1
|
Chaves MA, Ferst JG, Fiorenza MF, Vit FF, da Silveira JC. The Influence of Ovarian-Derived Extracellular Vesicles in Reproduction. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2025. [PMID: 39741214 DOI: 10.1007/102_2024_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
In this chapter, we explore the multifaceted roles of extracellular vesicles (EVs) in ovarian biology, focusing on their contributions to folliculogenesis, oocyte competence, corpus luteum function, and immune response regulation. EVs, particularly those derived from follicular fluid (ffEVs), are crucial mediators of cell-to-cell communication within the ovarian follicle, influencing processes such as meiotic progression, stress response, and hormonal regulation. We review preexisting literature, highlighting key findings on the molecular cargo of EVs, such as miRNAs and proteins, and their involvement in regulating the function of the follicle cells. Additionally, the influence of EVs on the immune responses within the ovary was also addressed. Some attention is given to the potential of EVs as non-invasive biomarkers and therapeutic tools, particularly in addressing conditions like premature ovarian insufficiency and polycystic ovary syndrome. By discussing the existing challenges and emerging research, we hope that this chapter will provide a deeper understanding of EVs' therapeutic potential and offer insights or suggestions for advancing assisted reproductive technologies.
Collapse
Affiliation(s)
- Matheus A Chaves
- Laboratory of Molecular Morphophysiology and Development, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Juliana G Ferst
- Laboratory of Molecular Morphophysiology and Development, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Mariani F Fiorenza
- Laboratory of Molecular Morphophysiology and Development, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Franciele F Vit
- Laboratory of Molecular Morphophysiology and Development, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Juliano C da Silveira
- Laboratory of Molecular Morphophysiology and Development, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
2
|
Mi C, Chen W, Zhang Y, Yang Y, Zhao J, Xu Z, Sun Y, Fan Q, Huang W, Guo G, Zhang H. BaP/BPDE suppresses human trophoblast cell migration/invasion and induces unexplained miscarriage by up-regulating a novel lnc-HZ11 in extracellular vesicles: An intercellular study. ENVIRONMENT INTERNATIONAL 2024; 188:108750. [PMID: 38788414 DOI: 10.1016/j.envint.2024.108750] [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: 01/21/2024] [Revised: 04/20/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024]
Abstract
Extracellular vesicles (EVs) mediate the intercellular crosstalk by transferring functional cargoes. Recently, we have discovered that BaP/BPDE exposure suppresses trophoblast cell migration/invasion and induces miscarriage, which are also regulate by lncRNAs at intracelluar levels. However, the EVs-mediated intercellular regulatory mechanisms are completely unexplored. Specifically, whether EVs might transfer BPDE-induced toxic lncRNA to fresh recipient trophoblast cells and suppress their migration/invasion to further induce miscarriage is completely unknown. In this study, we find that BPDE exposure up-regulates a novel lnc-HZ11, which suppresses EGR1/NF-κB/CXCL12 pathway and migration/invasion of trophoblast cells. Intercellular studies show that EV-HZ11 (lnc-HZ11 in EVs), which is highly expressed in BPDE-exposed donor cells, suppresses EGR1/NF-κB/CXCL12 pathway and migration/invasion in recipient cells by transferring lnc-HZ11 through EVs. Analysis of villous tissues collected from UM (unexplained miscarriage) patients and HC (healthy control) group shows that the levels of BPDE-DNA adducts, lnc-HZ11 or EV-lnc-HZ11, and EGR1/NF-κB/CXCL12 pathway are all associated with miscarriage. Mouse assays show that BaP exposure up-regulates the levels of lnc-Hz11 or EV-Hz11, suppresses Egr1/Nf-κb/Cxcl12 pathway, and eventually induces miscarriage. Knockdown of lnc-Hz11 by injecting EV-AS-Hz11 could effectively alleviate miscarriage in BaP-exposed mice. Furthermore, EV-HZ11 in serum samples could well predict the risk of miscarriage. Collectively, this study not only discovers EVs-HZ11-mediated intercellular mechanisms that BaP/BPDE suppresses trophoblast cell migration/invasion and induces miscarriage but also provides new approach for treatment against unexplained miscarriage through EV-HZ11.
Collapse
Affiliation(s)
- Chenyang Mi
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China; School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, 571199, China
| | - Weina Chen
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Ying Zhang
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Yang Yang
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Jingsong Zhao
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Zhongyan Xu
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Yi Sun
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Qigang Fan
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Wenxin Huang
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Geng Guo
- Department of Emergency, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China.
| | - Huidong Zhang
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China.
| |
Collapse
|
3
|
Fiorentino G, Merico V, Zanoni M, Comincini S, Sproviero D, Garofalo M, Gagliardi S, Cereda C, Lin CJ, Innocenti F, Taggi M, Vaiarelli A, Ubaldi FM, Rienzi L, Cimadomo D, Garagna S, Zuccotti M. Extracellular vesicles secreted by cumulus cells contain microRNAs that are potential regulatory factors of mouse oocyte developmental competence. Mol Hum Reprod 2024; 30:gaae019. [PMID: 38745364 DOI: 10.1093/molehr/gaae019] [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: 06/30/2023] [Revised: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
The role of cumulus cells (CCs) in the acquisition of oocyte developmental competence is not yet fully understood. In a previous study, we matured cumulus-denuded fully-grown mouse oocytes to metaphase II (MII) on a feeder layer of CCs (FL-CCs) isolated from developmentally competent (FL-SN-CCs) or incompetent (FL-NSN-CCs) SN (surrounded nucleolus) or NSN (not surrounding nucleolus) oocytes, respectively. We observed that oocytes cultured on the former could develop into blastocysts, while those matured on the latter arrested at the 2-cell stage. To investigate the CC factors contributing to oocyte developmental competence, here we focused on the CCs' release into the medium of extracellular vesicles (EVs) and on their miRNA content. We found that, during the 15-h transition to MII, both FL-SN-CCs and FL-NSN-CCs release EVs that can be detected, by confocal microscopy, inside the zona pellucida (ZP) or the ooplasm. The majority of EVs are <200 nm in size, which is compatible with their ability to cross the ZP. Next-generation sequencing of the miRNome of FL-SN-CC versus FL-NSN-CC EVs highlighted 74 differentially expressed miRNAs, with 43 up- and 31 down-regulated. Although most of these miRNAs do not have known roles in the ovary, in silico functional analysis showed that seven of these miRNAs regulate 71 target genes with specific roles in meiosis resumption (N = 24), follicle growth (N = 23), fertilization (N = 1), and the acquisition of oocyte developmental competence (N = 23). Overall, our results indicate CC EVs as emerging candidates of the CC-to-oocyte communication axis and uncover a group of miRNAs as potential regulatory factors.
Collapse
Affiliation(s)
- Giulia Fiorentino
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology 'Lazzaro Spallanzani', University of Pavia, Pavia, Italy
| | - Valeria Merico
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology 'Lazzaro Spallanzani', University of Pavia, Pavia, Italy
| | - Mario Zanoni
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology 'Lazzaro Spallanzani', University of Pavia, Pavia, Italy
| | - Sergio Comincini
- Functional Genomics Laboratory, Department of Biology and Biotechnology 'Lazzaro Spallanzani', University of Pavia, Pavia, Italy
| | - Daisy Sproviero
- IFOM, IFOM-The FIRC Institute of Molecular Oncology, Milan, Italy
| | - Maria Garofalo
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Stella Gagliardi
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Cristina Cereda
- Department of Pediatrics, Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, Milan, Italy
| | - Chih-Jen Lin
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Federica Innocenti
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Marilena Taggi
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Alberto Vaiarelli
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | | | - Laura Rienzi
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Danilo Cimadomo
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Silvia Garagna
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology 'Lazzaro Spallanzani', University of Pavia, Pavia, Italy
| | - Maurizio Zuccotti
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology 'Lazzaro Spallanzani', University of Pavia, Pavia, Italy
| |
Collapse
|
4
|
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
|
5
|
Wei S, Tang W, Chen D, Xiong J, Xue L, Dai Y, Guo Y, Wu C, Dai J, Wu M, Wang S. Multiomics insights into the female reproductive aging. Ageing Res Rev 2024; 95:102245. [PMID: 38401570 DOI: 10.1016/j.arr.2024.102245] [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: 11/09/2023] [Revised: 01/22/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024]
Abstract
The human female reproductive lifespan significantly diminishes with age, leading to decreased fertility, reduced fertility quality and endocrine function disorders. While many aspects of aging in general have been extensively documented, the precise mechanisms governing programmed aging in the female reproductive system remain elusive. Recent advancements in omics technologies and computational capabilities have facilitated the emergence of multiomics deep phenotyping. Through the application and refinement of various high-throughput omics methods, a substantial volume of omics data has been generated, deepening our comprehension of the pathogenesis and molecular underpinnings of reproductive aging. This review highlights current and emerging multiomics approaches for investigating female reproductive aging, encompassing genomics, epigenomics, transcriptomics, proteomics, metabolomics, and microbiomics. We elucidate their influence on fundamental cell biology and translational research in the context of reproductive aging, address the limitations and current challenges associated with multiomics studies, and offer a glimpse into future prospects.
Collapse
Affiliation(s)
- Simin Wei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Weicheng Tang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Dan Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Liru Xue
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Yican Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Chuqing Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China.
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China; Ministry of Education, Key Laboratory of Cancer Invasion and Metastasis, Wuhan, China.
| |
Collapse
|
6
|
Zhou Q, Liu Z, Liao Z, Zhang Y, Qu M, Wu F, Tian J, Zhao H, Peng Q, Zheng W, Huang M, Yang S. miRNA profiling of granulosa cell-derived exosomes reveals their role in promoting follicle development. J Cell Physiol 2024; 239:20-35. [PMID: 38149730 DOI: 10.1002/jcp.31140] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/23/2023] [Accepted: 10/03/2023] [Indexed: 12/28/2023]
Abstract
To explore whether granulosa cell (GC)-derived exosomes (GC-Exos) and follicular fluid-derived exosomes (FF-Exos) have functional similarities in follicle development and to establish relevant experiments to validate whether GC-Exos could serve as a potential substitute for follicular fluid-derived exosomes to improve folliculogenesis. GC-Exos were characterized. MicroRNA (miRNA) profiles of exosomes from human GCs and follicular fluid were analyzed in depth. The signature was associated with folliculogenesis, such as phosphatidylinositol 3 kinases-protein kinase B signal pathway, mammalian target of rapamycin signal pathway, mitogen-activated protein kinase signal pathway, Wnt signal pathway, and cyclic adenosine monophosphate signal pathway. A total of five prominent miRNAs were found to regulate the above five signaling pathways. These miRNAs include miRNA-486-5p, miRNA-10b-5p, miRNA-100-5p, miRNA-99a-5p, and miRNA-21-5p. The exosomes from GCs and follicular fluid were investigated to explore the effect on folliculogenesis by injecting exosomes into older mice. The proportion of follicles at each stage is counted to help us understand folliculogenesis. Exosomes derived from GCs were isolated successfully. miRNA profiles demonstrated a remarkable overlap between the miRNA profiles of FF-Exos and GC-Exos. The shared miRNA signature exhibited a positive influence on follicle development and activation. Furthermore, exosomes derived from GCs and follicular fluid promoted folliculogenesis in older female mice. Exosomes derived from GCs had similar miRNA profiles and follicle-promoting functions as follicular fluid exosomes. Consequently, GC-Exos are promising for replacing FF-Exos and developing new commercial reagents to improve female fertility.
Collapse
Affiliation(s)
- Qilin Zhou
- Department of Health Inspection and Quarantine, School of Public Health, Guangdong Medical University, Dongguan, China
- Department of Reproductive Medicine, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhen Liu
- Department of Reproductive Medicine, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Zhengdong Liao
- Department of Reproductive Medicine, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yangzhuohan Zhang
- School of Clinical Medicine, Hubei University of Science and Technology, Xianning, China
| | - Mengyuan Qu
- Department of Reproductive Medicine, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Fanggui Wu
- Department of Reproductive Medicine, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jingyan Tian
- Department of Reproductive Medicine, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Huan Zhao
- Department of Reproductive Medicine, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qianwen Peng
- Department of Reproductive Medicine, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Wenchao Zheng
- Department of Reproductive Medicine, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Mingyuan Huang
- Department of Health Inspection and Quarantine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Sheng Yang
- Department of Reproductive Medicine, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
7
|
Peng P, Wang X, Qiu C, Zheng W, Zhang H. Extracellular vesicles from human umbilical cord mesenchymal stem cells prevent steroid-induced avascular necrosis of the femoral head via the PI3K/AKT pathway. Food Chem Toxicol 2023; 180:114004. [PMID: 37634611 DOI: 10.1016/j.fct.2023.114004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
Extracellular vesicles (EVs) secreted by human umbilical cord mesenchymal stem cells (hucMSC) have excellent therapeutic potential for many diseases. The aim of this study was to define the role of hucMSC-EVs in the prevention and treatment of steroid-induced avascular necrosis of the femoral head (SANFH). After establishing the SANFH rat model, the effects of hucMSC-EVs were assessed by measuring the microstructure of the femoral head using HE staining, micro-computed tomography (micro-CT), and TUNEL staining. The administration of hucMSC-EVs caused a significant reduction to glucocorticoids (GCs)-induced osteoblast apoptosis and empty lacuna of the femoral head, while effectively improving the microstructure. HucMSC-EVs rescued the deactivation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway induced by GCs, and reversed the proliferation and migration of osteoblasts inhibited by GCs. In addition, hucMSC-EVs attenuated the inhibitory effects of GCs on rat osteoblast osteogenesis, angiogenesis of endothelial cells, and prevented osteoblast apoptosis. However, the promoting effects of hucMSC-EVs were abolished following the blockade of PI3K/AKT on osteoblasts. hucMSC-EVs were found to prevent glucocorticoid-induced femoral head necrosis in rats through the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Puji Peng
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, 450003, China; Department of Orthopedics, Zhengzhou University People's Hospital, Zhengzhou, 450003, China
| | - XueZhong Wang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chen Qiu
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430000, China
| | - Wendi Zheng
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, 450003, China; Department of Orthopedics, Zhengzhou University People's Hospital, Zhengzhou, 450003, China.
| | - Hongjun Zhang
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, 450003, China; Department of Orthopedics, Zhengzhou University People's Hospital, Zhengzhou, 450003, China.
| |
Collapse
|
8
|
Shoorei H, Seify M, Talebi SF, Majidpoor J, Dehaghi YK, Shokoohi M. Different types of bisphenols alter ovarian steroidogenesis: Special attention to BPA. Heliyon 2023; 9:e16848. [PMID: 37303564 PMCID: PMC10250808 DOI: 10.1016/j.heliyon.2023.e16848] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/27/2023] [Accepted: 05/31/2023] [Indexed: 06/13/2023] Open
Abstract
Endocrine disruptors such as bisphenol A (BPA) and some of its analogues, including BPS, BPAF, and BPE, are used extensively in the manufacture of plastics. These synthetic chemicals could seriously alter the functionality of the female reproductive system. Although the number of studies conducted on other types of bisphenols is smaller than the number of studies on BPA, the purpose of this review study was to evaluate the effects of bisphenol compounds, particularly BPA, on hormone production and on genes involved in ovarian steroidogenesis in both in vitro (human and animal cell lines) and in vivo (animal models) studies. The current data show that exposure to bisphenol compounds has adverse effects on ovarian steroidogenesis. For example, BPA, BPS, and BPAF can alter the normal function of the hypothalamic-pituitary-gonadal (HPG) axis by targeting kisspeptin neurons involved in steroid feedback signals to gonadotropin-releasing hormone (GnRH) cells, resulting in abnormal production of LH and FSH. Exposure to BPA, BPS, BPF, and BPB had adverse effects on the release of some hormones, namely 17-β-estradiol (E2), progesterone (P4), and testosterone (T). BPA, BPE, BPS, BPF, and BPAF are also capable of negatively altering the transcription of a number of genes involved in ovarian steroidogenesis, such as the steroidogenic acute regulatory protein (StAR, involved in the transfer of cholesterol from the outer to the inner mitochondrial membrane, where the steroidogenesis process begins), cytochrome P450 family 17 subfamily A member 1 (Cyp17a1, which is involved in the biosynthesis of androgens such as testosterone), 3 beta-hydroxysteroid dehydrogenase enzyme (3β-HSD, involved in the biosynthesis of P4), and cytochrome P450 family 19 subfamily A member 1 (Cyp19a1, involved in the biosynthesis of E2). Exposure to BPA, BPB, BPF, and BPS at prenatal or prepubertal stages could decrease the number of antral follicles by activating apoptosis and autophagy pathways, resulting in decreased production of E2 and P4 by granulosa cells (GCs) and theca cells (TCs), respectively. BPA and BPS impair ovarian steroidogenesis by reducing the function of some important cell receptors such as estrogens (ERs, including ERα and ERβ), progesterone (PgR), the orphan estrogen receptor gamma (ERRγ), the androgen receptor (AR), the G protein-coupled estrogen receptor (GPER), the FSHR (follicle-stimulating hormone receptor), and the LHCGR (luteinizing hormone/choriogonadotropin receptor). In animal models, the effects of bisphenol compounds depend on the type of animals, their age, and the duration and dose of bisphenols, while in cell line studies the duration and doses of bisphenols are the matter.
Collapse
Affiliation(s)
- Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Seify
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyedeh Fahimeh Talebi
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Department of Pharmacology, Birjand University of Medical Sciences, Birjand, Iran
| | - Jamal Majidpoor
- Department of Anatomy, Faculty of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Yeganeh Koohestani Dehaghi
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Majid Shokoohi
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
9
|
Costa CB, Fair T, Seneda MM. Review: Environment of the ovulatory follicle: modifications and use of biotechnologies to enhance oocyte competence and increase fertility in cattle. Animal 2023; 17 Suppl 1:100866. [PMID: 37567670 DOI: 10.1016/j.animal.2023.100866] [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: 11/23/2022] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 08/13/2023] Open
Abstract
The oocyte is the basis of life, supporting development from a fertilized cell to an independent multicellular organism. The oocyte's competence to drive the first cell cycles postfertilization are critical to embryonic survival and subsequent successful pregnancy. Coupled with the complex processes of follicle assembly, activation, differentiation, growth, and terminal maturation, oocyte developmental competence is gradually acquired during oocyte growth and meiotic maturation. Most reproduction management technologies and interventions are centered around these highly coordinated processes, targeting the ovarian follicle and the oocyte within. Thus, our objective was to highlight key aspects of oocyte and follicle development in cattle, and to discuss recent advances in oocyte and follicle-centered reproductive biotechnologies.
Collapse
Affiliation(s)
- Camila Bortoliero Costa
- Department of Biological Sciences, School of Sciences and Languages, São Paulo State University (UNESP), Campus Assis, São Paulo, Brazil; Graduate Program in Pharmacology and Biotechnology, Institute of Biosciences, UNESP, Botucatu, São Paulo, Brazil
| | - Trudee Fair
- School of Agriculture and Food Science, University College Dublin, Ireland
| | - Marcelo M Seneda
- State University of Londrina (UEL), Laboratory of Animal Reproduction, Londrina, PR, Brazil.
| |
Collapse
|
10
|
Nejabati HR, Roshangar L, Nouri M. Follicular fluid extracellular vesicle miRNAs and ovarian aging. Clin Chim Acta 2023; 538:29-35. [PMID: 36368351 DOI: 10.1016/j.cca.2022.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022]
Abstract
The decrease in the reproductive potential due to aging occurs as a gradual decline in the quantity and quality of the ovarian reserve, a phenomenon associated with risk of miscarriage, pregnancy loss, low ovarian stimulation, and oocyte abnormalities, such as chromosomal aneuploidies. Numerous studies have shown that the fertility potential of older women is decreased by changes to the cellular composition of the follicles. Additionally, a unique method of cellular communication has been identified which involves the release of extracellular vesicles (EVs) in various body fluids including follicular fluid (FF). The changing composition of EVs especially non-coding RNAs, such as miRNAs has been documented across a broad range of cell types during aging. Accordingly, alterations of miRNA cargo within FF-derived EVs due to increased age may serve as a potential predictor of oocyte quality. In this review we examine the relationship between FF EV miRNAs and ovarian aging.
Collapse
Affiliation(s)
- Hamid Reza Nejabati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Bastos NM, Goulart RS, Bambil DB, Bridi A, Mazzarella R, Alves L, da Silva Rosa PM, Neto AL, Silva SL, de Almeida Santana MH, Negrão JA, Pugliesi G, Meirelles FV, Perecin F, da Silveira JC. High body energy reserve influences extracellular vesicles miRNA contents within the ovarian follicle. PLoS One 2023; 18:e0280195. [PMID: 36626404 PMCID: PMC9831338 DOI: 10.1371/journal.pone.0280195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
Aiming to evaluate the effects of increased body energy reserve (BER) in Nellore cows' reproductive efficiency, cows were fed with different nutritional plans to obtain animals with high BER (HBER; Ad libitum diet) and moderate BER (MBER: cows fed 70% of HBER group ingestion). To evaluate the BER, cows were weekly weighted and evaluated for subcutaneous fat thickness and insulin serum concentration along the experimental period. At the end of the experimental period, animals were submitted to estrous synchronization and artificial insemination. Animals were slaughtered approximately 120 h after ovulation induction and the reproductive tracts were collected for embryo recovery and samples collection. Cumulus-oocyte-complexes (COC) and follicular fluid were collected from 3-6 mm in diameter ovarian follicles to perform miRNA analysis of cumulus cells (CC) and extracellular vesicles from follicular fluid (EV FF). As expected, differences were observed among MBER and HBER groups for body weight, fat thickness, and insulin serum concentration. HBER animals showed lower ovulation and embryo recovery rates compared to MBER animals. Different miRNAs were found among CC and EV FF within groups, suggesting that the BER may influence follicular communication. This suggests that small follicles (3-6 mm diameter) are already under BER effects, which may be greater on later stages of follicular development.
Collapse
Affiliation(s)
- Natália Marins Bastos
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Rodrigo Silva Goulart
- Department of Animal Science, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Danilo Brito Bambil
- Department of Animal Science, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Alessandra Bridi
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Rosane Mazzarella
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Luana Alves
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Paola Maria da Silva Rosa
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Adomar Laurindo Neto
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Saulo Luz Silva
- Department of Animal Science, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | | | - João Alberto Negrão
- Department of Basic Science, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Guilherme Pugliesi
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Flávio Vieira Meirelles
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Felipe Perecin
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Juliano Coelho da Silveira
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
12
|
Gurunathan S, Kang MH, Song H, Kim NH, Kim JH. The role of extracellular vesicles in animal reproduction and diseases. J Anim Sci Biotechnol 2022; 13:62. [PMID: 35681164 PMCID: PMC9185900 DOI: 10.1186/s40104-022-00715-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/05/2022] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EVs) are nanosized membrane-enclosed compartments that serve as messengers in cell-to-cell communication, both in normal physiology and in pathological conditions. EVs can transfer functional proteins and genetic information to alter the phenotype and function of recipient cells, which undergo different changes that positively affect their structural and functional integrity. Biological fluids are enriched with several subpopulations of EVs, including exosomes, microvesicles (MVs), and apoptotic bodies carrying several cargoes, such as lipids, proteins, and nucleic acids. EVs associated with the reproductive system are actively involved in the regulation of different physiological events, including gamete maturation, fertilization, and embryo and fetal development. EVs can influence follicle development, oocyte maturation, embryo production, and endometrial-conceptus communication. EVs loaded with cargoes are used to diagnose various diseases, including pregnancy disorders; however, these are dependent on the type of cell of origin and pathological characteristics. EV-derived microRNAs (miRNAs) and proteins in the placenta regulate inflammatory responses and trophoblast invasion through intercellular delivery in the placental microenvironment. This review presents evidence regarding the types of extracellular vesicles, and general aspects of isolation, purification, and characterization of EVs, particularly from various types of embryos. Further, we discuss EVs as mediators and messengers in reproductive biology, the effects of EVs on placentation and pregnancy disorders, the role of EVs in animal reproduction, in the male reproductive system, and mother and embryo cross-communication. In addition, we emphasize the role of microRNAs in embryo implantation and the role of EVs in reproductive and therapeutic medicine. Finally, we discuss the future perspectives of EVs in reproductive biology.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Min-Hee Kang
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Hyuk Song
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Nam Hyung Kim
- Guangdong Provincial Key Laboratory of Large Animal models for Biomedicine, Wuyi University, Jiangmen, 529020, China
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea.
| |
Collapse
|
13
|
Zhang G, Cui Z, Li J, Zhang D, Li Z, Lin Z, Yin H, Ran J, Wang Y, Liu Y. miR-122-5p regulates proliferation and apoptosis of chicken granulosa cells of hierarchal follicles by targeting MAPK3. Gene X 2022; 824:146397. [PMID: 35276240 DOI: 10.1016/j.gene.2022.146397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 01/22/2023] Open
Abstract
Chicken follicles plays a crucial role in the reproductive performance, especially in laying period. Recently, miR-122-5p has been found to be differentially expressed in the ovaries of rats with polycystic ovary syndrome and normal rats, indicating the potential role of miR-122-5p in the development of granulosa cells (GCs). In present study, we found that miR-122-5p was highly expressed in chicken atrophic ovaries. Herein, we investigated its function on GC proliferation and apoptosis of chicken in vitro. We found that overexpression of miR-122-5p significantly inhibited proliferation and promoted apoptosis of GCs, whereas the opposite effects were detected in miR-122-5p knockdown GCs. Meanwhile, mitogen-activated protein kinase 3 (MAPK3) was confirmed as a new target gene of miR-122-5p by bioinformatics software prediction and the dual-luciferase reporter assay verification. Furthermore, after knockdown of MAPK3, the function of MAPK3 for GC proliferation and apoptosis was opposite to that of miR-122-5p. Collectively, our results indicated that miR-122-5p impeded chicken GC proliferation and promoted apoptosis through the post-transcriptional downregulation of MAPK3.
Collapse
Affiliation(s)
- Guangfa Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211 Huimin Road, Wenjiang, Sichuan province, Chengdu 611130, China
| | - Zhifu Cui
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211 Huimin Road, Wenjiang, Sichuan province, Chengdu 611130, China
| | - Jingjing Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211 Huimin Road, Wenjiang, Sichuan province, Chengdu 611130, China
| | - Donghao Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211 Huimin Road, Wenjiang, Sichuan province, Chengdu 611130, China
| | - Zhiqiang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211 Huimin Road, Wenjiang, Sichuan province, Chengdu 611130, China
| | - Zhongzhen Lin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211 Huimin Road, Wenjiang, Sichuan province, Chengdu 611130, China
| | - Huadong Yin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211 Huimin Road, Wenjiang, Sichuan province, Chengdu 611130, China
| | - Jinshan Ran
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211 Huimin Road, Wenjiang, Sichuan province, Chengdu 611130, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211 Huimin Road, Wenjiang, Sichuan province, Chengdu 611130, China
| | - Yiping Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 211 Huimin Road, Wenjiang, Sichuan province, Chengdu 611130, China.
| |
Collapse
|
14
|
Aleksejeva E, Zarovni N, Dissanayake K, Godakumara K, Vigano P, Fazeli A, Jaakma Ü, Salumets A. Extracellular vesicle research in reproductive science- Paving the way for clinical achievements. Biol Reprod 2022; 106:408-424. [PMID: 34982163 DOI: 10.1093/biolre/ioab245] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/13/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
Mammalian conception involves a multitude of reciprocal interactions via a molecular dialogue between mother and conceptus. Extracellular vesicles (EVs) are secreted membrane-encapsulated particles that mediate cell-to-cell communication in various contexts. EVs, which are present in seminal, follicular, oviductal, and endometrial fluids, as well as in embryo secretions, carry molecular constituents that impact gamete maturation, fertilization, early embryo development, and embryo-maternal communication. The distribution, concentration, and molecular cargo of EVs are regulated by steroid hormones and the health status of the tissue of origin, and thus are influenced by menstrual phase, stage of conception, and the presence of infertility-associated diseases. EVs have been recognized as a novel source of biomarkers and potential reproductive medicine therapeutics, particularly for assisted reproductive technology (ART). There are still many technological and scientific hindrances to be overcome before EVs can be used in clinical diagnostic and therapeutic ART applications. Issues to be resolved include the lack of standardized measurement protocols and an absence of absolute EV quantification technologies. Additionally, clinically suitable and robust EV isolation methods have yet to be developed. In this review, we provide an overview of EV-mediated interactions during the early stages of reproduction from gamete maturation to embryo implantation and then outline the technological progress that must be made for EV applications to be translated to clinical settings.
Collapse
Affiliation(s)
- Elina Aleksejeva
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51014 Tartu, Estonia.,Competence Centre on Health Technologies, 50411 Tartu, Estonia
| | | | - Keerthie Dissanayake
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51014 Tartu, Estonia.,Department of Anatomy, Faculty of Medicine, University of Peradeniya, 20400 Peradeniya, Sri Lanka.,Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Kasun Godakumara
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51014 Tartu, Estonia.,Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Paola Vigano
- Reproductive Sciences Laboratory, Gynecology/Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Alireza Fazeli
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51014 Tartu, Estonia.,Department of Anatomy, Faculty of Medicine, University of Peradeniya, 20400 Peradeniya, Sri Lanka.,Academic Unit of Reproductive and Developmental Medicine, Department of Oncology and Metabolism, Medical School, University of Sheffield, S10 2TN Sheffield, UK
| | - Ülle Jaakma
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51014 Tartu, Estonia
| | - Andres Salumets
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51014 Tartu, Estonia.,Competence Centre on Health Technologies, 50411 Tartu, Estonia.,Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia.,Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 14186 Stockholm, Sweden
| |
Collapse
|
15
|
Bastos NM, Ferst JG, Goulart RS, Coelho da Silveira J. The role of the oviduct and extracellular vesicles during early embryo development in bovine. Anim Reprod 2022; 19:e20220015. [PMID: 35493787 PMCID: PMC9037602 DOI: 10.1590/1984-3143-ar2022-0015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/17/2022] [Indexed: 11/21/2022] Open
Abstract
The oviduct is an important reproductive structure that connects the ovary to the uterus and takes place to important events such as oocyte final maturation, fertilization and early embryonic development. Thus, gametes and embryo can be directly influenced by the oviductal microenvironment composed by epithelial cells such secretory and ciliated cells and oviductal fluid. The oviduct composition is anatomically dynamic and is under ovarian hormones control. The oviductal fluid provides protection, nourishment and transport to gametes and embryo and allows interaction to oviductal epithelial cells. All these functions together allows the oviduct to provides the ideal environment to the early reproductive events. Extracellular vesicles (EVs) are biological nanoparticles that mediates cell communication and are present at oviductal fluid and plays an important role in gametes/embryo - oviductal cells communication. This review will present the ability of the oviducts based on its dynamic and systemic changes during reproductive events, as well as the contribution of EVs in this process.
Collapse
|
16
|
Lee SH, Saadeldin IM. Exosomes as a Potential Tool for Supporting Canine Oocyte Development. Animals (Basel) 2020; 10:E1971. [PMID: 33121043 PMCID: PMC7693116 DOI: 10.3390/ani10111971] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/24/2020] [Accepted: 10/25/2020] [Indexed: 12/27/2022] Open
Abstract
The canine oviduct is a unique reproductive organ where the ovulated immature oocytes complete their maturation, while the other mammals ovulate matured gametes. Due to their peculiar reproductive characteristics, the in vitro maturation of dog oocytes is still not wellestablished compared with other mammals. Investigations of the microenvironment conditions in the oviductal canal are required to establish a reliable in vitro maturation system in the dog. Previous studies have suggested that the oviduct and its derivatives play a key role in improving fertilization as well as embryo development. In particular, the biological function of oviduct-derived exosomes on sperm and early embryo development has been investigated in porcine, bovine, and murine species. However, the information about their functions on canine cumulus-oocyte complexes is still elusive. Recent canine reproductive studies demonstrated how oviduct-derived extracellular vesicles such as microvesicles and exosomes interact with oocyte-cumulus complexes and how they can play roles in regulating canine cumulus/oocyte communications. In this review, we summarize the physiological characteristics of canine oviduct-derived exosomes and their potential effects on cumulus cells development as well as oocyte in vitro maturation via molecular signaling pathways.
Collapse
Affiliation(s)
- Seok Hee Lee
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Islam M. Saadeldin
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 44511, Saudi Arabia;
- Department of Comparative Medicine, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
17
|
Qasemi M, Amidi F. Extracellular microRNA profiling in human follicular fluid: new biomarkers in female reproductive potential. J Assist Reprod Genet 2020; 37:1769-1780. [PMID: 32642870 PMCID: PMC7468023 DOI: 10.1007/s10815-020-01860-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are small, about 22 nucleotides, non-coding RNAs which regulate a wide range of gene expression during post-transcriptional stage. They are released into intra- and extracellular microenvironments and play vital roles in different physiological and pathological pathways. Due to easy accessibility, detection of extracellular miRNAs in body fluids, e.g. serum, plasma, cerebrospinal fluid, and follicular fluid, has been explored in recent years. Since miRNAs are stable at unsuitable conditions, scientists have been investigating to use them as biomarkers in different fields of medicines. It goes without saying that experienced biomarkers would be required in reproductive medicine as well. Biomarkers can help clinicians and embryologists to diagnose disorders and assess the embryo quality via molecular pattern which is more reliable than nowadays routine methods. Follicular fluid as a noninvasive fluid in assisted reproductive techniques (ART) has attracted researchers as a rich pool for biomarkers, and miRNAs are not exception. Although miRNA biomarkers in reproduction field are located on their initial stage and there is a long path to move forward, several meticulous studies have been performed and discovered their associations with various conditions. In this regard, we summarize the reported miRNAs in follicular fluid and their correlations with female infertility and ART success rate, while subsequent investigations are required.
Collapse
Affiliation(s)
- Maryam Qasemi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fardin Amidi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Pioltine EM, Machado MF, da Silveira JC, Fontes PK, Botigelli RC, Quaglio AEV, Costa CB, Nogueira MFG. Can extracellular vesicles from bovine ovarian follicular fluid modulate the in-vitro oocyte meiosis progression similarly to the CNP-NPR2 system? Theriogenology 2020; 157:210-217. [PMID: 32814248 DOI: 10.1016/j.theriogenology.2020.06.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 06/26/2020] [Accepted: 06/27/2020] [Indexed: 11/28/2022]
Abstract
C-type natriuretic peptide (CNP) and its natriuretic peptide receptors subtype 2 (NPR2) are essential for the maintenance of oocyte meiotic arrest in different species. Extracellular vesicles (EVs) in bovine follicular fluid (FF) are important for cell communication within the ovarian follicle. This study investigated the involvement of EVs from FF of bovine ovarian follicles in the CNP-NPR2 system, first by analyzing the presence of CNP in the EV contents, followed by addition of EVs to in-vitro maturation (IVM) medium, to evaluate the effect on maintenance of oocyte meiosis arrest and improvements in in-vitro embryo production. As expected, CNP was observed in FF and granulosa cells from the ovarian follicles. To the best of our knowledge, this is the first time that CNP has been found in the EV contents. To evaluate the possible effect of EVs on the progression of oocyte meiosis, the IVM was performed under three conditions: CNP and EV supplementation and control condition. Both the CNP and EV treatments inhibited meiosis resumption in the oocyte within 9 h of IVM. CNP treatment increased cGMP levels in cumulus cells within 6 h of IVM compared to the control group, but the EV treatment did not. In contrast, the relative mRNA abundance of adenylate cyclase 3 and 9 (ADCY3 and ADCY9) was upregulated in oocytes after 6 h of IVM under EV treatment compared to the control group, but not under CNP treatment. Last, these treatments in the IVM medium had no significant effect on the in-vitro embryo production. In conclusion, we demonstrated the presence of endogenous CNP in bovine reproductive structures, especially in the EVs from the FF of antral follicles. The presence of CNP in the EVs suggests an important involvement of this cell-communication system in the CNP-NPR2 system. Therefore, we indeed observed that the EVs from FF can modulate the arrest of oocyte meiosis, acting similarly to the CNP-NPR2 system to block the oocyte in the GV state. However, the mechanism of each system might be different; the CNP-NPR2 system seems to be involved in modulating the cGMP levels, while the contents of EVs might be involved in modulating the cAMP levels.
Collapse
Affiliation(s)
- Elisa M Pioltine
- São Paulo State University (UNESP), Institute of Biosciences, Department of Pharmacology, Botucatu, São Paulo, 18618-689, Brazil.
| | - Mariana F Machado
- São Paulo State University (UNESP), Institute of Biosciences, Department of Pharmacology, Botucatu, São Paulo, 18618-689, Brazil
| | - Juliano C da Silveira
- University of São Paulo (USP), Faculty of Animal Science and Food Engineering, Department of Veterinary Medicine, Pirassununga, São Paulo, 13635-900, Brazil
| | - Patrícia K Fontes
- São Paulo State University (UNESP), Institute of Biosciences, Department of Pharmacology, Botucatu, São Paulo, 18618-689, Brazil
| | - Ramon C Botigelli
- São Paulo State University (UNESP), Institute of Biosciences, Department of Pharmacology, Botucatu, São Paulo, 18618-689, Brazil
| | - Ana Elisa V Quaglio
- São Paulo State University (UNESP), Institute of Biosciences, Department of Pharmacology, Botucatu, São Paulo, 18618-689, Brazil
| | - Camila B Costa
- São Paulo State University (UNESP), Institute of Biosciences, Department of Pharmacology, Botucatu, São Paulo, 18618-689, Brazil
| | - Marcelo F G Nogueira
- São Paulo State University (UNESP), Institute of Biosciences, Department of Pharmacology, Botucatu, São Paulo, 18618-689, Brazil; São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Department of Biological Sciences, Assis, São Paulo, 19806-900, Brazil
| |
Collapse
|
19
|
Shomali N, Hemmatzadeh M, Yousefzadeh Y, Soltani-Zangbar MS, Hamdi K, Mehdizadeh A, Yousefi M. Exosomes: Emerging biomarkers and targets in folliculogenesis and endometriosis. J Reprod Immunol 2020; 142:103181. [PMID: 32717674 DOI: 10.1016/j.jri.2020.103181] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 06/14/2020] [Accepted: 07/18/2020] [Indexed: 12/31/2022]
Abstract
An appropriate connection of the cells in the ovary follicles is vital for a healthy ovule maturation and fertilization, and also for endometrium preparation for implantation that can cause endometriosis. Cellular communication within the follicle and endometrial epithelium involve many signaling molecules. Recent studies indicate that cellular communication can be enclosed by secretion and absorption of small membrane carriers which are named extracellular vesicles including exosomes and microvesicles. Understanding and defining these EVs (Extracellular vesicles) population are important for future studies and clinical translation. Here, we describe the various important cargos which are carried by exosomes during folliculogenesis and endometriosis. Additionally, the current knowledge of exosomes and their cargo within the FF (Follicular fluid) during the folliculogenesis and also in the intrauterine cavity which are involved in endometriosis lesions have also been summarized. Considering the potential importance of this form of the cell to cell communication in the reproductive system, the vital issues under discussion lead to a new insight in this rapidly expanding field and it may be an interesting approach for diagnostic, prognostic and especially therapeutic strategies in the field of infertility and assisted reproductive technology (ART).
Collapse
Affiliation(s)
- Navid Shomali
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hemmatzadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Yousefzadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Kobra Hamdi
- Reproductive Biology Department, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
20
|
Huang X, Wu B, Chen M, Hong L, Kong P, Wei Z, Teng X. Depletion of exosomal circLDLR in follicle fluid derepresses miR-1294 function and inhibits estradiol production via CYP19A1 in polycystic ovary syndrome. Aging (Albany NY) 2020; 12:15414-15435. [PMID: 32651991 PMCID: PMC7467373 DOI: 10.18632/aging.103602] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder in reproductive women and is characterized by polycystic ovaries, hyperandrogenism and chronic anovulation. Abnormal folliculogenesis is considered as a common characteristic of PCOS. Our aim is to identify the altered circRNA expression profile in exosomes isolated from follicular fluid (FF) of PCOS patients to investigate the molecular function of exosomal circRNA, as a vital mediator in follicular microenvironment, in the aetiology and pathobiology of PCOS. In this study, the circRNA expression profile of FF exosomes were compared between PCOS and control patients by RNA sequencing (N=5 vs 5). Sixteen circRNAs showed significantly different expression. GO and KEGG pathway analyses indicated that their parental genes were enriched in PCOS-related pathways, including ovarian steroidogenesis, aldosterone synthesis and secretion, and Jak-STAT signaling. Among sixteen differentially expressed circRNAs, hsa_circ_0006877 (circLDLR) was processed from its parental LDLR (low density lipoprotein receptor) transcript, which participated in ovarian steroidogenesis. Its depletion in PCOS FF exosomes was further verified in an additional cohort (N=25 vs 25) by qRT-PCR. And a circLDLR-miR-1294-CYP19A1 competing endogenous RNA (ceRNA) network was predicted by cytoscape software, and confirmed by luciferase assay and correlative expression in the cumulus cells of PCOS patients. Mechanistically, the intercellular transfer of functional circLDLR assay and its withdrawal experiments in KGN cells showed that depleting circLDLR in exosomes increased miR-1294 expression and inhibited CYP19A1 expression in recipient cells, as well as reduced their estrogen (E2) secretion. Our findings revealed a ceRNA network of circLDLR and provided new information on abnormal follicle development in PCOS.
Collapse
Affiliation(s)
- Xin Huang
- Department of Assisted Reproduction, and Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bi Wu
- Department of Assisted Reproduction, and Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Miaoxin Chen
- Department of Assisted Reproduction, and Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ling Hong
- Department of Assisted Reproduction, and Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Pengcheng Kong
- Department of Assisted Reproduction, and Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhiyun Wei
- Department of Assisted Reproduction, and Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoming Teng
- Department of Assisted Reproduction, and Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Bartolucci AF, Uliasz T, Peluso JJ. MicroRNA-21 as a regulator of human cumulus cell viability and its potential influence on the developmental potential of the oocyte. Biol Reprod 2020; 103:94-103. [PMID: 32333014 DOI: 10.1093/biolre/ioaa058] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/18/2020] [Accepted: 04/22/2020] [Indexed: 12/26/2022] Open
Abstract
MicroRNA-21 is expressed in bovine, murine, and human cumulus cells with its expression in murine and bovine cumulus cells correlated with oocyte developmental potential. The aim of this study was to assess the relationship between cumulus cell MIR-21 and human oocyte developmental potential. These studies revealed that both the immature and mature forms of MicroRNA-21 (MIR-21-5p) were elevated in cumulus cells of oocytes that developed into blastocysts compared to cumulus cells of oocytes that arrested prior to blastocyst formation. This increase in MicroRNA-21 was observed regardless of whether the oocytes developed into euploid or aneuploid blastocysts. Moreover, MIR-21-5p levels in cumulus cells surrounding oocytes that either failed to mature or matured to metaphase II but failed to fertilize, were ≈50% less than the MIR-21-5p levels associated with oocytes that arrested prior to blastocyst formation. Why cumulus cells associated with oocytes of reduced developmental potential expressed less MIR-21-5p is unknown. It is unlikely due to reduced expression of either the receptors of growth differentiation factor 9 or rosha Ribonuclease III (DROSHA) and Dicer Ribonuclease III (DICER) which sequentially promote the conversion of immature forms of MicroRNA-21 to mature MicroRNA-21. Furthermore, cultured cumulus cells treated with a MIR-21-5p inhibitor had an increase in apoptosis and a corresponding increase in the expression of PTEN, a gene known to inhibit the AKT-dependent survival pathway in cumulus cells. These studies provide evidence for a role of MicroRNA-21 in human cumulus cells that influences the developmental potential of human oocytes.
Collapse
Affiliation(s)
- Alison F Bartolucci
- Department of Obstetrics and Gynecology, Farmington, CT 06030, USA.,The Center for Advanced Reproductive Services, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Tracy Uliasz
- Department of Cell Biology, University of CT Health Center, Farmington, CT 06030, USA
| | - John J Peluso
- Department of Obstetrics and Gynecology, Farmington, CT 06030, USA.,Department of Cell Biology, University of CT Health Center, Farmington, CT 06030, USA
| |
Collapse
|
22
|
Rodosthenous RS, Baccarelli AA, Mansour A, Adir M, Israel A, Racowsky C, Hauser R, Bollati V, Machtinger R. Supraphysiological Concentrations of Bisphenol A Alter the Expression of Extracellular Vesicle-Enriched miRNAs From Human Primary Granulosa Cells. Toxicol Sci 2020; 169:5-13. [PMID: 30690568 DOI: 10.1093/toxsci/kfz020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bisphenol A (BPA) is a widely used chemical that has been detected in follicular fluid and associated with adverse reproductive effects. Granulosa cells have an important role in follicular growth and oocyte maturation, however, little is known about the biological mechanisms of BPA toxicity on human granulosa cells. In this study, we exposed primary granulosa cells to different concentrations of BPA (0, 20, 200, 2000, and 20 000 ng/ml) and used quantitative polymerase chain reaction to measure the expression levels of miRNAs enriched in extracellular vesicles (EV-enriched miRNAs), and cellular levels of selected target genes of differentially expressed EV-enriched miRNAs. We found that exposure to 20 000 ng/ml BPA was associated with decreased levels of EV-miR-27b-3p (FC = 0.58, p = .04) and increased levels of its biologically relevant target genes FADD (FC = 1.22, p = .01), IGF1 (FC = 1.59, p = .06), and PPARG (FC = 1.73, p = .001) as compared with the control. In addition, we observed that under the same exposure conditions, the expression levels of miR-27b-3p in granulosa cells were also downregulated (FC = 0.65, p = .03) as compared with the control. Our findings suggest that both cellular and extracellular changes in gene expression may mediate BPA toxicity in granulosa cells.
Collapse
Affiliation(s)
| | - Andrea A Baccarelli
- Human Epigenetics Laboratory, Columbia University, Mailman School of Public Health, New York, New York
| | - Abdallah Mansour
- Sheba Medical Center, Ramat-Gan and Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Michal Adir
- Sheba Medical Center, Ramat-Gan and Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Ariel Israel
- Department of Family Medicine, Clalit Health Services, Jerusalem, Israel
| | - Catherine Racowsky
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Russ Hauser
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Valentina Bollati
- EPIGET - Epidemiology, Epigenetics and Toxicology Lab, Department of Clinical Sciences and Community Health, University of Milan, Milano, Italy
| | - Ronit Machtinger
- Sheba Medical Center, Ramat-Gan and Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
23
|
Andrade GM, del Collado M, Meirelles FV, da Silveira JC, Perecin F. Intrafollicular barriers and cellular interactions during ovarian follicle development. Anim Reprod 2019; 16:485-496. [PMID: 32435292 PMCID: PMC7234062 DOI: 10.21451/1984-3143-ar2019-0051] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 05/03/2019] [Indexed: 12/20/2022] Open
Abstract
Follicles are composed of different interdependent cell types including oocytes, cumulus, granulosa, and theca cells. Follicular cells and oocytes exchange signaling molecules from the beginning of the development of the primordial follicles until the moment of ovulation. The follicular structure transforms during folliculogenesis; barriers form between the germ and the somatic follicular cells, and between the somatic follicular cells. As such, communication systems need to adapt to maintain the exchange of signaling molecules. Two critical barriers are established at different stages of development: the zona pellucida, separating the oocyte and the cumulus cells limiting the communication through specific connections, and the antrum, separating subpopulations of follicular cells. In both situations, communication is maintained either by the development of specialized connections as transzonal projections or by paracrine signaling and trafficking of extracellular vesicles through the follicular fluid. The bidirectional communication between the oocytes and the follicle cells is vital for driving folliculogenesis and oogenesis. These communication systems are associated with essential functions related to follicular development, oocyte competence, and embryonic quality. Here, we discuss the formation of the zona pellucida and antrum during folliculogenesis, and their importance in follicle and oocyte development. Moreover, this review discusses the current knowledge on the cellular mechanisms such as the movement of molecules via transzonal projections, and the exchange of extracellular vesicles by follicular cells to overcome these barriers to support female gamete development. Finally, we highlight the undiscovered aspects related to intrafollicular communication among the germ and somatic cells, and between the somatic follicular cells and give our perspective on manipulating the above-mentioned cellular communication to improve reproductive technologies.
Collapse
Affiliation(s)
- Gabriella Mamede Andrade
- Faculty of Animal Sciences and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Maite del Collado
- Faculty of Animal Sciences and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Flávio Vieira Meirelles
- Faculty of Animal Sciences and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Juliano Coelho da Silveira
- Faculty of Animal Sciences and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Felipe Perecin
- Faculty of Animal Sciences and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, São Paulo, Brazil.
| |
Collapse
|
24
|
Characterization of mRNA profiles of the exosome-like vesicles in porcine follicular fluid. PLoS One 2019; 14:e0217760. [PMID: 31188849 PMCID: PMC6561635 DOI: 10.1371/journal.pone.0217760] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/18/2019] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles such as exosomes contain several types of transcripts, including mRNAs and micro RNAs (miRNAs), and have emerged as important mediators of cell-to-cell communication. Exosome-like vesicles were identified in the ovarian follicles of several mammalian species. Although the miRNA contents have been extensively characterized, the detailed investigation of their mRNA profiles is lacking. Here, we characterize the mRNA profiles of exosome-like vesicles in ovarian follicles in a pig model. The mRNA contents of the exosome-like vesicles isolated from porcine follicular fluid were analyzed and compared with those from mural granulosa cells (MGCs) using the Illumina HiSeq platform. Bioinformatics studies suggested that the exosomal mRNAs are enriched in those encoding proteins involved in metabolic, phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) -protein kinase B (AKT), and mitogen-activated protein kinase (MAPK) pathways. While the mRNA profile of the exosome-like vesicles resembled that of MGCs, the vesicles contained mRNAs barely detectable in MGCs. Thus, while the majority of the vesicles are likely to be secreted from MGCs, some may originate from other cell types, including theca cells and oocytes, as well as the cells of non-ovarian organs/tissues. Therefore, the mRNA profiles unveiled several novel characteristics of the exosome-like vesicles in ovarian follicles.
Collapse
|
25
|
Reza AMMT, Choi YJ, Han SG, Song H, Park C, Hong K, Kim JH. Roles of microRNAs in mammalian reproduction: from the commitment of germ cells to peri-implantation embryos. Biol Rev Camb Philos Soc 2018; 94:415-438. [PMID: 30151880 PMCID: PMC7379200 DOI: 10.1111/brv.12459] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRNAs) are active regulators of numerous biological and physiological processes including most of the events of mammalian reproduction. Understanding the biological functions of miRNAs in the context of mammalian reproduction will allow a better and comparative understanding of fertility and sterility in male and female mammals. Herein, we summarize recent progress in miRNA‐mediated regulation of mammalian reproduction and highlight the significance of miRNAs in different aspects of mammalian reproduction including the biogenesis of germ cells, the functionality of reproductive organs, and the development of early embryos. Furthermore, we focus on the gene expression regulatory feedback loops involving hormones and miRNA expression to increase our understanding of germ cell commitment and the functioning of reproductive organs. Finally, we discuss the influence of miRNAs on male and female reproductive failure, and provide perspectives for future studies on this topic.
Collapse
Affiliation(s)
- Abu Musa Md Talimur Reza
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Yun-Jung Choi
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Sung Gu Han
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, 05029, Republic of Korea
| | - Hyuk Song
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Chankyu Park
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| |
Collapse
|
26
|
Del Collado M, Andrade GM, Meirelles FV, da Silveira JC, Perecin F. Contributions from the ovarian follicular environment to oocyte function. Anim Reprod 2018; 15:261-270. [PMID: 34178149 PMCID: PMC8202235 DOI: 10.21451/1984-3143-ar2018-0082] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The magnitude of oocyte's role for embryo development is categorical. This unique cell contains the machineries and cellular components necessary to remodel male and female chromatin, to sustain early development and to, ultimately, generate a complete and complex individual. However, to gain these competences before fertilization, the oocyte undergoes several morphological, cellular and molecular changes during its lifetime enclosed in the ovarian follicle. This review will briefly revisit how the oocyte orchestrate the follicular cells, and how molecules transit to the oocyte from the innermost (cumulus) and outermost (antrum and granulosa cells) layers surrounding the follicle-enclosed oocyte. Finally, we will discuss the interferences of in vitro culture conditions in the communication of the oocyte with its surrounding cells and the potential strategies to modulate these communication systems to increase oocyte competence.
Collapse
Affiliation(s)
- Maite Del Collado
- Faculty of Animal Sciences and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, SP, Brazil
| | - Gabriella Mamede Andrade
- Faculty of Animal Sciences and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, SP, Brazil
| | - Flávio Vieira Meirelles
- Faculty of Animal Sciences and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, SP, Brazil
| | - Juliano Coelho da Silveira
- Faculty of Animal Sciences and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, SP, Brazil
| | - Felipe Perecin
- Faculty of Animal Sciences and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, SP, Brazil
| |
Collapse
|
27
|
Da Broi MG, Giorgi VSI, Wang F, Keefe DL, Albertini D, Navarro PA. Influence of follicular fluid and cumulus cells on oocyte quality: clinical implications. J Assist Reprod Genet 2018; 35:735-751. [PMID: 29497954 PMCID: PMC5984887 DOI: 10.1007/s10815-018-1143-3] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 02/19/2018] [Indexed: 01/03/2023] Open
Abstract
An equilibrium needs to be established by the cellular and acellular components of the ovarian follicle if developmental competence is to be acquired by the oocyte. Both cumulus cells (CCs) and follicular fluid (FF) are critical determinants for oocyte quality. Understanding how CCs and FF influence oocyte quality in the presence of deleterious systemic or pelvic conditions may impact clinical decisions in the course of managing infertility. Given that the functional integrities of FF and CCs are susceptible to concurrent pathological conditions, it is important to understand how pathophysiological factors influence natural fertility and the outcomes of pregnancy arising from the use of assisted reproduction technologies (ARTs). Accordingly, this review discusses the roles of CCs and FF in ensuring oocyte competence and present new insights on pathological conditions that may interfere with oocyte quality by altering the intrafollicular environment.
Collapse
Affiliation(s)
- M. G. Da Broi
- Division of Human Reproduction, Department of Gynecology and Obstetrics, Ribeirão Preto School of Medicine, University of São Paulo, Avenida Bandeirantes, 3900, Ribeirao Preto, SP CEP: 14049-900 Brazil
| | - V. S. I. Giorgi
- Division of Human Reproduction, Department of Gynecology and Obstetrics, Ribeirão Preto School of Medicine, University of São Paulo, Avenida Bandeirantes, 3900, Ribeirao Preto, SP CEP: 14049-900 Brazil
| | - F. Wang
- Department of Obstetrics and Gynecology, Laboratory of Reproductive Medicine, NYU School of Medicine, 180 Varick Street, New York, NY 10014 USA
| | - D. L. Keefe
- Department of Obstetrics and Gynecology, Laboratory of Reproductive Medicine, NYU School of Medicine, 180 Varick Street, New York, NY 10014 USA
- Department of Obstetrics and Gynecology, New York University, Langone Medical Center, New York, NY 10016 USA
| | - D. Albertini
- The Center for Human Reproduction, New York, NY USA
| | - P. A. Navarro
- Division of Human Reproduction, Department of Gynecology and Obstetrics, Ribeirão Preto School of Medicine, University of São Paulo, Avenida Bandeirantes, 3900, Ribeirao Preto, SP CEP: 14049-900 Brazil
| |
Collapse
|
28
|
Latifi Z, Fattahi A, Ranjbaran A, Nejabati HR, Imakawa K. Potential roles of metalloproteinases of endometrium-derived exosomes in embryo-maternal crosstalk during implantation. J Cell Physiol 2017; 233:4530-4545. [PMID: 29115666 DOI: 10.1002/jcp.26259] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/09/2017] [Indexed: 12/15/2022]
Abstract
During embryo implantation, crosstalk between the endometrial epithelium and the blastocyst, especially the trophoblasts, is a prerequisite for successful implantation. During this crosstalk, various molecular and functional changes occur to promote synchrony between the embryo and the endometrium as well as the uterine cavity microenvironment. In the past few years, growing evidence has shown that endometrium-derived exosomes play pivotal roles in the embryonic-maternal crosstalk during implantation, although the exact mechanism of this crosstalk has yet to be determined. The presence of metalloproteinases has been reported in endometrium-derived exosomes, implying the importance of these enzymes in exosome-based crosstalk. Thus, in this review, we describe the potential roles of the metalloproteinases of endometrium-derived exosomes in promoting embryo attachment and implantation. This study could provide a better understanding of the potential roles of exosomal metalloproteinases in embryo implantation and pave the way for developing novel exosome-based regulatory agents to support early pregnancy.
Collapse
Affiliation(s)
- Zeinab Latifi
- Animal Resource Science Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Ibaraki, Japan.,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Ranjbaran
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Reza Nejabati
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kazuhiko Imakawa
- Animal Resource Science Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Ibaraki, Japan
| |
Collapse
|
29
|
Lefebvre FA, Lécuyer E. Small Luggage for a Long Journey: Transfer of Vesicle-Enclosed Small RNA in Interspecies Communication. Front Microbiol 2017; 8:377. [PMID: 28360889 PMCID: PMC5352665 DOI: 10.3389/fmicb.2017.00377] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/23/2017] [Indexed: 12/25/2022] Open
Abstract
In the evolutionary arms race, symbionts have evolved means to modulate each other's physiology, oftentimes through the dissemination of biological signals. Beyond small molecules and proteins, recent evidence shows that small RNA molecules are transferred between organisms and transmit functional RNA interference signals across biological species. However, the mechanisms through which specific RNAs involved in cross-species communication are sorted for secretion and protected from degradation in the environment remain largely enigmatic. Over the last decade, extracellular vesicles have emerged as prominent vehicles of biological signals. They can stabilize specific RNA transcripts in biological fluids and selectively deliver them to recipient cells. Here, we review examples of small RNA transfers between plants and bacterial, fungal, and animal symbionts. We also discuss the transmission of RNA interference signals from intestinal cells to populations of the gut microbiota, along with its roles in intestinal homeostasis. We suggest that extracellular vesicles may contribute to inter-species crosstalk mediated by small RNA. We review the mechanisms of RNA sorting to extracellular vesicles and evaluate their relevance in cross-species communication by discussing conservation, stability, stoichiometry, and co-occurrence of vesicles with alternative communication vehicles.
Collapse
Affiliation(s)
- Fabio A. Lefebvre
- Institut de Recherches Cliniques de Montréal (IRCM), RNA Biology DepartmentMontreal, QC, Canada
- Département de Biochimie, Université de MontréalMontreal, QC, Canada
| | - Eric Lécuyer
- Institut de Recherches Cliniques de Montréal (IRCM), RNA Biology DepartmentMontreal, QC, Canada
- Département de Biochimie, Université de MontréalMontreal, QC, Canada
- Divison of Experimental Medicine, McGill UniversityMontreal, QC, Canada
| |
Collapse
|
30
|
Machtinger R, Rodosthenous RS, Adir M, Mansour A, Racowsky C, Baccarelli AA, Hauser R. Extracellular microRNAs in follicular fluid and their potential association with oocyte fertilization and embryo quality: an exploratory study. J Assist Reprod Genet 2017; 34:525-533. [PMID: 28188594 DOI: 10.1007/s10815-017-0876-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 01/18/2017] [Indexed: 12/25/2022] Open
Abstract
PURPOSE The purpose of this study is to determine the profile of extracellular microRNAs (exmiRNAs) in follicular fluid (FF) and explore their association with fertilization potential and embryo quality. METHODS We collected FF from single follicles containing mature oocytes from 40 women undergoing IVF and we screened for the expression of 754 exmiRNAs in FF using the TaqMan OpenArray® qPCR platform. To determine the association of exmiRNAs and IVF outcomes, we compared their expression levels in FF samples that differ by fertilization status (normally, abnormally, and failed to fertilize) and embryo quality (top vs. non-top). RESULTS We detected 207 exmiRNAs, of which miR-30d-5p, miR-320b, miR-10b-3p, miR-1291, and miR-720 were most prevalent. We identified four exmiRNAs with significant fold change (FC) when FF that contained normally fertilized was compared to failed to fertilize oocytes [miR-202-5p (FC = 1.82, p = 0.01), miR-206 (FC = 2.09, p = 0.04), miR-16-1-3p (FC = 1.88, p = 0.05), and miR-1244 (FC = 2.72, p = 0.05)]. We also found four exmiRNAs to be significantly differentially expressed in FF that yielded top quality versus non-top quality embryos [(miR-766-3p (FC = 1.95, p = 0.01), miR-663b (FC = 0.18, p = 0.02), miR-132-3p (FC = 2.45, p = 0.05), and miR-16-5p (FC = 3.80, p = 0.05)]. In-silico analysis revealed that several of these exmiRNAs are involved in pathways implicated in reproductive system diseases, organismal abnormalities, and organ development. CONCLUSIONS Our findings suggest that exmiRNAs in the follicular fluid can lead to downstream events that will affect fertilization and day 3 embryo morphology. We encourage further observational and experimental studies to confirm our findings and to determine the role of exmiRNAs in human reproduction.
Collapse
Affiliation(s)
- Ronit Machtinger
- Department of Obstetrics and Gynecology, Sheba Medical Center, Ramat-Gan and Sackler School of Medicine Tel- Aviv University, Tel Aviv, Israel.
| | | | - Michal Adir
- Department of Obstetrics and Gynecology, Sheba Medical Center, Ramat-Gan and Sackler School of Medicine Tel- Aviv University, Tel Aviv, Israel
| | - Abdallah Mansour
- Department of Obstetrics and Gynecology, Sheba Medical Center, Ramat-Gan and Sackler School of Medicine Tel- Aviv University, Tel Aviv, Israel
| | - Catherine Racowsky
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrea A Baccarelli
- Human Epigenetics Laboratory, Columbia University, Mailman School of Public Health, New York, NY, USA
| | - Russ Hauser
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
31
|
Matsuno Y, Onuma A, Fujioka YA, Yasuhara K, Fujii W, Naito K, Sugiura K. Effects of exosome-like vesicles on cumulus expansion in pigs in vitro. J Reprod Dev 2017; 63:51-58. [PMID: 28163264 PMCID: PMC5320430 DOI: 10.1262/jrd.2016-124] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cell-secreted vesicles, such as exosomes, have recently been recognized as mediators of cell communication. A recent study in cattle showed the involvement of exosome-like vesicles in the control of cumulus expansion, a prerequisite process for normal ovulation; however, whether this is the case in other mammalian species is not known. Therefore, this study aimed to examine the presence of exosome-like vesicles in ovarian follicles and their effects on cumulus expansion in vitro in pigs. The presence of exosome-like vesicles in porcine follicular fluid (pFF) was confirmed by transmission electron microscopic observation, the detection of marker proteins, and RNA profiles specific to exosomes. Fluorescently labeled exosome-like vesicles isolated from pFF were incorporated into both cumulus and mural granulosa cells in vitro. Exosome-like vesicles were not capable of inducing cumulus expansion to a degree comparable to that induced by follicle-stimulating hormone (FSH). Moreover, exosome-like vesicles had no significant effects on the expression levels of transcripts required for the normal expansion process (HAS2, TNFAIP6, and PTGS2). Interestingly, FSH-induced expression of HAS2 and TNFAIP6 mRNA, but not of PTGS2 mRNA, was significantly increased by the presence of exosome-like vesicles; however, the degree of FSH-induced expansion was not affected. In addition, porcine exosome-like vesicles had no significant effects on the expansion of mouse cumulus-oocyte complexes. Collectively, the present results suggest that exosome-like vesicles are present in pFF, but they are not efficient in inducing cumulus expansion in pigs.
Collapse
Affiliation(s)
- Yuta Matsuno
- Laboratory of Applied Genetics, Department of Animal Resource Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | | | | | | | | | | | | |
Collapse
|
32
|
Botigelli RC, Razza EM, Pioltine EM, Nogueira MFG. New approaches regarding the in vitro maturation of oocytes: manipulating cyclic nucleotides and their partners in crime. JBRA Assist Reprod 2017; 21:35-44. [PMID: 28333031 PMCID: PMC5365199 DOI: 10.5935/1518-0557.20170010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Several discoveries have been described recently (5-10 years) about the biology of ovarian follicles (oocyte, cumulus cells and granulosa cells), including new aspects of cellular communication, the control of oocyte maturation and the acquisition of oocyte competence for fertilization and further embryo development. These advances are nourishing assisted reproduction techniques (ART) with new possibilities, in which novel culture systems are being developed and tested to improve embryo yield and quality. This mini-review aims to describe how the recent knowledge on the physiological aspects of mammalian oocyte is reflecting as original or revisited approaches into the context of embryo production. These new insights include recent findings on the mechanisms that control oocyte maturation, especially modulating intraoocyte levels of cyclic nucleotides during in vitro maturation using endogenous or exogenous agents. In this mini-review we also discuss the positive and negative effects of these manipulations on the outcoming embryo.
Collapse
Affiliation(s)
- Ramon Cesar Botigelli
- Department of Pharmacology, Institute of Bioscience, University of São Paulo State, Botucatu, São Paulo, Brazil
| | - Eduardo Montanari Razza
- Department of Pharmacology, Institute of Bioscience, University of São Paulo State, Botucatu, São Paulo, Brazil
| | - Elisa Mariano Pioltine
- Department of Pharmacology, Institute of Bioscience, University of São Paulo State, Botucatu, São Paulo, Brazil
| | - Marcelo Fábio Gouveia Nogueira
- Department of Pharmacology, Institute of Bioscience, University of São Paulo State, Botucatu, São Paulo, Brazil.,Department of Biological Sciences, Faculty of Sciences and Letters, University of São Paulo State, Assis, São Paulo, Brazil
| |
Collapse
|
33
|
Exosome-mediated communication in the ovarian follicle. J Assist Reprod Genet 2016; 33:303-311. [PMID: 26814471 DOI: 10.1007/s10815-016-0657-9] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 01/07/2016] [Indexed: 01/14/2023] Open
Abstract
Cells are able to produce and release different types of vesicles, such as microvesicles and exosomes, in the extracellular microenvironment. According to the scientific community, both microvesicles and exosomes are able to take on and transfer different macromolecules from and to other cells, and in this way, they can influence the recipient cell function. Among the different macromolecule cargos, the most studied are microRNAs. MicroRNAs are a large family of non-coding RNAs involved in the regulation of gene expression. They control every cellular process and their altered regulation is involved in human diseases. Their presence in mammalian follicular fluid has been recently demonstrated, and here, they are enclosed within microvesicles and exosomes or they can also be associated to protein complexes. The presence of microvesicles and exosomes carrying microRNAs in follicular fluid could represent an alternative mechanism of autocrine and paracrine communication inside the ovarian follicle. The outcomes from these studies could be important in basic reproductive research but could also be useful for clinical application. In fact, the characterization of extracellular vesicles in follicular fluid could improve reproductive disease diagnosis and provide biomarkers of oocyte quality in ART (Assisted Reproductive Treatment).
Collapse
|
34
|
Maalouf SW, Liu WS, Pate JL. MicroRNA in ovarian function. Cell Tissue Res 2015; 363:7-18. [PMID: 26558383 DOI: 10.1007/s00441-015-2307-4] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/29/2015] [Indexed: 01/14/2023]
Abstract
The mammalian ovary is a dynamic organ. The coordination of follicle recruitment, selection, and ovulation and the timely development and regression of the corpus luteum are essential for a functional ovary and fertility. Deregulation of any of these processes results in ovarian dysfunction and potential infertility. MicroRNA (miRNA) are short noncoding RNA that regulate developmental processes and time-sensitive functions. The expression of miRNA in the ovary varies with cell type, function, and stage of the estrous cycle. miRNA are involved in the formation of primordial follicles, follicular recruitment and selection, follicular atresia, oocyte-cumulus cell interaction, granulosal cell function, and luteinization. miRNA are differentially expressed in luteal cells at the various stages of the estrous cycle and during maternal recognition of pregnancy, suggesting a role in luteal development, maintenance, and regression. An understanding of the patterns of expression and functions of miRNA in the ovary will lead to novel therapeutics to treat ovarian dysfunction and improve fertility and, potentially, to the development of better contraceptives.
Collapse
Affiliation(s)
- S W Maalouf
- Department of Animal Science, Center for Reproductive Biology and Health, The Pennsylvania State University, 324 Henning Building, University Park, PA 16802, USA
| | - W S Liu
- Department of Animal Science, Center for Reproductive Biology and Health, The Pennsylvania State University, 324 Henning Building, University Park, PA 16802, USA
| | - J L Pate
- Department of Animal Science, Center for Reproductive Biology and Health, The Pennsylvania State University, 324 Henning Building, University Park, PA 16802, USA.
| |
Collapse
|
35
|
Hung WT, Hong X, Christenson LK, McGinnis LK. Extracellular Vesicles from Bovine Follicular Fluid Support Cumulus Expansion. Biol Reprod 2015; 93:117. [PMID: 26423123 DOI: 10.1095/biolreprod.115.132977] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/21/2015] [Indexed: 01/04/2023] Open
Abstract
Expansion of the cumulus complex surrounding the oocyte is critical for ovulation of a fertilizable egg. The ovulation-inducing surge of luteinizing hormone leads to an increased expression of genes such as prostaglandin-endoperoxide synthase 2 (Ptgs2), pentraxin-related protein 3 (Ptx3), and tumor necrosis factor alpha-induced protein 6 (Tnfaip6) that support cumulus expansion. Factors released by mural granulosa and cumulus granulosa cells into the follicular fluid induce paracrine signaling within the follicular compartment. The follicular fluid that separates these distinct granulosa cell types is an enriched fluid containing numerous proteins, nucleic acids, and other macromolecules. Extracellular vesicles (EVs) are also present; however, no physiologically relevant functions of follicular EVs have yet been demonstrated. In our study, the effect of follicular EVs on cumulus-oocyte complex (COC) expansion and relevant gene expression was assayed. Follicular EVs were isolated using ultracentrifugation from follicular fluid of small (3-5 mm) and large (>9 mm) antral bovine follicles, then characterized by nanoparticle tracking analysis, electron microscopy, and Western blot analysis. To test for bioactivity, mouse and bovine COCs were cultured with follicular EVs. Cumulus expansion and Ptgs2, Ptx3, and Tnfaip6 gene expression were measured following COC maturation culture. The results demonstrated that follicular EVs can support both measurable cumulus expansion and increased gene expression.
Collapse
Affiliation(s)
- Wei-Ting Hung
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Xioman Hong
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Lane K Christenson
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Lynda K McGinnis
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas Department of Obstetrics & Gynecology, Division of Reproductive Endocrinology and Infertility, University of Southern California, Los Angeles, California
| |
Collapse
|