1
|
Yan H, Wang L, Zhang G, Li N, Zhao Y, Liu J, Jiang M, Du X, Zeng Q, Xiong D, He L, Zhou Z, Luo M, Liu W. Oxidative stress and energy metabolism abnormalities in polycystic ovary syndrome: from mechanisms to therapeutic strategies. Reprod Biol Endocrinol 2024; 22:159. [PMID: 39722030 DOI: 10.1186/s12958-024-01337-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024] Open
Abstract
Polycystic ovary syndrome (PCOS), as a common endocrine and metabolic disorder, is often regarded as a primary cause of anovulatory infertility in women. The pathogenesis of PCOS is complex and influenced by multiple factors. Emerging evidence highlights that energy metabolism dysfunction and oxidative stress in granulosa cells (GCs) are pivotal contributors to aberrant follicular development and impaired fertility in PCOS patients. Mitochondrial dysfunction, increased oxidative stress, and disrupted glucose metabolism are frequently observed in individuals with PCOS, collectively leading to compromised oocyte quality. This review delves into the mechanisms linking oxidative stress and energy metabolism abnormalities in PCOS, analyzing their adverse effects on reproductive function. Furthermore, potential therapeutic strategies to mitigate oxidative stress and metabolic disturbances are proposed, providing a theoretical basis for advancing clinical management of PCOS.
Collapse
Affiliation(s)
- Heqiu Yan
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Woman's and Children's Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, 610045, China
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, 610000, China
| | - Li Wang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Woman's and Children's Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, 610045, China
| | - Guohui Zhang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Woman's and Children's Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, 610045, China
| | - Ningjing Li
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 611137, China
| | - Yuhong Zhao
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, 610000, China
| | - Jun Liu
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 611137, China
| | - Min Jiang
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 611137, China
| | - Xinrong Du
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 611137, China
| | - Qin Zeng
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Woman's and Children's Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, 610045, China
| | - Dongsheng Xiong
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Woman's and Children's Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, 610045, China
| | - Libing He
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Woman's and Children's Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, 610045, China
| | - Zhuoting Zhou
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 611137, China
| | - Mengjun Luo
- Department of Clinical Laboratory, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, No. 1617 Ri Yue Street, Chengdu, Sichuan, 611731, China.
| | - Weixin Liu
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Woman's and Children's Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, 610045, China.
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, 610000, China.
| |
Collapse
|
2
|
Dubey P, Reddy S, Sharma K, Johnson S, Hardy G, Dwivedi AK. Polycystic Ovary Syndrome, Insulin Resistance, and Cardiovascular Disease. Curr Cardiol Rep 2024; 26:483-495. [PMID: 38568339 DOI: 10.1007/s11886-024-02050-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/20/2024] [Indexed: 06/26/2024]
Abstract
PURPOSE OF REVIEW Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder in women of reproductive age. It has been associated with metabolic, reproductive, and psychiatric disorders. Despite its association with insulin resistance (IR) and cardiovascular disease (CVD) risk factors, the association between PCOS and CVD outcomes has been conflicting. This review reports the updated evidence between PCOS, insulin resistance, and CVD events. RECENT FINDINGS IR is highly prevalent occurring in 50 to 95% of general and obese PCOS women. The etiology of PCOS involves IR and hyperandrogenism, which lead to CVD risk factors, subclinical CVD, and CVD outcomes. Multiple studies including meta-analysis confirmed a strong association between PCOS and CVD events including ischemic heart disease, stroke, atrial fibrillation, and diabetes, particularly among premenopausal women, and these associations were mediated by metabolic abnormalities. PCOS is highly familial and has substantial CVD risk and transgenerational effects regardless of obesity. A personalized approach to the CVD risk assessment and management of symptom manifestations should be conducted according to its phenotypes. Lifestyle modifications and reduction in environmental stressors should be encouraged for CVD prevention among PCOS women.
Collapse
Affiliation(s)
- Pallavi Dubey
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA.
| | - Sireesha Reddy
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
| | - Kunal Sharma
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
| | - Sarah Johnson
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA
| | - Ghislain Hardy
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
| | - Alok Kumar Dwivedi
- Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, USA
| |
Collapse
|
3
|
Khan H, Rafi Z, Khan MY, Maarfi F, Rehman S, Kaur K, Ahmad MK, Shahab U, Ahmad N, Ahmad S. Epigenetic contributions to cancer: Exploring the role of glycation reactions. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 387:143-193. [PMID: 39179346 DOI: 10.1016/bs.ircmb.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2024]
Abstract
Advanced Glycation End-products (AGEs), with their prolonged half-life in the human body, are emerging as potent diagnostic indicators. Early intervention studies, focusing on AGE cross-link breakers, have shown encouraging results in heart failure patients, paving the way for disease progression monitoring and therapy effectiveness evaluation. AGEs are the byproducts of a non-enzymatic reaction where sugars interact with proteins, lipids, and nucleic acids. These compounds possess the power to alter numerous biological processes, ranging from disrupting molecular conformation and promoting cross-linking to modifying enzyme activity, reducing clearance, and impairing receptor recognition. The damage inflicted by AGEs through the stimulation of intracellular signaling pathways is associated with the onset of chronic diseases across various organ systems. This review consolidates the characteristics of AGEs and the challenges posed by their expression in diverse physiological and pathological states. Furthermore, it highlights the clinical relevance of AGEs and the latest research breakthroughs aimed at reducing AGE accumulation.
Collapse
Affiliation(s)
- Hamda Khan
- Department of Biochemistry, Faculty of Medicine, Jawahar Lal Nehru Medical College, Aligarh Muslim University, Aligarh, India
| | - Zeeshan Rafi
- Department of Bioengineering, Integral University, Lucknow, India
| | - Mohd Yasir Khan
- School of Applied & Life Sciences, Uttaranchal University, Dehradun, India
| | - Farah Maarfi
- School of Applied & Life Sciences, Uttaranchal University, Dehradun, India
| | | | - Kirtanjot Kaur
- University Centre for Research and Development, Chandigarh University, Mohali, India
| | | | - Uzma Shahab
- Department of Biochemistry, King George Medical University, Lucknow, India
| | - Naved Ahmad
- Department of Computer Science and Information System, College of Applied Sciences, AlMaarefa University, Riyadh, Saudi Arabia
| | - Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Saudi Arabia.
| |
Collapse
|
4
|
Fu LW, Gao Z, Zhang N, Yang N, Long HY, Kong LY, Li XY. Traditional Chinese medicine formulae: A complementary method for the treatment of polycystic ovary syndrome. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117698. [PMID: 38171464 DOI: 10.1016/j.jep.2023.117698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/04/2023] [Accepted: 12/30/2023] [Indexed: 01/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Polycystic ovary syndrome (PCOS) is a prevalent female endocrine condition that significantly affects women of all age groups and is characterized by metabolic dysfunction. The efficacy of existing pharmaceutical interventions for the treatment of PCOS remains inadequate. With a rich history and cultural significance spanning thousands of years, Traditional Chinese Medicine (TCM) is extensively employed for treating a variety of ailments and can serve as a supplementary therapy for managing PCOS. Multiple clinical observations and laboratory tests have unequivocally demonstrated the substantial effectiveness and safety of TCM formulae in treating PCOS, and further investigations are currently in progress. AIM OF THE STUDY To summarize the TCM formulae commonly employed in the clinical management of PCOS, examine their therapeutic benefits, investigate their mechanism of action, active constituents, and establish the correlation between efficacy, mechanism of action, and active constituents. MATERIALS AND METHODS We conducted a comprehensive search on PubMed, Web of Science, and China national knowledge infrastructure (CNKI) using the following keywords: "Polycystic Ovary Syndrome", "Traditional Chinese Medicine Decoctions", "Traditional Chinese Medicine formulae", "Traditional Chinese Medicine", "Clinical Observation", "Mechanism", "Treatment", "Pharmacology", and various combinations of these terms. From January 1, 2006 until October 7, 2023, (inclusive). RESULTS This paper summarized the clinical effectiveness, mechanism of action, and active components of 8 TCM formulae for the treatment of PCOS. Our research indicates that TCM formulae can potentially treat PCOS by enhancing the levels of hyperandrogenism and other endocrine hormones, decreasing insulin resistance and hyperinsulinemia, and controlling chronic low-grade inflammation, among other modes of action. In addition, we found an association between epigenetics and TCM formulae for the treatment of PCOS. CONCLUSION TCM formulae have specific advantages in the treatment of Polycystic Ovary Syndrome (PCOS). They achieve therapeutic benefits by targeting several pathways and connections, attracting considerable interest and playing a vital role in the treatment of PCOS. TCM formulae can be used as an adjunctive therapy for the treatment of PCOS.
Collapse
Affiliation(s)
- Li-Wen Fu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zu Gao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Ning Zhang
- Department of Reproduction and Genetics, Shandong Province Hospital of Traditional Chinese, Affiliated Hospital, Shandong University of Traditional Chinese Medicine, Jinan, 250000, China
| | - Nan Yang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Hui-Yan Long
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Ling-Yuan Kong
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiu-Yang Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
5
|
Hou X, Ling Z, Guo Y, Su Y, Wang H, Li H, Lu Y, Chen X, Ji C, Shen R. Peptide derived from RAGE efficiently improves oocyte development through attenuating oxidative stress in oocytes of mice with polycystic ovary syndrome. FASEB J 2024; 38:e23553. [PMID: 38470398 DOI: 10.1096/fj.202302038rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 02/12/2024] [Accepted: 02/27/2024] [Indexed: 03/13/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a common and complex endocrine disorder in reproductive-aged women that frequently leads to infertility due to poor oocyte quality. In this study, we identified a new active peptide (advanced glycation end products receptors RAGE344-355 ) from PCOS follicular fluid using mass spectrometry. We found that supplementing PCOS-like mouse oocytes with RAGE344-355 attenuated both meiotic defects and oxidative stress levels, ultimately preventing developmental defects. Additionally, our results suggest that RAGE344-355 may interact with eEF1a1 to mitigate oxidative meiotic defects in PCOS-like mouse oocytes. These findings highlight the potential for further clinical development of RAGE344-355 as a potent supplement and therapeutic option for women with PCOS. This research addresses an important clinical problem and offers promising opportunities for improving oocyte quality in PCOS patients.
Collapse
Affiliation(s)
- Xiaojing Hou
- Nanjing Women and Children's HealthCare Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhonghui Ling
- Nanjing Women and Children's HealthCare Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Yaping Guo
- Nanjing Women and Children's HealthCare Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Su
- Nanjing Women and Children's HealthCare Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Hanbin Wang
- Nanjing Women and Children's HealthCare Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Hang Li
- Nanjing Women and Children's HealthCare Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Yuxia Lu
- Nanjing Women and Children's HealthCare Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaojiao Chen
- Nanjing Women and Children's HealthCare Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Chenbo Ji
- Nanjing Women and Children's HealthCare Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Rong Shen
- Nanjing Women and Children's HealthCare Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
6
|
Mirani M, Bahmanpour S, Masjedi F, Derakhshan Z, Dara M, Nasr-Esfahani MH, Tabei SMB. Pyridoxamine protects human granulosa cells against advanced glycation end-products-induced steroidogenesis disturbances. Mol Biol Rep 2023; 50:8537-8549. [PMID: 37642758 DOI: 10.1007/s11033-023-08723-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/31/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Ovarian advanced glycation end-products (AGEs) accumulation is associated with ovarian granulosa cells (GCs) dysfunction. Vitamin B6 derivatives positively affected reproduction. The current study was conducted to elucidate the AGEs effects on human luteinized mural GCs steroidogenesis in the presence or absence of pyridoxamine (PM). METHODS AND RESULTS Isolated GCs of 50 healthy women were divided into four parts and treated with media alone (Control), PM alone, or human glycated albumin (HGA) with/without PM. Main steroidogenic enzymes and hormones were assessed by qRT-PCR and ELISA. The AGE receptor (RAGE) protein was also determined using Western blotting. The non-toxic concentration of HGA increased the expression of RAGE, StAR, 3β-HSD, and 17β-HSD (P < 0.0001 for all) but decreased the expression of CYP19A1 at mRNA levels. The increased RAGE protein expression was also confirmed by western blot analysis. These effects resulted in declined estradiol (E2), slightly, and a sharp rise in progesterone (P4) and testosterone (T) levels, respectively. PM, on its own, ameliorated the HGA-altered enzyme expression and, thereby, corrected the aberrant levels of E2, P4, and T. These effects are likely mediated by regulating the RAGE gene and protein expression. CONCLUSION This study indicates that hormonal dysfunctions induced by the AGEs-RAGE axis in luteinized GCs are likely rectified by PM treatment. This effect is likely acquired by reduced expression of RAGE. A better understanding of how AGEs and PM interact in ovarian physiology and pathology may lead to more targeted therapy for treating ovarian dysfunction.
Collapse
Affiliation(s)
- Maryam Mirani
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soghra Bahmanpour
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Masjedi
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Derakhshan
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahintaj Dara
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - Seyed Mohammad Bagher Tabei
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, 7134845794, Iran.
| |
Collapse
|
7
|
Tian Z, Chen S, Shi Y, Wang P, Wu Y, Li G. Dietary advanced glycation end products (dAGEs): An insight between modern diet and health. Food Chem 2023; 415:135735. [PMID: 36863235 DOI: 10.1016/j.foodchem.2023.135735] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 02/06/2023] [Accepted: 02/15/2023] [Indexed: 03/04/2023]
Abstract
Advanced glycation end products (AGEs) are formed by a series of chemical reactions of amino acids, peptides, proteins, and ketones at normal temperature or heated non-enzymatic conditions. A large amount of AGEs derived from Maillard Reaction (MR) during the process of food heat-processing. After oral intake, dietary AGEs are converted into biological AGEs through digestion and absorption, and accumulated in almost all organs. The safety and health risk of dietary AGEs have attracted wide attention. Increasing evidence have shown that uptake of dietary AGEs is closely related to the occurrence of many chronic diseases, such as diabetes, chronic kidney disease, osteoporosis, and Alzheimer's disease. This review summarized the most updated information of production, bio-transport in vivo, detection technologies, and physiological toxicity of dietary AGEs, and also discussed approaches to inhibit dietary AGEs generation. Impressively, the future opportunities and challenges on the detection, toxicity, and inhibition of dietary AGEs are raised.
Collapse
Affiliation(s)
- Zhaoqing Tian
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Shasha Chen
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Yiheng Shi
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Panpan Wang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Yongning Wu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China; NHC Key Laboratory of Food Safety Risk Assessment, Food Safety Research Unit (2019RU014) of Chinese Academy of Medical Science, China National Center for Food Safety Risk Assessment, Beijing 100021, China
| | - Guoliang Li
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| |
Collapse
|
8
|
Yifu P. Evidence for causal effects of polycystic ovary syndrome on oxidative stress: a two-sample mendelian randomisation study. BMC Med Genomics 2023; 16:141. [PMID: 37337194 DOI: 10.1186/s12920-023-01581-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 06/13/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is often accompanied by increased oxidative stress levels; however, it is still unclear whether PCOS itself is causally related to oxidative stress (OS), whether OS can increase the occurrence of PCOS, and which characteristics of PCOS increase OS levels. Therefore, this study explored the causal relationship between PCOS, its characteristics, and OS. METHODS Two-sample bidirectional and two-sample Mendelian randomisation studies were performed based on publicly available statistics from genome-wide association studies. PCOS; its characteristics, such as testosterone, low-density lipoprotein, high-density lipoprotein; and 11 major OS markers (superoxide dismutase, glutathione S-transferase, glutathione peroxidase, catalase, uric acid, zinc, tocopherol, ascorbic acid, retinol, albumin, and total bilirubin), were studied. The main analytical method used was inverse variance weighting (IVW). Pleiotropy was evaluated using the Mendelian randomisation-Egger intercept. Q and P values were used to assess heterogeneity. RESULTS There was no causal relationship between PCOS and the OS indices (all P > 0.05). There was a causal relationship between the OS index, ascorbate level, and PCOS (IVW, odds ratio: 2.112, 95% confidence interval: 1.257-3.549, P = 0.005). In addition, there was a causal relationship between testosterone, low-density lipoprotein, high-density lipoprotein, sex hormone-binding globulin, body mass index, triacylglycerol, age at menarche, and most OS indices according to the IVW method. The F statistics showed that there was no weak instrumental variable. A sensitivity analysis was performed using the leave-one-out method. No pleiotropy was observed. The results were robust, and the conclusions were reliable. CONCLUSIONS This study showed for the first time that there was no causal relationship between PCOS and OS. However, there was a causal relationship between the OS index, ascorbate level, and PCOS. It revealed that PCOS itself could not increase OS, and the increase in OS in PCOS was related to other potential factors, such as testosterone, low-density lipoprotein, high-density lipoprotein, sex hormone-binding globulin, body mass index, triacylglycerol, and age at menarche.
Collapse
Affiliation(s)
- Pu Yifu
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
9
|
Zhao H, Zhang J, Cheng X, Nie X, He B. Insulin resistance in polycystic ovary syndrome across various tissues: an updated review of pathogenesis, evaluation, and treatment. J Ovarian Res 2023; 16:9. [PMID: 36631836 PMCID: PMC9832677 DOI: 10.1186/s13048-022-01091-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/26/2022] [Indexed: 01/12/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine disorder characterized by chronic ovulation dysfunction and overabundance of androgens; it affects 6-20% of women of reproductive age. PCOS involves various pathophysiological factors, and affected women usually have significant insulin resistance (IR), which is a major cause of PCOS. IR and compensatory hyperinsulinaemia have differing pathogeneses in various tissues, and IR varies among different PCOS phenotypes. Genetic and epigenetic changes, hyperandrogenaemia, and obesity aggravate IR. Insulin sensitization drugs are a new treatment modality for PCOS. We searched PubMed, Google Scholar, Elsevier, and UpToDate databases in this review, and focused on the pathogenesis of IR in women with PCOS and the pathophysiology of IR in various tissues. In addition, the review provides a comprehensive overview of the current progress in the efficacy of insulin sensitization therapy in the management of PCOS, providing the latest evidence for the clinical treatment of women with PCOS and IR.
Collapse
Affiliation(s)
- Han Zhao
- grid.412467.20000 0004 1806 3501Department of Endocrinology, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000 People’s Republic of China
| | - Jiaqi Zhang
- grid.412467.20000 0004 1806 3501Department of Endocrinology, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000 People’s Republic of China
| | - Xiangyi Cheng
- grid.412467.20000 0004 1806 3501Department of Endocrinology, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000 People’s Republic of China
| | - Xiaozhao Nie
- grid.412467.20000 0004 1806 3501Department of Endocrinology, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110000 People’s Republic of China
| | - Bing He
- Department of Endocrinology, Shengjing Hospital, China Medical University, Shenyang, Liaoning, 110000, People's Republic of China.
| |
Collapse
|
10
|
Luo ED, Jiang HM, Chen W, Wang Y, Tang M, Guo WM, Diao HY, Cai NY, Yang X, Bian Y, Xing SS. Advancements in lead therapeutic phytochemicals polycystic ovary syndrome: A review. Front Pharmacol 2023; 13:1065243. [PMID: 36699064 PMCID: PMC9868606 DOI: 10.3389/fphar.2022.1065243] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/12/2022] [Indexed: 01/11/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrine diseases in women of reproductive age and features complex pathological symptoms and mechanisms. Existing medical treatments have, to some extent, alleviated the deterioration of PCOS. However, these strategies only temporarily control symptoms, with a few side effects and no preventive effect. Phytochemicals extracted from medicinal herbs and plants are vital for discovering novel drugs. In recent years, many kinds of research have proven that phytochemicals isolated from traditional Chinese medicine (TCM) and medicinal plants show significant potential in preventing, alleviating, and treating PCOS. Nevertheless, compared to the abundance of experimental literature and minimal specific-topic reviews related to PCOS, there is a lack of systematic reviews to summarize these advancements in this promising field. Under this background, we systematically document the progress of bioactive phytochemicals from TCM and medicinal plants in treating PCOS, including flavonoids, polyphenols, and alkaloids. According to the literature, these valuable phytochemicals demonstrated therapeutic effects on PCOS supported by in vivo and in vitro experiments, mainly depending on anti-inflammatory, antioxidation, improvement of hormone disorder and insulin resistance (IR), and alleviation of hyperinsulinemia. Based on the current progress, future research directions should emphasize 1) exploring bioactive phytochemicals that potentially mediate bone metabolism for the treatment of PCOS; 2) improving unsatisfactory bioavailability by using advanced drug delivery systems such as nanoparticles and antibody-conjugated drugs, as well as a chemical modification; 3) conducting in-depth research on the pathogenesis of PCOS to potentially impact the gut microbiota and its metabolites in the evolution of PCOS; 4) revealing the pharmacological effects of these bioactive phytochemicals on PCOS at the genetic level; and 5) exploring the hypothetical and unprecedented functions in regulating PCOS by serving as proteolysis-targeting chimeras and molecular glues compared with traditional small molecule drugs. In brief, this review aims to provide detailed mechanisms of these bioactive phytochemicals and hopefully practical and reliable insight into clinical applications concerning PCOS.
Collapse
Affiliation(s)
- Er-Dan Luo
- GCP Institution, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hai-Mei Jiang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Chen
- Traditional Chinese Medicine Department, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Chengdu, China
| | - Mi Tang
- GCP Institution, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wen-Mei Guo
- GCP Institution, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hao-Yang Diao
- GCP Institution, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ning-Yuan Cai
- GCP Institution, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiao Yang
- GCP Institution, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ying Bian
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Chengdu, China
| | - Sha-Sha Xing
- GCP Institution, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
11
|
Cyproterone Acetate Mediates IRE1α Signaling Pathway to Alleviate Pyroptosis of Ovarian Granulosa Cells Induced by Hyperandrogen. BIOLOGY 2022; 11:biology11121761. [PMID: 36552271 PMCID: PMC9775519 DOI: 10.3390/biology11121761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/09/2022]
Abstract
OBJECTIVE Hyperandrogenemia (HA) is the main pathophysiological change that takes place in polycystic ovary syndrome (PCOS). Cyproterone acetate (CYA) is a drug commonly used to reduce androgen in patients with PCOS. Long-term and continuous exposure to HA can cause ovarian granulosa cells (GCs), pyroptotic death, and follicular dysfunction in PCOS mice. The aim of this study was to investigate whether CYA could ameliorate the hyperandrogenemia-induced pyroptosis of PCOS ovarian GCs by alleviating the activation of the IRE1α signaling pathway. METHODS Firstly, thirty PCOS patients with HA as their main clinical manifestation were selected as the study group, and thirty non-PCOS patients were selected as the control group. The GCs and follicular fluid of the patients were collected, and the expression of pyroptosis-related proteins was detected. Secondly, a PCOS mouse model induced by dehydroepiandrosterone (DHEA) was constructed, and the treatment group model was constructed with the subcutaneous injection of cyproterone acetate in PCOS mice. The expression of pyroptosis-related protein in ovarian GCs was detected to explore the alleviating effect of CYA on the pyroptosis of ovarian GCs in PCOS mice. Thirdly, KGN cells-i.e., from the human GC line-were cultured with dihydrotestosterone, CYA, and ERN1 (IRE1α gene) small interfering RNA in vitro to explore whether CYA can alleviate the activation of the IRE1α signaling pathway and ameliorate the hyperandrogenemia-induced pyroptosis of PCOS ovarian GCs. RESULTS The expression of pyroptosis-related proteins was significantly increased in ovarian GCs of PCOS patients with HA as the main clinical manifestation, and in the PCOS mouse model induced by DHEA. After treatment with CYA, the expression of pyroptosis-related proteins in the ovarian GCs of mice was significantly lower than that in PCOS mice. In vitro experiments showed that CYA could ameliorate KGN cells' pyroptosis by alleviating the activation of the IRE1α signaling pathway. CONCLUSION This study showed that CYA could ameliorate the activation of the IRE1α signaling pathway in mouse GCs and KGN cells, and also alleviate pyroptosis in ovarian GCs. This study provides a new mechanism and evidential support for CYA in the treatment of PCOS patients.
Collapse
|
12
|
Szukiewicz D, Trojanowski S, Kociszewska A, Szewczyk G. Modulation of the Inflammatory Response in Polycystic Ovary Syndrome (PCOS)-Searching for Epigenetic Factors. Int J Mol Sci 2022; 23:ijms232314663. [PMID: 36498989 PMCID: PMC9736994 DOI: 10.3390/ijms232314663] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women of reproductive age. Despite its incidence, the syndrome is poorly understood and remains underdiagnosed, and female patients are diagnosed with a delay. The heterogenous nature of this complex disorder results from the combined occurrence of genetic, environmental, endocrine, and behavioral factors. Primary clinical manifestations of PCOS are derived from the excess of androgens (anovulation, polycystic ovary morphology, lack of or scanty, irregular menstrual periods, acne and hirsutism), whereas the secondary manifestations include multiple metabolic, cardiovascular, and psychological disorders. Dietary and lifestyle factors play important roles in the development and course of PCOS, which suggests strong epigenetic and environmental influences. Many studies have shown a strong association between PCOS and chronic, low-grade inflammation both in the ovarian tissue and throughout the body. In the vast majority of PCOS patients, elevated values of inflammatory markers or their gene markers have been reported. Development of the vicious cycle of the chronic inflammatory state in PCOS is additionally stimulated by hyperinsulinemia and obesity. Changes in DNA methylation, histone acetylation and noncoding RNA levels are presented in this review in the context of oxidative stress, reactive oxygen species, and inflammatory signaling in PCOS. Epigenetic modulation of androgenic activity in response to inflammatory signaling is also discussed.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
- Correspondence:
| | - Seweryn Trojanowski
- Chair and Department of Obstetrics, Gynecology and Gynecological Oncology, Medical University of Warsaw, 03-242 Warsaw, Poland
| | - Anna Kociszewska
- Chair and Department of Obstetrics, Gynecology and Gynecological Oncology, Medical University of Warsaw, 03-242 Warsaw, Poland
| | - Grzegorz Szewczyk
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
13
|
Song X, Ge L, Wang D, Li L, Ma D, Li X, Song X. Family-based analysis of -675 4G/5G polymorphism in the PAI-1 gene of polycystic ovary syndrome in Chinese population. J OBSTET GYNAECOL 2022; 42:3304-3308. [PMID: 36065507 DOI: 10.1080/01443615.2022.2114326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Previous studies have shown that 4G/5G polymorphism in promoter region of plasminogen activator inhibitor-1 (PAI-1) gene can affect insulin sensitivity by elevating the level and activity of plasma PAI-1. In order to elucidate the relationship between the polymorphism of PAI-1 gene and polycystic ovary syndrome (PCOS), we used transmission disequilibrium test (TDT) to study the family of PCOS. Eight hundred and fifty-five participants consisting of 285 trios (mother, father and offspring with PCOS) were recruited at the Center of Reproductive Medicine, Shandong University from July 2007 to August 2014. 4G/5G polymorphism of PAI-1 gene was genotyped using direct sequencing protocol and TDT was used to analyse the association between PAI-1 gene and PCOS. Though the 5G allele in PAI-1 gene was overtransmitted in families, no statistical significance existed and there was no association between PAI-1 gene and PCOS, indicating that PAI-1 gene was unlikely to play a major role in the aetiology of PCOS in Chinese population.Impact StatementWhat is already known on this subject? Some studies have shown that 4G/5G polymorphism in promoter region of plasminogen activator inhibitor-1 (PAI-1) gene can affect insulin sensitivity by elevating the level and activity of plasma PAI-1, participating in the formation of insulin resistance (IR).What do the results of this study add? Though the 5G allele in PAI-1 gene was overtransmitted in families, no statistical significance existed and there was no association between PAI-1 gene and polycystic ovary syndrome (PCOS).What are the implications of these findings for clinical practice and/or further research? PAI-1 gene was unlikely to play a major role in the aetiology of PCOS.
Collapse
Affiliation(s)
- Xiaocui Song
- Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Li Ge
- Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Dongsha Wang
- Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Li Li
- Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Dongmei Ma
- Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Xiu Li
- Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Xiaocui Song
- Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| |
Collapse
|
14
|
Wu M, Zhang J, Gu R, Dai F, Yang D, Zheng Y, Tan W, Jia Y, Li B, Cheng Y. The role of Sirtuin 1 in the pathophysiology of polycystic ovary syndrome. Eur J Med Res 2022; 27:158. [PMID: 36030228 PMCID: PMC9419382 DOI: 10.1186/s40001-022-00746-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/28/2022] [Indexed: 11/21/2022] Open
Abstract
Polycystic ovarian syndrome (PCOS) is the most common multifactor heterogeneous endocrine and metabolic disease in women of childbearing age. PCOS is a group of clinical syndromes characterized by reproductive disorders, metabolic disorders, and mental health problems that seriously impact the physical and mental health of patients. At present, new studies suggest that human evolution leads to the body changes and the surrounding environment mismatch adaptation, but the understanding of the disease is still insufficient, the pathogenesis is still unclear. Sirtuin 1 (SIRT1), a member of the Sirtuin family, is expressed in various cells and plays a crucial role in cell energy conversion and physiological metabolism. Pathophysiological processes such as cell proliferation and apoptosis, autophagy, metabolism, inflammation, antioxidant stress and insulin resistance play a crucial role. Moreover, SIRT1 participates in the pathophysiological processes of oxidative stress, autophagy, ovulation disturbance and insulin resistance, which may be a vital link in the occurrence of PCOS. Hence, the study of the role of SIRT1 in the pathogenesis of PCOS and related complications will contribute to a more thorough understanding of the pathogenesis of PCOS and supply a basis for the treatment of patients.
Collapse
Affiliation(s)
- Mali Wu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jie Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Ran Gu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yajing Zheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wei Tan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yifan Jia
- Department of Pain, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Bingshu Li
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
15
|
Chen CY, Zhang JQ, Li L, Guo MM, He YF, Dong YM, Meng H, Yi F. Advanced Glycation End Products in the Skin: Molecular Mechanisms, Methods of Measurement, and Inhibitory Pathways. Front Med (Lausanne) 2022; 9:837222. [PMID: 35646963 PMCID: PMC9131003 DOI: 10.3389/fmed.2022.837222] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/21/2022] [Indexed: 12/19/2022] Open
Abstract
Advanced glycation end products (AGEs) are a series of stable compounds produced under non-enzymatic conditions by the amino groups of biomacromolecules and the free carbonyl groups of glucose or other reducing sugars commonly produced by thermally processed foods. AGEs can cause various diseases, such as diabetes, atherosclerosis, neurodegeneration, and chronic kidney disease, by triggering the receptors of AGE (RAGEs) in the human body. There is evidence that AGEs can also affect the different structures and physiological functions of the skin. However, the mechanism is complicated and cumbersome and causes various harms to the skin. This article aims to identify and summarise the formation and characteristics of AGEs, focussing on the molecular mechanisms by which AGEs affect the composition and structure of normal skin substances at different skin layers and induce skin issues. We also discuss prevention and inhibition pathways, provide a systematic and comprehensive method for measuring the content of AGEs in human skin, and summarise and analyse their advantages and disadvantages. This work can help researchers acquire a deeper understanding of the relationship between AGEs and the skin and provides a basis for the development of effective ingredients that inhibit glycation.
Collapse
Affiliation(s)
- Chun-Yu Chen
- Beijing Key Laboratory of Plant Resources Research and Development, Beijing Technology and Business University, Beijing, China.,Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, China.,Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijng, China
| | - Jia-Qi Zhang
- Beijing Key Laboratory of Plant Resources Research and Development, Beijing Technology and Business University, Beijing, China.,Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, China.,Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijng, China
| | - Li Li
- Beijing Key Laboratory of Plant Resources Research and Development, Beijing Technology and Business University, Beijing, China.,Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, China.,Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijng, China
| | - Miao-Miao Guo
- Beijing Key Laboratory of Plant Resources Research and Development, Beijing Technology and Business University, Beijing, China.,Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, China.,Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijng, China
| | - Yi-Fan He
- Beijing Key Laboratory of Plant Resources Research and Development, Beijing Technology and Business University, Beijing, China.,Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, China.,Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijng, China
| | - Yin-Mao Dong
- Beijing Key Laboratory of Plant Resources Research and Development, Beijing Technology and Business University, Beijing, China.,Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, China.,Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijng, China
| | - Hong Meng
- Beijing Key Laboratory of Plant Resources Research and Development, Beijing Technology and Business University, Beijing, China.,Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, China.,Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijng, China
| | - Fan Yi
- Beijing Key Laboratory of Plant Resources Research and Development, Beijing Technology and Business University, Beijing, China.,Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, China.,Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijng, China
| |
Collapse
|
16
|
Mouanness M, Merhi Z. Impact of Dietary Advanced Glycation End Products on Female Reproduction: Review of Potential Mechanistic Pathways. Nutrients 2022; 14:nu14050966. [PMID: 35267940 PMCID: PMC8912317 DOI: 10.3390/nu14050966] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/04/2023] Open
Abstract
Advanced glycation end products (AGEs), a heterogenous group of products formed by the reaction between protein and reducing sugars, can form endogenously due to non-enzymatic reactions or by exogenous sources such as diet where considerable increase in AGEs is observed due to the modification of food mainly by thermal processing. Recent studies have suggested that AGEs could impact, via inducing inflammation and oxidative stress, the reproductive health and fertility in both males and females. This review presents a summary of recently published data pertaining to the pathogenesis of dietary AGEs and their receptors as well as their potential impact on female reproductive health. More specifically, it will present data pertaining to dietary AGEs’ involvement in the mechanistic pathogenesis of polycystic ovary syndrome, ovarian dysfunction, as well as the AGEs’ effect perinatally on the female offspring reproduction. Understanding the mechanistic impact of dietary AGEs on female reproduction can help contribute to the development of targeted pharmacological therapies that will help curb rising female infertility.
Collapse
Affiliation(s)
- Marco Mouanness
- Rejuvenating Fertility Center, 315 W 57th Street, Suite 208, New York, NY 10019, USA;
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Maimonides Medical Center, Brooklyn, NY 11219, USA
| | - Zaher Merhi
- Rejuvenating Fertility Center, 315 W 57th Street, Suite 208, New York, NY 10019, USA;
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Maimonides Medical Center, Brooklyn, NY 11219, USA
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Correspondence:
| |
Collapse
|
17
|
Merhi Z, Du XQ, Charron MJ. Postnatal weaning to different diets leads to different reproductive phenotypes in female offspring following perinatal exposure to high levels of dietary advanced glycation end products. F&S SCIENCE 2022; 3:95-105. [PMID: 35559999 DOI: 10.1016/j.xfss.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To examine, following perinatal exposure to a diet high in advanced glycation end products (AGEs), whether the use of standard AGE-free mouse chow during the postweaning period alters metabolism and reproduction differently than exposure to a diet low in AGEs. DESIGN Experimental animal study. SETTING University-based research laboratory. ANIMAL(S) Female CD1 mice. INTERVENTION(S) Seven-week-old mice were placed on a diet either low or high in AGEs perinatally, before mating and then during pregnancy and lactation. All offspring were weaned onto an AGE-free normal chow. MAIN OUTCOME MEASURE(S) Growth curve, liver and abdominal fat weight, insulin and glucose tolerance tests, vaginal opening, estrous cyclicity, and serum levels of antimüllerian hormone, leptin, and adiponectin were assessed. Ovarian histologic examination for follicular count and gene expression was also performed. RESULT(S) Compared with the mice exposed to a diet low in AGEs, the mice exposed to a diet high in AGEs showed lower body weight in pups, lower liver weight, delayed vaginal opening, higher serum antimüllerian hormone levels, lower primordial and secondary follicle pools, and higher ovarian Fshr messenger RNA levels. CONCLUSION(S) Following weaning, perinatal AGEs can target puberty onset and folliculogenesis differently to standard mouse chow.
Collapse
Affiliation(s)
- Zaher Merhi
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, SUNY Downstate Health Sciences University, Brooklyn, New York; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York.
| | - Xiu Quan Du
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Maureen J Charron
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York; Department of Obstetrics, Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, New York; Department of Medicine and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
18
|
Lin L, Wang L. Knockdown of DPP4 promotes the proliferation and the activation of the CREB/aromatase pathway in ovarian granulosa cells. Mol Med Rep 2022; 25:73. [PMID: 35014677 PMCID: PMC8767454 DOI: 10.3892/mmr.2022.12589] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/21/2021] [Indexed: 11/23/2022] Open
Abstract
Dipeptidyl peptidase 4 (DPP4) has been revealed to be upregulated in women suffering from polycystic ovary syndrome (PCOS), which is a common reproductive disorder. The present study was designed to investigate the effects of inhibition of DPP4 expression on the proliferation of ovarian granulosa cells as well as on the activation of the cAMP response element‑binding protein (CREB)/aromatase pathway. The expression levels of DPP4 in rat serum samples with or without PCOS and ovarian granulosa cells (KGN cells) were detected using reverse transcription‑quantitative PCR (RT‑qPCR) and western blot analyses. Cell viability and cell cycle progression were detected using the Cell Counting Kit‑8 assay and flow cytometric analysis, respectively. The 5‑ethynyl‑2'‑deoxyuridine assay was employed to detect the proliferation of glycolaldehyde‑bovine serum albumin (GOA‑BSA)‑treated KGN cells. In addition, RT‑qPCR and western blot analyses were applied to detect the expression levels of CREB, specific cell cycle‑associated proteins and cytochrome P450 (CYP) 19A1 and CYP11A1 enzymes in KGN cells. The expression levels of DPP4 were upregulated in rats with PCOS. Inhibition of DPP4 expression promoted the proliferation and cell cycle arrest of KGN cells. It was also revealed that the expression levels of cell cycle‑associated proteins were upregulated in DPP4‑silenced KGN cells. In addition, their proliferation was decreased following treatment with GOA‑BSA, while the addition of sitagliptin partially reversed these effects. Additionally, sitagliptin reversed the inhibitory effects caused by GOA‑BSA treatment on the cell cycle progression and on the activation of the CREB/aromatase pathway in KGN cells, as determined by the increased expression levels of the cell cycle‑associated proteins as well as those of the CREB protein and the CYP19A1 and CYP11A1 enzymes. In conclusion, inhibition of DPP4 expression promoted the proliferation of KGN cells and the activation of the CREB/aromatase pathway.
Collapse
Affiliation(s)
- Lina Lin
- Center for Reproductive Medicine, Shantou Central Hospital, Shantou, Guangdong 515041, P.R. China
| | - Liman Wang
- Center for Reproductive Medicine, Shantou Central Hospital, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
19
|
Livadas S, Anagnostis P, Bosdou JK, Bantouna D, Paparodis R. Polycystic ovary syndrome and type 2 diabetes mellitus: A state-of-the-art review. World J Diabetes 2022; 13:5-26. [PMID: 35070056 PMCID: PMC8771268 DOI: 10.4239/wjd.v13.i1.5] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/30/2021] [Accepted: 12/25/2021] [Indexed: 02/06/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) often coexists with a wide spectrum of dysglycemic conditions, ranging from impaired glucose tolerance to type 2 diabetes mellitus (T2D), which occur to a greater extent compared to healthy body mass index-matched women. This concurrence of disorders is mainly attributed to common pathogenetic pathways linking the two entities, such as insulin resistance. However, due to methodological flaws in the available studies and the multifaceted nature of the syndrome, there has been substantial controversy as to the exact association between T2D and PCOS which has not yet been elucidated. The aim of this review is to present the best available evidence regarding the epidemiology of dysglycemia in PCOS, the unique pathophysiological mechanisms underlying the progression of dysglycemia, the most appropriate methods for assessing glycemic status and the risk factors for T2D development in this population, as well as T2D risk after transition to menopause. Proposals for application of a holistic approach to enable optimal management of T2D risk in PCOS are also provided. Specifically, adoption of a healthy lifestyle with adherence to improved dietary patterns, such the Mediterranean diet, avoidance of consumption of endocrine-disrupting foods and beverages, regular exercise, and the effect of certain medications, such as metformin and glucagon-like peptide 1 receptor agonists, are discussed. Furthermore, the maintenance of a healthy weight is highlighted as a key factor in achievement of a significant reduction of T2D risk in women with PCOS.
Collapse
Affiliation(s)
| | - Panagiotis Anagnostis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki 54636, Greece
| | - Julia K Bosdou
- Unit for Human Reproduction, 1st Department of Obstetrics and Gynaecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki 54636, Greece
| | - Dimitra Bantouna
- Department of Pathology and Cytology, University of Patras School of Medicine, Patras 10563, Greece
| | - Rodis Paparodis
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH 23456, United States
| |
Collapse
|
20
|
Saadat N, Puttabyatappa M, Elangovan VR, Dou J, Ciarelli JN, Thompson RC, Bakulski KM, Padmanabhan V. Developmental Programming: Prenatal Testosterone Excess on Liver and Muscle Coding and Noncoding RNA in Female Sheep. Endocrinology 2022; 163:6413684. [PMID: 34718504 PMCID: PMC8667859 DOI: 10.1210/endocr/bqab225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Indexed: 11/19/2022]
Abstract
Prenatal testosterone (T)-treated female sheep manifest peripheral insulin resistance, ectopic lipid accumulation, and insulin signaling disruption in liver and muscle. This study investigated transcriptional changes and transcriptome signature of prenatal T excess-induced hepatic and muscle-specific metabolic disruptions. Genome-wide coding and noncoding (nc) RNA expression in liver and muscle from 21-month-old prenatal T-treated (T propionate 100 mg intramuscular twice weekly from days 30-90 of gestation; term: 147 days) and control females were compared. Prenatal T (1) induced differential expression of messenger RNAs (mRNAs) in liver (15 down, 17 up) and muscle (66 down, 176 up) (false discovery rate < 0.05, absolute log2 fold change > 0.5); (2) downregulated mitochondrial pathway genes in liver and muscle; (3) downregulated hepatic lipid catabolism and peroxisome proliferator-activated receptor (PPAR) signaling gene pathways; (4) modulated noncoding RNA (ncRNA) metabolic processes gene pathway in muscle; and (5) downregulated 5 uncharacterized long noncoding RNA (lncRNA) in the muscle but no ncRNA changes in the liver. Correlation analysis showed downregulation of lncRNAs LOC114112974 and LOC105607806 was associated with decreased TPK1, and LOC114113790 with increased ZNF470 expression. Orthogonal projections to latent structures discriminant analysis identified mRNAs HADHA and SLC25A45, and microRNAs MIR154A, MIR25, and MIR487B in the liver and ARIH1 and ITCH and miRNAs MIR369, MIR10A, and MIR10B in muscle as potential biomarkers of prenatal T excess. These findings suggest downregulation of mitochondria, lipid catabolism, and PPAR signaling genes in the liver and dysregulation of mitochondrial and ncRNA gene pathways in muscle are contributors of lipotoxic and insulin-resistant hepatic and muscle phenotype. Gestational T excess programming of metabolic dysfunctions involve tissue-specific ncRNA-modulated transcriptional changes.
Collapse
Affiliation(s)
- Nadia Saadat
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan 48019-5718, USA
| | - Muraly Puttabyatappa
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan 48019-5718, USA
| | | | - John Dou
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan 48019-5718, USA
| | - Joseph N Ciarelli
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan 48019-5718, USA
| | - Robert C Thompson
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan 48019-5718, USA
| | - Kelly M Bakulski
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan 48019-5718, USA
| | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan 48019-5718, USA
- Correspondence: Vasantha Padmanabhan, PhD, MS, Department of Pediatrics, University of Michigan, 7510 MSRB1, 1150 W Medical Center Dr, Ann Arbor, MI 48019-5718, USA.
| |
Collapse
|
21
|
Harada M. Pathophysiology of polycystic ovary syndrome revisited: Current understanding and perspectives regarding future research. Reprod Med Biol 2022; 21:e12487. [PMID: 36310656 PMCID: PMC9601867 DOI: 10.1002/rmb2.12487] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/01/2022] [Accepted: 09/15/2022] [Indexed: 11/05/2022] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among reproductive-age women and has lifelong effects on health. Methods In this review, I discuss the pathophysiology of PCOS. First, I summarize our current understanding of the etiology and pathology of PCOS, then, discuss details of two representative environmental factors involved in the pathogenesis of PCOS. Finally, I present perspectives regarding the directions of future research. Main findings The pathophysiology of PCOS is heterogeneous and shaped by the interaction of reproductive dysfunction and metabolic disorders. Hyperandrogenism and insulin resistance exacerbate one another during the development of PCOS, which is also affected by dysfunction of the hypothalamus-pituitary-ovarian axis. PCOS is a highly heritable disorder, and exposure to certain environmental factors causes individuals with predisposing genetic factors to develop PCOS. The environmental factors that drive the development of PCOS pathophysiology make a larger contribution than the genetic factors, and may include the intrauterine environment during the prenatal period, the follicular microenvironment, and lifestyle after birth. Conclusion On the basis of this current understanding, three areas are proposed to be subjects for future research, with the ultimate goals of developing therapeutic and preventive strategies and providing appropriate lifelong management, including preconception care.
Collapse
Affiliation(s)
- Miyuki Harada
- Department of Obstetrics and Gynecology, Faculty of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
22
|
Merhi Z, Du XQ, Charron MJ. Perinatal exposure to high dietary advanced glycation end products affects the reproductive system in female offspring in mice. Mol Hum Reprod 2021; 26:615-623. [PMID: 32609365 DOI: 10.1093/molehr/gaaa046] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/28/2020] [Indexed: 12/14/2022] Open
Abstract
Maternal nutrition and the intrauterine environment are important in determining susceptibility to reproductive and metabolic disturbances. Advanced glycation end products (AGEs) are widely consumed in Western diet. The purpose of this study was to determine whether perinatal exposure to a high levels of dietary AGEs affect metabolic and reproductive parameters in female mice offspring. Female CD1 mice, 7 weeks old, were placed on either a diet low (L-AGE) or high (H-AGE) in AGEs before mating and then during pregnancy and lactation. All offspring were weaned onto the L-AGE diet and studied through to 16 weeks of age; they were counted and weighed at birth and then every week for a total of 11 weeks. Vaginal opening, litter size, growth curve, liver and abdominal fat weights, serum levels of anti-Mullerian hormone, leptin and adiponectin, as well as insulin and glucose tolerance tests were compared. Ovaries were harvested for follicular count and gene expression by real-time polymerase chain reaction. Compared to perinatal exposure to the L-AGE diet, perinatal exposure to the H-AGE diet caused lower body weight at birth, and adult offspring exhibited delayed growth, lower serum leptin and adiponectin levels, delayed vaginal opening, irregular oestrous cyclicity, arrested follicular development and significant alterations in the expression of genes involved in folliculogenesis (Amh and Amhr2) and steroidogenesis (Cyp19a1). These results indicate that perinatal exposure to a diet elevated in AGEs causes deficits in perinatal growth, pubertal onset, and reproductive organ development in female mice. Whether these findings translate to humans remains to be determined in future studies.
Collapse
Affiliation(s)
- Zaher Merhi
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA.,Department of Obstetrics and Gynecology NYU School of Medicine, New York, NY 10016, USA
| | - Xiu Quan Du
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Maureen J Charron
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Department of Obstetrics, Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Department of Medicine & the Fleischer Institute for Diabetes & Metabolism, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
23
|
Zhang FF, Zhang Q, Wang YL, Wang FF, Hardiman PJ, Qu F. Intergenerational Influences between Maternal Polycystic Ovary Syndrome and Offspring: An Updated Overview. J Pediatr 2021; 232:272-281. [PMID: 33482217 DOI: 10.1016/j.jpeds.2021.01.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 11/16/2022]
Affiliation(s)
- Fang-Fang Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qing Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuan-Lin Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fang-Fang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Paul J Hardiman
- Institute for Women's Health, University College London, London, United Kingdom
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
24
|
Tkaczuk-Włach J, Kędzierski W, Jonik I, Sadok I, Filip A, Kankofer M, Polkowski W, Ziółkowski P, Gamian A, Staniszewska M. Immunomodulatory Factors in Primary Endometrial Cell Cultures Isolated from Cancer and Noncancerous Human Tissue-Focus on RAGE and IDO1. Cells 2021; 10:cells10051013. [PMID: 33922995 PMCID: PMC8145962 DOI: 10.3390/cells10051013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Immune modulatory factors like indoleamine 2,3-dioxygenase 1 (IDO1) generating kynurenine (Kyn) and receptor for advanced glycation end-products (RAGE) contribute to endometrial and cancer microenvironment. Using adequate experimental models is needed to learn about the significance of these molecular factors in endometrial biology. In this paper we study IDO1 activity and RAGE expression in the in vitro cultured primary human endometrial cells derived from cancerous and noncancerous tissue. Methods: The generated primary cell cultures from cancer and noncancerous endometrial tissues were characterized using immunofluorescence and Western Blot for expression of endometrial and cancer markers. IDO1 activity was studied by Kyn quantification with High Performance Liquid Chromatography with Diode Array Detector. Results: The primary cultures of endometrial cells were obtained with 80% success rate and no major genetic aberrations. The cells retained in vitro expression of markers (mucin MUC1 and HER2) or immunomodulatory factors (RAGE and IDO1). Increased Kyn secretion was associated with cancer endometrial cell culture in contrast to the control one. Conclusions: Primary endometrial cells express immune modulatory factors RAGE and IDO1 in vitro associated with cancer phenotype of endometrium.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Case-Control Studies
- Endometrial Neoplasms/genetics
- Endometrial Neoplasms/immunology
- Endometrial Neoplasms/metabolism
- Endometrial Neoplasms/pathology
- Endometrium/immunology
- Endometrium/metabolism
- Endometrium/pathology
- Female
- Follow-Up Studies
- Gene Expression Regulation, Neoplastic
- Humans
- Immunomodulation
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Kynurenine/metabolism
- Middle Aged
- Mitogen-Activated Protein Kinases/genetics
- Mitogen-Activated Protein Kinases/metabolism
- Primary Cell Culture
- Prognosis
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Joanna Tkaczuk-Włach
- Diagnostic Techniques Unit, Collegium Maximum, Medical University of Lublin, Staszica 4/6, 20-081 Lublin, Poland;
| | - Witold Kędzierski
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 12, 20-033 Lublin, Poland; (W.K.); (I.J.); (M.K.)
| | - Ilona Jonik
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 12, 20-033 Lublin, Poland; (W.K.); (I.J.); (M.K.)
| | - Ilona Sadok
- Laboratory of Separation and Spectroscopic Method Applications, Centre for Interdisciplinary Research, The John Paul II Catholic University of Lublin, Konstantynow 1J, 20-708 Lublin, Poland;
| | - Agata Filip
- Department of Cancer Genetics with Cytogenetic Laboratory, Medical University of Lublin, Radziwillowska 11, 20-080 Lublin, Poland;
| | - Marta Kankofer
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 12, 20-033 Lublin, Poland; (W.K.); (I.J.); (M.K.)
| | - Wojciech Polkowski
- Department of Surgical Oncology, Medical University of Lublin, Radziwillowska 13, 20-080 Lublin, Poland;
| | - Piotr Ziółkowski
- Department of Pathomorphology, Wroclaw Medical University, Marcinkowskiego 1, 50-368 Wroclaw, Poland;
| | - Andrzej Gamian
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114 Wroclaw, Poland;
| | - Magdalena Staniszewska
- Laboratory of Separation and Spectroscopic Method Applications, Centre for Interdisciplinary Research, The John Paul II Catholic University of Lublin, Konstantynow 1J, 20-708 Lublin, Poland;
- SDS Optic S.A., Centrum ECOTECH-COMPLEX, Block A, 20-612 Lublin, Poland
- Correspondence: or ; Tel.: +48-814-545-621
| |
Collapse
|
25
|
Puttabyatappa M, Ciarelli JN, Chatoff AG, Padmanabhan V. Developmental programming: Metabolic tissue-specific changes in endoplasmic reticulum stress, mitochondrial oxidative and telomere length status induced by prenatal testosterone excess in the female sheep. Mol Cell Endocrinol 2021; 526:111207. [PMID: 33607270 PMCID: PMC8005473 DOI: 10.1016/j.mce.2021.111207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023]
Abstract
Prenatal testosterone (T) excess-induced metabolic dysfunctions involve tissue specific changes in insulin sensitivity with insulin resistant, oxidative and lipotoxic state in liver/muscle and insulin sensitive but inflammatory and oxidative state in visceral adipose tissues (VAT). We hypothesized that mitochondrial dysfunction, endoplasmic reticulum (ER) stress and premature cellular senescence are contributors to the tissue-specific changes in insulin sensitivity. Markers of mitochondrial number, function, and oxidative phosphorylation (OxPhos), ER stress and cellular senescence (telomere length) were assessed in liver, muscle and 4 adipose (VAT, subcutaneous [SAT], epicardiac [ECAT] and perirenal [PRAT]) depots collected from control and prenatal T-treated female sheep at 21 months of age. Prenatal T treatment led to: (a) reduction in mitochondrial number and OxPhos complexes and increase in ER stress markers in muscle; (b) increase in fibrosis with trend towards increase in short telomere fragments in liver (c) depot-specific mitochondrial changes with OxPhos complexes namely increase in SAT and reduction in PRAT and increase in mitochondrial number in ECAT; (d) depot-specific ER stress marker changes with increase in VAT, reduction in SAT, contrasting changes in ECAT and no changes in PRAT; and (d) reduced shorter telomere fragments in SAT, ECAT and PRAT. These changes indicate insulin resistance may be driven by mitochondrial and ER dysfunction in muscle, fibrosis and premature senescence in liver, and depot-specific changes in mitochondrial function and ER stress without involving cellular senescence in adipose tissue. These findings provide mechanistic insights into pathophysiology of metabolic dysfunction among female offspring from hyperandrogenic pregnancies.
Collapse
Affiliation(s)
| | - Joseph N Ciarelli
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Adam G Chatoff
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
26
|
Bruni V, Capozzi A, Lello S. The Role of Genetics, Epigenetics and Lifestyle in Polycystic Ovary Syndrome Development: the State of the Art. Reprod Sci 2021; 29:668-679. [DOI: 10.1007/s43032-021-00515-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/21/2021] [Indexed: 12/11/2022]
|
27
|
Kooshki F, Tutunchi H, Vajdi M, Karimi A, Niazkar HR, Shoorei H, Pourghassem Gargari B. A Comprehensive insight into the effect of chromium supplementation on oxidative stress indices in diabetes mellitus: A systematic review. Clin Exp Pharmacol Physiol 2021; 48:291-309. [PMID: 33462845 DOI: 10.1111/1440-1681.13462] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 04/16/2020] [Accepted: 12/24/2020] [Indexed: 02/06/2023]
Abstract
Diabetes mellitus is a metabolic disorder defined as an increase in blood glucose levels (hyperglycaemia) and insufficient production or action of insulin produced by the pancreas. Chronic hyperglycaemia leads to increased reactive oxygen species (ROS) production and oxidative stress, which consequently results in insulin resistance, beta cell degeneration, dyslipidaemia, and glucose intolerance in diabetic patients. Chromium has an essential role in the metabolism of proteins, lipids, and carbohydrates through increasing insulin efficiency. This systematic review aimed to evaluate chromium supplementation's potential roles in oxidative stress indices in diabetes mellitus. A systematic search was performed in PubMed, Scopus, Google Scholar, Cochrane, and Science Direct databases until November 2020. All clinical trials and animal studies that assessed chromium's effect on oxidative stress indices in diabetes mellitus and were published in English-language journals were included. Finally, only 33 out of 633 articles met the required criteria for further analysis. Among 33 papers, 25 studies were performed on animals, and eight investigations were conducted on humans. Twenty-eight studies of chromium supplementation lead to reducing oxidative stress indices. Also, 23 studies showed that chromium supplementation markedly increased antioxidant enzymes' activity and improved levels of antioxidant indices. In conclusion, chromium supplementation decreased oxidative stress in diabetes mellitus. However, further clinical trials are suggested in a bid to determine the exact mechanisms.
Collapse
Affiliation(s)
- Fateme Kooshki
- Student Research Committee, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Helda Tutunchi
- Nutrition Research Center, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Vajdi
- Student Research Committee, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arash Karimi
- Student Research Committee, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Reza Niazkar
- Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Bahram Pourghassem Gargari
- Nutrition Research Center, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
28
|
Di Bari F, Catalano A, Bellone F, Martino G, Benvenga S. Vitamin D, Bone Metabolism, and Fracture Risk in Polycystic Ovary Syndrome. Metabolites 2021; 11:metabo11020116. [PMID: 33670644 PMCID: PMC7922814 DOI: 10.3390/metabo11020116] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 02/06/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among premenopausal women. PCOS may have reproductive, metabolic, cardiovascular, and psychological implications. Vitamin D deficit is often encountered in PCOS women and may contribute to the pathophysiology of this disorder. As of the key role of vitamin D in bone and mineral metabolism, and because the vitamin D status appears to be closely linked with the PCOS manifestations including insulin resistance, obesity, ovulatory and menstrual irregularities, oxidative stress and PTH elevation, hypovitaminosis D may directly and indirectly via the different facets of PCOS impair bone health in these women. Although limited data are available on life-long fracture risk in women with PCOS, the importance of preserving bone health in youth and adults to prevent osteoporosis and related fractures is also recognized in PCOS women. Evidence of the association between vitamin D and the clinical hallmarks of PCOS are summarized and discussed. Vitamin D arises as a cornerstone in women with PCOS and contributes to the pathophysiological link between PCOS and bone metabolism.
Collapse
Affiliation(s)
- Flavia Di Bari
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Viale Gazzi, 98125 Messina, Italy; (F.D.B.); (F.B.); (G.M.); (S.B.)
| | - Antonino Catalano
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Viale Gazzi, 98125 Messina, Italy; (F.D.B.); (F.B.); (G.M.); (S.B.)
- Correspondence: ; Tel.: +39-090-2213987
| | - Federica Bellone
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Viale Gazzi, 98125 Messina, Italy; (F.D.B.); (F.B.); (G.M.); (S.B.)
| | - Gabriella Martino
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Viale Gazzi, 98125 Messina, Italy; (F.D.B.); (F.B.); (G.M.); (S.B.)
| | - Salvatore Benvenga
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Viale Gazzi, 98125 Messina, Italy; (F.D.B.); (F.B.); (G.M.); (S.B.)
- Master Program on Childhood, Adolescent and Women’s Endocrine Health, University of Messina, Viale Gazzi, 98125 Messina, Italy
- Interdepartmental Program of Molecular & Clinical Endocrinology and Women’s Endocrine Health, University Hospital, A.O.U. Policlinico G. Martino, Viale Gazzi, 98125 Messina, Italy
| |
Collapse
|
29
|
Harada M, Takahashi N, Azhary JM, Kunitomi C, Fujii T, Osuga Y. Endoplasmic reticulum stress: a key regulator of the follicular microenvironment in the ovary. Mol Hum Reprod 2021; 27:gaaa088. [PMID: 33543293 DOI: 10.1093/molehr/gaaa088] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/04/2020] [Indexed: 12/17/2022] Open
Abstract
Intra-ovarian local factors regulate the follicular microenvironment in coordination with gonadotrophins, thus playing a crucial role in ovarian physiology as well as pathological states such as polycystic ovary syndrome (PCOS). One recently recognized local factor is endoplasmic reticulum (ER) stress, which involves the accumulation of unfolded or misfolded proteins in the ER related to various physiological and pathological conditions that increase the demand for protein folding or attenuate the protein-folding capacity of the organelle. ER stress results in activation of several signal transduction cascades, collectively termed the unfolded protein response (UPR), which affect a wide variety of cellular functions. Recent studies have revealed diverse roles of ER stress in physiological and pathological conditions in the ovary. In this review, we summarize the most current knowledge of the regulatory roles of ER stress in the ovary, in the context of reproduction. The physiological roles of ER stress and the UPR in the ovary remain largely undetermined. On the contrary, activation of ER stress is known to impair follicular and oocyte health in various pathological conditions; moreover, ER stress also contributes to the pathogenesis of several ovarian diseases, including PCOS. Finally, we discuss the potential of ER stress as a novel therapeutic target. Inhibition of ER stress or UPR activation, by treatment with existing chemical chaperones, lifestyle intervention, or the development of small molecules that target the UPR, represents a promising therapeutic strategy.
Collapse
Affiliation(s)
- Miyuki Harada
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Hongo, Bunkyo, Tokyo 113-8655, Japan
| | - Nozomi Takahashi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Hongo, Bunkyo, Tokyo 113-8655, Japan
| | - Jerilee Mk Azhary
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Hongo, Bunkyo, Tokyo 113-8655, Japan
| | - Chisato Kunitomi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Hongo, Bunkyo, Tokyo 113-8655, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Hongo, Bunkyo, Tokyo 113-8655, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Hongo, Bunkyo, Tokyo 113-8655, Japan
| |
Collapse
|
30
|
LNK promotes granulosa cell apoptosis in PCOS via negatively regulating insulin-stimulated AKT-FOXO3 pathway. Aging (Albany NY) 2021; 13:4617-4633. [PMID: 33495419 PMCID: PMC7906173 DOI: 10.18632/aging.202421] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS), which is often accompanied by insulin resistance, is closely related to increased apoptosis of ovarian granulosa cells. LNK is an important regulator of the insulin signaling pathway. When insulin binds to the receptor, the PI3K/AKT/FOXO signaling pathway is activated, and FOXO translocates from the nucleus to the cytoplasm, thereby inhibiting the expression of pro-apoptotic genes. METHODS Granulosa cells were collected from PCOS patients to investigate the relationship between LNK, cell apoptosis and insulin resistance. KGN cells underwent LNK overexpression/silence and insulin stimulation. The AKT/FOXO3 pathway was studied by western blot and immunofluorescence. LNK knockout mice were used to investigate the effect of LNK on the pathogenesis of PCOS. RESULTS The level of LNK was higher in PCOS group than control group. LNK was positively correlated with granulosa cell apoptosis and insulin resistance, and negatively correlated with oocyte maturation rate. LNK overexpression in KGN cells inhibited insulin-induced AKT/FOXO3 signaling pathway, causing nucleus translocation of FOXO3 and promoting granulosa cell apoptosis. LNK knockout partially restored estrous cycle and improved glucose metabolism in PCOS mice. CONCLUSIONS LNK was closely related to insulin resistance and apoptosis of granulosa cells via the AKT/FOXO3 pathway. LNK knockout partially restored estrous cycle and improved glucose metabolism in PCOS mice, suggesting LNK might become a potential biological target for the clinical treatment of PCOS.
Collapse
|
31
|
Jiang Q, Pan Y, Li P, Zheng Y, Bian Y, Wang W, Wu G, Song T, Shi Y. ANGPTL4 Expression in Ovarian Granulosa Cells Is Associated With Polycystic Ovary Syndrome. Front Endocrinol (Lausanne) 2021; 12:799833. [PMID: 35140683 PMCID: PMC8820586 DOI: 10.3389/fendo.2021.799833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES This study aims to characterize the expression of ANGPTL4 in ovarian granulosa cells (GCs) and its association with polycystic ovary syndrome (PCOS). METHODS This study included 104 PCOS patients and 112 women in control group undergoing in vitro fertilization-embryo transfer (IVF-ET) from the reproductive hospital affiliated with Shandong University from 2019 to 2021. By reverse transcription and real-time quantitative (RT-q) PCR, the mRNA expression of ANGPTL4 in GCs was assessed, and clinical information for these patients were then reviewed and analyzed. RESULTS The RT-qPCR results showed that ANGPTL4 expression in the control group was significantly lower than that in the PCOS group (p = 0.000) and had positive association with AMH (r = 0.211), HOMA-IR (r = 0.174), LDL/HDL (r = 0.176), ApoB/ApoAI (r = 0.155), and TC/HDL (r = 0.189). Additionally, the high expression of ANGPTL4 in the ovarian granulosa cells might be an independent predictor in PCOS (OR: 3.345; 95% CI: 1.951-5.734) with a close contact with incidence of PCOS (AUC: 0.704; 95% CI: 0.633-0.774, p < 0.001). CONCLUSIONS Our study revealed higher ANGPTL4 expression in ovarian GCs with PCOS. Its association with glucose and lipid metabolism showed that ANGPTL4 might play an important role in PCOS metabolism and pathogenesis.
Collapse
Affiliation(s)
- Qi Jiang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Ye Pan
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Ping Li
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Yanjun Zheng
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Yuehong Bian
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Wenqi Wang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Guihua Wu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tian Song
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Yuhua Shi
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
- *Correspondence: Yuhua Shi,
| |
Collapse
|
32
|
Papadakis G, Kandaraki EA, Garidou A, Koutsaki M, Papalou O, Diamanti-Kandarakis E, Peppa M. Tailoring treatment for PCOS phenotypes. Expert Rev Endocrinol Metab 2021; 16:9-18. [PMID: 33382003 DOI: 10.1080/17446651.2021.1865152] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/14/2020] [Indexed: 12/17/2022]
Abstract
Introduction: Polycystic ovary syndrome (PCOS) is one of the most common endocrinopathies in reproductive-aged women. Hyperandrogenism, polycystic ovaries, chronic anovulation, and metabolic aberrations are its common features. The treatment approach focuses on the main aberrations, which characterize the different phenotypes. Areas covered: Management strategies targeting the metabolic phenotype include lifestyle modifications for weight loss and improvement of dietary habits, as well as medication, such as insulin-sensitizers. The treatment of hyperandrogenic phenotype includes cosmetic procedures and the combined oral contraceptives with or without antiandrogens. The therapeutic approach to reproductive phenotype includes diet and lifestyle modifications, clomiphene citrate, and aromatase inhibitors. Alternative treatments include dietary supplements, herbs, resveratrol, myo-inositol, and acupuncture. Expert opinion: New studies have shown that higher anti-Müllerian hormone levels, gut microbiome composition, and plasma metabolomics are new parameters that are related to the most severe phenotypes. The clinical phenotypes can change over the lifespan with weight gain and can coexist in the same individual. Individualized treatment remains the main approach but grouping the phenotypes and following therapeutic recommendations may prove to be also clinically appropriate.
Collapse
Affiliation(s)
- Georgios Papadakis
- Endocrinology and Diabetes, STEPS Stoffwechselzentrum , Biel/Bienne, Switzerland
| | - Eleni A Kandaraki
- Endocrinology &Diabetes, European University Cyprus (EUC) , Nicosia, Cyprus
- Department of Endocrinology and Diabetes, HYGEIA Hospital Athens , Greece
| | - Anna Garidou
- Private Practice, Endocrinologist and Diabetologist , Chalandri, Athens, Greece
| | - Maria Koutsaki
- Private Practice, Endocrinologist and Diabetologist , Kesariani, Athens, Greece
| | - Olga Papalou
- Department of Endocrinology and Diabetes, HYGEIA Hospital Athens , Greece
| | | | - Melpomeni Peppa
- Endocrine Unit, 2nd Department of Internal Medicine Propaedeutic, Research Institute and Diabetes Center, National and Kapodistrian University of Athens, Attikon University Hospital , Greece
| |
Collapse
|
33
|
Ye W, Xie T, Song Y, Zhou L. The role of androgen and its related signals in PCOS. J Cell Mol Med 2020; 25:1825-1837. [PMID: 33369146 PMCID: PMC7882969 DOI: 10.1111/jcmm.16205] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 11/29/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women at reproductive age. However, the underlying pathogenic mechanisms have not been completely understood. Hyperandrogenism is an important clinic feature in patients with PCOS, suggesting its pathologic role in the development and progression of PCOS. However, the actual role of androgen and the related signals in PCOS and PCOS-related complications have not yet been clarified. In this review, we surveyed the origin and effects of androgen on PCOS and the related complications, highlighted the cellular signals affecting androgen synthesis and summarized the pathological processes caused by hyperandrogenism. Our review well reveals the important mechanisms referring the pathogenesis of PCOS and provides important clues to the clinic strategies in patients with PCOS.
Collapse
Affiliation(s)
- Wenting Ye
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tingting Xie
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yali Song
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| |
Collapse
|
34
|
Karmash OI, Liuta MY, Korobov AM, Sybirna NO. Effect of Photomodulation Therapy on Development of Oxidative Stress in Blood Leukocytes of Rats with Streptozocin-Induced Diabetes Mellitus. CYTOL GENET+ 2020. [DOI: 10.3103/s0095452720050114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
35
|
Takahashi N, Harada M, Azhary JMK, Kunitomi C, Nose E, Terao H, Koike H, Wada-Hiraike O, Hirata T, Hirota Y, Koga K, Fujii T, Osuga Y. Accumulation of advanced glycation end products in follicles is associated with poor oocyte developmental competence. Mol Hum Reprod 2020; 25:684-694. [PMID: 31504800 DOI: 10.1093/molehr/gaz050] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/14/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022] Open
Abstract
Advanced glycation end products (AGEs) affect the follicular microenvironment. The close relationship between AGEs, proinflammatory cytokine production and activation of the unfolded protein response (UPR), which involves activating transcription factor 4 (ATF4), is crucial for regulation of various cellular functions. We examined whether accumulation of AGEs in follicles was associated with proinflammatory cytokine production and activation of the UPR in granulosa cells and decreased oocyte developmental competence. Concentrations of AGEs, soluble receptor for AGE (sRAGE), interleukin (IL)-6 and IL-8 in follicular fluid (FF) were examined by ELISAs in 50 follicles. mRNA expression of ATF4, IL-6 and IL-8 in cumulus cells (CCs) were examined by quantitative RT-PCR in 77 samples. Cultured human granulosa-lutein cells (GLCs) were treated with AGE-bovine serum albumin (BSA) alone or following transfection of ATF4-targeting small interfering RNA. The AGE concentration and the AGE/sRAGE ratio in FF were significantly higher in follicles containing oocytes that developed into poor-morphology embryos (group I) than those with good-morphology embryos (group II). When compared with sibling follicles from the same patients, the AGE/sRAGE and concentrations of IL-6 and IL-8 in FF, as well as ATF4, IL-6 and IL-8 mRNA expression in CCs, were significantly higher in group I follicles than group II. AGE treatment increased mRNA expression of ATF4, IL-6 and IL-8 in cultured GLCs. Knockdown of ATF4 abrogated the stimulatory effects of AGE on mRNA expression and protein secretion of IL-6 and IL-8. Our findings support the idea that accumulation of AGEs in follicles reduces oocyte competence by triggering inflammation via activation of ATF4 in the follicular microenvironment.
Collapse
Affiliation(s)
- Nozomi Takahashi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Miyuki Harada
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Jerilee M K Azhary
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Chisato Kunitomi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Emi Nose
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Hiromi Terao
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Hiroshi Koike
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Osamu Wada-Hiraike
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Tetsuya Hirata
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Kaori Koga
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| |
Collapse
|
36
|
Thornton K, Merhi Z, Jindal S, Goldsammler M, Charron MJ, Buyuk E. Dietary Advanced Glycation End Products (AGEs) could alter ovarian function in mice. Mol Cell Endocrinol 2020; 510:110826. [PMID: 32339649 DOI: 10.1016/j.mce.2020.110826] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/20/2020] [Accepted: 04/20/2020] [Indexed: 12/17/2022]
Abstract
Nutrition is an important source of exogenous AGEs and thermally processed foods present in western-style diets contain a large amount of these pro-inflammatory AGEs. Additionally, the intake of dietary AGEs could upregulate ovarian gene expression of inflammatory macrophage markers. The objective of this study was to investigate the effect of diet rich in AGEs on estrous cyclicity and ovarian function in a mouse model. Six-week old C57BL/6 J female mice were randomly subjected to either a diet low in AGEs (L-AGE) or a diet high in AGEs (H-AGE) for a total of 13 weeks. Experiments performed included daily vaginal smears to assess estrous cyclicity, ovarian superovulation with gonadotropins to assess the number of oocytes released, whole ovarian tissue mRNA quantification by RT-PCR to quantify genes involved in folliculogenesis, steroidogenesis, and macrophage markers, and ovarian morphology for follicle count. Outcome measures included estrous cyclicity, number of oocytes following superovulation, expression of genes involved in folliculogenesis, steroidogenesis, and macrophage infiltration as well as the number of primordial, primary, secondary, antral follicles and corpora lutea. Compared to mice on L-AGE diet, mice on H-AGE spent significantly longer time in the diestrus phase, had similar number of oocytes released following ovarian superovulation, and showed significant alterations in genes involved in steroidogenesis (increase in Star mRNA expression levels) and folliculogenesis (increase in Gdf-9 and Fshr mRNA expression levels). Mouse macrophage marker F4/80 mRNA expression was upregulated in mice on H-AGE diet compared to mice on L-AGE diet. Finally, mice on H-AGE diet had significantly fewer corpora lutea in their ovaries. These results indicate that the ingestion of high amounts of dietary AGEs could disrupt folliculogenesis and steroidogenesis that might lead to abnormal estrous cyclicity. Intake of dietary AGEs could also upregulate ovarian gene expression of inflammatory macrophage markers.
Collapse
Affiliation(s)
- Kimberly Thornton
- Montefiore's Institute for Reproductive Medicine and Health, Hartsdale, NY, 10530, USA; Department of Obstetrics & Gynecology and Women's Health, Division of Reproductive Endocrinology and Infertility, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Currently at Reproductive Medicine Associates of New York, Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY, 10022, USA
| | - Zaher Merhi
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, SUNY Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Sangita Jindal
- Montefiore's Institute for Reproductive Medicine and Health, Hartsdale, NY, 10530, USA; Department of Obstetrics & Gynecology and Women's Health, Division of Reproductive Endocrinology and Infertility, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Michelle Goldsammler
- Montefiore's Institute for Reproductive Medicine and Health, Hartsdale, NY, 10530, USA; Department of Obstetrics & Gynecology and Women's Health, Division of Reproductive Endocrinology and Infertility, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Maureen J Charron
- Department of Obstetrics & Gynecology and Women's Health, Division of Reproductive Endocrinology and Infertility, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Erkan Buyuk
- Montefiore's Institute for Reproductive Medicine and Health, Hartsdale, NY, 10530, USA; Department of Obstetrics & Gynecology and Women's Health, Division of Reproductive Endocrinology and Infertility, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Currently at Reproductive Medicine Associates of New York, Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY, 10022, USA.
| |
Collapse
|
37
|
Azhary JMK, Harada M, Kunitomi C, Kusamoto A, Takahashi N, Nose E, Oi N, Wada-Hiraike O, Urata Y, Hirata T, Hirota Y, Koga K, Fujii T, Osuga Y. Androgens Increase Accumulation of Advanced Glycation End Products in Granulosa Cells by Activating ER Stress in PCOS. Endocrinology 2020; 161:5724441. [PMID: 32020188 DOI: 10.1210/endocr/bqaa015] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 02/04/2020] [Indexed: 12/15/2022]
Abstract
Polycystic ovary syndrome (PCOS) is associated with hyperandrogenism, and we previously found that androgens activate endoplasmic reticulum (ER) stress in granulosa cells from patients with PCOS. In addition, recent studies demonstrated the accumulation of advanced glycation end products (AGEs) in granulosa cells from PCOS patients, which contribute to the pathology. Therefore, we hypothesized that androgens upregulate the receptor for AGEs (RAGE) expression in granulosa cells by activating ER stress, thereby increasing the accumulation of AGEs in these cells and contributing to the pathology. In the present study, we show that testosterone increases RAGE expression and AGE accumulation in cultured human granulosa-lutein cells (GLCs), and this is reduced by pretreatment with tauroursodeoxycholic acid (TUDCA), an ER stress inhibitor in clinical use. Knockdown of the transcription factor C/EBP homologous protein (CHOP), an unfolded protein response factor activated by ER stress, inhibits testosterone-induced RAGE expression and AGE accumulation. The expression of RAGE and the accumulation of AGEs are upregulated in granulosa cells from PCOS patients and dehydroepiandrosterone-induced PCOS mice. Administration of the RAGE inhibitor FPS-ZM1 or TUDCA to PCOS mice reduces RAGE expression and AGE accumulation in granulosa cells, improves their estrous cycle, and reduces the number of atretic antral follicles. In summary, our findings indicate that hyperandrogenism in PCOS increases the expression of RAGE and accumulation of AGEs in the ovary by activating ER stress, and that targeting the AGE-RAGE system, either by using a RAGE inhibitor or a clinically available ER stress inhibitor, may represent a novel approach to PCOS therapy.
Collapse
Affiliation(s)
- Jerilee M K Azhary
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Miyuki Harada
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Chisato Kunitomi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Akari Kusamoto
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Nozomi Takahashi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Emi Nose
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Nagisa Oi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Osamu Wada-Hiraike
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Yoko Urata
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Tetsuya Hirata
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Kaori Koga
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| |
Collapse
|
38
|
Gill V, Kumar V, Singh K, Kumar A, Kim JJ. Advanced Glycation End Products (AGEs) May Be a Striking Link Between Modern Diet and Health. Biomolecules 2019; 9:biom9120888. [PMID: 31861217 PMCID: PMC6995512 DOI: 10.3390/biom9120888] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/30/2019] [Accepted: 12/02/2019] [Indexed: 12/12/2022] Open
Abstract
The Maillard reaction is a simple but ubiquitous reaction that occurs both in vivo and ex vivo during the cooking or processing of foods under high-temperature conditions, such as baking, frying, or grilling. Glycation of proteins is a post-translational modification that forms temporary adducts, which, on further crosslinking and rearrangement, form permanent residues known as advanced glycation end products (AGEs). Cooking at high temperature results in various food products having high levels of AGEs. This review underlines the basis of AGE formation and their corresponding deleterious effects on the body. Glycated Maillard products have a direct association with the pathophysiology of some metabolic diseases, such as diabetes mellitus type 2 (DM2), acute renal failure (ARF), Alzheimer’s disease, dental health, allergies, and polycystic ovary syndrome (PCOS). The most glycated and structurally abundant protein is collagen, which acts as a marker for diabetes and aging, where decreased levels indicate reduced skin elasticity. In diabetes, high levels of AGEs are associated with carotid thickening, ischemic heart disease, uremic cardiomyopathy, and kidney failure. AGEs also mimic hormones or regulate/modify their receptor mechanisms at the DNA level. In women, a high AGE diet directly correlates with high levels of androgens, anti-Müllerian hormone, insulin, and androstenedione, promoting ovarian dysfunction and/or infertility. Vitamin D3 is well-associated with the pathogenesis of PCOS and modulates steroidogenesis. It also exhibits a protective mechanism against the harmful effects of AGEs. This review elucidates and summarizes the processing of infant formula milk and the associated health hazards. Formulated according to the nutritional requirements of the newborn as a substitute for mother’s milk, formula milk is a rich source of primary adducts, such as carboxy-methyl lysine, which render an infant prone to inflammation, dementia, food allergies, and other diseases. We therefore recommend that understanding this post-translational modification is the key to unlocking the mechanisms and physiology of various metabolic syndromes.
Collapse
Affiliation(s)
- Vidhu Gill
- Central Research Station, Subharti Medical College, Swami Vivekanand Subharti University, Meerut 250002, India; (V.G.); (K.S.)
| | - Vijay Kumar
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea
- Correspondence: (V.K.); (J.-J.K.); Tel.: +82-53-810-3027 or +82-10-9668-3464 (J.-J.K.); Fax: +82-53-801-3027 (J.-J.K.)
| | - Kritanjali Singh
- Central Research Station, Subharti Medical College, Swami Vivekanand Subharti University, Meerut 250002, India; (V.G.); (K.S.)
| | - Ashok Kumar
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow 226014, India;
| | - Jong-Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea
- Correspondence: (V.K.); (J.-J.K.); Tel.: +82-53-810-3027 or +82-10-9668-3464 (J.-J.K.); Fax: +82-53-801-3027 (J.-J.K.)
| |
Collapse
|
39
|
Wang J, Wu D, Guo H, Li M. Hyperandrogenemia and insulin resistance: The chief culprit of polycystic ovary syndrome. Life Sci 2019; 236:116940. [PMID: 31604107 DOI: 10.1016/j.lfs.2019.116940] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/23/2019] [Accepted: 10/06/2019] [Indexed: 02/07/2023]
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common systemic reproductive endocrine diseases, which has a variety of effects on a woman's health. Because of the involvement of multiple pathways and the lack of common clues, PCOS demonstrates multifactorial properties and heterogeneity of symptoms. Recent studies have demonstrated that the core etiology and primary endocrine characteristics of PCOS are hyperandrogenemia (HA) and insulin resistance (IR). HA and IR are the main causes of PCOS and they can interplay each other in the occurrence and development of PCOS. Just because of this, the study about the effects of HA and IR on pathophysiology of various related symptoms of PCOS is very important to understand the pathogenesis of PCOS. This paper reviews the main symptoms of PCOS, including neuroendocrine disorders, reproductive processes, dyslipidemia, obesity, hypertension, nonalcoholic fatty liver disease (NAFLD), and sleep disordered breathing, which seriously affect the physical and mental health of PCOS women. The increasing knowledge of the development pattern of HA and IR in PCOS suggests that changes in diet and lifestyle, and the discovery of potential therapeutic agents may improve PCOS. However, further studies are needed to clarify the mutual influence and relation of HA and IR in development of PCOS. This review provides an overview of the current knowledge about the effects of HA and IR on PCOS.
Collapse
Affiliation(s)
- Juan Wang
- Department of Histology and Embryology, University of South China, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang, 421001, Hunan, China
| | - Daichao Wu
- Department of Histology and Embryology, University of South China, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang, 421001, Hunan, China
| | - Hui Guo
- Department of Histology and Embryology, University of South China, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang, 421001, Hunan, China
| | - Meixiang Li
- Department of Histology and Embryology, University of South China, Institute of Clinical Anatomy & Reproductive Medicine, Hengyang, 421001, Hunan, China.
| |
Collapse
|
40
|
Asadipooya K, Uy EM. Advanced Glycation End Products (AGEs), Receptor for AGEs, Diabetes, and Bone: Review of the Literature. J Endocr Soc 2019; 3:1799-1818. [PMID: 31528827 PMCID: PMC6734192 DOI: 10.1210/js.2019-00160] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/03/2019] [Indexed: 12/24/2022] Open
Abstract
Diabetes compromises bone cell metabolism and function, resulting in increased risk of fragility fracture. Advanced glycation end products (AGEs) interact with the receptor for AGEs (RAGE) and can make a meaningful contribution to bone cell metabolism and/or alter function. Searches in PubMed using the key words "advanced glycation end-product," "RAGE," "sRAGE," "bone," and "diabetes" were made to explain some of the clinical outcomes of diabetes in bone metabolism through the AGE-RAGE signaling pathway. All published clinical studies were included in tables. The AGE-RAGE signaling pathway participates in diabetic complications, including diabetic osteopathy. Some clinical results in diabetic patients, such as reduced bone density, suppressed bone turnover markers, and bone quality impairment, could be potentially due to AGE-RAGE signaling consequences. However, the AGE-RAGE signaling pathway has some helpful roles in the bone, including an increase in osteogenic function. Soluble RAGE (sRAGE), as a ligand decoy, may increase in either conditions of RAGE production or destruction, and then it cannot always reflect the AGE-RAGE signaling. Recombinant sRAGE can block the AGE-RAGE signaling pathway but is associated with some limitations, such as accessibility to AGEs, an increase in other RAGE ligands, and a long half-life (24 hours), which is associated with losing the beneficial effect of AGE/RAGE. As a result, sRAGE is not a helpful marker to assess activity of the RAGE signaling pathway. The recombinant sRAGE cannot be translated into clinical practice due to its limitations.
Collapse
Affiliation(s)
- Kamyar Asadipooya
- Division of Endocrinology and Molecular Medicine, Department of Medicine, University of Kentucky, Lexington, Kentucky
| | - Edilfavia Mae Uy
- Division of Endocrinology and Molecular Medicine, Department of Medicine, University of Kentucky, Lexington, Kentucky
| |
Collapse
|