1
|
Zheng X, Wu YJ, Wu LM, Zhang L, Zhang L, Jin Z, Gao F, Li QQ, Wang Y, Wu YD. Development and validation of a HPLC-MS/MS method the determination of genistein and equol in serum, urine and follicular fluid. J Pharm Biomed Anal 2025; 260:116800. [PMID: 40086052 DOI: 10.1016/j.jpba.2025.116800] [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: 10/16/2024] [Revised: 02/17/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Soy isoflavones exert estrogen-like synergistic or antagonistic effects by binding to estrogen receptors, and potentially impact the function of female reproductive system, but their distribution profile in human remains little clarified. To determination of genistein (GEN) and equol (EQ) in human urine, serum and follicular fluid (FF), an analytical method based on high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) was developed and validated. The enrichment and clean-up are performed on a solid-phase extraction (SPE) column; the elution is a gradient one, with the mobile phase (A) of 0.1 % (v/v) formic acid aqueous solution and the mobile phase (B) of 0.1 % (v/v) formic acid in acetonitrile; the column temperature is 40 °C. Mass spectrometry is performed using negative ion mode electrospray ionization (ESI -) in multiple reaction monitoring (MRM) mode. The method was validated over the linear ranges of 7.8-1000.0 ng/mL and 39.1-5000.0 ng/mL, for serum and urine, with correlation coefficients (r) of 0.9948-0.9984. The precision, accuracy and stability meet the U.S. Food and Drug Administration guidance. This method has been used to detect genistein (GEN) and equol (EQ) in serum, follicular fluid, and urine, to report equol in follicular fluid for the first time, and to study the correlation between genistein and equol in three body fluids. The study showed that the average concentration of EQ in follicular fluid was 18.5 ng/mL and there was a significant positive Spearman's correlation between concentrations of GEN in serum and FF (r = 0.44, p ≤ 0.05).
Collapse
Affiliation(s)
- Xia Zheng
- Institute of Food Science and Engineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yue-Jin Wu
- Institute of Food Science and Engineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Li-Mei Wu
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Ling Zhang
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Lin Zhang
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Zhen Jin
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Fang Gao
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Qing-Qing Li
- Science and Research Department, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yin Wang
- Institute of Food Science and Engineering, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Yi-Dan Wu
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
2
|
Zhang Y, He C, He Y, Zhu Z. Follicular Fluid Metabolomics: Tool for Predicting IVF Outcomes of Different Infertility Causes. Reprod Sci 2025; 32:921-934. [PMID: 39090336 PMCID: PMC11978680 DOI: 10.1007/s43032-024-01664-y] [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: 03/16/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024]
Abstract
Infertility affects approximately 15% of couples at child-bearing ages and assisted reproductive technologies (ART), especially in vitro fertilization and embryo transfer (IVF-ET), provided infertile patients with an effective solution. The current paradox is that multiple embryo transfer that may leads to severe obstetric and perinatal complications seems to be the most valid measure to secure high success rate in the majority of clinic centers. Therefore, to avoid multiple transfer of embryos, it is urgent to explore biomarkers for IVF prognosis to select high-quality oocytes and embryos. Follicular fluid (FF), a typical biofluid constituted of the plasma effusion and granulosa-cell secretion, provides essential intracellular substances for oocytes maturation and its variation in composition reflects oocyte developmental competence and embryo viability. With the advances in metabolomics methodology, metabolomics, as an accurate and sensitive analyzing method, has been utilized to explore predictors in FF for ART success. Although FF metabolomics has provided a great possibility for screening markers with diagnostic and predictive value, its effectiveness is still doubted by some researchers. This may be resulted from the ignorance of the impact of sterility causes on the FF metabolomic profiles and thus its predictive ability might not be rightly illustrated. Therefore, in this review, we categorically demonstrate the study of FF metabolomics according to specific infertility causes, expecting to reveal the predicting value of metabolomics for IVF outcomes.
Collapse
Affiliation(s)
- Yijing Zhang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Chenyan He
- Sichuan Normal University, Chengdu, Sichuan, China
| | - Yuedong He
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Zhongyi Zhu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.
| |
Collapse
|
3
|
Wang Y, Wu Y, Jiang H, Li S, Li J, Wang C, Yang L, Zhou X, Yu J, Zhai J, Chen ZJ, Du Y. L-Kynurenine activates the AHR-PCSK9 pathway to mediate the lipid metabolic and ovarian dysfunction in polycystic ovary syndrome. Metabolism 2025; 168:156238. [PMID: 40169085 DOI: 10.1016/j.metabol.2025.156238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/03/2025]
Abstract
Dysregulated amino acid metabolism is a key contributor to polycystic ovary syndrome (PCOS). This cross-sectional study revealed that serum levels of L-kynurenine (L-Kyn) were significantly elevated in women with PCOS, whereas pyridoxal 5'-phosphate (PLP) levels were markedly reduced. Moreover, human serum L-Kyn levels exhibited a positive correlated with low-density lipoprotein cholesterol (LDL-C) and a negative correlation with high-density lipoprotein cholesterol (HDL-C). Additionally, letrozole (LET) induced PCOS-like mice displayed increased hepatic L-Kyn levels. Mechanistically, both in vivo and in vitro experiments demonstrated that the upregulation of indoleamine 2,3-dioxygenase (IDO1) activates the aryl hydrocarbon receptor (AHR) - proprotein convertase subtilisin/kexin type 9 (PCSK9) pathway in the liver of PCOS-like mice, thereby contributing to dyslipidemia. Treatment with epacadostat, an inhibitor of the enzyme IDO1, or PLP, a cofactor for L-Kyn catabolism, effectively restored ovarian function, improved glucose tolerance, and ameliorated lipid profile abnormalities in PCOS-like mice.
Collapse
Affiliation(s)
- Yujiao Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Yifan Wu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Hongwei Jiang
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Key Laboratory of Hereditary Rare Diseases of Health Commission of Henan Province, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Shang Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Jingjing Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Cong Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Lexin Yang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Xiying Zhou
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Juanjuan Yu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Junyu Zhai
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China; Center for Reproductive Medicine, Shandong University, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, Shandong 250012, China; NMU-SD Suzhou Collaborative Innovation Center for Reproductive Medicine, China
| | - Yanzhi Du
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China.
| |
Collapse
|
4
|
Albeitawi S, Bani-Mousa SU, Jarrar B, Aloqaily I, Al-Shlool N, Alsheyab G, Kassab A, Qawasmi B, Awaisheh A. Associations Between Follicular Fluid Biomarkers and IVF/ICSI Outcomes in Normo-Ovulatory Women-A Systematic Review. Biomolecules 2025; 15:443. [PMID: 40149979 PMCID: PMC11940193 DOI: 10.3390/biom15030443] [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: 01/12/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
(1) Background: The follicular fluid (FF) comprises a large portion of ovarian follicles, and serves as both a communication and growth medium for oocytes, and thus should be representative of the metabolomic status of the follicle. This review aims to explore FF biomarkers as well as their effects on fertilization, oocyte, and embryo development, and later on implantation and maintenance of pregnancy. (2) Methods: This review was registered in the PROSPERO database with the ID: CRD42025633101. We parsed PubMed, Scopus, and Google Scholar for research on the effects of different FF biomarkers on IVF/ICSI outcomes in normo-ovulatory women. Included studies were assessed for risk of bias using the NOS scale. Data were extracted and tabulated by two independent researchers. (3) Results: 22 included articles, with a sample size range of 31 to 414 and a median of 60 participants, contained 61 biomarkers, including proteins, growth factors, steroid and polypeptide hormones, inflammation and oxidative stress markers, amino acids, vitamins, lipids of different types, and miRNAs. Most of the biomarkers studied had significant effects on IVF/ICSI outcomes, and seem to have roles in various cellular pathways responsible for oocyte and embryo growth, implantation, placental formation, and maintenance of pregnancy. The FF metabolome also seems to be interconnected, with its various components influencing the levels and activities of each other through feedback loops. (4) Conclusions: FF biomarkers can be utilized for diagnostic and therapeutic purposes in IVF; however, further studies are required for choosing the most promising ones due to heterogeneity of results. Widespread adoption of LC-MS and miRNA microarrays can help quantify a representative FF metabolome, and we see great potential for in vitro supplementation (IVS) of some FF biomarkers in improving IVF/ICSI outcomes.
Collapse
Affiliation(s)
- Soha Albeitawi
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | | | - Baraa Jarrar
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Ibrahim Aloqaily
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Nour Al-Shlool
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Ghaida Alsheyab
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Ahmad Kassab
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Baha’a Qawasmi
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Abdalrahman Awaisheh
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| |
Collapse
|
5
|
Wu J, Zhao X, Fang Y, Wang C, Tian Y, Tu W, Wu Q, Yan L, Yang X. GPD1L-Mediated Glycerophospholipid Metabolism Dysfunction in Women With Diminished Ovarian Reserve: Insights From Pseudotargeted Metabolomic Analysis of Follicular Fluid. Cell Prolif 2025:e70024. [PMID: 40108995 DOI: 10.1111/cpr.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/16/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
Diminished ovarian reserve (DOR) is a pathological condition characterised by reduced ovarian function, which refers to the decreased quality and quantity of oocytes, potentially causing female infertility and various health issues. Follicular fluid (FF) serves as the microenvironment for follicular development and oocyte maturation, gaining an in-depth understanding of the metabolic state of FF will help us uncover the key biological processes involved in ovarian aging, while the specific underlying pathogenic mechanisms are not fully understood. In this study, we utilised pseudotargeted metabolomic analysis of FF to reveal the glycerophospholipid metabolism dysfunction mediated by GPD1L in DOR patients. We also found that GPD1L was downregulated in granulosa cells (GCs) of DOR patients, resulting in increased cell apoptosis and mitochondrial dysfunction. Moreover, our results demonstrated that the downregulated expression of GPD1L could induce follicular atresia and impair oocyte quality in mouse ovaries. Altogether, our research suggested that GPD1L in GCs and the key metabolites in the glycerophospholipid metabolism pathway could potentially act as novel biomarkers of DOR diagnosis, paving the way for a new theoretical basis for understanding the pathogenesis of DOR.
Collapse
Affiliation(s)
- Jiaqi Wu
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xuehan Zhao
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| | - Ying Fang
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| | - Cong Wang
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| | - Yichang Tian
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| | - Wan Tu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Qiqian Wu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Long Yan
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Xiaokui Yang
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Wang H, Yang L. Ovarian Mechanobiology: Understanding the Interplay Between Mechanics and Follicular Development. Cells 2025; 14:355. [PMID: 40072084 PMCID: PMC11898978 DOI: 10.3390/cells14050355] [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: 02/02/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/15/2025] Open
Abstract
The ovary is a dynamic organ where mechanical forces profoundly regulate follicular development, oocyte maturation, and overall reproductive function. These forces, originating from the extracellular matrix (ECM), granulosa and theca cells, and ovarian stroma, influence cellular behavior through mechanotransduction, translating mechanical stimuli into biochemical responses. This review explores the intricate interplay between mechanical cues and ovarian biology, focusing on key mechanosensitive pathways such as Hippo signaling, the PI3K/AKT pathway, and cytoskeletal remodeling, which govern follicular dormancy, activation, and growth. Additionally, it examines how ovarian aging disrupts the mechanical microenvironment, with ECM stiffening and altered mechanotransduction contributing to a decline in ovarian reserve and reproductive potential. Emerging technologies, including 3D culture systems and organ-on-chip platforms, are highlighted for their ability to replicate the ovarian microenvironment and advance drug discovery and therapeutic interventions. By integrating mechanobiological principles, this review aims to enhance our understanding of ovarian function and provide new strategies for preserving fertility and combating infertility.
Collapse
Affiliation(s)
- Haiyang Wang
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Liuqing Yang
- NUS Bia-Echo Asia Centre of Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| |
Collapse
|
7
|
Klimowska A, Jurewicz J, Radwan M, Radwan P, Pol P, Wielgomas B. Distribution of Environmental Phenols into Follicular Fluid and Urine of Women Attending Infertility Clinic. J Xenobiot 2025; 15:17. [PMID: 39997360 PMCID: PMC11856404 DOI: 10.3390/jox15010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 02/26/2025] Open
Abstract
Infertility and environmental pollution are two globally prevalent and related issues. To explore women's reproductive health, the composition of follicular fluid (FF) has been studied and it was found that changes to its composition, including the presence of exogenous chemicals, can adversely affect the fertilization process. Two groups of women (idiopathic infertility and controls) who were patients at a fertility clinic were recruited for this study. Samples of urine and FF were gathered from each participant to determine the concentration of 14 common phenols (four parabens, six bisphenols, two benzophenones, and two naphthols). Associations between phenol concentrations (free and total) in both matrices were described using Spearman's correlation coefficient and were compared between two groups by the Mann-Whitney U test. Eight phenols were quantified in more than 50% of the urine samples, while only three parabens were quantified in hydrolyzed FF samples, and only methylparaben was quantified in non-hydrolyzed FF samples. Conjugates were the predominant form in FF samples. However, a significant correlation of 0.533 (p < 0.0001) was observed between free and total methylparaben concentrations in FF. Differences in concentrations between cases and controls in both matrices were not statistically significant, except for benzophenone-3 in urine, with a higher median observed in the control group (p = 0.04). The total paraben concentrations in urine and FF samples were rather weakly correlated (r = 0.232-0.473), implying that urine concentrations may not be appropriate for predicting their concentration in FF.
Collapse
Affiliation(s)
- Anna Klimowska
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, 107 Hallera Street, 80-416 Gdańsk, Poland;
| | - Joanna Jurewicz
- Department of Chemical Safety, Nofer Institute of Occupational Medicine, 91-348 Lodz, Poland;
| | - Michał Radwan
- Department of Gynecology and Reproduction, Gameta Hospital, 34/36 Rudzka St., 95-030 Rzgow, Poland;
- Faculty of Health Sciences, Mazovian State University in Plock, 2 Dabrowskiego Sq., 09-402 Plock, Poland
| | - Paweł Radwan
- Gameta Health Centre, 7 Cybernetyki St., 02-677 Warsaw, Poland;
- Gameta, Kielce-Regional Science-Technology Centre, 45 Podzamcze St., Chęciny, 26-060 Kielce, Poland;
| | - Paweł Pol
- Gameta, Kielce-Regional Science-Technology Centre, 45 Podzamcze St., Chęciny, 26-060 Kielce, Poland;
| | - Bartosz Wielgomas
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, 107 Hallera Street, 80-416 Gdańsk, Poland;
| |
Collapse
|
8
|
Song W, Chen X, Wu H, Rahimian N. Circular RNAs as a novel class of potential therapeutic and diagnostic biomarkers in reproductive biology/diseases. Eur J Med Res 2024; 29:643. [PMID: 39741306 DOI: 10.1186/s40001-024-02230-7] [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: 06/29/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Abstract
Infertility is a prevalent problem among 10% of people within their reproductive years. Sometimes, even advanced treatment options like assisted reproduction technology have the potential to result in failed implantation. Because of the expected changes in gene expression during both in vitro and in vivo fertilization processes, these methods of assisting fertility have also been associated with undesirable pregnancy outcomes related to infertility. In this aspect, Circular RNAs (circRNAs) play a crucial role as epigenetic modifiers in a wide range of biological and pathological activities, including problems with fertility. CircRNAs are integral pieces in multiple cellular functions, including moving substances within the nucleus, silencing one X chromosome, cell death, the ability of stem cells to differentiate into different cell types, and the process of gene expression inherited from parental genes. Due to the progress made in high-speed gene sequencing, a large amount of circRNA molecules have been detected, revealing their significant functions in diverse biological functions like enhancing testicular development, preserving the differentiation and renewal of spermatogonial cells, and controlling spermatocyte meiosis. Moreover, these non-coding RNAs contribute in different aspects of female reproductive system including pregnancy-related diseases, gynecologic cancers, and endometriosis. In conclusion, there is no denying that circRNAs have immense potential to be used as biomarkers and treatments for reproductive disorders in males and females. In this research, we provide a comprehensive analysis of the multiple circRNAs associated with women's infertility.
Collapse
Affiliation(s)
- Wanyu Song
- Department of Obstetrics, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
- People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Xiuli Chen
- Department of Obstetrics, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
- People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Haiying Wu
- Department of Obstetrics, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China.
- People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China.
| | - Neda Rahimian
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Pan Y, Pan C, Zhang C. Unraveling the complexity of follicular fluid: insights into its composition, function, and clinical implications. J Ovarian Res 2024; 17:237. [PMID: 39593094 PMCID: PMC11590415 DOI: 10.1186/s13048-024-01551-9] [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: 07/05/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
Follicular fluid (FF) plays a vital role in the bidirectional communication between oocytes and granulosa cells (GCs), regulating and promoting oocyte growth and development. This fluid constitutes a complex microenvironment, rich in various molecules including hormones, growth factors, cytokines, lipids, proteins, and extracellular vesicles. Understanding the composition and metabolic profile of follicular fluid is important for investigating ovarian pathologies such as polycystic ovary syndrome (PCOS) and endometriosis. Additionally, analyzing follicular fluid can offer valuable insights into oocyte quality, aiding in optimal oocyte selection for in vitro fertilization (IVF). This review provides an overview of follicular fluid composition, classification of its components and discusses the influential components of oocyte development. It also highlights the role of follicular fluid in the pathogenesis and diagnosis of ovarian diseases, along with potential follicular fluid biomarkers for assessing oocyte quality. By understanding the intricate relationship between follicular fluid and oocyte development, we can advance fertility research and improve clinical outcomes for infertility patients.
Collapse
Affiliation(s)
- Yurong Pan
- Nanchang University Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Chenyu Pan
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Chunping Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330019, China.
| |
Collapse
|
10
|
Przewocki J, Łukaszuk A, Jakiel G, Wocławek-Potocka I, Kłosińska K, Olszewska J, Łukaszuk K. Proteomic Analysis of Follicular Fluid in Polycystic Ovary Syndrome: Insights into Protein Composition and Metabolic Pathway Alterations. Int J Mol Sci 2024; 25:11749. [PMID: 39519300 PMCID: PMC11546118 DOI: 10.3390/ijms252111749] [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: 10/16/2024] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
This study explores the proteomic composition of follicular fluid (FF) from women undergoing oocyte retrieval for in vitro fertilisation (IVF), with a focus on the effects of polycystic ovary syndrome (PCOS). FF samples were collected from 74 patients, including 34 with PCOS and 40 oocyte donors. Proteomic profiling using machine learning identified significant differences in protein abundance between the PCOS and control groups. Of the 484 quantified proteins, 20 showed significantly altered levels in the PCOS group. Functional annotation and pathway enrichment analysis pointed to the involvement of protease inhibitors and immune-related proteins in the pathophysiology of PCOS, suggesting that inflammation and immune dysregulation may play a key role. Additionally, HDL assembly was identified as a significant pathway, with apolipoprotein-AI (APOA1) and alpha-2-macroglobulin (A2M) as the major proteins involved. Notably, myosin light polypeptide 6 was the most downregulated protein, showing the highest absolute fold change, and may serve as a novel independent biomarker for PCOS.
Collapse
Affiliation(s)
- Janusz Przewocki
- Institute of Mathematics, University of Gdansk, 80-308 Gdańsk, Poland
- iYoni App—For Fertility Treatment, LifeBite, 10-763 Olsztyn, Poland;
| | - Adam Łukaszuk
- Edinburgh Medical School, College of Medicine and Veterinary Medicine, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK;
| | - Grzegorz Jakiel
- Invicta Research and Development Center, 81-740 Sopot, Poland;
- First Department of Obstetrics and Gynaecology, Centre of Postgraduate Medical Education, 01-004 Warsaw, Poland
| | - Izabela Wocławek-Potocka
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland;
| | - Karolina Kłosińska
- Department of Obstetrics and Gynecology, Oncological Gynecology and Gynecological Endocrinology, Medical University of Gdansk, 80-214 Gdańsk, Poland;
| | - Jolanta Olszewska
- Department of Obstetrics and Gynecology Nursing, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| | - Krzysztof Łukaszuk
- iYoni App—For Fertility Treatment, LifeBite, 10-763 Olsztyn, Poland;
- Department of Obstetrics and Gynecology Nursing, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| |
Collapse
|
11
|
Töhönen V, Antonson P, Boggavarapu NR, Ali H, Motaholi LA, Gustafsson JÅ, Varshney M, Rodriguez-Wallberg KA, Katayama S, Nalvarte I, Inzunza J. Transcriptomic profiling of the oocyte-cumulus-granulosa cell complex from estrogen receptor β knockout mice. F&S SCIENCE 2024; 5:306-317. [PMID: 39168303 DOI: 10.1016/j.xfss.2024.08.004] [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: 07/15/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
OBJECTIVE To study the role of estrogen receptor β in follicle development and maturation and the response to gonadotropin stimulation aiming at superovulation. DESIGN Experimental study and transcriptomic analysis. SETTING Karolinka Institutet, medical university. ANIMAL(S) Healthy wild-type (WT) and estrogen receptor β knockout (Esr2-KO) female mice undergoing superovulation at 4 weeks, 7 weeks, and 6 months of age. INTERVENTION(S) Not applicable. MAIN OUTCOME MEASURE(S) Oocyte yield after superovulation, transcriptomic profiling of cumulus-granulosa cell complexes and oocytes, and immunohistochemical analyses. RESULT(S) Superovulation of estrogen receptor β (ERβ) knockout mice resulted in reduced oocyte yield at 6 months of age compared with WT mice, but younger mice had similar yields. RNA-seq analysis of cumulus cells from superovulated WT and Esr2-KO mice identified genes and pathways associated with among others adhesion, proliferation, Wnt-signaling, and placed ERβ in bipotential granulosa cell cluster. Loss of ERβ increased expression of the other estrogen receptors Esr1 and Gper1. CONCLUSION(S) Our results show that ERβ has an important role in regulating ovulation in response to exogenous gonadotropins in 6-month-old mice, but not in younger mice. Our transcriptomic and immunohistochemical observations suggest a dysregulation of the granulosa cell communication and lack of tight coordination between granulosa cell replication and antrum expansion. A significant upregulation of other estrogen receptors may support a compensatory mechanism sustaining fertility during younger age in Esr2-KO mice.
Collapse
Affiliation(s)
- Virpi Töhönen
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Per Antonson
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | | | - Heba Ali
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Visionsgatan, Solna, Sweden; Department of Biochemistry, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | | | - Jan-Åke Gustafsson
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden; Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas
| | - Mukesh Varshney
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology-Pathology, Laboratory of Translational Fertility Preservation, Karolinska Institutet, Stockholm, Sweden; Division of Gynecology and Reproduction, Department of Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Shintaro Katayama
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden; Folkhälsan Research Center, Helsinki, Finland; Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Ivan Nalvarte
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Visionsgatan, Solna, Sweden
| | - Jose Inzunza
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
12
|
Ma XF, Liu AJ, Zheng Z, Hu BX, Zhi YX, Liu C, Tian SJ. Resolving and functional analysis of RNA editing sites in sheep ovaries and associations with litter size. Animal 2024; 18:101342. [PMID: 39471744 DOI: 10.1016/j.animal.2024.101342] [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: 12/05/2023] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 11/01/2024] Open
Abstract
Sheep litter size is a critical trait in mutton production. While litter size regulation in relation to DNA transcription have been rigorously investigated, the function of RNA editing remains less explored. To elucidate the mechanisms controlling sheep fecundity at the RNA editing level and identify pivotal RNA editing sites, this study scrutinised RNA editing sites (RESs) in follicular and luteal phases of ovaries from sheep with high and low fecundity, and the functions of population-specific RESs were subsequently analysed. A total of 2 182 475 RESs, 74.61% of which were A-to-I and C-to-U sites, were identified. These RESs were fairly evenly dispersed over the chromosomes, with 46.8% showing close clustering (inter-site distance < 300 bp). Notably, 93% were primarily situated in intronic and intergenic regions. In the follicular phase, pivotal RESs were found in the introns of genes including LPS responsive beige-like anchor, MCC regulator of Wnt signalling, and RWD domain containing 3, among others, and in the exon region of EvC ciliary complex subunit 2. In the luteal phase, RESs were observed in the introns of genes such as H/ACA ribonucleoprotein assembly factor and SDA1 domain-containing 1, and the exon and 3'UTR regions of polypeptide N-acetylgalactosaminyltransferase 15 and ilvB acetolactate synthase-like, respectively. High-fecundity sheep showed RESs in the follicular phase in genes such as fibrillin 1, cyclin-dependent kinase 6, and roundabout 1, and in genes such as autophagy-related 2B and versican in the luteal phase. Thirteen RESs specific to the follicular phase and eight specific to the luteal phase were identified in high-fecundity sheep ovaries. These RESs offer promising molecular targets and enhance understanding of multiple births in sheep from the perspective of posttranscriptional alterations.
Collapse
Affiliation(s)
- X F Ma
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - A J Liu
- Department of Agricultural and Animal Husbandry Engineering, Cangzhou Technical College, Hebei, Cangzhou, China
| | - Z Zheng
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - B X Hu
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - Y X Zhi
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - C Liu
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - S J Tian
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China; The Research Center of Cattle and Sheep Embryonic Technique of Hebei Province, Hebei, Baoding, 071000 Baoding, China.
| |
Collapse
|
13
|
Zaniker EJ, Zhang J, Russo D, Huang R, Suritis K, Drake RS, Barlow-Smith E, Shalek AK, Woodruff TK, Xiao S, Goods BA, Duncan FE. Follicle-intrinsic and spatially distinct molecular programs drive follicle rupture and luteinization during ex vivo mammalian ovulation. Commun Biol 2024; 7:1374. [PMID: 39443665 PMCID: PMC11500180 DOI: 10.1038/s42003-024-07074-9] [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: 05/09/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
During ovulation, the apical wall of the preovulatory follicle breaks down to facilitate gamete release. In parallel, the residual follicle wall differentiates into a progesterone-producing corpus luteum. Disruption of ovulation, whether through contraceptive intervention or infertility, has implications for women's health. In this study, we harness the power of an ex vivo ovulation model and machine-learning guided microdissection to identify differences between the ruptured and unruptured sides of the follicle wall. We demonstrate that the unruptured side exhibits clear markers of luteinization after ovulation while the ruptured side exhibits cell death signals. RNA-sequencing of individual follicle sides reveals 2099 differentially expressed genes (DEGs) between follicle sides without ovulation induction, and 1673 DEGs 12 h after induction of ovulation. Our model validates molecular patterns consistent with known ovulation biology even though this process occurs in the absence of the ovarian stroma, vasculature, and immune cells. We further identify previously unappreciated pathways including amino acid transport and Jag-Notch signaling on the ruptured side and glycolysis, metal ion processing, and IL-11 signaling on the unruptured side of the follicle. This study yields key insights into follicle-inherent, spatially-defined pathways that underlie follicle rupture, which may further understanding of ovulation physiology and advance women's health.
Collapse
Affiliation(s)
- Emily J Zaniker
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jiyang Zhang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Daniela Russo
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute, Harvard University & Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Ruixu Huang
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Kristine Suritis
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Riley S Drake
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute, Harvard University & Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | | | - Alex K Shalek
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute, Harvard University & Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Teresa K Woodruff
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Department of Obstetrics and Gynecology, Michigan State University, East Lansing, MI, USA
| | - Shuo Xiao
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Brittany A Goods
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
14
|
Thaler J, Tripisciano C, Nieuwland R. Investigations on the Hemostatic Potential of Physiological Body Fluids. Hamostaseologie 2024; 44:377-385. [PMID: 39442510 DOI: 10.1055/a-2374-2903] [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: 10/25/2024] Open
Abstract
Current blood coagulation models consider the interactions between blood, the vessel wall, and other tissues that expose tissue factor (TF), the main initiator of coagulation. A potential role of body fluids other than blood is generally not considered. In this review, we summarize the evidence that body fluids such as mother's milk saliva, urine, semen, and amniotic fluid trigger coagulation. The ability of these body fluids to trigger coagulation is explained by the presence of extracellular vesicles (EVs). These EVs expose extrinsic tenase complexes (i.e., complexes of TF and activated factor VII) that can trigger coagulation. Why these body fluids share this activity, however, is unknown. Possible explanations are that these body fluids contribute to hemostatic protection and/or to the regulation of the epithelial barrier function. Further investigations may help understand the underlying cellular and biochemical pathways regulating or contributing to coagulation and innate immunity, which may be directly relevant to medical conditions such as gastrointestinal bleeding and chronic inflammatory bowel disease.
Collapse
Affiliation(s)
- Johannes Thaler
- Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Carla Tripisciano
- Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Amsterdam University Medical Centers, location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Kim CW, Kim EJ, Woo MS, Cao DL, Cirunduzi AC, Ryu JH, Kong IK, Lee DK, Hong SG, Han J, Kang D. Downregulation of TASK-3 Channel Induces Senescence in Granulosa Cells of Bovine Cystic Ovarian Follicles. Int J Mol Sci 2024; 25:10199. [PMID: 39337686 PMCID: PMC11432027 DOI: 10.3390/ijms251810199] [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: 08/29/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Ovarian cysts are linked to hormone imbalances and altered gene expressions, but the connection between cysts and ion channel expression is understudied. This study explored the role of TWIK-related acid-sensitive K+ (TASK) channels in bovine ovarian cyst formation. The ovarian follicles were split into small (5 to 10 mm in diameter) and large (>25 mm in diameter) groups. Among the measured K+, Na+, and Cl- concentrations in follicular fluid (FF) obtained from small-sized follicles (SFs) and large-sized follicles (LFs), the K+ concentration was significantly lower in LFFF. Quantitative PCR, Western blot, and immunocytochemistry data revealed that TASK-3 expression levels significantly decreased by approximately 50% in LFs and granulosa cells obtained from LFs (LFGCs) compared to the corresponding controls. The TASK-3 protein was localized to the plasma membranes of GCs. The diameters of LFGCs were larger than those of SFGCs. The cell swelling response to exposure to a hypotonic solution (200 mOsm/L) was highly reduced in TASK-3-overexpressing cells compared to vector-transfected cells. TASK-3-knockdown cells showed arrested growth. Senescence markers were detected in LFGCs and TASK-3-knockdown cells. These findings suggest that reduced TASK-3 expression in LFs is associated with the inhibition of GC growth, leading to senescence and cyst formation.
Collapse
Affiliation(s)
- Chang-Woon Kim
- Department of Obstetrics and Gynecology, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Republic of Korea
| | - Eun-Jin Kim
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Min Seok Woo
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Dang Long Cao
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Asifiwe Clarisse Cirunduzi
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Ji Hyeon Ryu
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Il-Keun Kong
- Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Dong Kun Lee
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Seong-Geun Hong
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Jaehee Han
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Dawon Kang
- Department of Physiology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Republic of Korea
| |
Collapse
|
16
|
Wang H, Huang Z, Shen X, Lee Y, Song X, Shu C, Wu LH, Pakkiri LS, Lim PL, Zhang X, Drum CL, Zhu J, Li R. Rejuvenation of aged oocyte through exposure to young follicular microenvironment. NATURE AGING 2024; 4:1194-1210. [PMID: 39251866 DOI: 10.1038/s43587-024-00697-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/30/2024] [Indexed: 09/11/2024]
Abstract
Reproductive aging is a major cause of fertility decline, attributed to decreased oocyte quantity and developmental potential. A possible cause is aging of the surrounding follicular somatic cells that support oocyte growth and development by providing nutrients and regulatory factors. Here, by creating chimeric follicles, whereby an oocyte from one follicle was transplanted into and cultured within another follicle whose native oocyte was removed, we show that young oocytes cultured in aged follicles exhibited impeded meiotic maturation and developmental potential, whereas aged oocytes cultured within young follicles were significantly improved in rates of maturation, blastocyst formation and live birth after in vitro fertilization and embryo implantation. This rejuvenation of aged oocytes was associated with enhanced interaction with somatic cells, transcriptomic and metabolomic remodeling, improved mitochondrial function and higher fidelity of meiotic chromosome segregation. These findings provide the basis for a future follicular somatic cell-based therapy to treat female infertility.
Collapse
Affiliation(s)
- HaiYang Wang
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.
| | - Zhongwei Huang
- NUS Bia Echo Asia Centre for Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xingyu Shen
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Yaelim Lee
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - XinJie Song
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Chang Shu
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Lik Hang Wu
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Leroy Sivappiragasam Pakkiri
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Poh Leong Lim
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xi Zhang
- Center for Cell Dynamics and Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chester Lee Drum
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jin Zhu
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Rong Li
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.
- Center for Cell Dynamics and Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
17
|
Franko R, de Almeida Monteiro Melo Ferraz M. Exploring the potential of in vitro extracellular vesicle generation in reproductive biology. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70007. [PMID: 39238549 PMCID: PMC11375532 DOI: 10.1002/jex2.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 09/07/2024]
Abstract
The interest in the growing field of extracellular vesicle (EV) research highlights their significance in intercellular signalling and the selective transfer of biological information between donor and recipient cells. EV studies have provided valuable insights into intercellular communication mechanisms, signal identification and their involvement in disease states, offering potential avenues for manipulating pathological conditions, detecting biomarkers and developing drug-delivery systems. While our understanding of EV functions in reproductive tissues has significantly progressed, exploring their potential as biomarkers for infertility, therapeutic interventions and enhancements in assisted reproductive technologies remains to be investigated. This knowledge gap stems partly from the difficulties associated with large-scale EV production relevant to clinical applications. Most existing studies on EV production rely on conventional 2D cell culture systems, characterized by suboptimal EV yields and a failure to replicate in vivo conditions. This results in the generation of EVs that differ from their in vivo counterparts. Hence, this review firstly delves into the importance of EVs in reproduction to then expand on current techniques for in vitro EV production, specifically examining diverse methods of culture and the potential of bioengineering technologies to establish innovative systems for enhanced EV production.
Collapse
Affiliation(s)
- Roksan Franko
- Clinic of Ruminants, Faculty of Veterinary Medicine Ludwig-Maximilians-Universität München Oberschleißheim Germany
- Gene Center Ludwig-Maximilians-Universität München Munich Germany
| | - Marcia de Almeida Monteiro Melo Ferraz
- Clinic of Ruminants, Faculty of Veterinary Medicine Ludwig-Maximilians-Universität München Oberschleißheim Germany
- Gene Center Ludwig-Maximilians-Universität München Munich Germany
| |
Collapse
|
18
|
Duval C, Wyse BA, Tsang BK, Librach CL. Extracellular vesicles and their content in the context of polycystic ovarian syndrome and endometriosis: a review. J Ovarian Res 2024; 17:160. [PMID: 39103867 DOI: 10.1186/s13048-024-01480-7] [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: 04/30/2024] [Accepted: 07/18/2024] [Indexed: 08/07/2024] Open
Abstract
Extracellular vesicles (EVs), particles enriched in bioactive molecules like proteins, nucleic acids, and lipids, are crucial mediators of intercellular communication and play key roles in various physiological and pathological processes. EVs have been shown to be involved in ovarian follicular function and to be altered in two prevalent gynecological disorders; polycystic ovarian syndrome (PCOS) and endometriosis.Ovarian follicles are complex microenvironments where folliculogenesis takes place with well-orchestrated interactions between granulosa cells, oocytes, and their surrounding stromal cells. Recent research unveiled the presence of EVs, including exosomes and microvesicles, in the follicular fluid (FFEVs), which constitutes part of the developing oocyte's microenvironment. In the context of PCOS, a multifaceted endocrine, reproductive, and metabolic disorder, studies have explored the dysregulation of these FFEVs and their cargo. Nine PCOS studies were included in this review and two miRNAs were commonly reported in two different studies, miR-379 and miR-200, both known to play a role in female reproduction. Studies have also demonstrated the potential use of EVs as diagnostic tools and treatment options.Endometriosis, another prevalent gynecological disorder characterized by ectopic growth of endometrial-like tissue, has also been linked to aberrant EV signaling. EVs in the peritoneal fluid of women with endometriosis carry molecules that modulate the immune response and promote the establishment and maintenance of endometriosis lesions. EVs derived from endometriosis lesions, serum and peritoneal fluid obtained from patients with endometriosis showed no commonly reported biomolecules between the eleven reviewed studies. Importantly, circulating EVs have been shown to be potential biomarkers, also reflecting the severity of the pathology.Understanding the interplay of EVs within human ovarian follicles may provide valuable insights into the pathophysiology of both PCOS and endometriosis. Targeting EV-mediated communication may open avenues for novel diagnostic and therapeutic approaches for these common gynecological disorders. More research is essential to unravel the mechanisms underlying EV involvement in folliculogenesis and its dysregulation in PCOS and endometriosis, ultimately leading to more effective and personalized interventions.
Collapse
Affiliation(s)
- Cyntia Duval
- CReATe Fertility Center, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | | | - Benjamin K Tsang
- Inflammation and Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Departments of Obstetrics and Gynecology & Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Clifford L Librach
- CReATe Fertility Center, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada
- Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
19
|
Benedetti C, Pavani KC, Gansemans Y, Azari-Dolatabad N, Pascottini OB, Peelman L, Six R, Fan Y, Guan X, Deserranno K, Fernández-Montoro A, Hamacher J, Van Nieuwerburgh F, Fair T, Hendrix A, Smits K, Van Soom A. From follicle to blastocyst: microRNA-34c from follicular fluid-derived extracellular vesicles modulates blastocyst quality. J Anim Sci Biotechnol 2024; 15:104. [PMID: 39097731 PMCID: PMC11298084 DOI: 10.1186/s40104-024-01059-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/04/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND Within the follicular fluid, extracellular vesicles (EVs) guide oocyte growth through their cargo microRNAs (miRNAs). Here, we investigated the role of EVs and their cargo miRNAs by linking the miRNAs found in EVs, derived from the fluid of an individual follicle, to the ability of its oocyte to become a blastocyst (competent) or not (non-competent). METHODS Bovine antral follicles were dissected, categorized as small (2-4 mm) or large (5-8 mm) and the corresponding oocytes were subjected to individual maturation, fertilization and embryo culture to the blastocyst stage. Follicular fluid was pooled in 4 groups (4 replicates) based on follicle size and competence of the corresponding oocyte to produce a blastocyst. Follicular fluid-derived EVs were isolated, characterized, and subjected to miRNA-sequencing (Illumina Miseq) to assess differential expression (DE) in the 4 groups. Functional validation of the effect of miR-34c on embryo development was performed by supplementation of mimics and inhibitors during in vitro maturation (IVM). RESULTS We identified 16 DE miRNAs linked to oocyte competence when follicular size was not considered. Within the large and small follicles, 46 DE miRNAs were driving blastocyst formation in each group. Comparison of EVs from competent small and large follicles revealed 90 DE miRNAs. Cell regulation, cell differentiation, cell cycle, and metabolic process regulation were the most enriched pathways targeted by the DE miRNAs from competent oocytes. We identified bta-miR-34c as the most abundant in follicular fluid containing competent oocytes. Supplementation of miR-34c mimic and inhibitor during IVM did not affect embryo development. However, blastocyst quality, as evidenced by higher cell numbers, was significantly improved following oocyte IVM in the presence of miR-34c mimics, while miR-34c inhibitors resulted in the opposite effect. CONCLUSION This study demonstrates the regulatory effect of miRNAs from follicular fluid-derived EVs on oocyte competence acquisition, providing a further basis for understanding the significance of miRNAs in oocyte maturation and embryonic development. Up-regulation of miR-34c in EVs from follicular fluid containing competent oocytes and the positive impact of miR-34c mimics added during IVM on the resulting blastocysts indicate its pivotal role in oocyte competence.
Collapse
Affiliation(s)
- Camilla Benedetti
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Krishna Chaitanya Pavani
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
- Department for Reproductive Medicine, Ghent University Hospital, 9000, Ghent, Belgium
| | - Yannick Gansemans
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, B-9000, Ghent, Belgium
| | | | - Osvaldo Bogado Pascottini
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Luc Peelman
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, B-9000, Ghent, Belgium
| | - Rani Six
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, B-9000, Ghent, Belgium
| | - Yuan Fan
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, B-9000, Ghent, Belgium
| | - Xuefeng Guan
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, B-9000, Ghent, Belgium
| | - Koen Deserranno
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, B-9000, Ghent, Belgium
| | - Andrea Fernández-Montoro
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Joachim Hamacher
- Institute of Crop Science and Resource Conservation, Plant Pathology, Rheinische Friedrich-Wilhelms-University of Bonn, 53115, Bonn, Germany
| | - Filip Van Nieuwerburgh
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, B-9000, Ghent, Belgium
| | - Trudee Fair
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, Ireland
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, B-9000, Ghent, Belgium
| | - Katrien Smits
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium.
| | - Ann Van Soom
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium.
| |
Collapse
|
20
|
Lava Kumar S, Kushawaha B, Mohanty A, Kumari A, Kumar A, Beniwal R, Kiran Kumar P, Athar M, Krishna Rao D, Rao HBDP. Glutathione peroxidase (GPX1) - Selenocysteine metabolism preserves the follicular fluid's (FF) redox homeostasis via IGF-1- NMD cascade in follicular ovarian cysts (FOCs). Biochim Biophys Acta Mol Basis Dis 2024; 1870:167235. [PMID: 38744343 DOI: 10.1016/j.bbadis.2024.167235] [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: 11/03/2023] [Revised: 04/18/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
Follicular ovarian cysts (FOCs) are characterized by follicles in the ovaries that are >20 mm in diameter and persist for >10 days without the corpus luteum, leading to anovulation, dysregulation of folliculogenesis and subfertility in humans and livestock species. Despite their clinical significance, the precise impact of FOCs on oocyte reserve, maturation, and quality still needs to be explored. While FOCs are observed in both human and livestock populations, they are notably prevalent in livestock species. Consequently, livestock species serve as valuable models for investigating the molecular intricacies of FOCs. Thus, in this study, using goat FOCs, we performed integrated proteomic, metabolomic and functional analyses to demonstrate that oocyte maturation is hampered due to increased reactive oxygen species (ROS) in FOCs follicular fluid (FF) via downregulation of glutathione peroxidase (GPX1), a critical antioxidant seleno enzyme required to negate oxidative stress. Notably, GPX1 reduction was positively correlated with the FF's decline of free selenium and selenocysteine metabolic enzymes, O-phosphoryl-tRNA (Sec) selenium transferase (SEPSECS) and selenocysteine lyase (SCLY) levels. Adding GPX1, selenocysteine, or selenium to the culture media rescued the oocyte maturation abnormalities caused by FOCs FF by down-regulating the ROS. Additionally, we demonstrate that substituting GPX1 regulator, Insulin-like growth factor-I (IGF-1) in the in vitro maturation media improved the oocyte maturation in the cystic FF by down-regulating the ROS activity via suppressing Non-sense-mediated decay (NMD) of GPX1. In contrast, inhibition of IGF-1R and the target of rapamycin complex 1 (mTORC1) hampered the oocyte maturation via NMD up-regulation. These findings imply that the GPX1 regulation via selenocysteine metabolism and the IGF-1-mediated NMD may be critical for the redox homeostasis of FF. We propose that GPX1 enhancers hold promise as therapeutics for enhancing the competence of FOCs oocytes. However, further in vivo studies are necessary to validate these findings observed in vitro.
Collapse
Affiliation(s)
- S Lava Kumar
- National Institute of Animal Biotechnology, Hyderabad, Telangana 500032, India; Graduate studies, Regional Center for Biotechnology, Faridabad 121 001, India
| | - Bhawna Kushawaha
- National Institute of Animal Biotechnology, Hyderabad, Telangana 500032, India
| | - Aradhana Mohanty
- National Institute of Animal Biotechnology, Hyderabad, Telangana 500032, India; Graduate studies, Regional Center for Biotechnology, Faridabad 121 001, India
| | - Anjali Kumari
- National Institute of Animal Biotechnology, Hyderabad, Telangana 500032, India; Graduate studies, Regional Center for Biotechnology, Faridabad 121 001, India
| | - Ajith Kumar
- National Institute of Animal Biotechnology, Hyderabad, Telangana 500032, India; Graduate studies, Regional Center for Biotechnology, Faridabad 121 001, India
| | - Rohit Beniwal
- National Institute of Animal Biotechnology, Hyderabad, Telangana 500032, India; Graduate studies, Regional Center for Biotechnology, Faridabad 121 001, India
| | - P Kiran Kumar
- National Institute of Animal Biotechnology, Hyderabad, Telangana 500032, India
| | - Mohd Athar
- National Institute of Animal Biotechnology, Hyderabad, Telangana 500032, India; Graduate studies, Regional Center for Biotechnology, Faridabad 121 001, India
| | - D Krishna Rao
- Tata Institute of Fundamental Research, Hyderabad, Telangana 500032, India
| | - H B D Prasada Rao
- National Institute of Animal Biotechnology, Hyderabad, Telangana 500032, India.
| |
Collapse
|
21
|
Huang R, Kratka CE, Pea J, McCann C, Nelson J, Bryan JP, Zhou LT, Russo DD, Zaniker EJ, Gandhi AH, Shalek AK, Cleary B, Farhi SL, Duncan FE, Goods BA. Single-cell and spatiotemporal profile of ovulation in the mouse ovary. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.594719. [PMID: 38826447 PMCID: PMC11142086 DOI: 10.1101/2024.05.20.594719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Ovulation is a spatiotemporally coordinated process that involves several tightly controlled events, including oocyte meiotic maturation, cumulus expansion, follicle wall rupture and repair, and ovarian stroma remodeling. To date, no studies have detailed the precise window of ovulation at single-cell resolution. Here, we performed parallel single-cell RNA-seq and spatial transcriptomics on paired mouse ovaries across an ovulation time course to map the spatiotemporal profile of ovarian cell types. We show that major ovarian cell types exhibit time-dependent transcriptional states enriched for distinct functions and have specific localization profiles within the ovary. We also identified gene markers for ovulation-dependent cell states and validated these using orthogonal methods. Finally, we performed cell-cell interaction analyses to identify ligand-receptor pairs that may drive ovulation, revealing previously unappreciated interactions. Taken together, our data provides a rich and comprehensive resource of murine ovulation that can be mined for discovery by the scientific community.
Collapse
|
22
|
He S, Li H, Zhang Q, Zhao W, Li W, Dai C, Li B, Cheng J, Wu S, Zhou Z, Yang J, Li S. Berberine alleviates inflammation in polycystic ovary syndrome by inhibiting hyaluronan synthase 2 expression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155456. [PMID: 38537446 DOI: 10.1016/j.phymed.2024.155456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/03/2024] [Accepted: 02/14/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a heterogeneous metabolic and endocrine disorder that causes anovulatory infertility and abnormal folliculogenesis in women of reproductive age. Several studies have revealed inflammation in PCOS follicles, and recent evidence suggests that Berberine (BBR) effectively reduces inflammatory responses in PCOS, however, the underlying mechanisms remain unclear. PURPOSE To determine the underlying mechanisms by which BBR alleviates inflammation in PCOS. STUDY DESIGN Primary human GCs from healthy women and women with PCOS, and KGN cells were used for in vitro studies. ICR mice were used for in vivo studies. METHODS Gene expression was measured using RT-qPCR. HAS2, inflammatory cytokines, and serum hormones were assayed by ELISA. Protein expression profiles were assayed by Western blot. Chronic low-grade inflammatory mouse models were developed by intraperitoneal injection with LPS, and PCOS mouse models were established by subcutaneous intraperitoneal injection of DHEA. BBR and 4-MU were administered by gavage. Ovarian morphologic changes were evaluated using H&E staining. HAS2 expression in the ovary was assayed using Western blot and immunohistochemistry. RESULTS Our results confirmed that HAS2 expression and hyaluronan (HA) accumulation are closely associated with inflammatory responses in PCOS. Data obtained from in vitro studies showed that HAS2 and inflammatory genes (e.g., MCP-1, IL-1β, and IL-6) are significantly upregulated in PCOS samples and LPS-induced KGN cells compared to their control groups. In addition, these effects were reversed by blocking HAS2 expression or HA synthesis using BBR or 4-MU, respectively. Furthermore, HAS2 overexpression induces the expression of inflammatory genes in PCOS. These results were further confirmed in LPS- and DHEA-induced mouse models, where inflammatory genes were reduced by BBR or 4-MU, and ovarian morphology was restored. CONCLUSIONS Our results define previously unknown links between HAS2 and chronic low-grade inflammation in the follicles of women with PCOS. BBR exerts its anti-inflammatory effects by down-regulating HAS2. This study provides a novel therapeutic target for alleviating ovarian inflammation in women with PCOS.
Collapse
Affiliation(s)
- Shaojing He
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China
| | - Hui Li
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China; Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing, 210014, China
| | - Qianjie Zhang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China
| | - Weimin Zhao
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China; Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing, 210014, China
| | - Wei Li
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Chaohui Dai
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China; Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing, 210014, China
| | - Bixia Li
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China; Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing, 210014, China
| | - Jinhua Cheng
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China; Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing, 210014, China
| | - Shuang Wu
- Hubei Provincial Hospital of Traditional Chinese Medicine Affiliated to Hubei University of Traditional Chinese Medicine, Wuhan, 430060, China
| | - Zhongming Zhou
- Hubei Provincial Hospital of Traditional Chinese Medicine Affiliated to Hubei University of Traditional Chinese Medicine, Wuhan, 430060, China
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Saijiao Li
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China.
| |
Collapse
|
23
|
Zhang G, Lin W, Gao N, Lan C, Ren M, Yan L, Pan B, Xu J, Han B, Hu L, Chen Y, Wu T, Zhuang L, Lu Q, Wang B, Fang M. Using Machine Learning to Construct the Blood-Follicle Distribution Models of Various Trace Elements and Explore the Transport-Related Pathways with Multiomics Data. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:7743-7757. [PMID: 38652822 DOI: 10.1021/acs.est.3c10904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Permeabilities of various trace elements (TEs) through the blood-follicle barrier (BFB) play an important role in oocyte development. However, it has not been comprehensively described as well as its involved biological pathways. Our study aimed to construct a blood-follicle distribution model of the concerned TEs and explore their related biological pathways. We finally included a total of 168 women from a cohort of in vitro fertilization-embryo transfer conducted in two reproductive centers in Beijing City and Shandong Province, China. The concentrations of 35 TEs in both serum and follicular fluid (FF) samples from the 168 women were measured, as well as the multiomics features of the metabolome, lipidome, and proteome in both plasma and FF samples. Multiomics features associated with the transfer efficiencies of TEs through the BFB were selected by using an elastic net model and further utilized for pathway analysis. Various machine learning (ML) models were built to predict the concentrations of TEs in FF. Overall, there are 21 TEs that exhibited three types of consistent BFB distribution characteristics between Beijing and Shandong centers. Among them, the concentrations of arsenic, manganese, nickel, tin, and bismuth in FF were higher than those in the serum with transfer efficiencies of 1.19-4.38, while a reverse trend was observed for the 15 TEs with transfer efficiencies of 0.076-0.905, e.g., mercury, germanium, selenium, antimony, and titanium. Lastly, cadmium was evenly distributed in the two compartments with transfer efficiencies of 0.998-1.056. Multiomics analysis showed that the enrichment of TEs was associated with the synthesis and action of steroid hormones and the glucose metabolism. Random forest model can provide the most accurate predictions of the concentrations of TEs in FF among the concerned ML models. In conclusion, the selective permeability through the BFB for various TEs may be significantly regulated by the steroid hormones and the glucose metabolism. Also, the concentrations of some TEs in FF can be well predicted by their serum levels with a random forest model.
Collapse
Affiliation(s)
- Guohuan Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, P. R. China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of the People's Republic of China, Beijing 100191, P. R. China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, P. R. China
| | - Weinan Lin
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, P. R. China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of the People's Republic of China, Beijing 100191, P. R. China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, P. R. China
| | - Ning Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, P. R. China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of the People's Republic of China, Beijing 100191, P. R. China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, P. R. China
| | - Changxin Lan
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, P. R. China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of the People's Republic of China, Beijing 100191, P. R. China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, P. R. China
| | - Mengyuan Ren
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, P. R. China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of the People's Republic of China, Beijing 100191, P. R. China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, P. R. China
| | - Lailai Yan
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing 100191, P. R. China
| | - Bo Pan
- Yunnan Provincial Key Lab of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming 650500, P. R. China
| | - Jia Xu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, P. R. China
| | - Bin Han
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, P. R. China
| | - Ligang Hu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Science, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Yuanchen Chen
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, P. R. China
| | - Tianxiang Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, P. R. China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of the People's Republic of China, Beijing 100191, P. R. China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, P. R. China
| | - Lili Zhuang
- Reproductive Medicine Centre, Yuhuangding Hospital of Yantai, Affiliated Hospital of Qingdao University, Yantai 264000, P. R. China
| | - Qun Lu
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R China
- Center of Reproductive Medicine, Peking University People's Hospital, Beijing 100044, P. R. China
| | - Bin Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, P. R. China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of the People's Republic of China, Beijing 100191, P. R. China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, P. R. China
- Laboratory for Earth Surface Processes, College of Urban and Environmental Science, Peking University, Beijing 100871, China
| | - Mingliang Fang
- Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, P. R. China
| |
Collapse
|
24
|
Duan Y, Cai B, Guo J, Wang C, Mai Q, Xu Y, Zeng Y, Shi Y, Wang B, Ding C, Chen M, Zhou C, Xu Y. GDF9 His209GlnfsTer6/S428T and GDF9 Q321X/S428T bi-allelic variants caused female subfertility with defective follicle enlargement. Cell Commun Signal 2024; 22:235. [PMID: 38643161 PMCID: PMC11031944 DOI: 10.1186/s12964-024-01616-8] [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: 11/22/2023] [Accepted: 04/12/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Antral follicles consist of an oocyte cumulus complex surrounding by somatic cells, including mural granulosa cells as the inner layer and theca cells as the outsider layer. The communications between oocytes and granulosa cells have been extensively explored in in vitro studies, however, the role of oocyte-derived factor GDF9 on in vivo antral follicle development remains elusive due to lack of an appropriate animal model. Clinically, the phenotype of GDF9 variants needs to be determined. METHODS Whole-exome sequencing (WES) was performed on two unrelated infertile women characterized by an early rise of estradiol level and defect in follicle enlargement. Besides, WES data on 1,039 women undergoing ART treatment were collected. A Gdf9Q308X/S415T mouse model was generated based on the variant found in one of the patients. RESULTS Two probands with bi-allelic GDF9 variants (GDF9His209GlnfsTer6/S428T, GDF9Q321X/S428T) and eight GDF9S428T heterozygotes with normal ovarian response were identified. In vitro experiments confirmed that these variants caused reduction of GDF9 secretion, and/or alleviation in BMP15 binding. Gdf9Q308X/S415T mouse model was constructed, which recapitulated the phenotypes in probands with abnormal estrogen secretion and defected follicle enlargement. Further experiments in mouse model showed an earlier expression of STAR in small antral follicles and decreased proliferative capacity in large antral follicles. In addition, RNA sequencing of granulosa cells revealed the transcriptomic profiles related to defective follicle enlargement in the Gdf9Q308X/S415T group. One of the downregulated genes, P4HA2 (a collagen related gene), was found to be stimulated by GDF9 protein, which partly explained the phenotype of defective follicle enlargement. CONCLUSIONS GDF9 bi-allelic variants contributed to the defect in antral follicle development. Oocyte itself participated in the regulation of follicle development through GDF9 paracrine effect, highlighting the essential role of oocyte-derived factors on ovarian response.
Collapse
Affiliation(s)
- Yuwei Duan
- Department of Gynecology & Obstetrics, Center for Reproductive Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, Guangzhou, Guangdong, 510080, China
| | - Bing Cai
- Department of Gynecology & Obstetrics, Center for Reproductive Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, Guangzhou, Guangdong, 510080, China
| | - Jing Guo
- Department of Gynecology & Obstetrics, Center for Reproductive Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, Guangzhou, Guangdong, 510080, China
| | - Chen Wang
- Department of Gynecology & Obstetrics, Center for Reproductive Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, Guangzhou, Guangdong, 510080, China
| | - Qingyun Mai
- Department of Gynecology & Obstetrics, Center for Reproductive Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, Guangzhou, Guangdong, 510080, China
| | - Yan Xu
- Department of Gynecology & Obstetrics, Center for Reproductive Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, Guangzhou, Guangdong, 510080, China
| | - Yang Zeng
- Department of Gynecology & Obstetrics, Center for Reproductive Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, Guangzhou, Guangdong, 510080, China
| | - Yue Shi
- Department of Gynecology & Obstetrics, Center for Reproductive Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, Guangzhou, Guangdong, 510080, China
| | - Boyan Wang
- Department of Gynecology & Obstetrics, Center for Reproductive Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, Guangzhou, Guangdong, 510080, China
| | - Chenhui Ding
- Department of Gynecology & Obstetrics, Center for Reproductive Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, Guangzhou, Guangdong, 510080, China
| | - Minghui Chen
- Department of Gynecology & Obstetrics, Center for Reproductive Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, Guangzhou, Guangdong, 510080, China
| | - Canquan Zhou
- Department of Gynecology & Obstetrics, Center for Reproductive Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, 510080, China
- Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, Guangzhou, Guangdong, 510080, China
| | - Yanwen Xu
- Department of Gynecology & Obstetrics, Center for Reproductive Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, 510080, China.
- Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
25
|
Xie Y, Chen J, Liu K, Huang J, Zeng Y, Gao M, Qian Y, Liu L, Tan Y, Nie X. Differential expression of follicular fluid exosomal microRNA in women with diminished ovarian reserve. J Assist Reprod Genet 2024; 41:1087-1096. [PMID: 38321265 PMCID: PMC11052957 DOI: 10.1007/s10815-024-03037-5] [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: 09/18/2023] [Accepted: 01/15/2024] [Indexed: 02/08/2024] Open
Abstract
PURPOSE Decreased ovarian reserve function is mainly characterized by female endocrine disorders and fertility decline. Follicular fluid (FF) exosomal microRNAs (miRNAs) have been shown to regulate the function of granulosa cells (GCs). The present study explored differentially expressed miRNAs (DEmiRNAs) in patients with diminished ovarian reserve (DOR). METHODS FF was collected from 12 DOR patients and 12 healthy controls. DEmiRNAs between the two groups were identified and analyzed using high-throughput sequencing technology and validated by real-time quantitative PCR (RT-qPCR). RESULTS A total of 592 DEmiRNAs were identified using high-throughput miRNA sequencing, of which 213 were significantly upregulated and 379 were significantly downregulated. The sequencing results were further validated by RT-qPCR. These DEmiRNA target genes were mainly involved in the cancer pathway, phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway, regulation of actin cytoskeleton signaling pathway, and biological processes related to protein binding, nucleoplasm, cytoplasm, and cell membrane. CONCLUSION FF exosomal miRNAs are significantly differentially expressed in DOR patients versus non-DOR patients, underscoring their crucial role in regulating the pathogenesis of DOR.
Collapse
Affiliation(s)
- Ying Xie
- Department of Reproductive Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Juan Chen
- Department of Reproductive Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Kailu Liu
- Department of Reproductive Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Jingyu Huang
- Department of Reproductive Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yaqiong Zeng
- Department of Reproductive Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Mengya Gao
- Department of Reproductive Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yu Qian
- Department of Reproductive Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Li Liu
- Department of Reproductive Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yong Tan
- Department of Reproductive Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Xiaowei Nie
- Department of Reproductive Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
26
|
Salvi A, Li W, Dipali SS, Cologna SM, Pavone ME, Duncan FE, Burdette JE. Follicular fluid aids cell adhesion, spreading in an age independent manner and shows an age-dependent effect on DNA damage in fallopian tube epithelial cells. Heliyon 2024; 10:e27336. [PMID: 38501015 PMCID: PMC10945186 DOI: 10.1016/j.heliyon.2024.e27336] [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: 11/27/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/20/2024] Open
Abstract
Ovarian cancer (OC) is deadly, and likely arises from the fallopian tube epithelium (FTE). Despite the association of OC with ovulation, OC typically presents in post-menopausal women who are no longer ovulating. The goal of this study was to understand how ovulation and aging interact to impact OC progression from the FTE. Follicular fluid released during ovulation induces DNA damage in the FTE, however, the role of aging on FTE exposure to follicular fluid is unexplored. Follicular fluid samples were collected from 14 women and its effects on FTE cells was assessed. Follicular fluid caused DNA damage and lipid oxidation in an age-dependent manner, but instead induced cell proliferation in a dose-dependent manner, independent of age in FTE cells. Follicular fluid regardless of age disrupted FTE spheroid formation and stimulated attachment and growth on ultra-low attachment plates. Proteomics analysis of the adhesion proteins in the follicular fluid samples identified vitronectin, a glycoprotein responsible for FTE cell attachment and spreading.
Collapse
Affiliation(s)
- Amrita Salvi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL, 60607, USA
| | - Wenping Li
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, 60607, USA
| | - Shweta S. Dipali
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Stephanie M. Cologna
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, 60607, USA
| | - Mary Ellen Pavone
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Francesca E. Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Joanna E. Burdette
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL, 60607, USA
| |
Collapse
|
27
|
Gabryś J, Gurgul A, Szmatoła T, Kij-Mitka B, Andronowska A, Karnas E, Kucharski M, Wojciechowska-Puchałka J, Kochan J, Bugno-Poniewierska M. Follicular Fluid-Derived Extracellular Vesicles Influence on In Vitro Maturation of Equine Oocyte: Impact on Cumulus Cell Viability, Expansion and Transcriptome. Int J Mol Sci 2024; 25:3262. [PMID: 38542236 PMCID: PMC10970002 DOI: 10.3390/ijms25063262] [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: 01/30/2024] [Revised: 02/29/2024] [Accepted: 03/08/2024] [Indexed: 07/14/2024] Open
Abstract
Cumulus cell (CC) expansion is pivotal for oocyte maturation, during which CCs release factors that initiate paracrine signaling within the follicular fluid (FF). The FF is abundant in extracellular vesicles (EVs) that facilitate intercellular communication. Although bovine and murine EVs can control cumulus expansion, these effects have not been observed in equines. This study aimed to assess the impact of FF-derived EVs (ffEVs) on equine CC expansion, viability, and transcriptome. Cumulus-oocyte complexes (COCs) that underwent in vitro maturation (IVM) in the presence (200 µg protein/mL) or absence (control) of ffEVs were assessed for cumulus expansion and viability. CCs were isolated after 12 h of IVM, followed by RNA extraction, cDNA library generation, and subsequent transcriptome analysis using next-generation sequencing. Confocal microscopy images illustrated the internalization of labeled ffEVs by CCs. Supplementation with ffEVs significantly enhanced cumulus expansion in both compacted (Cp, p < 0.0001) and expanded (Ex, p < 0.05) COCs, while viability increased in Cp groups (p < 0.01), but decreased in Ex groups (p < 0.05), compared to the controls. Although transcriptome analysis revealed a subtle effect on CC RNA profiles, differentially expressed genes encompassed processes (e.g., MAPK and Wnt signaling) potentially crucial for cumulus properties and, consequently, oocyte maturation.
Collapse
Affiliation(s)
- Julia Gabryś
- Department of Animal Reproduction, Anatomy and Genomics, Faculty of Animal Science, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059 Krakow, Poland; (J.G.); (B.K.-M.); (J.W.-P.); (J.K.); (M.B.-P.)
| | - Artur Gurgul
- Center for Experimental and Innovative Medicine, University of Agriculture in Krakow, Rędzina 1c, 30-248 Krakow, Poland;
| | - Tomasz Szmatoła
- Center for Experimental and Innovative Medicine, University of Agriculture in Krakow, Rędzina 1c, 30-248 Krakow, Poland;
| | - Barbara Kij-Mitka
- Department of Animal Reproduction, Anatomy and Genomics, Faculty of Animal Science, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059 Krakow, Poland; (J.G.); (B.K.-M.); (J.W.-P.); (J.K.); (M.B.-P.)
| | - Aneta Andronowska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland;
| | - Elżbieta Karnas
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland;
| | - Mirosław Kucharski
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059 Krakow, Poland;
| | - Joanna Wojciechowska-Puchałka
- Department of Animal Reproduction, Anatomy and Genomics, Faculty of Animal Science, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059 Krakow, Poland; (J.G.); (B.K.-M.); (J.W.-P.); (J.K.); (M.B.-P.)
| | - Joanna Kochan
- Department of Animal Reproduction, Anatomy and Genomics, Faculty of Animal Science, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059 Krakow, Poland; (J.G.); (B.K.-M.); (J.W.-P.); (J.K.); (M.B.-P.)
| | - Monika Bugno-Poniewierska
- Department of Animal Reproduction, Anatomy and Genomics, Faculty of Animal Science, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059 Krakow, Poland; (J.G.); (B.K.-M.); (J.W.-P.); (J.K.); (M.B.-P.)
| |
Collapse
|
28
|
Xu Y, Zhou Z, Zhang G, Yang Z, Shi Y, Jiang Z, Liu Y, Chen H, Huang H, Zhang Y, Pan J. Metabolome implies increased fatty acid utilization and histone methylation in the follicles from hyperandrogenic PCOS women. J Nutr Biochem 2024; 125:109548. [PMID: 38104867 DOI: 10.1016/j.jnutbio.2023.109548] [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: 07/10/2023] [Revised: 11/21/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Well-balanced metabolism is essential for the high-quality of oocytes, and metabolic fluctuations of follicular microenvironment potentially encourage functional changes in follicle cells, ultimately impacting the developmental potential of oocytes. Here, the global metabolomic profiles of follicular fluid from PCOS women with ovarian hyperandrogenism and nonhyperandrogenism were depicted by untargeted metabolome and transcriptome. In parallel, functional methods were employed to evaluate the possible impact of dysregulated metabolites on oocyte and embryo development. Our findings demonstrated that PCOS women exhibited distinct metabolic features in follicles, such as the increase in fatty acid utilization and the downregulation in amino acid metabolism. And intrafollicular androgen levels were positively correlated with contents of multiple fatty acids, suggesting androgen as one of the contributing factors to the metabolic abnormalities in PCOS follicles. Moreover, we further demonstrated that elevated levels of α-linolenic acid and H3K27me3 could hinder oocyte maturation, fertilization, and early embryo development. Hopefully, our data serve as a broad resource on the metabolic abnormalities of PCOS follicles, and advances in the relevant knowledge will allow the identification of biomarkers that predict the progression of PCOS and its poor pregnancy outcomes.
Collapse
Affiliation(s)
- Yue Xu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
| | - Zhiyang Zhou
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
| | - Gaochen Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
| | - Zuwei Yang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China; The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Shi
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
| | - Zhaoying Jiang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ye Liu
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huixi Chen
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China; The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China; The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yu Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China.
| | - Jiexue Pan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China; The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
29
|
Dai M, Hong L, Yin T, Liu S. Disturbed Follicular Microenvironment in Polycystic Ovary Syndrome: Relationship to Oocyte Quality and Infertility. Endocrinology 2024; 165:bqae023. [PMID: 38375912 DOI: 10.1210/endocr/bqae023] [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: 10/22/2023] [Revised: 02/15/2024] [Accepted: 02/15/2024] [Indexed: 02/21/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine disorder associated with infertility and poor reproductive outcomes. The follicular fluid (FF) microenvironment plays a crucial role in oocyte development. This review summarizes evidence elucidating the alterations in FF composition in PCOS. Various studies demonstrated a pronounced proinflammatory milieu in PCOS FF, characterized by increased levels of cytokines, including but not limited to interleukin-6 (IL-6), tumor necrosis factor α, C-reactive protein, and IL-1β, concomitant with a reduction in anti-inflammatory IL-10. T lymphocytes and antigen-presenting cells are dysregulated in PCOS FF. PCOS FF exhibit heightened reactive oxygen species production and the accumulation of lipid peroxidation byproducts, and impaired antioxidant defenses. Multiple microRNAs are dysregulated in PCOS FF, disrupting signaling critical to granulosa cell function. Proteomic analysis reveals changes in pathways related to immune responses, metabolic perturbations, angiogenesis, and hormone regulation. Metabolomics identify disturbances in glucose metabolism, amino acids, lipid profiles, and steroid levels with PCOS FF. Collectively, these pathological alterations may adversely affect oocyte quality, embryo development, and fertility outcomes. Further research on larger cohorts is needed to validate these findings and to forge the development of prognostic biomarkers of oocyte developmental competence within FF. Characterizing the follicular environment in PCOS is key to elucidating the mechanisms underlying subfertility in this challenging disorder.
Collapse
Affiliation(s)
- Mengyang Dai
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan 430061, China
| | - Ling Hong
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen 518000, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen 518000, China
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan 430061, China
| | - Su Liu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen 518000, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen 518000, China
| |
Collapse
|
30
|
Lecová M, Babjáková D, Sopková D, Andrejčáková Z, Hertelyová Z, Petrilla V, Polláková M, Vlčková R. Different Lengths of Diet Supplementation with 10% Flaxseed Alter the Hormonal Profile and the Follicular Fluid Fatty Acid Content of Fattening Gilts. Life (Basel) 2024; 14:240. [PMID: 38398749 PMCID: PMC10890391 DOI: 10.3390/life14020240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
The effect of 10% dietary flaxseed fed for 3 and 6 weeks on serum hormone levels of fattening gilts, the fatty acid (FA) follicular fluid (FF) composition of small and large antral follicles, and the steroidogenesis and IGF-I secretion by isolated small antral follicles and their response to regulatory hormones (LH, FSH, IGF-I) was studied using immunoassay and gas chromatography analyses. Both supplemental periods increased levels of P4 and IGF-I in blood serum. A shorter period inhibited steroidogenesis (P4, T, E2) and IGF-I secretion by small antral follicles, which was associated with decreased levels of monounsaturated FAs (MUFA) and preferred n-6 polyunsaturated FA (PUFA) metabolism. A longer period stimulated hormone secretion at elevated levels of saturated FAs (SFA) at the expense of MUFAs and PUFAs preferring the n-3 PUFA metabolism. Out of ovarian regulators, only LH and IGF-I were able to alter the secretion of steroids and IGF-I by small follicles of fattening pigs fed a basal diet. The effect of flaxseed on the secretion of follicular hormones after both supplemental periods was altered by all regulatory hormones in a dose-dependent manner. The level of SFAs and PUFAs in FF of large follicles increased with the length of flaxseed feeding, suggesting the suppression of ovulation.
Collapse
Affiliation(s)
- Martina Lecová
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia; (M.L.); (D.B.); (D.S.); (Z.A.); (V.P.); (M.P.)
| | - Diana Babjáková
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia; (M.L.); (D.B.); (D.S.); (Z.A.); (V.P.); (M.P.)
| | - Drahomíra Sopková
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia; (M.L.); (D.B.); (D.S.); (Z.A.); (V.P.); (M.P.)
| | - Zuzana Andrejčáková
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia; (M.L.); (D.B.); (D.S.); (Z.A.); (V.P.); (M.P.)
| | - Zdenka Hertelyová
- Department of Experimental Medicine, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Šrobárova 2, 041 80 Košice, Slovakia;
| | - Vladimír Petrilla
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia; (M.L.); (D.B.); (D.S.); (Z.A.); (V.P.); (M.P.)
| | - Magdaléna Polláková
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia; (M.L.); (D.B.); (D.S.); (Z.A.); (V.P.); (M.P.)
| | - Radoslava Vlčková
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia; (M.L.); (D.B.); (D.S.); (Z.A.); (V.P.); (M.P.)
| |
Collapse
|
31
|
Sachs MK, Makieva S, Velasco Gil A, Xie M, Ille F, Salvadori V, Schmidhauser M, Saenz-de-Juano MD, Ulbrich SE, Leeners B. Transcriptomic signature of luteinized cumulus cells of oocytes developing to live birth after women received intracytoplasmic sperm injection. F&S SCIENCE 2024; 5:24-38. [PMID: 38036000 DOI: 10.1016/j.xfss.2023.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/02/2023]
Abstract
OBJECTIVE To compare the transcriptome of human cumulus cells (CCs) from oocytes with different outcomes (pregnancy yes/no, live birth [LB] yes/no), to identify noninvasive biomarkers for oocyte selection as well as new therapeutic targets to increase LB rates from assisted reproductive technologies (ART). DESIGN Retrospective observational study. SETTINGS This study was conducted at a University Hospital in Switzerland. PATIENTS Subfertile couples undergoing controlled ovarian superstimulation and intracytoplasmic sperm injection with subsequent unbiopsied embryo transfer below the female age of 43 years. INTERVENTION(S) RNA sequencing of CCs from oocytes results in a pregnancy, no pregnancy, LB, or no LB. MAIN OUTCOME MEASURES Differential gene expression (DEG) between CCs of oocytes results in "no pregnancy" vs. "pregnancy" and "pregnancy only" vs. "live birth." RESULTS Although RNA sequencing did not reveal DEGs when comparing the transcriptomic profiles of the groups "no pregnancy" with "pregnancy," we identified 139 DEGs by comparing "pregnancy only" with "live birth," of which 28 belonged to clusters relevant to successful ART outcomes (i.e., CTGF, SERPINE2, PCK1, HHIP, HS3ST, and BIRC5). A functional enrichment analysis revealed that the transcriptome of CCs associated with LB depicts pathways of extracellular matrix, inflammatory cascades leading to ovulation, cell patterning, proliferation, and differentiation, and silencing pathways leading to apoptosis. CONCLUSION We identified a CCs transcriptomic profile associated with LB after embryo transfer that, after further validation, could serve to predict successful ART outcomes. The definition of relevant pathways of CCs related to oocyte competency contributes to a broader understanding of the cumulus oocyte complex and helps identify further therapeutic targets for improving ART success.
Collapse
Affiliation(s)
- Maike K Sachs
- Department of Reproductive Endocrinology, University Hospital Zurich, Frauenklinikstrasse, Zurich, Switzerland.
| | - Sofia Makieva
- Department of Reproductive Endocrinology, University Hospital Zurich, Frauenklinikstrasse, Zurich, Switzerland
| | - Ana Velasco Gil
- Department of Reproductive Endocrinology, University Hospital Zurich, Frauenklinikstrasse, Zurich, Switzerland
| | - Min Xie
- Department of Reproductive Endocrinology, University Hospital Zurich, Frauenklinikstrasse, Zurich, Switzerland
| | - Fabian Ille
- Center of Bioscience and Medical Engineering, Institute of Medical Engineering, Lucerne University of Applied Sciences and Arts, Obermattweg, Hergiswil, Switzerland
| | - Vincent Salvadori
- Center of Bioscience and Medical Engineering, Institute of Medical Engineering, Lucerne University of Applied Sciences and Arts, Obermattweg, Hergiswil, Switzerland
| | - Meret Schmidhauser
- ETH Zürich, Animal Physiology, Institute of Agricultural Sciences, Universitätsstrasse Zurich, Switzerland
| | - Mara D Saenz-de-Juano
- ETH Zürich, Animal Physiology, Institute of Agricultural Sciences, Universitätsstrasse Zurich, Switzerland
| | - Susanne E Ulbrich
- ETH Zürich, Animal Physiology, Institute of Agricultural Sciences, Universitätsstrasse Zurich, Switzerland
| | - Brigitte Leeners
- Department of Reproductive Endocrinology, University Hospital Zurich, Frauenklinikstrasse, Zurich, Switzerland
| |
Collapse
|
32
|
Athar F, Karmani M, Templeman N. Metabolic hormones are integral regulators of female reproductive health and function. Biosci Rep 2024; 44:BSR20231916. [PMID: 38131197 PMCID: PMC10830447 DOI: 10.1042/bsr20231916] [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: 11/03/2023] [Revised: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023] Open
Abstract
The female reproductive system is strongly influenced by nutrition and energy balance. It is well known that food restriction or energy depletion can induce suppression of reproductive processes, while overnutrition is associated with reproductive dysfunction. However, the intricate mechanisms through which nutritional inputs and metabolic health are integrated into the coordination of reproduction are still being defined. In this review, we describe evidence for essential contributions by hormones that are responsive to food intake or fuel stores. Key metabolic hormones-including insulin, the incretins (glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1), growth hormone, ghrelin, leptin, and adiponectin-signal throughout the hypothalamic-pituitary-gonadal axis to support or suppress reproduction. We synthesize current knowledge on how these multifaceted hormones interact with the brain, pituitary, and ovaries to regulate functioning of the female reproductive system, incorporating in vitro and in vivo data from animal models and humans. Metabolic hormones are involved in orchestrating reproductive processes in healthy states, but some also play a significant role in the pathophysiology or treatment strategies of female reproductive disorders. Further understanding of the complex interrelationships between metabolic health and female reproductive function has important implications for improving women's health overall.
Collapse
Affiliation(s)
- Faria Athar
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Muskan Karmani
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Nicole M. Templeman
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| |
Collapse
|
33
|
Jin LY, Yu JE, Xu HY, Chen B, Yang Q, Liu Y, Guo MX, Zhou CL, Cheng Y, Pang HY, Wu HY, Sheng JZ, Huang HF. Overexpression of Pde4d in rat granulosa cells inhibits maturation and atresia of antral follicles to induce polycystic ovary. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166869. [PMID: 37673361 DOI: 10.1016/j.bbadis.2023.166869] [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: 04/06/2023] [Revised: 08/08/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND Follicle dysplasia can cause polycystic ovary syndrome, which can lead to anovulatory infertility. This study explored gene(s) that may contribute to polycystic ovary syndrome. METHODS Three animal models of polycystic ovary syndrome were created by treating 3-week-old rats respectively with estradiol valerate, testosterone propionate, or constant illumination for 8 weeks. Granulosa cells from the three disease groups and from healthy controls were transcriptionally profiled to identify differentially expressed genes. The phosphodiesterase-4d (Pde4d) was screened as the most promising candidate pathogenic gene. The Pde4d was overexpressed in rats via intrabursal infection with recombinant lentivirus to see the effect of Pde4d on ovarian morphology. The potential roles of the candidate gene and interactors of the encoded protein were explored using polymerase chain reaction, western blotting, transfection and co-immunoprecipitation. RESULTS All three rat models of polycystic ovary syndrome showed polycystic ovary phenotype. Seven promising candidate genes were obtained by transcriptomics and verifications. Pde4d was further investigated because it could trigger downstream signaling pathways. The Pde4d overexpression in rat ovary induced cystic follicles. It inhibited follicle maturation through a mechanism involving inhibition of cAMP-PKA-CREB signaling. The Pde4d also inhibited phosphorylation of c-Jun N-terminal kinase to reduce apoptosis in the ovary, through a mechanism involving interaction of its poly-proline domain with the protein POSH. CONCLUSION Upregulation of Pde4d may contribute to polycystic ovary syndrome by impeding follicle maturation and preventing apoptotic atresia.
Collapse
Affiliation(s)
- Lu-Yang Jin
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Department of Gynecology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jia-En Yu
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Hai-Yan Xu
- Reproductive Medicine Center, Ningbo First Hospital, School of Medicine, Zhejiang University, Ningbo, Zhejiang, China
| | - Bin Chen
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, Zhejiang 310016, China
| | - Qian Yang
- International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ye Liu
- International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Meng-Xi Guo
- International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng-Liang Zhou
- International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Cheng
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China
| | - Hai-Yan Pang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Hai-Yan Wu
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Jian-Zhong Sheng
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - He-Feng Huang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200030, China.
| |
Collapse
|
34
|
Nagyova E, Mlynarcikova AB, Nemcova L, Scsukova S. Unique hyaluronan structure of expanded oocyte-cumulus extracellular matrix in ovarian follicles. Endocr Regul 2024; 58:174-180. [PMID: 39121477 DOI: 10.2478/enr-2024-0020] [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] [Indexed: 08/11/2024] Open
Abstract
In preovulatory follicles, after the endogenous gonadotropin surge, the oocyte-cumulus complexes (OCCs) produce hyaluronan (HA) in a process called "cumulus expansion". During this process, the heavy chains (HCs) of the serum-derived inter-alpha-trypsin inhibitor (IαI) family bind covalently to synthesized HA and form a unique structure of the expanded cumulus HA-rich extracellular matrix. Understanding the biochemical mechanism of the covalent linkage between HA and the HCs of the IαI family is one of the most significant discoveries in reproductive biology, since it explains basis of the cumulus expansion process running in parallel with the oocyte maturation, both essential for ovulation. Two recent studies have supported the above-mentioned findings: in the first, seven components of the extracellular matrix were detected by proteomic, evolutionary, and experimental analyses, and in the second, the essential role of serum in the process of cumulus expansion in vitro was confirmed. We have previously demonstrated the formation of unique structure of the covalent linkage of HA to HCs of IαI in the expanded gonadotropin-stimulated OCC, as well as interactions with several proteins produced by the cumulus cells: tumor necrosis factor-alpha-induced protein 6, pentraxin 3, and versican. Importantly, deletion of these genes in the mice produces female infertility due to defects in the oocyte-cumulus structure.
Collapse
Affiliation(s)
- Eva Nagyova
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Libechov, Czech Republic
| | | | - Lucie Nemcova
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Libechov, Czech Republic
| | - Sona Scsukova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
35
|
Fattahi A, Zarezadeh R, Rastgar Rezaei Y, Mettler L, Nouri M, Schmutzler AG, Salmassi A. Expression of interleukin-1β and its receptor in human granulosa cells and their association with steroidogenesis. Tissue Cell 2023; 85:102230. [PMID: 37820554 DOI: 10.1016/j.tice.2023.102230] [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: 03/27/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
This study aimed to investigate whether interleukin 1β (IL-1β) and soluble IL-1 receptor 2 (sIL-1R2) are expressed in human granulosa cells (GCs) and relate to ovarian steroidogenesis. Ninety-six women undergoing in vitro fertilization (IVF) were recruited. RT-PCR and immunocytochemistry were used to detect mRNAs and proteins of IL-1β and IL-1R2, respectively. The steroidogenesis of primary cultured GCs was evaluated following treatment with either IL-1β alone or IL-1β and FSH in combination. There were positive correlations between serum IL-1β and serum progesterone (r = 0.220, p = 0.032) and follicular fluid (FF) estradiol (r = 0.242, p = 0.018). Additionally, serum and FF sIL-1R2 were negatively and positively correlated with FF estradiol (r = -0.376, p = 0.005) and FF progesterone (r = 0.434, p = 0.001), respectively. The mRNA and protein expression of IL-1β and IL-1R2 became evident in GCs. IL-1β alone significantly increased estradiol secretion from GCs, but in the presence of FSH, it could notably promote progesterone secretion in addition to estradiol. In conclusion, IL-1β and sIL-1R2 are expressed in human GCs and substantially contribute to ovarian steroidogenesis, suggesting that the IL-1β system may be a potential target for optimizing ovarian hyperstimulation and steroidogenesis in IVF cycles.
Collapse
Affiliation(s)
- Amir Fattahi
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yeganeh Rastgar Rezaei
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Liselotte Mettler
- Department of Gynecology & Obstetrics, University Hospitals Schleswig-Holstein, Campus Kiel, Germany
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Andreas G Schmutzler
- Department of Gynecology & Obstetrics, University Hospitals Schleswig-Holstein, Campus Kiel, Germany.
| | - Ali Salmassi
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Gynecology & Obstetrics, University Hospitals Schleswig-Holstein, Campus Kiel, Germany.
| |
Collapse
|
36
|
Yao W, Liu C, Qin DY, Yuan XQ, Yao QY, Li NJ, Huang Y, Rao WT, Li YY, Deng YL, Zeng Q, Li YF. Associations between Phthalate Metabolite Concentrations in Follicular Fluid and Reproductive Outcomes among Women Undergoing in Vitro Fertilization/Intracytoplasmic Sperm Injection Treatment. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:127019. [PMID: 38150316 PMCID: PMC10752415 DOI: 10.1289/ehp11998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Phthalates have been reported to impair fertility in various studies. However, evidence exploring the associations between phthalate metabolites in follicular fluid (FF) and reproductive outcomes is lacking. OBJECTIVES To investigate the associations between phthalate metabolite concentrations in FF and in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) outcomes among women recruited from a fertility clinic. METHODS We included 641 women undergoing IVF/ICSI treatment from December 2018 to January 2020. The levels of eight phthalate metabolites, including monoethyl phthalate (MEP), mono-isobutyl phthalate (MiBP), mono-n -butyl phthalate (MBP), monobenzyl phthalate (MBzP), mono(2-ethylhexyl) phthalate (MEHP), mono(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP), mono(2-ethyl-5-oxohexyl) phthalate (MEOHP), and mono(2-ethyl-5-carboxypentyl) phthalate (MECPP), were quantified in FF collected on the oocyte retrieval day. Associations between quartiles of individual phthalate metabolite concentrations and nine IVF/ICSI outcomes, including oocyte yield, mature oocyte number, two distinct pronuclei (2PN) zygote number, fertilization rate, blastocyst formation rate, implantation, clinical pregnancy, miscarriage, and live birth, were estimated with generalized linear models. The effects of phthalate mixtures on IVF/ICSI outcomes were assessed using Bayesian kernel machine regression (BKMR) models. RESULTS After adjusting for relevant confounders, elevated quartiles of MBzP, MEHHP, and MEHP in FF were inversely associated with the numbers of retrieved oocytes, mature oocytes, and 2PN zygotes (all p for trends < 0.10 ). In comparison with the lowest quartile, the highest quartile of molar sum of di(2-ethylhexyl) phthalate metabolites (Σ DEHP ) was associated with a reduction of 9.1% [95% confidence interval (CI): - 17.1 % , - 0.37 % ] and 10.3% (95% CI: - 18.8 % , - 0.94 % ) in yielded oocyte and mature oocyte numbers, respectively. Furthermore, the BKMR models revealed inverse associations between phthalate mixtures and the numbers of retrieved oocytes and mature oocytes. We generally found null results for implantation, clinical pregnancy, miscarriage, and live birth. DISCUSSION Certain phthalate metabolites in FF are inversely associated with the numbers of retrieved oocytes, mature oocytes, and 2PN zygotes among women undergoing IVF/ICSI treatment. https://doi.org/10.1289/EHP11998.
Collapse
Affiliation(s)
- Wen Yao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Chong Liu
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan-Yu Qin
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Xiao-Qiong Yuan
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Qing-Yun Yao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Ni-Jie Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yong Huang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Wen-Tao Rao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yu-Ying Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yan-Ling Deng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Qiang Zeng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yu-Feng Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| |
Collapse
|
37
|
Sun N, Chen Y, Lu L, Yan H, Zhou J, Li K, Zhang W, Yuan L, Heng BC, Zeng W, Shi Y, Tong G, Yin P. Peptidomic analysis of follicular fluid in patients with polycystic ovarian syndrome. Front Cell Dev Biol 2023; 11:1289063. [PMID: 38020909 PMCID: PMC10666747 DOI: 10.3389/fcell.2023.1289063] [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: 09/05/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Objective: The aim of this study was to analyze and compare the differential expression of peptides within the follicular fluid of polycystic ovary syndrome (PCOS) patients versus normal women by using peptidomics techniques. The underlying mechanisms involved in PCOS pathogenesis will be explored, together with screening and identification of potential functional peptides via bioinformatics analysis. Materials and methods: A total of 12 patients who underwent in vitro fertilization and embryo transfer (IVF-ET) at the Reproductive Medicine Center of Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine from 1 September 2022 to 1 November 2022 were included in this study. The follicular fluid of PCOS patients (n = 6) and normal women (n = 6) were collected. The presence and concentration differences of various peptides were detected by the LC-MS/MS method. GO and KEGG analysis were performed on the precursor proteins of the differentially-expressed peptides, and protein network interaction analysis was carried out to identify functionally-relevant peptides among the various peptides. Results: A variety of peptides within the follicular fluid of PCOS versus normal patients were detected by peptidomics techniques. Altogether, 843 upregulated peptides and 236 downregulated peptides were detected (absolute fold change ≥2 and p < 0.05). Of these, 718 (718 = 488 + 230) peptides were only detected in the PCOS group, while 205 (205 = 174 + 31) were only detected in the control group. Gene Ontology enrichment and pathway analysis were performed to characterize peptides through their precursor proteins. We identified 18 peptides from 7 precursor proteins associated with PCOS, and 4 peptide sequences were located in the functional domains of their corresponding precursor proteins. Conclusion: In this study, differences in the follicular development of PCOS versus normal patients were revealed from the polypeptidomics of follicular development, which thus provided new insights for future studies on the pathological mechanisms of PCOS development.
Collapse
Affiliation(s)
- Ningyu Sun
- Department of Reproductive Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanyuan Chen
- Department of Reproductive Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Lu
- Department of Reproductive Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Yan
- Department of Reproductive Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Zhou
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Kai Li
- Department of Reproductive Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wuwen Zhang
- Department of Reproductive Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lihua Yuan
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Boon Chin Heng
- School of Stomatology, Peking University, Beijing, China
| | - Weiwei Zeng
- Department of Gynecology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yin Shi
- Key Laboratory of Acupuncture and Immunological Effects, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guoqing Tong
- Department of Reproductive Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Ping Yin
- Department of Reproductive Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
38
|
Cadenas J, Pors SE, Hansen CP, Olufsen SM, Subiran C, Bøtkjær JA, La Cour Poulsen L, Fedder J, Dueholm M, Colmorn LB, Kristensen SG, Mamsen LS, Andersen CY. Midkine characterization in human ovaries: potential new variants in follicles. F&S SCIENCE 2023; 4:294-301. [PMID: 37739342 DOI: 10.1016/j.xfss.2023.09.003] [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: 07/14/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
OBJECTIVE To characterize the growth factor midkine (MDK) in the human ovary to determine whether MDK is produced locally within the ovary, examine whether different ovarian cell types are more likely to produce MDK, and determine whether there are any stage-specific variations during follicle growth. Previous studies have revealed that MDK potentially affects human follicle growth and oocyte maturation. Proteomic analyses in follicular fluid (FF) have identified MDK to functionally cluster together and follow a similar expression profile to that of well-known proteins involved in ovarian follicle development. Midkine has not yet been characterized in the human ovary. DESIGN Descriptive study. SETTING University Hospital. PATIENTS The study included samples from 121 patients: 71 patients (aged 17-37 years) who underwent ovarian tissue cryopreservation provided granulosa cells (GC), cumulus cells, ovarian cortex, medulla tissue, and FF from small antral follicles (SAF); and 50 patients (aged 20-35 years) receiving in vitro fertilization treatment provided FF from preovulatory follicles before and after induction of final follicle maturation. INTERVENTIONS None. MAIN OUTCOME MEASURES MDK relative gene expression was quantified using a real-time quantitative polymerase chain reaction in cumulus cells, GC, and medulla tissue. Additionally, immunostaining and western blotting assays were used to detect MDK protein in the ovarian cortex, which contains preantral follicles, SAF, and medulla tissue. Furthermore, enzyme-linked immunosorbent assay analyses were performed to measure the concentration of MDK in FF aspirated from SAF and preovulatory follicles both before and 36 hours after inducing the final maturation of follicles. RESULTS Immunostaining and reverse transcription-quantitative polymerase chain reaction revealed a more prominent expression of MDK in GC compared with other ovarian cell types. Intrafollicular MDK concentration was significantly higher in SAF compared with preovulatory follicles. In addition, different molecular weight species of MDK were detected using western blotting in various ovarian sample types: GC and FF samples presented primarily one band of approximately 15 kDa and an additional band of approximately 13 kDa, although other bands with higher molecular weight (between 30 and 38 kDa) were detected in medulla tissue. CONCLUSIONS This is the first time that MDK has been immunolocalized in human ovarian cells at the protein level and that potentially different MDK variants have been detected in human FF, GC, and ovarian medulla tissue. Future studies are needed to sequence and identify the different potential MDK variants found to determine their functional importance for ovary and oocyte competence.
Collapse
Affiliation(s)
- Jesús Cadenas
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark.
| | - Susanne Elisabeth Pors
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | | | - Sarah Maria Olufsen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Cristina Subiran
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Jane Alrø Bøtkjær
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Liv La Cour Poulsen
- The Fertility Clinic, Copenhagen University Hospital, Herlev Hospital, Borgmester Ib Juuls Vej 9, Herlev, Denmark
| | - Jens Fedder
- Centre of Andrology and Fertility Clinic, Odense University Hospital, Odense, Denmark; Research Unit of Human Reproduction, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Margit Dueholm
- Department of Gynecology and Obstetrics, Aarhus University Hospital, Aarhus, Denmark
| | | | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Linn Salto Mamsen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Claus Yding Andersen
- The Department of Clinical Medicine, Faculty of Health and Medical Science, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
39
|
Babayev E, Suebthawinkul C, Gokyer D, Parkes WS, Rivas F, Pavone ME, Hall AR, Pritchard MT, Duncan FE. Cumulus expansion is impaired with advanced reproductive age due to loss of matrix integrity and reduced hyaluronan. Aging Cell 2023; 22:e14004. [PMID: 37850336 PMCID: PMC10652338 DOI: 10.1111/acel.14004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 10/19/2023] Open
Abstract
Reproductive aging is associated with ovulatory defects. Age-related ovarian fibrosis partially contributes to this phenotype as short-term treatment with anti-fibrotic compounds improves ovulation in reproductively old mice. However, age-dependent changes that are intrinsic to the follicle may also be relevant. In this study, we used a mouse model to demonstrate that reproductive aging is associated with impaired cumulus expansion which is accompanied by altered morphokinetic behavior of cumulus cells as assessed by time-lapse microscopy. The extracellular matrix integrity of expanded cumulus-oocyte complexes is compromised with advanced age as evidenced by increased penetration of fluorescent nanoparticles in a particle exclusion assay and larger open spaces on scanning electron microscopy. Reduced hyaluronan (HA) levels, decreased expression of genes encoding HA-associated proteins (e.g., Ptx3 and Tnfaip6), and increased expression of inflammatory genes and matrix metalloproteinases underlie this loss of matrix integrity. Importantly, HA levels are decreased with age in follicular fluid of women, indicative of conserved reproductive aging mechanisms. These findings provide novel mechanistic insights into how defects in cumulus expansion contribute to age-related infertility and may serve as a target to extend reproductive longevity.
Collapse
Affiliation(s)
- Elnur Babayev
- Department of Obstetrics and Gynecology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Chanakarn Suebthawinkul
- Department of Obstetrics and Gynecology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Department of Obstetrics and Gynecology, Faculty of MedicineChulalongkorn UniversityBangkokThailand
| | - Dilan Gokyer
- Department of Obstetrics and Gynecology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Wendena S. Parkes
- Department of Pharmacology, Toxicology, & Therapeutics, Institute for Reproductive and Developmental SciencesUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Felipe Rivas
- Virginia Tech‐Wake Forest University School of Biomedical Engineering and SciencesWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Mary Ellen Pavone
- Department of Obstetrics and Gynecology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Adam R. Hall
- Virginia Tech‐Wake Forest University School of Biomedical Engineering and SciencesWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Michele T. Pritchard
- Department of Pharmacology, Toxicology, & Therapeutics, Institute for Reproductive and Developmental SciencesUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Francesca E. Duncan
- Department of Obstetrics and Gynecology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
40
|
Dipali SS, Suebthawinkul C, Burdette JE, Pavone ME, Duncan FE. Human follicular fluid elicits select dose- and age-dependent effects on mouse oocytes and cumulus-oocyte complexes in a heterologous in vitro maturation assay. Mol Hum Reprod 2023; 29:gaad039. [PMID: 37950499 PMCID: PMC10674105 DOI: 10.1093/molehr/gaad039] [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: 01/20/2023] [Revised: 10/11/2023] [Indexed: 11/12/2023] Open
Abstract
Follicular fluid (FF) is a primary microenvironment of the oocyte within an antral follicle. Although several studies have defined the composition of human FF in normal physiology and determined how it is altered in disease states, the direct impacts of human FF on the oocyte are not well understood. The difficulty of obtaining suitable numbers of human oocytes for research makes addressing such a question challenging. Therefore, we used a heterologous model in which we cultured mouse oocytes in human FF. To determine whether FF has dose-dependent effects on gamete quality, we performed in vitro maturation of denuded oocytes from reproductively young mice (6-12 weeks) in 10%, 50%, or 100% FF from participants of mid-reproductive age (32-36 years). FF impacted meiotic competence in a dose-dependent manner, with concentrations >10% inhibiting meiotic progression and resulting in spindle and chromosome alignment defects. We previously demonstrated that human FF acquires a fibro-inflammatory cytokine signature with age. Thus, to determine whether exposure to an aging FF microenvironment contributes to the age-dependent decrease in gamete quality, we matured denuded oocytes and cumulus-oocyte complexes (COCs) in FF from reproductively young (28-30 years) and old (40-42 years) participants. FF decreased meiotic progression of COCs, but not oocytes, from reproductively young and old (9-12 months) mice in an age-dependent manner. Moreover, FF had modest age-dependent impacts on mitochondrial aggregation in denuded oocytes and cumulus layer expansion dynamics in COCs, which may influence fertilization or early embryo development. Overall, these findings demonstrate that acute human FF exposure can impact select markers of mouse oocyte quality in both dose- and age-dependent manners.
Collapse
Affiliation(s)
- Shweta S Dipali
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Chanakarn Suebthawinkul
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Joanna E Burdette
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Mary Ellen Pavone
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
41
|
Zaniker EJ, Babayev E, Duncan FE. Common mechanisms of physiological and pathological rupture events in biology: novel insights into mammalian ovulation and beyond. Biol Rev Camb Philos Soc 2023; 98:1648-1667. [PMID: 37157877 PMCID: PMC10524764 DOI: 10.1111/brv.12970] [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: 08/11/2022] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/10/2023]
Abstract
Ovulation is a cyclical biological rupture event fundamental to fertilisation and endocrine function. During this process, the somatic support cells that surround the germ cell undergo a remodelling process that culminates in breakdown of the follicle wall and release of a mature egg. Ovulation is driven by known proteolytic and inflammatory pathways as well as structural alterations to the follicle vasculature and the fluid-filled antral cavity. Ovulation is one of several types of systematic remodelling that occur in the human body that can be described as rupture. Although ovulation is a physiological form of rupture, other types of rupture occur in the human body which can be pathological, physiological, or both. In this review, we use intracranial aneurysms and chorioamniotic membrane rupture as examples of rupture events that are pathological or both pathological and physiological, respectively, and compare these to the rupture process central to ovulation. Specifically, we compared existing transcriptomic profiles, immune cell functions, vascular modifications, and biomechanical forces to identify common processes that are conserved between rupture events. In our transcriptomic analysis, we found 12 differentially expressed genes in common among two different ovulation data sets and one intracranial aneurysm data set. We also found three genes that were differentially expressed in common for both ovulation data sets and one chorioamniotic membrane rupture data set. Combining analysis of all three data sets identified two genes (Angptl4 and Pfkfb4) that were upregulated across rupture systems. Some of the identified genes, such as Rgs2, Adam8, and Lox, have been characterised in multiple rupture contexts, including ovulation. Others, such as Glul, Baz1a, and Ddx3x, have not yet been characterised in the context of ovulation and warrant further investigation as potential novel regulators. We also identified overlapping functions of mast cells, macrophages, and T cells in the process of rupture. Each of these rupture systems share local vasoconstriction around the rupture site, smooth muscle contractions away from the site of rupture, and fluid shear forces that initially increase and then decrease to predispose one specific region to rupture. Experimental techniques developed to study these structural and biomechanical changes that underlie rupture, such as patient-derived microfluidic models and spatiotemporal transcriptomic analyses, have not yet been comprehensively translated to the study of ovulation. Review of the existing knowledge, transcriptomic data, and experimental techniques from studies of rupture in other biological systems yields a better understanding of the physiology of ovulation and identifies avenues for novel studies of ovulation with techniques and targets from the study of vascular biology and parturition.
Collapse
Affiliation(s)
- Emily J. Zaniker
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 10-109, Chicago, IL 60611, USA
| | - Elnur Babayev
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 10-109, Chicago, IL 60611, USA
| | - Francesca E. Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 10-109, Chicago, IL 60611, USA
| |
Collapse
|
42
|
Zhou Z, Zhang Y, Zhang X, Zhang J, Yi G, Wan B, Li Y, Lu H, Tan C, Lu W. Follicular Fluid-Derived Small Extracellular Vesicles Alleviate DHEA-Induced Granulosa Cell Apoptosis by Delivering LINC00092. Reprod Sci 2023; 30:3092-3102. [PMID: 37188981 DOI: 10.1007/s43032-023-01251-7] [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: 11/29/2022] [Accepted: 04/23/2023] [Indexed: 05/17/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a perplexing condition in females of reproductive age. Dysplasia of ovarian granulosa cell (GC) is implicated in PCOS. Follicular fluid (FF)-extracellular vesicles (Evs) are important in cell-cell communication during follicular development. The current study elaborated on the function and mechanism of FF-Evs in the viability and apoptosis of GC cells in PCOS development. Human GC cells KGN were treated with dehydroepiandrosterone (DHEA) to mimic a PCOS-like condition in vitro, which were further co-cultured with the FF-derived Evs (FF-Evs). The FF-Evs treatment significantly reduced DHEA-induced apoptosis of KGN cells while promoting cell viability and migration. The lncRNA microarray analysis showed that FF-Evs mainly deliver LINC00092 into the KGN cells. Knockdown of LINC00092 negated the protective effect of FF-Evs against DHEA-induced damage on KGN cells. Moreover, by performing bioinformatics analyses and biotin-labeled RNA pull-down assay, we found that LINC00092 could bind to the RNA binding protein LIN28B and inhibit its binding to pre-microRNA-18-5p, which allowed biogenesis of pre-miR-18-5p and increased the expression of miR-18b-5p, a miRNA with known alleviating role in PCOS by suppressing the PTEN mRNA. Collectively, the present work demonstrates that FF-Evs can alleviate DHEA-induced GC damage by delivering LINC00092.
Collapse
Affiliation(s)
- Zhi Zhou
- Reproductive Medical Center, Hainan Women and Children's Medical Center, 75 South Longkun Road, Haikou, 570206, Hainan, People's Republic of China
| | - Yong Zhang
- Department of Pharmacology, School of Basic Medicine and Life Science, Hainan Medical University, Haikou, 571199, Hainan, People's Republic of China
| | - Xiaopo Zhang
- Key Laboratory of Tropical Translational Medicine of the Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, Hainan, People's Republic of China
| | - Juan Zhang
- Reproductive Medical Center, Zhuzhou Central Hospital, Zhuzhou, 412007, Hunan, People's Republic of China
| | - Guohui Yi
- Public Research Laboratory, Hainan Medical University, Haikou, 571199, Hainan, People's Republic of China
| | - Bangbei Wan
- Reproductive Medical Center, Hainan Women and Children's Medical Center, 75 South Longkun Road, Haikou, 570206, Hainan, People's Republic of China
| | - Yejuan Li
- Reproductive Medical Center, Hainan Women and Children's Medical Center, 75 South Longkun Road, Haikou, 570206, Hainan, People's Republic of China
| | - Hui Lu
- Reproductive Medical Center, Hainan Women and Children's Medical Center, 75 South Longkun Road, Haikou, 570206, Hainan, People's Republic of China
| | - Can Tan
- Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL, 60611, USA
| | - Weiying Lu
- Reproductive Medical Center, Hainan Women and Children's Medical Center, 75 South Longkun Road, Haikou, 570206, Hainan, People's Republic of China.
| |
Collapse
|
43
|
Bongaerts E, Mamia K, Rooda I, Björvang RD, Papaikonomou K, Gidlöf SB, Olofsson JI, Ameloot M, Alfaro-Moreno E, Nawrot TS, Damdimopoulou P. Ambient black carbon particles in human ovarian tissue and follicular fluid. ENVIRONMENT INTERNATIONAL 2023; 179:108141. [PMID: 37603992 DOI: 10.1016/j.envint.2023.108141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/10/2023] [Accepted: 08/09/2023] [Indexed: 08/23/2023]
Abstract
Evidence indicates a link between exposure to ambient air pollution and decreased female fertility. The ability of air pollution particles to reach human ovarian tissue and follicles containing the oocytes in various maturation stages has not been studied before. Particulate translocation might be an essential step in explaining reproductive toxicity and assessing associated risks. Here, we analysed the presence of ambient black carbon particles in (i) follicular fluid samples collected during ovum pick-up from 20 women who underwent assisted reproductive technology treatment and (ii) adult human ovarian tissue from 5 individuals. Follicular fluid and ovarian tissue samples were screened for the presence of black carbon particles from ambient air pollution using white light generation by carbonaceous particles under femtosecond pulsed laser illumination. We detected black carbon particles in all follicular fluid (n = 20) and ovarian tissue (n = 5) samples. Black carbon particles from ambient air pollution can reach the ovaries and follicular fluid, directly exposing the ovarian reserve and maturing oocytes. Considering the known link between air pollution and decreased fertility, the impact of such exposure on oocyte quality, ovarian ageing and fertility needs to be clarified urgently.
Collapse
Affiliation(s)
- Eva Bongaerts
- Centre for Environmental Sciences, Hasselt University, 3590 Hasselt, Belgium
| | - Katariina Mamia
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 86 Huddinge, Sweden; Department of Gynaecology and Reproductive Medicine, Karolinska University Hospital, 141 86 Huddinge, Sweden
| | - Ilmatar Rooda
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 86 Huddinge, Sweden; Department of Gynaecology and Reproductive Medicine, Karolinska University Hospital, 141 86 Huddinge, Sweden
| | - Richelle D Björvang
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 86 Huddinge, Sweden; Department of Gynaecology and Reproductive Medicine, Karolinska University Hospital, 141 86 Huddinge, Sweden; Department of Women's and Children's Health, Uppsala University, 75185 Uppsala, Sweden
| | - Kiriaki Papaikonomou
- Department of Gynaecology and Reproductive Medicine, Karolinska University Hospital, 141 86 Huddinge, Sweden; Department of Women's and Children's Health, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Sebastian B Gidlöf
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 86 Huddinge, Sweden; Department of Gynaecology and Reproductive Medicine, Karolinska University Hospital, 141 86 Huddinge, Sweden; Department of Women's and Children's Health, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Jan I Olofsson
- Department of Women's and Children's Health, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Marcel Ameloot
- Biomedical Research Institute, Hasselt University, 3590 Hasselt, Belgium
| | - Ernesto Alfaro-Moreno
- Nanosafety Group, International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University, 3590 Hasselt, Belgium; Department of Public Health and Primary Care, KU Leuven, 3000 Leuven, Belgium
| | - Pauliina Damdimopoulou
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 86 Huddinge, Sweden; Department of Gynaecology and Reproductive Medicine, Karolinska University Hospital, 141 86 Huddinge, Sweden.
| |
Collapse
|
44
|
Bastos DCDS, Chiamolera MI, Silva RE, Souza MDCBD, Antunes RA, Souza MM, Mancebo ACA, Arêas PCF, Reis FM, Lo Turco EG, Bloise FF, Ortiga-Carvalho TM. Metabolomic analysis of follicular fluid from women with Hashimoto thyroiditis. Sci Rep 2023; 13:12497. [PMID: 37532758 PMCID: PMC10397241 DOI: 10.1038/s41598-023-39514-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/26/2023] [Indexed: 08/04/2023] Open
Abstract
Hashimoto thyroiditis is an autoimmune disease characterized by hypothyroidism and a high level of anti-thyroid autoantibodies. It has shown to negatively impact female fertility; however, the mechanisms are unclear. Ovarian follicular fluid appears to be the key to understanding how Hashimoto thyroiditis affecst fertility. Thus, we aimed to evaluated the metabolic profile of follicular fluid and antithyroid autoantibody levels in the context of Hashimoto thyroiditis. We collected follicular fluid from 61 patients, namely 38 women with thyroid autoantibody positivity and 23 women as negative controls, undergoing in vitro fertilization treatment. Follicular fluid samples were analyzed using metabolomics, and thyroid autoantibodies were measured. Fifteen metabolites with higher concentrations in the follicular fluid samples from Hashimoto thyroiditis were identified, comprising five possible affected pathways: the glycerophospholipid, arachidonic acid, linoleic acid, alpha-linolenic acid, and sphingolipid metabolism pathways. These pathways are known to regulate ovarian functions. In addition, antithyroglobulin antibody concentrations in both serum and follicular fluid were more than tenfold higher in women with Hashimoto thyroiditis than in controls. Our data showed that the metabolic profile of follicular fluid is altered in women with Hashimoto thyroiditis, suggesting a potential mechanistic explanation for the association of this disease with female infertility.
Collapse
Affiliation(s)
- Diana Caroline da Silva Bastos
- Laboratório de Endocrinologia Translacional, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Maria Izabel Chiamolera
- Laboratório de Endocrinologia Molecular e Translacional, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brasil
| | - Renata Elen Silva
- Laboratório de Endocrinologia Molecular e Translacional, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brasil
| | | | - Roberto Azevedo Antunes
- Fertipraxis Centro de Reproducao Humana, Rio de Janeiro, Brasil
- Maternidade Escola, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | | | | | | | - Fernando M Reis
- Departamento de Ginecologia e Obstetrícia, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | - Edson Guimarães Lo Turco
- Ion Medicine, São Paulo, Brasil
- Departamento de Cirurgia, Disciplina de Urologia, Setor de Reprodução Assistida Universidade Federal de São Paulo, Universidade Federal de São Paulo, São Paulo, Brasil
| | - Flavia Fonseca Bloise
- Laboratório de Endocrinologia Translacional, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Tania M Ortiga-Carvalho
- Laboratório de Endocrinologia Translacional, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
| |
Collapse
|
45
|
Wang X, Zhou S, Wu Z, Liu R, Ran Z, Liao J, Shi H, Wang F, Chen J, Liu G, Liang A, Yang L, Zhang S, Li X, He C. The FSH-mTOR-CNP signaling axis initiates follicular antrum formation by regulating tight junction, ion pumps, and aquaporins. J Biol Chem 2023; 299:105015. [PMID: 37414146 PMCID: PMC10424218 DOI: 10.1016/j.jbc.2023.105015] [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: 03/30/2023] [Revised: 06/20/2023] [Accepted: 06/28/2023] [Indexed: 07/08/2023] Open
Abstract
The initial formation of the follicular antrum (iFFA) serves as a dividing line between gonadotropin-independent and gonadotropin-dependent folliculogenesis, enabling the follicle to sensitively respond to gonadotropins for its further development. However, the mechanism underlying iFFA remains elusive. Herein, we reported that iFFA is characterized by enhanced fluid absorption, energy consumption, secretion, and proliferation and shares a regulatory mechanism with blastula cavity formation. By use of bioinformatics analysis, follicular culture, RNA interference, and other techniques, we further demonstrated that the tight junction, ion pumps, and aquaporins are essential for follicular fluid accumulation during iFFA, as a deficiency of any one of these negatively impacts fluid accumulation and antrum formation. The intraovarian mammalian target of rapamycin-C-type natriuretic peptide pathway, activated by follicle-stimulating hormone, initiated iFFA by activating tight junction, ion pumps, and aquaporins. Building on this, we promoted iFFA by transiently activating mammalian target of rapamycin in cultured follicles and significantly increased oocyte yield. These findings represent a significant advancement in iFFA research, further enhancing our understanding of folliculogenesis in mammals.
Collapse
Affiliation(s)
- Xiaodong Wang
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shanshan Zhou
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zian Wu
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ruiyan Liu
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zaohong Ran
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jianning Liao
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Hongru Shi
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Feng Wang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, USA
| | - Jianguo Chen
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Guoshi Liu
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Aixin Liang
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Liguo Yang
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shujun Zhang
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiang Li
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Changjiu He
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
46
|
Zhou XY, Yang YZ, Zhang J, Zhang XF, Liu YD, Wang Z, Chen SL. Elevated cell-free mitochondria DNA level of patients with premature ovarian insufficiency. BMC Pregnancy Childbirth 2023; 23:462. [PMID: 37349693 DOI: 10.1186/s12884-023-05769-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) patients present with a chronic inflammatory state. Cell-free mitochondria DNA (cf-mtDNA) has been explored as a reliable biomarker for estimating the inflammation-related disorders, however, the cf-mtDNA levels in POI patients have never been measured. Therefore, in the presenting study, we aimed to evaluate the levels of cf-mtDNA in plasma and follicular fluid (FF) of POI patients and to determine a potential role of cf-mtDNA in predicting the disease progress and pregnancy outcomes. METHODS We collected plasma and FF samples from POI patients, biochemical POI (bPOI) patients and control women. Quantitative real-time PCR was used to measure the ratio of mitochondrial genome to nuclear genome of cf-DNAs extracted from the plasma and FF samples. RESULTS The plasma cf-mtDNA levels, including COX3, CYB, ND1 and mtDNA79, were significantly higher in overt POI patients than those in bPOI patients or control women. The plasma cf-mtDNA levels were weakly correlated with ovarian reserve, and could not be improved by regular hormone replacement therapy. The levels of cf-mtDNA in FF, rather than those in plasma, exhibited the potential to predict the pregnancy outcomes, although they were comparable among overt POI, bPOI and control groups. CONCLUSIONS The increased plasma cf-mtDNA levels in overt POI patients indicated its role in the progress of POI and the FF cf-mtDNA content may hold the value in predicting pregnancy outcomes of POI patients.
Collapse
Affiliation(s)
- Xing-Yu Zhou
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China
| | - Yi-Zhen Yang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China
| | - Jun Zhang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China
| | - Xiao-Fei Zhang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China
| | - Yu-Dong Liu
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China
| | - Zhe Wang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China
| | - Shi-Ling Chen
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
47
|
Wyse BA, Salehi R, Russell SJ, Sangaralingam M, Jahangiri S, Tsang BK, Librach CL. Obesity and PCOS radically alters the snRNA composition of follicular fluid extracellular vesicles. Front Endocrinol (Lausanne) 2023; 14:1205385. [PMID: 37404312 PMCID: PMC10315679 DOI: 10.3389/fendo.2023.1205385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/29/2023] [Indexed: 07/06/2023] Open
Abstract
Introduction The ovarian follicle consists of the oocyte, somatic cells, and follicular fluid (FF). Proper signalling between these compartments is required for optimal folliculogenesis. The association between polycystic ovarian syndrome (PCOS) and extracellular vesicular small non-coding RNAs (snRNAs) signatures in follicular fluid (FF) and how this relates to adiposity is unknown. The purpose of this study was to determine whether FF extracellular vesicle (FFEV)-derived snRNAs are differentially expressed (DE) between PCOS and non-PCOS subjects; and if these differences are vesicle-specific and/or adiposity-dependent. Methods FF and granulosa cells (GC) were collected from 35 patients matched by demographic and stimulation parameters. FFEVs were isolated and snRNA libraries were constructed, sequenced, and analyzed. Results miRNAs were the most abundant biotype present, with specific enrichment in exosomes (EX), whereas in GCs long non-coding RNAs were the most abundant biotype. In obese PCOS vs. lean PCOS, pathway analysis revealed target genes involved in cell survival and apoptosis, leukocyte differentiation and migration, JAK/STAT, and MAPK signalling. In obese PCOS FFEVs were selectively enriched (FFEVs vs. GCs) for miRNAs targeting p53 signalling, cell survival and apoptosis, FOXO, Hippo, TNF, and MAPK signalling. Discussion We provide comprehensive profiling of snRNAs in FFEVs and GCs of PCOS and non-PCOS patients, highlighting the effect of adiposity on these findings. We hypothesize that the selective packaging and release of miRNAs specifically targeting anti-apoptotic genes into the FF may be an attempt by the follicle to reduce the apoptotic pressure of the GCs and stave off premature apoptosis of the follicle observed in PCOS.
Collapse
Affiliation(s)
- Brandon A. Wyse
- Research Department, CReATe Fertility Centre, Toronto, ON, Canada
| | - Reza Salehi
- Research Department, CReATe Fertility Centre, Toronto, ON, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Departments of Obstetrics and Gynecology & Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | | | | | - Sahar Jahangiri
- Research Department, CReATe Fertility Centre, Toronto, ON, Canada
- CReATe Biobank, Toronto, ON, Canada
| | - Benjamin K. Tsang
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Departments of Obstetrics and Gynecology & Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Clifford L. Librach
- Research Department, CReATe Fertility Centre, Toronto, ON, Canada
- CReATe Biobank, Toronto, ON, Canada
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Biological Sciences, DAN Women & Babies Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
48
|
Zhu M, Wang N, Wang S, Wang Y, Yang X, Fan J, Chen Y. Effects of Follicular Fluid on Physiological Characteristics and Differentiation of Fallopian Tube Epithelial Cells Implicating for Ovarian Cancer Pathogenesis. Int J Mol Sci 2023; 24:10154. [PMID: 37373301 DOI: 10.3390/ijms241210154] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The fallopian tube (FT) is an important reproductive organ in females. Ample evidence suggests that the distal end of FT is the original site of high-grade serous ovarian carcinoma (HGSC). FT may suffer from repeated injury and repair stimulated by follicular fluid (FF); however, this hypothesis has not been examined. In fact, the molecular mechanism of homeostasis, differentiation, and the transformation of fallopian tube epithelial cells (FTECs) resulting from the stimulation of FF are still enigmatic. In this study, we examined the effects of FF along with factors present in the FF on a variety of FTEC models, including primary cell culture, ALI (air-liquid interface) culture, and 3D organ spheroid culture. We found that FF plays a similar role to estrogen in promoting cell differentiation and organoid formation. Moreover, FF significantly promotes cell proliferation and induces cell injury and apoptosis in high concentrations. These observations may help us to investigate the mechanisms of the initiation of HGSC.
Collapse
Affiliation(s)
- Maobi Zhu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529000, China
| | - Na Wang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529000, China
| | - Sha Wang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529000, China
| | - Yao Wang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529000, China
| | - Xiawen Yang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529000, China
| | - Jianglin Fan
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529000, China
- Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Yajie Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529000, China
| |
Collapse
|
49
|
Viardot-Foucault V, Zhou J, Bi D, Takinami Y, Chan JKY, Lee YH. Dehydroepiandrosterone supplementation and the impact of follicular fluid metabolome and cytokinome profiles in poor ovarian responders. J Ovarian Res 2023; 16:107. [PMID: 37268990 PMCID: PMC10239139 DOI: 10.1186/s13048-023-01166-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/25/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Poor ovarian responders (POR) are women undergoing in-vitro fertilization who respond poorly to ovarian stimulation, resulting in the retrieval of lower number of oocytes, and subsequently lower pregnancy rates. The follicular fluid (FF) provides a crucial microenvironment for the proper development of follicles and oocytes through tightly controlled metabolism and cell signaling. Androgens such as dehydroepiandrosterone (DHEA) have been proposed to alter the POR follicular microenvironment, but the impact DHEA imposes on the FF metabolome and cytokine profiles is unknown. Therefore, the objective of this study is to profile and identify metabolomic changes in the FF with DHEA supplementation in POR patients. METHODS FF samples collected from 52 POR patients who underwent IVF with DHEA supplementation (DHEA +) and without (DHEA-; controls) were analyzed using untargeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) metabolomics and a large-scale multiplex suspension immunoassay covering 65 cytokines, chemokines and growth factors. Multivariate statistical modelling by partial least squares-discriminant regression (PLSR) analysis was performed for revealing metabolome-scale differences. Further, differential metabolite analysis between the two groups was performed by PLSR β-coefficient regression analysis and Student's t-test. RESULTS Untargeted metabolomics identified 118 FF metabolites of diverse chemistries and concentrations which spanned three orders of magnitude. They include metabolic products highly associated with ovarian function - amino acids for regulating pH and osmolarity, lipids such fatty acids and cholesterols for oocyte maturation, and glucocorticoids for ovarian steroidogenesis. Four metabolites, namely, glycerophosphocholine, linoleic acid, progesterone, and valine were significantly lower in DHEA + relative to DHEA- (p < 0.05-0.005). The area under the curves of progesterone glycerophosphocholine, linoleic acid and valine are 0.711, 0.730, 0.785 and 0.818 (p < 0.05-0.01). In DHEA + patients, progesterone positively correlated with IGF-1 (Pearson r: 0.6757, p < 0.01); glycerophosphocholine negatively correlated with AMH (Pearson r: -0.5815; p < 0.05); linoleic acid correlated with estradiol and IGF-1 (Pearson r: 0.7016 and 0.8203, respectively; p < 0.01 for both). In DHEA- patients, valine negatively correlated with serum-free testosterone (Pearson r: -0.8774; p < 0.0001). Using the large-scale immunoassay of 45 cytokines, we observed significantly lower MCP1, IFNγ, LIF and VEGF-D levels in DHEA + relative to DHEA. CONCLUSIONS In POR patients, DHEA supplementation altered the FF metabolome and cytokine profile. The identified four FF metabolites that significantly changed with DHEA may provide information for titrating and monitoring individual DHEA supplementation.
Collapse
Affiliation(s)
- Veronique Viardot-Foucault
- Department of Reproductive Medicine, KK Women’s and Children’s Hospital, 100 Bukit Timah Road, Singapore, 229899 Singapore
| | - Jieliang Zhou
- Translational ‘Omics and Biomarkers Group, KK Research Centre, KK Women’s and Children’s Hospital, 100 Bukit Timah Road, Singapore, 229899 Singapore
| | - Dexi Bi
- Department of Pathology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Yoshihiko Takinami
- Bruker Japan, 3-9 Yokohama City, Kanagawa, 220-0022 Japan
- Present Address: Kanomax Analytical Incorportated, Shimizu Suita City, Osaka Japan
| | - Jerry. K. Y. Chan
- Department of Reproductive Medicine, KK Women’s and Children’s Hospital, 100 Bukit Timah Road, Singapore, 229899 Singapore
- Obstetrics and Gynaecology Academic Clinical Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857 Singapore
| | - Yie Hou Lee
- Translational ‘Omics and Biomarkers Group, KK Research Centre, KK Women’s and Children’s Hospital, 100 Bukit Timah Road, Singapore, 229899 Singapore
- Obstetrics and Gynaecology Academic Clinical Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857 Singapore
- Singapore-MIT Alliance for Research and Technoology, 1 CREATE Way, Singapore, 138602 Singapore
| |
Collapse
|
50
|
Ullah A, Wang MJ, Wang YX, Shen B. CXC chemokines influence immune surveillance in immunological disorders: Polycystic ovary syndrome and endometriosis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166704. [PMID: 37001703 DOI: 10.1016/j.bbadis.2023.166704] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/13/2023] [Accepted: 03/22/2023] [Indexed: 04/05/2023]
Abstract
Reproductive health is a worldwide challenge, but it is of particular significance to women during their reproductive age. Several female reproductive problems, including polycystic ovary syndrome (PCOS) and endometriosis, affect about 10 % of women and have a negative impact on their health, fertility, and quality of life. Small, chemotactic, and secreted cytokines are CXC chemokines. Both PCOS and endometriosis demonstrate dysregulation of CXC chemokines, which are critical to the development and progression of both diseases. Recent research has shown that both in humans and animals, CXC chemokines tend to cause inflammation. It has also been found that CXC chemokines are necessary for promoting angiogenesis and inflammatory responses. CXC chemokine overexpression is frequently associated with poor survival and prognosis. CXC chemokine levels in PCOS and endometriosis patients impact their circumstances significantly. Hence, CXC chemokines have significant potential as diagnostic and prognostic biomarkers and therapeutic targets. The molecular mechanisms through which CXC chemokines promote inflammation and the development of PCOS and endometriosis are currently unknown. This article will discuss the functions of CXC chemokines in the promotion, development, and therapy of PCOS and endometriosis, as well as future research directions. The current state and future prospects of CXC chemokine -based therapeutic strategies in the management of PCOS and endometriosis are also highlighted.
Collapse
|