1
|
Mehta P, Saha D, Das A, Das BK. Gut microbiota in diabetic-linked polycystic ovarian syndrome: Mechanisms and therapeutic insights. Tissue Cell 2025; 95:102870. [PMID: 40154106 DOI: 10.1016/j.tice.2025.102870] [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: 11/05/2024] [Revised: 01/16/2025] [Accepted: 03/15/2025] [Indexed: 04/01/2025]
Abstract
Polycystic ovarian syndrome (PCOS) is a complex multisystem disorder prevalent among women of reproductive age, commonly marked by insulin resistance, hyperinsulinemia, and metabolic disruptions such as hypertension and dyslipidemia, which elevate risks of cardiovascular disease and hepatic steatosis. Recent advances underscore the gut microbiome's critical role in modulating insulin resistance and metabolic homeostasis in PCOS. This review highlights novel insights into gut dysbiosis-driven inflammation, gut-brain hormonal signaling, and immune modulation as underlying mechanisms connecting PCOS with metabolic dysfunction and diabetes. We comprehensively analyzed studies up to September 2024 on gut microbiota, diabetes, PCOS, and metformin, exploring emerging perspectives on the microbiome's therapeutic potential in managing PCOS. Metformin's dual role in insulin sensitivity improvement and gut microbiome modulation is emphasized, including its indirect effects on weight management. This review also identifies gaps in current research, urging a shift toward precision therapies targeting microbiome-related pathways in PCOS. Further exploration of the gut-brain axis, pathogen-associated molecular patterns, and the need for controlled clinical trials are discussed to enhance therapeutic approaches.
Collapse
Affiliation(s)
- Prachi Mehta
- Department of Pharmacology, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Azara, Guwahati, Assam 781017, India
| | - Dipankar Saha
- Department of Pharmacology, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Azara, Guwahati, Assam 781017, India
| | - Abinash Das
- Department of Pharmacology, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Azara, Guwahati, Assam 781017, India
| | - Bhrigu Kumar Das
- Department of Pharmacology, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Azara, Guwahati, Assam 781017, India.
| |
Collapse
|
2
|
Wang L, Jin Y, Zhi Y, Li Z, Wang M, Wang B, Wang X. Effects of melatonin in polycystic ovary syndrome: is there Hippo pathway crosstalk? J Ovarian Res 2025; 18:101. [PMID: 40369589 PMCID: PMC12076993 DOI: 10.1186/s13048-025-01642-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/06/2025] [Indexed: 05/16/2025] Open
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder among reproductive women, characterized by hyperandrogenism, oligo-ovulation and polycystic ovarian morphology. Incorporating complementary medicine alongside traditional lifestyle therapies for PCOS may offer additional benefits for affected women. Melatonin (MT), a hormone secreted by the pineal gland, has emerged as a potential treatment for regulating ovarian function in PCOS. However, the specific effects and underlying mechanisms of MT on PCOS need to be elucidated. METHODS This review consolidates evidence from randomized controlled trials, original research articles, systematic reviews, and meta-analyses regarding MT supplementation in PCOS, with a particular focus on its interaction with the Hippo pathway, to provide a comprehensive overview of current knowledge. RESULTS Current evidence suggests that MT plays a role in modulating PCOS through various mechanisms and is associated with the Hippo pathway. However, several uncertainties and key limitations in the existing literature must be addressed before these treatments can be integrated into standard clinical practice. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Yuanyuan Jin
- Department of Obstetrics and Gynecology, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Yuanyuan Zhi
- Department of Obstetrics and Gynecology, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Zhenzhen Li
- Department of Pathology, Shandong Provincial Maternal and Child Health Care Hospital, Qingdao University, Jinan, 250014, China
| | - Meili Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Boda Wang
- Emergency Department, Xinji Town Central Health Center, Guanxian County, Liaocheng, 252500, China
| | - Xinbo Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
| |
Collapse
|
3
|
Li Q, Zheng T, Chen J, Li B, Zhang Q, Yang S, Shao J, Guan W, Zhang S. Exploring melatonin's multifaceted role in female reproductive health: From follicular development to lactation and its therapeutic potential in obstetric syndromes. J Adv Res 2025; 70:223-242. [PMID: 38692429 PMCID: PMC11976432 DOI: 10.1016/j.jare.2024.04.025] [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: 03/27/2024] [Accepted: 04/25/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Melatonin is mainly secreted by the pineal gland during darkness and regulates biological rhythms through its receptors in the suprachiasmatic nucleus of the hypothalamus. In addition, it also plays a role in the reproductive system by affecting the function of the hypothalamic-pituitary-gonadal axis, and by acting as a free radical scavenger thus contributing to the maintenance of the optimal physiological state of the gonads. Besides, melatonin can freely cross the placenta to influence fetal development. However, there is still a lack of overall understanding of the role of melatonin in the reproductive cycle of female mammals. AIM OF REVIEW Here we focus the role of melatonin in female reproduction from follicular development to delivery as well as the relationship between melatonin and lactation. We further summarize the potential role of melatonin in the treatment of preeclampsia, polycystic ovary syndrome, endometriosis, and ovarian aging. KEY SCIENTIFIC CONCEPTS OF REVIEW Understanding the physiological role of melatonin in female reproductive processes will contribute to the advancement of human fertility and reproductive medicine research.
Collapse
Affiliation(s)
- Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiaming Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Baofeng Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qianzi Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiayuan Shao
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
4
|
Zhao Y, Zhao X, Jiang T, Xi H, Jiang Y, Feng X. A Retrospective Review on Dysregulated Autophagy in Polycystic Ovary Syndrome: From Pathogenesis to Therapeutic Strategies. Horm Metab Res 2024. [PMID: 38565184 DOI: 10.1055/a-2280-7130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The main purpose of this article is to explore the relationship between autophagy and the pathological mechanism of PCOS, and to find potential therapeutic methods that can alleviate the pathological mechanism of PCOS by targeting autophagy. Relevant literatures were searched in the following databases, including: PubMed, MEDLINE, Web of Science, Scopus. The search terms were "autophagy", "PCOS", "polycystic ovary syndrome", "ovulation", "hyperandrogenemia", "insulin resistance", "inflammatory state", "circadian rhythm" and "treatment", which were combined according to the retrieval methods of different databases. Through analysis, we uncovered that abnormal levels of autophagy were closely related to abnormal ovulation, insulin resistance, hyperandrogenemia, and low-grade inflammation in patients with PCOS. Lifestyle intervention, melatonin, vitamin D, and probiotics, etc. were able to improve the pathological mechanism of PCOS via targeting autophagy. In conclusion, autophagy disorder is a key pathological mechanism in PCOS and is also a potential target for drug development and design.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaoxuan Zhao
- Department of Traditional Chinese Medicine (TCM) Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Tianyue Jiang
- Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hongyan Xi
- Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yuepeng Jiang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaoling Feng
- Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
5
|
Bhattacharya K, Dey R, Sen D, Paul N, Basak AK, Purkait MP, Shukla N, Chaudhuri GR, Bhattacharya A, Maiti R, Adhikary K, Chatterjee P, Karak P, Syamal AK. Polycystic ovary syndrome and its management: In view of oxidative stress. Biomol Concepts 2024; 15:bmc-2022-0038. [PMID: 38242137 DOI: 10.1515/bmc-2022-0038] [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: 10/30/2023] [Accepted: 12/11/2023] [Indexed: 01/21/2024] Open
Abstract
In the past two decades, oxidative stress (OS) has drawn a lot of interest due to the revelation that individuals with many persistent disorders including diabetes, polycystic ovarian syndrome (PCOS), cardiovascular, and other disorders often have aberrant oxidation statuses. OS has a close interplay with PCOS features such as insulin resistance, hyperandrogenism, and chronic inflammation; there is a belief that OS might contribute to the development of PCOS. PCOS is currently recognized as not only one of the most prevalent endocrine disorders but also a significant contributor to female infertility, affecting a considerable proportion of women globally. Therefore, the understanding of the relationship between OS and PCOS is crucial to the development of therapeutic and preventive strategies for PCOS. Moreover, the mechanistic study of intracellular reactive oxygen species/ reactive nitrogen species formation and its possible interaction with women's reproductive health is required, which includes complex enzymatic and non-enzymatic antioxidant systems. Apart from that, our current review includes possible regulation of the pathogenesis of OS. A change in lifestyle, including physical activity, various supplements that boost antioxidant levels, particularly vitamins, and the usage of medicinal herbs, is thought to be the best way to combat this occurrence of OS and improve the pathophysiologic conditions associated with PCOS.
Collapse
Affiliation(s)
- Koushik Bhattacharya
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Khurda Road, Bhubaneswar, Odisha, India
| | - Rajen Dey
- Department of Medical Laboratory Technology, Swami Vivekananda University, Barrackpore, West Bengal, India
| | - Debanjana Sen
- Post-Graduate Department of Physiology, Hooghly Mohsin College, Chinsurah, West-Bengal, India
| | - Nimisha Paul
- Department of General Human Physiology and Biochemistry, Hitkarini Dental College and Hospital, Jabalpur, Madhya Pradesh, India
| | - Asim Kumar Basak
- School of Allied Health Sciences, Brainware University, Barasat, West-Bengal, India
| | | | - Nandini Shukla
- Department of Anatomy, Pt. J.N.M. Medical College, Raipur, Chhattisgarh, India
| | - Gargi Ray Chaudhuri
- Department of Physiotherapy, Nopany Institute of Health Care Studies, Kolkata, West-Bengal, India
| | - Aniruddha Bhattacharya
- Department of Physiology, International Medical School, Management and Science University, Selangor, Malaysia
| | - Rajkumar Maiti
- Department of Physiology, Bankura Christian College, Bankura, West Bengal, India
| | - Krishnendu Adhikary
- Department of Interdisciplinary Science, Centurion University of Technology and Management, Khurda Road, Bhubaneswar, Odisha, India
| | - Prity Chatterjee
- Department of Biotechnology, Paramedical College, Durgapur, West Bengal, India
| | - Prithviraj Karak
- Department of Physiology, Bankura Christian College, Bankura, West Bengal, India
| | - Alak Kumar Syamal
- Post-Graduate Department of Physiology, Hooghly Mohsin College, Chinsurah, West-Bengal, India
| |
Collapse
|
6
|
Petrillo T, Semprini E, Tomatis V, Arnesano M, Ambrosetti F, Battipaglia C, Sponzilli A, Ricciardiello F, Genazzani AR, Genazzani AD. Putative Complementary Compounds to Counteract Insulin-Resistance in PCOS Patients. Biomedicines 2022; 10:biomedicines10081924. [PMID: 36009471 PMCID: PMC9406066 DOI: 10.3390/biomedicines10081924] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/25/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most frequent endocrine-metabolic disorder among women at reproductive age. The diagnosis is based on the presence of at least two out of three criteria of the Rotterdam criteria (2003). In the last decades, the dysmetabolic aspect of insulin resistance and compensatory hyperinsulinemia have been taken into account as the additional key features in the etiopathology of PCOS, and they have been widely studied. Since PCOS is a complex and multifactorial syndrome with different clinical manifestations, it is difficult to find the gold standard treatment. Therefore, a great variety of integrative treatments have been reported to counteract insulin resistance. PCOS patients need a tailored therapeutic strategy, according to the patient’s BMI, the presence or absence of familiar predisposition to diabetes, and the patient’s desire to achieve pregnancy or not. The present review analyzes and discloses the main clinical insight of such complementary substances.
Collapse
Affiliation(s)
- Tabatha Petrillo
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Elisa Semprini
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Veronica Tomatis
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Melania Arnesano
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Fedora Ambrosetti
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Christian Battipaglia
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Alessandra Sponzilli
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Francesco Ricciardiello
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Andrea R. Genazzani
- Department of Obstetrics and Gynecology, University of Pisa, 56126 Pisa, Italy
| | - Alessandro D. Genazzani
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
- Correspondence:
| |
Collapse
|
7
|
Cheng X, He B. Clinical and Biochemical Potential of Antioxidants in Treating Polycystic Ovary Syndrome. Int J Womens Health 2022; 14:467-479. [PMID: 35392500 PMCID: PMC8982783 DOI: 10.2147/ijwh.s345853] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 03/18/2022] [Indexed: 11/23/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common cause of infertility in reproductive-age women. Increased reactive oxygen species levels and decreased antioxidant capacity in PCOS patients can lead to metabolic disorders and damage the ovarian tissues, resulting in the occurrence of related symptoms. Antioxidants have been used in the treatment of PCOS and have yielded satisfactory outcomes due to their ability to counter oxidative stress. Many experiments on PCOS patients have proved that antioxidants can not only improve the ovarian environment, promote follicular maturation, and elevate oocyte quantities but can also regulate lipid and glucose metabolism as well as vascular endothelial cell function in PCOS patients, thereby attenuating adiposity and reducing the occurrence rate of chronic complications to ensure that patients can obtain long-term benefits. This review describes the use of antioxidants in PCOS, which have been used in the treatment.
Collapse
Affiliation(s)
- Xiangyi Cheng
- Department of Endocrinology, Shengjing Hospital, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
| | - Bing He
- Department of Endocrinology, Shengjing Hospital, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
| |
Collapse
|
8
|
Sudhakaran G, Guru A, Hari Deva Muthu B, Murugan R, Arshad A, Arockiaraj J. Evidence-based hormonal, mutational, and endocrine-disrupting chemical-induced zebrafish as an alternative model to study PCOS condition similar to mammalian PCOS model. Life Sci 2022; 291:120276. [PMID: 34990650 DOI: 10.1016/j.lfs.2021.120276] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/15/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022]
Abstract
Polycystic ovarian syndrome (PCOS) causes swollen ovaries in women at reproductive age due to hormonal disorder with small cysts on the outer edges. The cause of the disorder is still yet to be found. Multiple factors have increased PCOS prevalence, hyperandrogenism, oxidative stress, inflammation, and insulin resistance. Various animal PCOS models have been developed to imitate the pathophysiology of PCOS in humans. Zebrafish is one of the most versatile animal experimental models because of the transparency of the embryos, small size, and rapid growth. The zebrafish similarity to higher vertebrates made it a useful non-mammalian model for PCOS drug testing and screening. This review provides an insight into the usage of zebrafish, a non-mammalian model for PCOS, as an opportunity for evaluating future initiatives in such a research domain.
Collapse
Affiliation(s)
- Gokul Sudhakaran
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - Ajay Guru
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - B Hari Deva Muthu
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - Raghul Murugan
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - Aziz Arshad
- International Institute of Aquaculture and Aquatic Sciences (I-AQUAS), Universiti Putra Malaysia, 71050 Port Dickson, Negeri Sembilan, Malaysia
| | - Jesu Arockiaraj
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India; Foundation for Aquaculture Innovations and Technology Transfer (FAITT), Thoraipakkam, Chennai 600 097, Tamil Nadu, India.
| |
Collapse
|
9
|
Guo R, Zheng H, Li Q, Qiu X, Zhang J, Cheng Z. Melatonin alleviates insulin resistance through the PI3K/AKT signaling pathway in ovary granulosa cells of polycystic ovary syndrome. Reprod Biol 2021; 22:100594. [PMID: 34953312 DOI: 10.1016/j.repbio.2021.100594] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 12/19/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine gynecological disorder. Insulin resistance (IR) is a major cause of PCOS. Melatonin, a critical endogenous hormone, has beneficial effects on the female reproductive system. This study aims to investigate the molecular effect of melatonin on IR in human ovarian granulosa cells (GCs). Hormone levels of the subjects were determined through clinical examination. The expression levels of insulin receptor substrate (IRS)-1 and glucose transporter (GLUT4) in GCs from PCOS patients and a human granulosa cell line (SVOG) were examined using qRT-PCR and western blot. The IR cell model was established by inducing SVOG cells with palmitic acid (PA). IR was detected in GCs of PCOS patients and SVOG by measuring glucose content and glucose uptake. Cell viability and apoptosis levels were detected by CCK-8 assay and flow cytometry. PI3K/Akt pathway expression in SVOG was assessed by western blot. PCOS patients had higher levels of luteinizing hormone (LH), testosterone, and LH/follicle-stimulating hormone. PA decreased cell viability, promoted apoptosis, and reduced glucose uptake in SVOG cells. IRS-1 and GLUT4 mRNA and protein expression was downregulated, and glucose uptake capacity was reduced in PCOS GCs and SVOG cells. Melatonin significantly upregulated IRS-1 and GLUT4 expression, downregulated p-IRS-1 (Ser307), and improved glucose uptake in PCOS patients' GCs and SVOG cells. PA decreased PI3K and Akt phosphorylation, whereas melatonin increased p-PI3K and p-Akt levels. Melatonin can reduce IR in GCs and PA-induced SVOG cells via the PI3K/Akt signaling pathway, providing more evidence for treating polycystic ovary syndrome.
Collapse
Affiliation(s)
- Rui Guo
- Reproductive Medicine Center, Shandong Maternal and Child Health Care Center, NO. 238, East Jingshi Road, Jinan 250014, Shandong, China
| | - Hong Zheng
- Department of Reproductive Medicine, Dezhou People's Hospital, NO. 1166, Dongfanghong West Road, Dezhou 253014, Shandong, China
| | - Qiuying Li
- Department of Radiology, Zhangqiu People's Hospital, NO. 1920, Huiquan Road, Jinan 250200, Shandong, China
| | - Xun Qiu
- Department of Radiology, Zhangqiu People's Hospital, NO. 1920, Huiquan Road, Jinan 250200, Shandong, China
| | - Jian Zhang
- Department of Radiology, Zhangqiu People's Hospital, NO. 1920, Huiquan Road, Jinan 250200, Shandong, China
| | - Zhaofang Cheng
- Department of Radiology, Zhangqiu People's Hospital, NO. 1920, Huiquan Road, Jinan 250200, Shandong, China; Department of Obstetrics and Gynecology, Zhangqiu People's Hospital, NO. 1920, Huiquan Road, Jinan 250200, Shandong, China.
| |
Collapse
|
10
|
Seymen CM, Yar Sağlam AS, Elmazoğlu Z, Arık GN, Take Kaplanoğlu G. Involvement of endometrial IGF-1R/IGF-1/Bcl-2 pathways in experimental polycystic ovary syndrome: Identification of the regulatory effect of melatonin. Tissue Cell 2021; 73:101585. [PMID: 34375824 DOI: 10.1016/j.tice.2021.101585] [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: 09/25/2020] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 11/25/2022]
Abstract
The involvement of endometrial IGF-1R/IGF-1/Bcl-2 pathways and the potential regulatory effects of exogenously administrated melatonin on this expression is investigated in the experimental PCOS model in the present study. Thirty-two 6-8 week old Sprague Dawley rats were divided into four groups: the Sham Control Group (1% CMC/day by oral gavage [o.g.]); the Melatonin Group (2 mg/kg/day melatonin by subcutaneous administration [s.c.]); the Experimental PCOS Group (1 mg/kg/day Letrozole by o.g.); and the Experimental PCOS + Melatonin Group (1 mg/kg/day Letrozole by o.g. and 2 mg/kg/day melatonin by s.c. administration). Vaginal smear samples were taken from the 14th day to the end of the experiment for colpocytological measurements. At the end of the 21 day experimental period, uterine tissues were taken; Hematoxylin-Eosin histochemical, IGF-1R/IGF-1/Bcl-2, PCNA immuno-histochemical stainings and western blot analyses were performed for related antibodies. All of the data was supported statistically. The epithelium of endometrium lost its single-layer structure in some parts, separation was observed between the epithelium and the basal membrane junction, intracellular edema was found in the uterine glands by the polycystic ovary-induction. Also this induction increased the expression of IGF-1R/IGF-1, Bcl-2, and PCNA proteins. Morphological degenerations returned to its normal appearance generally by the melatonin administrations and melatonin also regulated the increased expression of endometrial IGF-1R/IGF-1/Bcl-2 and PCNA pathways. It is concluded that additional studies are needed, using melatonin as a supporting agent may be appropriate in cases of PCOS.
Collapse
Affiliation(s)
- Cemile Merve Seymen
- Gazi University Faculty of Medicine, Department of Histology and Embryology, Ankara, Turkey.
| | - Atiye Seda Yar Sağlam
- Gazi University Faculty of Medicine, Department of Medical Biology and Genetics, Ankara, Turkey
| | - Zübeyir Elmazoğlu
- Gazi University Faculty of Medicine, Department of Medical Pharmacology, Ankara, Turkey
| | - Gökçe Nur Arık
- Gazi University Faculty of Medicine, Department of Histology and Embryology, Ankara, Turkey
| | - Gülnur Take Kaplanoğlu
- Gazi University Faculty of Medicine, Department of Histology and Embryology, Ankara, Turkey
| |
Collapse
|
11
|
Alizadeh M, Karandish M, Asghari Jafarabadi M, Heidari L, Nikbakht R, Babaahmadi Rezaei H, Mousavi R. Metabolic and hormonal effects of melatonin and/or magnesium supplementation in women with polycystic ovary syndrome: a randomized, double-blind, placebo-controlled trial. Nutr Metab (Lond) 2021; 18:57. [PMID: 34092248 PMCID: PMC8183043 DOI: 10.1186/s12986-021-00586-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/28/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders among women of reproductive age. This study was designed to investigate the effects of melatonin and/or magnesium supplementation on metabolic profile and levels of sex hormones in PCOS women. METHODS In an 8-week randomized double-blind placebo-controlled trial, 84 subjects with PCOS aged 18-40 years were randomly assigned based on the random block procedure to take magnesium, melatonin, magnesium plus melatonin, and placebo. Fasting blood samples were obtained at the beginning and end of the study. RESULTS After intervention, the mean Pittsburg Sleep Quality Index score decreased significantly in both co-supplementation and melatonin groups (P < 0.001). Magnesium supplementation in combination with melatonin resulted in a significant greater decrease in testosterone concentrations compared with the placebo (P < 0.05). Co-supplementation of magnesium-melatonin had significantly reduced serum insulin levels (geometric means difference: - 1.11 (mIU/mL) (percent change: - 15.99)), homeostasis model of assessment-insulin resistance (HOMA-IR) (- 0.28 (- 18.66)), serum cholesterol (mean difference: - 16.08 (mg/dl) [95% CI - 24.24, - 7.92]), low-density lipoprotein cholesterol (LDL-C) - 18.96 (mg/dl) [- 28.73, - 9.20]) and testosterone levels (- 0.09 (ng/ml) (- 25.00)), as compared to the baseline values (P < 0.05). An increase in serum high-density lipoprotein cholesterol (HDL-C) levels was also observed following the administration of the melatonin alone (2.76 (mg/dl) [0.57, 4.95]) or in combination with magnesium (2.19 (mg/dl) [0.61, 3.77]) (P < 0.05). CONCLUSIONS Co-supplementation with magnesium and melatonin had beneficial effects on sleep quality and total testosterone. Additionally, melatonin supplementation alone was found to be associated with a significant reduction in PSQI score. Moreover, combined melatonin and magnesium supplementation was more effective in improving serum levels of cholesterol, LDL-C, HDL-C and insulin, and HOMA-IR. TRIAL REGISTRATION Iranian Registry of Clinical Trial. http://www.irct.ir : IRCT20191130045556N1, January 2020.
Collapse
Affiliation(s)
- Mohammad Alizadeh
- Department of Clinical Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Science, Tabriz, Iran
| | - Majid Karandish
- Nutrition and Metabolic Disease Research Center, Clinical Science Research Institute, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
| | - Mohammad Asghari Jafarabadi
- Department of Statistics and Epidemiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.,Road Traffic Injury Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | | | - Roshan Nikbakht
- Fertility Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
| | - Hossein Babaahmadi Rezaei
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
| | - Reihaneh Mousavi
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. .,Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
12
|
Mojaverrostami S, Asghari N, Khamisabadi M, Heidari Khoei H. The role of melatonin in polycystic ovary syndrome: A review. Int J Reprod Biomed 2019; 17:865-882. [PMID: 31970309 PMCID: PMC6943797 DOI: 10.18502/ijrm.v17i12.5789] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/16/2019] [Accepted: 07/20/2019] [Indexed: 12/26/2022] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is a widespread endocrine disorder, affecting approximately 20% of women within reproductive age. It is associated with hyperandrogenism, obesity, menstrual irregularity, and anovulatory infertility. Melatonin is the main pineal gland hormone involved in the regulation of the circadian rhythm. In recent years, it has been observed that a reduction in melatonin levels of follicular fluid exists in PCOS patients. Melatonin receptors in the ovary and intra-follicular fluid adjust sex steroid secretion at different phases of ovarian follicular maturation. Moreover, melatonin is a strong antioxidant and an effective free radical scavenger, which protects ovarian follicles during follicular maturation. Objective In this paper, we conducted a literature review and the summary of the current research on the role of melatonin in PCOS. Materials and Methods Electronic databases including PubMed/MEDLINE, Web of Science, Scopus, and Reaxys were searched from their inception to October 2018 using the keywords “Melatonin” AND “Polycystic ovary syndrome” OR “PCOS.” Results Based on the data included in our review, it was found that the administration of melatonin can improve the oocyte and embryo quality in PCOS patients. It may also have beneficial effects in correcting the hormonal alterations in PCOS patients. Conclusion Since metabolic dysfunction is the major finding contributing to the initiation of PCOS, melatonin can hinder this process via its improving effects on metabolic functions.
Collapse
Affiliation(s)
- Sina Mojaverrostami
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Narjes Asghari
- Department of Molecular Medicine, Faculty of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | | | - Heidar Heidari Khoei
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Melatonin Reduces Androgen Production and Upregulates Heme Oxygenase-1 Expression in Granulosa Cells from PCOS Patients with Hypoestrogenia and Hyperandrogenia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8218650. [PMID: 31772710 PMCID: PMC6854986 DOI: 10.1155/2019/8218650] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/07/2019] [Accepted: 08/24/2019] [Indexed: 12/20/2022]
Abstract
Background/Aims Polycystic ovary syndrome (PCOS) is an endocrine disorder characterized by abnormal hormone levels in peripheral blood and poor-quality oocytes. PCOS is a pathophysiological syndrome caused by chronic inflammation and oxidative stress. The aim of this study was to investigate the mechanism of melatonin regulation on androgen production and antioxidative damage in granulosa cells from PCOS patients with hypoestrogenia and hyperandrogenia. Methods Cumulus-oocyte complexes were collected from PCOS patients who had low levels of estrogen in follicular fluids. Results Melatonin triggered upregulation of cytochrome P450 family 19 subfamily A member 1 (CYP19A1) expression via the extracellular signal-regulated kinase pathway in luteinized granulosa cells. As a result, conversion of androgen to 17β-estradiol was accelerated. We also found that melatonin significantly reduced the levels of inducible nitric oxide (NO) synthetase and NO in luteinized granulosa cells. Levels of transcripts encoding NF-E2-related factor-2 and its downstream target heme oxygenase-1 were also increased, leading to anti-inflammatory and antioxidant effects. We also found that melatonin could improve oocyte development potential. Conclusion Our preliminary results showed that melatonin had a positive impact on oocyte quality in PCOS patients with hypoestrogenia and hyperandrogenia.
Collapse
|
14
|
Zhuang Z, Pan X, Zhao K, Gao W, Liu J, Deng T, Qin W. The Effect of Interleukin-6 (IL-6), Interleukin-11 (IL-11), Signal Transducer and Activator of Transcription 3 (STAT3), and AKT Signaling on Adipocyte Proliferation in a Rat Model of Polycystic Ovary Syndrome. Med Sci Monit 2019; 25:7218-7227. [PMID: 31554782 PMCID: PMC6777385 DOI: 10.12659/msm.916385] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is associated with low-grade inflammation, adipocyte hypertrophy, hyperglycemia, increased serum testosterone levels, and reduced lipolysis. This study aimed to investigate the role of interleukin-6 (IL-6) and IL-11 in the pathophysiology of adipocyte hypertrophy in a rat model of PCOS. Material/Methods The rat model of PCOS was developed using a subcutaneous injection of dehydroepiandrosterone (DHEA). Histology of the rat ovaries was used to confirm the development of PCOS. Serum levels of testosterone and glucose were measured. Immunohistochemistry, immunofluorescence, quantitative real-time polymerase chain reaction (qRT-PCR), and Western blot were performed to measure IL-6 and IL-11 in the rat model of PCOS. Cell proliferation was measured using the cell counting kit-8 (CCK-8) assay. Results Serum levels of testosterone and glucose and the expression of IL-6 and IL-11 were significantly increased in the rat model of PCOS via the activation of AKT/STAT3 signaling. Following IL-6 and IL-11 stimulation of mesenchymal adipocytes isolated from adipose tissue, IL-6 and IL-11 induced cell proliferation through the STAT3/AKT signaling pathway. Conclusions In a rat model of PCOS, increased expression of IL-6 and IL-11 was associated with the AKT/STAT3 pathway. Increased levels of IL-6 and IL-11 stimulated adipocytes from adipose tissue of the rat model, which promoted cell proliferation by activating AKT/STAT3 signaling.
Collapse
Affiliation(s)
- Zhaohui Zhuang
- Department of Reproduction, Suqian Maternity Hospital, Suqian, Jiangsu, China (mainland)
| | - Xiaohong Pan
- Department of Treating Potential Diseases, Xuzhou City Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Xuzhou, Jiangsu, China (mainland)
| | - Kai Zhao
- Department of Gynecology, Xuzhou City Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Xuzhou, Jiangsu, China (mainland)
| | - Wei Gao
- Department of Gynecology and Obstetrics, Suqian Maternity Hospital, Suqian, Jiangsu, China (mainland)
| | - Juan Liu
- Department of Gynecology and Obstetrics, Suqian Maternity Hospital, Suqian, Jiangsu, China (mainland)
| | - Tianqi Deng
- Department of Gynecology, Xuzhou City Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Xuzhou, Jiangsu, China (mainland)
| | - Wenmin Qin
- Department of Gynecology, Xuzhou City Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Xuzhou, Jiangsu, China (mainland)
| |
Collapse
|
15
|
Simon SL, McWhirter L, Diniz Behn C, Bubar KM, Kaar JL, Pyle L, Rahat H, Garcia-Reyes Y, Carreau AM, Wright KP, Nadeau KJ, Cree-Green M. Morning Circadian Misalignment Is Associated With Insulin Resistance in Girls With Obesity and Polycystic Ovarian Syndrome. J Clin Endocrinol Metab 2019; 104:3525-3534. [PMID: 30888398 PMCID: PMC6610211 DOI: 10.1210/jc.2018-02385] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 03/13/2019] [Indexed: 02/08/2023]
Abstract
CONTEXT To our knowledge, circadian rhythms have not been examined in girls with polycystic ovarian syndrome (PCOS), despite the typical delayed circadian timing of adolescence, which is an emerging link between circadian health and insulin sensitivity (SI), and decreased SI in PCOS. OBJECTIVE To examine differences in the circadian melatonin rhythm between obese adolescent girls with PCOS and control subjects, and evaluate relationships between circadian variables and SI. DESIGN Cross-sectional study. PARTICIPANTS Obese adolescent girls with PCOS (n = 59) or without PCOS (n = 33). OUTCOME MEASURES Estimated sleep duration and timing from home actigraphy monitoring, in-laboratory hourly sampled dim-light, salivary-melatonin and fasting hormone analysis. RESULTS All participants obtained insufficient sleep. Girls with PCOS had later clock-hour of melatonin offset, later melatonin offset relative to sleep timing, and longer duration of melatonin secretion than control subjects. A later melatonin offset after wake time (i.e., morning wakefulness occurring during the biological night) was associated with higher serum free testosterone levels and worse SI regardless of group. Analyses remained significant after controlling for daytime sleepiness and sleep-disordered breathing. CONCLUSION Circadian misalignment in girls with PCOS is characterized by later melatonin offset relative to clock time and sleep timing. Morning circadian misalignment was associated with metabolic dysregulation in girls with PCOS and obesity. Clinical care of girls with PCOS and obesity would benefit from assessment of sleep and circadian health. Additional research is needed to understand mechanisms underlying the relationship between morning circadian misalignment and SI in this population.
Collapse
Affiliation(s)
- Stacey L Simon
- Division of Pulmonary Medicine, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado
| | - Laura McWhirter
- Division of Pulmonary Medicine, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado
| | - Cecilia Diniz Behn
- Division of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, Colorado
| | - Kate M Bubar
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, Colorado
| | - Jill L Kaar
- Division of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado
| | - Laura Pyle
- Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado
| | - Haseeb Rahat
- Division of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado
| | - Yesenia Garcia-Reyes
- Division of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado
| | - Anne-Marie Carreau
- Division of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado
| | - Kenneth P Wright
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kristen J Nadeau
- Division of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado
- Center for Women’s Health Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Melanie Cree-Green
- Division of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado
- Center for Women’s Health Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
16
|
Adeyanju OA, Falodun TO, Fabunmi OA, Olatunji LA, Soladoye AO. Very low dose spironolactone protects experimentally-induced polycystic ovarian syndrome from insulin-resistant metabolic disturbances by suppressing elevated circulating testosterone. Chem Biol Interact 2019; 310:108742. [PMID: 31295448 DOI: 10.1016/j.cbi.2019.108742] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/01/2019] [Accepted: 07/08/2019] [Indexed: 11/30/2022]
Abstract
Polycystic ovarian syndrome (PCOS) is the most common endocrinological disorder in women of reproductive age and hyperandrogenism is a prominent feature of PCOS resulting in infertility and increased risk of developing metabolic disorders including insulin resistance (IR), abdominal adiposity, glucose intolerance and cardiovascular diseases. Spironolactone (SPL), a non-selective mineralocorticoid receptor (MR) antagonist, has been in wide clinical use for several decades. In this study, we investigated the effects of SPL on IR and metabolic disturbances in letrozole-induced PCOS rats. Eighteen adults female Wistar rats were randomly divided into 3 groups and treated with vehicle, letrozole (LET; 1 mg/kg) and LET + SPL (SPL; 0.25 mg/kg), p.o. once daily for 21 consecutive days. Results showed that LET treatment induced PCOS characterised by elevated plasma testosterone and luteinizing hormone (LH) accompanied with increased body weight and visceral adiposity, IR, glucose intolerance, dyslipidemia and altered histomorphological ovaries. Treatment with SPL however attenuated the elevated testosterone in LET-induced PCOS model accompanied with a reversal in all the observed alterations. Taken together, analysis of the physical, biochemical and histological evidences shows that the protective effect of this very low dose spironolactone may be through its anti-androgenic mechanism.
Collapse
Affiliation(s)
- Oluwaseun A Adeyanju
- Cardiometabolic Research Unit, Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria; HOPE Cardiometabolic Research Team & Department of Physiology, University of Ilorin, Ilorin, Nigeria.
| | - Timothy O Falodun
- Cardiometabolic Research Unit, Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | | | - Lawrence A Olatunji
- HOPE Cardiometabolic Research Team & Department of Physiology, University of Ilorin, Ilorin, Nigeria
| | - Ayodele O Soladoye
- Cardiometabolic Research Unit, Department of Physiology, College of Health and Medical Sciences, Bowen University, Iwo, Nigeria
| |
Collapse
|
17
|
Jamilian M, Foroozanfard F, Mirhosseini N, Kavossian E, Aghadavod E, Bahmani F, Ostadmohammadi V, Kia M, Eftekhar T, Ayati E, Mahdavinia M, Asemi Z. Effects of Melatonin Supplementation on Hormonal, Inflammatory, Genetic, and Oxidative Stress Parameters in Women With Polycystic Ovary Syndrome. Front Endocrinol (Lausanne) 2019; 10:273. [PMID: 31139144 PMCID: PMC6527800 DOI: 10.3389/fendo.2019.00273] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/15/2019] [Indexed: 12/21/2022] Open
Abstract
Purpose: The aim of the current study was to evaluate the effect of melatonin administration on clinical, hormonal, inflammatory, and genetic parameters in women with polycystic ovarian syndrome (PCOS). Methods: The present randomized, double-blinded, placebo-controlled clinical trial was conducted among 56 patients with PCOS, aged 18-40 years old. Subjects were randomly allocated to take either 5 mg melatonin supplements (n = 28) or placebo (n = 28) twice a day for 12 weeks. Results: Melatonin administration significantly reduced hirsutism (β -0.47; 95% CI, -0.86, -0.09; P = 0.01), serum total testosterone (β -0.11 ng/mL; 95% CI, -0.21, -0.02; P = 0.01), high-sensitivity C-reactive protein (hs-CRP) (β -0.61 mg/L; 95% CI, -0.95, -0.26; P = 0.001), and plasma malondialdehyde (MDA) levels (β -0.25 μmol/L; 95% CI, -0.38, -0.11; P < 0.001), and significantly increased plasma total antioxidant capacity (TAC) levels (β 106.07 mmol/L; 95% CI, 62.87, 149.28; P < 0.001) and total glutathione (GSH) (β 81.05 μmol/L; 95% CI, 36.08, 126.03; P = 0.001) compared with the placebo. Moreover, melatonin supplementation downregulated gene expression of interleukin-1 (IL-1) (P = 0.03) and tumor necrosis factor alpha (TNF-α) (P = 0.01) compared with the placebo. Conclusions: Overall, melatonin administration for 12 weeks to women with PCOS significantly reduced hirsutism, total testosterone, hs-CRP, and MDA, while increasing TAC and GSH levels. In addition, melatonin administration reduced gene expression of IL-1 and TNF-α. Clinical Trial Registration: www.irct.ir, identifier IRCT2017082733941N9, Available online at: https://www.irct.ir/trial/26051.
Collapse
Affiliation(s)
- Mehri Jamilian
- Traditional and Complementary Medicine Research Center, Arak University of Medical Sciences, Arak, Iran
| | - Fatemeh Foroozanfard
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Elham Kavossian
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Esmat Aghadavod
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Fereshteh Bahmani
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Vahidreza Ostadmohammadi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Mersedeh Kia
- Department of Midwifery, Gorgan Branch, Islamic Azad University, Gorgan, Iran
| | - Tahereh Eftekhar
- Reproductive Health Research Center, Tehran University of Medical Science, Tehran, Iran
- *Correspondence: Tahereh Eftekhar
| | - Elnaz Ayati
- Reproductive Health Research Center, Tehran University of Medical Science, Tehran, Iran
- Department of Gynecology and Obstetrics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Mahdavinia
- Department of Dermatology, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
- Zatollah Asemi
| |
Collapse
|
18
|
Ragy MM, Abdel-Hamid HA, Toni NDM. Pathophysiological changes in experimental polycystic ovary syndrome in female albino rats: Using either hemin or L-arginine. J Cell Physiol 2018; 234:8426-8435. [PMID: 30443939 DOI: 10.1002/jcp.27757] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 10/29/2018] [Indexed: 12/25/2022]
Abstract
Polycystic ovary syndrome (PCOS), one of the important endocrine disorders affecting females in the reproductive age, is caused mainly by an abnormal oxidation status that subsequently causes inflammatory conditions. Thus, this study aims to examine the possible individual prophylactic effects of gasotransmitters, hemin, or L-arginine in letrozole-induced PCOS. Fifty adult female albino rats were used and separated into a control group, which received the vehicle; a letrozole-induced PCOS group (L), which received letrozole orally at a dose level of 1 mg/kg for 21 days; a letrozole+hemin (L+H) group, which received letrozole plus hemin at a dose level of 25 mg/kg injected IP twice per week for 21 days; and a letrozole+L-arginine (L+A) group, which received letrozole plus L-arginine at a dose level of 200 mg/kg orally for 21 days. During PCO induction, the body weight and Lee index were measured. Serum glucose, insulin, lipid profile, gonadotrophic hormones, testosterone, estrogen, and tumor necrosis factor alpha were assayed, while ovarian tissues were analyzed to measure the oxidative state and histopathological changes. Our results proved that either hemin or L-arginine administration could improve the oxidative state, the inflammatory reaction, the hormonal imbalance, and the metabolic disturbances in PCO rats, which was confirmed by a histopathological examination of the rats' ovaries. In conclusion, either hemin or L-arginine had protective effects against PCOS with better pathophysiological changes with hemin.
Collapse
Affiliation(s)
- Merhan M Ragy
- Medical Physiology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - Heba A Abdel-Hamid
- Medical Physiology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - Nisreen D M Toni
- Medical Physiology Department, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
19
|
de Souza BR, de Almeida Chuffa LG, Simão VA, Camargo ICC. Histopathological changes in androgenized ovaries are recovered by melatonin treatment. Int J Exp Pathol 2018; 99:158-171. [PMID: 30256483 PMCID: PMC6157297 DOI: 10.1111/iep.12283] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/22/2018] [Accepted: 07/07/2018] [Indexed: 12/21/2022] Open
Abstract
Nandrolone decanoate (ND) is a synthetic steroid, which promotes adverse effects on the ovarian tissue, and melatonin (MLT) exhibits a number of beneficial properties in the reproductive system. This study evaluated the general features of the ovarian tissue and the immunoexpression of sex steroid receptors in ND-treated rats that were submitted to short-term melatonin treatment. Adult rats received mineral oil (control group) and ND at doses of 7.5 mg/kg for 15 days (ND-treated group). The treatment with MLT (10mg/kg for 7 days) was given alone, before or in combination with ND. All ND-treated animals showed persistent dioestrus. In the androgenized groups that received MLT, ovarian morphology and size, and the number/area of corpora lutea were recovered. The number of healthy and atretic follicles was recovered when MLT was administered prior to ND; this was similar to the ovaries of control and MLT groups. There was a decrease in estrogen receptors immunostaining in the follicles of androgenized rats that were treated with MLT, and pretreatment with MLT reduced the expression of androgen receptor in atretic follicles and corpora lutea, when compared with ND-treated group. We conclude that MLT treatment recovered the histopathological aspects of the androgenized ovaries, and MLT pretreatment was the most effective.
Collapse
Affiliation(s)
- Bianca R. de Souza
- Programa de Pós‐Graduação em BiociênciasDepartamento de BiotecnologiaUniversidade Estadual Paulista – UNESPAssisSão PauloBrazil
| | | | - Vinícius Augusto Simão
- Programa de Pós‐Graduação em BiociênciasDepartamento de BiotecnologiaUniversidade Estadual Paulista – UNESPAssisSão PauloBrazil
| | - Isabel C. C. Camargo
- Departamento de BiotecnologiaFaculdade de Ciências e LetrasUniversidade Estadual Paulista – UNESPAssisSão PauloBrazil
| |
Collapse
|
20
|
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
|
21
|
Walters KA, Bertoldo MJ, Handelsman DJ. Evidence from animal models on the pathogenesis of PCOS. Best Pract Res Clin Endocrinol Metab 2018; 32:271-281. [PMID: 29779581 DOI: 10.1016/j.beem.2018.03.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Polycystic ovarian syndrome (PCOS) is the most common endocrine condition in women, and is characterized by reproductive, endocrine and metabolic features. However, there is no simple unequivocal diagnostic test for PCOS, its etiology remains unknown and there is no cure. Hence, the management of PCOS is suboptimal as it relies on the ad hoc empirical management of its symptoms only. Decisive studies are required to unravel the origins of PCOS, but due to ethical and logistical reasons these are not possible in humans. Experimental animal models for PCOS have been established which have enhanced our understanding of the mechanisms underlying PCOS and propose novel mechanism-based therapies to treat the condition. This review examines the findings from various animal models to reveal the current knowledge of the mechanisms underpinning the development of PCOS, and also provides insights into the implications from these studies for improved clinical management of this disorder.
Collapse
Affiliation(s)
- K A Walters
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, NSW 2052, Australia.
| | - M J Bertoldo
- Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales, Sydney, NSW 2052, Australia.
| | - D J Handelsman
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales 2139, Australia.
| |
Collapse
|
22
|
Wang Y, He J, Yang J. Eicosapentaenoic Acid Improves Polycystic Ovary Syndrome in Rats via Sterol Regulatory Element-Binding Protein 1 (SREBP-1)/Toll-Like Receptor 4 (TLR4) Pathway. Med Sci Monit 2018; 24:2091-2097. [PMID: 29627845 PMCID: PMC5907624 DOI: 10.12659/msm.909098] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND This study aimed to investigate the protective effect of eicosapentaenoic acid (EPA) on rats with polycystic ovary syndrome (PCOS). MATERIAL AND METHODS Rats with PCOS were intraperitoneally injected with different doses of EPA. Levels of follicle stimulating hormone (FSH), luteinizing hormone (LH), and testosterone (T) were measured using corresponding kits. HE staining was used to observe lesions in ovarian tissue. Levels of inflammatory factors in ovarian tissue of rats were detected by ELISA. RT-PCR was to detect the expression of SREBP1 mRNA and Western blot was used to detect the expression of SREBP1 and TLR4 protein. RESULTS The levels of LH and T were significantly higher and FDH was significantly lower in the Model group compared with the Control group. EPA treatment increased the number of follicular cell layers and promoted maturation of oocytes. Levels of IL-1β, TNF-α, and IL-18 were significantly reduced after EPA treatment. Content of IL-10 was significantly increased after EPA treatment. Expression levels of SREBP1 and TLR4 were significantly deceased after EPA treatment. CONCLUSIONS EPA can improve PCOS through the SREBP1/TLR4 pathway.
Collapse
Affiliation(s)
- Yanting Wang
- Reproductive Medicine Center, People's Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Jinying He
- Reproductive Medicine Center, People's Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Jing Yang
- Reproductive Medicine Center, People's Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| |
Collapse
|
23
|
Drąg-Kozak E, Socha M, Gosiewski G, Łuszczek-Trojnar E, Chyb J, Popek W. Protective effect of melatonin on cadmium-induced changes in some maturation and reproductive parameters of female Prussian carp (Carassius gibelio B.). ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:9915-9927. [PMID: 29374378 PMCID: PMC5891563 DOI: 10.1007/s11356-018-1308-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 01/15/2018] [Indexed: 05/25/2023]
Abstract
The aim of this study was to determine whether melatonin (Mel), which is a known antioxidant and free radical scavenger, could perform the role of a preventive agent against the toxic effects of cadmium (Cd2+) on mortality, fish growth, gonadosomatic index (GSI), luteinizing hormone (LH) secretion, the response to hormonal stimulation of spawning, and also tissue accumulation of Cd in Prussian carp females. These females received melatonin implants and were exposed to 0.4 or 4.0 mg/L of Cd (as CdCl2·2.5H2O) over either a 5- or 3-month period, followed by further 2 months of purification in clear water. Negative changes caused by exposure to cadmium in the water were as follows: higher fish mortality, lower body weight, increased accumulation of cadmium in the brain and ovary, lowered GSI, impaired spontaneous LH secretion during exposure, and impaired LH secretion during stimulation of spawning. All of these effects were observed in the group of fish exposed to 0.4 and/or 4.0 mg Cd/L but did not occur or were less pronounced in the groups exposed to cadmium in the presence of melatonin released from the implants. During depuration, in the group of fish which had been exposed to the highest Cd concentration, we observed a significant improvement in fish survival rate, body growth, inhibition of further cadmium accumulation in tissues, and gradual return of spontaneous LH secretion as well as normalization of the GSI value to the control group levels. In conclusion, these findings indicate that melatonin can be a preventive agent for some toxic effects on fish reproduction induced by environmental cadmium contamination.
Collapse
Affiliation(s)
- Ewa Drąg-Kozak
- Department of Ichthyobiology and Fisheries, University of Agriculture in Krakow, ul. Spiczakowa 6, 30-199, Krakow-Mydlniki, Poland.
| | - Magdalena Socha
- Department of Ichthyobiology and Fisheries, University of Agriculture in Krakow, ul. Spiczakowa 6, 30-199, Krakow-Mydlniki, Poland
| | - Grzegorz Gosiewski
- Department of Ichthyobiology and Fisheries, University of Agriculture in Krakow, ul. Spiczakowa 6, 30-199, Krakow-Mydlniki, Poland
| | - Ewa Łuszczek-Trojnar
- Department of Ichthyobiology and Fisheries, University of Agriculture in Krakow, ul. Spiczakowa 6, 30-199, Krakow-Mydlniki, Poland
| | - Jarosław Chyb
- Department of Ichthyobiology and Fisheries, University of Agriculture in Krakow, ul. Spiczakowa 6, 30-199, Krakow-Mydlniki, Poland
| | - Włodzimierz Popek
- Department of Ichthyobiology and Fisheries, University of Agriculture in Krakow, ul. Spiczakowa 6, 30-199, Krakow-Mydlniki, Poland
| |
Collapse
|
24
|
Yang HL, Zhou WJ, Gu CJ, Meng YH, Shao J, Li DJ, Li MQ. Pleiotropic roles of melatonin in endometriosis, recurrent spontaneous abortion, and polycystic ovary syndrome. Am J Reprod Immunol 2018; 80:e12839. [PMID: 29493042 DOI: 10.1111/aji.12839] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 02/06/2018] [Indexed: 12/22/2022] Open
Abstract
Melatonin is a neurohormone synthesized from the aromatic amino acid tryptophan mainly by the pineal gland of mammals. Melatonin acts as a broad-spectrum antioxidant, powerful free radical scavenger, anti-inflammatory agent, anticarcinogenic factor, sleep inducer and regulator of the circadian rhythm, and potential immunoregulator. Melatonin and reproductive system are interrelated under both physiological and pathological conditions. Oxidative stress, inflammation, and immune dysregulation are associated with the pathogenesis of the female reproductive system which causes endometriosis (EMS), recurrent spontaneous abortion (RSA), and polycystic ovary syndrome (PCOS). Accumulating studies have indicated that melatonin plays pleiotropic and essential roles in these obstetrical and gynecological disorders and would be a candidate therapeutic drug to regulate inflammation and immune function and protect special cells or organs. Here, we systematically review the pleiotropic roles of melatonin in EMS, RSA, and PCOS to explore its pathological implications and treatment potential.
Collapse
Affiliation(s)
- Hui-Li Yang
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Wen-Jie Zhou
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Chun-Jie Gu
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Yu-Han Meng
- Reproductive Medical Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jun Shao
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Da-Jin Li
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| |
Collapse
|
25
|
Spinedi E, Cardinali DP. The Polycystic Ovary Syndrome and the Metabolic Syndrome: A Possible Chronobiotic-Cytoprotective Adjuvant Therapy. Int J Endocrinol 2018; 2018:1349868. [PMID: 30147722 PMCID: PMC6083563 DOI: 10.1155/2018/1349868] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/28/2018] [Indexed: 12/12/2022] Open
Abstract
Polycystic ovary syndrome is a highly frequent reproductive-endocrine disorder affecting up to 8-10% of women worldwide at reproductive age. Although its etiology is not fully understood, evidence suggests that insulin resistance, with or without compensatory hyperinsulinemia, and hyperandrogenism are very common features of the polycystic ovary syndrome phenotype. Dysfunctional white adipose tissue has been identified as a major contributing factor for insulin resistance in polycystic ovary syndrome. Environmental (e.g., chronodisruption) and genetic/epigenetic factors may also play relevant roles in syndrome development. Overweight and/or obesity are very common in women with polycystic ovary syndrome, thus suggesting that some polycystic ovary syndrome and metabolic syndrome female phenotypes share common characteristics. Sleep disturbances have been reported to double in women with PCOS and obstructive sleep apnea is a common feature in polycystic ovary syndrome patients. Maturation of the luteinizing hormone-releasing hormone secretion pattern in girls in puberty is closely related to changes in the sleep-wake cycle and could have relevance in the pathogenesis of polycystic ovary syndrome. This review article focuses on two main issues in the polycystic ovary syndrome-metabolic syndrome phenotype development: (a) the impact of androgen excess on white adipose tissue function and (b) the possible efficacy of adjuvant melatonin therapy to improve the chronobiologic profile in polycystic ovary syndrome-metabolic syndrome individuals. Genetic variants in melatonin receptor have been linked to increased risk of developing polycystic ovary syndrome, to impairments in insulin secretion, and to increased fasting glucose levels. Melatonin therapy may protect against several metabolic syndrome comorbidities in polycystic ovary syndrome and could be applied from the initial phases of patients' treatment.
Collapse
Affiliation(s)
- Eduardo Spinedi
- Centre for Experimental and Applied Endocrinology (CENEXA, UNLP-CONICET-FCM), CEAS-CICPBA, La Plata Medical School, La Plata, Argentina
| | - Daniel P. Cardinali
- BIOMED-UCA-CONICET and Department of Teaching and Research, Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| |
Collapse
|
26
|
Cardinali DP, Vigo DE. Melatonin, mitochondria, and the metabolic syndrome. Cell Mol Life Sci 2017; 74:3941-3954. [PMID: 28819865 PMCID: PMC11107716 DOI: 10.1007/s00018-017-2611-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 08/03/2017] [Indexed: 12/12/2022]
Abstract
A number of risk factors for cardiovascular disease including hyperinsulinemia, glucose intolerance, dyslipidemia, obesity, and elevated blood pressure are collectively known as metabolic syndrome (MS). Since mitochondrial activity is modulated by the availability of energy in cells, the disruption of key regulators of metabolism in MS not only affects the activity of mitochondria but also their dynamics and turnover. Therefore, a link of MS with mitochondrial dysfunction has been suspected since long. As a chronobiotic/cytoprotective agent, melatonin has a special place in prevention and treatment of MS. Melatonin levels are reduced in diseases associated with insulin resistance like MS. Melatonin improves sleep efficiency and has antioxidant and anti-inflammatory properties, partly for its role as a metabolic regulator and mitochondrial protector. We discuss in the present review the several cytoprotective melatonin actions that attenuate inflammatory responses in MS. The clinical data that support the potential therapeutical value of melatonin in human MS are reviewed.
Collapse
Affiliation(s)
- Daniel P Cardinali
- BIOMED-UCA-CONICET and Department of Teaching and Research, Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Av. Alicia Moreau de Justo 1500, 4o piso, 1107, Buenos Aires, Argentina.
| | - Daniel E Vigo
- BIOMED-UCA-CONICET and Department of Teaching and Research, Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Av. Alicia Moreau de Justo 1500, 4o piso, 1107, Buenos Aires, Argentina
| |
Collapse
|
27
|
Yaghmaei P, Dehestani B, Ghorbani S, Abbasi F, Ebrahim-Habibi A. Indole-based derivatives effect on rats with polycystic ovary syndrome. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-017-0317-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
28
|
Forrestel AC, Miedlich SU, Yurcheshen M, Wittlin SD, Sellix MT. Chronomedicine and type 2 diabetes: shining some light on melatonin. Diabetologia 2017; 60:808-822. [PMID: 27981356 DOI: 10.1007/s00125-016-4175-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/18/2016] [Indexed: 12/19/2022]
Abstract
In mammals, the circadian timing system drives rhythms of physiology and behaviour, including the daily rhythms of feeding and activity. The timing system coordinates temporal variation in the biochemical landscape with changes in nutrient intake in order to optimise energy balance and maintain metabolic homeostasis. Circadian disruption (e.g. as a result of shift work or jet lag) can disturb this continuity and increase the risk of cardiometabolic disease. Obesity and metabolic disease can also disturb the timing and amplitude of the clock in multiple organ systems, further exacerbating disease progression. As our understanding of the synergy between the timing system and metabolism has grown, an interest has emerged in the development of novel clock-targeting pharmaceuticals or nutraceuticals for the treatment of metabolic dysfunction. Recently, the pineal hormone melatonin has received some attention as a potential chronotherapeutic drug for metabolic disease. Melatonin is well known for its sleep-promoting effects and putative activity as a chronobiotic drug, stimulating coordination of biochemical oscillations through targeting the internal timing system. Melatonin affects the insulin secretory activity of the pancreatic beta cell, hepatic glucose metabolism and insulin sensitivity. Individuals with type 2 diabetes mellitus have lower night-time serum melatonin levels and increased risk of comorbid sleep disturbances compared with healthy individuals. Further, reduced melatonin levels, and mutations and/or genetic polymorphisms of the melatonin receptors are associated with an increased risk of developing type 2 diabetes. Herein we review our understanding of molecular clock control of glucose homeostasis, detail the influence of circadian disruption on glucose metabolism in critical peripheral tissues, explore the contribution of melatonin signalling to the aetiology of type 2 diabetes, and discuss the pros and cons of melatonin chronopharmacotherapy in disease management.
Collapse
Affiliation(s)
- Andrew C Forrestel
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 693, Rochester, NY, 14642, USA
| | - Susanne U Miedlich
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 693, Rochester, NY, 14642, USA
| | - Michael Yurcheshen
- UR Medicine Sleep Center, Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Steven D Wittlin
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 693, Rochester, NY, 14642, USA
| | - Michael T Sellix
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 693, Rochester, NY, 14642, USA.
| |
Collapse
|
29
|
Hardeland R. Melatonin and the pathologies of weakened or dysregulated circadian oscillators. J Pineal Res 2017; 62. [PMID: 27763686 DOI: 10.1111/jpi.12377] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/18/2016] [Indexed: 12/20/2022]
Abstract
Dynamic aspects of melatonin's actions merit increasing future attention. This concerns particularly entirely different effects in senescent, weakened oscillators and in dysregulated oscillators of cancer cells that may be epigenetically blocked. This is especially obvious in the case of sirtuin 1, which is upregulated by melatonin in aged tissues, but strongly downregulated in several cancer cells. These findings are not at all controversial, but are explained on the basis of divergent changes in weakened and dysregulated oscillators. Similar findings can be expected to occur in other accessory oscillator components that are modulated by melatonin, among them several transcription factors and metabolic sensors. Another cause of opposite effects concerns differences between nocturnally active laboratory rodents and the diurnally active human. This should be more thoroughly considered in the field of metabolic syndrome and related pathologies, especially with regard to type 2 diabetes and other aspects of insulin resistance. Melatonin was reported to impair glucose tolerance in humans, especially in carriers of the risk allele of the MT2 receptor gene, MTNR1B, that contains the SNP rs10830963. These findings contrast with numerous reports on improvements of glucose tolerance in preclinical studies. However, the relationship between melatonin and insulin may be more complex, as indicated by loss-of-function mutants of the MT2 receptor that are also prodiabetic, by the age-dependent time course of risk allele overexpression, by progressive reduction in circadian amplitudes and melatonin secretion, which are aggravated in diabetes. By supporting high-amplitude rhythms, melatonin may be beneficial in preventing or delaying diabetes.
Collapse
Affiliation(s)
- Rüdiger Hardeland
- Johann Friedrich Blumenbach Institute of Zoology and Anthropology, University of Göttingen, Göttingen, Germany
| |
Collapse
|
30
|
Jahan S, Munir F, Razak S, Mehboob A, Ain QU, Ullah H, Afsar T, Shaheen G, Almajwal A. Ameliorative effects of rutin against metabolic, biochemical and hormonal disturbances in polycystic ovary syndrome in rats. J Ovarian Res 2016; 9:86. [PMID: 27923406 PMCID: PMC5142269 DOI: 10.1186/s13048-016-0295-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 11/29/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is the most prevalent endocrinopathy in women of reproductive age. The study was commenced to assess the favorable effects of Rutin against metabolic, biochemical, histological, and androgenic aspects of polycystic ovary syndrome in rats. METHODS Female Sprague-Dawley rats were administered letrozole (1 mg/kg) per orally (p.o) for a period of 21 days for the induction of PCOS, followed by dose of rutin (100 mg/kg and 150 mg/kg, p.o) for 15 days using 0.5% w/v CMC as vehicle. Metformin was also given as a standard control to one of the rat groups. Serum estradiol, progesterone, testosterone, serum lipid parameters, CRP and glucose levels were evaluated. Furthermore, antioxidant activity was tested using superoxide dismutase, catalase, glutathione per-oxidase and reactive-oxygen species level. RESULTS Rutin flavonoid had a dose-dependent effect on androgenic levels depicting more recovery in the rutin-I treated group, while rutin-II treated groups showed better antioxidant and lipid profiles as compared with PCOS groups. A decrease in the value of C reactive protein (CRP) and a restoration in the proportion of estrous phase smears were observed in the rutin treated groups. Histopathological examination of ovary revealed a significant decrease in the number of cystic follicles in post treated groups. The effects observed with rutin were moderately similar to that with standard metformin, a widely used treatment drug for PCOS. CONCLUSION The study provides evidence for the potential ameliorative effects of rutin against clinical and biochemical features of PCOS.
Collapse
Affiliation(s)
- Sarwat Jahan
- Reproductive Physiology Laboratory, Department Of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Faryal Munir
- Reproductive Physiology Laboratory, Department Of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Suhail Razak
- Reproductive Physiology Laboratory, Department Of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Anam Mehboob
- Reproductive Physiology Laboratory, Department Of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Qurat Ul Ain
- Reproductive Physiology Laboratory, Department Of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Hizb Ullah
- Reproductive Physiology Laboratory, Department Of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Tayyaba Afsar
- Department of Biochemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ghazala Shaheen
- Reproductive Physiology Laboratory, Department Of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ali Almajwal
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
31
|
Sen A, Sellix MT. The Circadian Timing System and Environmental Circadian Disruption: From Follicles to Fertility. Endocrinology 2016; 157:3366-73. [PMID: 27501186 DOI: 10.1210/en.2016-1450] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The internal or circadian timing system is deeply integrated in female reproductive physiology. Considerable details of rheostatic timing function in the neuroendocrine control of pituitary hormone secretion, adenohypophyseal hormone gene expression and secretion, gonadal steroid hormone biosynthesis and secretion, ovulation, implantation, and parturition have been reported. The molecular clock, an autonomous feedback loop oscillator of interacting transcriptional regulators, dictates the timing and amplitude of gene expression in each tissue of the female hypothalamic-pituitary-gonadal (HPG) axis. Although multiple targets of the molecular clock have been identified, many associated with critical physiological functions in the HPG axis, the full extent of clock-driven gene expression and physiology in this critical system remains unknown. Environmental circadian disruption (ECD), the disturbance of temporal relationships within and between internal clocks (brain and periphery), and external timing cues (eg, light, nutrients, social cues) due to rotating/night shift work or transmeridian travel have been linked to reproductive dysfunction and subfertility. Moreover, ECD resulting from exposure to endocrine disrupting chemicals, environmental toxins, and/or irregular hormone levels during sexual development can also reduce fertility. Thus, perturbations that disturb clock function at the molecular, cellular or systemic level correlate with significant declines in female reproductive function. Here we briefly review the evidence for molecular clock function in each tissue of the female HPG axis (GnRH neuron, pituitary, uterus, oviduct, and ovary), describe the human epidemiological and animal data supporting the negative effects of ECD on fertility, and explore the potential for novel chronotherapeutics in women's health and fertility.
Collapse
Affiliation(s)
- Aritro Sen
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester New York 14642
| | - Michael T Sellix
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester New York 14642
| |
Collapse
|
32
|
Nirwane A, Majumdar A. Resveratrol and pterostilbene ameliorate the metabolic derangements associated with smokeless tobacco in estrogen deficient female rats. J Funct Foods 2016. [DOI: 10.1016/j.jff.2015.12.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
33
|
Nohara K, Yoo SH, Chen Z(J. Manipulating the circadian and sleep cycles to protect against metabolic disease. Front Endocrinol (Lausanne) 2015; 6:35. [PMID: 25852644 PMCID: PMC4369727 DOI: 10.3389/fendo.2015.00035] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/03/2015] [Indexed: 11/30/2022] Open
Abstract
Modernization of human society parallels an epidemic of metabolic disorders including obesity. Apart from excess caloric intake, a 24/7 lifestyle poses another important challenge to our metabolic health. Recent research under both laboratory and epidemiological settings has indicated that abnormal temporal organization of sleep and wakeful activities including food intake is a significant risk factor for metabolic disease. The circadian clock system is our intrinsic biological timer that regulates internal rhythms such as the sleep/wake cycle and also responses to external stimuli including light and food. Initially thought to be mainly involved in the timing of sleep, the clock, and/or clock genes may also play a role in sleep architecture and homeostasis. Importantly, an extensive body of evidence has firmly established a master regulatory role of the clock in energy balance. Together, a close relationship between well-timed circadian/sleep cycles and metabolic health is emerging. Exploiting this functional connection, an important holistic strategy toward curbing the epidemic of metabolic disorders (e.g., obesity) involves corrective measures on the circadian clock and sleep. In addition to behavioral and environmental interventions including meal timing and light control, pharmacological agents targeting sleep and circadian clocks promise convenient and effective applications. Recent studies, for example, have reported small molecules targeting specific clock components and displaying robust beneficial effects on sleep and metabolism. Furthermore, a group of clock-amplitude-enhancing small molecules (CEMs) identified via high-throughput chemical screens are of particular interest for future in vivo studies of their metabolic and sleep efficacies. Elucidating the functional relationship between clock, sleep, and metabolism will also have far-reaching implications for various chronic human diseases and aging.
Collapse
Affiliation(s)
- Kazunari Nohara
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zheng (Jake) Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- *Correspondence: Zheng (Jake) Chen, Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 6.200, Houston, TX 77030, USA e-mail:
| |
Collapse
|