levels of estradiol, progesterone and testosterone in CSF and serum. Results Samples of serum and CSF of 42 patients with aSAH were collected concomitantly daily or every other day via the arterial line and the external ventricular drainage for two weeks after the hemorrhage. Blood flow velocities in the cerebral arteries were determined by TCD. Total estradiol, progesterone and testosterone concentrations were measured by electro-chemiluminescence immunoassay. The strength of correlation was assessed by Spearman’s rank correlation coefficient. The correlation analysis revealed very weak correlations between cerebral blood flow velocities and concentrations of estradiol, progesterone and testosterone levels in both compartments with correlation coefficients below 0.2. Conclusions In humans with aSAH, merely very weak correlations between flow velocities in cerebral arteries and concentrations of estradiol, progesterone and testosterone in serum and CSF were demonstrated. These results suggest a limited influence of the respective steroids on cerebral vascular tone although vasodilatory effects were described in rodent studies. Thus, the implication of steroids in processes of neurological deterioration warrants further clarification.
Collapse
|
31
|
Ishii M, Senju A, Oguro A, Shimono M, Araki S, Kusuhara K, Itoh K, Tsuji M, Ishihara Y. Measurement of the Estradiol Concentration in Cerebrospinal Fluid from Infants and Its Correlation with Serum Estradiol and Exosomal MicroRNA-126-5p. Biol Pharm Bull 2020; 43:1966-1968. [PMID: 33268717 DOI: 10.1248/bpb.b20-00549] [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] [Indexed: 11/22/2022]
Abstract
Estradiol has an important role in the brain, such as in neuronal development and protection, but estradiol levels in the human brain have not been well investigated. In this study, we measured the estradiol concentration in the cerebrospinal fluid (CSF) of infants to reveal the relationships between the estradiol concentrations in the serum and the CSF and further determined exosomal microRNAs in serum. Estradiol in the CSF was strongly correlated with serum estradiol and moderately correlated with miR-126-5p in the serum exosomes. This report is the first to determine the estradiol concentration in CSF from infants and showed that the levels of miR-126-5p as well as serum estradiol can be candidates to predict brain estrogen status.
Collapse
Affiliation(s)
- Masahiro Ishii
- Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health
| | - Ayako Senju
- Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health
| | - Ami Oguro
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University
| | - Masayuki Shimono
- Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health
| | - Shunsuke Araki
- Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health
| | - Koichi Kusuhara
- Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health
| | - Kouichi Itoh
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University
| | - Mayumi Tsuji
- Department of Environmental Health, School of Medicine, University of Occupational and Environmental Health
| | - Yasuhiro Ishihara
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University
| |
Collapse
|
32
|
Teubel J, Parr MK. Determination of neurosteroids in human cerebrospinal fluid in the 21st century: A review. J Steroid Biochem Mol Biol 2020; 204:105753. [PMID: 32937199 DOI: 10.1016/j.jsbmb.2020.105753] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022]
Abstract
Determination of steroid hormones synthesized by the human body plays an important role in various fields of endocrinology. Neurosteroids (NS) are steroids that are synthesized in the central (CNS) or peripheral nervous system (PNS), which is not only a source but also a target for neurosteroids. They are discussed as possible biomarkers in various cognitive disorders and research interest in this topic raises continuously. Nevertheless, knowledge on functions and metabolism is still limited, although the concept of neurosteroids was already introduced in the 1980s. Until today, the analysis of neurosteroids is truly challenging. The only accessible matrix for investigations of brain metabolism in living human beings is cerebrospinal fluid (CSF), which therefore becomes a very interesting specimen for analysis. However, neurosteroid concentrations are expected to be very low and the available amount of cerebrospinal fluid is limited. Further, high structural similarities of endogenous neurosteroids challenges analysis. Therefore, comprehensive methods, highly selective and sensitive for a large range of concentrations for different steroids in one aliquot are required and under continuous development. Although research has been increasingly intensified, still only few data are available on reference levels of neurosteroids in human cerebrospinal fluid. In this review, published literature of the last twenty years, as a period with relatively contemporary analytical methods, was systematically investigated. Considerations on human cerebrospinal fluid, different analytical approaches, and available data on levels of in analogy to periphery conceivable occurring neurosteroids, including (pro-) gestagens, androgens, corticoids, estrogens, and steroid conjugates, and their interpretation are intensively discussed.
Collapse
Affiliation(s)
- Juliane Teubel
- Freie Universität Berlin, Institute of Pharmacy, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - Maria Kristina Parr
- Freie Universität Berlin, Institute of Pharmacy, Königin-Luise-Str. 2+4, 14195 Berlin, Germany.
| |
Collapse
|
33
|
Witchel SF, Plant TM. Intertwined reproductive endocrinology: Puberty and polycystic ovary syndrome. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2020; 14:127-136. [PMID: 33102929 PMCID: PMC7583558 DOI: 10.1016/j.coemr.2020.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous familial disorder often emerging during the peri-pubertal years concomitantly with the onset of gonadarche and adrenarche. Both gonadarche and PCOS reflect functional changes in the hypothalamic-pituitary-ovarian axis. During this transition, normal girls manifest features consistent with PCOS such as irregular menses, mild hyperandrogenism, and multi-follicular ovary morphology. Themes common to puberty and PCOS, neuroendocrine features, androgen exposure, and insulin sensitivity, will be considered to address the possibility that PCOS interferes with the normal pubertal transition.
Collapse
Affiliation(s)
- Selma Feldman Witchel
- Division of Pediatric Endocrinology, UPMC Children's Hospital of Pittsburgh/University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224 USA
| | - Tony M Plant
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, PA 15213, USA
| |
Collapse
|
34
|
Wawrzkiewicz-Jałowiecka A, Kowalczyk K, Trybek P, Jarosz T, Radosz P, Setlak M, Madej P. In Search of New Therapeutics-Molecular Aspects of the PCOS Pathophysiology: Genetics, Hormones, Metabolism and Beyond. Int J Mol Sci 2020; 21:ijms21197054. [PMID: 32992734 PMCID: PMC7582580 DOI: 10.3390/ijms21197054] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/20/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022] Open
Abstract
In a healthy female reproductive system, a subtle hormonal and metabolic dance leads to repetitive cyclic changes in the ovaries and uterus, which make an effective ovulation and potential implantation of an embryo possible. However, that is not so in the case of polycystic ovary syndrome (PCOS), in which case the central mechanism responsible for entraining hormonal and metabolic rhythms during the menstrual cycle is notably disrupted. In this review we provide a detailed description of the possible scenario of PCOS pathogenesis. We begin from the analysis of how a set of genetic disorders related to PCOS leads to particular malfunctions at a molecular level (e.g., increased enzyme activities of cytochrome P450 (CYP) type 17A1 (17α-hydroxylase), 3β-HSD type II and CYP type 11A1 (side-chain cleavage enzyme) in theca cells, or changes in the expression of aquaporins in granulosa cells) and discuss further cellular- and tissue-level consequences (e.g., anovulation, elevated levels of the advanced glycation end products in ovaries), which in turn lead to the observed subsequent systemic symptoms. Since gene-editing therapy is currently out of reach, herein special emphasis is placed on discussing what kinds of drug targets and which potentially active substances seem promising for an effective medication, acting on the primary causes of PCOS on a molecular level.
Collapse
Affiliation(s)
- Agata Wawrzkiewicz-Jałowiecka
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, 44-100 Gliwice, Poland;
- Correspondence: ; Tel.: +48-32-237-12-85
| | - Karolina Kowalczyk
- Department of Obstetrics and Gynecology, Medical University of Silesia in Katowice, 40-752 Katowice, Poland; (K.K.); (P.R.); (P.M.)
| | - Paulina Trybek
- Faculty of Science and Technology, University of Silesia in Katowice, 41-500 Chorzow, Poland;
| | - Tomasz Jarosz
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, 44-100 Gliwice, Poland;
| | - Patrycja Radosz
- Department of Obstetrics and Gynecology, Medical University of Silesia in Katowice, 40-752 Katowice, Poland; (K.K.); (P.R.); (P.M.)
| | - Marcin Setlak
- Department of Neurosurgery, Medical University of Silesia, 40-752 Katowice, Poland;
| | - Paweł Madej
- Department of Obstetrics and Gynecology, Medical University of Silesia in Katowice, 40-752 Katowice, Poland; (K.K.); (P.R.); (P.M.)
| |
Collapse
|
35
|
Stener-Victorin E, Padmanabhan V, Walters KA, Campbell RE, Benrick A, Giacobini P, Dumesic DA, Abbott DH. Animal Models to Understand the Etiology and Pathophysiology of Polycystic Ovary Syndrome. Endocr Rev 2020; 41:bnaa010. [PMID: 32310267 PMCID: PMC7279705 DOI: 10.1210/endrev/bnaa010] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/14/2020] [Indexed: 12/14/2022]
Abstract
More than 1 out of 10 women worldwide are diagnosed with polycystic ovary syndrome (PCOS), the leading cause of female reproductive and metabolic dysfunction. Despite its high prevalence, PCOS and its accompanying morbidities are likely underdiagnosed, averaging > 2 years and 3 physicians before women are diagnosed. Although it has been intensively researched, the underlying cause(s) of PCOS have yet to be defined. In order to understand PCOS pathophysiology, its developmental origins, and how to predict and prevent PCOS onset, there is an urgent need for safe and effective markers and treatments. In this review, we detail which animal models are more suitable for contributing to our understanding of the etiology and pathophysiology of PCOS. We summarize and highlight advantages and limitations of hormonal or genetic manipulation of animal models, as well as of naturally occurring PCOS-like females.
Collapse
Affiliation(s)
| | - Vasantha Padmanabhan
- Departments of Pediatrics, Obstetrics and Gynecology, and Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan
| | - Kirsty A Walters
- Fertility & Research Centre, School of Women’s and Children’s Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Anna Benrick
- Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- School of Health Sciences and Education, University of Skövde, Skövde, Sweden
| | - Paolo Giacobini
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, California
| | - David H Abbott
- Department of Obstetrics and Gynecology, Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
36
|
Abstract
Although the fundamental symptoms of polycystic ovary syndrome (PCOS) relate most directly to ovarian dysfunction, central neuroendocrine systems play a prominent role in its pathophysiology. Gonadotropin-releasing hormone (GnRH) pulse generator resistance to negative feedback contributes to rapid GnRH pulse secretion, which promotes gonadotropin abnormalities that foster ovarian hyperandrogenemia and ovulatory dysfunction. The causes of GnRH neuron dysfunction, however, have remained enigmatic. In this review, we highlight a number of recent preclinical and clinical studies pertinent to the neuroendocrine abnormalities of PCOS, including those that have provided important insights into the relevance of animal models with PCOS-like features, the potential roles of kisspeptin and γ-aminobutyric acid (GABA)-ergic neurons, and the potential role of anti-Müllerian hormone.
Collapse
|
37
|
Zhu HL, Chen YQ, Zhang ZF. Downregulation of lncRNA ZFAS1 and upregulation of microRNA-129 repress endocrine disturbance, increase proliferation and inhibit apoptosis of ovarian granulosa cells in polycystic ovarian syndrome by downregulating HMGB1. Genomics 2020; 112:3597-3608. [PMID: 32320818 DOI: 10.1016/j.ygeno.2020.04.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 01/17/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The objective was to find the role of long-non-coding RNA zinc finger antisense 1 (lncRNA ZFAS1)/microRNA (miR)-129/high-mobility group box protein 1 (HMGB1) axis in polycystic ovary syndrome (PCOS). METHODS Ovarian granulosa cells from non-PCOS patients and PCOS patients were collected, and HMGB1, miR-129 and lncRNA ZFAS1 expression were detected. Ovarian granulosa cells were transfected with si-ZFAS1 or miR-129 mimics to verify their roles in P4 and E2 secretion, and the biological functions of ovarian granulosa cells. RESULTS LncRNA ZFAS1 and HMGB1 were elevated, while miR-129 was down-regulated in ovarian granulosa cells of PCOS patients. Down-regulated lncRNA ZFAS1 or overexpressed miR-129 could decrease HMGB1 expression, increase P4 and E2 secretion, promote proliferation activity while inhibit apoptosis of ovarian granulosa cells in PCOS. CONCLUSION LncRNA ZFAS1 could bind to miR-129 to promote HMGB1 expression, thereby affecting the endocrine disturbance, proliferation and apoptosis of ovarian granulosa cells in PCOS.
Collapse
Affiliation(s)
- Hong-Li Zhu
- Department of Gynecology and Obstetrics, Affiliated Hangzhou People's Hospital of Nanjing Medical University, Hangzhou 310010, Zhejiang, PR China
| | - Yue-Qun Chen
- Department of Gynecology and Obstetrics, Affiliated Hangzhou First People's Hospital of Zhejiang University, Hangzhou 310010, Zhejiang, PR China
| | - Zhi-Fen Zhang
- Department of Gynecology and Obstetrics, Affiliated Hangzhou People's Hospital of Nanjing Medical University, Hangzhou 310010, Zhejiang, PR China.
| |
Collapse
|
38
|
Wu G, Yang Z, Chen Y, Li X, Yang J, Yin T. Downregulation of Lnc-OC1 attenuates the pathogenesis of polycystic ovary syndrome. Mol Cell Endocrinol 2020; 506:110760. [PMID: 32070768 DOI: 10.1016/j.mce.2020.110760] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/13/2020] [Accepted: 02/13/2020] [Indexed: 02/06/2023]
Abstract
Long non-coding RNAs (lncRNAs) play a vital role in the progression of many human diseases. The aim of this study is to explore the relationship between lncRNA-ovarian cancer associated 1 (Lnc-OC1) and PCOS. In this study, we found that Lnc-OC1 was significantly higher in PCOS granulosa cells (GCs) compared to non-PCOS GCs. Lnc-OC1 knockdown inhibited cell viability and promoted cell apoptosis, expression of aromatase mRNA and production of estradiol in KGN cells. In PCOS mice, Lnc-OC1 promoted the serum insulin release, production of angiogenesis-related factors and IκBα phosphorylation, which could be partially restored by Lnc-OC1 shRNA. These results suggest that Lnc-OC1 plays an important part in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Gengxiang Wu
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Zhe Yang
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Yajie Chen
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Xiaoling Li
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Jing Yang
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China.
| | - Tailang Yin
- Reproductive Medical Centre, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China.
| |
Collapse
|
39
|
Abstract
The neuroendocrinology of reproduction focuses on the neuromodulation of gonadotropin-releasing hormone (GnRH), the ontogeny of the hypothalamic-pituitary-gonadal axis, and common reproductive events and conditions, namely, puberty, the menstrual cycle, and disorders of reproductive function. The core concept underpinning the neuroendocrinology of reproduction is neuroregulation of hypothalamic GnRH drive. In both men and women, reproductive function requires that GnRH input elicit appropriate secretion of follicle-stimulating hormone and luteinizing hormone from the anterior pituitary and that the gonads respond to such input appropriately. Moreover, insufficient GnRH drive causes hypothalamic hypogonadism and secondary insufficiency of gonadal sex steroid hormone synthesis and release in both sexes. Alterations in GnRH drive also reflect gonadal conditions such as dysgenesis, hyperandrogenism, gonadotropin mutations, and aging and loss or absence of oocytes or Sertoli cells. The most common cause of insufficient GnRH drive is functional, that is, due to the endocrine effects of psychologic or behavioral variables. Rarely does reduced GnRH drive reflect organic or congenital causes such as developmental defects, brain tumors, or celiac disease. Despite a common neuropathogenesis the heterogeneity of behavioral variables associated with reduced GnRH drive has resulted in a variety of names, including functional hypothalamic amenorrhea, stress-induced anovulation, and psychogenic amenorrhea.
Collapse
Affiliation(s)
- Deepika Garg
- (1)Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Sarah L Berga
- Department of Obstetrics and Gynecology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States.
| |
Collapse
|
40
|
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
|
41
|
Li Y, Liu YD, Zhou XY, Chen SL, Chen X, Zhe J, Zhang J, Zhang QY, Chen YX. MiR-29a regulates the proliferation, aromatase expression, and estradiol biosynthesis of human granulosa cells in polycystic ovary syndrome. Mol Cell Endocrinol 2019; 498:110540. [PMID: 31421163 DOI: 10.1016/j.mce.2019.110540] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/09/2019] [Accepted: 08/12/2019] [Indexed: 01/05/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility in reproductive-aged women; however, its etiology remains poorly understood. This study aimed to reveal the role of miR-29a in PCOS. MiR-29a levels were measured in the granulosa cells (GCs) of forty-seven PCOS patients and forty-seven controls. A receiver operating characteristic (ROC) curve was drawn to evaluate the diagnostic value of miR-29a in non-hyperandrogenism PCOS. MiR-29a was overexpressed in KGN and COV434 cells to examine its roles in proliferation, cell-cycle progression, and steroidogenesis. MiR-29a was significantly down-regulated in PCOS patients, and associated with an increased antral follicle count. The ROC curve showed a major separation between PCOS patients and controls. MiR-29a overexpression in KGN and COV434 cells inhibited cell proliferation, arrested cell-cycle progression, and decreased aromatase expression and estradiol production. These findings suggest that miR-29a is involved in GC proliferation and steroidogenesis, providing insights into PCOS pathogenesis.
Collapse
Affiliation(s)
- Ying Li
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yu-Dong Liu
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xing-Yu Zhou
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Shi-Ling Chen
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| | - Xin Chen
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jing Zhe
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jun Zhang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Qing-Yan Zhang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Ying-Xue Chen
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| |
Collapse
|
42
|
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: 55] [Impact Index Per Article: 9.2] [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
|
43
|
Martin J, Plank E, Jungwirth B, Hapfelmeier A, Podtschaske A, Kagerbauer SM. Weak correlations between serum and cerebrospinal fluid levels of estradiol, progesterone and testosterone in males. BMC Neurosci 2019; 20:53. [PMID: 31619164 PMCID: PMC6794746 DOI: 10.1186/s12868-019-0535-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 10/05/2019] [Indexed: 01/16/2023] Open
Abstract
Background Neuroactive steroids seem to be implicated in a variety of neurophysiological and behavioral processes, such as sleep, learning, memory, stress, feeding and aging. Numerous studies have also addressed this implication in various cerebral disorders and diseases. Yet, the correlation and association between steroids in the periphery, e.g. blood, and the central compartments, e.g. cerebrospinal fluid (CSF), have not yet been comprehensively assessed. As the brain is not directly accessible, and the collection of human CSF usually requires invasive procedures, easier accessible compartments, such as blood, have always attracted attention. However, studies in humans are scarce. In the present study we determined estradiol, progesterone and testosterone levels in CSF and serum of 22 males without cerebral disorders or diseases. Results Samples were taken under conditions corresponding closest to basal conditions with patients expecting only spinal anesthesia and minor surgery. All samples per patient were collected concomitantly. Total estradiol, progesterone and testosterone concentrations were measured by electro-chemiluminescence immunoassay. The strength of correlation was assessed by Spearman’s rank correlation coefficient. Correlation analysis revealed merely weak to very weak correlations for estradiol, progesterone and testosterone respectively between the CSF and serum compartments. Conclusions Total steroid levels of estradiol, progesterone and testosterone in CSF and serum of males without neurological disorders were determined. Weak to very weak correlations between CSF and serum were found thus suggesting that concentrations in the periphery do not parallel concentrations in the central compartments. Further research is needed to clarify to what extent and under which conditions serum levels of estradiol, progesterone and testosterone may possibly serve as a biomarker reflecting the respective concentrations in the CSF or in the brain.
Collapse
Affiliation(s)
- Jan Martin
- Department of Anesthesiology and Intensive Care Medicine, Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Ismaninger Strasse 22, 81675, Munich, Germany.
| | - Eva Plank
- Department of Anesthesiology and Intensive Care Medicine, Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Bettina Jungwirth
- Department of Anesthesiology and Intensive Care Medicine, Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Alexander Hapfelmeier
- Institute of Medical Informatics, Statistics und Epidemiology, Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany
| | - Armin Podtschaske
- Department of Anesthesiology and Intensive Care Medicine, Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Ismaninger Strasse 22, 81675, Munich, Germany
| | - Simone M Kagerbauer
- Department of Anesthesiology and Intensive Care Medicine, Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Ismaninger Strasse 22, 81675, Munich, Germany
| |
Collapse
|
44
|
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
|
45
|
Coutinho EA, Kauffman AS. The Role of the Brain in the Pathogenesis and Physiology of Polycystic Ovary Syndrome (PCOS). Med Sci (Basel) 2019; 7:E84. [PMID: 31382541 PMCID: PMC6722593 DOI: 10.3390/medsci7080084] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/25/2019] [Accepted: 07/30/2019] [Indexed: 12/20/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disorder, affecting at least 10% of women of reproductive age. PCOS is typically characterized by the presence of at least two of the three cardinal features of hyperandrogenemia (high circulating androgen levels), oligo- or anovulation, and cystic ovaries. Hyperandrogenemia increases the severity of the condition and is driven by increased luteinizing hormone (LH) pulse secretion from the pituitary. Indeed, PCOS women display both elevated mean LH levels, as well as an elevated frequency of LH pulsatile secretion. The abnormally high LH pulse frequency, reflective of a hyperactive gonadotropin-releasing hormone (GnRH) neural circuit, suggests a neuroendocrine basis to either the etiology or phenotype of PCOS. Several studies in preclinical animal models of PCOS have demonstrated alterations in GnRH neurons and their upstream afferent neuronal circuits. Some rodent PCOS models have demonstrated an increase in GnRH neuron activity that correlates with an increase in stimulatory GABAergic innervation and postsynaptic currents onto GnRH neurons. Additional studies have identified robust increases in hypothalamic levels of kisspeptin, another potent stimulator of GnRH neurons. This review outlines the different brain and neuroendocrine changes in the reproductive axis observed in PCOS animal models, discusses how they might contribute to either the etiology or adult phenotype of PCOS, and considers parallel findings in PCOS women.
Collapse
Affiliation(s)
- Eulalia A Coutinho
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Alexander S Kauffman
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
46
|
Witchel SF, Oberfield SE, Peña AS. Polycystic Ovary Syndrome: Pathophysiology, Presentation, and Treatment With Emphasis on Adolescent Girls. J Endocr Soc 2019; 3:1545-1573. [PMID: 31384717 PMCID: PMC6676075 DOI: 10.1210/js.2019-00078] [Citation(s) in RCA: 266] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/30/2019] [Indexed: 02/06/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous disorder characterized by hyperandrogenism and chronic anovulation. Depending on diagnostic criteria, 6% to 20% of reproductive aged women are affected. Symptoms of PCOS arise during the early pubertal years. Both normal female pubertal development and PCOS are characterized by irregular menstrual cycles, anovulation, and acne. Owing to the complicated interwoven pathophysiology, discerning the inciting causes is challenging. Most available clinical data communicate findings and outcomes in adult women. Whereas the Rotterdam criteria are accepted for adult women, different diagnostic criteria for PCOS in adolescent girls have been delineated. Diagnostic features for adolescent girls are menstrual irregularity, clinical hyperandrogenism, and/or hyperandrogenemia. Pelvic ultrasound findings are not needed for the diagnosis of PCOS in adolescent girls. Even before definitive diagnosis of PCOS, adolescents with clinical signs of androgen excess and oligomenorrhea/amenorrhea, features of PCOS, can be regarded as being "at risk for PCOS." Management of both those at risk for PCOS and those with a confirmed PCOS diagnosis includes education, healthy lifestyle interventions, and therapeutic interventions targeting their symptoms. Interventions can include metformin, combined oral contraceptive pills, spironolactone, and local treatments for hirsutism and acne. In addition to ascertaining for associated comorbidities, management should also include regular follow-up visits and planned transition to adult care providers. Comprehensive knowledge regarding the pathogenesis of PCOS will enable earlier identification of girls with high propensity to develop PCOS. Timely implementation of individualized therapeutic interventions will improve overall management of PCOS during adolescence, prevent associated comorbidities, and improve quality of life.
Collapse
Affiliation(s)
- Selma Feldman Witchel
- UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sharon E Oberfield
- Division of Pediatric Endocrinology, Columbia University Medical Center, New York–Presbyterian Morgan Stanley Children’s Hospital, New York, New York
| | - Alexia S Peña
- Robinson Research Institute, University of Adelaide, North Adelaide, South Australia, Australia
| |
Collapse
|
47
|
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: 68] [Impact Index Per Article: 11.3] [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
|
48
|
O'Reilly MW, Westgate CS, Hornby C, Botfield H, Taylor AE, Markey K, Mitchell JL, Scotton WJ, Mollan SP, Yiangou A, Jenkinson C, Gilligan LC, Sherlock M, Gibney J, Tomlinson JW, Lavery GG, Hodson DJ, Arlt W, Sinclair AJ. A unique androgen excess signature in idiopathic intracranial hypertension is linked to cerebrospinal fluid dynamics. JCI Insight 2019; 4:125348. [PMID: 30753168 PMCID: PMC6483000 DOI: 10.1172/jci.insight.125348] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/05/2019] [Indexed: 12/14/2022] Open
Abstract
Idiopathic intracranial hypertension (IIH) is a condition of unknown etiology, characterized by elevated intracranial pressure frequently manifesting with chronic headaches and visual loss. Similar to polycystic ovary syndrome (PCOS), IIH predominantly affects obese women of reproductive age. In this study, we comprehensively examined the systemic and cerebrospinal fluid (CSF) androgen metabolome in women with IIH in comparison with sex-, BMI-, and age-matched control groups with either simple obesity or PCOS (i.e., obesity and androgen excess). Women with IIH showed a pattern of androgen excess distinct to that observed in PCOS and simple obesity, with increased serum testosterone and increased CSF testosterone and androstenedione. Human choroid plexus expressed the androgen receptor, alongside the androgen-activating enzyme aldoketoreductase type 1C3. We show that in a rat choroid plexus cell line, testosterone significantly enhanced the activity of Na+/K+-ATPase, a surrogate of CSF secretion. We demonstrate that IIH patients have a unique signature of androgen excess and provide evidence that androgens can modulate CSF secretion via the choroid plexus. These findings implicate androgen excess as a potential causal driver and therapeutic target in IIH. Women with idiopathic intracranial hypertension have a unique androgen excess signature that is associated with modulation of cerebrospinal fluid secretion dynamics.
Collapse
Affiliation(s)
- Michael W O'Reilly
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Edgbaston, Birmingham, United Kingdom
| | - Connar Sj Westgate
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Catherine Hornby
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Hannah Botfield
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Edgbaston, Birmingham, United Kingdom
| | - Angela E Taylor
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Edgbaston, Birmingham, United Kingdom
| | - Keira Markey
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Edgbaston, Birmingham, United Kingdom.,Department of Neurology, and
| | - James L Mitchell
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Edgbaston, Birmingham, United Kingdom.,Department of Neurology, and
| | - William J Scotton
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Edgbaston, Birmingham, United Kingdom.,Department of Neurology, and
| | - Susan P Mollan
- Birmingham Neuro-Ophthalmology Unit, Ophthalmology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Andreas Yiangou
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Edgbaston, Birmingham, United Kingdom.,Department of Neurology, and
| | - Carl Jenkinson
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Lorna C Gilligan
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Mark Sherlock
- Department of Endocrinology and Diabetes Mellitus, Tallaght Hospital, Tallaght, Dublin, Ireland
| | - James Gibney
- Department of Endocrinology and Diabetes Mellitus, Tallaght Hospital, Tallaght, Dublin, Ireland
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, National Institute of Health Research (NIHR) Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Edgbaston, Birmingham, United Kingdom
| | - David J Hodson
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Edgbaston, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors, University of Birmingham, and University of Warwick, Birmingham, United Kingdom
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Edgbaston, Birmingham, United Kingdom
| | - Alexandra J Sinclair
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Edgbaston, Birmingham, United Kingdom.,Department of Neurology, and
| |
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: 10] [Impact Index Per Article: 1.7] [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
|
Xu XH, Kou LC, Wang HM, Bo CM, Song XC. Genetic polymorphisms of melatonin receptors 1A and 1B may result in disordered lipid metabolism in obese patients with polycystic ovary syndrome. Mol Med Rep 2019; 19:2220-2230. [PMID: 30664204 PMCID: PMC6390034 DOI: 10.3892/mmr.2019.9872] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 11/28/2018] [Indexed: 02/07/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a condition in which a woman's levels of the sex hormones (estrogen and progesterone) are out of balance, leading to the growth of ovarian cysts. PCOS can affect the menstrual cycle, fertility, cardiac function and even appearance of women. Therefore, we aimed to explore the genetic polymorphism of the melatonin receptors 1A and 1B in obese patients with PCOS to identify a new theoretical basis for its treatment. Patients presenting with PCOS (n=359) were enrolled and classified into an obese OB-PCOS group [body mass index (BMI) of PCOS patients ≥25 kg/m2] or a nonobese NOB-PCOS group, and 215 oviduct infertile patients who experienced normal ovulation were used as the control group. All baseline characteristics, endocrine hormone levels, lipid and glucose metabolism, and insulin indices were measured. The genotypes of rs2119882 within the MTNR1A gene and of rs10830963 within the MTNR1B gene were determined by PCR-RFLP; the genotype frequency and the difference in the distribution of allele frequency were compared. For rs2119882, C allele carriers who were not diagnosed with PCOS had an increased risk of developing PCOS, and C allele carriers with PCOS had an increased risk of developing OB-PCOS. For rs10830963, G allele carriers who were not diagnosed with PCOS had an increased risk of developing PCOS. The TT genotype in rs2119882 and the CC genotype in rs10830963 were protective factors for OB-PCOS, and increased levels of LH, testosterone, and estradiol and abnormal menstruation were key risk factors for PCOS. Furthermore, the TT genotype at the rs2119882 site was the key protective factor for OB-PCOS patients. Our study found that MTNR1A rs2119882 and MTNR1B rs10830963 could increase the risk for PCOS and cause glycolipid metabolism disorder in PCOS patients.
Collapse
Affiliation(s)
- Xiu-Hua Xu
- Obstetrics and Gynecology Clinic, Dongying People's Hospital, Dongying, Shandong 257091, P.R. China
| | - Lian-Cui Kou
- Department of Blood Rheumatism, Dongying People's Hospital, Dongying, Shandong 257091, P.R. China
| | - Hai-Mei Wang
- Marketing and Customer Service, Dongying People's Hospital, Dongying, Shandong 257091, P.R. China
| | - Chun-Mei Bo
- Obstetrics and Gynecology Clinic, Dongying People's Hospital, Dongying, Shandong 257091, P.R. China
| | - Xiao-Cui Song
- Department of Reproductive Medicine, Dongying People's Hospital, Dongying, Shandong 257091, P.R. China
| |
Collapse
|