1
|
Mahabamunuge J, Sekula NM, Lepore C, Kudrimoti M, Upadhyay A, Alshowaikh K, Li HJ, Seifer DB, AlAshqar A. The Molecular Basis of Polycystic Ovary Syndrome and Its Cardiometabolic Correlates: Exploring the Intersection and Its Clinical Implications-A Narrative Review. Biomedicines 2025; 13:709. [PMID: 40149685 PMCID: PMC11940587 DOI: 10.3390/biomedicines13030709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/04/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Recent studies have highlighted the association between polycystic ovary syndrome (PCOS) and cardiometabolic diseases, leading to an improved understanding of the underlying mechanistic factors. PCOS significantly increases cardiovascular risk by predisposing individuals to various subclinical and clinical conditions, including atherosclerosis and type 2 diabetes mellitus. Additionally, it interacts synergistically with other traditional cardiovascular risk factors, such as obesity, hyperlipidemia, and insulin resistance. Several molecular mechanisms involving genetics, epigenetics, adipokine secretion, hyperandrogenemia, and hyperinsulinemia play a role in the relationship between PCOS and these comorbidities. For instance, androgen excess has been implicated in the development of hypertension, type 2 diabetes mellitus, endothelial dysfunction, and ultimately, broader cardiovascular disease. A deeper understanding of these underlying mechanisms facilitates the development of diagnostic, preventative, and therapeutic strategies directed at reducing cardiometabolic morbidity. This narrative review summarizes the current evidence, explores the potential clinical implications of these findings, and discusses emerging therapies to reduce cardiometabolic morbidity in women with PCOS.
Collapse
Affiliation(s)
- Jasmin Mahabamunuge
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; (J.M.); (N.M.S.); (C.L.); (M.K.); (A.U.); (K.A.)
| | - Nicole M. Sekula
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; (J.M.); (N.M.S.); (C.L.); (M.K.); (A.U.); (K.A.)
| | - Christina Lepore
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; (J.M.); (N.M.S.); (C.L.); (M.K.); (A.U.); (K.A.)
| | - Meghana Kudrimoti
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; (J.M.); (N.M.S.); (C.L.); (M.K.); (A.U.); (K.A.)
| | - Animesh Upadhyay
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; (J.M.); (N.M.S.); (C.L.); (M.K.); (A.U.); (K.A.)
| | - Khadija Alshowaikh
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; (J.M.); (N.M.S.); (C.L.); (M.K.); (A.U.); (K.A.)
| | - Howard J. Li
- Division of Reproductive Endocrinology and Infertility, National Institutes of Health, Bethesda, MD 20892, USA;
| | - David B. Seifer
- Division of Reproductive Endocrinology and Infertility, Yale School of Medicine, New Haven, CT 06510, USA;
| | - Abdelrahman AlAshqar
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; (J.M.); (N.M.S.); (C.L.); (M.K.); (A.U.); (K.A.)
| |
Collapse
|
2
|
Tinano FR, Machado IFR, Latronico AC, Gomes LG. Shared Pathophysiological Mechanisms and Genetic Factors in Early Menarche and Polycystic Ovary Syndrome. J Neurosci 2025; 45:e1681242024. [PMID: 40074331 PMCID: PMC11905354 DOI: 10.1523/jneurosci.1681-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 03/14/2025] Open
Abstract
Early age at menarche (early AAM) and polycystic ovary syndrome (PCOS) are reproductive and metabolic disorders with overlapping pathophysiological and genetic features. Epidemiological studies suggest a link between these two conditions, both of which are characterized by dysregulation of the neuroendocrine pathways that control pulsatile gonadotropin-releasing hormone secretion, thus affecting gonadotropin release, particularly luteinizing hormone secretion. A common pathophysiology involving positive energy balance and abnormal metabolic status is evident in both disorders. Genetic and epigenetic factors influence the onset of puberty and reproductive outcomes. Genome-wide association studies have identified common genetic variants associated with AAM and PCOS, particularly in genes related to the neuroendocrine axis (e.g., FSHB) and obesity (e.g., FTO). In addition, high-throughput sequencing has revealed rare loss-of-function variants in the DLK1 gene in women with central precocious puberty (CPP), early menarche, and PCOS, who experienced adverse metabolic outcomes in adulthood. This review explores the shared pathophysiological mechanisms between CPP/early AAM and PCOS, examines potential genetic and epigenetic factors that may link these neuroendocrine reproductive conditions, and offers insights into future research and treatment strategies. Understanding these connections may provide new targets for therapeutic interventions and improve outcomes for individuals with these reproductive disorders.
Collapse
Affiliation(s)
- Flavia Rezende Tinano
- Discipline of Endocrinology & Metabolism, Department of Internal Medicine, University of Sao Paulo Medical School, University of Sao Paulo, Sao Paulo, Sao Paulo 01246 903, Brazil
| | - Iza Franklin Roza Machado
- Discipline of Endocrinology & Metabolism, Department of Internal Medicine, University of Sao Paulo Medical School, University of Sao Paulo, Sao Paulo, Sao Paulo 01246 903, Brazil
| | - Ana Claudia Latronico
- Discipline of Endocrinology & Metabolism, Department of Internal Medicine, University of Sao Paulo Medical School, University of Sao Paulo, Sao Paulo, Sao Paulo 01246 903, Brazil
| | - Larissa Garcia Gomes
- Discipline of Endocrinology & Metabolism, Department of Internal Medicine, University of Sao Paulo Medical School, University of Sao Paulo, Sao Paulo, Sao Paulo 01246 903, Brazil
| |
Collapse
|
3
|
Abdul-Ameer F, AlAsadi IJA, Hosseini A, Bahreini E. The relationship between serum CTRP-5, C3a/desArg, and complement-C3 levels and hypothyroidism in women with polycystic ovary syndrome. BMC Endocr Disord 2024; 24:272. [PMID: 39696270 DOI: 10.1186/s12902-024-01801-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
INTRODUCTION Many patients with polycystic ovary syndrome (PCOS) also experience thyroid disorders. There is a notable similarity in energy metabolism among PCOS, C1q/tumor necrosis factor (TNF)-related proteins (CTRP-5)deficiency, C3a/desArg (also known as acylation-stimulating protein (ASP)) deficiency, and hypothyroidism. This study aimed to investigate the relationship between serum levels of these factors and hypothyroidism in patients with PCOS. Improved clarity and vocabulary, corrected minor grammatical issues, and enhanced readability. METHODS This case-control study involved three groups: healthy women (control group), women with PCOS and hypothyroidism, and women with PCOS without hypothyroidism. Serum levels of FBS, total cholesterol, triglycerides, and HDL-C were measured using enzymatic and colorimetric methods. TSH, T4, T3, and anti-thyroid peroxidase (Anti-TPO) levels were determined by ELISA to screen for hypothyroidism in women with PCOS. Additionally, serum levels of luteinizing hormone (LH), follicle-stimulating hormone (FSH), CTRP-5, ASP, and complement C3 were assessed using the ELISA method. RESULTS The results indicated that reduced blood levels of CTRP-5, along with elevated levels of ASP (C3a/desArg) and complement C3 in patients with PCOS, may be linked to dysregulation of the thyroid gland. Furthermore, the study observed that changes in these parameters, in conjunction with thyroid dysfunction, are associated with pathological alterations in lipid profiles and blood glucose levels. CONCLUSION While changes in CTRP-5, ASP, and complement C3 can influence energy expenditure and storage in PCOS and thyroid function, the complex nature of PCOS requires further research to investigate the prevalence of hypothyroidism in individuals with PCOS. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Fatima Abdul-Ameer
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614525, Tehran, Iran
- College of Medicine, University of Karbala, Karbala, Iraq
| | | | - Asieh Hosseini
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Bahreini
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614525, Tehran, Iran.
| |
Collapse
|
4
|
Pich K, Pietroń K, Szlaga A, Billert M, Skrzypski M, Pawlicki P, Kotula-Balak M, Dupont J, Błasiak A, Rak A. Adipokines level in plasma, hypothalamus, ovaries and adipose tissue of rats with polycystic ovary syndrome. Reprod Biomed Online 2024:104693. [PMID: 40199655 DOI: 10.1016/j.rbmo.2024.104693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 04/10/2025]
Abstract
RESEARCH QUESTION Do the levels of adipokines (adiponectin, apelin, chemerin and vaspin) in plasma, hypothalamus, ovaries and periovarian adipose tissue differ during polycystic ovarian syndrome (PCOS)? DESIGN The PCOS was induced in rats by oral administration of non-steroidal aromatase inhibitor letrozole. To determine the plasma levels of adiponectin, apelin, chemerin and vaspin enzyme-linked immunosorbent assays were carried out. To assess the expression (gene and protein) and immunolocalization of these adipokines and their receptors, namely Adipor1 and Adipor2 for adiponectin, Aplnr for apelin, Ccrl2, Cmklr1 and Gpr1 for chemerin and Grp78 for vaspin in the hypothalamus, real-time polymerase chain reaction, and Western blot and immunohistochemistry were used to analyse ovaries and periovarian adipose tissue respectively. RESULTS In PCOS, the plasma level of adiponectin decreased (P = 0.0003), whereas apelin, chemerin and vaspin increased (P ≤ 0.0479). Moreover, PCOS modulates the expression of adipokines and their receptors in the hypothalamus, ovaries and periovarian adipose tissue compared with healthy rats (P ≤ 0.487). CONCLUSIONS A strong relationship was found between PCOS and adipokines, which suggests that adipokines may be a biomarker of PCOS.
Collapse
Affiliation(s)
- Karolina Pich
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Kraków, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Poland
| | - Klaudia Pietroń
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Kraków, Poland
| | - Agata Szlaga
- Departament of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Poland
| | - Maria Billert
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, Poznań, Poland
| | - Marek Skrzypski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, Poznań, Poland
| | - Piotr Pawlicki
- Center of Experimental and Innovative Medicine, University of Agriculture in Kraków, Kraków, Poland
| | - Małgorzata Kotula-Balak
- Department of Animal Anatomy and Preclinical Sciences, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Kraków, Kraków Poland
| | - Joëlle Dupont
- INRAE, Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Anna Błasiak
- Departament of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Poland
| | - Agnieszka Rak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Kraków, Poland.
| |
Collapse
|
5
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
6
|
Moore T. X centromeric drive may explain the prevalence of polycystic ovary syndrome and other conditions: Genomic structure of the human X chromosome pericentromeric region is consistent with meiotic drive associated with PCOS and other conditions. Bioessays 2024; 46:e2400056. [PMID: 39072829 DOI: 10.1002/bies.202400056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/30/2024]
Abstract
X chromosome centromeric drive may explain the prevalence of polycystic ovary syndrome and contribute to oocyte aneuploidy, menopause, and other conditions. The mammalian X chromosome may be vulnerable to meiotic drive because of X inactivation in the female germline. The human X pericentromeric region contains genes potentially involved in meiotic mechanisms, including multiple SPIN1 and ZXDC paralogs. This is consistent with a multigenic drive system comprising differential modification of the active and inactive X chromosome centromeres in female primordial germ cells and preferential segregation of the previously inactivated X chromosome centromere to the polar body at meiosis I. The drive mechanism may explain differences in X chromosome regulation in the female germlines of the human and mouse and, based on the functions encoded by the genes in the region, the transmission of X pericentromeric genetic or epigenetic variants to progeny could contribute to preeclampsia, autism, and differences in sexual differentiation.
Collapse
Affiliation(s)
- Tom Moore
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| |
Collapse
|
7
|
Khademi Z, Pourreza S, Hamedi-Shahraki S, Amirkhizi F. Association Between Selenium and Circulating Adipokine Levels in Patients with Polycystic Ovary Syndrome. Biol Trace Elem Res 2024; 202:3442-3448. [PMID: 37910262 DOI: 10.1007/s12011-023-03935-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/24/2023] [Indexed: 11/03/2023]
Abstract
There is increasing evidence that selenium (Se) and its major transport protein, selenoprotein-P (SePP), may be associated with polycystic ovary syndrome (PCOS). However, the association of serum Se and SePP levels with circulating adipokines in this population has not received sufficient attention. In the present study, we aimed to investigate the associations of serum Se and SePP with circulating adipokine levels in patients with PCOS. In this cross-sectional study, 115 patients aged 18-45 years with PCOS diagnosed according to the Rotterdam Consensus Criteria were recruited. The general characteristics of the participants were collected using a general questionnaire and anthropometric measurements were taken. Blood samples were obtained and serum levels of leptin, adiponectin, visfatin, resistin, and omentin-1, as well as markers of glucose metabolism, were measured. Serum levels of Se and SePP were inversely correlated with fasting blood glucose (FBS), serum insulin, and homeostatic model assessment for insulin resistance (HOMA-IR). In addition, serum levels of Se and SePP were positively correlated with serum levels of adiponectin and visfatin. Although there was no significant correlation between serum Se and serum omentin-1 levels, a significant positive correlation was found between serum SePP levels and this adipokine. The present study found that serum Se and SePP levels were positively correlated with serum adiponectin and visfatin levels. Further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Zeinab Khademi
- Department of Public Health, Sirjan School of Medical Sciences, Sirjan, Iran
| | - Sanaz Pourreza
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Soudabeh Hamedi-Shahraki
- Department of Epidemiology and Biostatistics, Faculty of Public Health, Zabol University of Medical Sciences, Zabol, Iran
| | - Farshad Amirkhizi
- Department of Nutrition, Faculty of Public Health, Zabol University of Medical Sciences, Zabol, Iran.
| |
Collapse
|
8
|
Zhuang J, Wang Y, Wang S, Hu R, Wu Y. Association between visceral adiposity index and infertility in reproductive-aged women in the United States. Sci Rep 2024; 14:14230. [PMID: 38902300 PMCID: PMC11189895 DOI: 10.1038/s41598-024-64849-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 06/13/2024] [Indexed: 06/22/2024] Open
Abstract
Over the years, obesity has become more commonplace and has had a substantial impact on several medical specialties, including reproductive medicine. The potential correlation between the visceral adiposity index (VAI) and infertility has yet to be determined. Women between the ages of 18 and 45 were included in this cross-sectional study, which was conducted as part of the National Health and Nutrition Examination Survey (NHANES) between 2015 and 2020. Three tertiles were used to group VAI levels. Subgroup analysis and weighted binary logistic regression were employed to investigate the independent relationship between VAI and infertility. Smooth curve fitting was used to explore nonlinear relationships. This cross-sectional study followed the criteria of the STROBE guidelines. Of the 1231 participants, 127 were infertile women aged 18-45 years. A higher VAI was associated with a higher prevalence of infertility (OR = 1.22, 95% CI:1.03-1.45), which remained consistent across all subgroups (p > 0.05 for all interactions). We demonstrated a positive nonlinear association between VAI and infertility using a smooth curve fit. A higher visceral adiposity index level is positively correlated with a higher incidence of infertility among women in the United States. Women who are infertile can be identified using the visceral obesity index, and controlling visceral obesity may help lower the chances of becoming infertile.
Collapse
Affiliation(s)
- Jiaru Zhuang
- Human Reproductive Medicine Center, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214026, Jiangsu, People's Republic of China
- Department of Laboratory Medicine, Jiangnan University Medical Center, 68 Zhongshan Road, Wuxi, 214000, Jiangsu, People's Republic of China
| | - Yuan Wang
- Human Reproductive Medicine Center, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214026, Jiangsu, People's Republic of China
| | - Shan Wang
- Human Reproductive Medicine Center, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214026, Jiangsu, People's Republic of China
| | - Renjing Hu
- Department of Laboratory Medicine, Jiangnan University Medical Center, 68 Zhongshan Road, Wuxi, 214000, Jiangsu, People's Republic of China.
| | - Yibo Wu
- Human Reproductive Medicine Center, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214026, Jiangsu, People's Republic of China.
| |
Collapse
|
9
|
Tan H, Long P, Xiao H. Dissecting the shared genetic architecture between endometriosis and polycystic ovary syndrome. Front Endocrinol (Lausanne) 2024; 15:1359236. [PMID: 38742190 PMCID: PMC11089172 DOI: 10.3389/fendo.2024.1359236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Background Previous study suggested evidence for coexistence and similarities between endometriosis and polycystic ovary syndrome (PCOS), but it is unclear regarding the shared genetic architecture and causality underlying the phenotypic similarities observed for endometriosis and PCOS. Methods By leveraging summary statistics from public genome-wide association studies regarding endometriosis (European-based: N=470,866) and PCOS (European-based: N=210,870), we explored the genetic correlation that shared between endometriosis and PCOS using linkage disequilibrium score regression. Shared risk SNPs were derived using PLACO (Pleiotropic analysis under composite null hypothesis) and FUMA (Functional Mapping and Annotation of Genetic Associations). The potential causal association between endometriosis and PCOS was investigated using two-sample Mendelian randomization (MR). Linkage disequilibrium score for the specific expression of genes analysis (LDSC-SEG) were performed for tissue enrichment analysis. The expression profiles of the risk gene in tissues were further examined. Results A positive genetic association was observed between endometriosis and PCOS. 12 significant pleiotropic loci shared between endometriosis and PCOS were identified. Genetic associations between endometriosis and PCOS were particularly enriched in uterus, endometrium and fallopian tube. Two-sample MR analysis further indicated a potential causative effect of endometriosis on PCOS, and vice versa. Microarray and RNA-seq verified the expressions of SYNE1 and DNM3 were significantly altered in the endometrium of patients with endometriosis or PCOS compared to those of control subjects. Conclusion Our study indicates the genetic correlation and shared risk genes between PCOS and endometriosis. These findings provide insights into the potential mechanisms behind their comorbidity and the future development of therapeutics.
Collapse
Affiliation(s)
- Hangjing Tan
- Institute of Reproductive & Stem Cell Engineering, Center of Reproductive Health, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Panpan Long
- Institute of Reproductive & Stem Cell Engineering, Center of Reproductive Health, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Center of Genetics, Changsha Jiangwan Maternity Hospital, Changsha, Hunan, China
| | - Hongmei Xiao
- Institute of Reproductive & Stem Cell Engineering, Center of Reproductive Health, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| |
Collapse
|
10
|
Stener-Victorin E, Teede H, Norman RJ, Legro R, Goodarzi MO, Dokras A, Laven J, Hoeger K, Piltonen TT. Polycystic ovary syndrome. Nat Rev Dis Primers 2024; 10:27. [PMID: 38637590 DOI: 10.1038/s41572-024-00511-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/20/2024]
Abstract
Despite affecting ~11-13% of women globally, polycystic ovary syndrome (PCOS) is a substantially understudied condition. PCOS, possibly extending to men's health, imposes a considerable health and economic burden worldwide. Diagnosis in adults follows the International Evidence-based Guideline for the Assessment and Management of Polycystic Ovary Syndrome, requiring two out of three criteria - clinical or biochemical hyperandrogenism, ovulatory dysfunction, and/or specific ovarian morphological characteristics or elevated anti-Müllerian hormone. However, diagnosing adolescents omits ovarian morphology and anti-Müllerian hormone considerations. PCOS, marked by insulin resistance and hyperandrogenism, strongly contributes to early-onset type 2 diabetes, with increased odds for cardiovascular diseases. Reproduction-related implications include irregular menstrual cycles, anovulatory infertility, heightened risks of pregnancy complications and endometrial cancer. Beyond physiological manifestations, PCOS is associated with anxiety, depression, eating disorders, psychosexual dysfunction and negative body image, collectively contributing to diminished health-related quality of life in patients. Despite its high prevalence persisting into menopause, diagnosing PCOS often involves extended timelines and multiple health-care visits. Treatment remains ad hoc owing to limited understanding of underlying mechanisms, highlighting the need for research delineating the aetiology and pathophysiology of the syndrome. Identifying factors contributing to PCOS will pave the way for personalized medicine approaches. Additionally, exploring novel biomarkers, refining diagnostic criteria and advancing treatment modalities will be crucial in enhancing the precision and efficacy of interventions that will positively impact the lives of patients.
Collapse
Affiliation(s)
| | - Helena Teede
- Monash Centre for Health Research and Implementation, Monash Health and Monash University, Melbourne, Victoria, Australia
| | - Robert J Norman
- Robinson Research Institute, Adelaide Medical School, Adelaide, South Australia, Australia
| | - Richard Legro
- Department of Obstetrics and Gynecology, Penn State College of Medicine, Hershey, PA, USA
- Department of Public Health Science, Penn State College of Medicine, Hershey, PA, USA
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anuja Dokras
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Joop Laven
- Division of Reproductive Endocrinology & Infertility, Department of Obstetrics and Gynecology, Erasmus MC, Rotterdam, Netherlands
| | - Kathleen Hoeger
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| |
Collapse
|
11
|
Salmeri N, Viganò P, Cavoretto P, Marci R, Candiani M. The kisspeptin system in and beyond reproduction: exploring intricate pathways and potential links between endometriosis and polycystic ovary syndrome. Rev Endocr Metab Disord 2024; 25:239-257. [PMID: 37505370 DOI: 10.1007/s11154-023-09826-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/18/2023] [Indexed: 07/29/2023]
Abstract
Endometriosis and polycystic ovary syndrome (PCOS) are two common female reproductive disorders with a significant impact on the health and quality of life of women affected. A novel hypothesis by evolutionary biologists suggested that these two diseases are inversely related to one another, representing a pair of diametrical diseases in terms of opposite alterations in reproductive physiological processes but also contrasting phenotypic traits. However, to fully explain the phenotypic features observed in women with these conditions, we need to establish a potential nexus system between the reproductive system and general biological functions. The recent discovery of kisspeptin as pivotal mediator of internal and external inputs on the hypothalamic-pituitary-gonadal axis has led to a new understanding of the neuroendocrine upstream regulation of the human reproductive system. In this review, we summarize the current knowledge on the physiological roles of kisspeptin in human reproduction, as well as its involvement in complex biological functions such as metabolism, inflammation and pain sensitivity. Importantly, these functions are known to be dysregulated in both PCOS and endometriosis. Within the evolving scientific field of "kisspeptinology", we critically discuss the clinical relevance of these discoveries and their potential translational applications in endometriosis and PCOS. By exploring the possibilities of manipulating this complex signaling system, we aim to pave the way for novel targeted therapies in these reproductive diseases.
Collapse
Affiliation(s)
- Noemi Salmeri
- Gynecology and Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Paola Viganò
- Infertility Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via M. Fanti 6, 20122, Milan, Italy.
| | - Paolo Cavoretto
- Gynecology and Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Roberto Marci
- Gynecology & Obstetrics, University of Ferrara, 44121, Ferrara, Italy
| | - Massimo Candiani
- Gynecology and Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| |
Collapse
|
12
|
Camili FE, Akis M, Adali E, Hismiogullari AA, Taskin MI, Guney G, Afsar S. Oncostatin M Is Related to Polycystic Ovary Syndrome-Case Control Study. Biomedicines 2024; 12:355. [PMID: 38397957 PMCID: PMC10886802 DOI: 10.3390/biomedicines12020355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Background: Oncostatin M, a novel adipokine, plays a role in oogenesis, lipogenesis, and inflammation and may contribute to polycystic ovary syndrome pathogenesis and related metabolic problems. Adipokines are believed to contribute to developing polycystic ovary syndrome and its accompanying metabolic parameters, such as dyslipidemia, insulin resistance, and cardiovascular diseases. Methods: In this case-control study, the patients were grouped in a 1:1 ratio into either the polycystic ovary syndrome (n = 32) or the control group (n = 32). Serum levels of fasting glucose, insulin, C-reactive protein, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, triglyceride, white blood cell count, thyroid-stimulating hormone, luteinizing hormone, follicle-stimulating hormone, total testosterone, prolactin, estradiol, homeostasis model assessment of insulin resistance, and oncostatin M were analyzed. Results: Oncostatin M levels were significantly lower, but C-reactive protein levels were substantially higher in the polycystic ovary syndrome group than in the control group (p = 0.002, p = 0.001, respectively). Oncostatin M was inversely correlated with total cholesterol, non-high-density lipoprotein cholesterol, fasting glucose, and the luteinizing hormone/follicle-stimulating hormone ratio (ρ = -0.329, p =0.017; ρ = -0.386, p = 0.005; ρ = -0.440, p = 0.001; ρ = -0.316, p = 0.023, respectively). Conversely, there was no correlation between oncostatin M and total testosterone level (ρ = 0.220; p = 0.118). In the context of inflammation and metabolic parameters, oncostatin M was inversely correlated with C-reactive protein, homeostatic model assessment for insulin resistance score, and low-density lipoprotein cholesterol (ρ = -0.353, p = 0.019; ρ = -0.275, p = 0.048; ρ = -0.470, p < 0.001, respectively). Conclusions: Plasma oncostatin M levels were considerably lower in patients with polycystic ovary syndrome than in the control group, and this was inversely correlated with the hormonal and metabolic parameters of polycystic ovary syndrome. Thus, oncostatin M may be a novel therapeutic target for polycystic ovary syndrome and its metabolic parameters.
Collapse
Affiliation(s)
- Figen Efe Camili
- Department of Obstetrics and Gynecology, School of Medicine, Balikesir University, 10145 Balikesir, Türkiye; (F.E.C.); (E.A.); (M.I.T.); (G.G.)
| | - Merve Akis
- Department of Medical Biochemistry, School of Medicine, Balikesir University, 10145 Balikesir, Türkiye; (M.A.); (A.A.H.)
| | - Ertan Adali
- Department of Obstetrics and Gynecology, School of Medicine, Balikesir University, 10145 Balikesir, Türkiye; (F.E.C.); (E.A.); (M.I.T.); (G.G.)
| | - Adnan Adil Hismiogullari
- Department of Medical Biochemistry, School of Medicine, Balikesir University, 10145 Balikesir, Türkiye; (M.A.); (A.A.H.)
| | - Mine Islimye Taskin
- Department of Obstetrics and Gynecology, School of Medicine, Balikesir University, 10145 Balikesir, Türkiye; (F.E.C.); (E.A.); (M.I.T.); (G.G.)
| | - Gurhan Guney
- Department of Obstetrics and Gynecology, School of Medicine, Balikesir University, 10145 Balikesir, Türkiye; (F.E.C.); (E.A.); (M.I.T.); (G.G.)
| | - Selim Afsar
- Department of Obstetrics and Gynecology, School of Medicine, Balikesir University, 10145 Balikesir, Türkiye; (F.E.C.); (E.A.); (M.I.T.); (G.G.)
| |
Collapse
|
13
|
Bril F, Ezeh U, Amiri M, Hatoum S, Pace L, Chen YH, Bertrand F, Gower B, Azziz R. Adipose Tissue Dysfunction in Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2023; 109:10-24. [PMID: 37329216 PMCID: PMC10735305 DOI: 10.1210/clinem/dgad356] [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: 01/19/2023] [Revised: 05/15/2023] [Accepted: 06/12/2023] [Indexed: 06/18/2023]
Abstract
PURPOSE Polycystic ovary syndrome (PCOS) is a complex genetic trait and the most common endocrine disorder of women, clinically evident in 5% to 15% of reproductive-aged women globally, with associated cardiometabolic dysfunction. Adipose tissue (AT) dysfunction appears to play an important role in the pathophysiology of PCOS even in patients who do not have excess adiposity. METHODS We undertook a systematic review concerning AT dysfunction in PCOS, and prioritized studies that assessed AT function directly. We also explored therapies that targeted AT dysfunction for the treatment of PCOS. RESULTS Various mechanisms of AT dysfunction in PCOS were identified including dysregulation in storage capacity, hypoxia, and hyperplasia; impaired adipogenesis; impaired insulin signaling and glucose transport; dysregulated lipolysis and nonesterified free fatty acids (NEFAs) kinetics; adipokine and cytokine dysregulation and subacute inflammation; epigenetic dysregulation; and mitochondrial dysfunction and endoplasmic reticulum and oxidative stress. Decreased glucose transporter-4 expression and content in adipocytes, leading to decreased insulin-mediated glucose transport in AT, was a consistent abnormality despite no alterations in insulin binding or in IRS/PI3K/Akt signaling. Adiponectin secretion in response to cytokines/chemokines is affected in PCOS compared to controls. Interestingly, epigenetic modulation via DNA methylation and microRNA regulation appears to be important mechanisms underlying AT dysfunction in PCOS. CONCLUSION AT dysfunction, more than AT distribution and excess adiposity, contributes to the metabolic and inflammation abnormalities of PCOS. Nonetheless, many studies provided contradictory, unclear, or limited data, highlighting the urgent need for additional research in this important field.
Collapse
Affiliation(s)
- Fernando Bril
- Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL 35233, USA
| | - Uche Ezeh
- California IVF Fertility Center, Sacramento, CA 95833, USA
- Department of Obstetrics & Gynecology, Heersink School of Medicine, UAB, Birmingham, AL 35233, USA
| | - Mina Amiri
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1516745811, Iran
| | - Sana Hatoum
- Foundation for Research and Education Excellence, Vestavia, AL 35243, USA
| | - Lauren Pace
- Department of Obstetrics & Gynecology, Heersink School of Medicine, UAB, Birmingham, AL 35233, USA
| | - Yen-Hao Chen
- Department of Research, Biomere-West, Richmond, CA 94806, USA
| | - Fred Bertrand
- Department of Clinical and Diagnostic Sciences, School of Health Professions, UAB, Birmingham, AL 35294, USA
| | - Barbara Gower
- Department of Nutrition Sciences, School of Health Professions, UAB, Birmingham, AL 35294, USA
| | - Ricardo Azziz
- Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL 35233, USA
- Department of Obstetrics & Gynecology, Heersink School of Medicine, UAB, Birmingham, AL 35233, USA
- Department of Healthcare Organization and Policy, School of Public Health, UAB, Birmingham, AL 35233, USA
- Department of Health Policy, Management and Behavior, School of Public Health, University at Albany, SUNY, Rensselaer, NY 12144, USA
| |
Collapse
|
14
|
Pant P, Sircar R, Prasad R, Prasad HO, Chitme HR. Protein Expression and Bioinformatics Study of Granulosa Cells of Polycystic Ovary Syndrome Expressed Under the Influence of DHEA. Clin Med Insights Endocrinol Diabetes 2023; 16:11795514231206732. [PMID: 38023736 PMCID: PMC10644732 DOI: 10.1177/11795514231206732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 09/22/2023] [Indexed: 12/01/2023] Open
Abstract
Background The reproductive system is heavily dependent on ovarian follicles, which are made up of germ cells (oocytes) and granulosa cells (GCs), including cumulus granulosa cells (CGCs) and mural granulosa cells (MGCs). Understanding their normal and steroid-induced functions is the key to understanding the pathophysiology of endocrinal diseases in women. Objective This study investigated the differentially expressed proteins by CGCs and MGCs of patients with polycystic ovarian syndrome (PCOS) and without subsequent exposure to dehydroepiandrosterone sulfate (DHEAS) and functional differentiation. Design The present study was observational and experimental study carried out in hospital involving 80 female patients undergoing IVF for infertility. Methods In this study, we isolated CGCs and MGCs from the follicular fluid of both PCOS and non-PCOS patients undergoing in vitro fertilization (IVF). The cells were cultured and treated with DHEAS for 48 hours, and these cells were extracted, digested, and analyzed by tandem mass spectrometry followed by processing of the results using open-source bioinformatics tools. Results The present investigation discovered 276 and 341 proteins in CGCs and MGCs, respectively. DHEAS reduced the number of proteins expressed by CGCs and MGCs to 34 and 57 from 91 and 94, respectively. Venn results of CGCs revealed 49, 53, 36, and 21 proteins in normal CGCs, PCOS-CGCs, post-DHEAS, and PCOS-CGCs, respectively. Venn analysis of MGCs showed 51 proteins specific to PCOS and 29 shared by normal and PCOS samples after DHEAS therapy. MGCs express the most binding and catalytic proteins, whereas CGCs express transporter-related proteins. A protein pathway study demonstrated considerable differences between normal and PCOS samples, while DHEAS-treated samples of both cell lines showed distinct pathways. String findings identified important network route components such as albumin, actin, apolipoprotein, complement component C3, and heat shock protein. Conclusion This is the first study to show how DHEAS-induced stress affects the expression of proteins by MGCs and CGCs isolated from normal and PCOS patients. Further studies are recommended to identify PCOS biomarkers from CGCs and MGCs expressed under the influence of DHEAS.
Collapse
Affiliation(s)
- Pankaj Pant
- Faculty of Pharmacy, DIT University, Dehradun, Uttarakhand, India
| | - Reema Sircar
- Indira IVF Hospital, Dehradun, Uttarakhand, India
| | - Ritu Prasad
- Morpheus Prasad International Hospital, Dehradun, Uttarakhand, India
| | - Hari Om Prasad
- Morpheus Prasad International Hospital, Dehradun, Uttarakhand, India
| | | |
Collapse
|
15
|
Garella R, Bernacchioni C, Chellini F, Tani A, Palmieri F, Parigi M, Guasti D, Cassioli E, Castellini G, Ricca V, Bani D, Sassoli C, Donati C, Squecco R. Adiponectin Modulates Smooth Muscle Cell Morpho-Functional Properties in Murine Gastric Fundus via Sphingosine Kinase 2 Activation. Life (Basel) 2023; 13:1812. [PMID: 37763216 PMCID: PMC10532860 DOI: 10.3390/life13091812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/31/2023] [Accepted: 08/15/2023] [Indexed: 09/29/2023] Open
Abstract
Adipokines are peptide hormones produced by the adipose tissue involved in several biological functions. Among adipokines, adiponectin (ADPN) has antidiabetic and anti-inflammatory properties. It can also modulate food intake at central and peripheral levels, acting on hypothalamus and facilitating gastric relaxation. ADPN exerts its action interacting with two distinct membrane receptors and triggering some well-defined signaling cascades. The ceramidase activity of ADPN receptor has been reported in many tissues: it converts ceramide into sphingosine. In turn, sphingosine kinase (SK) phosphorylates it into sphingosine-1 phosphate (S1P), a crucial mediator of many cellular processes including contractility. Using a multidisciplinary approach that combined biochemical, electrophysiological and morphological investigations, we explored for the first time the possible role of S1P metabolism in mediating ADPN effects on the murine gastric fundus muscle layer. By using a specific pharmacological inhibitor of SK2, we showed that ADPN affects smooth muscle cell membrane properties and contractile machinery via SK2 activation in gastric fundus, adding a piece of knowledge to the action mechanisms of this hormone. These findings help to identify ADPN and its receptors as new therapeutic targets or as possible prognostic markers for diseases with altered energy balance and for pathologies with fat mass content alterations.
Collapse
Affiliation(s)
- Rachele Garella
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (F.P.); (R.S.)
| | - Caterina Bernacchioni
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | - Flaminia Chellini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Alessia Tani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Francesco Palmieri
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (F.P.); (R.S.)
| | - Martina Parigi
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Daniele Guasti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Emanuele Cassioli
- Psychiatry Unit, Department of Health Sciences, University of Florence, 50134 Florence, Italy; (E.C.); (G.C.); (V.R.)
| | - Giovanni Castellini
- Psychiatry Unit, Department of Health Sciences, University of Florence, 50134 Florence, Italy; (E.C.); (G.C.); (V.R.)
| | - Valdo Ricca
- Psychiatry Unit, Department of Health Sciences, University of Florence, 50134 Florence, Italy; (E.C.); (G.C.); (V.R.)
| | - Daniele Bani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Chiara Sassoli
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (M.P.); (D.G.); (D.B.); (C.S.)
| | - Chiara Donati
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | - Roberta Squecco
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (F.P.); (R.S.)
| |
Collapse
|
16
|
Kicińska AM, Maksym RB, Zabielska-Kaczorowska MA, Stachowska A, Babińska A. Immunological and Metabolic Causes of Infertility in Polycystic Ovary Syndrome. Biomedicines 2023; 11:1567. [PMID: 37371662 PMCID: PMC10295970 DOI: 10.3390/biomedicines11061567] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Infertility has been recognized as a civilizational disease. One of the most common causes of infertility is polycystic ovary syndrome (PCOS). Closely interrelated immunometabolic mechanisms underlie the development of this complex syndrome and lead to infertility. The direct cause of infertility in PCOS is ovulation and implantation disorders caused by low-grade inflammation of ovarian tissue and endometrium which, in turn, result from immune and metabolic system disorders. The systemic immune response, in particular the inflammatory response, in conjunction with metabolic disorders, insulin resistance (IR), hyperadrenalism, insufficient secretion of progesterone, and oxidative stress lead not only to cardiovascular diseases, cancer, autoimmunity, and lipid metabolism disorders but also to infertility. Depending on the genetic and environmental conditions as well as certain cultural factors, some diseases may occur immediately, while others may become apparent years after an infertility diagnosis. Each of them alone can be a significant factor contributing to the development of PCOS and infertility. Further research will allow clinical management protocols to be established for PCOS patients experiencing infertility so that a targeted therapy approach can be applied to the factor underlying and driving the "vicious circle" alongside symptomatic treatment and ovulation stimulation. Hence, therapy of fertility for PCOS should be conducted by interdisciplinary teams of specialists as an in-depth understanding of the molecular relationships and clinical implications between the immunological and metabolic factors that trigger reproductive system disorders is necessary to restore the physiology and homeostasis of the body and, thus, fertility, among PCOS patients.
Collapse
Affiliation(s)
- Aleksandra Maria Kicińska
- Department of Physiology, Faculty of Medicine, Medical University of Gdansk, ul. Debinki 1, 80-210 Gdansk, Poland; (A.M.K.); (M.A.Z.-K.)
| | - Radoslaw B. Maksym
- 1st Department of Obstetrics and Gynecology, Centre for Postgraduate Medical Education, ul. Żelazna 90, 02-004 Warsaw, Poland;
| | - Magdalena A. Zabielska-Kaczorowska
- Department of Physiology, Faculty of Medicine, Medical University of Gdansk, ul. Debinki 1, 80-210 Gdansk, Poland; (A.M.K.); (M.A.Z.-K.)
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, ul. Debinki 1, 80-210 Gdansk, Poland
| | - Aneta Stachowska
- Department of Physiology, Faculty of Medicine, Medical University of Gdansk, ul. Debinki 1, 80-210 Gdansk, Poland; (A.M.K.); (M.A.Z.-K.)
| | - Anna Babińska
- Department of Endocrinology and Internal Medicine, Medical University of Gdansk, 80-210 Gdansk, Poland
| |
Collapse
|
17
|
Thaqi G, Berisha B, Pfaffl MW. Expression of Locally Produced Adipokines and Their Receptors during Different Physiological and Reproductive Stages in the Bovine Corpus Luteum. Animals (Basel) 2023; 13:1782. [PMID: 37889693 PMCID: PMC10251875 DOI: 10.3390/ani13111782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 10/05/2023] Open
Abstract
This study aimed to determine the gene expression of different local novel adipokines, such as vaspin, adiponectin, visfatin, and resistin, and their known receptors, namely, heat shock 70 protein 5, adiponectin receptor 1, and adiponectin receptor 2, in the bovine corpus luteum (CL) during different phases of the estrous cycle (on days 1-2, 3-4, 5-7, 8-12, 13-18, >18) and pregnancy (at months 1-2, 3-4, 5-7, >7). The mRNA expression was measured by reverse transcription polymerase chain reaction (RT-qPCR). The mRNA expression levels were normalized to the geometric mean of all three constantly expressed reference genes (cyclophilin A, ubiquitin, ubiquitin C). Our findings suggest that adipokines are expressed and present in all investigated groups, and are specifically up- or downregulated during the estrus cycle and during pregnancy. Vaspin and adiponectin levels were upregulated in the middle and late cycle stages. Resistin was abundant during the CL regression stage and in the first months of pregnancy. The specific expression of adipokine receptors indicates their involvement in the local mechanisms that regulate CL function. Further investigations are required to elucidate the regulative mechanisms underlying the different local effects of adipokines on the ovarian physiology of cows.
Collapse
Affiliation(s)
- Granit Thaqi
- Chair of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, 85354 Weihenstephan, Germany; (B.B.); (M.W.P.)
| | - Bajram Berisha
- Chair of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, 85354 Weihenstephan, Germany; (B.B.); (M.W.P.)
- Department of Animal Biotechnology, Faculty of Agriculture and Veterinary, University of Prishtina, 10000 Prishtina, Kosovo
| | - Michael W. Pfaffl
- Chair of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, 85354 Weihenstephan, Germany; (B.B.); (M.W.P.)
| |
Collapse
|
18
|
Grewal T, Buechler C. Adipokines as Diagnostic and Prognostic Markers for the Severity of COVID-19. Biomedicines 2023; 11:1302. [PMID: 37238973 PMCID: PMC10215701 DOI: 10.3390/biomedicines11051302] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Accumulating evidence implicates obesity as a risk factor for increased severity of disease outcomes in patients infected with severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2). Obesity is associated with adipose tissue dysfunction, which not only predisposes individuals to metabolic complications, but also substantially contributes to low-grade systemic inflammation, altered immune cell composition, and compromised immune function. This seems to impact the susceptibility and outcome of diseases caused by viruses, as obese people appear more vulnerable to developing infections and they recover later from infectious diseases than normal-weight individuals. Based on these findings, increased efforts to identify suitable diagnostic and prognostic markers in obese Coronavirus disease 2019 (COVID-19) patients to predict disease outcomes have been made. This includes the analysis of cytokines secreted from adipose tissues (adipokines), which have multiple regulatory functions in the body; for instance, modulating insulin sensitivity, blood pressure, lipid metabolism, appetite, and fertility. Most relevant in the context of viral infections, adipokines also influence the immune cell number, with consequences for overall immune cell activity and function. Hence, the analysis of the circulating levels of diverse adipokines in patients infected with SARS-CoV-2 have been considered to reveal diagnostic and prognostic COVID-19 markers. This review article summarizes the findings aimed to correlate the circulating levels of adipokines with progression and disease outcomes of COVID-19. Several studies provided insights on chemerin, adiponectin, leptin, resistin, and galectin-3 levels in SARS-CoV-2-infected patients, while limited information is yet available on the adipokines apelin and visfatin in COVID-19. Altogether, current evidence points at circulating galectin-3 and resistin levels being of diagnostic and prognostic value in COVID-19 disease.
Collapse
Affiliation(s)
- Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93053 Regensburg, Germany
| |
Collapse
|
19
|
Tews HC, Elger T, Grewal T, Weidlich S, Vitali F, Buechler C. Fecal and Urinary Adipokines as Disease Biomarkers. Biomedicines 2023; 11:biomedicines11041186. [PMID: 37189804 DOI: 10.3390/biomedicines11041186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
The use of biomarkers is of great clinical value for the diagnosis and prognosis of disease and the assessment of treatment efficacy. In this context, adipokines secreted from adipose tissue are of interest, as their elevated circulating levels are associated with a range of metabolic dysfunctions, inflammation, renal and hepatic diseases and cancers. In addition to serum, adipokines can also be detected in the urine and feces, and current experimental evidence on the analysis of fecal and urinary adipokine levels points to their potential as disease biomarkers. This includes increased urinary adiponectin, lipocalin-2, leptin and interleukin-6 (IL-6) levels in renal diseases and an association of elevated urinary chemerin as well as urinary and fecal lipocalin-2 levels with active inflammatory bowel diseases. Urinary IL-6 levels are also upregulated in rheumatoid arthritis and may become an early marker for kidney transplant rejection, while fecal IL-6 levels are increased in decompensated liver cirrhosis and acute gastroenteritis. In addition, galectin-3 levels in urine and stool may emerge as a biomarker for several cancers. With the analysis of urine and feces from patients being cost-efficient and non-invasive, the identification and utilization of adipokine levels as urinary and fecal biomarkers could become a great advantage for disease diagnosis and predicting treatment outcomes. This review article highlights data on the abundance of selected adipokines in urine and feces, underscoring their potential to serve as diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Hauke C Tews
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Tanja Elger
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Simon Weidlich
- Department of Internal Medicine II, School of Medicine, University Hospital Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Francesco Vitali
- Department of Medicine 1, Gastroenterology, Pneumology and Endocrinology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christa Buechler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
20
|
Expression and Function of BMP and Activin Membrane-Bound Inhibitor (BAMBI) in Chronic Liver Diseases and Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:ijms24043473. [PMID: 36834884 PMCID: PMC9964332 DOI: 10.3390/ijms24043473] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
BAMBI (bone morphogenetic protein and activin membrane-bound inhibitor) is a transmembrane pseudoreceptor structurally related to transforming growth factor (TGF)-β type 1 receptors (TGF-β1Rs). BAMBI lacks a kinase domain and functions as a TGF-β1R antagonist. Essential processes such as cell differentiation and proliferation are regulated by TGF-β1R signaling. TGF-β is the best-studied ligand of TGF-βRs and has an eminent role in inflammation and fibrogenesis. Liver fibrosis is the end stage of almost all chronic liver diseases, such as non-alcoholic fatty liver disease, and at the moment, there is no effective anti-fibrotic therapy available. Hepatic BAMBI is downregulated in rodent models of liver injury and in the fibrotic liver of patients, suggesting that low BAMBI has a role in liver fibrosis. Experimental evidence convincingly demonstrated that BAMBI overexpression is able to protect against liver fibrosis. Chronic liver diseases have a high risk of hepatocellular carcinoma (HCC), and BAMBI was shown to exert tumor-promoting as well as tumor-protective functions. This review article aims to summarize relevant studies on hepatic BAMBI expression and its role in chronic liver diseases and HCC.
Collapse
|
21
|
Chiang YF, Huang KC, Chen HY, Huang TC, Ali M, Chang HY, Shieh TM, Shih YH, Wang KL, Huang YJ, Chung CP, Hsia SM. The Adipokine Visfatin Modulates Cancer Stem Cell Properties in Triple-Negative Breast Cancer. Biomedicines 2023; 11:biomedicines11020297. [PMID: 36830834 PMCID: PMC9953233 DOI: 10.3390/biomedicines11020297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/30/2022] [Accepted: 01/05/2023] [Indexed: 01/26/2023] Open
Abstract
Obesity is a cancer progression risk factor; excessive adipocytes increase adipokine secretion. Visfatin, a novel adipokine highly expressed in cancer patients, is related to breast cancer risk. The modulation of nicotinamide adenine dinucleotide (NAD+) metabolism and the induction of a tumorigenic environment plays a vital role in cancer progression. Among cancer cell types, cancer stem-like cells (CSCs) with self-renewal and chemotherapy-resistance abilities could modulate tumor progression and cancer recurrence ability. In this study, we focused on visfatin's modulation effect on stemness-related properties using the high-malignancy breast cancer cell line MDA-MB-231 in in vitro and in vivo studies. Visfatin treatment significantly increased both the sphere number and sphere diameter and increased the protein expression of NANOG homeobox (NANOG), sex-determining region Y-box 2 (SOX2), and octamer-binding transcription factor 4 (OCT4), as well as SIRT1 protein levels. The serum angiogenesis marker VEGF and extracellular nicotinamide phosphoribosyl transferase (NAMPT, visfatin) were induced after visfatin treatment, increasing the stemness and angiogenesis environment, which were significantly reduced by the visfatin inhibitor FK866. Our results demonstrate that the visfatin-activated SIRT-SOX2 axis promotes triple-negative breast cancer stemness and enriches the tumorigenic microenvironment.
Collapse
Affiliation(s)
- Yi-Fen Chiang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
| | - Ko-Chieh Huang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
| | - Hsin-Yuan Chen
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
| | - Tsui-Chin Huang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
| | - Mohamed Ali
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Hsin-Yi Chang
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei 114201, Taiwan
| | - Tzong-Ming Shieh
- School of Dentistry, College of Dentistry, China Medical University, Taichung 40402, Taiwan
| | - Yin-Hwa Shih
- Department of Healthcare Administration, Asia University, Taichung 41354, Taiwan
| | - Kai-Lee Wang
- Department of Nursing, Ching Kuo Institute of Management and Health, Keelung 20301, Taiwan
| | - Yun-Ju Huang
- Department of Biotechnology and Food Technology, Southern Taiwan University of Science and Technology, Tainan City 710301, Taiwan
| | - Cheng-Pei Chung
- Department of Nutrition and Health Sciences, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333324, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333324, Taiwan
| | - Shih-Min Hsia
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
- School of Food and Safety, Taipei Medical University, Taipei 110301, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 110301, Taiwan
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Correspondence: ; Tel.: +886-2-27361661 (ext. 6558)
| |
Collapse
|