1
|
Guo D, Wang R. Association between visceral adipose surrogates and infertility among reproductive-aged females: a cross-sectional study. Front Endocrinol (Lausanne) 2024; 15:1488309. [PMID: 39726843 PMCID: PMC11669517 DOI: 10.3389/fendo.2024.1488309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Background Visceral adipose tissue (VAT) exerts a substantial influence on female infertility. Nevertheless, the relationship between VAT surrogates and female infertility remains ambiguous. Methods This study employed a cross-sectional design and analyzed data from the National Health and Nutrition Examination Survey (NHANES, 2012-2016). Weighted logistic regression models were utilized to examine the association between VAT surrogates and infertility. Furthermore, Receiver Operating Characteristic (ROC) curve analysis was conducted to assess the diagnostic efficacy of these surrogates for infertility. Results Individuals experiencing infertility exhibited markedly elevated levels of the Chinese Visceral Adiposity Index (CVAI) (108.30 vs. 69.86, P<0.001) and Visceral Adiposity Index (VAI) (1.68 vs. 1.35, P<0.001). When considered as a continuous variable, CVAI (odds ratio [OR]: 1.06, 95% confidence interval [CI]: 1.03-1.09, P<0.001), rather than VAI (OR:1.02, 95%CI: 0.98-1.06, P=0.259), demonstrated a significant association with the risk of female infertility. Consistent findings were also evident after dividing participants into 4 subgroups based on CVAI quartiles. Additionally, ROC curves indicated that CVAI exhibited the most robust diagnostic value for female infertility compared to other indices. Subgroup analyses revealed a robust association between CVAI and infertility across different populations. Conclusion Females with elevated CVAI levels faced a significantly heightened risk of infertility in the United States. CVAI holds promise as a valuable tool for stratifying the risk of infertility.
Collapse
Affiliation(s)
- Dongli Guo
- Department of Physiological Obstetrics, Women and Children’s Hospital, Zhumadian Central Hospital, Zhumadian, Henan, China
| | | |
Collapse
|
2
|
Liu F, Tian L, Zhang Y, Deng W, Xu X, Zou Y, An R. DIA proteomic and PRM validation through human granulose cells profiles screen suitable biomarkers for polycystic ovary syndrome patients. J Proteomics 2024; 309:105332. [PMID: 39424224 DOI: 10.1016/j.jprot.2024.105332] [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: 06/14/2024] [Revised: 10/12/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
The aim of this study is to identify differentially expressed proteins (DEPs) in granulose cells (GCs) from women with or withoutpolycystic ovary syndrome (PCOS) via data independent acquisition (DIA) proteomic analysis.A total of 63 women were recruited for this study, 34 PCOS patients as experimental group (P), and 29 women without PCOS as Normal group (NP). DIA-based proteomic analysis was performed to identify DEPs in GCs between the P and NP samples. Certain typical DEPs were further validated by Parallel reaction monitoring (PRM), and correlation analysis was performed between these DEPs and the clinical characteristics.Cell vitality was measured by CCK-8 assay. DIA analysis revealed 174 significantly DEPs, of which 7 were upregulated and 167 downregulated. Bioinformatics analysis was performed to analysis the significantly DEPs. The PRM experiment confirmed TOP2A and SPHKAP were upregulated significantly in P by comparing to NP, while GM2A, MRPS16, APOA2 and FGF2 were downregulated significantly. Most notably, Correlation analysis revealed that TOP2A, SPHKAP, MRPS16 and FGF2were positively correlated with TG, AMH and Age, but negatively correlated with Menarche age, DBIL, FT3, Basal serum FSH and LH.Meanwhile, CCK-8 assay has shown that downregulation of FGF2 could weaken cell viability. Finally, a panel of DEPs were identified in the GCs of patients with PCOS, of which certain significant DEPs might play essential roles in the pathogenesis of PCOS, could be regarded as candidate biomarkers for PCOS.
Collapse
Affiliation(s)
- Faying Liu
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Reproductive Health, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China
| | - Lifeng Tian
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China
| | - Ying Zhang
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China
| | - Wei Deng
- Department of Obstetrics and Gynecology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321099, China
| | - Xiaoyun Xu
- Department of Quality Control, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China
| | - Yang Zou
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Reproductive Health, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China
| | - Ruifang An
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
3
|
Oteri V, Galeano F, Panebianco S, Piticchio T, Le Moli R, Frittitta L, Vella V, Baratta R, Gullo D, Frasca F, Tumminia A. Influence of Mediterranean Diet on Sexual Function in People with Metabolic Syndrome: A Narrative Review. Nutrients 2024; 16:3397. [PMID: 39408364 PMCID: PMC11479179 DOI: 10.3390/nu16193397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 10/20/2024] Open
Abstract
Metabolic syndrome (MS), a cluster of cardiometabolic disorders, and sexual dysfunction are two conditions that impact a large proportion of the general population. Although they can occur independently, they are frequently linked and significantly affect people's quality of life. In recent years, research has increasingly focused on the importance of diet, particularly the Mediterranean diet (MD), in modulating sexual function due to its anti-inflammatory, antioxidant, and vasodilatory properties. In this narrative review, we examined the relationship between MS and sexual function in both men and women, with a special emphasis on the MD's therapeutic efficacy in improving sexual dysfunction. In men, MD has been shown to ameliorate erectile dysfunction, as well as several sperm parameters, perhaps leading to improved fertility. On the other hand, adherence to MD has been demonstrated to partially recover several sexual dysfunctions in women, such as those related to their menstrual cycle, menopause, endometriosis, and polycystic ovary syndrome. These favorable effects of MD have been demonstrated in both sexes also among people affected by MS. However, more targeted studies are needed to validate these data for different dietary approaches as well.
Collapse
Affiliation(s)
- Vittorio Oteri
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, CT, Italy; (V.O.); (F.G.); (S.P.); (T.P.); (R.L.M.); (L.F.); (V.V.)
| | - Francesco Galeano
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, CT, Italy; (V.O.); (F.G.); (S.P.); (T.P.); (R.L.M.); (L.F.); (V.V.)
| | - Stefania Panebianco
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, CT, Italy; (V.O.); (F.G.); (S.P.); (T.P.); (R.L.M.); (L.F.); (V.V.)
| | - Tommaso Piticchio
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, CT, Italy; (V.O.); (F.G.); (S.P.); (T.P.); (R.L.M.); (L.F.); (V.V.)
- Department of Medicine and Surgery, University Kore of Enna, 94100 Enna, EN, Italy
| | - Rosario Le Moli
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, CT, Italy; (V.O.); (F.G.); (S.P.); (T.P.); (R.L.M.); (L.F.); (V.V.)
- Department of Medicine and Surgery, University Kore of Enna, 94100 Enna, EN, Italy
| | - Lucia Frittitta
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, CT, Italy; (V.O.); (F.G.); (S.P.); (T.P.); (R.L.M.); (L.F.); (V.V.)
- Diabetes and Obesity Center, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, CT, Italy
| | - Veronica Vella
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, CT, Italy; (V.O.); (F.G.); (S.P.); (T.P.); (R.L.M.); (L.F.); (V.V.)
| | - Roberto Baratta
- Endocrine Unit, Garibaldi-Nesima Hospital, 95122 Catania, CT, Italy; (R.B.); (D.G.); (A.T.)
| | - Damiano Gullo
- Endocrine Unit, Garibaldi-Nesima Hospital, 95122 Catania, CT, Italy; (R.B.); (D.G.); (A.T.)
| | - Francesco Frasca
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, CT, Italy; (V.O.); (F.G.); (S.P.); (T.P.); (R.L.M.); (L.F.); (V.V.)
- Endocrine Unit, Garibaldi-Nesima Hospital, 95122 Catania, CT, Italy; (R.B.); (D.G.); (A.T.)
| | - Andrea Tumminia
- Endocrine Unit, Garibaldi-Nesima Hospital, 95122 Catania, CT, Italy; (R.B.); (D.G.); (A.T.)
| |
Collapse
|
4
|
Kordowitzki P, Graczyk S, Haghani A, Klutstein M. Oocyte Aging: A Multifactorial Phenomenon in A Unique Cell. Aging Dis 2024; 15:5-21. [PMID: 37307833 PMCID: PMC10796106 DOI: 10.14336/ad.2023.0527] [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: 05/06/2023] [Accepted: 05/27/2023] [Indexed: 06/14/2023] Open
Abstract
The oocyte is considered to be the largest cell in mammalian species. Women hoping to become pregnant face a ticking biological clock. This is becoming increasingly challenging as an increase in life expectancy is accompanied by the tendency to conceive at older ages. With advancing maternal age, the fertilized egg will exhibit lower quality and developmental competence, which contributes to increased chances of miscarriage due to several causes such as aneuploidy, oxidative stress, epigenetics, or metabolic disorders. In particular, heterochromatin in oocytes and with it, the DNA methylation landscape undergoes changes. Further, obesity is a well-known and ever-increasing global problem as it is associated with several metabolic disorders. More importantly, both obesity and aging negatively affect female reproduction. However, among women, there is immense variability in age-related decline of oocytes' quantity, developmental competence, or quality. Herein, the relevance of obesity and DNA-methylation will be discussed as these aspects have a tremendous effect on female fertility, and it is a topic of continuous and widespread interest that has yet to be fully addressed for the mammalian oocyte.
Collapse
Affiliation(s)
- Pawel Kordowitzki
- Department of Preclinical and Basic Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Torun, Poland.
| | - Szymon Graczyk
- Department of Preclinical and Basic Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Torun, Poland.
| | - Amin Haghani
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Altos Labs, San Diego, CA, USA.
| | - Michael Klutstein
- Institute of Biomedical and Oral Research, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
5
|
Chen MJ, Chou CH, Hsiao TH, Wu TY, Li CY, Chen YL, Chao KH, Lee TH, Gicana RG, Shih CJ, Brandon-Mong GJ, Lai YL, Li PT, Tseng YL, Wang PH, Chiang YR. Clostridium innocuum, an opportunistic gut pathogen, inactivates host gut progesterone and arrests ovarian follicular development. Gut Microbes 2024; 16:2424911. [PMID: 39508647 PMCID: PMC11545266 DOI: 10.1080/19490976.2024.2424911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/04/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
\Levels of progesterone, an endogenous female hormone, increase after ovulation; progesterone is crucial in the luteal phase to maintain successful pregnancy and prevent early miscarriage. Both endogenous and exogenous progesterone are recycled between the liver and gut; thus, the gut microbiota regulate host progesterone levels by inhibiting enterohepatic progesterone circulation. Our data indicated Clostridium innocuum as a major species involved in gut progesterone metabolism in women with infertility. C. innocuum converts progesterone into the neurosteroid epipregnanolone (with negligible progestogenic activity). We purified and characterized the corresponding enzyme, namely NADPH-dependent 5β-dihydroprogesterone reductase, which is highly oxygen sensitive and whose corresponding genes are prevalent in C. innocuum. Moreover, C. innocuum-administered female C57BL/6 mice (aged 7 weeks) exhibited decreased plasma progesterone levels (~35%). Clostridium-specific antibiotics (metronidazole) restored low plasma progesterone levels in these mice. Furthermore, prolonged C. innocuum administration (12 weeks) arrested ovarian follicular development in female mice. Cytological and histological analyses indicated that C. innocuum may cause luteal phase insufficiency and affect menstrual regularity. Our findings suggest C. innocuum as a causal factor of progesterone resistance in women taking progesterone.
Collapse
Affiliation(s)
- Mei-Jou Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- Livia Shan-Yu Wan Chair Professor of Obstetrics and Gynecology, National Taiwan University, Taipei, Taiwan
| | - Chia-Hung Chou
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tsun-Hsien Hsiao
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Tien-Yu Wu
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Chi-Ying Li
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Yi-Lung Chen
- Department of Microbiology, Soochow University, Taipei, Taiwan
- Department of Agricultural Chemistry, National Taiwan University, Taipei, Taiwan
| | - Kuang-Han Chao
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzong-Huei Lee
- Institute of Fisheries Science, National Taiwan University, Taipei, Taiwan
| | | | - Chao-Jen Shih
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan
| | | | - Yi-Li Lai
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Po-Ting Li
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Lin Tseng
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Hsiang Wang
- Graduate Institute of Environmental Engineering, National Central University, Taoyuan, Taiwan
- Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan
| | - Yin-Ru Chiang
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
- Department of Agricultural Chemistry, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
6
|
Wang Y, Du YY, Yao W, Deng TR, Guo N, Yin L, Yuan XQ, Guo QC, Li J, Liao HM, Qin DY, Li YF. Associations between phthalate metabolites and cytokines in the follicular fluid of women undergoing in vitro fertilization. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 267:115616. [PMID: 37871386 DOI: 10.1016/j.ecoenv.2023.115616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
Many studies have showed that phthalates have reproductive and embryonic toxicity, while the potential mechanisms are mostly unknown. Inflammation may play a mediating part in phthalate exposure and adverse reproductive endpoints. A cross-sectional survey was conducted to investigate the associations of phthalate metabolites with inflammatory cytokines in the follicular fluid (FF) of women undergoing in vitro fertilization (IVF). We determined the levels of eight phthalate metabolites and five cytokines in the FF of 76 women, including interleukin (IL)- 6, IL-8, IL-10, monocyte chemoattractant protein-1 (MCP-1) and tumor necrosis factor-α (TNF-α). The associations of individual phthalate exposure with cytokines in FF samples were explored by multiple linear regression. We further evaluated the combined effects of multiple phthalate exposures on FF levels of cytokines by using Bayesian kernel machine regression (BKMR) models. We found that there was a positive relationship between mono-ethyl phthalate (MEP) and IL-6 in the FF (percent change:12.4%; 95% CI: 1.3%, 24.9%). In contrast, elevated mono-benzyl phthalate (MBzP), mono(2-ethylhexyl) phthalate (MEHP) and %MEHP levels were associated with decreased MCP-1. In the BKMR models, phthalate metabolite mixtures were positively associated with TNF-α when the mixtures were lower than 65th percentile compared with their medians. In the stratified analyses, MEHP was inversely associated with MCP-1 among women with BMI ≥ 23 kg/m2 (test for interaction <0.05). Our results suggest that certain phthalate metabolites or their mixtures may alter levels of inflammatory cytokines in the FF, and further research is necessary to elucidate the mechanisms underlying the relationship between phthalates exposure, ovarian dysfunction and adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Yi Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yao-Yao Du
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wen Yao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao-Ran Deng
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Na Guo
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li Yin
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiao-Qiong Yuan
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qing-Chun Guo
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Juan Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hong-Mei Liao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dan-Yu Qin
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu-Feng Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
7
|
Ramadan AG, Abdel-Rehim WM, El-Tahan RA, Elblehi SS, Kamel MA, Shaker SA. Maternal and paternal obesity differentially reprogram the ovarian mitochondrial biogenesis of F1 female rats. Sci Rep 2023; 13:15480. [PMID: 37726284 PMCID: PMC10509203 DOI: 10.1038/s41598-023-42468-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/11/2023] [Indexed: 09/21/2023] Open
Abstract
Obesity has harmful consequences on reproductive outcomes and the rapid increase in obesity is assumed to be influenced by epigenetics and trans-generation effects. Our study aimed to explore the effect of maternal and/or paternal obesity on the ovarian tissues of the first-generation female offspring in rats. The study was conducted on 40 adult Wistar albino rats (20 males and 20 females). Obesity was induced by feeding them an obesogenic diet for 3 months. The pregnancy was induced in the females by mating with males in four combinations: healthy mother with healthy father (control parents, CP), healthy mother with obese fathers (OF), obese mothers with healthy father (OM), and obese mother with obese father (obese parents, OP). After delivery, the female offspring at two months were sacrificed, and the blood and ovarian tissues were collected to assess the studied parameters. Our result showed differential impacts of maternal and paternal obesity on the ovarian health of the female offspring. The female offspring of obese OM or OP showed early signs of obesity. These metabolic abnormalities were associated with signs of ovarian lesions, impaired folliculogenesis, and decreased oocyte quality and also showed significant alterations in mitochondrial biogenesis, redox status, inflammation, and microRNAs expression (miR-149 and miR-494). In conclusion, altered ovarian expression of microRNAs and associated impaired mitochondrial biogenesis pathways may be the root causes for the observed intergeneration transmission of the obesogenic phenotype.
Collapse
Affiliation(s)
- Amina G Ramadan
- Department of Biochemistry, Medical Research Institute, Alexandria University, 165 El-Horreya Avenue, EL-Hadara, POB: 21561, Alexandria, Egypt.
| | - Wafaa M Abdel-Rehim
- Department of Biochemistry, Medical Research Institute, Alexandria University, 165 El-Horreya Avenue, EL-Hadara, POB: 21561, Alexandria, Egypt
| | - Rasha A El-Tahan
- Department of Biochemistry, Medical Research Institute, Alexandria University, 165 El-Horreya Avenue, EL-Hadara, POB: 21561, Alexandria, Egypt
| | - Samar S Elblehi
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Maher A Kamel
- Department of Biochemistry, Medical Research Institute, Alexandria University, 165 El-Horreya Avenue, EL-Hadara, POB: 21561, Alexandria, Egypt
| | - Sara A Shaker
- Department of Biochemistry, Medical Research Institute, Alexandria University, 165 El-Horreya Avenue, EL-Hadara, POB: 21561, Alexandria, Egypt.
| |
Collapse
|
8
|
Velazquez C, Herrero Y, Bianchi MS, Cohen DJ, Cuasnicu P, Prost K, Marinoni R, Pascuali N, Parborell F, Abramovich D. Beneficial effects of metformin on mice female fertility after a high-fat diet intake. Mol Cell Endocrinol 2023; 575:111995. [PMID: 37364632 DOI: 10.1016/j.mce.2023.111995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/08/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023]
Abstract
Female fertility is highly dependent on energy balance. High fat diet (HFD) intake entails a risk of infertility and ovulatory disorders. Considering the increase in the prevalence of overweight and obesity over the last decades, it is crucial to understand the mechanisms involved in overweight-associated infertility. In this study, we evaluated the reproductive performance of female mice fed with a HFD and the effects of metformin administration on ovarian function in these mice. We hypothesized that one of the mechanisms involved in subfertility due to a HFD intake is the alteration of ovarian blood vessel formation. We found that mice fed with HFD had altered estrous cycles and steroidogenesis, increased ovarian fibrosis, fewer pups per litter and require more time to achieve pregnancy. HFD-fed mice also presented dysregulated ovarian angiogenesis and an increase in nuclear DNA damage in ovarian cells. Ovulation rates were lower in these animals, as evidenced both in natural mating and after ovulation induction with gonadotropins. Metformin ameliorated ovarian angiogenesis, improved steroidogenesis, fibrosis, and ovulation, decreased the time to pregnancy and increased litter sizes in HFD-fed mice. We conclude that ovarian angiogenesis is one of the mechanisms detrimentally affected by HFD intake. Since metformin could improve ovarian microvasculature, it may be an interesting strategy to study in women to shed light on new targets for patients with metabolic disturbances.
Collapse
Affiliation(s)
- Candela Velazquez
- Instituto de Biología y Medicina Experimental, IByME-CONICET, Vuelta de Obligado 2490, C1428ADL Ciudad Autónoma de Buenos Aires, Argentina
| | - Yamila Herrero
- Instituto de Biología y Medicina Experimental, IByME-CONICET, Vuelta de Obligado 2490, C1428ADL Ciudad Autónoma de Buenos Aires, Argentina
| | - María Silvia Bianchi
- Instituto de Biología y Medicina Experimental, IByME-CONICET, Vuelta de Obligado 2490, C1428ADL Ciudad Autónoma de Buenos Aires, Argentina
| | - Débora Juana Cohen
- Instituto de Biología y Medicina Experimental, IByME-CONICET, Vuelta de Obligado 2490, C1428ADL Ciudad Autónoma de Buenos Aires, Argentina
| | - Patricia Cuasnicu
- Instituto de Biología y Medicina Experimental, IByME-CONICET, Vuelta de Obligado 2490, C1428ADL Ciudad Autónoma de Buenos Aires, Argentina
| | - Katherine Prost
- Hospital Interzonal General de Agudos Pedro Fiorito, sector de Endocrinología, Av. Manuel Belgrano 827, B1870 Avellaneda, Provincia de Buenos Aires, Argentina
| | - Rocío Marinoni
- Instituto de Biología y Medicina Experimental, IByME-CONICET, Vuelta de Obligado 2490, C1428ADL Ciudad Autónoma de Buenos Aires, Argentina
| | - Natalia Pascuali
- Instituto de Biología y Medicina Experimental, IByME-CONICET, Vuelta de Obligado 2490, C1428ADL Ciudad Autónoma de Buenos Aires, Argentina; Department of Pathology, College of Medicine, University of Illinois at Chicago (UIC), Chicago, IL, United States
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental, IByME-CONICET, Vuelta de Obligado 2490, C1428ADL Ciudad Autónoma de Buenos Aires, Argentina
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental, IByME-CONICET, Vuelta de Obligado 2490, C1428ADL Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
9
|
The Implication of Mechanistic Approaches and the Role of the Microbiome in Polycystic Ovary Syndrome (PCOS): A Review. Metabolites 2023; 13:metabo13010129. [PMID: 36677054 PMCID: PMC9863528 DOI: 10.3390/metabo13010129] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
As a complex endocrine and metabolic condition, polycystic ovarian syndrome (PCOS) affects women's reproductive health. These common symptoms include hirsutism, hyperandrogenism, ovulatory dysfunction, irregular menstruation, and infertility. No one knows what causes it or how to stop it yet. Alterations in gut microbiota composition and disruptions in secondary bile acid production appear to play a causative role in developing PCOS. PCOS pathophysiology and phenotypes are tightly related to both enteric and vaginal bacteria. Patients with PCOS exhibit changed microbiome compositions and decreased microbial diversity. Intestinal microorganisms also alter PCOS patient phenotypes by upregulating or downregulating hormone release, gut-brain mediators, and metabolite synthesis. The human body's gut microbiota, also known as the "second genome," can interact with the environment to improve metabolic and immunological function. Inflammation is connected to PCOS and may be caused by dysbiosis in the gut microbiome. This review sheds light on the recently discovered connections between gut microbiota and insulin resistance (IR) and the potential mechanisms of PCOS. This study also describes metabolomic studies to obtain a clear view of PCOS and ways to tackle it.
Collapse
|
10
|
Di Berardino C, Peserico A, Capacchietti G, Zappacosta A, Bernabò N, Russo V, Mauro A, El Khatib M, Gonnella F, Konstantinidou F, Stuppia L, Gatta V, Barboni B. High-Fat Diet and Female Fertility across Lifespan: A Comparative Lesson from Mammal Models. Nutrients 2022; 14:nu14204341. [PMID: 36297035 PMCID: PMC9610022 DOI: 10.3390/nu14204341] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/18/2022] Open
Abstract
Female reproduction focuses mainly on achieving fully grown follicles and competent oocytes to be successfully fertilized, as well as on nourishing the developing offspring once pregnancy occurs. Current evidence demonstrates that obesity and/or high-fat diet regimes can perturbate these processes, leading to female infertility and transgenerational disorders. Since the mechanisms and reproductive processes involved are not yet fully clarified, the present review is designed as a systematic and comparative survey of the available literature. The available data demonstrate the adverse influences of obesity on diverse reproductive processes, such as folliculogenesis, oogenesis, and embryo development/implant. The negative reproductive impact may be attributed to a direct action on reproductive somatic and germinal compartments and/or to an indirect influence mediated by the endocrine, metabolic, and immune axis control systems. Overall, the present review highlights the fragmentation of the current information limiting the comprehension of the reproductive impact of a high-fat diet. Based on the incidence and prevalence of obesity in the Western countries, this topic becomes a research challenge to increase self-awareness of dietary reproductive risk to propose solid and rigorous preventive dietary regimes, as well as to develop targeted pharmacological interventions.
Collapse
Affiliation(s)
- Chiara Di Berardino
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Alessia Peserico
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Correspondence:
| | - Giulia Capacchietti
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Alex Zappacosta
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Nicola Bernabò
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council, A. Buzzati-Traverso Campus, via E. Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy
| | - Valentina Russo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Annunziata Mauro
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Mohammad El Khatib
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Francesca Gonnella
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Fani Konstantinidou
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Liborio Stuppia
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Valentina Gatta
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Barbara Barboni
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
11
|
Chen W, Pang Y. Metabolic Syndrome and PCOS: Pathogenesis and the Role of Metabolites. Metabolites 2021; 11:metabo11120869. [PMID: 34940628 PMCID: PMC8709086 DOI: 10.3390/metabo11120869] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/29/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrine diseases among women of reproductive age and is associated with many metabolic manifestations, such as obesity, insulin resistance (IR) and hyperandrogenism. The underlying pathogenesis of these metabolic symptoms has not yet been fully elucidated. With the application of metabolomics techniques, a variety of metabolite changes have been observed in the serum and follicular fluid (FF) of PCOS patients and animal models. Changes in metabolites result from the daily diet and occur during uncommon physiological routines. However, some of these metabolite changes may provide evidence to explain possible mechanisms and new approaches for prevention and therapy. This article reviews the pathogenesis of PCOS metabolic symptoms and the relationship between metabolites and the pathophysiology of PCOS. Furthermore, the potential clinical application of some specific metabolites will be discussed.
Collapse
Affiliation(s)
- Weixuan Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China;
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing 100191, China
| | - Yanli Pang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China;
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing 100191, China
- Correspondence:
| |
Collapse
|
12
|
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrinopathy in women of reproductive age. Although its essential clinical manifestation includes a plethora of symptoms and signs, which largely reflects the underlying hyperandrogenism, oligo/anovulation, and polycystic ovarian morphology, PCOS may also be associated with many metabolic derangements. These metabolic derangements happen to overlap with many of the core constituents of the metabolic syndrome (MBS)—increased insulin resistance, central obesity, and dyslipidemia. The two disorders also display similarly increased risks for certain metabolic and vascular diseases, such as type 2 diabetes mellitus, hypertension, and cardiovascular diseases. Due to the many similarities between metabolic syndrome and PCOS, this review aims to examine the evidence concerning the overlapping features, the risks for comorbidities, possible shared mechanisms, and treatment strategies in patients with coexisting PCOS and MBS.
Collapse
Affiliation(s)
- Yun-Chiao Hsieh
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Po-Kai Yang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Mei-Jou Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
- Livia ShangYu Wan Chair Professor of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
13
|
Wang N, Si C, Xia L, Wu X, Zhao S, Xu H, Ding Z, Niu Z. TRIB3 regulates FSHR expression in human granulosa cells under high levels of free fatty acids. Reprod Biol Endocrinol 2021; 19:139. [PMID: 34503515 PMCID: PMC8428109 DOI: 10.1186/s12958-021-00823-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/01/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Granulosa cells (GCs) in cumulus oophorus highly express follicle stimulating hormone receptor (FSHR), which is the most important mediator of both estradiol synthesis and oocyte maturation. Obese women have elevated free fatty acids (FFAs) levels in their follicular fluids and decreased FSHR expression in GCs, which is related to an altered protein kinase B/glycogen synthase kinase 3β (Akt/GSK3β) signaling pathway. Such FFA increases accompany 3-fold rises in pseudokinase 3 (TRIB3) expression and reduce the Akt phosphorylation status in both the human liver and in insulinoma cell lines. Therefore, in a high FFA environment, we determined if TRIB3 mediates regulation of FSHR via the Akt/GSK3β signaling pathway in human GCs. METHODS GCs from women undergoing in vitro fertilization were collected and designated as high and low FFAs cohorts based on their follicular fluid FFA content. GCs with low FFA levels and a human granulosa-like tumor (KGN) cell line were exposed to palmitic acid (PA), which is a dominate FFA follicular fluid constituent. The effects were assessed of this substitution on the Akt/GSK3β signaling pathway activity as well as the expressions of TRIB3 and FSHR at both the gene and protein levels by qPCR, Western blot and immunofluorescence staining analyses. Meanwhile, the individual effects of TRIB3 knockdown in KGN cells and p-AKT inhibitors were compared to determine the mechanisms of FFA-induced FSHR downregulation. RESULTS The average FSH dose consuming per oocyte (FSH dose/oocyte) was elevated and Top embryo quality ratio was decreased in women with high levels of FFAs in their follicular fluid. In these women, the GC TRIB3 and ATF4 protein expression levels were upregulated which was accompanied by FSHR downregulation. Such upregulation was confirmed based on corresponding increases in their gene expression levels. On the other hand, the levels of p-Akt decreased while p-GSK3β increased in the GCs. Moreover, TRIB3 knockdown reversed declines in FSHR expression and estradiol (E2) production in KGN cells treated with PA, which also resulted in increased p-Akt levels and declines in the p-GSK3β level. In contrast, treatment of TRIB3-knockdown cells with an inhibitor of p-Akt (Ser473) resulted in rises in the levels of both p-GSK3β as well as FSHR expression whereas E2 synthesis fell. CONCLUSIONS During exposure to a high FFA content, TRIB3 can reduce FSHR expression through stimulation of the Akt/GSK3β pathway in human GCs. This response may contribute to inducing oocyte maturation.
Collapse
Affiliation(s)
- Nan Wang
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated with the Medical School of Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Chenchen Si
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated with the Medical School of Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Lan Xia
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated with the Medical School of Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Xian Wu
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated with the Medical School of Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Sheng Zhao
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated with the Medical School of Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Huihui Xu
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated with the Medical School of Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Zhide Ding
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Zhihong Niu
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated with the Medical School of Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|