1
|
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
|
2
|
Pavli P, Kalampokas T, Eleftheriades M, Lambrinoudaki I, Vlahos NF, Valsamakis G. Daughters of PCOS mothers and AMH plasma concentrations in pre-puberty and puberty: A meta-analysis. Early Hum Dev 2025; 201:106194. [PMID: 39799804 DOI: 10.1016/j.earlhumdev.2025.106194] [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: 11/17/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
PURPOSE to compare the Anti-Müllerian hormone (AMH) plasma concentrations of pre-pubertal and pubertal daughters born to polycystic ovary syndrome (PCOS) mothers to daughters born to control mothers and to investigate their alterations during pre-puberty and all stages of puberty. METHODS We critically investigated and meta-analyzed observational studies, which compared the plasma concentrations of AMH in pre-pubertal and pubertal daughters of PCOS pregnancies. A search of the literature was completed till the end of June of 2024 in the PubMed, Scopus, and Medline for the eligible studies. The meta-analysis followed the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA). The primary outcome included AMH plasma concentrations. RESULTS Our search yielded 961 potentially eligible studies, 5 of which were finally studied. Pre-pubertal female offsprings of PCOS mothers present higher plasma concentrations of AMH, with the pooled mean difference 10.08, while also pubertal daughters of PCOS mothers present higher plasma concentrations of AMH, with the pooled mean difference 15.79. CONCLUSION Pre-pubertal and pubertal female offsprings of PCOS mothers show higher AMH plasma concentrations, when compared to daughters of healthy mothers without PCOS. The main pathophysiological pathways of these findings seem to be hyperandrogenism, hyperinsulinemia, and also genetic, epigenetic and intrauterine factors.
Collapse
Affiliation(s)
- Polina Pavli
- 2nd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, "Aretaieion" University Hospital, 11528 Athens, Greece.
| | - Theodoros Kalampokas
- 2nd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, "Aretaieion" University Hospital, 11528 Athens, Greece
| | - Makarios Eleftheriades
- 2nd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, "Aretaieion" University Hospital, 11528 Athens, Greece
| | - Irene Lambrinoudaki
- 2nd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, "Aretaieion" University Hospital, 11528 Athens, Greece
| | - Nikolaos F Vlahos
- 2nd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, "Aretaieion" University Hospital, 11528 Athens, Greece
| | - George Valsamakis
- 2nd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, "Aretaieion" University Hospital, 11528 Athens, Greece
| |
Collapse
|
3
|
Watanabe Y, Fisher L, Campbell RE, Jasoni CL. Developmental expression patterns of gonadal hormone receptors in arcuate kisspeptin and GABA neurons of the postnatal female mouse. J Neuroendocrinol 2025; 37:e13477. [PMID: 39605295 DOI: 10.1111/jne.13477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/29/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
The arcuate nucleus of the hypothalamus (ARC) is central in the neuronal regulation of fertility and reproduction through translating gonadal steroid hormone cues into the GnRH signaling pathway in the brain. Evidence suggests that circulating gonadal steroids play an important role in modulating female reproduction via kisspeptin and γ-aminobutyric acid (GABA) neurons in the ARC in both development and adulthood. However, the temporal onset of these ARC neurons' sensitivity to gonadal steroids is unknown. Using RNAscope® in situ hybridization, we localized androgen receptor (Ar), estrogen receptor alpha (Esr1), and progesterone receptor (Pgr) expression in ARC kisspeptin or GABA neurons of female mice at postnatal day (P)4, P8, P12, P20, and P60. A probe that binds to kiss1 mRNA or vGat mRNA was used to produce signal in kisspeptin or GABA neurons, respectively. In adult, we identified that the vast majority of kisspeptin neurons coexpressed Esr1 (95%) and Pgr (93%), while a smaller proportion coexpressed Ar (66%). Similar proportions of Ar- or Esr1-positive kisspeptin neurons were seen from P4, suggesting that kisspeptin neurons develop adult-like sensitivity to androgen and estrogen in early postnatal life. In contrast, the proportion of Pgr-positive kisspeptin cells in early life was significantly lower than in adulthood, suggesting that progesterone sensitivity develops over time in the ARC kisspeptin population. ARC GABA neurons also colocalized with Ar (70%), Esr1 (64%), or Pgr (85%) in adulthood. GABA neurons continuously expressed Esr1 or Pgr from the postnatal stages to adulthood, while the proportion of Ar-positive GABA neurons gradually increased from P4 (24%) to P20 (59%). These results suggest that while ARC GABA neurons can respond to circulating estrogen and progesterone from early postnatal ages, this same population may become more sensitive to androgens during later postnatal life. Our findings identified the expression patterns of Ar, Esr1, and Pgr by ARC kisspeptin and GABA neurons during early postnatal life. These data provide the understanding for the hormone sensitivity of these populations during early postnatal life, the critical time for the formation and regulation of female reproductive physiology.Esr1 (95%) and Pgr (93%), while a smaller proportion coexpressed Ar (66%). Similar proportions of Ar- or Esr1-positive kisspeptin neurons were seen from P4, suggesting that kisspeptin neurons develop adult-like sensitivity to androgen and estrogen in early postnatal life. In contrast, the proportion of Pgr-positive kisspeptin cells in early life was significantly lower than in adulthood, suggesting that progesterone sensitivity develops over time in the ARC kisspeptin population. ARC GABA neurons also colocalized with Ar (70%), Esr1 (64%), or Pgr (85%) in adulthood. GABA neurons continuously expressed Esr1 or Pgr from the postnatal stages to adulthood, while the proportion of Ar-positive GABA neurons gradually increased from P4 (24%) to P20 (59%). These results suggest that while ARC GABA neurons can respond to circulating estrogen and progesterone from early postnatal ages, this same population may become more sensitive to androgens during later postnatal life. Our findings identified the expression patterns of Ar, Esr1, and Pgr by ARC kisspeptin and GABA neurons during early postnatal life. These data provide the understanding for the hormone sensitivity of these populations during early postnatal life, the critical time for the formation and regulation of female reproductive physiology.
Collapse
Affiliation(s)
- Yugo Watanabe
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Department of Biochemistry, School of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Lorryn Fisher
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Christine L Jasoni
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| |
Collapse
|
4
|
Quignon C, Backer A, Kearney J, Bow H, Wray S. Mild Gestational Hypothyroidism in Mice Has Transient Developmental Effects and Long-Term Consequences on Neuroendocrine Systems. Thyroid 2025; 35:97-110. [PMID: 39728609 DOI: 10.1089/thy.2024.0512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Background: Thyroid hormones (TH) play a key role in fetal brain development. While severe thyroid dysfunction, has been shown to cause neurodevelopmental and reproductive disorders, the rising levels of TH-disruptors in the environment in the past few decades have increased the need to assess effects of subclinical (mild) TH insufficiency during gestation. Since embryos do not produce their own TH before mid-gestation, early development processes rely on maternal production. Notably, the reproductive network governed by gonadotropin-releasing hormone (GnRH) neurons develops during this critical period. Methods: The risk of mild maternal hypothyroidism on the development of GnRH neurons and long-term effect on neuroendocrine function in the offspring was investigated using a mouse model of gestational hypothyroidism induced by methimazole (MMI) treatment. Results: MMI treatment during gestation led to reduced litter size, consistent with increased miscarriages due to hypothyroidism. E12/13 embryos, collected from MMI-treated dams, had a decreased number of GnRH neurons, but the migration of the remaining GnRH neurons was normal. Cell proliferation was reduced in the vomeronasal organ (VNO), correlating with the reduced number of GnRH neurons detected in this region. Using a GnRH cell line confirmed attenuated proliferation in the absence of T3. Pups born from hypothyroid mothers had normal postweaning growth and estrus cycles, yet adult offspring had significantly more cells expressing estrogen receptor alpha in the arcuate nucleus. Notably, by adulthood, GnRH cell number and distribution was comparable with nontreated controls indicating that compensatory mechanisms occurred after E13. Conclusion: Overall, our work shows that mild TH disruption during gestation transiently affects proliferation of the pool of GnRH neurons within the VNO and has a long-term impact on neuroendocrine systems.
Collapse
Affiliation(s)
- Clarisse Quignon
- National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Annika Backer
- National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Jessica Kearney
- National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Hannah Bow
- National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Susan Wray
- National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
5
|
Stener-Victorin E, Deng Q. Epigenetic inheritance of PCOS by developmental programming and germline transmission. Trends Endocrinol Metab 2024:S1043-2760(24)00324-2. [PMID: 39732517 DOI: 10.1016/j.tem.2024.12.002] [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: 10/29/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/30/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine and metabolic disorder, affecting approximately 11-13% of women of reproductive age. Women with PCOS experience a higher prevalence of infertility, pregnancy complications, and cardiometabolic disorders such as obesity, insulin resistance, and type 2 diabetes mellitus. Furthermore, psychiatric comorbidities, including depression and anxiety, significantly impact the quality of life in this population. Although obesity exacerbates these health risks, the exact etiology and pathophysiology of PCOS remain complex and only partially understood. Emerging research suggests potential transgenerational inheritance through genetic and epigenetic mechanisms, highlighting the possibility of PCOS-related risks affecting subsequent generations, including sons. This review synthesizes recent findings on PCOS inheritance patterns and underscores areas for future clinical and research exploration.
Collapse
Affiliation(s)
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|
6
|
Yang Q, Jia S, Tao J, Zhang J, Fan Z. Multiple effects of kisspeptin on neuroendocrine, reproduction, and metabolism in polycystic ovary syndrome. J Neuroendocrinol 2024:e13482. [PMID: 39694850 DOI: 10.1111/jne.13482] [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: 06/03/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a highly prevalent and heterogeneous disease characterized by a combination of reproductive and endocrine abnormalities, often associated with metabolic and mental health disorders. The etiology and pathogenesis of PCOS remain unclear, but recent research has increasingly focused on the upstream mechanisms underlying its development. Among these, kisspeptin (KISS) signaling has emerged as a pivotal component in the regulation of the hypothalamic-pituitary-gonadal axis, with significant roles in reproductive function, energy regulation, and metabolism. Women with PCOS commonly exhibit disruptions in gonadotropin secretion, including elevated luteinizing hormone (LH) levels, imbalanced LH/follicle-stimulating hormone (FSH) ratios, and increased androgen levels, all of which are usually parallel with abnormal KISS signaling. Furthermore, alterations in the KISS/KISS1R system within the central and circulatory systems, as well as peripheral tissues, have been implicated in the development of PCOS. These changes affect multiple pathophysiological domains, including reproductive function, energy regulation, metabolic homeostasis, inflammatory response, and emotional disorders, and are further influenced by lifestyle and environmental factors. This review aims to comprehensively summarize the existing experimental and clinical evidence supporting these roles of KISS in PCOS, with the goal of establishing a foundation for future research and potential clinical applications.
Collapse
Affiliation(s)
- Qiaorui Yang
- Department of Gynecology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shengxiao Jia
- Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Jing Tao
- Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Jinfu Zhang
- Department of Gynecology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Gynecology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai, China
| | - Zhenliang Fan
- Nephrology Department, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Zhejiang, China
| |
Collapse
|
7
|
Lafci NG, Yilmaz B, Yildiz BO. PCOS - the many faces of a disorder in women and men. J Endocrinol Invest 2024:10.1007/s40618-024-02512-1. [PMID: 39680364 DOI: 10.1007/s40618-024-02512-1] [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: 12/30/2023] [Accepted: 12/01/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE Polycystic ovary syndrome (PCOS) is a very common endocrine, metabolic and reproductive disorder. The underlying pathophysiology is not yet fully understood and both genetic and environmental factors contribute to its development. We aimed to explore clinical and genetic aspects of familial clustering in PCOS, shedding light on its reproductive and metabolic consequences in both male and female first-degree relatives of the affected women. METHODS Searching the electronic database of PubMed up to October 2023, we synthesized findings from available prospective and retrospective studies and review articles, investigating the familial clustering of PCOS and incorporating data on its metabolic consequences and genetic associations. RESULTS There is a significant clustering of reproductive and metabolic abnormalities in first-degree relatives of women with PCOS. Genetic studies, including genome-wide association studies (GWAS), reveal a complex molecular etiology, emphasizing polygenic architecture. This is supported by the identification of two distinct PCOS subtypes, termed "reproductive" and "metabolic" which exhibit differential genetic underpinnings. CONCLUSION Clinicians should be aware of increased reproductive and metabolic dysfunction both in female and male first-degree relatives of PCOS probands. Current challenges include refining genetic risk scores and understanding the impact of PCOS genetic factors on diverse outcomes, necessitating a sex-specific approach in research and clinical practice. Future directions should address causality, improve diagnostic capability, and unravel the long-term consequences in both genders, emphasizing the importance of proactive clinical assessment in PCOS probands and their families.
Collapse
Affiliation(s)
- Naz Guleray Lafci
- Department of Medical Genetics, Hacettepe University School of Medicine, Ankara, Turkey
| | - Bulent Yilmaz
- Department of Obstetrics and Gynecology, Divison of Reproductive Endocrinology and Infertility, Recep Tayyip Erdoğan University School of Medicine, Rize, Turkey
| | - Bulent Okan Yildiz
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Hacettepe University School of Medicine, Ankara, Turkey.
| |
Collapse
|
8
|
Yıldırım E, Görkem Ü. Association between serum copeptin levels and non-obese normoglycemic polycystic ovary syndrome: A case control study. Turk J Obstet Gynecol 2024; 21:280-285. [PMID: 39663787 PMCID: PMC11635721 DOI: 10.4274/tjod.galenos.2024.65703] [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: 10/04/2024] [Accepted: 11/13/2024] [Indexed: 12/13/2024] Open
Abstract
Objective Copeptin is a glycopeptide that increases under stress and is present in polycystic ovary syndrome (PCOS) patients with metabolic system disorders. We examined the relationship between copeptin and reproductive function in patients with normoglycemic PCOS with anovulatory cycles and normal weight. Materials and Methods Women with unexplained infertility (n=52) and women with PCOS (n=57) were included in the study. PCOS was determined using the Rotterdam criteria. Biochemical tests including estradiol, follicle-stimulating hormone, luteinizing hormone, anti-Müllerian hormone (AMH), insulin, and copeptin were performed. Serum copeptin concentrations were measured using enzyme immunoassay. Results There were no significant differences in demographic data, insulin levels, and insulin resistance between the PCOS and healthy volunteers. Copeptin levels were lower in the PCOS group (p<0.001). A significant negative correlation was observed between AMH and copeptin in the control group (r=-0.402, p= 0.013). In the PCOS group, a negative correlation was observed between antral follicle count and copeptin, as well as between AMH and copeptin (r=-0.544, p<0.01). Receiver operating characteristic (ROC) curve analysis was performed to determine the predictive value of copeptin levels. The estimated areas under the ROC curves for serum concentration were found to be statistically significant (p<0.001) with a cut-off value of 2.78 (95% confidence interval 0.701-0.896), sensitivity of 0.87, and specificity of 0.70. Conclusion This study showed that copeptin levels are lower in patients with PCOS in the absence of insulin resistance and obesity than in healthy volunteers, and there is a negative correlation between copeptin and reproductive markers.
Collapse
Affiliation(s)
- Engin Yıldırım
- Malatya Turgut Özal University Faculty of Medicine, Department of Obstetrics and Gynecology, Malatya, Turkey
| | - Ümit Görkem
- Hitit University Faculty of Medicine, Department of Obstetrics and Gynecology, Çorum, Turkey
| |
Collapse
|
9
|
Wang D, Zhang M, Wang WS, Chu W, Zhai J, Sun Y, Chen ZJ, Du Y. Decreased neurotensin induces ovulatory dysfunction via the NTSR1/ERK/EGR1 axis in polycystic ovary syndrome. Front Med 2024:10.1007/s11684-024-1089-z. [PMID: 39648233 DOI: 10.1007/s11684-024-1089-z] [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: 02/09/2024] [Accepted: 05/19/2024] [Indexed: 12/10/2024]
Abstract
Polycystic ovary syndrome (PCOS) is the predominant cause of subfertility in reproductive-aged women; however, its pathophysiology remains unknown. Neurotensin (NTS) is a member of the gut-brain peptide family and is involved in ovulation; its relationship with PCOS is unclear. Here, we found that NTS expression in ovarian granulosa cells and follicular fluids was markedly decreased in patients with PCOS. In the in vitro culture of cumulus-oocyte complexes, the neurotensin receptor 1 (NTSR1) antagonist SR48692 blocked cumulus expansion and oocyte meiotic maturation by inhibiting metabolic cooperation and damaging the mitochondrial structure in oocytes and surrounding cumulus cells. Furthermore, the ERK1/2-early growth response 1 pathway was found to be a key downstream mediator of NTS/NTSR1 in the ovulatory process. Animal studies showed that in vivo injection of SR48692 in mice reduced ovulation efficiency and contributed to irregular estrus cycles and polycystic ovary morphology. By contrast, NTS partially ameliorated the ovarian abnormalities in mice with dehydroepiandrosterone-induced PCOS. Our findings highlighted the critical role of NTS reduction and consequent abnormal NTSR1 signaling in the ovulatory dysfunction of PCOS, suggesting a potential strategy for PCOS treatment.
Collapse
Affiliation(s)
- Dongshuang Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Meiling Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Wang-Sheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Weiwei Chu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Junyu Zhai
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Yun Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory for Reproductive Endocrinology of Ministry of Education, Shandong Provincial Key Laboratory of Reproductive Medicine, Center for Reproductive Medicine, Shandong Provincial Hospital, Shandong University, Jinan, 250012, China.
| | - Yanzhi Du
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China.
| |
Collapse
|
10
|
Ren C, Zhang S, Ma J, Huang J, Huang P, Qu M, Zhao H, Zhou Z, Gong A. Nicotinamide Mononucleotide Alleviates Bile Acid Metabolism and Hormonal Dysregulation in Letrozole-Induced PCOS Mice. BIOLOGY 2024; 13:1028. [PMID: 39765695 PMCID: PMC11673032 DOI: 10.3390/biology13121028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/20/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025]
Abstract
Polycystic ovary syndrome (PCOS) involves complex genetic, metabolic, endocrine, and environmental factors. This study explores the effects of nicotinamide mononucleotide (NMN) in a letrozole-induced PCOS mouse model, focusing on metabolic regulation. Letrozole-induced aromatase inhibition elevated androgen and reduced bile acid levels, linking liver dysfunction and gut imbalance to PCOS. Letrozole-treated mice exhibited disrupted estrous cycles, ovarian congestion, and elevated testosterone. NMN intervention alleviated hyperandrogenism, ovarian abnormalities, and bile acid decline but did not fully restore the estrous cycle or improve lipid profiles. Metabolomic analysis showed that NMN partially reversed bile acid and lipid metabolism disturbances. These findings highlight NMN's protective role in reducing hyperandrogenism and ovarian cyst formation. However, effective PCOS treatment should target liver and gut metabolism, not just ovarian symptoms, to mitigate systemic effects. Bile acid dysregulation may play a key role in PCOS progression and warrants further investigation.
Collapse
Affiliation(s)
- Caifang Ren
- School of Medicine, Jiangsu University, Zhenjiang 212013, China; (S.Z.); (J.H.); (P.H.); (M.Q.); (H.Z.); (Z.Z.)
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China;
| | - Shuang Zhang
- School of Medicine, Jiangsu University, Zhenjiang 212013, China; (S.Z.); (J.H.); (P.H.); (M.Q.); (H.Z.); (Z.Z.)
| | - Jianyu Ma
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China;
| | - Junjie Huang
- School of Medicine, Jiangsu University, Zhenjiang 212013, China; (S.Z.); (J.H.); (P.H.); (M.Q.); (H.Z.); (Z.Z.)
| | - Pan Huang
- School of Medicine, Jiangsu University, Zhenjiang 212013, China; (S.Z.); (J.H.); (P.H.); (M.Q.); (H.Z.); (Z.Z.)
| | - Mingzi Qu
- School of Medicine, Jiangsu University, Zhenjiang 212013, China; (S.Z.); (J.H.); (P.H.); (M.Q.); (H.Z.); (Z.Z.)
| | - Haoyue Zhao
- School of Medicine, Jiangsu University, Zhenjiang 212013, China; (S.Z.); (J.H.); (P.H.); (M.Q.); (H.Z.); (Z.Z.)
| | - Zhengrong Zhou
- School of Medicine, Jiangsu University, Zhenjiang 212013, China; (S.Z.); (J.H.); (P.H.); (M.Q.); (H.Z.); (Z.Z.)
| | - Aihua Gong
- School of Medicine, Jiangsu University, Zhenjiang 212013, China; (S.Z.); (J.H.); (P.H.); (M.Q.); (H.Z.); (Z.Z.)
- Hematological Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212003, China
| |
Collapse
|
11
|
Zhang H, Ye Y, Zhao Y, Li S, Jiao P, Yang Y, Jin Y, Zeng L, Zhang H, Chen M, Jiang H, Zhou L, Li J, Li D, Li R. Obesity is associated with lower levels of negative emotions in polycystic ovary syndrome in clinical and animal studies. Ann Med 2024; 56:2373199. [PMID: 38956857 PMCID: PMC11225633 DOI: 10.1080/07853890.2024.2373199] [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: 04/18/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is one of the most common endocrine and metabolic disorders in women of reproductive age. It is frequently comorbid with obesity and negative emotions. Currently, there are few reports on the relationship between obesity and negative emotions in patients with PCOS. Here we performed both basic and clinical studies to study the relationship between obesity and negative emotions in PCOS. METHODS We performed a cross-sectional study including 608 patients with PCOS and 184 healthy participants to assess the mental health status of people with different body mass indices (BMI). Self-rated anxiety, depression, and perceived stress scales were used for subjective mood evaluations. Rat PCOS models fed 45 and 60% high-fat diets were used to confirm the results of the clinical study. Elevated plus maze and open field tests were used to assess anxiety- and depression-like behaviors in rats. RESULTS We observed overweight/obesity, increased depression, anxiety, and perceived stress in women with PCOS, and found that anxiety and depression were negatively correlated with BMI in patients with severe obesity and PCOS. Similar results were confirmed in the animal study; the elevated plus maze test and open field test demonstrated that only 60% of high fat diet-induced obesity partly reversed anxiety- and depression-like behaviors in PCOS rats. A high-fat diet also modulated rat hypothalamic and hippocampal luteinizing hormone and testosterone levels. CONCLUSION These results reveal a potential relationship between obesity and negative emotions in PCOS and prompt further investigation. The interactions between various symptoms of PCOS may be targeted to improve the overall well-being of patients.
Collapse
Affiliation(s)
- Haolin Zhang
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yang Ye
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Yue Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Shi Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Peijie Jiao
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yang Yang
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Yuxin Jin
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Lin Zeng
- Research Centre of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | - Hua Zhang
- Research Centre of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | - Meishuang Chen
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Hong Jiang
- Department of Neurobiology, School of Basic Medical Sciences, Neuroscience Research Institute, Peking University, Beijing, China
| | - Lifei Zhou
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Jiayi Li
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Dong Li
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Rong Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| |
Collapse
|
12
|
Palomba S, Costanzi F, Caserta D, Vitagliano A. Pharmacological and non-pharmacological interventions for improving endometrial receptivity in infertile patients with polycystic ovary syndrome: a comprehensive review of the available evidence. Reprod Biomed Online 2024; 49:104381. [PMID: 39454320 DOI: 10.1016/j.rbmo.2024.104381] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 10/28/2024]
Abstract
Direct and indirect evidence suggests that endometrial receptivity may play a crucial role in the reduced fertility rate of women with polycystic ovary syndrome (PCOS). Various pharmacological and non-pharmacological strategies with potential effects on endometrial receptivity in patients with PCOS have been proposed. The aim of this study was to summarize the rationale and the clinical and experimental evidence of interventions tested for improving endometrial receptivity in infertile patients with PCOS. A systematic review was conducted by consulting electronic databases. All interventions with a potential influence on endometrial receptivity in infertile patients with PCOS were evaluated, and their main biological mechanisms were analysed. In total, 24 interventions related to endometrial receptivity were identified. Notwithstanding a strong biological rationale, no intervention aimed at improving endometrial receptivity in women with PCOS is supported by an adequate body of evidence, limiting their use in clinical practice. Further high-quality research is needed in this field to limit potentially ineffective and unsafe add-on treatments in infertile patients with PCOS.
Collapse
Affiliation(s)
- Stefano Palomba
- Unit of Gynaecology, Department of Medical-Surgical Sciences and Translational Medicine, University 'Sapienza' of Rome, Sant'Andrea Hospital, Rome, Italy.
| | - Flavia Costanzi
- Unit of Gynaecology, Department of Medical-Surgical Sciences and Translational Medicine, University 'Sapienza' of Rome, Sant'Andrea Hospital, Rome, Italy; University 'Sapienza' of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Donatella Caserta
- Unit of Gynaecology, Department of Medical-Surgical Sciences and Translational Medicine, University 'Sapienza' of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Amerigo Vitagliano
- Unit of Obstetrics and Gynaecology, Department of Interdisciplinary Medicine, University of Bari, Bari, Italy
| |
Collapse
|
13
|
Dou Y, Zhang M, Zhang H, Zhang C, Feng L, Hu J, Gao Y, Yuan XZ, Zhao Y, Zhao H, Chen ZJ. Lactating exposure to microplastics at the dose of infants ingested during artificial feeding induced reproductive toxicity in female mice and their offspring. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 949:174972. [PMID: 39053555 DOI: 10.1016/j.scitotenv.2024.174972] [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: 05/02/2024] [Revised: 07/17/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024]
Abstract
Microplastics (MPs) pollution poses a global environmental challenge with significant concerns regarding its potential impact on human health. Toxicological investigations have revealed multi-system impairments caused by MPs in various organisms. However, the specific reproductive hazards in human contexts remain elusive, and understanding the transgenerational reproductive toxicity of MPs remains limited. This study delves into the reproductive toxicity resulting from lactational exposure to polystyrene MPs (PS-MPs) in female mice, extending the inquiry to assess the reproductive effects on their offspring bred by rigorous natural mating. The MPs dosage corresponds to the detected concentration in infant formula prepared using plastic bottles. By systematically evaluating the reproductive phenotypes of F0 female mice from birth to adulthood, we found that female mice exposed to PS-MPs exhibited delayed puberty, disturbed estrous cyclicity, diminished fertility, elevated testosterone, abnormal follicle development, disrupted ovarian steroidogenesis, and ovarian inflammation. Importantly, the observed inheritable reproductive toxicity manifested with gender specificity, showcasing more pronounced abnormalities in male offspring. Specifically, reproductive disorders did not manifest in female offspring; however, a significant decrease in sperm count and viability was observed in PS-MPs-exposed F1 males. Testicular transcriptomics analysis of F1 males significantly enriched pathways associated with reproductive system development and epigenetic modification, such as male germ cell proliferation, DNA methylation, and histone modification. In summary, real-life exposure to PS-MPs impaired the reproductive function of female mice and threateningly disrupted the spermatogenesis of their F1 male offspring, which raises serious concerns about inter- and trans-generational reproductive toxicities of MPs in mammals. These findings underscore the potential threats of MPs to human reproductive health, emphasizing the need for continued vigilance and research in this critical area.
Collapse
Affiliation(s)
- Yunde Dou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China; Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Mengge Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
| | - Honghui Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China; The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China; Suzhou Municipal Hospital, Suzhou, China; Gusu School, Nanjing Medical University, Suzhou, China
| | - Changlong Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
| | - Lijuan Feng
- College of Geography and Environment, Shandong Normal University, Jinan, Shandong, China
| | - Jingmei Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
| | - Yuan Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
| | - Xian-Zheng Yuan
- Shandong Key Laboratory of Water Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Shandong University, Qingdao, Shandong, China
| | - Yueran Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong 250012, China
| | - Han Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong 250012, China
| | - Zi-Jiang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong 250012, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China; Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
14
|
Quan K, Ning S, You Z, Deng G. Bioinformatics analysis of the role of RNA modification regulators in polycystic ovary syndrome. Heliyon 2024; 10:e36706. [PMID: 39281527 PMCID: PMC11400967 DOI: 10.1016/j.heliyon.2024.e36706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/18/2024] Open
Abstract
Purpose Polycystic ovary syndrome (PCOS) is the most common metabolic and endocrine disorder affecting women of reproductive age. The pathogenesis of PCOS is influenced by factors such as race, genetics, environment, hyperandrogenemia, hyperinsulinemia, and obesity. However, the molecular mechanisms linking RNA modification and PCOS remain underexplored. This study aims to investigate the potential genetic and molecular pathways connecting RNA modification with PCOS through bioinformatics analyses. Methods The GSE34526, GSE5850, and GSE98421 datasets were obtained from the National Center for Biotechnology Information Gene Expression Omnibus database. We identified intersecting differentially expressed genes (DEGs) and RNA modification-related genes within the GSE34526 dataset and visualized the overlaps using a Venn diagram. Subsequent analyses included Gene Ontology (GO), pathway enrichment (Kyoto Encyclopedia of Genes and Genomes), gene set enrichment analysis (GSEA), gene set variation analysis (GSVA), and immune infiltration analysis. Additionally, we constructed a protein-protein interaction network as well as mRNA-miRNA, mRNA-RNA binding protein, and mRNA-transcription factor (TF) regulatory networks. The expression and receiver operating characteristic curves of hub genes were also identified. Results The expression of several RNA modification-related DEGs (RMRDEGs) (ALYREF, NUDT1, AGO2, TET2, YTHDF2, and TRMT61B) showed significant differences in PCOS patients. GSEA and GSVA indicated that RMRDEGs were enriched in the hedgehog, MAPK, JAK STAT, and Notch pathways. Key transcription factors, including SP7, KLF8, HCFC1, IRF1, and MLLT1, were identified in the TF regulatory networks. Conclusions These findings suggest that there are gene and miRNA profile alterations exist in PCOS patients and highlight immune-related differences. This knowledge could pave the way for new research directions in the diagnosis and treatment of PCOS.
Collapse
Affiliation(s)
- Kewei Quan
- Department of Obstetrics and Gynecology, Dongguan Hospital of Guangzhou University of Chinese Medicine, Dongguan, 523000, China
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Shuting Ning
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Zilin You
- Department of Obstetrics and Gynecology, Dongguan Hospital of Guangzhou University of Chinese Medicine, Dongguan, 523000, China
| | - Gaopi Deng
- Department of Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510080, Guangdong Province, China
| |
Collapse
|
15
|
Coutinho EA, Esparza LA, Rodriguez J, Yang J, Schafer D, Kauffman AS. Targeted inhibition of kisspeptin neurons reverses hyperandrogenemia and abnormal hyperactive LH secretion in a preclinical mouse model of polycystic ovary syndrome. Hum Reprod 2024; 39:2089-2103. [PMID: 38978296 PMCID: PMC11373419 DOI: 10.1093/humrep/deae153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/11/2024] [Indexed: 07/10/2024] Open
Abstract
STUDY QUESTION Do hyperactive kisspeptin neurons contribute to abnormally high LH secretion and downstream hyperandrogenemia in polycystic ovary syndrome (PCOS)-like conditions and can inhibition of kisspeptin neurons rescue such endocrine impairments? SUMMARY ANSWER Targeted inhibition of endogenous kisspeptin neuron activity in a mouse model of PCOS reduced the abnormally hyperactive LH pulse secretion and hyperandrogenemia to healthy control levels. WHAT IS KNOWN ALREADY PCOS is a reproductive disorder characterized by hyperandrogenemia, anovulation, and/or polycystic ovaries, along with a hallmark feature of abnormal LH hyper-pulsatility, but the mechanisms underlying the endocrine impairments remain unclear. A chronic letrozole (LET; aromatase inhibitor) mouse model recapitulates PCOS phenotypes, including polycystic ovaries, anovulation, high testosterone, and hyperactive LH pulses. LET PCOS-like females also have increased hypothalamic kisspeptin neuronal activation which may drive their hyperactive LH secretion and hyperandrogenemia, but this has not been tested. STUDY DESIGN, SIZE, DURATION Transgenic KissCRE+/hM4Di female mice or littermates Cre- controls were treated with placebo, or chronic LET (50 µg/day) to induce a PCOS-like phenotype, followed by acute (once) or chronic (2 weeks) clozapine-N-oxide (CNO) exposure to chemogenetically inhibit kisspeptin cells (n = 6 to 10 mice/group). PARTICIPANTS/MATERIALS, SETTING, METHODS Key endocrine measures, including in vivo LH pulse secretion patterns and circulating testosterone levels, were assessed before and after selective kisspeptin neuron inhibition and compared between PCOS groups and healthy controls. Alterations in body weights were measured and pituitary and ovarian gene expression was determined by qRT-PCR. MAIN RESULTS AND THE ROLE OF CHANCE Acute targeted inhibition of kisspeptin neurons in PCOS mice successfully lowered the abnormally hyperactive LH pulse secretion (P < 0.05). Likewise, chronic selective suppression of kisspeptin neuron activity reversed the previously high LH and testosterone levels (P < 0.05) down to healthy control levels and rescued reproductive gene expression (P < 0. 05). LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Ovarian morphology was not assessed in this study. Additionally, mouse models can offer mechanistic insights into neuroendocrine processes in PCOS-like conditions but may not perfectly mirror PCOS in women. WIDER IMPLICATIONS OF THE FINDINGS These data support the hypothesis that overactive kisspeptin neurons can drive neuroendocrine PCOS-like impairments, and this may occur in PCOS women. Our findings complement recent clinical investigations using NKB receptor antagonists to lower LH in PCOS women and suggest that pharmacological dose-dependent modulation of kisspeptin neuron activity may be a valuable future therapeutic target to clinically treat hyperandrogenism and lower elevated LH in PCOS women. STUDY FUNDING/COMPETING INTEREST(S) This research was supported by NIH grants R01 HD111650, R01 HD090161, R01 HD100580, P50 HD012303, R01 AG078185, and NIH R24 HD102061, and a pilot project award from the British Society for Neuroendocrinology. There are no competing interests.
Collapse
Affiliation(s)
- Eulalia A Coutinho
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Lourdes A Esparza
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Julian Rodriguez
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jason Yang
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Danielle Schafer
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Alexander S Kauffman
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
16
|
Bazzano MV, Köninger A, Solano ME. Beyond defence: Immune architects of ovarian health and disease. Semin Immunopathol 2024; 46:11. [PMID: 39134914 PMCID: PMC11319434 DOI: 10.1007/s00281-024-01021-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/23/2024] [Indexed: 08/15/2024]
Abstract
Throughout the individual's reproductive period of life the ovary undergoes continues changes, including cyclic processes of cell death, tissue regeneration, proliferation, and vascularization. Tissue-resident leucocytes particularly macrophages, play a crucial role in shaping ovarian function and maintaining homeostasis. Macrophages crucially promote angiogenesis in the follicles and corpora lutea, thereby supporting steroidogenesis. Recent research on macrophage origins and early tissue seeding has unveiled significant insights into their role in early organogenesis, e.g. in the testis. Here, we review evidence about the prenatal ovarian seeding of leucocytes, primarily macrophages with angiogenic profiles, and its connection to gametogenesis. In the prenatal ovary, germ cells proliferate, form cysts, and undergo changes that, following waves of apoptosis, give rice to the oocytes contained in primordial follicles. These follicles constitute the ovarian reserve that lasts throughout the female's reproductive life. Simultaneously, yolk-sac-derived primitive macrophages colonizing the early ovary are gradually replaced or outnumbered by monocyte-derived fetal macrophages. However, the cues indicating how macrophage colonization and follicle assembly are related are elusive. Macrophages may contribute to organogenesis by promoting early vasculogenesis. Whether macrophages contribute to ovarian lymphangiogenesis or innervation is still unknown. Ovarian organogenesis and gametogenesis are vulnerable to prenatal insults, potentially programming dysfunction in later life, as observed in polycystic ovary syndrome. Experimental and, more sparsely, epidemiological evidence suggest that adverse stimuli during pregnancy can program defective folliculogenesis or a diminished follicle reserve in the offspring. While the ovary is highly sensitive to inflammation, the involvement of local immune responses in programming ovarian health and disease remains to be thoroughly investigated.
Collapse
Affiliation(s)
- Maria Victoria Bazzano
- Laboratory of Translational Perinatology, University of Regensburg, Biopark 1-3, D-93053, Regensburg, Germany
| | - Angela Köninger
- University Department of Obstetrics and Gynecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstr. 1-3, D-93049, Regensburg, Germany
| | - Maria Emilia Solano
- Laboratory of Translational Perinatology, University of Regensburg, Biopark 1-3, D-93053, Regensburg, Germany.
| |
Collapse
|
17
|
Dou Y, Zhao R, Wu H, Yu Z, Yin C, Yang J, Yang C, Luan X, Cheng Y, Huang T, Bian Y, Han S, Zhang Y, Xu X, Chen ZJ, Zhao H, Zhao S. DENND1A desensitizes granulosa cells to FSH by arresting intracellular FSHR transportation. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1620-1634. [PMID: 38709439 DOI: 10.1007/s11427-023-2438-4] [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: 07/11/2023] [Accepted: 11/27/2023] [Indexed: 05/07/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a complex disorder. Genome-wide association studies (GWAS) have identified several genes associated with this condition, including DENND1A. DENND1A encodes a clathrin-binding protein that functions as a guanine nucleotide exchange factor involved in vesicular transport. However, the specific role of DENND1A in reproductive hormone abnormalities and follicle development disorders in PCOS remain poorly understood. In this study, we investigated DENND1A expression in ovarian granulosa cells (GCs) from PCOS patients and its correlation with hormones. Our results revealed an upregulation of DENND1A expression in GCs from PCOS cases, which was positively correlated with testosterone levels. To further explore the functional implications of DENND1A, we generated a transgenic mouse model overexpressing Dennd1a (TG mice). These TG mice exhibited subfertility, irregular estrous cycles, and increased testosterone production following PMSG stimulation. Additionally, the TG mice displayed diminished responsiveness to FSH, characterized by smaller ovary size, less well-developed follicles, and abnormal expressions of FSH-priming genes. Mechanistically, we found that Dennd1a overexpression disrupted the intracellular trafficking of follicle stimulating hormone receptor (FSHR), promoting its internalization and inhibiting recycling. These findings shed light on the reproductive role of DENND1A and uncover the underlying mechanisms, thereby contributing valuable insights into the pathogenesis of PCOS and providing potential avenues for drug design in PCOS treatment.
Collapse
Affiliation(s)
- Yunde Dou
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Rusong Zhao
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, 215008, China
- Gusu School, Nanjing Medical University, Suzhou, 215000, China
| | - Han Wu
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Zhiheng Yu
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Changjian Yin
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Jie Yang
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Chaoyan Yang
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Xiaohua Luan
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Yixiao Cheng
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Tao Huang
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Yuehong Bian
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Shan Han
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, 250012, China
| | - Yuqing Zhang
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Xin Xu
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Zi-Jiang Chen
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, 250012, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200127, China
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Han Zhao
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China.
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China.
| | - Shigang Zhao
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, 250012, China.
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong University, Jinan, 250012, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China.
| |
Collapse
|
18
|
Sobczuk J, Paczkowska K, Andrusiów S, Bolanowski M, Daroszewski J. Are Women with Polycystic Ovary Syndrome at Increased Risk of Alzheimer Disease? Lessons from Insulin Resistance, Tryptophan and Gonadotropin Disturbances and Their Link with Amyloid-Beta Aggregation. Biomolecules 2024; 14:918. [PMID: 39199306 PMCID: PMC11352735 DOI: 10.3390/biom14080918] [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: 06/29/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
Alzheimer disease, the leading cause of dementia, and polycystic ovary syndrome, one of the most prevalent female endocrine disorders, appear to be unrelated conditions. However, studies show that both disease entities have common risk factors, and the amount of certain protein marker of neurodegeneration is increased in PCOS. Reports on the pathomechanism of both diseases point to the possibility of common denominators linking them. Dysregulation of the kynurenine pathway, insulin resistance, and impairment of the hypothalamic-pituitary-gonadal axis, which are correlated with amyloid-beta aggregation are these common areas. This article discusses the relationship between Alzheimer disease and polycystic ovary syndrome, with a particular focus on the role of disorders of tryptophan metabolism in both conditions. Based on a review of the available literature, we concluded that systemic changes occurring in PCOS influence the increased risk of neurodegeneration.
Collapse
Affiliation(s)
- Joachim Sobczuk
- Department of Endocrinology, Diabetes and Isotope Therapy, University Clinical Hospital, 50-367 Wroclaw, Poland
| | | | - Szymon Andrusiów
- Department of Neurology, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Marek Bolanowski
- Department of Endocrinology, Diabetes and Isotope Therapy, University Clinical Hospital, 50-367 Wroclaw, Poland
- Department of Endocrinology, Diabetes and Isotope Therapy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Jacek Daroszewski
- Department of Endocrinology, Diabetes and Isotope Therapy, University Clinical Hospital, 50-367 Wroclaw, Poland
- Department of Endocrinology, Diabetes and Isotope Therapy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
19
|
Oliveira VEDM, Evrard F, Faure MC, Bakker J. Social isolation and aggression training lead to escalated aggression and hypothalamus-pituitary-gonad axis hyperfunction in mice. Neuropsychopharmacology 2024; 49:1266-1275. [PMID: 38337026 PMCID: PMC11224373 DOI: 10.1038/s41386-024-01808-3] [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: 11/24/2023] [Revised: 12/30/2023] [Accepted: 01/17/2024] [Indexed: 02/12/2024]
Abstract
Although the participation of sex hormones and sex hormone-responsive neurons in aggressive behavior has been extensively studied, the role of other systems within the hypothalamus-pituitary-gonadal (HPG) axis remains elusive. Here we assessed how the gonadotropin-releasing hormone (GnRH) and kisspeptin systems are impacted by escalated aggression in male mice. We used a combination of social isolation and aggression training (IST) to exacerbate mice's aggressive behavior. Next, low-aggressive (group-housed, GH) and highly aggressive (IST) mice were compared regarding neuronal activity in the target populations and hormonal levels, using immunohistochemistry and ELISA, respectively. Finally, we used pharmacological and viral approaches to manipulate neuropeptide signaling and expression, subsequently evaluating its effects on behavior. IST mice exhibited enhanced aggressive behavior compared to GH controls, which was accompanied by elevated neuronal activity in GnRH neurons and arcuate nucleus kisspeptin neurons. Remarkably, IST mice presented an increased number of kisspeptin neurons in the anteroventral periventricular nucleus (AVPV). In addition, IST mice exhibited elevated levels of luteinizing hormone (LH) in serum. Accordingly, activation and blockade of GnRH receptors (GnRHR) exacerbated and reduced aggression, respectively. Surprisingly, kisspeptin had intricate effects on aggression, i.e., viral ablation of AVPV-kisspeptin neurons impaired the training-induced rise in aggressive behavior whereas kisspeptin itself strongly reduced aggression in IST mice. Our results indicate that IST enhances aggressive behavior in male mice by exacerbating HPG-axis activity. Particularly, increased GnRH neuron activity and GnRHR signaling were found to underlie aggression whereas the relationship with kisspeptin remains puzzling.
Collapse
Affiliation(s)
- Vinícius Elias de Moura Oliveira
- Laboratory of Neuroendocrinology, GIGA-Neurosciences, University of Liege, 4000, Liege, Belgium.
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128, Mainz, Germany.
| | - Florence Evrard
- Laboratory of Neuroendocrinology, GIGA-Neurosciences, University of Liege, 4000, Liege, Belgium
| | - Melanie C Faure
- Laboratory of Neuroendocrinology, GIGA-Neurosciences, University of Liege, 4000, Liege, Belgium
| | - Julie Bakker
- Laboratory of Neuroendocrinology, GIGA-Neurosciences, University of Liege, 4000, Liege, Belgium.
| |
Collapse
|
20
|
Vatier C, Christin-Maitre S. Epigenetic/circadian clocks and PCOS. Hum Reprod 2024; 39:1167-1175. [PMID: 38600622 DOI: 10.1093/humrep/deae066] [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: 12/13/2023] [Revised: 03/04/2024] [Indexed: 04/12/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) affects 6-20% of reproductive-aged women. It is associated with increased risks of metabolic syndrome, Type 2 diabetes, cardiovascular diseases, mood disorders, endometrial cancer and non-alcoholic fatty liver disease. Although various susceptibility loci have been identified through genetic studies, they account for ∼10% of PCOS heritability. Therefore, the etiology of PCOS remains unclear. This review explores the role of epigenetic changes and modifications in circadian clock genes as potential contributors to PCOS pathogenesis. Epigenetic alterations, such as DNA methylation, histone modifications, and non-coding RNA changes, have been described in diseases related to PCOS, such as diabetes, cardiovascular diseases, and obesity. Furthermore, several animal models have illustrated a link between prenatal exposure to androgens or anti-Müllerian hormone and PCOS-like phenotypes in subsequent generations, illustrating an epigenetic programming in PCOS. In humans, epigenetic changes have been reported in peripheral blood mononuclear cells (PBMC), adipose tissue, granulosa cells (GC), and liver from women with PCOS. The genome of women with PCOS is globally hypomethylated compared to healthy controls. However, specific hypomethylated or hypermethylated genes have been reported in the different tissues of these women. They are mainly involved in hormonal regulation and inflammatory pathways, as well as lipid and glucose metabolism. Additionally, sleep disorders are present in women with PCOS and disruptions in clock genes' expression patterns have been observed in their PBMC or GCs. While epigenetic changes hold promise as diagnostic biomarkers, the current challenge lies in distinguishing whether these changes are causes or consequences of PCOS. Targeting epigenetic modifications potentially opens avenues for precision medicine in PCOS, including lifestyle interventions and drug therapies. However, data are still lacking in large cohorts of well-characterized PCOS phenotypes. In conclusion, understanding the interplay between genetics, epigenetics, and circadian rhythms may provide valuable insights for early diagnosis and therapeutic strategies in PCOS in the future.
Collapse
Affiliation(s)
- Camille Vatier
- Department of Endocrine and Reproductive Medicine, Center of Endocrine Rare Diseases of Growth and Development (CRESCENDO), FIRENDO, Endo-ERN, Hôpital Saint-Antoine, Assistance-Publique-Hôpitaux de Paris, Sorbonne University, Paris, France
- Institut National de la Santé et de la Recherche Medicale (INSERM) UMR 938, Centre de Recherche Saint-Antoine et Institut de Cardio-Métabolisme et Nutrition (ICAN), Paris, France
| | - Sophie Christin-Maitre
- Department of Endocrine and Reproductive Medicine, Center of Endocrine Rare Diseases of Growth and Development (CRESCENDO), FIRENDO, Endo-ERN, Hôpital Saint-Antoine, Assistance-Publique-Hôpitaux de Paris, Sorbonne University, Paris, France
- INSERM UMR U933, Paris, France
| |
Collapse
|
21
|
Serra L, Estienne A, Bongrani A, Ramé C, Caria G, Froger C, Jolivet C, Henriot A, Amalric L, Corbin E, Guérif F, Froment P, Dupont J. The epoxiconazole and tebuconazole fungicides impair granulosa cells functions partly through the aryl hydrocarbon receptor (AHR) signalling with contrasted effects in obese, normo-weight and polycystic ovarian syndrome (PCOS) patients. Toxicol Rep 2024; 12:65-81. [PMID: 38259722 PMCID: PMC10801249 DOI: 10.1016/j.toxrep.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/24/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
Polycystic ovarian syndrome (PCOS), frequently associated to obesity, is the main reproductive disorder in women in age to procreate. Some evidence suggests that pesticides can result in alterations of the female reproductive system, including polycystic ovary syndrome (PCOS). Here, we detected two fungicides, Tebuconazole (Tb) and Epoxiconazole (Epox) in the soils and waters of French area. Our hypothesis is that these two triazoles could be associated to the etiology of PCOS. We used the human KGN cell line and primary human granulosa cells (hGCs) from different group of patients: normal weight non PCOS (NW), normal weight PCOS (PCOS NW), obese (obese) and obese PCOS (PCOS obese). We exposed in vitro these cells to Tb and Epox from 0 up to 10 mM for 24 and 48 h and analysed cell viability and steroidogenesis. In hGCs NW, cell viability was reduced from 12.5 µM for Tb and 75 µM for Epox. In hGCs NW, Epox decreased progesterone (Pg) and estradiol (E2) secretions and inhibited STAR, HSD3B and CYP19A1 mRNA expressions from 25 µM and increased AHR mRNA expression from 75 µM. Tb exposure also reduced steroid secretion and STAR and CYP19A1 mRNA expressions and increased AHR mRNA expression but at cytotoxic concentrations. Silencing of AHR in KGN cells reduced inhibitory effects of Tb and Epox on steroid secretion. Tb and Epox exposure decreased more steroid secretion in hGCs from obese, PCOS NW and PCOS obese groups than in NW group. Moreover, we found a higher gene expression of AHR within these three groups. Taken together, both Epox and Tb reduced steroidogenesis in hGCs through partly AHR and Tb was more cytotoxic than Epox. These triazoles alter more strongly PCOS and/or obese hGCs suggesting that human with reproductive disorders are more sensitive to triazoles exposure.
Collapse
Affiliation(s)
- Loise Serra
- CNRS, IFCE, INRAE, University of Tours, PRC, F-37380 Nouzilly, France
| | - Anthony Estienne
- CNRS, IFCE, INRAE, University of Tours, PRC, F-37380 Nouzilly, France
| | - Alice Bongrani
- CNRS, IFCE, INRAE, University of Tours, PRC, F-37380 Nouzilly, France
| | - Christelle Ramé
- CNRS, IFCE, INRAE, University of Tours, PRC, F-37380 Nouzilly, France
| | - Giovanni Caria
- INRAE, Laboratoire d'Analyses des Sols, 273, rue de Cambrai, 62000 Arras, France
| | - Claire Froger
- INRAE Orléans - US 1106, Unité INFOSOL, Orléans, France
| | | | - Abel Henriot
- Division Laboratoires, BRGM, 3 Avenue Claude Guillemin, 45060 Orleans Cedex 2, France
| | - Laurence Amalric
- Division Laboratoires, BRGM, 3 Avenue Claude Guillemin, 45060 Orleans Cedex 2, France
| | - Emilie Corbin
- CNRS, IFCE, INRAE, University of Tours, PRC, F-37380 Nouzilly, France
| | - Fabrice Guérif
- Service de Médecine et Biologie de la Reproduction, CHRU de Tours, F-37044 Tours, France
| | - Pascal Froment
- CNRS, IFCE, INRAE, University of Tours, PRC, F-37380 Nouzilly, France
| | - Joëlle Dupont
- CNRS, IFCE, INRAE, University of Tours, PRC, F-37380 Nouzilly, France
| |
Collapse
|
22
|
Azumah R, Hummitzsch K, Anderson RA, Rodgers RJ. Expression of transforming growth factor β signalling molecules and their correlations with genes in loci linked to polycystic ovary syndrome in human foetal and adult tissues. Reprod Fertil Dev 2024; 36:RD23174. [PMID: 38894494 DOI: 10.1071/rd23174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Context Altered signalling of androgens, anti-Müllerian hormone or transforming growth factor beta (TGFβ) during foetal development have been implicated in the predisposition to polycystic ovary syndrome (PCOS) in later life, aside from its genetic predisposition. In foetal ovarian fibroblasts, TGFβ1 has been shown to regulate androgen signalling and seven genes located in loci associated with PCOS. Since PCOS exhibits a myriad of symptoms, it likely involves many different organs. Aims To identify the relationships between TGFβ signalling molecules and PCOS candidate genes in different tissues associated with PCOS. Methods Using RNA sequencing data, we examined the expression patterns of TGFβ signalling molecules in the human ovary, testis, heart, liver, kidney, brain tissue, and cerebellum from 4 to 20weeks of gestation and postnatally. We also examined the correlations between gene expression of TGFβ signalling molecules and PCOS candidate genes. Key results TGFβ signalling molecules were dynamically expressed in most tissues prenatally and/or postnatally. FBN3 , a PCOS candidate gene involved in TGFβ signalling, was expressed during foetal development in all tissues. The PCOS candidate genes HMGA2, YAP1 , and RAD50 correlated significantly (P TGFBR1 in six out of the seven tissues examined. Conclusions This study suggests that possible crosstalk occurs between genes in loci associated with PCOS and TGFβ signalling molecules in multiple tissues, particularly during foetal development. Implications Thus, alteration in TGFβ signalling during foetal development could affect many tissues contributing to the multiple phenotypes of PCOS in later life.
Collapse
Affiliation(s)
- Rafiatu Azumah
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Katja Hummitzsch
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Richard A Anderson
- Medical Research Council Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Raymond J Rodgers
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
23
|
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
|
24
|
Kobayashi H, Shigetomi H, Matsubara S, Yoshimoto C, Imanaka S. Role of the mitophagy-apoptosis axis in the pathogenesis of polycystic ovarian syndrome. J Obstet Gynaecol Res 2024; 50:775-792. [PMID: 38417972 DOI: 10.1111/jog.15916] [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: 01/07/2024] [Accepted: 02/15/2024] [Indexed: 03/01/2024]
Abstract
AIM Polycystic ovary syndrome (PCOS) is a common endocrine disorder characterized by menstrual irregularities, androgen excess, and polycystic ovarian morphology, but its pathogenesis remains largely unknown. This review focuses on how androgen excess influences the molecular basis of energy metabolism, mitochondrial function, and mitophagy in granulosa cells and oocytes, summarizes our current understanding of the pathogenesis of PCOS, and discuss perspectives on future research directions. METHODS A search of PubMed and Google Scholar databases were used to identify relevant studies for this narrative literature review. RESULTS Female offspring born of pregnant animals exposed to androgens recapitulates the PCOS phenotype. Abnormal mitochondrial morphology, altered expression of genes related to glycolysis, mitochondrial biogenesis, fission/fusion dynamics, and mitophagy have been identified in PCOS patients and androgenic animal models. Androgen excess causes uncoupling of the electron transport chain and depletion of the cellular adenosine 5'-triphosphate pool, indicating further impairment of mitochondrial function. A shift toward mitochondrial fission restores mitochondrial quality control mechanisms. However, prolonged mitochondrial fission disrupts autophagy/mitophagy induction due to loss of compensatory reserve for mitochondrial biogenesis. Disruption of compensatory mechanisms that mediate the quality control switch from mitophagy to apoptosis may cause a disease phenotype. Furthermore, genetic predisposition, altered expression of genes related to glycolysis and oxidative phosphorylation, or a combination of these factors may also contribute to the development of PCOS. CONCLUSION In conclusion, fetuses exposed to a hyperandrogenemic intrauterine environment may cause the PCOS phenotype possibly through disruption of the compensatory regulation of the mitophagy-apoptosis axis.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, Kashihara, Japan
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
- Department of Gynecology and Reproductive Medicine, Aska Ladies Clinic, Nara, Japan
| | - Sho Matsubara
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
- Department of Medicine, Kei Oushin Clinic, Nishinomiya, Japan
| | - Chiharu Yoshimoto
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
- Department of Obstetrics and Gynecology, Nara Prefecture General Medical Center, Nara, Japan
| | - Shogo Imanaka
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, Kashihara, Japan
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| |
Collapse
|
25
|
Mimouni NEH, Giacobini P. Polycystic ovary syndrome (PCOS): progress towards a better understanding and treatment of the syndrome. C R Biol 2024; 347:19-25. [PMID: 38639155 DOI: 10.5802/crbiol.147] [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: 06/22/2023] [Revised: 11/06/2023] [Accepted: 11/17/2023] [Indexed: 04/20/2024]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine and metabolic disorder in women of reproductive age. It has a strong hereditary component estimated at 60 to 70% in daughters. It has been suggested that environmental factors during the fetal period may be involved in the development of the syndrome in adulthood. However, the underlying mechanisms of its transmission remain unknown, thus limiting the development of effective therapeutic strategies.This article highlights how an altered fetal environment (prenatal exposure to high levels of anti-Müllerian hormone) can contribute to the onset of PCOS in adulthood and lead to the transgenerational transmission of neuroendocrine and metabolic traits through alterations in the DNA methylation process.The originality of the translational findings summarized here involves the identification of potential biomarkers for early diagnosis of the syndrome, in addition to the validation of a promising therapeutic avenue in a preclinical model of PCOS, which can improve the management of patients suffering from the syndrome.
Collapse
|
26
|
Stener-Victorin E, Teede H, Norman RJ, Legro R, Goodarzi MO, Dokras A, Laven J, Hoeger K, Piltonen TT. Polycystic ovary syndrome. Nat Rev Dis Primers 2024; 10:27. [PMID: 38637590 DOI: 10.1038/s41572-024-00511-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/20/2024]
Abstract
Despite affecting ~11-13% of women globally, polycystic ovary syndrome (PCOS) is a substantially understudied condition. PCOS, possibly extending to men's health, imposes a considerable health and economic burden worldwide. Diagnosis in adults follows the International Evidence-based Guideline for the Assessment and Management of Polycystic Ovary Syndrome, requiring two out of three criteria - clinical or biochemical hyperandrogenism, ovulatory dysfunction, and/or specific ovarian morphological characteristics or elevated anti-Müllerian hormone. However, diagnosing adolescents omits ovarian morphology and anti-Müllerian hormone considerations. PCOS, marked by insulin resistance and hyperandrogenism, strongly contributes to early-onset type 2 diabetes, with increased odds for cardiovascular diseases. Reproduction-related implications include irregular menstrual cycles, anovulatory infertility, heightened risks of pregnancy complications and endometrial cancer. Beyond physiological manifestations, PCOS is associated with anxiety, depression, eating disorders, psychosexual dysfunction and negative body image, collectively contributing to diminished health-related quality of life in patients. Despite its high prevalence persisting into menopause, diagnosing PCOS often involves extended timelines and multiple health-care visits. Treatment remains ad hoc owing to limited understanding of underlying mechanisms, highlighting the need for research delineating the aetiology and pathophysiology of the syndrome. Identifying factors contributing to PCOS will pave the way for personalized medicine approaches. Additionally, exploring novel biomarkers, refining diagnostic criteria and advancing treatment modalities will be crucial in enhancing the precision and efficacy of interventions that will positively impact the lives of patients.
Collapse
Affiliation(s)
| | - Helena Teede
- Monash Centre for Health Research and Implementation, Monash Health and Monash University, Melbourne, Victoria, Australia
| | - Robert J Norman
- Robinson Research Institute, Adelaide Medical School, Adelaide, South Australia, Australia
| | - Richard Legro
- Department of Obstetrics and Gynecology, Penn State College of Medicine, Hershey, PA, USA
- Department of Public Health Science, Penn State College of Medicine, Hershey, PA, USA
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anuja Dokras
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Joop Laven
- Division of Reproductive Endocrinology & Infertility, Department of Obstetrics and Gynecology, Erasmus MC, Rotterdam, Netherlands
| | - Kathleen Hoeger
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| |
Collapse
|
27
|
Cotellessa L, Giacobini P. Role of Anti-Müllerian Hormone in the Central Regulation of Fertility. Semin Reprod Med 2024; 42:34-40. [PMID: 38608673 DOI: 10.1055/s-0044-1786050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
In recent years, the expanding roles of anti-Müllerian hormone (AMH) in various aspects of reproductive health have attracted significant attention. Initially recognized for its classical role in male sexual differentiation, AMH is produced postnatally by the Sertoli cells in the male testes and by the granulosa cells in the female ovaries. Traditionally, it was believed to primarily influence gonadal development and function. However, research over the last decade has unveiled novel actions of AMH beyond the gonads, specifically all along the hypothalamic-pituitary-gonadal axis. This review will focus on the emerging roles of AMH within the hypothalamus and discusses its potential implications in reproductive physiology. Additionally, recent preclinical and clinical studies have suggested that elevated levels of AMH may disrupt the hypothalamic network regulating reproduction, which could contribute to the central pathophysiology of polycystic ovary syndrome. These findings underscore the intricate interplay between AMH and the neuroendocrine system, offering new avenues for understanding the mechanisms underlying fertility and reproductive disorders.
Collapse
Affiliation(s)
- Ludovica Cotellessa
- Inserm, CHU Lille, Unit 1172, Lille Neuroscience & Cognition (LilNCog), University of Lille, Lille, France
| | - Paolo Giacobini
- Inserm, CHU Lille, Unit 1172, Lille Neuroscience & Cognition (LilNCog), University of Lille, Lille, France
| |
Collapse
|
28
|
Piltonen TT, Viita-aho J, Saarela U, Melin J, Forslund M. Utility of Serum Anti-Müllerian Hormone Measurement as Part of Polycystic Ovary Syndrome Diagnosis. Semin Reprod Med 2024; 42:49-59. [PMID: 38776986 PMCID: PMC11257749 DOI: 10.1055/s-0044-1786731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
The 2023 international evidence-based guideline update for the assessment and management of polycystic ovary syndrome (PCOS) recommends using the Rotterdam criteria for the diagnosis of PCOS. The updated guideline has evidence-based recommendation for the diagnosis, and it now also includes serum anti-Müllerian hormone (AMH) measurement as an alternative tool for gynecological ultrasound to diagnose polycystic ovary morphology (PCOM). The aim of this new recommendation was to facilitate PCOS diagnostic workup in primary care and other disciplines, as currently most diagnosing is done in gynecology and infertility clinics. Here, we review factors affecting AMH levels as well as the utility of AMH in PCOS diagnosis. We identified relevant studies that report different cut-offs for AMH to diagnose PCOM as part of PCOS diagnosis. There are, however, some limitations when using AMH that should be acknowledged. These include physiological aspects like age, ethnicity, and obesity and iatrogenic causes like hormonal medication and ovarian surgery. Also reference ranges are different depending on AMH assay used. As a summary, we conclude that AMH is a usable tool in PCOM diagnostics, but it does not have a single cut-off. Therefore, further studies are needed to establish age and assay-based reference ranges.
Collapse
Affiliation(s)
- Terhi T. Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center Oulu, University of Oulu, Oulu University Hospital, Oulu, Finland
| | - Johanna Viita-aho
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center Oulu, University of Oulu, Oulu University Hospital, Oulu, Finland
| | - Ulla Saarela
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center Oulu, University of Oulu, Oulu University Hospital, Oulu, Finland
| | - Johanna Melin
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Maria Forslund
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
29
|
Abbott DH, Hutcherson BA, Dumesic DA. Anti-Müllerian Hormone: A Molecular Key to Unlocking Polycystic Ovary Syndrome? Semin Reprod Med 2024; 42:41-48. [PMID: 38908381 DOI: 10.1055/s-0044-1787525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Anti-Müllerian hormone (AMH) is an important component within androgen receptor (AR)-regulated pathways governing the hyperandrogenic origin of polycystic ovary syndrome (PCOS). In women with PCOS, granulosa cell AMH overexpression in developing ovarian follicles contributes to elevated circulating AMH levels beginning at birth and continuing in adolescent daughters of PCOS women. A 6 to 7% incidence among PCOS women of gene variants coding for AMH or its receptor, AMHR2, suggests genetic contributions to AMH-related pathogenesis. Discrete gestational AMH administration to pregnant mice induces hypergonadotropic hyperandrogenic, PCOS-like female offspring with high circulating AMH levels that persist over three generations, suggesting epigenetic contributions to PCOS through developmental programming. Moreover, adult-onset, selective hyperactivation of hypothalamic neurons expressing gonadotropin-releasing hormone (GnRH) induces hypergonadotropic hyperandrogenism and PCOS-like traits in female mice. Both gestational and adult AMH inductions of PCOS-like traits are prevented by GnRH antagonist coadministration, implicating luteinizing hormone-dependent ovarian theca cell testosterone (T) action, mediated through the AR in AMH-induced pathogenesis. Interestingly, gestational or peripubertal exogenous T or dihydrotestosterone induction of PCOS-like traits in female mice, rats, sheep, and monkeys fails to elicit ovarian AMH hypersecretion; thus, AMH excess per se may lead to a distinct pathogenic contribution to hyperandrogenic PCOS origins.
Collapse
Affiliation(s)
- David H Abbott
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin
- Endocrinology and Reproductive Physiology Training Program, University of Wisconsin, Madison, Wisconsin
| | - Beverly A Hutcherson
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin
- Endocrinology and Reproductive Physiology Training Program, University of Wisconsin, Madison, Wisconsin
- Dean's Office, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, University of California, Los Angeles, California
| |
Collapse
|
30
|
Markantes GK, Panagodimou E, Koika V, Mamali I, Kaponis A, Adonakis G, Georgopoulos NA. Placental mRNA Expression of Neurokinin B Is Increased in PCOS Pregnancies with Female Offspring. Biomedicines 2024; 12:334. [PMID: 38397936 PMCID: PMC10886712 DOI: 10.3390/biomedicines12020334] [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: 01/05/2024] [Revised: 01/21/2024] [Accepted: 01/28/2024] [Indexed: 02/25/2024] Open
Abstract
Current research suggests that polycystic ovary syndrome (PCOS) might originate in utero and implicates the placenta in its pathogenesis. Kisspeptin (KISS1) and neurokinin B (NKB) are produced by the placenta in high amounts, and they have been implicated in several pregnancy complications associated with placental dysfunction. However, their placental expression has not been studied in PCOS. We isolated mRNA after delivery from the placentae of 31 PCOS and 37 control women with term, uncomplicated, singleton pregnancies. The expression of KISS1, NKB, and neurokinin receptors 1, 2, and 3 was analyzed with real-time polymerase chain reaction, using β-actin as the reference gene. Maternal serum and umbilical cord levels of total testosterone, sex hormone-binding globulin (SHBG), free androgen index (FAI), androstenedione, dehydroepiandrosterone sulfate (DHEAS), Anti-Mullerian hormone (AMH), and estradiol were also assessed. NKB placental mRNA expression was higher in PCOS women versus controls in pregnancies with female offspring. NKB expression depended on fetal gender, being higher in pregnancies with male fetuses, regardless of PCOS. NKB was positively correlated with umbilical cord FAI and AMH, and KISS1 was positively correlated with cord testosterone and FAI; there was also a strong positive correlation between NKB and KISS1 expression. Women with PCOS had higher serum AMH and FAI and lower SHBG than controls. Our findings indicate that NKB might be involved in the PCOS-related placental dysfunction and warrant further investigation. Studies assessing the placental expression of NKB should take fetal gender into consideration.
Collapse
Affiliation(s)
- Georgios K Markantes
- Division of Endocrinology, Department of Internal Medicine, School of Health Sciences, University of Patras, 26504 Patras, Greece
| | - Evangelia Panagodimou
- Department of Obstetrics and Gynecology, School of Health Sciences, University of Patras, 26504 Patras, Greece
| | - Vasiliki Koika
- Division of Endocrinology, Department of Internal Medicine, School of Health Sciences, University of Patras, 26504 Patras, Greece
| | - Irene Mamali
- Division of Endocrinology, Department of Internal Medicine, School of Health Sciences, University of Patras, 26504 Patras, Greece
| | - Apostolos Kaponis
- Department of Obstetrics and Gynecology, School of Health Sciences, University of Patras, 26504 Patras, Greece
| | - George Adonakis
- Department of Obstetrics and Gynecology, School of Health Sciences, University of Patras, 26504 Patras, Greece
| | - Neoklis A Georgopoulos
- Division of Endocrinology, Department of Internal Medicine, School of Health Sciences, University of Patras, 26504 Patras, Greece
| |
Collapse
|
31
|
Aref Y, Fat SC, Ray E. Recent insights into the role of hormones during development and their functional regulation. Front Endocrinol (Lausanne) 2024; 15:1340432. [PMID: 38318293 PMCID: PMC10841574 DOI: 10.3389/fendo.2024.1340432] [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: 11/18/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Introduction Hormones play a vital role in development from conception to birth and throughout the human lifespan. These periods are logically divided into fetal development, pre-pubertal growth, puberty, and adulthood. Deviations from standard physiological levels and release patterns of constituent hormones can lead to pathology affecting the normal developmental trajectory. Research is ongoing to better understand the mechanisms of these hormones and how their modulation affects development. Methods This article focuses on recent developments in understanding the role hormones play in development. We also cover recent discoveries in signaling pathways and hormonal regulation. Results New and continuing research into functional hormone regulation focuses on sex hormones, gonadotropic hormones, growth hormones, insulin-like growth factor, thyroid hormone, and the interconnectedness of each of these functional axes. Currently, the abundance of work focuses on fertility and correction of sex hormone levels based on an individual's condition and stage in life. Discussion Continuing research is needed to fully understand the long-term effects of hormone modulation in growth and sexual development. The role of each hormone in parallel endocrine axes should also be more thoroughly investigated to help improve the safety and efficacy in endocrine pharmacotherapeutics.
Collapse
Affiliation(s)
| | | | - Edward Ray
- Cedars-Sinai Medical Center, Department of Surgery, Division of Plastic and Reconstructive Surgery, Los Angeles, CA, United States
| |
Collapse
|
32
|
Ban M, Sun Y, Chen X, Zhou X, Zhang Y, Cui L. Association between maternal polycystic ovarian syndrome undergoing assisted reproductive technology and pregnancy complications and neonatal outcomes: a systematic review and meta-analysis. J Ovarian Res 2024; 17:6. [PMID: 38184624 PMCID: PMC10770902 DOI: 10.1186/s13048-023-01331-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/17/2023] [Indexed: 01/08/2024] Open
Abstract
BACKGROUND Polycystic ovarian syndrome (PCOS) is recognized as the most prevalent endocrine disorder among women of reproductive age. While the utilization of assisted reproductive technology (ART) has resulted in favorable outcomes for infertility treatment in PCOS patients, the inherent pathophysiological features of the condition give rise to complications and consequences during pregnancy and delivery for both the mother and offspring. This study was to assess the correlation between maternal PCOS and various pregnancy complications and neonatal outcomes undergone ART. METHODS A systematic search was conducted on PubMed, EmBase, and the Cochrane Library to identify observational studies that investigated the association between PCOS and the risk of various pregnancy complications and neonatal outcomes, including gestational diabetes mellitus (GDM), hypertension in pregnancy (PIH), preeclampsia (PE), preterm birth, abortion, congenital malformations (CA), small for gestational age (SGA), large for gestational age (LGA), low birth weight (LBW), macrosomia, neonatal intensive care unit (NICU) admission and birth weight. Eligible studies were selected based on predetermined inclusion and exclusion criteria. The meta-analysis was conducted using Review Manager and Stata software, with odds ratios (ORs) or mean difference (MD), confidence intervals (CIs), and heterogeneity (I2) being calculated. The search was conducted up to March 2023. RESULTS A total of 33 studies with a combined sample size of 92,810 participants were identified. The findings indicate that PCOS is significantly associated with an increased risk of GDM (OR 1.51, 95% CI:1.17-1.94), PIH (OR 1.72, 95% CI:1.25-2.39), PE (OR 2.12, 95% CI:1.49-3.02), preterm birth (OR 1.29, 95% CI:1.21-1.39), and LBW (OR 1.29, 95% CI:1.14-1.47). In subgroup analyses, the risks of GDM (OR 1.80, 95% CI:1.23-2.62) and abortion (OR 1.41, 95% CI:1.08-1.84) were elevated in fresh embryo transferred (ET) subgroup, whereas elevated risk of PE (OR 1.82, 95% CI:1.17-2.83) and preterm birth (OR 1.31, 95% CI:1.21-1.42) was identified in frozen ET subgroup. Whatever with or without hyperandrogenism, patients with PCOS had a higher risk in preterm birth (OR 1.69, 95% CI: 1.31-2.18; OR 1.24, 95% CI:1.02-1.50) and abortion (OR 1.38, 95% CI:1.12-1.71; OR 1.23, 95% CI:1.06-1.43). CONCLUSION Our findings suggest that individuals with PCOS undergone ART are at a notably elevated risk for experiencing pregnancy complications and unfavorable neonatal outcomes. Nevertheless, to establish a definitive association between PCOS and pregnancy-related outcomes, it is necessary to conduct extensive prospective, blinded cohort studies and effectively control for confounding variables.
Collapse
Affiliation(s)
- Miaomiao Ban
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, No.2021RU001), Jinan, 250012, Shandong, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yifei Sun
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, No.2021RU001), Jinan, 250012, Shandong, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Xiaojing Chen
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, No.2021RU001), Jinan, 250012, Shandong, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Xiaoqian Zhou
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, No.2021RU001), Jinan, 250012, Shandong, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yiyuan Zhang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, No.2021RU001), Jinan, 250012, Shandong, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Linlin Cui
- Center for Reproductive Medicine, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, No.2021RU001), Jinan, 250012, Shandong, China.
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250012, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
33
|
Chen-Patterson A, Bernier A, Burgert T, Davis V, Khan T, Geller D, Paprocki E, Shah R, Witchel SF, Pereira-Eshraghi C, Sopher AB, Cree MG, Torchen LC. Distinct Reproductive Phenotypes Segregate With Differences in Body Weight in Adolescent Polycystic Ovary Syndrome. J Endocr Soc 2024; 8:bvad169. [PMID: 38213910 PMCID: PMC10783242 DOI: 10.1210/jendso/bvad169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Indexed: 01/13/2024] Open
Abstract
Introduction Polycystic ovary syndrome (PCOS) is a heterogenous clinical syndrome defined by hyperandrogenism and irregular menses. In adult women with PCOS, discrete metabolic and reproductive subgroups have been identified. We hypothesize that distinct phenotypes can be distinguished between adolescent girls who are lean (LN-G) and girls with obesity (OB-G) at the time of PCOS diagnosis. Methods Data were extracted from the CALICO multisite PCOS database. Clinical data collected at the time of diagnosis were available in 354 patients (81% with obesity) from 7 academic centers. Patients with body mass index (BMI) < 85th percentile for age and sex were characterized as lean (LN-G) and those with BMI percentile ≥ 95th percentile as obese (OB-G). We compared metabolic and reproductive phenotypes in LN-G and OB-G. Results Reproductive phenotypes differed between the groups, with LN-G having higher total testosterone, androstenedione, and LH levels, while OB-G had lower sex hormone binding globulin (SHBG) and higher free testosterone. Metabolic profiles differed as expected, with OB-G having higher hemoglobin A1c, alanine aminotransferase, and serum triglycerides and more severe acanthosis nigricans. Conclusion LN-G with PCOS had a distinct reproductive phenotype characterized by increased LH, total testosterone, and androstenedione levels, suggesting neuroendocrine-mediated ovarian androgen production. In contrast, phenotypes in OB-G suggest hyperandrogenemia is primarily driven by insulin resistance with low SHBG levels. These observations support the existence of distinct metabolic and reproductive subtypes in adolescent PCOS characterized by unique mechanisms for hyperandrogenemia.
Collapse
Affiliation(s)
| | - Angelina Bernier
- Pediatric Endocrinology, University of Florida, Gainesville, FL 32608, USA
| | - Tania Burgert
- Pediatric Endocrinology, Children's Mercy Hospital, Kansas City, MO 64108, USA
| | - Vanessa Davis
- Pediatric Endocrinology, John H. Stroger, Jr. Hospital, Chicago, IL 60612, USA
| | - Tazeena Khan
- University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - David Geller
- Pediatric Endocrinology, Children's Hospital, Los Angeles, CA 90027, USA
| | - Emily Paprocki
- Pediatric Endocrinology, Children's Mercy Hospital, Kansas City, MO 64108, USA
| | - Rachana Shah
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Selma F Witchel
- Pediatric Endocrinology, University of Pittsburgh, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | | | - Aviva B Sopher
- Pediatric Endocrinology, Columbia University, NewYork, NY 10032, USA
| | - Melanie G Cree
- Pediatric Endocrinology, University of Colorado Anschutz, Aurora, CO 80045, USA
| | - Laura C Torchen
- Pediatric Endocrinology, Lurie Children's Hospital, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
34
|
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: 0] [Impact Index Per Article: 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
|
35
|
Khatun M, Lundin K, Naillat F, Loog L, Saarela U, Tuuri T, Salumets A, Piltonen TT, Tapanainen JS. Induced Pluripotent Stem Cells as a Possible Approach for Exploring the Pathophysiology of Polycystic Ovary Syndrome (PCOS). Stem Cell Rev Rep 2024; 20:67-87. [PMID: 37768523 PMCID: PMC10799779 DOI: 10.1007/s12015-023-10627-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the most prevalent endocrine condition among women with pleiotropic sequelae possessing reproductive, metabolic, and psychological characteristics. Although the exact origin of PCOS is elusive, it is known to be a complex multigenic disorder with a genetic, epigenetic, and environmental background. However, the pathogenesis of PCOS, and the role of genetic variants in increasing the risk of the condition, are still unknown due to the lack of an appropriate study model. Since the debut of induced pluripotent stem cell (iPSC) technology, the ability of reprogrammed somatic cells to self-renew and their potential for multidirectional differentiation have made them excellent tools to study different disease mechanisms. Recently, researchers have succeeded in establishing human in vitro PCOS disease models utilizing iPSC lines from heterogeneous PCOS patient groups (iPSCPCOS). The current review sets out to summarize, for the first time, our current knowledge of the implications and challenges of iPSC technology in comprehending PCOS pathogenesis and tissue-specific disease mechanisms. Additionally, we suggest that the analysis of polygenic risk prediction based on genome-wide association studies (GWAS) could, theoretically, be utilized when creating iPSC lines as an additional research tool to identify women who are genetically susceptible to PCOS. Taken together, iPSCPCOS may provide a new paradigm for the exploration of PCOS tissue-specific disease mechanisms.
Collapse
Affiliation(s)
- Masuma Khatun
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki University Central Hospital, Haartmaninkatu 8, Helsinki, 00029 HUS, Finland.
| | - Karolina Lundin
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki University Central Hospital, Haartmaninkatu 8, Helsinki, 00029 HUS, Finland
| | - Florence Naillat
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Liisa Loog
- Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
- Department of Genetics, University of Cambridge, Cambridge, CB2 3EH, UK
| | - Ulla Saarela
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki University Central Hospital, Haartmaninkatu 8, Helsinki, 00029 HUS, Finland
| | - Andres Salumets
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, 50406, Estonia
- Competence Centre of Health Technologies, Tartu, 50411, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Huddinge, Stockholm, 14186, Sweden
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki University Central Hospital, Haartmaninkatu 8, Helsinki, 00029 HUS, Finland
- Department of Obstetrics and Gynecology, HFR - Cantonal Hospital of Fribourg and University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
36
|
Gao L, Zhao Y, Wu H, Lin X, Guo F, Li J, Long Y, Zhou B, She J, Zhang C, Sheng J, Jin L, Wu Y, Huang H. Polycystic Ovary Syndrome Fuels Cardiovascular Inflammation and Aggravates Ischemic Cardiac Injury. Circulation 2023; 148:1958-1973. [PMID: 37937441 PMCID: PMC10713005 DOI: 10.1161/circulationaha.123.065827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 10/03/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Reducing cardiovascular disease burden among women remains challenging. Epidemiologic studies have indicated that polycystic ovary syndrome (PCOS), the most common endocrine disease in women of reproductive age, is associated with an increased prevalence and extent of coronary artery disease. However, the mechanism through which PCOS affects cardiac health in women remains unclear. METHODS Prenatal anti-Müllerian hormone treatment or peripubertal letrozole infusion was used to establish mouse models of PCOS. RNA sequencing was performed to determine global transcriptomic changes in the hearts of PCOS mice. Flow cytometry and immunofluorescence staining were performed to detect myocardial macrophage accumulation in multiple PCOS models. Parabiosis models, cell-tracking experiments, and in vivo gene silencing approaches were used to explore the mechanisms underlying increased macrophage infiltration in PCOS mouse hearts. Permanent coronary ligation was performed to establish myocardial infarction (MI). Histologic analysis and small-animal imaging modalities (eg, magnetic resonance imaging and echocardiography) were performed to evaluate the effects of PCOS on injury after MI. Women with PCOS and control participants (n=200) were recruited to confirm findings observed in animal models. RESULTS Transcriptomic profiling and immunostaining revealed that hearts from PCOS mice were characterized by increased macrophage accumulation. Parabiosis studies revealed that monocyte-derived macrophages were significantly increased in the hearts of PCOS mice because of enhanced circulating Ly6C+ monocyte supply. Compared with control mice, PCOS mice showed a significant increase in splenic Ly6C+ monocyte output, associated with elevated hematopoietic progenitors in the spleen and sympathetic tone. Plasma norepinephrine (a sympathetic neurotransmitter) levels and spleen size were consistently increased in women with PCOS when compared with those in control participants, and norepinephrine levels were significantly correlated with circulating CD14++CD16- monocyte counts. Compared with animals without PCOS, PCOS animals showed significantly exacerbated atherosclerotic plaque development and post-MI cardiac remodeling. Conditional Vcam1 silencing in PCOS mice significantly suppressed cardiac inflammation and improved cardiac injury after MI. CONCLUSIONS Our data documented previously unrecognized mechanisms through which PCOS could affect cardiovascular health in women. PCOS may promote myocardial macrophage accumulation and post-MI cardiac remodeling because of augmented splenic myelopoiesis.
Collapse
Affiliation(s)
- Ling Gao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China (L.G., X.L., F.G., J.L., Y.L., B.Z., C.Z., L.J., Y.W., H.H.)
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China (L.G., H.W., J. She, H.H.)
| | - Yichao Zhao
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.Z.)
| | - Haiyan Wu
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China (L.G., H.W., J. She, H.H.)
| | - Xianhua Lin
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China (L.G., X.L., F.G., J.L., Y.L., B.Z., C.Z., L.J., Y.W., H.H.)
| | - Fei Guo
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China (L.G., X.L., F.G., J.L., Y.L., B.Z., C.Z., L.J., Y.W., H.H.)
| | - Jie Li
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China (L.G., X.L., F.G., J.L., Y.L., B.Z., C.Z., L.J., Y.W., H.H.)
| | - Yuhang Long
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China (L.G., X.L., F.G., J.L., Y.L., B.Z., C.Z., L.J., Y.W., H.H.)
| | - Bokang Zhou
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China (L.G., X.L., F.G., J.L., Y.L., B.Z., C.Z., L.J., Y.W., H.H.)
| | - Junsen She
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China (L.G., H.W., J. She, H.H.)
| | - Chen Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China (L.G., X.L., F.G., J.L., Y.L., B.Z., C.Z., L.J., Y.W., H.H.)
| | - Jianzhong Sheng
- International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China (J. Sheng, H.H.)
| | - Li Jin
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China (L.G., X.L., F.G., J.L., Y.L., B.Z., C.Z., L.J., Y.W., H.H.)
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China (L.J., Y.W., H.H.)
| | - Yanting Wu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China (L.G., X.L., F.G., J.L., Y.L., B.Z., C.Z., L.J., Y.W., H.H.)
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China (L.J., Y.W., H.H.)
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China (L.G., X.L., F.G., J.L., Y.L., B.Z., C.Z., L.J., Y.W., H.H.)
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China (L.G., H.W., J. She, H.H.)
- International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China (J. Sheng, H.H.)
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China (L.J., Y.W., H.H.)
- State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China (H.H.)
| |
Collapse
|
37
|
Jiang L, Ruan X, Li Y, Gu M, Cheng J, Wang Y, Yang Y, Xu C, Wang Z, Liu L, Mueck AO. Diagnostic value of anti-Müllerian hormone combined with androgen-levels in Chinese patients with polycystic ovary syndrome. Gynecol Endocrinol 2023; 39:2206927. [PMID: 37141919 DOI: 10.1080/09513590.2023.2206927] [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] [Indexed: 05/06/2023] Open
Abstract
OBJECTIVE To establish a cutoff level of AMH which could help for the diagnosis of PCOS, to investigate the predictive value of AMH combined with androgens in Chinese women to diagnose PCOS. MATERIALS AND METHODS This is a prospective case control study, 550 women recruited (aged 20-40 years), in which 450 PCOS women recruited according to the Rotterdam criteria and 100 non-PCOS women in the control group were from the women for the pregnancy preparation examination. AMH were measured by the Elecsys AMH Plus immunoassay. Androgens and other sex hormone were measured. The validity of AMH toward the diagnosis of PCOS, or AMH combined with total testosterone, free testosterone, bioavailable testosterone and androstenedione was estimated by receiver operating characteristic (ROC)curves, and correlations between paired variables was estimated by Spearman's rank correlation coefficient. RESULTS The cutoff value of AMH in Chinese reproductive-age women with PCOS is 4.64 ng/mL, AUC under the curve is 0.938, with 81.6% sensitivity, and 92.0% specificity. Total testosterone, free testosterone, bioactive testosterone, and androstenedione are significantly higher in women with PCOS of reproductive age than in controls. The combination of AMH and free testosterone resulted in a higher AUC of 94.8%, with higher sensitivity (86.1%) and excellent specificity (90.3%) for the prediction of PCOS. CONCLUSION The Elecsys AMH Plus immunoassay, with a cutoff of 4.64 ng/mL, is a robust method for identifying PCOM to aid in PCOS diagnosis. The combination of AMH and free testosterone resulted in a higher AUC of 94.8% for the diagnose of PCOS.
Collapse
Affiliation(s)
- Lingling Jiang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xiangyan Ruan
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
- Department for Women's Health, University Women's Hospital and Research Centre for Women's Health, University of Tuebingen, Tuebingen, Germany
| | - Yanqiu Li
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Muqing Gu
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Jiaojiao Cheng
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yuejiao Wang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yu Yang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Che Xu
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Zhikun Wang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Lili Liu
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Alfred O Mueck
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
- Department for Women's Health, University Women's Hospital and Research Centre for Women's Health, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
38
|
Kulkarni S, Gupta K, Ratre P, Mishra PK, Singh Y, Biharee A, Thareja S. Polycystic ovary syndrome: Current scenario and future insights. Drug Discov Today 2023; 28:103821. [PMID: 37935329 DOI: 10.1016/j.drudis.2023.103821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/17/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023]
Abstract
Polycystic ovary syndrome (PCOS) prevails in approximately 33% of females of reproductive age globally. Although the root cause of the disease is unknown, attempts are made to clinically manage the disturbed hormone levels and symptoms arising due to hyperandrogenism, a hallmark of PCOS. This review presents detailed insights on the etiology, risk factors, current treatment strategies, and challenges therein. Medicinal agents currently in clinical trials and those in the development pipeline are emphasized. The significance of the inclusion of herbal supplements in PCOS and the benefits of improved lifestyle are also explained. Last, emerging therapeutic targets for treating PCOS are elaborated. The present review will assist the research fraternity working in the concerned domain to access significant knowledge associated with PCOS.
Collapse
Affiliation(s)
- Swanand Kulkarni
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Khushi Gupta
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Pooja Ratre
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab 151401, India; Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh 462030, India
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh 462030, India
| | - Yogesh Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Avadh Biharee
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab 151401, India.
| |
Collapse
|
39
|
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
|
40
|
Yang Y, Ruan X, Cheng J, Xu X, Mueck AO. Effects of different volumes of ovariectomy on fertility and offspring development of rats. Front Endocrinol (Lausanne) 2023; 14:1279610. [PMID: 38027202 PMCID: PMC10666792 DOI: 10.3389/fendo.2023.1279610] [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: 08/18/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Objective The purpose of this study was to explore the effect of removal of different volumes of ovarian tissue on fertility and offspring development of SD rats. Methods SD rats were randomly divided into 6 groups according to different volumes of ovariectomy: Sham group (n=6), non-ovariectomized; 25%-OVX group (n=6), with half of the left ovary excised; 50%-OVX group (n=5), with the left ovary excised; 75%-OVX group (n=5), with the left ovary and half of the right ovary excised; 87.5%-OVX group (n=6), with the left ovary and three quarters of the right ovary excised; 100%-OVX group (n=6), with bilateral ovaries excised. These female rats (F0) were mated with healthy male rats one and four months after the surgery, and the offspring of F0 rats were named F11mon and F14mon, respectively. The number of days from mating to delivery and number of live cubs were recorded. At postnatal day 21 (P21), the body weight, length and anogenital distance (AGD) of the cubs were measured. Results There were no differences in the number of live cubs between 25%-OVX, 50%-OVX and sham groups. Rats in the 87.5%-OVX group did not give birth at 1 month and 4 months after the operation. When compared with the sham group, the body weight and length of F11mon at P21 were increased in 25%-OVX group and 50%-OVX group. However, after the second delivery, we controlled each mother's lactation to no more than eight pups. As a result, there were no differences in the body weight, length and AGD of F14mon compared with sham group. Conclusion Removal of less than 50% of the ovaries did not affect the fertility of rats and offspring development of rats.
Collapse
Affiliation(s)
- Yu Yang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xiangyan Ruan
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
- Department for Women’s Health, University Women’s Hospital and Research Center for Women’s Health, University of Tuebingen, Tuebingen, Germany
| | - Jiaojiao Cheng
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xin Xu
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Alfred O. Mueck
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
- Department for Women’s Health, University Women’s Hospital and Research Center for Women’s Health, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
41
|
Iwase A, Hasegawa Y, Tsukui Y, Kobayashi M, Hiraishi H, Nakazato T, Kitahara Y. Anti-Müllerian hormone beyond an ovarian reserve marker: the relationship with the physiology and pathology in the life-long follicle development. Front Endocrinol (Lausanne) 2023; 14:1273966. [PMID: 38027144 PMCID: PMC10657644 DOI: 10.3389/fendo.2023.1273966] [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: 08/07/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Anti-Müllerian hormone (AMH), an indirect indicator of the number of remaining follicles, is clinically used as a test for ovarian reserve. Typically, a decline suggests a decrease in the number of remaining follicles in relation to ovarian toxicity caused by interventions, which may implicate fertility. In contrast, serum AMH levels are elevated in patients with polycystic ovary syndrome. AMH is produced primarily in the granulosa cells of the preantral and small antral follicles. Thus it varies in association with folliculogenesis and the establishment and shrinking of the follicle cohort. Ovarian activity during the female half-life, from the embryonic period to menopause, is based on folliculogenesis and maintenance of the follicle cohort, which is influenced by developmental processes, life events, and interventions. AMH trends over a woman's lifetime are associated with in vivo follicular cohort transitions that cannot be observed directly.
Collapse
Affiliation(s)
- Akira Iwase
- Department of Obstetrics and Gynecology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Guo F, Mao S, Long Y, Zhou B, Gao L, Huang H. The Influences of Perinatal Androgenic Exposure on Cardiovascular and Metabolic Disease of Offspring of PCOS. Reprod Sci 2023; 30:3179-3189. [PMID: 37380913 DOI: 10.1007/s43032-023-01286-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/08/2023] [Indexed: 06/30/2023]
Abstract
Hyperandrogenism is an endocrine disorder affecting a large population of reproductive-aged women, thus proportionally high number of fetuses are subjected to prenatal androgenic exposure (PNA). The short-term stimulations at critical ontogenetic stages can wield lasting influences on the health. The most commonly diagnosed conditions in reproductive age women is polycystic ovary syndrome (PCOS). PNA may affect the growth and development of many systems in the whole body and disrupts the normal metabolic trajectory in the offspring of PCOS, contributing to the prevalence of cardiovascular and metabolic diseases (CVMD), including myocardial hypertrophy, hypertension, hyperinsulinemia, insulin resistance, hyperglycemia, obesity, and dyslipidemia, which are the leading causes of hospitalizations in young PCOS offspring. In this review, we focus on the effects of prenatal androgenic exposure on the cardiovascular and metabolic diseases in offspring, discuss the possible pathogenesis respectively, and summarize potential management strategies to improve metabolic health of PCOS offspring. It is expected that the incidence of CVMD and the medical burden will be reduced in the future.
Collapse
Affiliation(s)
- Fei Guo
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Suqing Mao
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Yuhang Long
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Bokang Zhou
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Ling Gao
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Hefeng Huang
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.
| |
Collapse
|
43
|
Silva MSB, Decoster L, Delpouve G, Lhomme T, Ternier G, Prevot V, Giacobini P. Overactivation of GnRH neurons is sufficient to trigger polycystic ovary syndrome-like traits in female mice. EBioMedicine 2023; 97:104850. [PMID: 37898094 PMCID: PMC10630624 DOI: 10.1016/j.ebiom.2023.104850] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/18/2023] [Accepted: 10/12/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is the most common endocrine disorder leading to anovulatory infertility. Abnormalities in the central neuroendocrine system governed by gonadotropin-releasing hormone (GnRH) neurons might be related to ovarian dysfunction in PCOS, although the link in this disordered brain-to-ovary communication remains unclear. Here, we manipulated GnRH neurons using chemogenetics in adult female mice to unveil whether chronic overaction of these neurons would trigger PCOS-like hormonal and reproductive impairments. METHODS We used adult Gnrh1cre female mice to selectively target and express the designer receptors exclusively activated by designer drugs (DREADD)-based chemogenetic tool hM3D(Gq) in hypophysiotropic GnRH neurons. Chronic chemogenetic activation protocol was carried out with clozapine N-oxide (CNO) i.p. injections every 48 h over a month. We evaluated the reproductive and hormonal profile before, during, and two months after chemogenetic manipulations. FINDINGS We discovered that the overactivation of GnRH neurons was sufficient to disrupt reproductive cycles, promote hyperandrogenism, and induce ovarian dysfunction. These PCOS features were detected with a long-lasting neuroendocrine dysfunction through abnormally high luteinizing hormone (LH) pulse secretion. Additionally, the GnRH-R blockade prevented the establishment of long-term neuroendocrine dysfunction and androgen excess in these animals. INTERPRETATION Taken together, our results show that hyperactivity of hypothalamic GnRH neurons is a major driver of reproductive and hormonal impairments in PCOS and suggest that antagonizing the aberrant GnRH signaling could be an efficient therapeutic venue for the treatment of PCOS. FUNDING European Research Council (ERC) under the European Union's Horizon 2020 research and innovation program (grant agreement n◦ 725149).
Collapse
Affiliation(s)
- Mauro S B Silva
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France; Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Laurine Decoster
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France; Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Gaspard Delpouve
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France; Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Tori Lhomme
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France; Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Gaetan Ternier
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France; Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France; Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Paolo Giacobini
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France; Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France.
| |
Collapse
|
44
|
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
|
45
|
Halder A, Kumar H, Sharma M, Jain M, Kalsi AK, Pandey S. Serum anti-Müllerian hormone: A potential biomarker for polycystic ovary syndrome. Indian J Med Res 2023; 158:397-406. [PMID: 37991331 DOI: 10.4103/ijmr.ijmr_4608_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND OBJECTIVES Polycystic ovary syndrome (PCOS) is characterized by chronic ovulatory dysfunction, hyperandrogenism and polycystic ovary morphology (PCOM). Although hyperandrogenism is one of the major features of PCOS, it is rarely observed in southeast Asia. Recently, however, there has been growing evidence on association of anti-Müllerian hormone (AMH) with PCOS. The objective of this study was to investigate the diagnostic potentials of AMH in PCOS individuals. METHODS This case-control study included a total of 131 women with PCOS and 49 healthy controls who were enrolled after the exclusion of secondary causes of PCOS. Serum AMH was measured using an ultra-sensitive AMH ELISA kit in addition to other diagnostic biomarkers. Statistical analyses was carried out using the Student's t test, Wilcoxon rank-sum test, receiver operating characteristic (ROC) curve analysis, Spearman's rank correlation test and multivariable binary logistic regression analysis. RESULTS The median AMH values were 8.5 ng/ml and 2.5 ng/ml in the study group and controls, respectively ( P <0.001). The normal cutoff value of 4.1 ng/ml for AMH was derived from ROC curve analysis. With a 4.1 ng/ml cut-off value, high levels of AMH was found in about 84 per cent of PCOS cases. However, no significant difference in AMH level was noted between age groups (<20 vs . ≥20 yr), body mass index (BMI) (<25 vs . ≥25 kg/m 2 ) and PCOM types. The area under the ROC curve (AUC) for AMH yielded diagnostic range values. In total PCOS cases, AUC was 0.93 (95% CI: 0.88 and 0.96), and in phenotype A PCOS cases, AUC was 0.96 (95% CI: 0.91 and 0.98). The correlation test also showed no association with BMI, the FG score, PCOM, free androgen index, androstenedione, dehydroepiandrosterone sulphate and luteinizing hormone. However, a weak correlation was observed with testosterone in total PCOS cases and with DHT as well as age in phenotype A PCOS cases. The prediction model for PCOS using multivariable binary logistic regression analysis showed AMH as the best marker. INTERPRETATION CONCLUSIONS The results of this study suggest that AMH can be considered as the most promising biomarker in PCOS women, particularly with phenotype A and phenotype D.
Collapse
Affiliation(s)
| | - Hemant Kumar
- Department of Reproductive Biology, AIIMS, New Delhi, India
| | - Mona Sharma
- Department of Reproductive Biology, AIIMS, New Delhi, India
| | - Manish Jain
- Department of Reproductive Biology, AIIMS, New Delhi, India
| | | | - Shivam Pandey
- Department of Biostatistics, AIIMS, New Delhi, India
| |
Collapse
|
46
|
Sumji S, Bhat A, Rashid A, Bashir R, Wani IA, Vasudevan V, Sehar T, Ganie MA. Efficacy of serum anti-mullerian hormone (AMH) levels for prediction of polycystic ovary syndrome (PCOS) and its association with clinical, biochemical and hormonal parameters. Indian J Clin Biochem 2023; 38:457-465. [PMID: 37746534 PMCID: PMC10516813 DOI: 10.1007/s12291-022-01058-4] [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: 04/29/2022] [Accepted: 06/06/2022] [Indexed: 10/14/2022]
Abstract
Anti-mullerian hormone (AMH) has been proposed to add significance to diagnosis of PCOS in case of ambiguity. However, variable cutoffs of AHM among PCOS women have been reported. Using case-control design, this study investigated the diagnostic threshold of serum AMH levels among age matched 113 PCOS and 75 normo-ovulatory women and its correlation with clinical, hormonal and ultrasonographic parameters.PCOS was defined as per Rotterdam criteria 2003. Results depicted the mean serum AMH level to be significantly higher in PCOS group (7.84 ± 3.67vs. 3.23 ± 1.56 ng/mL) than controls. The AMH levels were positively(p = 0.001) associated with ovarian volume (r = 0.521) as well as number of ovarian follicles(r = 0.461). Further, serum AMH levels showed a positive correlation with luteinizing hormone/follicle stimulating hormone (LH/FSH) ratio (r = 0.206, p = 0.02), but no correlation significant with age, BMI,FG score and testosterone levels. As per receiver operating characteristic (ROC) curve, cut-off was worked out to be 3.76 ng/ml with 86.7% sensitivity and 62.7% specificity. The mean level of AMH were highest among PCOS women with phenotype A (12.67 ± 3.46 ng/ml) with least among PCOS women displaying phenotype B(7.28 ± 1.60 ng/ml) where there is absence of PCOM. In conclusion, serum AMH levels are highly predictive of PCOM and high LH/FSH ratio among PCOS women and may be a potent diagnostic marker of ovarian dysfunction either alone or in conjunction with other tools. Supplementary Information The online version contains supplementary material available at 10.1007/s12291-022-01058-4.
Collapse
Affiliation(s)
- Sajad Sumji
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir India
| | - Abid Bhat
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir India
| | - Aafia Rashid
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir India
- Department of Clinical Research, Sher-i-Kashmir Institute of Medical Sciences, 190011 Srinagar, India
| | - Rohina Bashir
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir India
- Department of Clinical Research, Sher-i-Kashmir Institute of Medical Sciences, 190011 Srinagar, India
| | - Imtiyaz A Wani
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir India
- Department of Clinical Research, Sher-i-Kashmir Institute of Medical Sciences, 190011 Srinagar, India
| | - Vishnu Vasudevan
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir India
| | - Tajali Sehar
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir India
- Department of Clinical Research, Sher-i-Kashmir Institute of Medical Sciences, 190011 Srinagar, India
| | - Mohd. Ashraf Ganie
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir India
- Department of Clinical Research, Sher-i-Kashmir Institute of Medical Sciences, 190011 Srinagar, India
| |
Collapse
|
47
|
Dumesic DA, Abbott DH, Chazenbalk GD. An Evolutionary Model for the Ancient Origins of Polycystic Ovary Syndrome. J Clin Med 2023; 12:6120. [PMID: 37834765 PMCID: PMC10573644 DOI: 10.3390/jcm12196120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrinopathy of reproductive-aged women, characterized by hyperandrogenism, oligo-anovulation and insulin resistance and closely linked with preferential abdominal fat accumulation. As an ancestral primate trait, PCOS was likely further selected in humans when scarcity of food in hunter-gatherers of the late Pleistocene additionally programmed for enhanced fat storage to meet the metabolic demands of reproduction in later life. As an evolutionary model for PCOS, healthy normal-weight women with hyperandrogenic PCOS have subcutaneous (SC) abdominal adipose stem cells that favor fat storage through exaggerated lipid accumulation during development to adipocytes in vitro. In turn, fat storage is counterbalanced by reduced insulin sensitivity and preferential accumulation of highly lipolytic intra-abdominal fat in vivo. This metabolic adaptation in PCOS balances energy storage with glucose availability and fatty acid oxidation for optimal energy use during reproduction; its accompanying oligo-anovulation allowed PCOS women from antiquity sufficient time and strength for childrearing of fewer offspring with a greater likelihood of childhood survival. Heritable PCOS characteristics are affected by today's contemporary environment through epigenetic events that predispose women to lipotoxicity, with excess weight gain and pregnancy complications, calling for an emphasis on preventive healthcare to optimize the long-term, endocrine-metabolic health of PCOS women in today's obesogenic environment.
Collapse
Affiliation(s)
- Daniel A. Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA;
| | - David H. Abbott
- Department of Obstetrics and Gynecology, Wisconsin National Primate Research Center, University of Wisconsin, 1223 Capitol Court, Madison, WI 53715, USA;
| | - Gregorio D. Chazenbalk
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA;
| |
Collapse
|
48
|
Concepción-Zavaleta MJ, Coronado-Arroyo JC, Quiroz-Aldave JE, Durand-Vásquez MDC, Ildefonso-Najarro SP, Rafael-Robles LDP, Concepción-Urteaga LA, Gamarra-Osorio ER, Suárez-Rojas J, Paz-Ibarra J. Endocrine factors associated with infertility in women: an updated review. Expert Rev Endocrinol Metab 2023; 18:399-417. [PMID: 37702309 DOI: 10.1080/17446651.2023.2256405] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/09/2023] [Accepted: 09/04/2023] [Indexed: 09/14/2023]
Abstract
INTRODUCTION Infertility is defined as the inability to conceive after unprotected sexual intercourse for at least 12 consecutive months. Our objective is to present an updated narrative review on the endocrine causes of infertility in women. AREAS COVERED A comprehensive review was conducted using Scielo, Scopus, and EMBASE databases, comprising 245 articles. The pathophysiology of infertility in women was described, including endocrinopathies such as hypothalamic amenorrhea, hyperprolactinemia, polycystic ovary syndrome, primary ovarian insufficiency, obesity, thyroid dysfunction, and adrenal disorders. The diagnostic approach was outlined, emphasizing the necessity of hormonal studies and ovarian response assessments. Additionally, the treatment plan was presented, commencing with non-pharmacological interventions, encompassing the adoption of a Mediterranean diet, vitamin supplementation, moderate exercise, and maintaining a healthy weight. Subsequently, pharmacological treatment was discussed, focusing on the management of associated endocrine disorders and ovulatory dysfunction. EXPERT OPINION This comprehensive review highlights the impact of endocrine disorders on fertility in women, providing diagnostic and therapeutic algorithms. Despite remaining knowledge gaps that hinder more effective treatments, ongoing research and advancements show promise for improved fertility success rates within the next five years. Enhanced comprehension of the pathophysiology behind endocrine causes and the progress in genetic research will facilitate the delivery of personalized treatments, thus enhancing fertility rates.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - José Paz-Ibarra
- Department of Medicine, School of Medicine, Universidad Nacional Mayor de San Marcos, Lima, Perú
| |
Collapse
|
49
|
Calcaterra V, Cena H, Sottotetti F, Hruby C, Madini N, Zelaschi N, Zuccotti G. Low-Calorie Ketogenic Diet: Potential Application in the Treatment of Polycystic Ovary Syndrome in Adolescents. Nutrients 2023; 15:3582. [PMID: 37630772 PMCID: PMC10459579 DOI: 10.3390/nu15163582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/05/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women of reproductive age. Hyperandrogenism, hyperinsulinism and insulin resistance (IR) are the main drivers of clinical, metabolic and reproductive phenotypes of PCOS. In adolescence, the cornerstones of PCOS treatment are lifestyle and dietary interventions. In particular, the quality and quantity of carbohydrates introduced with the diet play a crucial role in the benefits of diet on PCOS. Recently, the ketogenic diet (KD) has attracted significant interest for the treatment of IR and for the control of carbohydrate metabolism, which has proven to be beneficial for several dysmetabolic conditions, including PCOS. The goal of the KD is to induce a fasting-like metabolism with production of chetonic bodies. Ketosis is a good regulator of calorie intake and mimics the starvation effect in the body, leading to body weight control and consequent metabolism. Additionally, during ketogenesis, insulin receptor sensitivity is also promoted. We proposed a broad overview of the available literature regarding KD indications and considered its metabolic benefits useful for improving PCOS management. The reported data support that a low-calorie ketogenic diet (LCKD) plays a positive role as a regulator of control weight, IR, glucose and lipid homeostasis and hormonal profile. Unfortunately, the evidence concerning the benefits of the very LCKD in adolescents with PCOS and excessive body weight is still numerically scarce. Further studies are necessary to understand whether these effects are due to weight loss or to the nutritional characteristics of this diet. Considering the long-term consequences of PCOS, it is crucial to detect the prospects of nutritional interventions to protect fertility, starting in adolescence.
Collapse
Affiliation(s)
- Valeria Calcaterra
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy;
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (C.H.); (G.Z.)
| | - Hellas Cena
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (F.S.); (N.M.); (N.Z.)
- Clinical Nutrition and Dietetics Service, Unit of Internal Medicine and Endocrinology, ICS Maugeri IRCCS, 27100 Pavia, Italy
| | - Francesca Sottotetti
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (F.S.); (N.M.); (N.Z.)
| | - Chiara Hruby
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (C.H.); (G.Z.)
| | - Nagaia Madini
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (F.S.); (N.M.); (N.Z.)
| | - Noemi Zelaschi
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (F.S.); (N.M.); (N.Z.)
| | - Gianvincenzo Zuccotti
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (C.H.); (G.Z.)
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy; (F.S.); (N.M.); (N.Z.)
- Department of Biomedical and Clinical Science, University of Milano, 20157 Milano, Italy
| |
Collapse
|
50
|
Li Q, Sang Y, Chen Q, Ye B, Zhou X, Zhu Y. Integrated bioinformatics analysis elucidates granulosa cell whole-transcriptome landscape of PCOS in China. J Ovarian Res 2023; 16:154. [PMID: 37537636 PMCID: PMC10398987 DOI: 10.1186/s13048-023-01223-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/20/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common reproductive, neuroendocrine, and metabolic disorder in women of reproductive age that affects up to 5-10% of women of reproductive age. The aetiology of follicle development arrest and critical issues regarding the abnormal follicular development in PCOS remain unclear. The present study aims to systematically evaluate granulosa cell whole-transcriptome sequencing data to gain more insights into the transcriptomic landscape and molecular mechanism of PCOS in China. METHODS In the present study, the microarray datasets GSE138518, GSE168404, GSE193123, GSE138572, GSE95728, and GSE145296 were downloaded from the Gene Expression Omnibus (GEO) database. Subsequently, differential expression analysis was performed on the PCOS and control groups, followed by functional interaction prediction analysis to investigate gene-regulatory circuits in PCOS. Finally, hub genes and their associated ncRNAs were validated by qPCR in human-luteinized granulosa (hGL) cells and were correlated with the clinical characteristics of the patients. RESULTS A total of 200 differentially expressed mRNAs, 3 differentially expressed miRNAs, 52 differentially expressed lncRNAs, and 66 differentially expressed circRNAs were found in PCOS samples compared with controls. GO and KEGG enrichment analyses indicated that the DEGs were mostly enriched in phospholipid metabolic processes, steroid biosynthesis and inflammation related pathways. In addition, the upregulated miRNA hsa-miR-205-5p was significantly enriched in the ceRNA network, and two hub genes, MVD and PNPLA3, were regulated by hsa-miR-205-5p, which means that hsa-miR-205-5p may play a fundamental role in the pathogenesis of PCOS. We also found that MVD and PNPLA3 were related to metabolic processes and ovarian steroidogenesis, which may be the cause of the follicle development arrest in PCOS patients. CONCLUSIONS In summary, we systematically constructed a ceRNA network depicting the interactions between the ncRNAs and the hub genes in PCOS and control subjects and correlated the hub genes with the clinical characteristics of the patients, which provides valuable insights into the granulosa cell whole-transcriptome landscape of PCOS in China.
Collapse
Affiliation(s)
- Qingfang Li
- School of Medicine, Women’s Hospital, Zhejiang University, 1 Xueshi Road, Shangcheng District, Hangzhou, 310006 China
- Key Laboratory of Reproductive Genetics, Ministry of Education Zhejiang University, Hangzhou, 310006 China
| | - Yimiao Sang
- School of Medicine, Women’s Hospital, Zhejiang University, 1 Xueshi Road, Shangcheng District, Hangzhou, 310006 China
- Key Laboratory of Reproductive Genetics, Ministry of Education Zhejiang University, Hangzhou, 310006 China
| | - Qingqing Chen
- School of Medicine, Women’s Hospital, Zhejiang University, 1 Xueshi Road, Shangcheng District, Hangzhou, 310006 China
- Key Laboratory of Reproductive Genetics, Ministry of Education Zhejiang University, Hangzhou, 310006 China
| | - Bingru Ye
- School of Medicine, Women’s Hospital, Zhejiang University, 1 Xueshi Road, Shangcheng District, Hangzhou, 310006 China
- Key Laboratory of Reproductive Genetics, Ministry of Education Zhejiang University, Hangzhou, 310006 China
| | - Xiaoqian Zhou
- School of Medicine, Women’s Hospital, Zhejiang University, 1 Xueshi Road, Shangcheng District, Hangzhou, 310006 China
- Key Laboratory of Reproductive Genetics, Ministry of Education Zhejiang University, Hangzhou, 310006 China
| | - Yimin Zhu
- School of Medicine, Women’s Hospital, Zhejiang University, 1 Xueshi Road, Shangcheng District, Hangzhou, 310006 China
- Key Laboratory of Reproductive Genetics, Ministry of Education Zhejiang University, Hangzhou, 310006 China
- Department of Reproductive Endocrinology, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006 China
| |
Collapse
|