1
|
An J, Zhou Q, Guo X, Xu C, Jia X, Cao Z, Lu Q. From Pathophysiology to Treatment: The Role of Ferroptosis in PCOS. FRONT BIOSCI-LANDMRK 2025; 30:25586. [PMID: 40018919 DOI: 10.31083/fbl25586] [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: 07/08/2024] [Revised: 08/18/2024] [Accepted: 08/29/2024] [Indexed: 03/01/2025]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent gynecological endocrine and metabolic disorder in women, with an incidence rate of 10-13%. The etiology of PCOS is multifaceted, involving genetic predisposition, environmental influences, lifestyle factors, and endocrine metabolic dysregulation. Iron, a critical mineral, not only plays a role in regulating female physiological functions and the progression of PCOS but also requires careful management to avoid deficiency. However, excess iron can trigger ferroptosis, a form of nonapoptotic cell death characterized by the accumulation of lipid peroxides. While numerous studies have explored ferroptosis in patients with PCOS and animal models, the precise mechanisms and therapeutic implications remain inadequately understood. This review seeks to elucidate the pathophysiology of PCOS and the contributory factors of ferroptosis. Additionally, we examine the diverse manifestations of ferroptosis in PCOS and evaluate its role. Furthermore, we introduce ferroptosis-related traditional Chinese medicines that may enhance the understanding of PCOS pathogenesis and aid in the development of targeted therapies for ferroptosis in PCOS.
Collapse
Affiliation(s)
- Jie An
- Nanjing University of Chinese Medicine, 210029 Nanjing, Jiangsu, China
- Department of Gynecology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, 215300 Kunshan, Jiangsu, China
| | - Qin Zhou
- Department of Gynecology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, 215300 Kunshan, Jiangsu, China
| | - Xiaojing Guo
- Department of Gynecology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, 215300 Kunshan, Jiangsu, China
| | - Congya Xu
- Department of Gynecology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, 215300 Kunshan, Jiangsu, China
| | - XiaoFang Jia
- Department of Gynecology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, 215300 Kunshan, Jiangsu, China
| | - Zhenzhen Cao
- Department of Gynecology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, 215300 Kunshan, Jiangsu, China
| | - Qibin Lu
- Nanjing University of Chinese Medicine, 210029 Nanjing, Jiangsu, China
- Department of Gynecology of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 210029 Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Li X, Lin S, Yang X, Chen C, Cao S, Zhang Q, Ma J, Zhu G, Zhang Q, Fang Q, Zheng C, Liang W, Wu X. When IGF-1 Meets Metabolic Inflammation and Polycystic Ovary Syndrome. Int Immunopharmacol 2024; 138:112529. [PMID: 38941670 DOI: 10.1016/j.intimp.2024.112529] [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: 04/12/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 06/30/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder associated with insulin resistance (IR) and hyperandrogenaemia (HA). Metabolic inflammation (MI), characterized by a chronic low-grade inflammatory state, is intimately linked with chronic metabolic diseases such as IR and diabetes and is also considered an essential factor in the development of PCOS. Insulin-like growth factor 1 (IGF-1) plays an essential role in PCOS pathogenesis through its multiple functions in regulating cell proliferation metabolic processes and reducing inflammatory responses. This review summarizes the molecular mechanisms by which IGF-1, via MI, participates in the onset and progression of PCOS, aiming to provide insights for studies and clinical treatment of PCOS.
Collapse
Affiliation(s)
- Xiushen Li
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China; Department of Traditional Chinese Medicine, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Sailing Lin
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Xiaolu Yang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Can Chen
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Shu Cao
- Xin'an Academy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Qi Zhang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Jingxin Ma
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Guli Zhu
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Qi Zhang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Qiongfang Fang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Weizheng Liang
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China.
| | - Xueqing Wu
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China.
| |
Collapse
|
3
|
Stefanaki K, Karagiannakis DS, Peppa M, Vryonidou A, Kalantaridou S, Goulis DG, Psaltopoulou T, Paschou SA. Food Cravings and Obesity in Women with Polycystic Ovary Syndrome: Pathophysiological and Therapeutic Considerations. Nutrients 2024; 16:1049. [PMID: 38613082 PMCID: PMC11013286 DOI: 10.3390/nu16071049] [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: 01/12/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Polycystic ovary syndrome (PCOS), the most common endocrine disorder in women of reproductive age, constitutes a metabolic disorder frequently associated with obesity and insulin resistance (IR). Furthermore, women with PCOS often suffer from excessive anxiety and depression, elicited by low self-esteem due to obesity, acne, and hirsutism. These mood disorders are commonly associated with food cravings and binge eating. Hypothalamic signaling regulates appetite and satiety, deteriorating excessive food consumption. However, the hypothalamic function is incapable of compensating for surplus food in women with PCOS, leading to the aggravation of obesity and a vicious circle. Hyperandrogenism, IR, the reduced secretion of cholecystokinin postprandially, and leptin resistance defined by leptin receptors' knockout in the hypothalamus have been implicated in the pathogenesis of hypothalamic dysfunction and appetite dysregulation. Diet modifications, exercise, and psychological and medical interventions have been applied to alleviate food disorders, interrupting the vicious circle. Cognitive-behavioral intervention seems to be the mainstay of treatment, while the role of medical agents, such as GLP-1 analogs and naltrexone/bupropion, has emerged.
Collapse
Affiliation(s)
- Katerina Stefanaki
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.S.); (T.P.); (S.A.P.)
| | - Dimitrios S. Karagiannakis
- Academic Department of Gastroenterology, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Melpomeni Peppa
- Endocrine Unit and Diabetes Center, Second Department of Internal Medicine, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- 3rd Department of Internal Medicine, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Andromachi Vryonidou
- Department of Endocrinology and Diabetes Center, Hellenic Red Cross Hospital, 11526 Athens, Greece;
| | - Sophia Kalantaridou
- 3rd Department of Obstetrics and Gynecology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Dimitrios G. Goulis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, School of Medicine, Aristotle University of Thessaloniki, 57001 Thessaloniki, Greece
| | - Theodora Psaltopoulou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.S.); (T.P.); (S.A.P.)
| | - Stavroula A. Paschou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.S.); (T.P.); (S.A.P.)
| |
Collapse
|
4
|
Wang K, Li Y, Chen Y. Androgen excess: a hallmark of polycystic ovary syndrome. Front Endocrinol (Lausanne) 2023; 14:1273542. [PMID: 38152131 PMCID: PMC10751361 DOI: 10.3389/fendo.2023.1273542] [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: 08/06/2023] [Accepted: 11/22/2023] [Indexed: 12/29/2023] Open
Abstract
Polycystic ovarian syndrome (PCOS) is a metabolic, reproductive, and psychological disorder affecting 6-20% of reproductive women worldwide. However, there is still no cure for PCOS, and current treatments primarily alleviate its symptoms due to a poor understanding of its etiology. Compelling evidence suggests that hyperandrogenism is not just a primary feature of PCOS. Instead, it may be a causative factor for this condition. Thus, figuring out the mechanisms of androgen synthesis, conversion, and metabolism is relatively important. Traditionally, studies of androgen excess have largely focused on classical androgen, but in recent years, adrenal-derived 11-oxygenated androgen has also garnered interest. Herein, this Review aims to investigate the origins of androgen excess, androgen synthesis, how androgen receptor (AR) signaling mediates adverse PCOS traits, and the role of 11-oxygenated androgen in the pathophysiology of PCOS. In addition, it provides therapeutic strategies targeting hyperandrogenism in PCOS.
Collapse
Affiliation(s)
- Kexin Wang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yanhua Li
- Department of General Practice, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Chen
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
5
|
Olaniyi KS, Areloegbe SE. Alleviation of adipose-hepatic glycolipid dysregulation by acetate in experimental PCOS model is associated with NF-κB/NLRP3 repression. Can J Physiol Pharmacol 2023; 101:630-641. [PMID: 37590975 DOI: 10.1139/cjpp-2023-0169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
This study hypothesized that acetate breaks the vicious cycle driving adipose-hepatic metabolic dysregulation in a rat model of polycystic ovarian syndrome (PCOS), possibly by suppression of nuclear factor-kappaB (NF-κB)/NOD-like receptor protein 3 (NLRP3) inflammasome. Female Wistar rats (8-week-old) were randomly allocated into four groups of n =6/group, which received vehicle, sodium acetate (200 mg), letrozole (1 mg/kg), and letrozole plus sodium acetate, respectively. The animals were treated by oral gavage, once daily for a period of 21 days. The PCOS animals were insulin-resistant, hyperandrogenic, and hypoestrogenic with decreased sex-hormone binding globulin. In addition, the hepatic tissue had increased lipid profile and decreased glycogen synthesis, while the adipose tissue showed decreased lipid profile with elevated glycogen synthesis. Besides, the results also showed increased malondialdehyde, γ-glutamyl transferase, lactate dehydrogenase, and inflammatory mediators with corresponding decrease in antioxidant defense in the hepatic and adipose tissues. Immunohistochemical evaluation also demonstrated severe expression with Bcl2-associated X protein/NLRP3 antibodies. Nonetheless, concomitant acetate supplementation attenuated these derangements. The present data collectively suggest that acetate ameliorates adipose-hepatic glycolipid dysregulation in experimental PCOS model by attenuating androgen excess and NF-κB/NLRP3 immunoreactivity.
Collapse
Affiliation(s)
| | - Stephanie E Areloegbe
- Cardio/Endo-metabolic and Microbiome Research Unit, Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti 360101, Nigeria
| |
Collapse
|
6
|
Rashid R, Tripathi R, Singh A, Sarkar S, Kawale A, Bader GN, Gupta S, Gupta RK, Jha RK. Naringenin improves ovarian health by reducing the serum androgen and eliminating follicular cysts in letrozole-induced polycystic ovary syndrome in the Sprague Dawley rats. Phytother Res 2023; 37:4018-4041. [PMID: 37165686 DOI: 10.1002/ptr.7860] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 04/03/2023] [Accepted: 04/17/2023] [Indexed: 05/12/2023]
Abstract
Polycystic ovary syndrome (PCOS) is most common in women of reproductive age, giving rise to androgen excess and anovulation, leading to infertility and non-reproductive complications. We explored the ameliorating effect of naringenin in PCOS using the Sprague Dawley (SD) rat model and human granulosa cells. Letrozole-induced PCOS rats were given either naringenin (50 mg/kg/day) alone or in combination with metformin (300 mg/kg/day), followed by the estrous cycle, hormonal analysis, and glucose sensitivity test. To evaluate the effect of naringenin on granulosa cell (hGC) steroidogenesis, we treated cells with naringenin (2.5 μM) alone or in combination with metformin (1 mM) in the presence of forskolin (10 μM). To determine the steroidogenesis of CYP-17A1, -19A1, and 3βHSD2, the protein expression levels were examined. Treatment with naringenin in the PCOS animal groups increased ovulation potential and decreased cystic follicles and levels of androgens. The expression levels of CYP-17A1, -19A1, and 3βHSD2, were seen restored in the ovary of PCOS SD rats' model and in the human ovarian cells in response to the naringenin. We found an increased expression level of phosphorylated-AKT in the ovary and hGCs by naringenin. Naringenin improves ovulation and suppress androgens and cystic follicles, involving AKT activation.
Collapse
Affiliation(s)
- Rumaisa Rashid
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Department of Pharmaceutical Sciences, University of Kashmir, Jammu and Kashmir, India
| | - Rupal Tripathi
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Akanksha Singh
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sudarsan Sarkar
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ajaykumar Kawale
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - G N Bader
- Department of Pharmaceutical Sciences, University of Kashmir, Jammu and Kashmir, India
| | - Satish Gupta
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rakesh Kumar Gupta
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rajesh Kumar Jha
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
7
|
Zhang H, Butoyi C, Yuan G, Jia J. Exploring the role of gut microbiota in obesity and PCOS: Current updates and future prospects. Diabetes Res Clin Pract 2023; 202:110781. [PMID: 37331521 DOI: 10.1016/j.diabres.2023.110781] [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: 04/14/2023] [Revised: 05/26/2023] [Accepted: 06/13/2023] [Indexed: 06/20/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine gynecological disorder, and the specific pathogenesis of PCOS has not been elucidated. Obesity is a current major public health problem, which is also vital to PCOS. It can exacerbate PCOS symptoms via insulin resistance and hyperandrogenemia. The treatment of PCOS patients depends on the prevailing symptoms. Lifestyle interventions and weight loss remain first-line treatments for women with PCOS. The gut microbiota, which is a current research hot spot, has a substantial influence on PCOS and is closely related to obesity. The present study aimed to elucidate the function of the gut microbiota in obesity and PCOS to provide new ideas for the treatment of PCOS.
Collapse
Affiliation(s)
- Hui Zhang
- First Clinical Medical College, Jiangsu University, Zhenjiang, Jiangsu, China; Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Claudette Butoyi
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China; School of Medicine , Jiangsu University, Zhenjiang, Jiangsu, China
| | - Guoyue Yuan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Jue Jia
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
8
|
Sharafieh G, Salmanifarzaneh F, Gharbi N, Sarvestani FM, Rahmanzad F, Razlighi MR, Bakhtari A, Nazari N. Histological and molecular evaluation of Mentha arvensis extract on a polycystic ovary syndrome rat model. JBRA Assist Reprod 2023; 27:247-253. [PMID: 36630609 PMCID: PMC10279426 DOI: 10.5935/1518-0557.20220052] [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: 01/01/2022] [Accepted: 09/13/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE This study aimed to investigate the impact of Mentha arvensis on a rat model of polycystic ovary syndrome (PCOS). METHODS The PCOS rat model was made by the daily subcutaneous injection of testosterone enanthate (250mg/kg) for 21 days. Thirty rats were divided into five groups, including a healthy control group and four PCOS groups treated with various concentrations of hydroalcoholic extract of Mentha arvensis (0, 50, 100 and 200mg/kg). LH and FSH were measured in the blood. The ovaries were used for histological investigation, Cyp17 and Ptgs2 genes expression and total antioxidant capacity. RESULTS Our results indicated that the level of LH and FSH hormones in treated PCOS rats with various concentrations of M. arvensis were reduced in comparison with the untreated PCOS group (p>0.01). Mentha arvensis in the highest concentration (200mg/kg) decreased the number of cysts in this group in comparison with the untreated PCOS group (p<0.01). The expression of Cyp17 and Ptgs2 genes in the treated group with the highest concentration of hydroalcoholic extract were decreased in comparison with the untreated PCOS group (p<0.05). Moreover, the antioxidant capacity in the rats receiving Mentha arvensis hydroalcoholic extract was significantly increased in comparison with that from the untreated PCOS rats (p<0.05). CONCLUSIONS For the first time, Mentha arvensis hydroalcoholic extract proved to reduce some polycystic ovary syndrome symptoms. In the present experiment, a dose of 200mg/kg of Mentha arvensis hydroalcoholic extract was regarded as the most efficient dose.
Collapse
Affiliation(s)
- Golnoosh Sharafieh
- Department of Clinical Biochemistry, School of Medicine, Tehran
University of Medical Sciences, Tehran, Iran
- Department of Clinical Biochemistry, Islamic Azad University,
Shahr-e-Qods Branch, Qods, Tehran, Iran
| | - Fatemeh Salmanifarzaneh
- Department of Clinical Biochemistry, School of Medicine, Tehran
University of Medical Sciences, Tehran, Iran
- Department of Clinical Biochemistry, Islamic Azad University,
Shahr-e-Qods Branch, Qods, Tehran, Iran
| | - Negin Gharbi
- Department of Biology, Zarghan Branch, Islamic Azad University,
Zarghan, Iran
- Clinical Neurology Research Center, Shiraz University of Medical
Sciences, Shiraz, Iran
| | | | - Fatemeh Rahmanzad
- Department of immunology, College of Veterinary Medicine, Science
and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Azizollah Bakhtari
- Reprodutive Biology Department, School of Advanced Medical Sciences
and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nazanin Nazari
- Department of Immunology, Shiraz University of Medical Sciences,
Shiraz, Iran
| |
Collapse
|
9
|
Takeda Y, Demura M, Kometani M, Karashima S, Yoneda T, Takeda Y. Molecular and Epigenetic Control of Aldosterone Synthase, CYP11B2 and 11-Hydroxylase, CYP11B1. Int J Mol Sci 2023; 24:ijms24065782. [PMID: 36982850 PMCID: PMC10054571 DOI: 10.3390/ijms24065782] [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: 12/23/2022] [Revised: 03/15/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Aldosterone and cortisol serve important roles in the pathogenesis of cardiovascular diseases and metabolic disorders. Epigenetics is a mechanism to control enzyme expression by genes without changing the gene sequence. Steroid hormone synthase gene expression is regulated by transcription factors specific to each gene, and methylation has been reported to be involved in steroid hormone production and disease. Angiotensin II or potassium regulates the aldosterone synthase gene, CYP11B2. The adrenocorticotropic hormone controls the 11b-hydroxylase, CYP11B1. DNA methylation negatively controls the CYP11B2 and CYP11B1 expression and dynamically changes the expression responsive to continuous stimulation of the promoter gene. Hypomethylation status of the CYP11B2 promoter region is seen in aldosterone-producing adenomas. Methylation of recognition sites of transcription factors, including cyclic AMP responsive element binding protein 1 or nerve growth factor-induced clone B, diminish their DNA-binding activity. A methyl-CpG-binding protein 2 cooperates directly with the methylated CpG dinucleotides of CYP11B2. A low-salt diet, treatment with angiotensin II, and potassium increase the CYP11B2 mRNA levels and induce DNA hypomethylation in the adrenal gland. A close association between a low DNA methylation ratio and an increased CYP11B1 expression is seen in Cushing's adenoma and aldosterone-producing adenoma with autonomous cortisol secretion. Epigenetic control of CYP11B2 or CYP11B1 plays an important role in autonomic aldosterone or cortisol synthesis.
Collapse
Affiliation(s)
- Yoshimichi Takeda
- Endocrinology and Metabolism, Kanazawa University Hospital, Kanazawa 920-8641, Japan
- Department of Hygiene, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-1192, Japan
| | - Masashi Demura
- Department of Hygiene, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-1192, Japan
| | - Mitsuhiro Kometani
- Endocrinology and Metabolism, Kanazawa University Hospital, Kanazawa 920-8641, Japan
| | - Shigehiro Karashima
- Institute of Liberal Arts and Science, Kanazawa University, Kanazawa 920-1192, Japan
| | - Takashi Yoneda
- Institute of Liberal Arts and Science, Kanazawa University, Kanazawa 920-1192, Japan
| | - Yoshiyu Takeda
- Endocrinology and Metabolism, Kanazawa University Hospital, Kanazawa 920-8641, Japan
- Endocrine and Diabetes Center, Asanogawa General Hospital, Kanazawa 920-0811, Japan
| |
Collapse
|
10
|
Khamoshina MB, Artemenko YS, Bayramova AA, Ryabova VA, Orazov MR. Polycystic ovary syndrome and obesity: a modern paradigm. RUDN JOURNAL OF MEDICINE 2022. [DOI: 10.22363/2313-0245-2022-26-4-382-395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Polycystic ovary syndrome is a heterogeneous endocrine disease that affects women of childbearing age. The pathogenesis of polycystic ovary syndrome has not been fully studied to date, its paradigm considers the genetic determinism of the manifestation of hormonal and metabolic disorders, which are considered to be criteria for the verification of the disease (hyperandrogenism, oligo/anovulation and/or polycystic ovarian transformation during ultrasound examination (ultrasound). This review discusses the main ways of interaction between hyperandrogenism, insulin resistance and obesity and their role in the pathogenesis of polycystic ovary syndrome, as well as possible methods of treatment for this category of patients. The review analyzes the role of hyperandrogenism and insulin resistance in the implementation of the genetic scenario of polycystic ovary syndrome and finds out the reasons why women with polycystic ovary syndrome often demonstrate the presence of a «metabolic trio» - hyperinsulinemia, insulin resistance and type 2 diabetes mellitus. It is noted that obesity is not included in the criteria for the diagnosis of polycystic ovary syndrome, but epidemiological data confirm the existence of a relationship between these diseases. Obesity, especially visceral, which is often found in women with polycystic ovary syndrome, enhances and worsens metabolic and reproductive outcomes with polycystic ovary syndrome, as well as increases insulin resistance and compensatory hyperinsulinemia, which, in turn, stimulates adipogenesis and suppresses lipolysis. Obesity increases the sensitivity of tech cells to luteinizing hormone stimulation and enhances functional hyperandrogenism of the ovaries, increasing the production of androgens by the ovaries. Excess body weight is associated with a large number of inflammatory adipokines, which, in turn, contribute to the growth of insulin resistance and adipogenesis. Obesity and insulin resistance exacerbate the symptoms of hyperandrogenism, forming a vicious circle that contributes to the development of polycystic ovary syndrome. These data allow us to conclude that bariatric surgery can become an alternative to drugs (metformin, thiazolidinedione analogs of glucagon-like peptide-1), which has shown positive results in the treatment of patients with polycystic ovary syndrome and obesity.
Collapse
|
11
|
Differentiating Polycystic Ovary Syndrome from Adrenal Disorders. Diagnostics (Basel) 2022; 12:diagnostics12092045. [PMID: 36140452 PMCID: PMC9498167 DOI: 10.3390/diagnostics12092045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/08/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Although polycystic ovary syndrome (PCOS) is primarily considered a hyperandrogenic disorder in women characterized by hirsutism, menstrual irregularity, and polycystic ovarian morphology, an endocrinological investigation should be performed to rule out other hyperandrogenic disorders (e.g., virilizing tumors, non-classical congenital adrenal hyperplasia (NCAH), hyperprolactinemia, and Cushing’s syndrome) to make a certain diagnosis. PCOS and androgen excess disorders share clinical features such as findings due to hyperandrogenism, findings of metabolic syndrome, and menstrual abnormalities. The diagnosis of a woman with these symptoms is generally determined based on the patient’s history and rigorous clinical examination. Therefore, distinguishing PCOS from adrenal-originated androgen excess is an indispensable step in diagnosis. In addition to an appropriate medical history and physical examination, the measurement of relevant basal hormone levels and dynamic tests are required. A dexamethasone suppression test is used routinely to make a differential diagnosis between Cushing’s syndrome and PCOS. The most important parameter for differentiating PCOS from NCAH is the measurement of basal and ACTH-stimulated 17-OH progesterone (17-OHP) when required in the early follicular period. It should be kept in mind that rapidly progressive hyperandrogenic manifestations such as hirsutism may be due to an androgen-secreting adrenocortical carcinoma. This review discusses the pathophysiology of androgen excess of both adrenal and ovarian origins; outlines the conditions which lead to androgen excess; and aims to facilitate the differential diagnosis of PCOS from certain adrenal disorders.
Collapse
|
12
|
Association of Insulin Resistance and Elevated Androgen Levels with Polycystic Ovarian Syndrome (PCOS): A Review of Literature. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:9240569. [PMID: 35356614 PMCID: PMC8959968 DOI: 10.1155/2022/9240569] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/08/2022] [Accepted: 02/12/2022] [Indexed: 12/14/2022]
Abstract
The polycystic ovary syndrome (PCOS) is the disease featured by elevated levels of androgens, ovulatory dysfunction, and morphological abnormalities. At reproductive stage of women, the rate of PCOS occurrence is measured as 6–10% and the prevalence rate may be double. There are different pathophysiological factors involved in PCOS, and they play a major role in various abnormalities in individual patient. It is clear that there is noteworthy elevation of androgen in PCOS, causing substantial misery and infertility problems. The overexposure of androgen is directly linked with insulin resistance and hyperinsulinaemia. It has been reported previously that PCOS is related to cardiac metabolic miseries and potently increases the risk of heart diseases. Endometrial cancer is also a serious concern which is reported with exceedingly high incidence in women with PCOS. However, the overexposure of androgen has direct and specific influence on the development of insulin resistance. Although many factors are involved, resistance to the insulin and enhanced level of androgen are considered the major causes of PCOS. In the present review, we have focused on the pathophysiology and major revolutions of insulin resistance and excessive levels of androgen in females with PCOS.
Collapse
|
13
|
Chen Y, Zhang Q, Ma J, Yu Y. Mapping research trends of insulin resistance in polycystic ovary syndrome from 2017 to 2021: A bibliometric analysis. Front Endocrinol (Lausanne) 2022; 13:963213. [PMID: 36589816 PMCID: PMC9797656 DOI: 10.3389/fendo.2022.963213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION To map publication trends and explore research hotspots of insulin resistance (IR) in polycystic ovary syndrome (PCOS) study. METHODS With the theme of "Polycystic ovary syndrome" AND "Insulin Resistance", the key data set of Science Core Literature Collection (WoSCC) web from 2017 to 2021 was extracted and bibliometric analysis was performed. Through VOSviewer v1.6.10 software, the research trend in this field is analyzed visually. RESULTS 2080 literatures about IR in PCOS from 2017 to 2021 were downloaded. The following basic information was collected for each article: country, author, institution, journal, references. The key words are divided into six categories: (1) The interaction between insulin resistance and chronic inflammation; (2) The relationship between insulin resistance and metabolic syndrome and nonalcoholic fatty liver disease; (3) The interaction between insulin resistance and hyperandrogenemia; (4) The relationship between insulin resistance and dyslipidemia; (5) Metformin may regulate insulin resistance in the treatment of PCOS; (6) The study of serum biomarkers in PCOS patients with insulin resistance. DISCUSSION The six key words extracted can provide an in-depth perspective for the study of IR in PCOS, and provide valuable information to help researchers identify potential research directions, collaborators and cooperative institutions.
Collapse
|
14
|
Wu LM, Wang YX, Zhan Y, Liu AH, Wang YX, Shen HF, Wang YF, Wang LY, Tao ZB, Wang YQ. Dulaglutide, a long-acting GLP-1 receptor agonist, can improve hyperandrogenemia and ovarian function in DHEA-induced PCOS rats. Peptides 2021; 145:170624. [PMID: 34375684 DOI: 10.1016/j.peptides.2021.170624] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/05/2021] [Accepted: 08/05/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVE The purpose of this study was to explore the effect of dulaglutide on DHEA induced PCOS rats and its mechanism, to provide new drugs and research directions for clinical treatment of PCOS. METHODS In this study, the PCOS model was established by giving female SD rats subcutaneous injection of DHEA for 21 consecutive days. After modeling, the treatment group was injected subcutaneously with three doses of dulaglutide for 3 weeks. The model group was injected with sterile ultrapure water, and the normal group did not get any intervention. The body weight changes of rats in each group were recorded from the first day when rats received the administration of dulaglutide. Three weeks later, the rats were fasted the night after the last treatment, determined fasting insulin and fasting glucose the next day. After the rats were anesthetized by chloral hydrate, more blood was collected from the heart of the rat. The serum insulin, testosterone and sex hormone binding globulin (SHBG) levels were detected by the enzyme-linked immunoassay method. After removing the adipose tissue, the obtained rat ovary tissue was used for subsequent experimental detection, using HE staining for morphology and follicular development analysis; qRT-PCR for the detection of 3βHSD, CYP17α1, CYP19α1, and StAR gene expression in ovarian tissue; and western blotting analysis of CYP17α1, CYP19α1, StAR protein expression and insulin level to verify whether dulaglutide has a therapeutic effect on PCOS in rats. RESULTS After treated with different concentrations of dulaglutide, we found that the body weight of rats in the treatment groups were reduced. Compared with the rats in PCOS group, the serum androgen level of rats in the treatment groups was significantly decreased, and the serum sex hormone binding protein content was significantly increased, and there was statistically significant difference between these groups and PCOS group. In terms of protein expression and gene regulation, the expression of 3βHSD, CYP19α1 and StAR in the ovarian tissue of rats in treatment groups were decreased significantly after received the treatment of dulaglutide, and there was statistically significant difference between these groups and PCOS group. In addition, dulaglutide reduced the insulin content in the ovarian tissue of PCOS rats. CONCLUSION Dulaglutide may reduce the hyperandrogenemia of PCOS rats by regulating the content of serum SHBG and the expression of 3βHSD, CYP19α1, and StAR related genes and proteins, thereby inhibiting the excessive development of small follicles and the formation of cystic follicles in the ovaries of PCOS rats, thereby improving polycystic ovary in PCOS rats. In addition, dulaglutide may reduce the weight of PCOS rats, further reducing the level of high androgen in PCOS rats, and improving the morphology of their polycystic ovaries.
Collapse
Affiliation(s)
- Lu-Ming Wu
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yin-Xue Wang
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yue Zhan
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - A-Hui Liu
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yi-Xiang Wang
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China
| | - Hao-Fei Shen
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yi-Fan Wang
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China
| | - Li-Yan Wang
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China; Gansu Key Laboratory of Reproductive Medicine and Embryo, Gansu International Scientific and Technological Cooperation Base of Reproductive medicine transformation application, Lanzhou, China
| | - Zhong-Bin Tao
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yi-Qing Wang
- The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, China; Gansu Key Laboratory of Reproductive Medicine and Embryo, Gansu International Scientific and Technological Cooperation Base of Reproductive medicine transformation application, Lanzhou, China.
| |
Collapse
|
15
|
Polycystic ovarian syndrome: Correlation between hyperandrogenism, insulin resistance and obesity. Clin Chim Acta 2019; 502:214-221. [PMID: 31733195 DOI: 10.1016/j.cca.2019.11.003] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/03/2019] [Accepted: 11/04/2019] [Indexed: 12/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a complex and heterogeneous endocrine disease characterized by clinical or laboratorial hyperandrogenism, oligo-anovulation and metabolic abnormalities, including insulin resistance, excessive weight or obesity, type II diabetes, dyslipidemia and an increased risk of cardiovascular disease. The most significant clinical manifestation of PCOS is hyperandrogenism. Excess androgen profoundly affects granulosa cell function and follicular development via complex mechanisms that lead to obesity and insulin resistance. Most PCOS patients with hyperandrogenism have steroid secretion defects that result in abnormal folliculogenesis and failed dominant follicle selection. Hyperandrogenism induces obesity, hairy, acne, and androgenetic alopecia. These symptoms can bring great psychological stress to women. Drugs such as combined oral contraceptive pills, metformin, pioglitazone and low-dose spironolactone help improve pregnancy rates by decreasing androgen levels in vivo. Notably, PCOS is heterogeneous, and hyperandrogenism is not the only pathogenic factor. Obesity and insulin resistance aggravate the symptoms of hyperandrogenism, forming a vicious cycle that promotes PCOS development. Although numerous studies have been conducted, the definitive pathogenic mechanisms of PCOS remain uncertain. This review summarizes and discusses previous and recent findings regarding the relationship between hyperandrogenism, insulin resistance, obesity and PCOS.
Collapse
|
16
|
Zhu JL, Chen Z, Feng WJ, Long SL, Mo ZC. Sex hormone-binding globulin and polycystic ovary syndrome. Clin Chim Acta 2019; 499:142-148. [PMID: 31525346 DOI: 10.1016/j.cca.2019.09.010] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 12/14/2022]
Abstract
Polycystic ovary syndrome (PCOS), one of the most common endocrine diseases that causes infertility in reproductive women, is characterized by hyperandrogenemia, chronic anovulation, and polycystic ovary morphology (PCOM), and most women with PCOS have metabolic abnormalities. A reduction in plasma sex hormone-binding globulin (SHBG), a transport carrier that binds estrogen and androgens and regulates their biological activities, is often used as an indicator of hyperandrogenism in women with PCOS. Low serum SHBG levels are considered a biomarker of abnormal metabolism and are related to insulin resistance (IR), compensatory hyperinsulinemia and abnormalities in glucose and lipid metabolism in PCOS patients. SHBG is also associated with the long-term prognosis of PCOS. SHBG gene polymorphism is correlated with the risk of PCOS. As SHBG plays a vital role in the occurrence and development of PCOS, knowledge regarding its role in PCOS is helpful for further understanding the molecular mechanism of SHBG in PCOS development and providing new ideas for the treatment of female infertility. Hepatocyte nuclear factor-4α (HNF-4α) is a vital transcription factor in the SHBG synthesis process. HNF-4α binds to the cis-type element DR1 in the SHBG promoter to initiate transcription and regulates hepatic SHBG levels by modulating glucose and lipid metabolism and inflammatory factors. However, it remains unclear whether HNF-4α is indirectly involved in the pathogenesis of PCOS via regulation of hepatic SHBG synthesis. Therefore, this review discusses the interaction between SHBG and the various complications of PCOS as well as the regulatory effect of HNF-4α on SHBG expression.
Collapse
Affiliation(s)
- Jing-Ling Zhu
- Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, University of South China, Hengyang, Hunan 421001, China; Hunan Province Innovative Training Base for Medical Postgraduates, University of South China and Yueyang Women & Children's Medical Center, Yueyang, Hunan 416000, China
| | - Zhuo Chen
- Hunan Province Innovative Training Base for Medical Postgraduates, University of South China and Yueyang Women & Children's Medical Center, Yueyang, Hunan 416000, China
| | - Wen-Jie Feng
- 2015 Grade Medical Imaging Class of Medical School, University of South China, Hengyang 421001, China
| | - Shuang-Lian Long
- Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, University of South China, Hengyang, Hunan 421001, China; Hunan Province Innovative Training Base for Medical Postgraduates, University of South China and Yueyang Women & Children's Medical Center, Yueyang, Hunan 416000, China.
| | - Zhong-Cheng Mo
- Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, University of South China, Hengyang, Hunan 421001, China; Hunan Province Innovative Training Base for Medical Postgraduates, University of South China and Yueyang Women & Children's Medical Center, Yueyang, Hunan 416000, China.
| |
Collapse
|
17
|
Perturbed ovarian and uterine glucocorticoid receptor signaling accompanies the balanced regulation of mitochondrial function and NFκB-mediated inflammation under conditions of hyperandrogenism and insulin resistance. Life Sci 2019; 232:116681. [PMID: 31344428 DOI: 10.1016/j.lfs.2019.116681] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/16/2019] [Accepted: 07/21/2019] [Indexed: 11/23/2022]
Abstract
AIM This study aimed to determine whether glucocorticoid receptor (GR) signaling, mitochondrial function, and local inflammation in the ovary and uterus are intrinsically different in rats with hyperandrogenism and insulin resistance compared to controls. MAIN METHODS Female Sprague Dawley rats were exposed to daily injections of human chorionic gonadotropin and/or insulin. KEY FINDINGS In both the ovary and the uterus, decreased expression of the two GR isoforms was concurrent with increased expression of Fkbp51 but not Fkbp52 mRNA in hCG + insulin-treated rats. However, these rats exhibited contrasting regulation of Hsd11b1 and Hsd11b2 mRNAs in the two tissues. Further, the expression of several oxidative phosphorylation-related proteins decreased in the ovary and uterus following hCG and insulin stimulation, in contrast to increased expression of many genes involved in mitochondrial function and homeostasis. Additionally, hCG + insulin-treated rats showed increased expression of ovarian and uterine NFκB signaling proteins and Tnfaip3 mRNA. The mRNA expression of Il1b, Il6, and Mmp2 was decreased in both tissues, while the mRNA expression of Tnfa, Ccl2, Ccl5, and Mmp3 was increased in the uterus. Ovaries and uteri from animals co-treated with hCG and insulin showed increased collagen deposition compared to controls. SIGNIFICANCE Our observations suggest that hyperandrogenism and insulin resistance disrupt ovarian and uterine GR activation and trigger compensatory or adaptive effects for mitochondrial homeostasis, allowing tissue-level maintenance of mitochondrial function in order to limit ovarian and uterine dysfunction. Our study also suggests that hyperandrogenism and insulin resistance activate NFκB signaling resulting in aberrant regulation of inflammation-related gene expression.
Collapse
|
18
|
Tamadon A, Hu W, Cui P, Ma T, Tong X, Zhang F, Li X, Shao LR, Feng Y. How to choose the suitable animal model of polycystic ovary syndrome? TRADITIONAL MEDICINE AND MODERN MEDICINE 2018. [DOI: 10.1142/s2575900018300047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a gynecological metabolic and endocrine disorder with uncertain etiology. To understand the etiology of PCOS or the evaluation of various therapeutic agents, different animal models have been introduced. Considering this fact that is difficult to develop an animal model that mimics all aspects of this syndrome, but, similarity of biological, anatomical, and/or biochemical features of animal model to the human PCOS phenotypes can increase its application. This review paper evaluates the recently researched animal models and introduced the best models for different research purposes in PCOS studies. During January 2013 to January 2017, 162 studies were identified which applied various kinds of animal models of PCOS including rodent, primate, ruminant and fish. Between these models, prenatal and pre-pubertal androgen rat models and then prenatal androgen mouse model have been studied in detail than others. The comparison of main features of these models with women PCOS demonstrates higher similarity of these three models to human conditions. Thereafter, letrozole models can be recommended for the investigation of various aspects of PCOS. Interestingly, similarity of PCOS features of post-pubertal insulin and human chorionic gonadotropin rat models with women PCOS were considerable which can make it as a good choice for future investigations.
Collapse
Affiliation(s)
- Amin Tamadon
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Wei Hu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Peng Cui
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Tong Ma
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Xiaoyu Tong
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| | - Feifei Zhang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P. R. China
| | - Xin Li
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P. R. China
| | - Linus R. Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P. R. China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, P. R. China
- Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|
19
|
Mahamed RR, Maganhin CC, Sasso GRS, de Jesus Simões M, Baracat MCP, Baracat EC, Soares- JM. Metformin improves ovarian follicle dynamics by reducing theca cell proliferation and CYP-17 expression in an androgenized rat model. J Ovarian Res 2018; 11:18. [PMID: 29490689 PMCID: PMC5831207 DOI: 10.1186/s13048-018-0392-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 02/25/2018] [Indexed: 01/22/2023] Open
Abstract
Background Metformin influences insulin receptor signaling, which might interfere with the proliferation of ovarian follicular structures and steroidogenesis. We hypothesize that reductions in glucose and insulin levels might interfere with CYP-17 expression and histomorphological changes in an androgenized rat model. The aim of this study was to analyze the effect of metformin on CYP-17 expression, follicular dynamics, and proliferative parameters in neonatally androgenized female rats. Methods Thirty-six newborn rats were randomly allocated to the following three groups on the third day of life: control (CG, n = 12), androgenized (GA, n = 12), and androgenized + metformin (GAmet, n = 12). The GA and GAmet animals were administered 0.1 mL of testosterone propionate (1.25 mg/animal) diluted in castor oil (vehicle) in a single dose; the CG rats received a subcutaneous injection of the vehicle in the dorsum. After 90 days, gavage treatment was initiated, distilled water was administered to the CG and GA rats, and metformin (150 mg/kg) was administered to the GAmet animals. The treatment was administered daily for six weeks. Following anesthesia, blood was drawn for biochemical measurements, and the ovaries were removed for histological and immunohistochemical analyses of Ki67, VEGFA and CYP17 expression. The glucose and insulin levels were also measured. Results The comparison of the GA and GAmet animals revealed that metformin decreased the weight as well as the glucose and insulin levels, slowed the proliferation of the theca interna and interstitial cells, as evidenced by Ki-67 and VEGF-A expression, and diminished CYP17 expression in the analyzed ovarian structures. In addition, metformin reduced the number of degenerating follicles and interstitial cells and improved angiogenesis. Conclusion Metformin improves the carbohydrate metabolism, reduces proliferation, and decreases CYP-17 expression in the follicular structures of androgenized rats.
Collapse
Affiliation(s)
- Roberta Rassi Mahamed
- Department of Gynecology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil.,Laboratory of Molecular and Structural Gynecology (LIM 58) of Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, Avenida Dr Arnaldo, 455- sala 2113, São Paulo, SP, Zip Code 01246903, Brazil
| | - Carla Cristina Maganhin
- Laboratory of Molecular and Structural Gynecology (LIM 58) of Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, Avenida Dr Arnaldo, 455- sala 2113, São Paulo, SP, Zip Code 01246903, Brazil.
| | - Gisela Rodrigues Silva Sasso
- Department of Morphology and Genetics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Manuel de Jesus Simões
- Department of Morphology and Genetics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Maria Candida Pinheiro Baracat
- Laboratory of Molecular and Structural Gynecology (LIM 58) of Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, Avenida Dr Arnaldo, 455- sala 2113, São Paulo, SP, Zip Code 01246903, Brazil
| | - Edmund Chada Baracat
- Laboratory of Molecular and Structural Gynecology (LIM 58) of Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, Avenida Dr Arnaldo, 455- sala 2113, São Paulo, SP, Zip Code 01246903, Brazil
| | - José Maria Soares-
- Laboratory of Molecular and Structural Gynecology (LIM 58) of Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, Avenida Dr Arnaldo, 455- sala 2113, São Paulo, SP, Zip Code 01246903, Brazil
| |
Collapse
|
20
|
Jiang J, Qi L, Wei Q, Shi F. Effects of daily exposure to saccharin sodium and rebaudioside A on the ovarian cycle and steroidogenesis in rats. Reprod Toxicol 2018; 76:35-45. [PMID: 29262312 DOI: 10.1016/j.reprotox.2017.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 10/18/2022]
|
21
|
Volk KM, Pogrebna VV, Roberts JA, Zachry JE, Blythe SN, Toporikova N. High-Fat, High-Sugar Diet Disrupts the Preovulatory Hormone Surge and Induces Cystic Ovaries in Cycling Female Rats. J Endocr Soc 2017; 1:1488-1505. [PMID: 29308444 PMCID: PMC5740526 DOI: 10.1210/js.2017-00305] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/30/2017] [Indexed: 12/16/2022] Open
Abstract
Diet-induced obesity has been associated with various metabolic and reproductive disorders, including polycystic ovary syndrome. However, the mechanisms by which obesity influences the reproductive system are still not fully known. Studies have suggested that impairments in hormone signaling are associated with the development of symptoms such as acyclicity and ovarian cysts. However, these studies have often failed to address how these hormonal changes arise and how they might contribute to the progression of reproductive diseases. In the present study, we used a high-fat, high-sugar (HFHS) diet to induce obesity in a female rodent model to determine the changes in critical reproductive hormones that might contribute to the development of irregular estrous cycling and reproductive cycle termination. The HFHS animals exhibited impaired estradiol, progesterone (P4), and luteinizing hormone (LH) surges before ovulation. The HFHS diet also resulted in altered basal levels of testosterone (T) and LH. Furthermore, alterations in the basal P4/T ratio correlated strongly with ovarian cyst formation in HFHS rats. Thus, this model provides a method to assess the underlying etiology of obesity-related reproductive dysfunction and to examine an acyclic reproductive phenotype as it develops.
Collapse
Affiliation(s)
- Katrina M. Volk
- Neuroscience Program, Washington and Lee University, Lexington, Virginia 24450
| | | | - Jackson A. Roberts
- Neuroscience Program, Washington and Lee University, Lexington, Virginia 24450
| | - Jennifer E. Zachry
- Neuroscience Program, Washington and Lee University, Lexington, Virginia 24450
| | - Sarah N. Blythe
- Neuroscience Program, Washington and Lee University, Lexington, Virginia 24450
- Department of Biology, Washington and Lee University, Lexington, Virginia 24450
| | - Natalia Toporikova
- Neuroscience Program, Washington and Lee University, Lexington, Virginia 24450
- Department of Biology, Washington and Lee University, Lexington, Virginia 24450
| |
Collapse
|
22
|
Huang Y, Gao JM, Zhang CM, Zhao HC, Zhao Y, Li R, Yu Y, Qiao J. Assessment of growth and metabolism characteristics in offspring of dehydroepiandrosterone-induced polycystic ovary syndrome adults. Reproduction 2017; 152:705-714. [PMID: 27798284 PMCID: PMC5097128 DOI: 10.1530/rep-16-0081] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 09/19/2016] [Indexed: 11/17/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common reproductive disorder that has many characteristic features including hyperandrogenemia, insulin resistance and obesity, which may have significant implications for pregnancy outcomes and long-term health of women. Daughters born to PCOS mothers constitute a high-risk group for metabolic and reproductive derangements, but no report has described potential growth and metabolic risk factors for such female offspring. Hence, we used a mouse model of dehydroepiandrosterone (DHEA)-induced PCOS to study the mechanisms underlying the pathology of PCOS by investigating the growth, developmental characteristics, metabolic indexes and expression profiles of key genes of offspring born to the models. We found that the average litter size was significantly smaller in the DHEA group, and female offspring had sustained higher body weight, increased body fat and triglyceride content in serum and liver; they also exhibited decreased energy expenditure, oxygen consumption and impaired glucose tolerance. Genes related to glucolipid metabolism such as Pparγ, Acot1/2, Fgf21, Pdk4 and Inhbb were upregulated in the liver of the offspring in DHEA group compared with those in controls, whereas Cyp17a1 expression was significantly decreased. However, the expression of these genes was not detected in male offspring. Our results show that female offspring in DHEA group exhibit perturbed growth and glucolipid metabolism that were not observed in male offspring.
Collapse
Affiliation(s)
- Ying Huang
- Reproductive Medical CenterDepartment of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,Key Laboratory of Assisted ReproductionMinistry of Education, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijing, China
| | - Jiang-Man Gao
- Reproductive Medical CenterDepartment of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijing, China
| | - Chun-Mei Zhang
- Reproductive Medical CenterDepartment of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,Key Laboratory of Assisted ReproductionMinistry of Education, Beijing, China
| | - Hong-Cui Zhao
- Reproductive Medical CenterDepartment of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijing, China
| | - Yue Zhao
- Reproductive Medical CenterDepartment of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China .,Key Laboratory of Assisted ReproductionMinistry of Education, Beijing, China
| | - Rong Li
- Reproductive Medical CenterDepartment of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China .,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijing, China
| | - Yang Yu
- Reproductive Medical CenterDepartment of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China .,Key Laboratory of Assisted ReproductionMinistry of Education, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijing, China
| | - Jie Qiao
- Reproductive Medical CenterDepartment of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,Key Laboratory of Assisted ReproductionMinistry of Education, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyBeijing, China
| |
Collapse
|
23
|
Liu Y, Du SY, Ding M, Dou X, Zhang FF, Wu ZY, Qian SW, Zhang W, Tang QQ, Xu CJ. The BMP4-Smad signaling pathway regulates hyperandrogenism development in a female mouse model. J Biol Chem 2017; 292:11740-11750. [PMID: 28572510 DOI: 10.1074/jbc.m117.781369] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/31/2017] [Indexed: 11/06/2022] Open
Abstract
Polycystic ovary syndrome is a common endocrine disorder and a major cause of anovulatory sterility in women at reproductive age. Most patients with polycystic ovary syndrome have hyperandrogenism, caused by excess androgen synthesis. Bone morphogenetic protein 4 (BMP4) is an essential regulator of embryonic development and organ formation, and recent studies have also shown that BMP4 may be involved in female steroidogenesis process. However, the effect of BMP4 on hyperandrogenism remains unknown. Here, using a female mouse model of hyperandrogenism, we found that ovarian BMP4 levels were significantly decreased in hyperandrogenism. Elevated androgens inhibited BMP4 expression via activation of androgen receptors. Moreover, BMP4 treatment suppressed androgen synthesis in theca cells and promoted estrogen production in granulosa cells by regulating the expression of steroidogenic enzymes, including CYP11A, HSD3B2, CYP17A1, and CYP19A1 Consistently, knockdown of BMP4 augmented androgen levels and inhibited estrogen levels. Mechanistically, Smad signaling rather than the p38 MAPK pathway regulated androgen and estrogen formation, thereby mediating the effect of BMP4. Of note, BMP4-transgenic mice were protected against hyperandrogenism. Our observations clarify a vital role of BMP4 in controlling sex hormone levels and offer new insights into intervention for managing hyperandrogenism by targeting the BMP4-Smad signaling pathway.
Collapse
Affiliation(s)
- Yang Liu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, China; Key Laboratory of Metabolism and Molecular Medicine, the Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Shao-Yue Du
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, China
| | - Meng Ding
- Key Laboratory of Metabolism and Molecular Medicine, the Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Xin Dou
- Key Laboratory of Metabolism and Molecular Medicine, the Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Fei-Fei Zhang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, China
| | - Zhi-Yong Wu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, China
| | - Shu-Wen Qian
- Key Laboratory of Metabolism and Molecular Medicine, the Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Wei Zhang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, China
| | - Qi-Qun Tang
- Key Laboratory of Metabolism and Molecular Medicine, the Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai 200032, China.
| | - Cong-Jian Xu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, China.
| |
Collapse
|
24
|
Zhang Y, Hu M, Meng F, Sun X, Xu H, Zhang J, Cui P, Morina N, Li X, Li W, Wu XK, Brännström M, Shao R, Billig H. Metformin Ameliorates Uterine Defects in a Rat Model of Polycystic Ovary Syndrome. EBioMedicine 2017; 18:157-170. [PMID: 28336389 PMCID: PMC5405166 DOI: 10.1016/j.ebiom.2017.03.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 03/14/2017] [Accepted: 03/14/2017] [Indexed: 01/24/2023] Open
Abstract
Adult rats treated concomitantly with insulin and human chorionic gonadotropin exhibit endocrine, metabolic, and reproductive abnormalities that are very similar to those observed in polycystic ovary syndrome (PCOS) patients. In this study, we used this rat model to assess the effects of metformin on PCOS-related uterine dysfunction. In addition to reducing androgen levels, improving insulin sensitivity, and correcting the reproductive cycle, metformin treatment induced morphological changes in the PCOS-like uterus. At the molecular and cellular levels, metformin normalized the androgen receptor-mediated transcriptional program and restored epithelial–stromal interactions. In contrast to glucose transport, uterine inflammatory gene expression was suppressed through the PI3K–Akt–NFκB network, but without affecting apoptosis. These effects appeared to be independent of AMPK subunit and autophagy-related protein regulation. We found that when metformin treatment partially restored implantation, several implantation-related genes were normalized in the PCOS-like rat uterus. These results improve our understanding of how metformin rescues the disruption of the implantation process due to the uterine defects that result from hyperandrogenism and insulin resistance. Our data provide insights into the molecular and functional clues that might help explain, at least in part, the potential therapeutic options of metformin in PCOS patients with uterine dysfunction. The therapeutic dose of metformin sufficiently suppresses hyperandrogenism and insulin resistance. Metformin inhibits uterine androgen receptor (AR)-dependent gene expression to restore epithelial–stromal interactions. Metformin reduces uterine inflammation through the PI3K–Akt–NFκB pathway. Metformin partially restores implantation in PCOS-like rats.
The systemic benefits of metformin therapy for women with polycystic ovary syndrome (PCOS) are widely appreciated, but knowledge of the molecular mechanisms of its action and to what extent it beneficially affects uterine function is limited. Using a PCOS-like rat model, we show that treatment with metformin can reverse the negative effects of androgenic and inflammatory conditions in the rat uterus. Importantly, we find that the sustained benefit of metformin is to rescue implantation failure in some PCOS-like rats. Thus, our data will be of translational value in the clinical management of metformin treatment in PCOS patients with uterine dysfunction.
Collapse
Affiliation(s)
- Yuehui Zhang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China; Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Min Hu
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Fanci Meng
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - Xiaoyan Sun
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - Hongfei Xu
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - Jiao Zhang
- Department of Acupuncture and Moxibustion, Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150001 Harbin, China
| | - Peng Cui
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; Department of Integrative Medicine and Neurobiology, State Key Lab of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institute of Brain Science, Fudan University, 200032 Shanghai, China
| | - Njomeza Morina
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Xin Li
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 200011 Shanghai, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, 200011 Shanghai, China
| | - Wei Li
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - Xiao-Ke Wu
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - Mats Brännström
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Ruijin Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden.
| | - Håkan Billig
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| |
Collapse
|
25
|
Garg D, Merhi Z. Relationship between Advanced Glycation End Products and Steroidogenesis in PCOS. Reprod Biol Endocrinol 2016; 14:71. [PMID: 27769286 PMCID: PMC5073880 DOI: 10.1186/s12958-016-0205-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 10/14/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Women with PCOS have elevated levels of the harmful Advanced Glycation End Products (AGEs), which are highly reactive molecules formed after glycation of lipids and proteins. Additionally, AGEs accumulate in the ovaries of women with PCOS potentially contributing to the well-documented abnormal steroidogenesis and folliculogenesis. MAIN BODY A systematic review of articles and abstracts available in PubMed was conducted and presented in a systemic manner. This article reports changes in steroidogenic enzyme activity in granulosa and theca cells in PCOS and PCOS-models. It also described the changes in AGEs and their receptors in the ovaries of women with PCOS and presents the underlying mechanism(s) whereby AGEs could be responsible for the PCOS-related changes in granulosa and theca cell function thus adversely impacting steroidogenesis and follicular development. AGEs are associated with hyperandrogenism in PCOS possibly by altering the activity of various enzymes such as cholesterol side-chain cleavage enzyme cytochrome P450, steroidogenic acute regulatory protein, 17α-hydroxylase, and 3β-hydroxysteroid dehydrogenase. AGEs also affect luteinizing hormone receptor and anti-Mullerian hormone receptor expression as well as their signaling pathways in granulosa cells. CONCLUSIONS A better understanding of how AGEs alter granulosa and theca cell function is likely to contribute meaningfully to a conceptual framework whereby new interventions to prevent and/or treat ovarian dysfunction in PCOS can ultimately be developed.
Collapse
Affiliation(s)
- Deepika Garg
- Department of Obstetrics and Gynecology, Maimonides Medical Center, Brooklyn, NY 11219 USA
| | - Zaher Merhi
- Division of Reproductive Biology, Department of Obstetrics and Gynecology, NYU School of Medicine, 180 Varick Street, sixth floor, New York City, NY 11014 USA
| |
Collapse
|
26
|
Zhang Y, Sun X, Sun X, Meng F, Hu M, Li X, Li W, Wu XK, Brännström M, Shao R, Billig H. Molecular characterization of insulin resistance and glycolytic metabolism in the rat uterus. Sci Rep 2016; 6:30679. [PMID: 27461373 PMCID: PMC4962087 DOI: 10.1038/srep30679] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 07/07/2016] [Indexed: 12/29/2022] Open
Abstract
Peripheral insulin resistance and hyperandrogenism are the primary features of polycystic ovary syndrome (PCOS). However, how insulin resistance and hyperandrogenism affect uterine function and contribute to the pathogenesis of PCOS are open questions. We treated rats with insulin alone or in combination with human chorionic gonadotropin (hCG) and showed that peripheral insulin resistance and hyperandrogenism alter uterine morphology, cell phenotype, and cell function, especially in glandular epithelial cells. These defects are associated with an aberration in the PI3K/Akt signaling pathway that is used as an indicator for the onset of insulin resistance in classical metabolic tissues. Concomitantly, increased GSK3β (Ser-9) phosphorylation and decreased ERK1/2 phosphorylation in rats treated with insulin and hCG were also observed. We also profiled the expression of glucose transporter (Glut) isoform genes in the uterus under conditions of insulin resistance and/or hyperandrogenism. Finally, we determined the expression pattern of glycolytic enzymes and intermediates during insulin resistance and hyperandrogenism in the uterus. These findings suggest that the PI3K/Akt and MAPK/ERK signaling pathways play a role in the onset of uterine insulin resistance, and they also suggest that changes in specific Glut isoform expression and alterations to glycolytic metabolism contribute to the endometrial dysfunction observed in PCOS patients.
Collapse
Affiliation(s)
- Yuehui Zhang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China.,Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Xue Sun
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - Xiaoyan Sun
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - Fanci Meng
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - Min Hu
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Xin Li
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden.,Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 200011 Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, 200011 Shanghai, China
| | - Wei Li
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - Xiao-Ke Wu
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, 150040 Harbin, China
| | - Mats Brännström
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital at Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Ruijin Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Håkan Billig
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| |
Collapse
|
27
|
Ambrosetti V, Guerra M, Ramírez LA, Reyes A, Álvarez D, Olguín S, González-Mañan D, Fernandois D, Sotomayor-Zárate R, Cruz G. Increase in endogenous estradiol in the progeny of obese rats is associated with precocious puberty and altered follicular development in adulthood. Endocrine 2016; 53:258-70. [PMID: 26767652 DOI: 10.1007/s12020-016-0858-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 01/04/2016] [Indexed: 01/07/2023]
Abstract
Maternal obesity during pregnancy has been related with several pathological states in offspring. However, the impact of maternal obesity on reproductive system on the progeny is beginning to be elucidated. In this work, we characterize the effect of maternal obesity on puberty onset and follicular development in adult offspring in rats. We also propose that alterations in ovarian physiology observed in offspring of obese mothers are due to increased levels of estradiol during early development. Offspring of control dams and offspring of dams exposed to a high-fat diet (HF) were studied at postnatal days (PND) 1, 7, 14, 30, 60, and 120. Body weight and onset of puberty were measured. Counting of ovarian follicles was performed at PND 60 and 120. Serum estradiol, estriol, androstenedione, FSH, LH, and insulin levels were measured by ELISA. Hepatic CYP3A2 expression was determined by Western blot. HF rats had a higher weight than controls at all ages and they also had a precocious puberty. Estradiol levels were increased while CYP3A2 expression was reduced from PND 1 until PND 60 in HF rats compared to controls. Estriol was decreased at PND60 in HF rats. Ovaries from HF rats had a decrease in antral follicles at PND60 and PND120 and an increase in follicular cysts at PND60 and PND120. In this work, we demonstrated that maternal obesity in rats alters follicular development and induces follicular cysts generation in the adult offspring. We observed that maternal obesity produces an endocrine disruption through increasing endogenous estradiol in early life. A programmed failure in hepatic metabolism of estradiol is probably the cause of its increase.
Collapse
Affiliation(s)
- Valery Ambrosetti
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Facultad de Ciencias, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, 2360102, Valparaiso, Chile
| | - Marcelo Guerra
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Facultad de Ciencias, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, 2360102, Valparaiso, Chile
| | - Luisa A Ramírez
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Facultad de Ciencias, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, 2360102, Valparaiso, Chile
| | - Aldo Reyes
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Facultad de Ciencias, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, 2360102, Valparaiso, Chile
| | - Daniela Álvarez
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Facultad de Ciencias, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, 2360102, Valparaiso, Chile
| | - Sofía Olguín
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Facultad de Ciencias, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, 2360102, Valparaiso, Chile
| | - Daniel González-Mañan
- Molecular and Clinical Pharmacology Program, Faculty of Medicine, Institute of Biomedical Sciences, University of Chile, 8380492, Santiago, Chile
| | - Daniela Fernandois
- Programa de Doctorado en Farmacología, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, 8380492, Santiago, Chile
| | - Ramón Sotomayor-Zárate
- Laboratorio de Neuroquímica y Neurofarmacología, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Facultad de Ciencias, Instituto de Fisiología, Universidad de Valparaíso, 2360102, Valparaiso, Chile
| | - Gonzalo Cruz
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Facultad de Ciencias, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, 2360102, Valparaiso, Chile.
| |
Collapse
|
28
|
Baskind NE, Balen AH. Hypothalamic-pituitary, ovarian and adrenal contributions to polycystic ovary syndrome. Best Pract Res Clin Obstet Gynaecol 2016; 37:80-97. [PMID: 27137106 DOI: 10.1016/j.bpobgyn.2016.03.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/05/2016] [Indexed: 11/18/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent heterogeneous disorder linked with disturbances of reproductive, endocrine and metabolic function. The definition and aetiological hypotheses of PCOS are continually developing to incorporate evolving evidence of the syndrome, which appears to be both multifactorial and polygenic. The pathophysiology of PCOS encompasses inherent ovarian dysfunction that is strongly influenced by external factors including the hypothalamic-pituitary axis and hyperinsulinaemia. Neuroendocrine abnormalities including increased gonadotrophin-releasing hormone (GnRH) pulse frequency with consequent hypersecretion of luteinising hormone (LH) affects ovarian androgen synthesis, folliculogenesis and oocyte development. Disturbed ovarian-pituitary and hypothalamic feedback accentuates the gonadotrophin abnormalities, and there is emerging evidence putatively implicating dysfunction of the Kiss 1 system. Within the follicle subunit itself, there are intra-ovarian paracrine modulators, cytokines and growth factors, which appear to play a role. Adrenally derived androgens may also contribute to the pathogenesis of PCOS, but their role is less defined.
Collapse
Affiliation(s)
- N Ellissa Baskind
- The Leeds Centre for Reproductive Medicine, Seacroft Hospital, York Road, Leeds LS14 6UH, UK.
| | - Adam H Balen
- The Leeds Centre for Reproductive Medicine, Seacroft Hospital, York Road, Leeds LS14 6UH, UK.
| |
Collapse
|
29
|
Zhang Q, Liu D, Zhang M, Li N, Lu S, Du Y, Chen ZJ. Effects of brain-derived neurotrophic factor on oocyte maturation and embryonic development in a rat model of polycystic ovary syndrome. Reprod Fertil Dev 2016; 28:1904-1915. [DOI: 10.1071/rd15131] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 05/21/2015] [Indexed: 01/09/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is expressed extensively in the mammalian female reproductive system and has been implicated in the development of follicles and oocytes. However, BDNF expression patterns in the ovary and its effects on oocyte maturation and embryonic development in polycystic ovary syndrome (PCOS) have not been established. In the present study, we established a PCOS model by treating the rats with insulin and human chorionic gonadotropin (hCG). Rats treated with insulin + hCG had heavier bodyweight and ovarian weight, higher circulating concentrations of luteinising hormone (LH) and testosterone (T), and greater homeostatic model assessment of insulin resistance (HOMA-IR) values compared with control rats (P < 0.05). BDNF and its receptor tyrosine kinase type B (TrkB) were located in cyst walls, granulosa and theca cells, and BDNF protein levels were lower in ovaries of insulin + hCG-treated rats (P < 0.05). The rate of oocyte maturation and formation of blastocysts and morulae was greatest in rats treated with 5 ng mL–1 BDNF (P < 0.05) compared to other BDNF groups (1 and 10 ng mL–1) and the control. The control rats were also PCOS rats and were treated without BDNF. There were no significant differences in the rate of germinal vesicle breakdown (GVBD) and fertilisation among the various treatment groups (1, 5 and 10 ng mL–1) and the control group (P > 0.05). The results indicate that in vitro treatment with an appropriate concentration of BDNF not only promotes oocyte maturation, but also rescues embryonic development in rats treated with insulin + hCG as a model of PCOS.
Collapse
|
30
|
Liu W, Liu W, Fu Y, Wang Y, Zhang Y. Bak Foong pills combined with metformin in the treatment of a polycystic ovarian syndrome rat model. Oncol Lett 2015; 10:1819-1825. [PMID: 26622758 DOI: 10.3892/ol.2015.3466] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 04/14/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the treatment effects and associated mechanism of Bak Foong pills (BFPs) combined with metformin in the treatment of polycystic ovarian syndrome (PCOS). BFPs and/or metformin were administrated to treat the PCOS rats, and the weights and morphologies of the ovary, uterus and adrenal gland were measured. The levels of fasting blood glucose (FBG), serum testosterone (T), luteinizing hormone, fasting insulin (FIN) and insulin-like growth factor-1 were also measured, and the homeostasis model assessment of insulin resistance (HOMA-IR) was calculated. The expression level of androgen receptor (AR) in the ovarian tissue, and the cytochrome P450 cholesterol side-chain cleavage enzyme (P450scc) mRNA levels in the ovary and adrenal tissues were detected. The levels of T, FIN, FBG and HOMA-IR in the combination group were significantly reduced; the wet weights of the ovary and the adrenal gland were decreased significantly, while that of the uterus was increased, and the histological morphology benignly recovered. The rats of each treatment group all experienced restored ovulation. The AR expression level in the treatment group was reduced, and the P450scc mRNA levels in the ovary and the adrenal gland of the combined treatment group were decreased. BFPs combined with metformin significantly affected PCOS, and the possible mechanism involved in the treatment may have been through the reduction of P450scc generation. BFPs may reduce the androgen levels, thus allowing the ovary to restore ovulation.
Collapse
Affiliation(s)
- Wenhui Liu
- Center of Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wenpei Liu
- Translational Medicine Institute, Affiliated to The First People's Hospital of Chenzhou, University of South China, Chenzhou, Hunan 423000, P.R. China
| | - Yuling Fu
- Department of Obstetrics and Gynecology, The Second Hospital of Yinzhou, Ningbo, Zhejiang 315000, P.R. China
| | - Yan Wang
- Center of Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yuanzhen Zhang
- Center of Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
31
|
Wu B, Shi Y, Gong X, Yu L, Chen Q, Wang J, Sun Z. Evaluation of follicular synchronization caused by estrogen administration and its reproductive outcome. PLoS One 2015; 10:e0127595. [PMID: 26010950 PMCID: PMC4444187 DOI: 10.1371/journal.pone.0127595] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/16/2015] [Indexed: 11/18/2022] Open
Abstract
To evaluate multiple follicular development synchronization after estrogen stimulation in prepubertal mice, follicular responsiveness to gonadotropin superovulation, the prospective reproductive potential and ovarian polycystic ovary syndrome (PCOS)-like symptoms at adulthood, prepubertal mice were intraperitoneally injected with estrogen to establish an animal model with solvent as control. When synchronized tertiary follicles in ovaries, in vitro oocyte maturation and fertilization rates, blastocyst formation rate, developmental potential into offspring by embryo transfer, adult fertility and PCOS-like symptoms, and involved molecular mechanisms were focused, it was found that estrogen stimulation (10 μg/gBW) leads to follicular development synchronization at the early tertiary stage in prepubertal mice; reproduction from oocytes to offspring could be realized by means of the artificial reproductive technology though the model mice lost their natural fertility when they were reared to adulthood; and typical symptoms of PCOS, except changes in inflammatory pathways, were not remained up to adulthood. So in conclusion, estrogen can lead to synchronization in follicular development in prepubertal mice, but does not affect reproductive outcome of oocytes, and no typical symptoms of PCOS remained at adulthood despite changes related to inflammation.
Collapse
Affiliation(s)
- Bi Wu
- Shanghai Medical College, Institute of Reproduction and Development, Fudan University, Shanghai, P. R. China
| | - Yan Shi
- Key Laboratory of Contraceptive Drugs and Devices of National Population and Family Planning Commission of China, Shanghai Institute of Planned Parenthood Research, Shanghai, P. R. China
| | - Xia Gong
- Department of Food Science, Shanghai Business School, Shanghai, P. R. China
| | - Lin Yu
- Key Laboratory of Contraceptive Drugs and Devices of National Population and Family Planning Commission of China, Shanghai Institute of Planned Parenthood Research, Shanghai, P. R. China
| | - Qiuju Chen
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China
| | - Jian Wang
- Key Laboratory of Contraceptive Drugs and Devices of National Population and Family Planning Commission of China, Shanghai Institute of Planned Parenthood Research, Shanghai, P. R. China
| | - Zhaogui Sun
- Key Laboratory of Contraceptive Drugs and Devices of National Population and Family Planning Commission of China, Shanghai Institute of Planned Parenthood Research, Shanghai, P. R. China
- * E-mail:
| |
Collapse
|
32
|
Xia Y, Shen S, Zhang X, Deng Z, Xiang Z, Wang H, Yi L, Gao Q, Wang Y. Epigenetic pattern changes in prenatal female Sprague-Dawley rats following exposure to androgen. Reprod Fertil Dev 2015; 28:RD14292. [PMID: 25823942 DOI: 10.1071/rd14292] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 01/28/2015] [Indexed: 12/31/2022] Open
Abstract
Androgen excess is generally considered to be one of the major characteristics of polycystic ovary syndrome (PCOS). Evidence from both clinical research and animal studies has revealed that this syndrome may have fetal origins, with epigenetics being proposed as the underlying mechanism. Our PCOS rat model induced by prenatal administration of 3mg testosterone from Embryonic Day (E) 16 to E19 showed polycystic ovaries, irregular oestrous cycles and endocrine disorders in adulthood. The methylation status of 16, 8 and 4 cytosine-phosphate-guanine (CpG) sites in the promoter regions of the androgen receptor (Ar), cytochrome P450 family 11, subfamily A, polypeptide 1 (Cyp11a1) and cytochrome P450, family 17, subfamily A, polypeptide 1 (Cyp17a1) genes, respectively, were measured by pyrosequencing. We identified three hypomethylated sites (CpG +58, +65 and +150) in Ar and one hypomethylated site (CpG +1016) in Cyp11a1 in peripheral blood cells of prenatally androgenised (PNA) rats. In ovarian tissue, five CpG sites of Ar (CpG +87, +91, +93, +98, +150) and one single CpG site in Cyp11a1 (CpG +953) were significantly hypomethylated in PNA rats, but the modified methylation of these two genes may not be sufficient to significantly alter levels of gene expression. Furthermore, tissue-specific methylation analysis revealed that both Ar and Cyp11a1 exhibited significant hypomethylation in testis in contrast with ovary and blood. PNA may lead to methylation pattern changes and the development of PCOS, but further studies are required to reveal causal relationships.
Collapse
|
33
|
Fu H, Qiu Y, Xia M, Wei F, He H, Yang L. Spleen-yang-deficiency patients with polycystic ovary syndrome have higher levels of visfatin. J TRADIT CHIN MED 2014; 34:42-7. [PMID: 25102689 DOI: 10.1016/s0254-6272(14)60052-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To study serum visfatin levels in women with polycystic ovary syndrome (PCOS) grouped by Traditional Chinese Medicine (TCM) patterns. To study the correlations of serum visfatin levels with homeostatic model assessment insulin resistance (HOMA-IR), fasting plasma glucose (FPG), fasting insulin (FINS), body mass index (BMI), testosterone (T), total cholesterol (TC), and triglycerides (TG). METHODS Two hundred and twelve PCOS patients were placed into the following TCM pattern subgroups: Kidney-Yang deficiency (KYD) group, Spleen-Yang deficiency (SYD) group, stagnant Liver-Qi transforming into heat (SLQTH) group, and Kidney-Yin deficiency (KYIND) group. The correlations between serum visfatin levels and HOMA-IR, FPG, FINS, BMI, T, TC, and TG were analyzed. RESULTS Of all patients with PCOS, there were 82 in the KYD group (38.6%), 67 in the SYD group (31.6%), 37 in the SLQTH group (17.5%), and 26 in the KYIND group (12.3%). Visfatin levels in all PCOS subgroups were higher than those in the control group (P < 0.01 or P < 0.05). Among these subgroups, the visfatin levels in the SYD group were significantly higher than those in the other three TCM pattern groups (P < 0.05). There were no statistical differences among the remaining three pattern groups. The levels of BMI, FINS, HOMA-IR, T, and TG were significantly higher in all subgroups than those in the control group (P < 0.05). There were no significant differences in FPG and TC between all PCOS subgroups and the control group (P > 0.05). The SYD group had higher levels of FINS and HOMA-IR compared with the KYD, SLQTH, and KYIND groups (P < 0.05). In all subgroups, after controlling for BMI, TG, TC, and age, visfatin was positively correlated with FINS (r = 0.197, P = 0.015) and HOMA-IR (r = 0.173, P = 0.033), and was not correlated with T. CONCLUSION KYD and SYD patterns are most common in PCOS patients. Increased visfatin is a common pathophysiologic manifestation in PCOS patients. The SYD group had the highest levels of visfatin, and visfatin was positively correlated with FINS and HOMA-IR.
Collapse
|
34
|
Pohl A, Cassidy S, Auyeung B, Baron-Cohen S. Uncovering steroidopathy in women with autism: a latent class analysis. Mol Autism 2014; 5:27. [PMID: 24717046 PMCID: PMC4022124 DOI: 10.1186/2040-2392-5-27] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 03/10/2014] [Indexed: 11/30/2022] Open
Abstract
Background Prenatal exposure to increased androgens has been implicated in both polycystic ovary syndrome (PCOS) and autism spectrum conditions (ASC), suggesting that PCOS may be increased among women with ASC. One study suggested elevated steroidopathic symptoms (‘steroidopathy’) in women with ASC. As the symptoms are not independent, we conducted a latent class analysis (LCA). The objectives of the current study are: (1) to test if these findings replicate in a larger sample; and (2) to use LCA to uncover affected clusters of women with ASC. Methods We tested two groups of women, screened using the Autism Spectrum Quotient - Group 1: n = 415 women with ASC (mean age 36.39 ± 11.98 years); and Group 2: n = 415 controls (mean age 39.96 ± 11.92 years). All participants completed the Testosterone-related Medical Questionnaire online. A multiple-group LCA was used to identify differences in latent class structure between women with ASC and controls. Results There were significant differences in frequency of steroid-related conditions and symptoms between women with ASC and controls. A two-class semi-constrained model best fit the data. Based on response patterns, we identified the classes as ‘Typical’ and ‘Steroidopathic’. The prevalence of the ‘Steroidopathic’ class was significantly increased within the ASC group (ΔG2 = 15, df =1, P = 0.0001). In particular, we confirmed higher frequencies of epilepsy, amenorrhea, dysmenorrhea, severe acne, gender dysphoria, and transsexualism, and differences in sexual preference in women with ASC. Conclusions Women with ASC are at increased risk for symptoms and conditions linked to steroids. LCA revealed this steroidopathy despite the apparent underdiagnosis of PCOS.
Collapse
Affiliation(s)
- Alexa Pohl
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge CB2 8AH, UK
| | - Sarah Cassidy
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge CB2 8AH, UK ; Department of Psychology and Behavioural Sciences, Coventry University, James Starley Building, Cox Street, Coventry CV1 5LW, UK
| | - Bonnie Auyeung
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge CB2 8AH, UK ; Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh EH8 9 AD, UK
| | - Simon Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge CB2 8AH, UK ; CLASS Clinic, Cambridgeshire and Peterborough Mental Health Foundation NHS Trust, The Chitra Sethia Autism Centre, The Gatehouse, Fulborn Hospital, Fulborn, Cambridge CB21 5EF, UK
| |
Collapse
|