1
|
Zhang F, Zhu M, Chen Y, Wang G, Yang H, Lu X, Li Y, Chang HM, Wu Y, Ma Y, Yuan S, Zhu W, Dong X, Zhao Y, Yu Y, Wang J, Mu L. Harnessing omics data for drug discovery and development in ovarian aging. Hum Reprod Update 2025; 31:240-268. [PMID: 39977580 DOI: 10.1093/humupd/dmaf002] [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: 09/29/2024] [Revised: 12/02/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Ovarian aging occurs earlier than the aging of many other organs and has a lasting impact on women's overall health and well-being. However, effective interventions to slow ovarian aging remain limited, primarily due to an incomplete understanding of the underlying molecular mechanisms and drug targets. Recent advances in omics data resources, combined with innovative computational tools, are offering deeper insight into the molecular complexities of ovarian aging, paving the way for new opportunities in drug discovery and development. OBJECTIVE AND RATIONALE This review aims to synthesize the expanding multi-omics data, spanning genome, transcriptome, proteome, metabolome, and microbiome, related to ovarian aging, from both tissue-level and single-cell perspectives. We will specially explore how the analysis of these emerging omics datasets can be leveraged to identify novel drug targets and guide therapeutic strategies for slowing and reversing ovarian aging. SEARCH METHODS We conducted a comprehensive literature search in the PubMed database using a range of relevant keywords: ovarian aging, age at natural menopause, premature ovarian insufficiency (POI), diminished ovarian reserve (DOR), genomics, transcriptomics, epigenomics, DNA methylation, RNA modification, histone modification, proteomics, metabolomics, lipidomics, microbiome, single-cell, genome-wide association studies (GWAS), whole-exome sequencing, phenome-wide association studies (PheWAS), Mendelian randomization (MR), epigenetic target, drug target, machine learning, artificial intelligence (AI), deep learning, and multi-omics. The search was restricted to English-language articles published up to September 2024. OUTCOMES Multi-omics studies have uncovered key mechanisms driving ovarian aging, including DNA damage and repair deficiencies, inflammatory and immune responses, mitochondrial dysfunction, and cell death. By integrating multi-omics data, researchers can identify critical regulatory factors and mechanisms across various biological levels, leading to the discovery of potential drug targets. Notable examples include genetic targets such as BRCA2 and TERT, epigenetic targets like Tet and FTO, metabolic targets such as sirtuins and CD38+, protein targets like BIN2 and PDGF-BB, and transcription factors such as FOXP1. WIDER IMPLICATIONS The advent of cutting-edge omics technologies, especially single-cell technologies and spatial transcriptomics, has provided valuable insights for guiding treatment decisions and has become a powerful tool in drug discovery aimed at mitigating or reversing ovarian aging. As technology advances, the integration of single-cell multi-omics data with AI models holds the potential to more accurately predict candidate drug targets. This convergence offers promising new avenues for personalized medicine and precision therapies, paving the way for tailored interventions in ovarian aging. REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Fengyu Zhang
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Ming Zhu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yi Chen
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Guiquan Wang
- Xiamen Key Laboratory of Reproduction and Genetics, Department of Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Haiyan Yang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinmei Lu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Li
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Yang Wu
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Yunlong Ma
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shuai Yuan
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Wencheng Zhu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xi Dong
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yang Yu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jia Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liangshan Mu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Smith P, Reader KL, French MC, Hurst PR, Juengel JL. Expression of platelet derived growth factor (PDGF) ligands and receptors in the ovine ovary and effects of PDGFBB on granulosa cell function. Reprod Fertil Dev 2025; 37:RD24174. [PMID: 40294218 DOI: 10.1071/rd24174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Context While platelet derived growth factors (PDGF) are expressed in the ovary of some species, relatively little is known regarding the potential role that PDGF play in regulating ovarian follicular development. Aims To determine the effects of PDGF on granulosa cell function from ovine antral follicles and characterise the expression of PDGF ligands and receptors in developing follicles. Methods The effects of the PDGF ligand PDGFBB on thymidine incorporation and progesterone production of granulosa cells were determined with established bioassays. Expression patterns of PDGF ligands, PDGFA , B , C , D , and receptors, PDGFRA and B , were determined using in situ hybridisation. The more sensitive technique of RT-PCR was used to confirm expression of PDGFRA and B in granulosa cells of antral follicles. Key results Thymidine incorporation was increased, and progesterone production decreased, from granulosa cells in response to PDGFBB. Oocytes, granulosa cells, thecal layers and corpora lutea expressed at least one PDGF family member. Granulosa cells had faint expression of PDGF receptors, with thecal and luteal tissue also expressing PDGF receptors. Expression of both ligands and receptors was observed in stroma around the follicle. Conclusions In sheep, ovarian follicles express both PDGF ligands and receptors, and PDGFBB regulated both thymidine incorporation and progesterone production. Implications In sheep, PDGF was identified as an additional locally produced growth factor that regulates follicular function, stimulating granulosa cell proliferation and inhibiting progesterone production. Future work to better understand the role of the different ligands and receptors at different stages of ovarian follicular development seems warranted.
Collapse
Affiliation(s)
- Peter Smith
- Agricultural Systems and Reproduction, AgResearch Ltd, Invermay Agricultural Centre, Mosgiel 9092, New Zealand
| | - Karen L Reader
- Department of Pathology, University of Otago, Dunedin 9016, New Zealand
| | - Michelle C French
- Animal Genomics, AgResearch Ltd, Invermay Agricultural Centre, Mosgiel 9092, New Zealand
| | - Peter R Hurst
- Department of Anatomy, School of Medical Sciences, University of Otago, Dunedin 9016, New Zealand
| | - Jennifer L Juengel
- Agricultural Systems and Reproduction, AgResearch Ltd, Invermay Agricultural Centre, Mosgiel 9092, New Zealand
| |
Collapse
|
3
|
Adamowski M, Sharma Y, Molcan T, Wołodko K, Kelsey G, Galvão AM. Leptin signalling regulates transcriptional differences in granulosa cells from genetically obese mice but not the activation of NLRP3 inflammasome. Sci Rep 2024; 14:8070. [PMID: 38580672 PMCID: PMC10997671 DOI: 10.1038/s41598-024-58181-w] [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: 09/04/2023] [Accepted: 03/26/2024] [Indexed: 04/07/2024] Open
Abstract
Obesity is associated with increased ovarian inflammation and the establishment of leptin resistance. We presently investigated the role of impaired leptin signalling on transcriptional regulation in granulosa cells (GCs) collected from genetically obese mice. Furthermore, we characterised the association between ovarian leptin signalling, the activation of the NOD-like receptor protein 3 (NLRP3) inflammasome and macrophage infiltration in obese mice. After phenotype characterisation, ovaries were collected from distinct group of animals for protein and mRNA expression analysis: (i) mice subjected to a diet-induced obesity (DIO) protocol, where one group was fed a high-fat diet (HFD) and another a standard chow diet (CD) for durations of 4 or 16 weeks; (ii) mice genetically deficient in the long isoform of the leptin receptor (ObRb; db/db); (iii) mice genetically deficient in leptin (ob/ob); and (iv) mice rendered pharmacologically hyperleptinemic (LEPT). Next, GCs from antral follicles isolated from db/db and ob/ob mice were subjected to transcriptome analysis. Transcriptional analysis revealed opposing profiles in genes associated with steroidogenesis and prostaglandin action between the genetic models, despite the similarities in body weight. Furthermore, we observed no changes in the mRNA and protein levels of NLRP3 inflammasome components in the ovaries of db/db mice or in markers of M1 and M2 macrophage infiltration. This contrasted with the downregulation of NLRP3 inflammasome components and M1 markers in ob/ob and 16-wk HFD-fed mice. We concluded that leptin signalling regulates NLRP3 inflammasome activation and the expression of M1 markers in the ovaries of obese mice in an ObRb-dependent and ObRb-independent manner. Furthermore, we found no changes in the expression of leptin signalling and NLRP3 inflammasome genes in GCs from db/db and ob/ob mice, which was associated with no effects on macrophage infiltration genes, despite the dysregulation of genes associated with steroidogenesis in homozygous obese db/db. Our results suggest that: (i) the crosstalk between leptin signalling, NLRP3 inflammasome and macrophage infiltration takes place in ovarian components other than the GC compartment; and (ii) transcriptional changes in GCs from homozygous obese ob/ob mice suggest structural rearrangement and organisation, whereas in db/db mice the impairment in steroidogenesis and secretory activity.
Collapse
Affiliation(s)
- Marek Adamowski
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Yashaswi Sharma
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Tomasz Molcan
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Karolina Wołodko
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Gavin Kelsey
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK
| | - António M Galvão
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland.
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK.
- Department of Comparative Biomedical Sciences, Royal Veterinary College, 4 Royal College Street, London, NW1 0TU, UK.
| |
Collapse
|
4
|
Lin Y, Wang G, Li Y, Yang H, Zhao Y, Liu J, Mu L. Circulating Inflammatory Cytokines and Female Reproductive Diseases: A Mendelian Randomization Analysis. J Clin Endocrinol Metab 2023; 108:3154-3164. [PMID: 37350485 DOI: 10.1210/clinem/dgad376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/19/2023] [Accepted: 06/22/2023] [Indexed: 06/24/2023]
Abstract
CONTEXT Extensive studies have provided considerable evidence suggesting the role of inflammation in the development of female reproductive diseases. However, causality has not been established. OBJECTIVE To explore whether genetically determined circulating levels of cytokines are causally associated with female reproductive diseases and discover potential novel drug targets for these diseases. METHODS Instrumental variables (IVs) for 47 circulating cytokines were obtained from a genome-wide association study (GWAS) meta-analysis of 31 112 European individuals. Protein quantitative trait loci and expression quantitative trait loci close to genes served as our IVs. Summary data of 9 female reproductive diseases were mainly derived from GWAS meta-analysis of the UK biobank and FinnGen. We elevated the association using the Wald ratio or inverse variance-weighted Mendelian randomization (MR) with subsequent assessments for MR assumptions in several sensitivity and colocalization analyses. We consider a false discovery rate <0.05 as statistical significance in MR analyses. Replication studies were conducted for further validation, and phenome-wide association studies were designed to explore potential side effects. RESULTS Our results indicated that high levels of macrophage colony-stimulating factor (MCSF), growth-regulated oncogene-alpha (GROα), and soluble intercellular adhesion molecule-1 were associated with increased risks of endometriosis, female infertility, and pre-eclampsia, respectively. High platelet-derived growth factor-BB (PDGF-BB) levels that reduced the risk of ovarian aging were also supported. Replication analysis supported the relationship between GROα and female infertility, and between MCSF and endometriosis. CONCLUSION We identified 4 correlated pairs that implied potential protein drug targets. Notably, we preferred highlighting the value of PDGF-BB as a drug target for ovarian aging, and MCSF as a drug target for endometriosis.
Collapse
Affiliation(s)
- Yiting Lin
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guiquan Wang
- Center for Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Yan Li
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haiyan Yang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yue Zhao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jun Liu
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Liangshan Mu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Insights into Transcriptomic Differences in Ovaries between Lambs and Adult Sheep after Superovulation Treatment. Animals (Basel) 2023; 13:ani13040665. [PMID: 36830452 PMCID: PMC9951745 DOI: 10.3390/ani13040665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Superovulation technology shows a great potential for shortening breeding time. Using the juvenile superovulation technology, juvenile animals can generate more follicles than adult animals. By sequencing using high-throughput methods, we studied and described differentially expressed (DE) long non-coding RNA (lncRNAs) and messenger RNAs (mRNAs) in the ovaries of young and adult sheep. Herein, 242 DE lncRNAs and 3150 DE mRNAs were screened. Through GO and KEGG analyses, we obtained genes related to ovarian/follicle development and ovulation in DE mRNAs, including OaFSHR, OaLHCGR, OaLDLR, OaZP3, OaSCARB1, and OaPDGFRA; through lncRNA-mRNA correlation analysis, we found that genes associated with ovarian/follicle development or ovulation include: XR_003585520.1, MSTRG.15652.1, XR_003588840.1, and their paired genes PDGFC, LRP5, and LRP1. We observed a synergistic effect between PDGFR and LRP1. PDGFR may play a leading role compared with LRP1. The induced LHCGR in lambs is higher than in adult sheep, showing more sensitivity to LH. The release of the oocytes was stimulated. Among the three lncRNAs, we found that XR_003588840.1 was significantly different and might perform a regulatory role in ovarian/follicle growth or ovulation.
Collapse
|
6
|
Pascuali N, Scotti L, Oubiña G, de Zúñiga I, Gomez Peña M, Pomilio C, Saravia F, Tesone M, Abramovich D, Parborell F. Platelet-derived growth factor B restores vascular barrier integrity and diminishes permeability in ovarian hyperstimulation syndrome. Mol Hum Reprod 2021; 26:585-600. [PMID: 32467982 DOI: 10.1093/molehr/gaaa038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 04/08/2020] [Indexed: 01/29/2023] Open
Abstract
Although advances in the prediction and management of ovarian hyperstimulation syndrome (OHSS) have been introduced, complete prevention is not yet possible. Previously, we and other authors have shown that vascular endothelial growth factor, angiopoietins (ANGPTs) and sphingosine-1-phosphate are involved in OHSS etiology. In addition, we have demonstrated that ovarian protein levels of platelet-derived growth factor (PDGF) ligands -B and -D decrease in an OHSS rat model, whilst PDGFR-β and ANGPT2 remain unchanged. In the present work, we investigated the role of PDGF-B in OHSS by evaluating ligand protein levels in follicular fluid (FF) from women at risk of developing OHSS and by using an immature rat model of OHSS. We demonstrated that PDGF-B and PDGF-D are lower in FF from women at risk of developing OHSS compared to control patients (P < 0.05). In the OHSS rat model, PDGF-B (0.5 µg/ovary) administration decreased ovarian weight (P < 0.05), reduced serum progesterone (P < 0.05) and lowered the percentage of cysts (P < 0.05), compared to untreated OHSS rats, but had no effect on the proportion of follicles or corpora lutea (CL). PDGF-B treatment also restored the expression of steroidogenic acute regulatory protein (P < 0.05) and P450 cholesterol side-chain cleavage enzyme (P < 0.01) to control levels. In addition, PDGF-B increased the peri-endothelial cell area in CL and cystic structures, and reduced vascular permeability compared to untreated OHSS ovaries. Lastly, PDGF-B increased the levels of junction proteins claudin-5 (P < 0.05), occludin (P < 0.05) and β-catenin (P < 0.05), while boosting the extracellular deposition of collagen IV surrounding the ovarian vasculature (PP < 0.01), compared to OHSS alone. In conclusion, our findings indicate that PDGF-B could be another crucial mediator in the onset and development of OHSS, which may lead to the development of novel prediction markers and therapeutic strategies.
Collapse
Affiliation(s)
- Natalia Pascuali
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Leopoldina Scotti
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Gonzalo Oubiña
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | | | | | - Carlos Pomilio
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Flavia Saravia
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marta Tesone
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| |
Collapse
|
7
|
Abbassi L, El-Hayek S, Carvalho KF, Wang W, Yang Q, Granados-Aparici S, Mondadori R, Bordignon V, Clarke HJ. Epidermal growth factor receptor signaling uncouples germ cells from the somatic follicular compartment at ovulation. Nat Commun 2021; 12:1438. [PMID: 33664246 PMCID: PMC7933413 DOI: 10.1038/s41467-021-21644-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 02/07/2021] [Indexed: 01/31/2023] Open
Abstract
Germ cells are physically coupled to somatic support cells of the gonad during differentiation, but this coupling must be disrupted when they are mature, freeing them to participate in fertilization. In mammalian females, coupling occurs via specialized filopodia that project from the ovarian follicular granulosa cells to the oocyte. Here, we show that signaling through the epidermal growth factor receptor (EGFR) in the granulosa, which becomes activated at ovulation, uncouples the germ and somatic cells by triggering a massive and temporally synchronized retraction of the filopodia. Although EGFR signaling triggers meiotic maturation of the oocyte, filopodial retraction is independent of the germ cell state, being regulated solely within the somatic compartment, where it requires ERK-dependent calpain-mediated loss of filopodia-oocyte adhesion followed by Arp2/3-mediated filopodial shortening. By uncovering the mechanism regulating germ-soma uncoupling at ovulation, our results open a path to improving oocyte quality in human and animal reproduction.
Collapse
Affiliation(s)
- Laleh Abbassi
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Stephany El-Hayek
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Biology, McGill University, Montreal, Canada
- Centre for Arab Genomic Studies, Dubai, United Arab Emirates
| | - Karen Freire Carvalho
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Wusu Wang
- Research Institute of the McGill University Health Centre, Montreal, Canada
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, PR China
| | - Qin Yang
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | | | - Rafael Mondadori
- Department of Animal Science, McGill University, Montreal, Canada
| | - Vilceu Bordignon
- Department of Animal Science, McGill University, Montreal, Canada
| | - Hugh J Clarke
- Research Institute of the McGill University Health Centre, Montreal, Canada.
- Division of Experimental Medicine, McGill University, Montreal, Canada.
- Department of Biology, McGill University, Montreal, Canada.
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada.
| |
Collapse
|
8
|
Liu A, Liu M, Li Y, Chen X, Zhang L, Tian S. Differential expression and prediction of function of lncRNAs in the ovaries of low and high fecundity Hanper sheep. Reprod Domest Anim 2021; 56:604-620. [PMID: 33475207 DOI: 10.1111/rda.13898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/18/2021] [Indexed: 12/13/2022]
Abstract
Litter size is an important trait that determines the production efficiency of sheep bred for meat. Its detailed investigation can reveal the molecular mechanisms that control the fecundity of sheep and possibly accelerate the breeding process of new varieties of sheep that have high prolificacy. Long non-coding RNAs (lncRNAs) have proven to be an important factor in the regulation of follicular development. However, the mechanisms by which lncRNAs regulate litter size in sheep remain unclear. In the present study, ovarian tissues from the follicular (F) or luteal phase (L) of Hanper sheep that were either monotocous (M) or polytocous (P; FM, FP, LM and LP groups) were collected and sequenced to identify differentially expressed lncRNAs and predict their function. The results indicate that the number of up- and down-regulated lncRNAs in the follicular phase (FM vs. FP) was 95 and 111 and 109 and 49, respectively, in the luteal phase (LM vs. LP). The functional enrichment of the different lncRNAs coexpressed with mRNA was analysed. The results demonstrated that the KISS1-GnRH-LH/FSH-E2 and EGF-EGFR-RAS-PI3K signalling pathways promoted the initiation of the primordial period, follicular development and ovulation in the follicular phase (FM vs. FP). During the luteal phase (LM vs. LP), the production and development of the corpus luteum in ewes was influenced by the KITLG-KIT/FGF-FGFR/HGF-MET-RAS-ERK signalling pathway. STEM clustering functional enrichment analysis of the differentially expressed lncRNAs indicated that profile11 was principally enriched in the Cytokine-Jak-STAT, PDGF-PDGFR-PI3K and KITLG-KIT-RAS-ERK signalling pathways. By analysis of the differential expression of the lncRNAs and their expression in each group, lncRNAs Xist (loc101112291) and Gtl2 (loc101123329) were found to be highly expressed, suggesting that regulation of follicular development was mediated through methylation processes.
Collapse
Affiliation(s)
- Aiju Liu
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Menghe Liu
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Yuexin Li
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Xiaoyong Chen
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Limeng Zhang
- Laboratory of Molecular Biology, Zhengzhou Normal University, Zhengzhou, China
| | - Shujun Tian
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China.,The Research Center of Cattle and Sheep, Embryonic Technique of Hebei Province, Baoding, China
| |
Collapse
|
9
|
Salem W, Ho JR, Woo I, Ingles SA, Chung K, Paulson RJ, McGinnis LK. Long-term imatinib diminishes ovarian reserve and impacts embryo quality. J Assist Reprod Genet 2020; 37:1459-1466. [PMID: 32372302 PMCID: PMC7311628 DOI: 10.1007/s10815-020-01778-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/08/2020] [Indexed: 10/24/2022] Open
Abstract
PURPOSE Tyrosine kinase inhibitors (TKIs) such as imatinib are commonly used chemotherapeutics, but the effects of long-term treatments on reproductive outlook for cancer survivors are unknown. The purpose of this study was to examine the effects of long-term imatinib treatments on follicle development and embryo quality. Since prospective studies are not possible in healthy humans, we have incorporated a commonly used mouse model. METHODS Adult female mice were treated with daily IP injections of imatinib for 4-6 weeks. Liquid chromatography-mass spectrometry was used to measure imatinib in serum and ovarian tissues. At the end of treatments, females were superovulated and mated to yield fertilized embryos. Oocytes and embryos were collected from oviducts, assessed for development by microscopy, and fertilized embryos were cultured in vitro. Blastocysts were fixed and stained for differential cell counts. RESULTS Long-term imatinib treatments caused a shift in follicle development, with imatinib-treated females having fewer primordial follicles, but an increase in primary and secondary follicles (P < 0.05). There was no effect on ovulation or fertilization rates. However, blastocysts from imatinib-treated females had fewer total cells (P < 0.05) and a significant shift from inner cell mass to increased trophectoderm cells. CONCLUSION This pilot study indicates that long-term TKI treatments may have significant impact on ovarian reserve and embryo developmental capacity. More studies are needed in other model systems to determine the long-term impact of TKIs in patients. Knowing the potential effects of chemotherapeutics on reproductive outlook is critical for quality of life and more research is needed.
Collapse
Affiliation(s)
- Wael Salem
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Southern California, Los Angeles, CA, USA
| | - Jacqueline R Ho
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Southern California, Los Angeles, CA, USA
| | - Irene Woo
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Southern California, Los Angeles, CA, USA
| | - Sue A Ingles
- Department of Preventative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Karine Chung
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Southern California, Los Angeles, CA, USA
| | - Richard J Paulson
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Southern California, Los Angeles, CA, USA
| | - Lynda K McGinnis
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
10
|
Bouet PE, Boueilh T, de la Barca JMC, Boucret L, Blanchard S, Ferré-L'Hotellier V, Jeannin P, Descamps P, Procaccio V, Reynier P, May-Panloup P. The cytokine profile of follicular fluid changes during ovarian ageing. J Gynecol Obstet Hum Reprod 2020; 49:101704. [PMID: 32028036 DOI: 10.1016/j.jogoh.2020.101704] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 12/14/2019] [Accepted: 01/21/2020] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Ovarian ageing is one of the commonest causes of infertility in patients consulting for assisted reproductive technology. The composition of the follicular fluid (FF), which reflects the exchanges between the oocyte and its microenvironment, has been extensively investigated to determine the metabolic pathways involved in various ovarian disorders. Considering the importance of cytokines in folliculogenesis, we focused on the cytokine profile of the FF during ovarian ageing. MATERIAL AND METHODS Our cross-sectional study assesses the levels of 27 cytokines and growth factors in the FF of two groups of women undergoing in vitro fertilization. One group included 28 patients with ovarian ageing clinically characterized by a diminished ovarian reserve (DOR), and the other group included 29 patients with a normal ovarian reserve (NOR), serving as controls. RESULTS With univariate analysis, the cytokine profile was found to differ significantly between the two groups. After adjustment of the p-values, platelet-derived growth factor-BB (PDGF-BB) was the only cytokine with a significantly lower concentration in the DOR group (7.34 ± 16.11 pg/mL) than in the NOR group (24.39 ± 41.38 pg/mL) (p = 0.005), independently of chronological age. CONCLUSION Thus, PDGF-BB would seem to be implicated in the physiopathology of DOR, potentially in relation to its role in folliculogenesis or in the protection against oxidative stress.
Collapse
Affiliation(s)
- Pierre-Emmanuel Bouet
- Department of Reproductive Medicine, Angers University Hospital, 49000, Angers, France; MITOVASC Institute, CNRS 6015, INSERM U1083, Angers University, 49000, Angers, France.
| | - Thomas Boueilh
- Reproductive Biology Unit, Angers University Hospital, 49000, Angers, France
| | - Juan Manuel Chao de la Barca
- Department of Biochemistry and Genetics, Angers University Hospital, 49000, Angers, France; MITOVASC Institute, CNRS 6015, INSERM U1083, Angers University, 49000, Angers, France
| | - Lisa Boucret
- Reproductive Biology Unit, Angers University Hospital, 49000, Angers, France; MITOVASC Institute, CNRS 6015, INSERM U1083, Angers University, 49000, Angers, France
| | - Simon Blanchard
- Immunology and Allergology Laboratory, Center of Immunology and Cancer Research Nantes-Anges, INSERM U1232, Angers University, 49000, Angers, France
| | | | - Pascale Jeannin
- Immunology and Allergology Laboratory, Center of Immunology and Cancer Research Nantes-Anges, INSERM U1232, Angers University, 49000, Angers, France
| | - Philippe Descamps
- Department of Reproductive Medicine, Angers University Hospital, 49000, Angers, France
| | - Vincent Procaccio
- Department of Biochemistry and Genetics, Angers University Hospital, 49000, Angers, France; MITOVASC Institute, CNRS 6015, INSERM U1083, Angers University, 49000, Angers, France
| | - Pascal Reynier
- Department of Biochemistry and Genetics, Angers University Hospital, 49000, Angers, France; MITOVASC Institute, CNRS 6015, INSERM U1083, Angers University, 49000, Angers, France
| | - Pascale May-Panloup
- Reproductive Biology Unit, Angers University Hospital, 49000, Angers, France; MITOVASC Institute, CNRS 6015, INSERM U1083, Angers University, 49000, Angers, France
| |
Collapse
|
11
|
Di Pietro M, Velazquez C, Matzkin ME, Frungieri MB, Peña MG, de Zúñiga I, Pascuali N, Irusta G, Bianchi MS, Parborell F, Abramovich D. Metformin has a direct effect on ovarian cells that is dependent on organic cation transporters. Mol Cell Endocrinol 2020; 499:110591. [PMID: 31546019 DOI: 10.1016/j.mce.2019.110591] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022]
Abstract
Metformin (MET) is the most widely prescribed hypoglycemic drug in type 2 diabetes and Polycystic Ovary Syndrome. Besides its effects on glucose metabolism, MET exerts beneficial effects on these patients' fertility. However, the exact mechanisms of action of MET on female fertility are still unclear. In this work, we analyzed a possible direct effect of MET on ovarian cells. We found expression of the organic cation transporters OCT1, OCT2 and OCT3, responsible for MET uptake into the cells, in rat granulosa cells and human cumulus cells. Furthermore, MET increased pAMPK and decreased VEGF levels both in vivo and in rat granulosa cells in culture. These last effects were reversed when OCTs were inhibited. Our results suggest that MET acts directly on ovarian cells regulating cell metabolism and VEGF expression. Our findings are relevant to optimize PCOS fertility treatment and to explore ovarian MET actions in other female pathologies.
Collapse
Affiliation(s)
- Mariana Di Pietro
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Candela Velazquez
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - María Eugenia Matzkin
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Mónica Beatriz Frungieri
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina; Cátedra de Química, Ciclo Básico Común, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Mariana Gómez Peña
- Centro Médico Pregna Medicina Reproductiva. Ciudad Autónoma de Buenos Aires, Argentina
| | - Ignacio de Zúñiga
- Centro Médico Pregna Medicina Reproductiva. Ciudad Autónoma de Buenos Aires, Argentina
| | - Natalia Pascuali
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Griselda Irusta
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - María Silvia Bianchi
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
12
|
Hernández-Coronado CG, Guzmán A, Castillo-Juárez H, Zamora-Gutiérrez D, Rosales-Torres AM. Sphingosine-1-phosphate (S1P) in ovarian physiology and disease. ANNALES D'ENDOCRINOLOGIE 2019; 80:263-272. [DOI: 10.1016/j.ando.2019.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/06/2019] [Accepted: 06/20/2019] [Indexed: 12/15/2022]
|
13
|
A report on three live births in women with poor ovarian response following intra-ovarian injection of platelet-rich plasma (PRP). Mol Biol Rep 2019; 46:1611-1616. [DOI: 10.1007/s11033-019-04609-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 01/16/2019] [Indexed: 11/27/2022]
|
14
|
Di Pietro M, Pascuali N, Parborell F, Abramovich D. Ovarian angiogenesis in polycystic ovary syndrome. Reproduction 2018; 155:R199-R209. [PMID: 29386378 DOI: 10.1530/rep-17-0597] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/30/2018] [Indexed: 12/16/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most prevalent endocrine pathology among women in reproductive age. Its main symptoms are oligo or amenorrhea, hyperandrogenism and the presence of ovarian cysts. It is also associated with infertility, obesity and insulin resistance. Mainly due to its heterogeneity, PCOS treatments are directed to manage its symptoms and to prevent associated diseases. The correct formation and regression of blood vessels during each ovarian cycle is indispensable for proper follicular development, ovulation and corpus luteum formation. The importance of these processes opened a new and promising field: ovarian angiogenesis. Vascular alterations characterize numerous pathologies, either with increased, decreased or abnormal angiogenesis. In the last years, several anomalies of ovarian angiogenesis have been described in women with PCOS. Therefore, it has been suggested that these alterations may be associated with the decreased - or lack of - ovulation rates and for the formation of cysts in the PCOS ovaries. Restoration of a proper vessel formation in the ovaries may lead to improved follicular development and ovulation in these patients. In the present review, we attempt to summarize the alterations in ovarian angiogenesis that have been described in women with PCOS. We also discuss the therapeutic approaches aimed to correct these alterations and their beneficial effects on the treatment of infertility in PCOS.
Collapse
Affiliation(s)
- Mariana Di Pietro
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| | - Natalia Pascuali
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| |
Collapse
|
15
|
Effects of vitrification and transplantation on follicular development and expression of EphrinB1 and PDGFA in mouse ovaries. Cryobiology 2017; 80:101-113. [PMID: 29154909 DOI: 10.1016/j.cryobiol.2017.11.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 11/08/2017] [Accepted: 11/14/2017] [Indexed: 12/31/2022]
Abstract
The aim of this study was to assess the follicular development and the patterns of EphrinB1 and PDGFA immunostaining in vitrified mouse ovarian tissue (OT) with and without transplantation. Histological evaluation was performed on fresh and vitrified OTs, whether transplanted or not. RT-PCR was performed on fresh and vitrified ovarian samples (OSs) and vitrified OS graft. Vitrification alone did not significantly reduce the normal primordial, primary, and secondary follicles except antral ones (p > 0.05). However, transplantation decreased all the follicle types. The EphrinB1 immunoexpression showed high intensity in all follicular types in vitrified OT and the significant increased was detected in secondary and antral follicles (p < 0.05). PDGFA protein immunoexpression of primordial and primary follicles was decreased in vitrified OT (p < 0.05). However, the lowest immunoexpression of EphrinB1 and PDGFA was detected after transplantation (p < 0.05). The levels of ephrinb1 and pdgfa mRNA expressions in vitrified OS and vitrified OS grafts were found as comparable to the fresh OS. In conclusion, vitrification has no detrimental effect on the follicles at the different developmental stages, majority of ovarian follicular loss takes place after transplantation rather than vitrification. Overall, vitrification and grafting do not change the ephrinb1 and pdgfa gene expressions. In addition, EphrinB1 and PDGFA are expressed during different stages of folliculogenesis in a different manner in fresh, vitrified, or grafted OTs. Vitrification and/or grafting appear to affect the follicular expression of EphrinB1 and PDGFA. These findings suggest that these proteins could have several functions related to the development of follicles and angiogenesis after transplantation.
Collapse
|
16
|
Barreto Amaral Teixeira D, Alves Fernandes Júnior G, Beraldo dos Santos Silva D, Bermal Costa R, Takada L, Gustavo Mansan Gordo D, Bresolin T, Carvalheiro R, Baldi F, Galvão de Albuquerque L. Genomic analysis of stayability in Nellore cattle. PLoS One 2017; 12:e0179076. [PMID: 28591167 PMCID: PMC5462402 DOI: 10.1371/journal.pone.0179076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/23/2017] [Indexed: 12/15/2022] Open
Abstract
Stayability, which can be defined as the probability of a cow calving at a certain age when given the opportunity, is an important reproductive trait in beef cattle because it is directly related to herd profitability. The objective of this study was to estimate genetic parameters and to identify possible genomic regions associated with the phenotypic expression of stayability in Nellore cows. The variance components were estimated by Bayesian inference using a threshold animal model that included the systematic effects of contemporary group and sexual precocity and the random effects of animal and residual. The SNP effects were estimated by the single-step genomic BLUP method using information of 2,838 animals (2,020 females and 930 sires) genotyped with the Illumina High-Density BeadChip Array (San Diego, CA, USA). The variance explained by windows formed by 200 consecutive SNPs was used to identify genomic regions of largest effect on the expression of stayability. The heritability was 0.11 ± 0.01 when A matrix (pedigree) was used and 0.14 ± 0.01 when H matrix (relationship matrix that combines pedigree information and SNP data) was used. A total of 147 candidate genes for stayability were identified on chromosomes 1, 2, 5, 6, 9 and 20 and on the X chromosome. New candidate regions for stayability were detected, most of them related to reproductive, immunological and central nervous system functions.
Collapse
Affiliation(s)
| | | | | | | | - Luciana Takada
- Faculdade de Ciências Agrárias e Veterinárias, UNESP, Jaboticabal, SP, Brazil
| | | | - Tiago Bresolin
- Faculdade de Ciências Agrárias e Veterinárias, UNESP, Jaboticabal, SP, Brazil
| | - Roberto Carvalheiro
- Faculdade de Ciências Agrárias e Veterinárias, UNESP, Jaboticabal, SP, Brazil
- CNPq Fellowship, Brasília, DF, Brazil
| | - Fernando Baldi
- Faculdade de Ciências Agrárias e Veterinárias, UNESP, Jaboticabal, SP, Brazil
- CNPq Fellowship, Brasília, DF, Brazil
| | - Lucia Galvão de Albuquerque
- Faculdade de Ciências Agrárias e Veterinárias, UNESP, Jaboticabal, SP, Brazil
- CNPq Fellowship, Brasília, DF, Brazil
- * E-mail:
| |
Collapse
|
17
|
|
18
|
Developmental effects of imatinib mesylate on follicle assembly and early activation of primordial follicle pool in postnatal rat ovary. Reprod Biol 2017; 17:25-33. [DOI: 10.1016/j.repbio.2016.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/25/2016] [Accepted: 11/28/2016] [Indexed: 12/26/2022]
|
19
|
Single-cell analysis of differences in transcriptomic profiles of oocytes and cumulus cells at GV, MI, MII stages from PCOS patients. Sci Rep 2016; 6:39638. [PMID: 28004769 PMCID: PMC5177934 DOI: 10.1038/srep39638] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 11/24/2016] [Indexed: 11/09/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common frequent endocrine disorder among women of reproductive age. Although assisted reproductive techniques (ARTs) are used to address subfertility in PCOS women, their effectiveness is not clear. Our aim was to compare transcriptomic profiles of oocytes and cumulus cells (CCs) between women with and without PCOS, and assess the effectiveness of ARTs in treating PCOS patients. We collected oocytes and CCs from 16 patients with and without PCOS patients to categorize them into 6 groups according to oocyte nuclear maturation. Transcriptional gene expression of oocyte and CCs was determined via single-cell RNA sequencing. The ratio of fertilization and cleavage was higher in PCOS patients than in non-PCOS patients undergoing ARTs, and there was no difference in the number of high-quality embryos between the groups. Differentially expressed genes including PPP2R1A, PDGFRA, EGFR, GJA1, PTGS2, TNFAIP6, TGF-β1, CAV1, INHBB et al. were investigated as potential causes of PCOS oocytes and CCs disorder at early stages, but their expression returned to the normal level at the metaphase II (MII) stage via ARTs. In conclusion, ARTs can improve the quality of cumulus-oocyte complex (COC) and increase the ratio of fertilization and cleavage in PCOS women.
Collapse
|
20
|
Di Pietro M, Scotti L, Irusta G, Tesone M, Parborell F, Abramovich D. Local administration of platelet-derived growth factor B (PDGFB) improves follicular development and ovarian angiogenesis in a rat model of Polycystic Ovary Syndrome. Mol Cell Endocrinol 2016; 433:47-55. [PMID: 27256152 DOI: 10.1016/j.mce.2016.05.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 05/04/2016] [Accepted: 05/27/2016] [Indexed: 11/18/2022]
Abstract
Alterations in ovarian angiogenesis are common features in Polycystic Ovary Syndrome (PCOS) patients; the most studied of these alterations is the increase in vascular endothelial growth factor (VEGF) production by ovarian cells. Platelet-derived growth factor B (PDGFB) and D (PDGFD) are decreased in follicular fluid of PCOS patients and in the ovaries of a rat model of PCOS. In the present study, we aimed to analyze the effects of local administration of PDGFB on ovarian angiogenesis, follicular development and ovulation in a DHEA-induced PCOS rat model. Ovarian PDGFB administration to PCOS rats partially restored follicular development, decreased the percentage of cysts, increased the percentage of corpora lutea, and decreased the production of anti-Müllerian hormone. In addition, PDGFB administration improved ovarian angiogenesis by reversing the increase in periendothelial cell area and restoring VEGF levels. Our results shed light into the mechanisms that lead to altered ovarian function in PCOS and provide new data for potential therapeutic strategies.
Collapse
Affiliation(s)
- Mariana Di Pietro
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, Buenos Aires, Argentina.
| | - Leopoldina Scotti
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, Buenos Aires, Argentina.
| | - Griselda Irusta
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, Buenos Aires, Argentina.
| | - Marta Tesone
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, Buenos Aires, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina.
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, Buenos Aires, Argentina.
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, Buenos Aires, Argentina.
| |
Collapse
|
21
|
Pascuali N, Scotti L, Abramovich D, Irusta G, Di Pietro M, Bas D, Tesone M, Parborell F. Inhibition of platelet-derived growth factor (PDGF) receptor affects follicular development and ovarian proliferation, apoptosis and angiogenesis in prepubertal eCG-treated rats. Mol Cell Endocrinol 2015; 412:148-58. [PMID: 25937181 DOI: 10.1016/j.mce.2015.04.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/24/2015] [Accepted: 04/24/2015] [Indexed: 11/21/2022]
Abstract
The platelet-derived growth factor (PDGF) system is crucial for blood vessel stability. In the present study, we evaluated whether PDGFs play a critical intraovarian survival role in gonadotropin-dependent folliculogenesis. We examined the effect of intrabursal administration of a selective platelet-derived growth factor receptor (PDGFR) inhibitor (AG1295) on follicular development, proliferation, apoptosis and blood vessel formation and stability in ovaries from rats treated with equine chorionic gonadotropin (eCG). The percentages of preantral follicles (PAFs) and early antral follicles (EAFs) were lower in AG1295-treated ovaries than in control ovaries (p < 0.01-0.05). The percentage of atretic follicles (AtrFs) increased in AG1295-treated ovaries compared to control (p < 0.05). The ovarian weight and estradiol concentrations were lower in AG1295-treated ovaries than in the control group (p < 0.01 and p < 0.05, respectively), whereas progesterone concentrations did not change. AG1295 decreased the proliferation index in EAFs (p < 0.05) and increased the percentage of nuclei positive for cleaved caspase-3 and apoptotic DNA fragmentation (p < 0.01-0.05). AG1295 increased the expression of Bax (p < 0.05) without changes in the expression of Bcl-2 protein. AG1295-treated ovaries increased the cleavage of caspase-8 (p < 0.05) and decreased AKT and BAD phosphorylation compared with control ovaries (p < 0.05). AG1295 caused a decrease not only in the endothelial cell area but also in the area of pericytes and vascular smooth muscle cells (VSMCs) in the ovary (p < 0.05). Our findings suggest that the local inhibition of PDGFs causes an increase in ovarian apoptosis through an imbalance in the ratio of antiapoptotic to proapoptotic proteins, thus leading a larger number of follicles to atresia. PDGFs could exert their mechanism of action through an autocrine/paracrine effect on granulosa and theca cells mediated by PDGFRs. In conclusion, these data clearly indicate that the PDGF system is necessary for follicular development induced by gonadotropins.
Collapse
Affiliation(s)
- Natalia Pascuali
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina
| | - Leopoldina Scotti
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina
| | - Griselda Irusta
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina
| | - Mariana Di Pietro
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina
| | - Diana Bas
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina
| | - Marta Tesone
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina.
| |
Collapse
|
22
|
miR-140-5p inhibits ovarian cancer growth partially by repression of PDGFRA. Biomed Pharmacother 2015; 75:117-22. [PMID: 26297547 DOI: 10.1016/j.biopha.2015.07.035] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 07/26/2015] [Indexed: 12/15/2022] Open
Abstract
Dysregulation of miRNAs is a common feature in human cancers, and miR-140-5p has been found to be down-regulated in cancer. However its role in ovarian cancer remains unclear. miR-140-5p was underexpressed in HCC tissues and cell lines compared with their normal controls. Additionally, PDGFRA was predicted the target gene of miR-140-5p. PDGFRA was inversely correlated with the expression of miR-140-5p in ovarian cancer cells. Importantly, we demonstrate that the over expression of miR-140-5p significantly inhibits ovarian cancer cell proliferation and induces apoptosis. Our results suggest the existence of a novel miR-140-5p-PDGFRA pathway and indicate that miR-140-5p acts as a tumor suppressor during ovarian carcinogenesis. These results may provide a promising alterative strategy for the therapeutic treatment of ovarian cancer.
Collapse
|
23
|
Petersen TS, Stahlhut M, Andersen CY. Phosphodiesterases in the rat ovary: effect of cAMP in primordial follicles. Reproduction 2015; 150:11-20. [PMID: 25861799 DOI: 10.1530/rep-14-0436] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 04/09/2015] [Indexed: 01/26/2023]
Abstract
Phosphodiesterases (PDEs) are important regulators of the intracellular cAMP concentration, which is a central second messenger that affects a multitude of intracellular functions. In the ovaries, cAMP exerts diverse functions, including regulation of ovulation and it has been suggested that augmented cAMP levels stimulate primordial follicle growth. The present study examined the gene expression, enzyme activity and immunolocalization of the different cAMP hydrolysing PDEs families in the rat ovary. Further, the effect of PDE4 inhibition on primordial follicle activation in cultured neonatal rat ovaries was also evaluated. We found varied expression of all eight families in the ovary with Pde7b and Pde8a having the highest expression each accounting for more than 20% of the total PDE mRNA. PDE4 accounted for 15-26% of the total PDE activity. Immunoreactive PDE11A was found in the oocytes and PDE2A in the corpora lutea. Incubating neonatal rat ovaries with PDE4 inhibitors did not increase primordial follicle activation or change the expression of the developing follicle markers Gdf9, Amh, Inha, the proliferation marker Mki67 or the primordial follicle marker Tmeff2. In addition, the cAMP analogue 8-bromo-cAMP did not increase AKT1 or FOXO3A phosphorylation associated with follicle activation or increase the expression of Kitlg known to be associated with follicle differentiation but did increase the Tmeff2, Mki67 and Inha expression in a dose-dependent manner. In conclusion, this study shows that both Pde7b and Pde8a are highly expressed in the rodent ovary and that PDE4 inhibition does not cause an increase in primordial follicle activation.
Collapse
Affiliation(s)
- Tonny Studsgaard Petersen
- Laboratory of Reproductive BiologyThe Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Copenhagen University, Department 5712, Blegdamsvej 9, Copenhagen 2100, DenmarkLEO PharmaBallerup 2750, Denmark Laboratory of Reproductive BiologyThe Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Copenhagen University, Department 5712, Blegdamsvej 9, Copenhagen 2100, DenmarkLEO PharmaBallerup 2750, Denmark
| | - Martin Stahlhut
- Laboratory of Reproductive BiologyThe Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Copenhagen University, Department 5712, Blegdamsvej 9, Copenhagen 2100, DenmarkLEO PharmaBallerup 2750, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive BiologyThe Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Copenhagen University, Department 5712, Blegdamsvej 9, Copenhagen 2100, DenmarkLEO PharmaBallerup 2750, Denmark
| |
Collapse
|
24
|
Brito IR, Sales AD, Rodrigues GQ, Lobo CH, Castro SV, Silva AWB, Moura AAA, Silva JRV, Rodrigues APR, Figueiredo JR. Differential gene expression and immunolocalization of platelet-derived growth factors and their receptors in caprine ovaries. Domest Anim Endocrinol 2015; 51:46-55. [PMID: 25498237 DOI: 10.1016/j.domaniend.2014.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 10/02/2014] [Accepted: 10/04/2014] [Indexed: 11/20/2022]
Abstract
This study evaluated the messenger RNA (mRNA) expression and immunolocalization of all members of the platelet-derived growth factor (PDGF) family in caprine ovaries by quantitative PCR and immunohistochemistry, respectively. Detectable levels of PDGF-A mRNA were not observed in primordial follicles. Higher levels of PDGF-B mRNA were observed in primary follicles than in primordial follicles (P < 0.05). PDGF-D mRNA levels were higher in secondary follicles than in the other preantral follicle categories (P < 0.05). PDGF-B mRNA expression was higher than PDGF-C mRNA expression in primary follicles (P < 0.05). In antral follicles, PDGF-A mRNA expression was higher in cumulus-oocyte complexes (COCs) from small antral follicles than in those from large antral follicles and their respective granulosa/theca (GT) cells (P < 0.05). Furthermore, in COCs from small and large antral follicles, PDGF-A mRNA expression was higher than that of the other PDGF isoforms (P < 0.05). The mRNA levels of PDGF-B and PDGF-D and PDGFR-α and PDGFR-β were higher in GT cells from large antral follicles than in GT cells from small antral follicles and in their respective COCs (P < 0.05). In COCs and GT cells from small antral follicles, the mRNA levels of PDGFR-α were higher than those of PDGFR-β (P < 0.05). All proteins were observed in the cytoplasm of oocytes from all follicular categories. In granulosa cells, all PDGFs and PDGFR-β were detected from starting at the secondary stage, and in theca cells, all proteins, except PDGF-C, were detected starting at the antral stage. In conclusion, PDGF and its receptors are differentially expressed in the oocytes and ovarian cells according to the stage of follicular development, suggesting their role in the regulation of folliculogenesis in goats.
Collapse
Affiliation(s)
- I R Brito
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil.
| | - A D Sales
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - G Q Rodrigues
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - C H Lobo
- Laboratory of Animal Physiology, Department of Animal Science, Federal University of Ceará, Fortaleza, CE, Brazil
| | - S V Castro
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - A W B Silva
- Biotechnology Nucleus of Sobral (NUBIS), Federal University of Ceará, Sobral, CE, Brazil
| | - A A A Moura
- Laboratory of Animal Physiology, Department of Animal Science, Federal University of Ceará, Fortaleza, CE, Brazil
| | - J R V Silva
- Biotechnology Nucleus of Sobral (NUBIS), Federal University of Ceará, Sobral, CE, Brazil
| | - A P R Rodrigues
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - J R Figueiredo
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
25
|
Peri LE, Koh BH, Ward GK, Bayguinov Y, Hwang SJ, Gould TW, Mullan CJ, Sanders KM, Ward SM. A novel class of interstitial cells in the mouse and monkey female reproductive tracts. Biol Reprod 2015; 92:102. [PMID: 25788664 DOI: 10.1095/biolreprod.114.124388] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 03/12/2015] [Indexed: 01/14/2023] Open
Abstract
Growing evidence suggests important roles for specialized platelet-derived growth factor receptor alpha-positive (PDGFRalpha(+)) cells in regulating the behaviors of visceral smooth muscle organs. Examination of the female reproductive tracts of mice and monkeys showed that PDGFRalpha(+) cells form extensive networks in ovary, oviduct, and uterus. PDGFRalpha(+) cells were located in discrete locations within these organs, and their distribution and density were similar in rodents and primates. PDGFRalpha(+) cells were distinct from smooth muscle cells and interstitial cells of Cajal (ICC). This was demonstrated with immunohistochemical techniques and by performing molecular expression studies on PDGFRalpha(+) cells from mice with enhanced green fluorescent protein driven off of the endogenous promoter for Pdgfralpha. Significant differences in gene expression were found in PDGFRalpha(+) cells from ovary, oviduct, and uterus. Differences in gene expression were also detected in cells from different tissue regions within the same organ (e.g., uterine myometrium vs. endometrium). PDGFRalpha(+) cells are unlikely to provide pacemaker activity because they lack significant expression of key pacemaker genes found in ICC (Kit and Ano1). Gja1 encoding connexin 43 was expressed at relatively high levels in PDGFRalpha(+) cells (except in the ovary), suggesting these cells can form gap junctions to one another and neighboring smooth muscle cells. PDGFRalpha(+) cells also expressed the early response transcription factor and proto-oncogene Fos, particularly in the ovary. These data demonstrate extensive distribution of PDGFRalpha(+) cells throughout the female reproductive tract. These cells are a heterogeneous population of cells that are likely to contribute to different aspects of physiological regulation in the various anatomical niches they occupy.
Collapse
Affiliation(s)
- Lauren E Peri
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Byoung H Koh
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Grace K Ward
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Yulia Bayguinov
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sung Jin Hwang
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Thomas W Gould
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Catrina J Mullan
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Kenton M Sanders
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sean M Ward
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
26
|
Scotti L, Parborell F, Irusta G, De Zuñiga I, Bisioli C, Pettorossi H, Tesone M, Abramovich D. Platelet-derived growth factor BB and DD and angiopoietin1 are altered in follicular fluid from polycystic ovary syndrome patients. Mol Reprod Dev 2014; 81:748-56. [PMID: 24889290 DOI: 10.1002/mrd.22343] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/15/2014] [Indexed: 01/17/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrinological pathology among women of reproductive age, and is characterized by abnormalities in ovarian angiogenesis, among other features. Consistent with this association, follicular fluid (FF) concentration and ovarian expression of vascular endothelial growth factor (VEGF) are increased in PCOS patients. In this study, we examined the protein levels of platelet-derived growth factor (PDGF) BB and DD (PDGFBB and PDGFDD), angiopoietin 1 and 2 (ANGPT1 and ANGPT2), and their soluble receptor sTIE2 in FF from PCOS and control patients undergoing assisted reproductive techniques. We also analyzed the effect of FF from PCOS and control patients on tight and adherens junction protein expression in an endothelial cell line. PDGFBB and PDGFDD were significantly lower whereas ANGPT1 concentration was significantly higher in FF from PCOS patients than from control patients. No changes were found in the concentration of ANGPT2 or sTIE2. Expression of claudin-5 was significantly increased in endothelial cells incubated for 24 hr in the presence of FF from PCOS versus from control patients, while vascular-endothelial cadherin, β-catenin, and zonula occludens 1 expression were unchanged. The changes observed in the levels of PDGF isoforms and ANGPT1 may prevent VEGF-induced vascular permeability in the PCOS ovary by regulating endothelial-cell-junction protein levels. Restoring the levels of angiogenic factors may provide new insights into PCOS treatment and the prevention of ovarian hyperstimulation syndrome in affected women.
Collapse
Affiliation(s)
- Leopoldina Scotti
- Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Emori C, Sugiura K. Role of oocyte-derived paracrine factors in follicular development. Anim Sci J 2014; 85:627-33. [PMID: 24717179 PMCID: PMC4271669 DOI: 10.1111/asj.12200] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 12/19/2013] [Indexed: 12/30/2022]
Abstract
Mammalian oocytes secrete transforming growth factor β (TGF-β) superfamily proteins, such as growth differentiation factor 9 (GDF9), bone morphogenetic protein 6 (BMP6) and BMP15, and fibroblast growth factors (FGFs). These oocyte-derived paracrine factors (ODPFs) play essential roles in regulating the differentiation and function of somatic granulosa cells as well as the development of ovarian follicles. In addition to the importance of individual ODPFs, emerging evidence suggests that the interaction of ODPF signals with other intra-follicular signals, such as estrogen, is critical for folliculogenesis. In this review, we will discuss the current understanding of the role of ODPFs in follicular development with an emphasis on their interaction with estrogen signaling in regulation of the differentiation and function of granulosa cells.
Collapse
Affiliation(s)
- Chihiro Emori
- Laboratory of Applied Genetics, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
28
|
Emori C, Wigglesworth K, Fujii W, Naito K, Eppig JJ, Sugiura K. Cooperative effects of 17β-estradiol and oocyte-derived paracrine factors on the transcriptome of mouse cumulus cells. Endocrinology 2013; 154:4859-72. [PMID: 24035995 PMCID: PMC3836066 DOI: 10.1210/en.2013-1536] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Oocyte-derived paracrine factors (ODPFs) and estrogens are both essential for the development and function of ovarian follicles in mammals. Cooperation of these two factors was assessed in vitro using intact cumulus-oocyte complexes, cumulus cells cultured after the removal of oocytes [oocytectomized (OOX) cumulus cells], and OOX cumulus cells cocultured with denuded oocytes, all in the presence or absence of 17β-estradiol (E2). Effects on the cumulus cell transcriptome were assessed by microarray analysis. There was no significant difference between the cumulus cell transcriptomes of either OOX cumulus cells cocultured with oocytes or intact cumulus-oocyte complexes. Therefore, oocyte-mediated regulation of the cumulus cell transcriptome is mediated primarily by ODPFs and not by gap junctional communication between oocytes and cumulus cells. Gene ontology analysis revealed that both ODPFs and E2 strongly affected the biological processes associated with cell proliferation in cumulus cells. E2 had limited effects on ODPF-regulated biological processes. However, in sharp contrast, ODPFs significantly affected biological processes regulated by E2 in cumulus cells. For example, only in the presence of ODPFs did E2 significantly promote the biological processes related to phosphorylation-mediated signal transduction in cumulus cells, such as the signaling pathways of epidermal growth factor, vascular endothelial growth factor, and platelet-derived growth factor. Therefore, ODPFs and E2 cooperate to regulate the cumulus cell transcriptome and, in general, oocytes modulate the effects of estrogens on cumulus cell function.
Collapse
Affiliation(s)
- Chihiro Emori
- PhD, Laboratory of Applied Genetics, Graduate School of Agriculture and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyoku, Tokyo, 113-8657, Japan.
| | | | | | | | | | | |
Collapse
|
29
|
Scotti L, Abramovich D, Pascuali N, de Zúñiga I, Oubiña A, Kopcow L, Lange S, Owen G, Tesone M, Parborell F. Involvement of the ANGPTs/Tie-2 system in ovarian hyperstimulation syndrome (OHSS). Mol Cell Endocrinol 2013; 365:223-30. [PMID: 23123737 DOI: 10.1016/j.mce.2012.10.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 10/09/2012] [Accepted: 10/22/2012] [Indexed: 11/23/2022]
Abstract
Ovarian hyperstimulation syndrome (OHSS) is a disorder associated with ovarian stimulation. OHSS features are ovarian enlargement with fluid shifting to the third space. Disturbances in the vasculature are considered the main changes that lead to OHSS. Our aim was to analyze the levels of angiopoietins 1 and 2 (ANGPT1 and 2) and their soluble and membrane receptors (s/mTie-2) in follicular fluid (FF) and in granulosa-lutein cells culture (GLCs) from women at risk of developing OHSS. We also evaluated the effect of ANGPT1 on endothelial cell migration. In ovaries from an OHSS rat model, we analyzed the protein concentration of ANGPTs, their mTie-2 receptor, and platelet-derived growth factor PDGF-B, -D and PDGFR-β. ANGPT1 levels were increased in both FF and GLCs from women at risk of OHSS. Incubation of these FF with an ANGPT1 neutralizing antibody decreased endothelial cell migration. In the ovaries of OHSS rat model, mTie-2 protein levels increased and PDGF-B and -D decreased. In summary, these results suggest that ANGPT1 could be another mediator in the development of OHSS.
Collapse
Affiliation(s)
- Leopoldina Scotti
- Instituto de Biología y Medicina Experimental - CONICET, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Imatinib mesylate at therapeutic doses has no impact on folliculogenesis or spermatogenesis in a leukaemic mouse model. Leuk Res 2012; 36:271-4. [DOI: 10.1016/j.leukres.2011.09.025] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 09/11/2011] [Accepted: 09/22/2011] [Indexed: 11/23/2022]
|
31
|
Brito IR, Lima IMT, Saraiva MVA, Silva CMG, Magalhães-Padilha DM, Araújo VR, Barreto Luz V, Barbalho Silva AW, Campello CC, Silva JRV, Figueiredo JR. Expression Levels of mRNA-Encoding PDGF Receptors in Goat Ovaries and the Influence of PDGF on the In Vitro Development of Caprine Pre-Antral Follicles. Reprod Domest Anim 2011; 47:695-703. [DOI: 10.1111/j.1439-0531.2011.01946.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
32
|
Kuo SW, Ke FC, Chang GD, Lee MT, Hwang JJ. Potential role of follicle-stimulating hormone (FSH) and transforming growth factor (TGFβ1) in the regulation of ovarian angiogenesis. J Cell Physiol 2011; 226:1608-19. [DOI: 10.1002/jcp.22491] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
33
|
Tilgner K, Atkinson SP, Yung S, Golebiewska A, Stojkovic M, Moreno R, Lako M, Armstrong L. Expression of GFP under the control of the RNA helicase VASA permits fluorescence-activated cell sorting isolation of human primordial germ cells. Stem Cells 2010; 28:84-92. [PMID: 19937754 DOI: 10.1002/stem.263] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The isolation of significant numbers of human primordial germ cells at several developmental stages is important for investigations of the mechanisms by which they are able to undergo epigenetic reprogramming. Only small numbers of these cells can be obtained from embryos of appropriate developmental stages, so the differentiation of human embryonic stem cells is essential to obtain sufficient numbers of primordial germ cells to permit epigenetic examination. Despite progress in the enrichment of human primordial germ cells using fluorescence-activated cell sorting (FACS), there is still no definitive marker of the germ cell phenotype. Expression of the widely conserved RNA helicase VASA is restricted to germline cells, but in contrast to species such as Mus musculus in which reporter constructs expressing green fluorescent protein (GFP) under the control of a Vasa promoter have been developed, such reporter systems are lacking in human in vitro models. We report here the generation and characterization of human embryonic stem cell lines stably carrying a VASA-pEGFP-1 reporter construct that expresses GFP in a population of differentiating human embryonic stem cells that show expression of characteristic markers of primordial germ cells. This population shows a different pattern of chromatin modifications to those obtained by FACS enrichment of Stage Specific Antigen one expressing cells in our previous publication.
Collapse
Affiliation(s)
- Katarzyna Tilgner
- North East Stem Cell Institute, Newcastle University, International Centre for Life, NE1 3BZ, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Hwu YM, Li SH, Lee RKK, Lin MH, Tsai YH, Yeh TS. Luteinizing hormone increases platelet-derived growth factor-D gene expression in human granulosa–luteal cells. Fertil Steril 2009; 92:2065-8. [DOI: 10.1016/j.fertnstert.2009.05.064] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 05/19/2009] [Accepted: 05/27/2009] [Indexed: 11/15/2022]
|
35
|
Woad KJ, Hammond AJ, Hunter M, Mann GE, Hunter MG, Robinson RS. FGF2 is crucial for the development of bovine luteal endothelial networks in vitro. Reproduction 2009; 138:581-8. [DOI: 10.1530/rep-09-0030] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The development of the corpus luteum requires angiogenesis, and involves the complex interplay between factors such as vascular endothelial growth factor A (VEGFA), fibroblast growth factor 2 (FGF2) and platelet-derived growth factor (PDGF). However, the relative role of these factors remains to be elucidated. This study used a new physiologically relevant mixed luteal cell culture system to test the hypotheses that: a) FGF2 and VEGFA are critical for bovine luteal angiogenesis; and b) local luteal PDGF signalling stimulates the formation of endothelial networks. Cells were treated with receptor tyrosine kinase inhibitors against VEGFA (SU1498), FGF2 (SU5402) or PDGF (AG1295) activity. After 9 days in culture, endothelial cells were immunostained for von Willebrand factor (VWF) and quantified by image analysis. Highly organised intricate endothelial networks were formed in the presence of exogenous VEGFA and FGF2. The inhibition of FGF2 activity reduced the total area of VWF staining versus controls (>95%; P<0.001). Inhibition of VEGF and PDGF activity reduced the endothelial network formation by more than 60 and 75% respectively (P<0.05). Progesterone production increased in all treatments from day 1 to 7 (P<0.001), and was unaffected by FGF2 or PDGF receptor kinase inhibition (P>0.05), but was reduced by the VEGF receptor inhibitor on days 5 and 7 (P<0.001). In conclusion, this study confirmed that VEGF signalling regulates both bovine luteal angiogenesis and progesterone production. However, FGF2 was crucial for luteal endothelial network formation. Also, for the first time, this study showed that local luteal PDGF activity regulates bovine luteal endothelial network formation in vitro.
Collapse
|
36
|
Abstract
In humans and other mammalian species, the pool of resting primordial follicles serves as the source of developing follicles and fertilizable ova for the entire length of female reproductive life. One question that has intrigued biologists is: what are the mechanisms controlling the activation of dormant primordial follicles. Studies from previous decades have laid a solid, but yet incomplete, foundation. In recent years, molecular mechanisms underlying follicular activation have become more evident, mainly through the use of genetically modified mouse models. As hypothesized in the 1990s, the pool of primordial follicles is now known to be maintained in a dormant state by various forms of inhibitory machinery, which are provided by several inhibitory signals and molecules. Several recently reported mutant mouse models have shown that a synergistic and coordinated suppression of follicular activation provided by multiple inhibitory molecules is necessary to preserve the dormant follicular pool. Loss of function of any of the inhibitory molecules for follicular activation, including PTEN (phosphatase and tensin homolog deleted on chromosome 10), Foxo3a, p27, and Foxl2, leads to premature and irreversible activation of the primordial follicle pool. Such global activation of the primordial follicle pool leads to the exhaustion of the resting follicle reserve, resulting in premature ovarian failure in mice. In this review, we summarize both historical and recent results on mammalian primordial follicular activation and focus on the up-to-date knowledge of molecular networks controlling this important physiological event. We believe that information obtained from mutant mouse models may also reflect the molecular machinery responsible for follicular activation in humans. These advances may provide a better understanding of human ovarian physiology and pathophysiology for future clinical applications.
Collapse
Affiliation(s)
- Deepak Adhikari
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | | |
Collapse
|
37
|
Schmahl J, Rizzolo K, Soriano P. The PDGF signaling pathway controls multiple steroid-producing lineages. Genes Dev 2009; 22:3255-67. [PMID: 19056881 DOI: 10.1101/gad.1723908] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The platelet-derived growth factor (PDGF) signaling pathway regulates numerous lineages of mesenchymal cell origin during development and in the adult. The transcriptional targets of this pathway have been shown to be required in several PDGF-dependent processes, but the roles of these targets in specific tissues is just beginning to be identified. In this study, we show that five different PDGF target genes are essential for male and/or female fertility. Mutations in each of these five different genes lead to defects in the steroid-producing cells in the testis and/or ovary and altered hormone production, suggesting that the PDGF pathway controls steroidogenesis through these genes in both sexes. Furthermore, conditional mutations of both PDGF receptors revealed a requirement in steroid-producing cells in multiple organs, including the testis, ovary, and adrenal cortex. Therefore, PDGF signaling may constitute a common mechanism in the control of multiple steroidogenic lineages.
Collapse
Affiliation(s)
- Jennifer Schmahl
- Program in Developmental Biology, Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | | | | |
Collapse
|