1
|
Xiao D, Sun X, Li W, Wen Z, Zhang W, Yang L. Associations of Dietary Index for Gut Microbiota and Flavonoid Intake With Female Infertility in the United States. Food Sci Nutr 2025; 13:e70098. [PMID: 40144557 PMCID: PMC11938295 DOI: 10.1002/fsn3.70098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
This study aimed to investigate the associations between a dietary index for gut microbiota (DI-GM), flavonoid intake, and female infertility, while exploring age-specific differences in these relationships to identify potential dietary strategies for female infertility prevention. This cross-sectional study focused on female participants aged 18-45 years, with data obtained from the 2017-2018 cycle of the National Health and Nutrition Examination Survey (NHANES). Weighted multivariable logistic regression models were employed to examine the associations between DI-GM, flavonoid intake, and self-reported female infertility. Age-stratified analyses were performed to evaluate whether these associations varied across reproductive life stages. Higher DI-GM scores were significantly associated with reduced infertility risk (aOR = 0.30, 95% CI: 0.13-0.71, p = 0.006), with the strongest protective effects observed in women aged < 35 years (Q3: aOR = 0.13, 95% CI: 0.03-0.58, p = 0.007; Q4: aOR = 0.27, 95% CI: 0.09-0.77, p = 0.015). Beneficial gut microbiota scores also showed a protective effect (aOR = 0.75, 95% CI: 0.57-0.98, p = 0.036). Among women aged ≥ 35 years, moderate flavonoid intake (Q2) showed a significant inverse association with female infertility risk (aOR = 0.19, 95% CI: 0.06-0.66, p = 0.009). Our findings reveal novel evidence that higher DI-GM scores and moderate flavonoid intake are significantly linked to a lower risk of female infertility, with age-specific patterns observed. Higher DI-GM scores showed significant protective effects in younger women (< 35 years), while moderate flavonoid intake was protective in women aged ≥ 35 years. These findings underscore the potential of personalized dietary strategies targeting gut microbiota composition and flavonoid intake as cost-effective approaches for female infertility prevention and management across different reproductive life stages.
Collapse
Affiliation(s)
- Di Xiao
- Department of Comprehensive Maternal and Child Health, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
| | - Xiang Sun
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
| | - Weidong Li
- Department of Comprehensive Maternal and Child Health, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
| | - Zihao Wen
- Department of Comprehensive Maternal and Child Health, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
| | - Wei‐Hong Zhang
- International Centre for Reproductive Health (ICRH), Department of Public Health and Primary CareGhent UniversityGentBelgium
- School of Public HealthUniversité Libre de Bruxelles (ULB)BruxellesBelgium
| | - Li Yang
- Department of Comprehensive Maternal and Child Health, Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
2
|
Zhu J, Qi X, Zhang Z, Zhou Q, Gu R, Wu X, Zhong L. Exploring Biomarkers Related to Cell Adhesion in Polycystic Ovary Syndrome Using Bioinformatics and Conducting Experimental Validation. Int J Gen Med 2025; 18:1777-1794. [PMID: 40177425 PMCID: PMC11963825 DOI: 10.2147/ijgm.s509651] [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: 12/04/2024] [Accepted: 03/13/2025] [Indexed: 04/05/2025] Open
Abstract
Purpose Related studies have pointed out that cell adhesion may play an important role for treating Polycystic Ovary Syndrome (PCOS). This study aimed to identify and analyze the biomarkers associated with cell adhesion-related genes (CRGs) for treating PCOS and their biological mechanisms. Patients and Methods In this study, GSE80432 was used to identify differentially expressed genes (DEGs) (PCOS vs control group) through differential expression analysis. Then, the DEGs were overlapped with 1531 CRGs to obtain the cross - genes. Subsequently, the Support Vector Machine-Recursive Feature Elimination combined with the least absolute shrinkage and selection operator was utilized to obtain candidate genes, and the genes with AUC greater than 0.7 and consistent expression trends in the two datasets were defined as biomarkers. Finally, a nomogram was constructed, and enrichment analysis, regulatory network, drug prediction, the association between biomarkers and PCOS, and reverse transcription quantitative PCR (RT-qPCR) were carried out respectively. Results A total of 10 cross-genes were identified, and 2 biomarkers (DSG2 and TH11) were screened out from them. RT-qPCR analysis showed that the expression of THBS1 was increased in PCOS samples, while there was no significant difference in DSG2. In addition, enrichment analysis indicated that both DSG2 and THBS1 were enriched in the B-cell receptor signaling pathway. Then, based on these two biomarkers, lncRNA-miRNA-mRNA (81 nodes and 135 edges) and TFs biomarker networks (38 nodes and 38 edges), such as MIR17HG'-has-miR-7-5p'-THBS1, TFDP1-DSG2, were constructed respectively. By predicting drugs targeting biomarkers, 61 drugs were predicted to target DSG2, while 133 drugs were predicted to target THBS1. Moreover, a stronger association between THBS1 and PCOS was detected (inference score = 27.15). Conclusion In this study, 2 biomarkers (DSG2 and THBS1) were identified, providing a potential theoretical basis for PCOS treatment.
Collapse
Affiliation(s)
- Jiani Zhu
- Department of Clinical Nutrition, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, People’s Republic of China
- Department of Clinical Nutrition, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, People’s Republic of China
| | - Xinyue Qi
- Department of Preventive Health Care, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, People’s Republic of China
- Department of Preventive Health Care, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, People’s Republic of China
| | - Zhenyu Zhang
- Department of Clinical Nutrition, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, People’s Republic of China
- Department of Clinical Nutrition, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, People’s Republic of China
| | - Qun Zhou
- Department of Internal Medicine No.3, Shilin Yi Autonomous County People’s Hospital, Kunming, Yunnan, People’s Republic of China
| | - Ran Gu
- Department of Clinical Nutrition, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, People’s Republic of China
- Department of Clinical Nutrition, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, People’s Republic of China
| | - Xiaorong Wu
- Department of Reproductive Medicine, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, People’s Republic of China
- Department of Reproductive Medicine, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, People’s Republic of China
| | - Lanping Zhong
- Department of Reproductive Medicine, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, People’s Republic of China
- Department of Reproductive Medicine, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, People’s Republic of China
| |
Collapse
|
3
|
Naqvi I, Bandyopadhyay A, Panda A, Hareramadas B. Polycystic Ovarian Syndrome: A Review of Multi-omics Analyses. Reprod Sci 2025; 32:618-646. [PMID: 39875694 DOI: 10.1007/s43032-025-01789-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 01/09/2025] [Indexed: 01/30/2025]
Abstract
Polycystic Ovary Syndrome (PCOS) is among the most prevalent endocrinological abnormalities of young females, posing a grave public health challenge to the society. The objective of the present literature review is to analyze the enormous amount of information available by way of numerous multi-omic studies, and to explore a meaningful relationship between various factors such as genetic, proteomic, environmental etc. to understand the multifactorial metabolic disorder in a proper manner. Detailed literature search was done in various science article repositories and biomedical databases such as PubMed, Google Scholar, BioMed Central, Embase etc. by using several keywords in whole gamut of combinations. PCOS is a heritable disease. It manifests as a result of a combination of several intricately inter-linked symptoms such as anovulation, obesity, type II diabetes, hyperandrogenism, polycystic ovaries etc., the last one being the main manifestation of the disease, thus leading to infertility among several other complications. Such a multifactorial metabolic disorder with extreme symptomatic heterogeneity cannot be fully explained solely based on symptoms or genetic variations; thus, giving some space of thought to other factors such as epigenetic, microbiomic factors etc. playing a role in the causation of the disease. The present scientific survey of literature extensively reviews various aspects of PCOS by critically looking into the vast multi-omic data, and concluded with suggesting treatment options as well as lifestyle changes required to deal with the psychological/ emotional impacts of the condition on affected women.
Collapse
Affiliation(s)
- Ilmas Naqvi
- Department of Zoology, Zakir Husain Delhi College (University of Delhi), J.L.N. Marg, New Delhi, 110002, India
| | | | - Amisha Panda
- Lab. No. 115, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - B Hareramadas
- Department of Zoology, Zakir Husain Delhi College (University of Delhi), J.L.N. Marg, New Delhi, 110002, India.
| |
Collapse
|
4
|
Talebi S, Shab-Bidar S, Askari G, Mohammadi H, Moini A, Djafarian K. Comparison of the impact of intermittent fasting diet alone or in conjunction with probiotic supplementation versus calorie-restricted diet on inflammatory, oxidative stress, and antioxidant capacity biomarkers in women with polycystic ovary syndrome: A randomized placebo-controlled trial. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2025; 30:5. [PMID: 40200973 PMCID: PMC11974602 DOI: 10.4103/jrms.jrms_280_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/08/2024] [Accepted: 07/10/2024] [Indexed: 04/10/2025]
Abstract
Background The objective of this study was to compare the effects of early time-restricted eating (eTRE) and eTRE plus probiotic supplementation to daily caloric restriction (DCR) alone in terms of biomarkers of oxidative stress (OS), antioxidant capacity, inflammation, and blood pressure (BP) in obese women with polycystic ovary syndrome (PCOS). Materials and Methods The research was conducted as a randomized, parallel, placebo-controlled clinical trial with an 8-week follow-up period. Participants were randomly assigned to one of three groups: 14:10 eTRE with probiotic supplementation (n = 30), 14:10 eTRE with placebo supplementation (n = 30), or DCR with placebo supplementation (n = 30). At the beginning and 8 weeks of the intervention, systolic blood pressure (SBP) and diastolic BP, inflammation, and OS parameters were evaluated. Results A total of 90 participants (mean age, 30.49 years and mean weight, 81.45 kg) were enrolled in this trial. After 8-week intervention, we observed SBP significantly decreased in both the eTRE + probiotic group (-0.31 mmHg [95% confidence interval (CI): -0.55, -0.07]) and the eTRE + placebo group (-0.24 mmHg [95% CI: -0.43, 0.04]), with no significant differences observed between groups. Moreover, C-reactive protein (CRP) levels were significantly reduced in all groups (P < 0.005). Total antioxidant capacity (TAC) also showed notable improvement in both the eTRE + probiotic group (P = 0.012) and the DCR group (P = 0.032). However, there were no significant differences between the three groups regarding BP, OS, TAC, and CRP markers. Conclusion It was not found that eTRE alone or eTRE with probiotics intervention resulted in improving BP, inflammatory, OS, and antioxidant capacity biomarkers than a standard DCR diet among obese women with PCOS. The present study did not reveal significant improvements in BP, inflammatory markers, OS, or antioxidant capacity with either eTRE alone or eTRE combined with probiotics compared to a standard DCR among obese women diagnosed with PCOS.Trial Register no: IRCT20121110011421N5.
Collapse
Affiliation(s)
- Sepide Talebi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Sakineh Shab-Bidar
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Askari
- Department of Community Nutrition, Nutrition and Food Security Research Center, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamed Mohammadi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashraf Moini
- Breast Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Obstetrics and Gynecology, Arash Women’s Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Kurosh Djafarian
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Ahmad F, Ahmed SH, Choucair F, Chouliaras S, Awwad J, Terranegra A. A disturbed communication between hypothalamic-pituitary-ovary axis and gut microbiota in female infertility: is diet to blame? J Transl Med 2025; 23:92. [PMID: 39838491 PMCID: PMC11749209 DOI: 10.1186/s12967-025-06117-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
Female infertility is a multifactorial condition influenced by various genetic, environmental, and lifestyle factors. Recent research has investigated the significant impact of gut microbiome dysbiosis on systemic inflammation, metabolic dysfunction, and hormonal imbalances, which can potentially impair fertility. The gut-brain axis, a bidirectional communication system between the gut and the brain, also plays a significant role in regulating reproductive functions. Emerging evidence suggests that the gut microbiome can influence brain functions and behavior, further emphasizing the importance of the microbiota-gut-brain axis in reproduction. Given their role as a major modulator of the gut microbiome, diet and dietary factors, including dietary patterns and nutrient intake, have been implicated in the development and management of female infertility. Hence, this review aims to highlight the impact of dietary patterns, such as the Western diet (WD) and Mediterranean diet (MD), and to decipher their modulatory action on the microbiota-gut-brain axis in infertile women. By contrasting the detrimental effects of WD with the therapeutic potential of MD, we emphasize the pivotal role of a balanced diet rich in nutrients in promoting a healthy gut microbiome. These insights underscore the potential of targeted dietary interventions and lifestyle modifications as promising strategies to enhance reproductive outcomes in subfertile women.
Collapse
Affiliation(s)
- Fatima Ahmad
- Translational Medicine Department, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad bin Khalifa University, Doha, Qatar
| | - Salma H Ahmed
- Translational Medicine Department, Sidra Medicine, Doha, Qatar
| | - Fadi Choucair
- Reproductive Medicine Unit, Sidra Medicine, Doha, Qatar
| | - Spyridon Chouliaras
- Reproductive Medicine Unit, Sidra Medicine, Doha, Qatar
- Weill Cornell Medicine, Ar-Rayyan, Qatar
| | - Johnny Awwad
- Reproductive Medicine Unit, Sidra Medicine, Doha, Qatar
- Vincent Memorial Obstetrics and Gynecology Service, Massachusetts General Hospital, Boston, MA, USA
| | - Annalisa Terranegra
- Translational Medicine Department, Sidra Medicine, Doha, Qatar.
- College of Health and Life Sciences, Hamad bin Khalifa University, Doha, Qatar.
| |
Collapse
|
6
|
Xavier-Santos D, Bedani R, de Almeida Vieira I, Padilha M, Lima CMG, Silva JDR, Ferreira BM, Giraldo PC, Pagnossa JP, Sivieri K, Antunes AEC, Sant'Ana AS. Exploring the Potential Use of Probiotics, Prebiotics, Synbiotics, and Postbiotics as Adjuvants for Modulating the Vaginal Microbiome: a Bibliometric Review. Probiotics Antimicrob Proteins 2025:10.1007/s12602-024-10444-8. [PMID: 39821884 DOI: 10.1007/s12602-024-10444-8] [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] [Accepted: 12/19/2024] [Indexed: 01/19/2025]
Abstract
Women's health is related to several factors that include physical, mental, and reproductive health. Additionally, the vaginal microbiota modulation performs a fundamental role in the regulation of physiological homeostasis and dysbiosis, which provides us a potential overview of the use of different biotic agents and their implications for female health. The objective of this work was propitiated insights and conception about the influence of probiotics, prebiotics, synbiotics, and postbiotics as adjuvants for prevention/treatment on the main infections that can affect women's health. Therefore, seventy-one studies published in the Web of Science Core Collection database from 1999 to 2024 were evaluated and performed to a bibliometric analysis employing the VOSviewer software for scientific mapping and network analysis. Our results suggest that administration of biotic agents as adjuvants are relevant for the prevention and/or treatment of the main diseases that affect female health, since they contribute to a healthy vaginal microbiota through anti-inflammatory and antimicrobial activities. Most clinical studies have demonstrated the effectiveness of intervention using probiotics to the detriment of other biotic agents in women's health, being bacterial vaginosis, polycystic ovary syndrome, and vulvovaginal candidiasis, the main diseases evaluated. However, preclinical studies have emphasized that the inhibition of pathogens responsible for the process of vaginal dysbiosis may be due to the formation of biofilm and the synthesis of compounds that could prevent the adhesion of these microorganisms. Future perspectives point to the beneficial modulation of the vaginal microbiota by biotic agents as a promising adjuvant approach to improve women's health.
Collapse
Affiliation(s)
- Douglas Xavier-Santos
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
- Fraunhofer Institute for Process Engineering and Packaging (IVV), Freising, Germany
| | - Raquel Bedani
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| | | | - Marina Padilha
- Department of Social and Applied Nutrition, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Clara Mariana Gonçalves Lima
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| | - Juliana Dara Rabêlo Silva
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| | - Beatriz Manfrinato Ferreira
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| | - Paulo César Giraldo
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Jorge Pamplona Pagnossa
- Department of Biological Sciences, Pontifical Catholic University, Poços de Caldas, MG, Brazil
| | - Katia Sivieri
- Department of Food and Nutrition, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, SP, Brazil
| | | | - Anderson S Sant'Ana
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil.
| |
Collapse
|
7
|
Gautam R, Maan P, Jyoti A, Kumar A, Malhotra N, Arora T. The Role of Lifestyle Interventions in PCOS Management: A Systematic Review. Nutrients 2025; 17:310. [PMID: 39861440 PMCID: PMC11767734 DOI: 10.3390/nu17020310] [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: 12/09/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most prevalent endocrine disorders among reproductive-aged women. It is characterized by hyperandrogenism, anovulation, and polycystic ovaries. Lifestyle changes are suggested as first-line interventions in managing PCOS. This systematic review aims to assess the scientific evidence regarding the role of lifestyle modifications (dietary changes, physical activity, and behavioral changes) in improving reproductive, anthropometric, metabolic, and psychological outcomes in women with PCOS. Dietary interventions such as foods with low glycemic index scores; caloric restrictions; high-fiber, omega three fatty acid-rich diets; ketogenic diets; Mediterranean diets; antioxidant-rich food; and anti-inflammatory diets improve insulin sensitivity and hormonal balance in women with PCOS. Physical activity, like aerobic and resistance exercise, enhances insulin sensitivity, helps weight loss, and improves metabolic and reproductive outcomes in women with PCOS. Further, behavioral and education modules can also be used to improve awareness, adherence, and the effectiveness of conventional treatment and to manage mental health issues related to PCOS. Collectively, lifestyle modifications not only improve the biochemical, hormonal, and anthropometric parameters in PCOS patients but also reduce the long-term risks of metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Rohit Gautam
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research (ICMR), New Delhi 110029, India; (R.G.); (P.M.)
| | - Pratibha Maan
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research (ICMR), New Delhi 110029, India; (R.G.); (P.M.)
| | - Anshu Jyoti
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India; (A.J.); (A.K.)
| | - Anshu Kumar
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India; (A.J.); (A.K.)
| | - Neena Malhotra
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India; (A.J.); (A.K.)
| | - Taruna Arora
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research (ICMR), New Delhi 110029, India; (R.G.); (P.M.)
| |
Collapse
|
8
|
Johnson C, Garipoğlu G, Jeanes Y, Frontino G, Costabile A. The Role of Diet, Glycaemic Index and Glucose Control in Polycystic Ovary Syndrome (PCOS) Management and Mechanisms of Progression. Curr Nutr Rep 2025; 14:8. [PMID: 39753786 PMCID: PMC11698792 DOI: 10.1007/s13668-024-00601-4] [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] [Accepted: 10/28/2024] [Indexed: 01/06/2025]
Abstract
PURPOSE OF REVIEW Polycystic Ovary Syndrome (PCOS) is a complex endocrine disorder with several causal pathways including impaired glucose tolerance, insulin resistance (IR), compensatory hyperinsulinemia and excess androgens (hyperandrogenism). This heterogeneous condition causes a range of reproductive, metabolic and psychological implications, the severity of which can differ between individuals depending on factors such as age, diet, ethnicity, genetics, medication, contraceptive use, adiposity, and Body Mass Index (BMI). RECENT FINDINGS Dietary interventions that focus on a low glycaemic index and glucose control are an efficient first-line dietary solution for the management of impaired glucose tolerance and IR, which subsequently improves weight management, quality of life and PCOS-related symptoms in individuals with this condition. This review aims to explore the relevance of nutrition and more specifically, the association of glycaemic index and glycaemic load with PCOS, as well as to assess the potential benefits of manipulating those indexes in the dietary approach for this syndrome.
Collapse
Affiliation(s)
- Claire Johnson
- School of Life and Health Sciences, University of Roehampton, London, UK
| | - Gökçen Garipoğlu
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Bahçeşehir University, Istanbul, Turkey
| | - Yvonne Jeanes
- School of Life and Health Sciences, University of Roehampton, London, UK
| | - Giada Frontino
- Consultant Obstetrician and Gynaecologist, London, England
| | - Adele Costabile
- School of Life and Health Sciences, University of Roehampton, London, UK.
| |
Collapse
|
9
|
Yu L, Chen X, Bai X, Fang J, Sui M. Microbiota Alters and Its Correlation with Molecular Regulation Underlying Depression in PCOS Patients. Mol Neurobiol 2024; 61:9977-9992. [PMID: 37995075 DOI: 10.1007/s12035-023-03744-7] [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: 08/15/2023] [Accepted: 10/27/2023] [Indexed: 11/24/2023]
Abstract
Depression is one of the complications in patients with polycystic ovary syndrome (PCOS) that leads to considerable mental health. Accumulating evidence suggests that human gut microbiomes are associated with the progression of PCOS and depression. However, whether microbiota influences depression development in PCOS patients is still uncharacterized. In this study, we employed metagenomic sequencing and transcriptome sequencing (RNA-seq) to profile the composition of the fecal microbiota and gene expression of peripheral blood mononuclear cells in depressed women with PCOS (PCOS-DP, n = 27) in comparison to mentally healthy women with PCOS (PCOS, n = 18) and compared with healthy control (HC, n = 27) and patients with major depressive disorder (MDD, n = 29). Gut microbiota assessment revealed distinct patterns in the relative abundance in the PCOS-DP compared to HC, MDD, and PCOS groups. Several gut microbes exhibited uniquely and significantly higher abundance in the PCOS-DP compared to PCOS patients, inducing EC Ruminococcus torques, Coprococcus comes, Megasphaera elsdenii, Acidaminococcus intestini, and Barnesiella viscericola. Bacteroides eggerthii was a potential gut microbial biomarker for the PCOS-DP. RNA-seq profiling identified that 35 and 37 genes were significantly elevated and downregulated in the PCOS-DP, respectively. The enhanced differential expressed genes (DEGs) in the PCOS-DP were enriched in pathways involved in signal transduction and endocrine and metabolic diseases, whereas several lipid metabolism pathways were downregulated. Intriguingly, genes correlated with the gut microbiota were found to be significantly enriched in pathways of neurodegenerative diseases and the immune system, suggesting that changes in the microbiota may have a systemic impact on the expression of neurodegenerative diseases and immune genes. Gut microbe-related DEGs of CREB3L3 and CCDC173 were possible molecular biomarkers and therapeutic targets of women with PCOS-DP. Our multi-omics data indicate shifts in the gut microbiome and host gene regulation in PCOS patients with depression, which is of possible etiological and diagnostic importance.
Collapse
Affiliation(s)
- Liying Yu
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China.
| | - Xiaoyu Chen
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Xuefeng Bai
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Jingping Fang
- College of Life Science, Fujian Normal University, Fuzhou, 350117, China
| | - Ming Sui
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China.
| |
Collapse
|
10
|
Resta SC, Guerra F, Talà A, Bucci C, Alifano P. Beyond Inflammation: Role of Pyroptosis Pathway Activation by Gram-Negative Bacteria and Their Outer Membrane Vesicles (OMVs) in the Interaction with the Host Cell. Cells 2024; 13:1758. [PMID: 39513865 PMCID: PMC11545737 DOI: 10.3390/cells13211758] [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: 09/27/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Pyroptosis is a gasdermin-mediated pro-inflammatory programmed cell death that, during microbial infections, aims to restrict the spreading of bacteria. Nevertheless, excessive pyroptosis activation leads to inflammation levels that are detrimental to the host. Pathogen-associated molecular patterns (PAMPs) present in bacteria and outer membrane vesicles (OMVs) can trigger pyroptosis pathways in different cell types with different outcomes. Moreover, some pathogens have evolved virulence factors that directly interfere with pyroptosis pathways, like Yersinia pestis YopM and Shigella flexneri IpaH7.8. Other virulence factors, such as those of Neisseria meningitidis, Neisseria gonorrhoeae, Salmonella enterica, and Helicobacter pylori affect pyroptosis pathways indirectly with important differences between pathogenic and commensal species of the same family. These pathogens deserve special attention because of the increasing antimicrobial resistance of S. flexneri and N. gonorrhoeae, the high prevalence of S. enterica and H. pylori, and the life-threatening diseases caused by N. meningitidis and Y. pestis. While inflammation due to macrophage pyroptosis has been extensively addressed, the effects of activation of pyroptosis pathways on modulation of cell cytoskeleton and cell-cell junctions in epithelia and endothelia and on the bacterial crossing of epithelial and endothelial barriers have only been partly investigated. Another important point is the diverse consequences of pyroptosis pathways on calcium influx, like activation of calcium-dependent enzymes and mitochondria dysregulation. This review will discuss the pyroptotic pathways activated by Gram-negative bacteria and their OMVs, analyzing the differences between pathogens and commensal bacteria. Particular attention will also be paid to the experimental models adopted and the main results obtained in the different models. Finally, strategies adopted by pathogens to modulate these pathways will be discussed with a perspective on the use of pyroptosis inhibitors as adjuvants in the treatment of infections.
Collapse
Affiliation(s)
- Silvia Caterina Resta
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy; (S.C.R.); (F.G.); (A.T.)
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy; (S.C.R.); (F.G.); (A.T.)
| | - Adelfia Talà
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy; (S.C.R.); (F.G.); (A.T.)
| | - Cecilia Bucci
- Department of Experimental Medicine (DiMeS), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy;
| | - Pietro Alifano
- Department of Experimental Medicine (DiMeS), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy;
| |
Collapse
|
11
|
Kukaev E, Kirillova E, Tokareva A, Rimskaya E, Starodubtseva N, Chernukha G, Priputnevich T, Frankevich V, Sukhikh G. Impact of Gut Microbiota and SCFAs in the Pathogenesis of PCOS and the Effect of Metformin Therapy. Int J Mol Sci 2024; 25:10636. [PMID: 39408965 PMCID: PMC11477200 DOI: 10.3390/ijms251910636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a complex disorder that impacts both the endocrine and metabolic systems, often resulting in infertility, obesity, insulin resistance, and cardiovascular complications. The aim of this study is to investigate the role of intestinal flora and its metabolites, particularly short-chain fatty acids (SCFAs), in the development of PCOS, and to assess the effects of metformin therapy on these components. SCFA levels in fecal and blood samples from women with PCOS (n=69) and healthy controls (n=18) were analyzed using Gas Chromatography-Mass Spectrometry (GC/MS) for precise measurement. Fecal microbiota were quantitatively detected by real-time polymerase chain reaction (PCR). To assess the efficacy of six months of metformin treatment, changes in the microbiota and SCFAs in the PCOS group (n=69) were also evaluated. The results revealed that women with PCOS exhibited a significant reduction in beneficial bacteria (namely, the C. leptum group and Prevotella spp.) alongside a notable overgrowth of opportunistic microorganisms (C. perfringens, C. difficile, Staphylococcus spp., and Streptococcus spp.). An overproduction of acetic acid (AA, FC=0.47, p<0.05) and valeric acid (VA, FC=0.54, p<0.05) suggests a link between elevated SCFAs and the development of obesity and PCOS. Interestingly, AA in the bloodstream might offer a protective effect against PCOS by ameliorating key symptoms such as high body mass index (r=-0.33, p=0.02), insulin resistance (r=-0.39, p=0.02), and chronic inflammation. Although serum SCFA levels showed non-significant changes following metformin treatment (p>0.05), the normalization of AA in the gut underscores that metformin exerts a more pronounced effect locally within the gastrointestinal tract. Furthermore, the study identified the most effective model for predicting the success of metformin therapy, based on serum concentrations of butyric acid (BA) and VA, achieving a 91% accuracy rate, 100% sensitivity, and 80% specificity. These promising findings highlight the potential for developing targeted interventions and personalized treatments, ultimately improving clinical outcomes for women with PCOS.
Collapse
Affiliation(s)
- Evgenii Kukaev
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (E.K.); (A.T.); (E.R.); (N.S.); (G.C.); (T.P.); (V.F.); (G.S.)
- V.L. Talrose Institute for Energy Problems of Chemical Physics, N.N. Semenov Federal Research Center of Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Ekaterina Kirillova
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (E.K.); (A.T.); (E.R.); (N.S.); (G.C.); (T.P.); (V.F.); (G.S.)
| | - Alisa Tokareva
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (E.K.); (A.T.); (E.R.); (N.S.); (G.C.); (T.P.); (V.F.); (G.S.)
| | - Elena Rimskaya
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (E.K.); (A.T.); (E.R.); (N.S.); (G.C.); (T.P.); (V.F.); (G.S.)
- Lebedev Physical Institute, 119991 Moscow, Russia
| | - Natalia Starodubtseva
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (E.K.); (A.T.); (E.R.); (N.S.); (G.C.); (T.P.); (V.F.); (G.S.)
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Galina Chernukha
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (E.K.); (A.T.); (E.R.); (N.S.); (G.C.); (T.P.); (V.F.); (G.S.)
| | - Tatiana Priputnevich
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (E.K.); (A.T.); (E.R.); (N.S.); (G.C.); (T.P.); (V.F.); (G.S.)
| | - Vladimir Frankevich
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (E.K.); (A.T.); (E.R.); (N.S.); (G.C.); (T.P.); (V.F.); (G.S.)
- Laboratory of Translational Medicine, Siberian State Medical University, 634050 Tomsk, Russia
| | - Gennady Sukhikh
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (E.K.); (A.T.); (E.R.); (N.S.); (G.C.); (T.P.); (V.F.); (G.S.)
| |
Collapse
|
12
|
Talebi S, Shab-Bidar S, Moini A, Mohammadi H, Djafarian K. The effects of time-restricted eating alone or in combination with probiotic supplementation in comparison with a calorie-restricted diet on endocrine and metabolic profiles in women with polycystic ovary syndrome: A randomized clinical trial. Diabetes Obes Metab 2024; 26:4468-4479. [PMID: 39143654 DOI: 10.1111/dom.15801] [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: 06/12/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 08/16/2024]
Abstract
AIM To assess the impact of intermittent fasting, with or without probiotic supplementation, versus a calorie-restricted diet on anthropometric measures, metabolic status and gonadal variables in women with polycystic ovary syndrome (PCOS). METHODS This is a randomized, placebo-controlled, parallel-arm clinical trial. The effects of the 14:10 early time-restricted eating (eTRE) strategy alone or combined with probiotics, on obese women with PCOS, were investigated. Participants were divided into three groups: eTRE plus probiotics (n = 30), eTRE plus placebo (n = 30) and a control group following a standard three-meal-per-day diet with daily calorie restriction (DCR) (n = 30). Over 8 weeks, various anthropometric, metabolic, menstrual and gonadal variables were assessed. RESULTS A total of 90 individuals were included in the study, with a mean body weight of 81.4 kg, and a mean age of 30 years. Mean (standard deviation) weight loss was not different between the groups at week 8 (TRE + probiotic: -2.2 [1.6] kg vs. TRE + placebo: -2.9 [2.7] kg vs. DCR: -2.5 [1.7] kg). Results revealed that, while all three regimes led to reductions in body weight, body mass index, vascular risk indicators, hirsutism and acne scores, there were no statistically significant differences between the eTRE groups and the control group in terms of weight loss, or improvements in metabolic, menstrual and gonadal variables (P > .05). Additionally, combining probiotics with eTRE did not benefit hormonal and cardiometabolic factors (P > .05). CONCLUSIONS The eTRE alone or eTRE plus probiotics did not result in significantly greater weight loss or improvements in metabolic, menstrual and gonadal variables compared with the standard three-meal DCR diet.
Collapse
Affiliation(s)
- Sepide Talebi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Sakineh Shab-Bidar
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashraf Moini
- Breast Disease Research Center (BDRC), Tehran University of Medical Sciences, Tehran, Iran
- Department of Obstetrics and Gynecology, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Hamed Mohammadi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Kurosh Djafarian
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Garmendia JV, De Sanctis CV, Hajdúch M, De Sanctis JB. Microbiota and Recurrent Pregnancy Loss (RPL); More than a Simple Connection. Microorganisms 2024; 12:1641. [PMID: 39203483 PMCID: PMC11357228 DOI: 10.3390/microorganisms12081641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Recurrent Pregnancy Loss (RPL) affects 1-2% of women, and its triggering factors are unclear. Several studies have shown that the vaginal, endometrial, and gut microbiota may play a role in RPL. A decrease in the quantity of Lactobacillus crispatus in local microbiota has been associated with an increase in local (vaginal and endometrial) inflammatory response and immune cell activation that leads to pregnancy loss. The inflammatory response may be triggered by gram-negative bacteria, lipopolysaccharides (LPS), viral infections, mycosis, or atypia (tumor growth). Bacterial structures and metabolites produced by microbiota could be involved in immune cell modulation and may be responsible for immune cell activation and molecular mimicry. Gut microbiota metabolic products may increase the amount of circulating pro-inflammatory lymphocytes, which, in turn, will migrate into vaginal or endometrial tissues. Local pro-inflammatory Th1 and Th17 subpopulations and a decrease in local Treg and tolerogenic NK cells are accountable for the increase in pregnancy loss. Local microbiota may modulate the local inflammatory response, increasing pregnancy success. Analyzing local and gut microbiota may be necessary to characterize some RPL patients. Although oral supplementation of probiotics has not been shown to modify vaginal or endometrial microbiota, the metabolites produced by it may benefit patients. Lactobacillus crispatus transplantation into the vagina may enhance the required immune tolerogenic response to achieve a normal pregnancy. The effect of hormone stimulation and progesterone to maintain early pregnancy on microbiota has not been adequately studied, and more research is needed in this area. Well-designed clinical trials are required to ascertain the benefit of microbiota modulation in RPL.
Collapse
Affiliation(s)
- Jenny Valentina Garmendia
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Hněvotínská 1333/5, 779 00 Olomouc, Czech Republic; (J.V.G.); (M.H.)
| | - Claudia Valentina De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Hněvotínská 1333/5, 779 00 Olomouc, Czech Republic; (J.V.G.); (M.H.)
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Hněvotínská 1333/5, 779 00 Olomouc, Czech Republic; (J.V.G.); (M.H.)
- Czech Advanced Technology and Research Institute, Palacky University, 779 00 Olomouc, Czech Republic
- Laboratory of Experimental Medicine, University Hospital Olomouc (FNOL), Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Hněvotínská 1333/5, 779 00 Olomouc, Czech Republic; (J.V.G.); (M.H.)
- Czech Advanced Technology and Research Institute, Palacky University, 779 00 Olomouc, Czech Republic
| |
Collapse
|
14
|
Çıtar Dazıroğlu ME, Acar Tek N, Cevher Akdulum MF, Yılmaz C, Yalınay AM. Effects of kefir consumption on gut microbiota and health outcomes in women with polycystic ovary syndrome. Food Sci Nutr 2024; 12:5632-5646. [PMID: 39139979 PMCID: PMC11317752 DOI: 10.1002/fsn3.4212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 08/15/2024] Open
Abstract
Polycystic Ovary Syndrome (PCOS), which is common among women of reproductive age, is characterized by low-grade chronic inflammation and is associated with several health problems and dysbiosis. Kefir has been shown to have many beneficial health effects; however, its effect on PCOS is unknown. This study aimed to examine the effect of kefir on the intestinal microbiota and health outcomes in PCOS. In this intervention study, 17 women with PCOS consumed 250 mL/day of kefir (containing Lactobacillus kefiranofaciens subsp. kefiranofaciens, Lactobacillus kefiranofaciens subsp. kefirgranum, Lactobacillus kefiri, Lactobacillus acidophilus, Lactobacillus parakefiri, Lactobacillus bulgaricus, Lactobacillus reuteri, Lactobacillus casei, Lactobacillus fermentum, Lactobacillus helveticus, Lactococcus lactis, Leuconostoc mesentereoides, Bifidobacterium bifidum, Streptococcus thermophilus, Kluyveromyces marxianus, Kluyveromyces lactis, Acetobacter pasteurianus, and Saccharomyces cerevisiae) for 8 weeks. Food consumption and physical activity records, anthropometrical measurements, quality of life, and fecal and blood samples were taken at the study's beginning and end. Quality of life in mental health (58.8 ± 15.08; 64.0 ± 15.23, respectively) and physical function (95.00 and 100.00, respectively) categories showed a significant increase after kefir intervention (p < .05). Additionally, Interleukin-6 (IL-6), one of the inflammatory cytokines, significantly decreased (174.00 and 109.10 ng/L, respectively) (p < .05). The intestinal barrier permeability was evaluated with zonulin, and no significant change was observed. Gut microbiota analysis showed that while the relative abundance of the class Bacilli and genus Lactococcus significantly increased, the genus Holdemania decreased with kefir consumption (p < .05). In conclusion, kefir appears to be beneficial for improving the microbiota and some health outcomes, like reducing inflammation and improving quality of life in PCOS. Therefore, kefir may be useful in the treatment of PCOS.
Collapse
Affiliation(s)
| | - Nilüfer Acar Tek
- Department of Nutrition and DieteticsGazi UniversityAnkaraTurkey
| | | | - Canan Yılmaz
- Department of Medical BiochemistryGazi UniversityAnkaraTurkey
| | | |
Collapse
|
15
|
Rizk FH, El Saadany AA, Elshamy AM, Abd Ellatif RA, El-Guindy DM, Helal DS, Hamama MG, El-Sharnoby JAEH, Abdel Ghafar MT, Faheem H. Ameliorating effects of adropin on letrozole-induced polycystic ovary syndrome via regulating steroidogenesis and the microbiota inflammatory axis in rats. J Physiol 2024; 602:3621-3639. [PMID: 38980987 DOI: 10.1113/jp285793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 06/26/2024] [Indexed: 07/11/2024] Open
Abstract
Growing evidence supports the role of gut microbiota in chronic inflammation, insulin resistance (IR) and sex hormone production in polycystic ovary syndrome (PCOS). Adropin plays a pivotal role in the regulation of glucose and lipid metabolism and is negatively correlated with IR, which affects intestinal microbiota and sex hormones. However, the effect of adropin administration in PCOS has yet to be investigated. The present study aimed to assess the effects of adropin on letrozole (LTZ)-induced PCOS in rats and the potential underlying mechanisms. The experimental groups were normal, adropin, letrozole and LTZ + adropin. At the end of the experiment, adropin significantly ameliorated PCOS, as evidenced by restoring the normal ovarian structure, decreasing the theca cell thickness in antral follicles, as well as serum testosterone and luteinizing hormone levels and luteinizing hormone/follicle-stimulating hormone ratios, at the same time as increasing granulosa cell thickness in antral follicles, oestradiol and follicle-stimulating hormone levels. The ameliorating effect could be attributed to its effect on sex hormone-binding globulin, key steroidogenic genes STAR and CYP11A1, IR, lipid profile, gut microbiota metabolites-brain-ovary axis components (short chain fatty acids, free fatty acid receptor 3 and peptide YY), intestinal permeability marker (zonulin and tight junction protein claudin-1), lipopolysaccharides/Toll-like receptor 4/nuclear factor kappa B inflammatory pathway and oxidative stress makers (malondialdehyde and total antioxidant capacity). In conclusion, adropin has a promising therapeutic effect on PCOS by regulating steroidogenesis, IR, lipid profile, the gut microbiota inflammatory axis and redox homeostasis. KEY POINTS: Adropin treatment reversed endocrine and ovarian morphology disorders in polycystic ovary syndrome (PCOS). Adropin regulated the ovarian steroidogenesis and sex hormone-binding globulin in PCOS. Adropin improved lipid profile and decreased insulin resistance in PCOS. Adropin modulated the components of the gut-brain-ovary axis (short chain fatty acids, free fatty acid receptor 3 and peptide YY) in PCOS. Adropin improved intestinal barrier integrity, suppressed of lipopolysaccharides/Toll-like receptor 4/nuclear factor kappa B signalling pathway and oxidative stress in PCOS.
Collapse
Affiliation(s)
- Fatma H Rizk
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Amira A El Saadany
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Amira Mostafa Elshamy
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Rasha A Abd Ellatif
- Department of Anatomy and Embryology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Dina M El-Guindy
- Department of Pathology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Duaa S Helal
- Department of Pathology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Mohamed G Hamama
- Department of Anatomy and Embryology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | | | - Heba Faheem
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
16
|
高 慧, 钱 贝, 倪 艳, 孙 莉, 傅 君. [Research Progress in the Pathogenesis of Polycystic Ovary Syndrome]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:1049-1054. [PMID: 39170002 PMCID: PMC11334293 DOI: 10.12182/20240760208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Indexed: 08/23/2024]
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common gynecological endocrine disorders. Most pathophysiological changes of PCOS begin in the peripubertal phase, and these pathophysiological changes will continuously affect women's health in the later stages of their lives. The pathogenic mechanisms of PCOS remain unclear, involving key aspects such as the regulation of hypothalamic-pituitary function, ovarian cellular functions, androgen levels, and insulin resistance. Herein, we summarized the latest findings on the pathogenesis of PCOS from the perspectives of the genetic background, intrauterine development, neuroendocrine function, inflammatory factors, gut microbiome, and environmental factors. This review will help provide new ideas for a deeper understanding of the disease, as well as its clinical diagnosis and treatment.
Collapse
Affiliation(s)
- 慧慧 高
- 浙江大学医学院附属儿童医院 小儿青少年妇科 国家儿童健康与疾病临床医学研究中心 (杭州 310052)National Clinical Research Center for Child Health, Department of Pediatric and Adolescent Gynecology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - 贝冉 钱
- 浙江大学医学院附属儿童医院 小儿青少年妇科 国家儿童健康与疾病临床医学研究中心 (杭州 310052)National Clinical Research Center for Child Health, Department of Pediatric and Adolescent Gynecology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - 艳 倪
- 浙江大学医学院附属儿童医院 小儿青少年妇科 国家儿童健康与疾病临床医学研究中心 (杭州 310052)National Clinical Research Center for Child Health, Department of Pediatric and Adolescent Gynecology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - 莉颖 孙
- 浙江大学医学院附属儿童医院 小儿青少年妇科 国家儿童健康与疾病临床医学研究中心 (杭州 310052)National Clinical Research Center for Child Health, Department of Pediatric and Adolescent Gynecology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - 君芬 傅
- 浙江大学医学院附属儿童医院 小儿青少年妇科 国家儿童健康与疾病临床医学研究中心 (杭州 310052)National Clinical Research Center for Child Health, Department of Pediatric and Adolescent Gynecology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| |
Collapse
|
17
|
Arvanitakis K, Chatzikalil E, Kalopitas G, Patoulias D, Popovic DS, Metallidis S, Kotsa K, Germanidis G, Koufakis T. Metabolic Dysfunction-Associated Steatotic Liver Disease and Polycystic Ovary Syndrome: A Complex Interplay. J Clin Med 2024; 13:4243. [PMID: 39064282 PMCID: PMC11278502 DOI: 10.3390/jcm13144243] [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: 06/08/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) and polycystic ovary syndrome (PCOS) are prevalent conditions that have been correlated with infertility through overlapped pathophysiological mechanisms. MASLD is associated with metabolic syndrome and is considered among the major causes of chronic liver disease, while PCOS, which is characterized by ovulatory dysfunction and hyperandrogenism, is one of the leading causes of female infertility. The pathophysiological links between PCOS and MASLD have not yet been fully elucidated, with insulin resistance, hyperandrogenemia, obesity, and dyslipidemia being among the key pathways that contribute to liver lipid accumulation, inflammation, and fibrosis, aggravating liver dysfunction. On the other hand, MASLD exacerbates insulin resistance and metabolic dysregulation in women with PCOS, creating a vicious cycle of disease progression. Understanding the intricate relationship between MASLD and PCOS is crucial to improving clinical management, while collaborative efforts between different medical specialties are essential to optimize fertility and liver health outcomes in individuals with MASLD and PCOS. In this review, we summarize the complex interplay between MASLD and PCOS, highlighting the importance of increasing clinical attention to the prevention, diagnosis, and treatment of both entities.
Collapse
Affiliation(s)
- Konstantinos Arvanitakis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (K.A.); (G.K.); (S.M.); (G.G.)
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Elena Chatzikalil
- Athens Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Georgios Kalopitas
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (K.A.); (G.K.); (S.M.); (G.G.)
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Dimitrios Patoulias
- Second Propaedeutic Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Djordje S. Popovic
- Clinic for Endocrinology, Diabetes and Metabolic Disorders, Clinical Centre of Vojvodina, 21000 Novi Sad, Serbia;
- Medical Faculty, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Symeon Metallidis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (K.A.); (G.K.); (S.M.); (G.G.)
- Division of Endocrinology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece;
| | - Kalliopi Kotsa
- Division of Endocrinology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece;
| | - Georgios Germanidis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (K.A.); (G.K.); (S.M.); (G.G.)
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Theocharis Koufakis
- Second Propaedeutic Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| |
Collapse
|
18
|
Fedeli V, Unfer V, Dinicola S, Laganà AS, Canipari R, Monti N, Querqui A, Galante E, Laurenzi G, Bizzarri M. Inositol Restores Appropriate Steroidogenesis in PCOS Ovaries Both In Vitro and In Vivo Experimental Mouse Models. Cells 2024; 13:1171. [PMID: 39056753 PMCID: PMC11275052 DOI: 10.3390/cells13141171] [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: 05/21/2024] [Revised: 06/29/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Androgen excess is a key feature of several clinical phenotypes of polycystic ovary syndrome (PCOS). However, the presence of FSH receptor (FSHR) and aromatase (CYP19A1) activity responses to physiological endocrine stimuli play a critical role in the pathogenesis of PCOS. Preliminary data suggest that myo-Inositol (myo-Ins) and D-Chiro-Inositol (D-Chiro-Ins) may reactivate CYP19A1 activity. We investigated the steroidogenic pathway of Theca (TCs) and Granulosa cells (GCs) in an experimental model of murine PCOS induced in CD1 mice exposed for 10 weeks to a continuous light regimen. The effect of treatment with different combinations of myo-Ins and D-Chiro-Ins on the expression of Fshr, androgenic, and estrogenic enzymes was analyzed by real-time PCR in isolated TCs and GCs and in ovaries isolated from healthy and PCOS mice. Myo-Ins and D-Chiro-Ins, at a ratio of 40:1 at pharmacological and physiological concentrations, positively modulate the steroidogenic activity of TCs and the expression of Cyp19a1 and Fshr in GCs. Moreover, in vivo, inositols (40:1 ratio) significantly increase Cyp19a1 and Fshr. These changes in gene expression are mirrored by modifications in hormone levels in the serum of treated animals. Myo-Ins and D-Chiro-Ins in the 40:1 formula efficiently rescued PCOS features by up-regulating aromatase and FSHR levels while down-regulating androgen excesses produced by TCs.
Collapse
Affiliation(s)
- Valeria Fedeli
- Department of Experimental Medicine, University La Sapienza, 00185 Rome, Italy; (N.M.); (A.Q.); (E.G.)
- Systems Biology Group Lab, University La Sapienza, 00185 Rome, Italy;
- The Experts Group on Inositol in Basic and Clinical Research, and on PCOS (EGOI-PCOS), 00161 Rome, Italy;
| | - Vittorio Unfer
- The Experts Group on Inositol in Basic and Clinical Research, and on PCOS (EGOI-PCOS), 00161 Rome, Italy;
- Dept. of Gynaecology, UniCamillus—Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| | - Simona Dinicola
- Systems Biology Group Lab, University La Sapienza, 00185 Rome, Italy;
- The Experts Group on Inositol in Basic and Clinical Research, and on PCOS (EGOI-PCOS), 00161 Rome, Italy;
| | - Antonio Simone Laganà
- Unit of Obstetrics and Gynecology, “Paolo Giaccone” Hospital, Department of Health Promotion, Mother and ChildCare, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy;
| | - Rita Canipari
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Unit of Histology and Medical Embryology, Sapienza, University of Rome, 00185 Rome, Italy; (R.C.); (G.L.)
| | - Noemi Monti
- Department of Experimental Medicine, University La Sapienza, 00185 Rome, Italy; (N.M.); (A.Q.); (E.G.)
- Systems Biology Group Lab, University La Sapienza, 00185 Rome, Italy;
| | - Alessandro Querqui
- Department of Experimental Medicine, University La Sapienza, 00185 Rome, Italy; (N.M.); (A.Q.); (E.G.)
- Systems Biology Group Lab, University La Sapienza, 00185 Rome, Italy;
| | - Emanuele Galante
- Department of Experimental Medicine, University La Sapienza, 00185 Rome, Italy; (N.M.); (A.Q.); (E.G.)
- Systems Biology Group Lab, University La Sapienza, 00185 Rome, Italy;
| | - Gaia Laurenzi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Unit of Histology and Medical Embryology, Sapienza, University of Rome, 00185 Rome, Italy; (R.C.); (G.L.)
| | - Mariano Bizzarri
- Department of Experimental Medicine, University La Sapienza, 00185 Rome, Italy; (N.M.); (A.Q.); (E.G.)
- Systems Biology Group Lab, University La Sapienza, 00185 Rome, Italy;
- The Experts Group on Inositol in Basic and Clinical Research, and on PCOS (EGOI-PCOS), 00161 Rome, Italy;
| |
Collapse
|
19
|
Yun C, Yan S, Liao B, Ding Y, Qi X, Zhao M, Wang K, Zhuo Y, Nie Q, Ye C, Xia P, Ma M, Li R, Jiang C, Qiao J, Pang Y. The microbial metabolite agmatine acts as an FXR agonist to promote polycystic ovary syndrome in female mice. Nat Metab 2024; 6:947-962. [PMID: 38769396 DOI: 10.1038/s42255-024-01041-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 04/02/2024] [Indexed: 05/22/2024]
Abstract
Polycystic ovary syndrome (PCOS), an endocrine disorder afflicting 6-20% of women of reproductive age globally, has been linked to alterations in the gut microbiome. We previously showed that in PCOS, elevation of Bacteroides vulgatus in the gut microbiome was associated with altered bile acid metabolism. Here we show that B. vulgatus also induces a PCOS-like phenotype in female mice via an alternate mechanism independent of bile acids. We find that B. vulgatus contributes to PCOS-like symptoms through its metabolite agmatine, which is derived from arginine by arginine decarboxylase. Mechanistically, agmatine activates the farnesoid X receptor (FXR) pathway to subsequently inhibit glucagon-like peptide-1 (GLP-1) secretion by L cells, which leads to insulin resistance and ovarian dysfunction. Critically, the GLP-1 receptor agonist liraglutide and the arginine decarboxylase inhibitor difluoromethylarginine ameliorate ovarian dysfunction in a PCOS-like mouse model. These findings reveal that agmatine-FXR-GLP-1 signalling contributes to ovarian dysfunction, presenting a potential therapeutic target for PCOS management.
Collapse
Affiliation(s)
- Chuyu Yun
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Sen Yan
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Baoying Liao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yong Ding
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xinyu Qi
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yingying Zhuo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Qixing Nie
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Chuan Ye
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Pengyan Xia
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing, China
| | - Ming Ma
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Rong Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China.
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China.
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing, China.
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China.
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China.
| | - Yanli Pang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China.
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
20
|
Ahmed M, Riaz U, Lv H, Yang L. A Molecular Perspective and Role of NAD + in Ovarian Aging. Int J Mol Sci 2024; 25:4680. [PMID: 38731898 PMCID: PMC11083308 DOI: 10.3390/ijms25094680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
The decline in female fecundity is linked to advancing chronological age. The ovarian reserve diminishes in quantity and quality as women age, impacting reproductive efficiency and the aging process in the rest of the body. NAD+ is an essential coenzyme in cellular energy production, metabolism, cell signaling, and survival. It is involved in aging and is linked to various age-related conditions. Hallmarks associated with aging, diseases, and metabolic dysfunctions can significantly affect fertility by disturbing the delicate relationship between energy metabolism and female reproduction. Enzymes such as sirtuins, PARPs, and CD38 play essential roles in NAD+ biology, which actively consume NAD+ in their enzymatic activities. In recent years, NAD+ has gained much attention for its role in aging and age-related diseases like cancer, Alzheimer's, cardiovascular diseases, and neurodegenerative disorders, highlighting its involvement in various pathophysiological processes. However, its impact on female reproduction is not well understood. This review aims to bridge this knowledge gap by comprehensively exploring the complex interplay between NAD+ biology and female reproductive aging and providing valuable information that could help develop plans to improve women's reproductive health and prevent fertility issues.
Collapse
Affiliation(s)
- Mehboob Ahmed
- Hubei Hongshan Laboratory, Wuhan 430070, China; (M.A.); (U.R.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Umair Riaz
- Hubei Hongshan Laboratory, Wuhan 430070, China; (M.A.); (U.R.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Haimiao Lv
- Hubei Hongshan Laboratory, Wuhan 430070, China; (M.A.); (U.R.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Liguo Yang
- Hubei Hongshan Laboratory, Wuhan 430070, China; (M.A.); (U.R.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
21
|
Zhou M, Yu J, Li X, Ruan Z, Yu S. Role of the gut microbiota and innate immunity in polycystic ovary syndrome: Current updates and future prospects. J Cell Mol Med 2024; 28:e18258. [PMID: 38546608 PMCID: PMC10977384 DOI: 10.1111/jcmm.18258] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/12/2024] [Accepted: 03/09/2024] [Indexed: 04/11/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the modern intractable reproductive diseases. The female irregular menstruation, infertility, obesity, and so forth caused by PCOS have become a hot issue affecting family harmony and social development. The aetiology of PCOS is complex. In recent years, many scholars have found that its pathogenesis was related to the imbalance of gut microbiota. Gut microbiota can form two-way communication with the brain through the 'gut-brain axis' and affect the host's metabolism. Current research has confirmed that the gut microbiota can interfere with glucose and lipid metabolism, insulin sensitivity, hormone secretion and follicular development in women by altering intestinal mucosal permeability and secreting metabolites. In addition, the diversity and composition of gut microbiota of PCOS patients changed, which may affect the metabolic function of the gut microbiota and the ability to produce metabolites, and may also directly or indirectly affect the endocrine function. This study reviewed recent research advances about the role of gut microbiota in PCOS. In order to provide basis for prevention and treatment of PCOS based on gut microbiota.
Collapse
Affiliation(s)
- Min Zhou
- Department of GynaecologyThe Affiliated Hospital to Changchun University of Chinese MedicineChangchunJilinChina
| | - Jing Yu
- Department of EndocrinologyThe Affiliated Hospital to Changchun University of Chinese MedicineChangchunJilinChina
| | - Xuewei Li
- Department of GynaecologyThe Affiliated Hospital to Changchun University of Chinese MedicineChangchunJilinChina
| | - Zheng Ruan
- Department of Traditional Chinese Medicine964th HospitalChangchunChina
| | - Shaohui Yu
- Department of GynaecologyThe Affiliated Hospital to Changchun University of Chinese MedicineChangchunJilinChina
| |
Collapse
|
22
|
Kumari N, Kumari R, Dua A, Singh M, Kumar R, Singh P, Duyar-Ayerdi S, Pradeep S, Ojesina AI, Kumar R. From Gut to Hormones: Unraveling the Role of Gut Microbiota in (Phyto)Estrogen Modulation in Health and Disease. Mol Nutr Food Res 2024; 68:e2300688. [PMID: 38342595 DOI: 10.1002/mnfr.202300688] [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: 09/24/2023] [Revised: 12/28/2023] [Indexed: 02/13/2024]
Abstract
The human gut microbiota regulates estrogen metabolism through the "estrobolome," the collection of bacterial genes that encode enzymes like β-glucuronidases and β-glucosidases. These enzymes deconjugate and reactivate estrogen, influencing circulating levels. The estrobolome mediates the enterohepatic circulation and bioavailability of estrogen. Alterations in gut microbiota composition and estrobolome function have been associated with estrogen-related diseases like breast cancer, enometrial cancer, and polycystic ovarian syndrome (PCOS). This is likely due to dysregulated estrogen signaling partly contributed by the microbial impacts on estrogen metabolism. Dietary phytoestrogens also undergo bacterial metabolism into active metabolites like equol, which binds estrogen receptors and exhibits higher estrogenic potency than its precursor daidzein. However, the ability to produce equol varies across populations, depending on the presence of specific gut microbes. Characterizing the estrobolome and equol-producing genes across populations can provide microbiome-based biomarkers. Further research is needed to investigate specific components of the estrobolome, phytoestrogen-microbiota interactions, and mechanisms linking dysbiosis to estrogen-related pathology. However, current evidence suggests that the gut microbiota is an integral regulator of estrogen status with clinical relevance to women's health and hormonal disorders.
Collapse
Affiliation(s)
- Nikki Kumari
- Post-Graduate Department of Zoology, Magadh University, Bodh Gaya, Bihar, 824234, India
| | - Rashmi Kumari
- Department of Zoology, College of Commerce, Arts & Science, Patliputra University, Patna, Bihar, 800020, India
| | - Ankita Dua
- Department of Zoology, Shivaji College, University of Delhi, New Delhi, 110027, India
| | - Mona Singh
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Roushan Kumar
- Post-Graduate Department of Zoology, Magadh University, Bodh Gaya, Bihar, 824234, India
| | - Poonam Singh
- Post-Graduate Department of Zoology, Magadh University, Bodh Gaya, Bihar, 824234, India
| | - Susan Duyar-Ayerdi
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Sunila Pradeep
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Akinyemi I Ojesina
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Roshan Kumar
- Post-Graduate Department of Zoology, Magadh University, Bodh Gaya, Bihar, 824234, India
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| |
Collapse
|
23
|
Ziaei R, Shahshahan Z, Ghasemi‐Tehrani H, Heidari Z, Nehls MS, Ghiasvand R. Inulin-type fructans with different degrees of polymerization improve insulin resistance, metabolic parameters, and hormonal status in overweight and obese women with polycystic ovary syndrome: A randomized double-blind, placebo-controlled clinical trial. Food Sci Nutr 2024; 12:2016-2028. [PMID: 38455215 PMCID: PMC10916604 DOI: 10.1002/fsn3.3899] [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: 03/13/2023] [Revised: 11/23/2023] [Accepted: 11/25/2023] [Indexed: 03/09/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is associated with reproductive disorders and adverse cardiometabolic risk factors that can negatively impact the general health of women. Inulin-type fructans (ITFs) are proposed to beneficially affect risk factors associated with metabolic disorders. Whether ITFs can help with the management of PCOS by modifying insulin resistance (IR) and androgen levels has not yet been explored. The aim of this study was to investigate the effects of ITFs with different degrees of polymerization on insulin resistance, blood lipids, anthropometric measures, and hormonal status in overweight and obese women with PCOS. In a randomized double-blind placebo-controlled trial, seventy-five women with PCOS aged 18-40 years old were randomly assigned to receive 10 g/day of high-performance inulin (HPI) or oligofructose-enriched inulin (OEI) or maltodextrin for 12 weeks. Biochemical and clinical outcomes were measured at baseline and after the intervention. Participants in the HPI and OEI groups experienced improvements in waist circumference, total testosterone, free androgen index, sex hormone-binding globulin, and triglycerides compared to the placebo group. Also, the number of women with irregular menses or oligomenorrhoea decreased significantly in both ITF groups. Participants in the HPI group reported lower body mass, fasting insulin, and HOMA-IR, as well as a higher quantitative insulin sensitivity check index. ITF supplementation, especially with long-chain ITFs, when given for 12 weeks may improve metabolic outcomes, androgen status and clinical manifestations in women with PCOS.
Collapse
Affiliation(s)
- Rahele Ziaei
- Department of Community Nutrition, School of Nutrition and Food ScienceIsfahan University of Medical SciencesIsfahanIran
| | - Zahra Shahshahan
- Department of Obstetrics and Gynecology, School of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Hatav Ghasemi‐Tehrani
- Fertility Department, School of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Zahra Heidari
- Department of Biostatistics and Epidemiology, School of HealthIsfahan University of Medical SciencesIsfahanIran
| | - Marilyn S. Nehls
- Department of Kinesiology and Health PromotionUniversity of KentuckyLexingtonKentuckyUSA
| | - Reza Ghiasvand
- Department of Community Nutrition, School of Nutrition and Food ScienceIsfahan University of Medical SciencesIsfahanIran
| |
Collapse
|
24
|
Dong YH, Luo YH, Liu CJ, Huang WY, Feng L, Zou XY, Zhou JY, Li XR. Changes in microbial composition and interaction patterns of female urogenital tract and rectum in response to HPV infection. J Transl Med 2024; 22:125. [PMID: 38303030 PMCID: PMC10832222 DOI: 10.1186/s12967-024-04916-2] [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: 08/18/2023] [Accepted: 01/21/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Previous studies have shown that changes in the microbial community of the female urogenital tract are associated with Human papillomavirus (HPV) infection. However, research on this association was mostly focused on a single site, and there are currently few joint studies on HPV infection and multiple sites in the female urogenital tract. METHODS We selected 102 healthy women from Yunnan Province as the research object, collected cervical exfoliation fluid, vaginal, urethral, and rectal swabs for microbial community analysis, and measured bacterial load, and related cytokine content. The link between HPV, microbiota, and inflammation was comprehensively evaluated using bioinformatics methods. FINDINGS The impact of HPV infection on the microbial composition of different parts varies. We have identified several signature bacterial genera that respond to HPV infection in several detection sites, such as Corynebacterium, Lactobacillus, Campylobacter, and Cutibacterium have been detected in multiple sites, reflecting their potential significance in cross body sites HPV infection responses. There was a solid microbial interaction network between the cervix, vagina, and urethra. The interrelationships between inflammatory factors and different bacterial genera might also affect the immune system's response to HPV infection. INTERPRETATION It might be an effective strategy to prevent and treat HPV infection by simultaneously understanding the correlation between the microbial changes in multiple parts of the female urogenital tract and rectum and HPV infection, and controlling the microbial network related to HPV infection in different parts.
Collapse
Affiliation(s)
- Yong-Hong Dong
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
| | - Yu-Hua Luo
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
| | - Chen-Jian Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
| | - Wen-Yu Huang
- Changchun Institute of Biological Products Co., Ltd., Changchun, 130012, Jilin, China
| | - Lin Feng
- Guangdong Hybribio Biotech Co., Ltd., Chaozhou, 521000, Guangdong, China
| | - Xing-Yuan Zou
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
| | - Jin-Yan Zhou
- Pediatrics Department, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, 650051, Yunnan, China.
| | - Xiao-Ran Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China.
| |
Collapse
|
25
|
Vine D, Ghosh M, Wang T, Bakal J. Increased Prevalence of Adverse Health Outcomes Across the Lifespan in Those Affected by Polycystic Ovary Syndrome: A Canadian Population Cohort. CJC Open 2024; 6:314-326. [PMID: 38487056 PMCID: PMC10935704 DOI: 10.1016/j.cjco.2023.12.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/11/2023] [Indexed: 03/17/2024] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is the most common metabolic-endocrine disorder impacting the health and quality of life of women over the lifespan. Evidence-based data on the scope of adverse health outcomes in those affected by PCOS is critical to improve healthcare and quality of life in this population. The aim of this study was to determine the prevalence of adverse health outcomes in those with PCOS compared to age-matched controls. Methods We conducted a retrospective observational case-control study in those diagnosed with PCOS and age-matched controls using the Alberta Health Services Health Analytics database and the International Classification of Diseases, for the period from 2002-2018 in Alberta, Canada. Results The cohort consisted of n = 16,531 exposed PCOS cases and n = 49,335 age-matched un-exposed controls. The prevalences of hypertension, renal disease, gastrointestinal disease, eating disorders, mental illness, depression-anxiety, rheumatoid arthritis, respiratory infections, and all malignancies were 20%-40% (P < 0.0001) higher in those with PCOS, compared to controls. The prevalence of obesity, dyslipidemia, nonalcoholic fatty liver disease, and type 2 diabetes was 2-3 fold higher in those with PCOS (P < 0.001). Cardiovascular, cerebrovascular, and peripheral vascular disease were 30%-50% higher, and they occurred 3-4 years earlier in those with PCOS (P < 0.0001); a 2-fold higher prevalence of dementia occurred in those with PCOS, compared to controls. Conclusion These findings provide evidence that PCOS is associated with a higher prevalence of morbidities over the lifespan, and the potential scope of the healthcare burden in women affected by PCOS.
Collapse
Affiliation(s)
- Donna Vine
- Metabolic and Cardiovascular Disease Laboratory, University of Alberta, Edmonton, Alberta, Canada
| | - Mahua Ghosh
- Division of Endocrinology and Metabolism, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ting Wang
- Alberta Strategy for Patient Orientated Research, Provincial Research Data Services, Alberta Health Services, Edmonton, Alberta, Canada
| | - Jeffrey Bakal
- Alberta Strategy for Patient Orientated Research, Provincial Research Data Services, Alberta Health Services, Edmonton, Alberta, Canada
| |
Collapse
|
26
|
Liu X, Liu J, Zhang T, Wang Q, Zhang H. Complex relationship between gut microbiota and thyroid dysfunction: a bidirectional two-sample Mendelian randomization study. Front Endocrinol (Lausanne) 2023; 14:1267383. [PMID: 38027113 PMCID: PMC10667917 DOI: 10.3389/fendo.2023.1267383] [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: 07/26/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Background Many studies have reported the link between gut microbiota and thyroid dysfunction. However, the causal effect of gut microbiota on thyroid dysfunction and the changes in gut microbiota after the onset of thyroid dysfunction are not clear. Methods A two-sample Mendelian randomization (MR) study was used to explore the complex relationship between gut microbiota and thyroid dysfunction. Data on 211 bacterial taxa were obtained from the MiBioGen consortium, and data on thyroid dysfunction, including hypothyroidism, thyroid-stimulating hormone alteration, thyroxine deficiency, and thyroid peroxidase antibodies positivity, were derived from several databases. Inverse variance weighting (IVW), weighted median, MR-Egger, weighted mode, and simple mode were applied to assess the causal effects of gut microbiota on thyroid dysfunction. Comprehensive sensitivity analyses were followed to validate the robustness of the results. Finally, a reverse MR study was conducted to explore the alteration of gut microbiota after hypothyroidism onset. Results Our bidirectional two-sample MR study revealed that the genera Intestinimonas, Eubacterium brachy group, Ruminiclostridium5, and Ruminococcaceae UCG004 were the risk factors for decreased thyroid function, whereas the genera Bifidobacterium and Lachnospiraceae UCG008 and phyla Actinobacteria and Verrucomicrobia were protective. The abundance of eight bacterial taxa varied after the onset of hypothyroidism. Sensitivity analysis showed that no heterogeneity or pleiotropy existed in the results of this study. Conclusion This novel MR study systematically demonstrated the complex relationship between gut microbiota and thyroid dysfunction, which supports the selection of more targeted probiotics to maintain thyroid-gut axis homeostasis and thus to prevent, control, and reverse the development of thyroid dysfunction.
Collapse
Affiliation(s)
| | | | | | - Qian Wang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Huawei Zhang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
27
|
Alsobaie S, Alageel AA, Ishfaq T, Ali Khan I, Alharbi KK. Examining the Genetic Role of rs8192675 Variant in Saudi Women Diagnosed with Polycystic Ovary Syndrome. Diagnostics (Basel) 2023; 13:3214. [PMID: 37892034 PMCID: PMC10606196 DOI: 10.3390/diagnostics13203214] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Polycystic ovary syndrome is a complex disorder defined by the Rotterdam criteria. Insulin resistance is a common factor for the development of type 2 diabetes mellitus among women with PCOS. The SLC2A2 gene has been identified as a T2DM gene by genome-wide association studies in the rs8192675 SNP. This study aimed to investigate the rs8192675 SNP in women diagnosed with PCOS on a molecular level and further for T2DM development in the Saudi women. In this case-control study, 100 PCOS women and 100 healthy controls were selected. Among 100 PCOS women, 28 women showed T2DM development. Genotyping for rs8192675 SNP was performed by PCR-RFLP analysis. Additionally, Sanger sequencing was performed to validate the RFLP analysis. The obtained data were used for a statistical analysis for the genotype and allele frequencies, logistic regression, and ANOVA analysis. The clinical data confirmed the positive association between FBG, FI, FSH, TT, TC, HDLc, LDLc, and family histories (p < 0.05). HWE analysis was associated in both the PCOS cases and the control individuals. Genotype and allele frequencies were associated in PCOS women and strongly associated with women with PCOS who developed T2DM (p < 0.05). No association was found in the logistic regression model or ANOVA analysis studied in women with PCOS (p > 0.05). A strong association was observed between the rs8192675 SNP and women with PCOS who developed T2DM using ANOVA analysis (p < 0.05). This study confirms that the rs8192675 SNP is associated with women with PCOS and strongly associated with women with PCOS with developed T2DM in Saudi Arabia.
Collapse
Affiliation(s)
- Sarah Alsobaie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia; (S.A.); (A.A.A.); (K.K.A.)
| | - Arwa A. Alageel
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia; (S.A.); (A.A.A.); (K.K.A.)
| | - Tahira Ishfaq
- Department of Obstetrics and Gynecology, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia;
| | - Imran Ali Khan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia; (S.A.); (A.A.A.); (K.K.A.)
| | - Khalid Khalaf Alharbi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia; (S.A.); (A.A.A.); (K.K.A.)
| |
Collapse
|
28
|
Canha-Gouveia A, Di Nisio V, Salumets A, Damdimopoulou P, Coy P, Altmäe S, Sola-Leyva A. The Upper Reproductive System Microbiome: Evidence beyond the Uterus. Semin Reprod Med 2023; 41:190-199. [PMID: 38320577 DOI: 10.1055/s-0043-1778056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
The microbiome of the female upper reproductive system has garnered increasing recognition and has become an area of interest in the study of women's health. This intricate ecosystem encompasses a diverse consortium of microorganisms (i.e., microbiota) and their genomes (i.e., microbiome) residing in the female upper reproductive system, including the uterus, the fallopian tubes, and ovaries. In recent years, remarkable advancements have been witnessed in sequencing technologies and microbiome research, indicating the potential importance of the microbial composition within these anatomical sites and its impact in women's reproductive health and overall well-being. Understanding the composition, dynamics, and functions of the microbiome of the female upper reproductive system opens up exciting avenues for improving fertility, treating gynecological conditions, and advancing our comprehension of the intricate interplay between the microbiome and the female reproductive system. The aim of this study is to compile currently available information on the microbial composition of the female upper reproductive system in humans, with a focus beyond the uterus, which has received more attention in recent microbiome studies compared with the fallopian tubes and ovaries. In conclusion, this review underscores the potential role of this microbiome in women's physiology, both in health and disease.
Collapse
Affiliation(s)
- Analuce Canha-Gouveia
- Department of Physiology, Faculty of Veterinary, University of Murcia, Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital "Virgen de la Arrixaca," Murcia, Spain
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Valentina Di Nisio
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Andres Salumets
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Pauliina Damdimopoulou
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Pilar Coy
- Department of Physiology, Faculty of Veterinary, University of Murcia, Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital "Virgen de la Arrixaca," Murcia, Spain
| | - Signe Altmäe
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Alberto Sola-Leyva
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden
- Competence Centre on Health Technologies, Tartu, Estonia
| |
Collapse
|
29
|
Schweighofer N, Strasser M, Obermayer A, Trummer O, Sourij H, Sourij C, Obermayer-Pietsch B. Identification of Novel Intronic SNPs in Transporter Genes Associated with Metformin Side Effects. Genes (Basel) 2023; 14:1609. [PMID: 37628660 PMCID: PMC10454417 DOI: 10.3390/genes14081609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Metformin is a widely used and effective medication in type 2 diabetes (T2DM) as well as in polycystic ovary syndrome (PCOS). Single nucleotide polymorphisms (SNPs) contribute to the occurrence of metformin side effects. The aim of the present study was to identify intronic genetic variants modifying the occurrence of metformin side effects and to replicate them in individuals with T2DM and in women with PCOS. We performed Next Generation Sequencing (Illumina Next Seq) of 115 SNPs in a discovery cohort of 120 metformin users and conducted a systematic literature review. Selected SNPs were analysed in two independent cohorts of individuals with either T2DM or PCOS, using 5'-3'exonucleaseassay. A total of 14 SNPs in the organic cation transporters (OCTs) showed associations with side effects in an unadjusted binary logistic regression model, with eight SNPs remaining significantly associated after appropriate adjustment in the discovery cohort. Five SNPs were confirmed in a combined analysis of both replication cohorts but showed different association patterns in subgroup analyses. In an unweighted polygenic risk score (PRS), the risk for metformin side effects increased with the number of risk alleles. Intronic SNPs in the OCT cluster contribute to the development of metformin side effects in individuals with T2DM and in women with PCOS and are therefore of interest for personalized therapy options.
Collapse
Affiliation(s)
- Natascha Schweighofer
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (N.S.); (M.S.); (A.O.); (H.S.); barbar (B.O.-P.)
- Center for Biomarker Research in Medicine, CBmed, 8010 Graz, Austria
| | - Moritz Strasser
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (N.S.); (M.S.); (A.O.); (H.S.); barbar (B.O.-P.)
- Department of Health Studies, Institute of Biomedical, FH Joanneum University of Applied Sciences, 8020 Graz, Austria
| | - Anna Obermayer
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (N.S.); (M.S.); (A.O.); (H.S.); barbar (B.O.-P.)
- Interdisciplinary Metabolic Medicine Trials Unit, Medical University of Graz, 8036 Graz, Austria
| | - Olivia Trummer
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (N.S.); (M.S.); (A.O.); (H.S.); barbar (B.O.-P.)
| | - Harald Sourij
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (N.S.); (M.S.); (A.O.); (H.S.); barbar (B.O.-P.)
- Interdisciplinary Metabolic Medicine Trials Unit, Medical University of Graz, 8036 Graz, Austria
| | - Caren Sourij
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria;
| | - Barbara Obermayer-Pietsch
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (N.S.); (M.S.); (A.O.); (H.S.); barbar (B.O.-P.)
| |
Collapse
|
30
|
Batóg G, Dołoto A, Bąk E, Piątkowska-Chmiel I, Krawiec P, Pac-Kożuchowska E, Herbet M. The interplay of oxidative stress and immune dysfunction in Hashimoto's thyroiditis and polycystic ovary syndrome: a comprehensive review. Front Immunol 2023; 14:1211231. [PMID: 37588599 PMCID: PMC10426741 DOI: 10.3389/fimmu.2023.1211231] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/10/2023] [Indexed: 08/18/2023] Open
Abstract
In recent years, there has been a significant increase in the concomitant incidence of Hashimoto's thyroiditis (HT) and polycystic ovary syndrome (PCOS), both in terms of incidence, etiology, and clinical consequences. PCOS patients suffering from autoimmune thyroid diseases show insulin resistance, impaired glucose tolerance, weight gain, and metabolic and reproductive complications. Studies have shown that chronic stress and its consequence, i.e. oxidative stress, play an important role in the pathomechanism of both disorders. It has also been shown that long-term exposure to stress triggers biological mechanisms, in particular related to the regulation of the inflammatory cascade, which plays a key role in autoimmune diseases. The paper is a review of the literature on the role of chronic stress, oxidative stress, and immune processes in the pathogenesis of HT and PCOS. In addition, the review is a source of knowledge about the treatment of these diseases, and in particular the use of antioxidants in therapeutic management.
Collapse
Affiliation(s)
- Gabriela Batóg
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Anna Dołoto
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Ewelina Bąk
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Iwona Piątkowska-Chmiel
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Paulina Krawiec
- Department of Paediatrics and Gastroenterology, Medical University of Lublin, Lublin, Poland
| | | | - Mariola Herbet
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
31
|
Chiantera V, Laganà AS, Basciani S, Nordio M, Bizzarri M. A Critical Perspective on the Supplementation of Akkermansia muciniphila: Benefits and Harms. Life (Basel) 2023; 13:1247. [PMID: 37374030 DOI: 10.3390/life13061247] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Akkermansia muciniphila is a mucin-degrading bacterium of the intestinal niche, exerting beneficial effects on the host metabolic profile. Accumulating evidence indicated Akkermansia as a promising therapeutic probiotic against metabolic disorders such as obesity, type 2 diabetes and cardiovascular diseases. However, in specific intestinal microenvironments, its excessive enrichment may be not beneficial. Conditions like inflammatory bowel disease (IBD), Salmonella typhimurium infection or post-antibiotic reconstitution may not benefit from Akkermansia supplementation. Furthermore, using Akkermansia in patients with endocrine and gynecological disorders-such as polycystic ovary syndrome (PCOS) or endometriosis-that have a higher risk of developing IBD, should be critically evaluated. In addition, a cautionary note comes from the neurological field, as the gut microbiota of patients suffering from Parkinson's disease or multiple sclerosis exhibits a characteristic signature of Akkermansia municiphila abundance. Overall, considering these controversial points, the use of Akkermansia should be evaluated on an individual basis, avoiding risking unexpected effects.
Collapse
Affiliation(s)
- Vito Chiantera
- Unit of Gynecologic Oncology, ARNAS "Civico-Di Cristina-Benfratelli", Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Antonio Simone Laganà
- Unit of Gynecologic Oncology, ARNAS "Civico-Di Cristina-Benfratelli", Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Sabrina Basciani
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, 00161 Rome, Italy
| | - Maurizio Nordio
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Mariano Bizzarri
- System Biology Group Laboratory, Sapienza University, 00161 Rome, Italy
| |
Collapse
|
32
|
Parker J. Pathophysiological Effects of Contemporary Lifestyle on Evolutionary-Conserved Survival Mechanisms in Polycystic Ovary Syndrome. Life (Basel) 2023; 13:life13041056. [PMID: 37109585 PMCID: PMC10145572 DOI: 10.3390/life13041056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is increasingly being characterized as an evolutionary mismatch disorder that presents with a complex mixture of metabolic and endocrine symptoms. The Evolutionary Model proposes that PCOS arises from a collection of inherited polymorphisms that have been consistently demonstrated in a variety of ethnic groups and races. In utero developmental programming of susceptible genomic variants are thought to predispose the offspring to develop PCOS. Postnatal exposure to lifestyle and environmental risk factors results in epigenetic activation of developmentally programmed genes and disturbance of the hallmarks of health. The resulting pathophysiological changes represent the consequences of poor-quality diet, sedentary behaviour, endocrine disrupting chemicals, stress, circadian disruption, and other lifestyle factors. Emerging evidence suggests that lifestyle-induced gastrointestinal dysbiosis plays a central role in the pathogenesis of PCOS. Lifestyle and environmental exposures initiate changes that result in disturbance of the gastrointestinal microbiome (dysbiosis), immune dysregulation (chronic inflammation), altered metabolism (insulin resistance), endocrine and reproductive imbalance (hyperandrogenism), and central nervous system dysfunction (neuroendocrine and autonomic nervous system). PCOS can be a progressive metabolic condition that leads to obesity, gestational diabetes, type two diabetes, metabolic-associated fatty liver disease, metabolic syndrome, cardiovascular disease, and cancer. This review explores the mechanisms that underpin the evolutionary mismatch between ancient survival pathways and contemporary lifestyle factors involved in the pathogenesis and pathophysiology of PCOS.
Collapse
Affiliation(s)
- Jim Parker
- School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|