1
|
Grudet F, Martinot E, Godin P, Bérubé M, Chédotal A, Boerboom D. Slit1 inhibits ovarian follicle development and female fertility in mice†. Biol Reprod 2024; 111:834-844. [PMID: 38943353 PMCID: PMC11473917 DOI: 10.1093/biolre/ioae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/17/2024] [Accepted: 06/27/2024] [Indexed: 07/01/2024] Open
Abstract
Previous in vitro studies have suggested that SLIT ligands could play roles in regulating ovarian granulosa cell proliferation and gene expression, as well as luteolysis. However, no in vivo study of Slit gene function has been conducted to date. Here, we investigated the potential role of Slit1 in ovarian biology using a Slit1-null mouse model. Female Slit1-null mice were found to produce larger litters than their wild-type counterparts due to increased ovulation rates. Increased ovarian weights in Slit1-null animals were found to be due to the presence of greater numbers of healthy antral follicles with similar numbers of atretic ones, suggesting both an increased rate of follicle recruitment and a decreased rate of atresia. Consistent with this, treatment of cultured granulosa cells with exogenous SLIT1 induced apoptosis in presence or absence of follicle-stimulating hormone, but had no effect on cell proliferation. Although few alterations in the messenger RNA levels of follicle-stimulating hormone-responsive genes were noted in granulosa cells of Slit1-null mice, luteinizing hormone target gene mRNA levels were greatly increased. Finally, increased phospho-AKT levels were found in granulosa cells isolated from Slit1-null mice, and SLIT1 pretreatment of cultured granulosa cells inhibited the ability of both follicle-stimulating hormone and luteinizing hormone to increase AKT phosphorylation, suggesting a mechanism whereby SLIT1 could antagonize gonadotropin signaling. These findings therefore represent the first evidence for a physiological role of a SLIT ligand in the ovary, and define Slit1 as a novel autocrine/paracrine regulator of follicle development.
Collapse
Affiliation(s)
- Florine Grudet
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Emmanuelle Martinot
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Philippe Godin
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Michael Bérubé
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Derek Boerboom
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| |
Collapse
|
2
|
Zhu Z, Huang B, Sun N, Yu X, Du Z, Li A, Huang C. Variations in gut microbiota composition and reproductive hormone levels between laying and broody Muscovy ducks. Poult Sci 2024; 103:104399. [PMID: 39490129 DOI: 10.1016/j.psj.2024.104399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/19/2024] [Accepted: 10/04/2024] [Indexed: 11/05/2024] Open
Abstract
High broodiness in Muscovy ducks impedes animal husbandry growth. The interaction between endocrine hormones and gut microbiota has been proven to play a crucial role in reproductive performance, and whether it can regulate the broody behavior of Muscovy ducks requires further research. Nine laying ducks (Laying group) and nine broody ducks (Broodiness group) were selected. Corresponding serum, ileum, and cecum chyme were collected for further research. The results showed that, compared to the laying group, the serum concentration of prolactin decreased, while the levels of Mullerian inhibiting substance, follicle-stimulating hormone, and follistatin increased in the broodiness group (P < 0.05). 16S rDNA sequencing showed that, the broodiness group exhibited lower abundance levels of Rothia, Streptococcus, and Lactobacillus, whereas the abundance of Turicibacter, Aliicoccus, and Facklamia was higher in the ileum compared to the laying group (P < 0.05). In the cecum, the broodiness group exhibits a significant reduction in the abundance of Butyricicoccus and unclassified_f_Rikenellaceae, while the abundance of Christensenellaceae_R-7_group, Ruminococcus_torques_group, Parabacteroides, norank_f_Oscillospiraceae, Cloacibacillus, Sellimonas, Shuttleworthia, norank_f_UCG-010, unclassified_f_Lachnospiraceae, Oscillospira, Synergistes, Family_XIII_AD3011_group and Eubacterium_nodatum_group is higher compared to the laying group. A Spearman correlation analysis reveals that both in the ileum and cecum, serum hormones exhibit significant correlations with the top 20 abundant intestinal microbial genera. Among these, serum follistatin has most entries of significant correlations with the detected microbial genera (P < 0.05). In conclusion, the broody behavior of Muscovy ducks can be modulated by the interaction between hormones and gut microbiota. Notably, the relationship between Follistatin and the composition of gut microbiota, specifically Firmicutes, is the most prominent.
Collapse
Affiliation(s)
- Zhihao Zhu
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Bingbing Huang
- Jiangxi Functional Feed Additive Engineering Laboratory, Institute of Biological Resource, Jiangxi Academy of Sciences, Nanchang, Jiangxi 330096, China
| | - Ningning Sun
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Xuanci Yu
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Ziyuan Du
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Ang Li
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Caiyun Huang
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| |
Collapse
|
3
|
Shpakov AO. Hormonal and Allosteric Regulation of the Luteinizing Hormone/Chorionic Gonadotropin Receptor. FRONT BIOSCI-LANDMRK 2024; 29:313. [PMID: 39344322 DOI: 10.31083/j.fbl2909313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/20/2024] [Accepted: 07/10/2024] [Indexed: 10/01/2024]
Abstract
Luteinizing hormone (LH) and human chorionic gonadotropin (CG), like follicle-stimulating hormone, are the most important regulators of the reproductive system. They exert their effect on the cell through the LH/CG receptor (LHCGR), which belongs to the family of G protein-coupled receptors. Binding to gonadotropin induces the interaction of LHCGR with various types of heterotrimeric G proteins (Gs, Gq/11, Gi) and β-arrestins, which leads to stimulation (Gs) or inhibition (Gi) of cyclic adenosine monophosphate-dependent cascades, activation of the phospholipase pathway (Gq/11), and also to the formation of signalosomes that mediate the stimulation of mitogen-activated protein kinases (β-arrestins). The efficiency and selectivity of activation of intracellular cascades by different gonadotropins varies, which is due to differences in their interaction with the ligand-binding site of LHCGR. Gonadotropin signaling largely depends on the status of N- and O-glycosylation of LH and CG, on the formation of homo- and heterodimeric receptor complexes, on the cell-specific microenvironment of LHCGR and the presence of autoantibodies to it, and allosteric mechanisms are important in the implementation of these influences, which is due to the multiplicity of allosteric sites in different loci of the LHCGR. The development of low-molecular-weight allosteric regulators of LHCGR with different profiles of pharmacological activity, which can be used in medicine for the correction of reproductive disorders and in assisted reproductive technologies, is promising. These and other issues regarding the hormonal and allosteric regulation of LHCGR are summarized and discussed in this review.
Collapse
Affiliation(s)
- Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| |
Collapse
|
4
|
Duval C, Wyse BA, Tsang BK, Librach CL. Extracellular vesicles and their content in the context of polycystic ovarian syndrome and endometriosis: a review. J Ovarian Res 2024; 17:160. [PMID: 39103867 DOI: 10.1186/s13048-024-01480-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/18/2024] [Indexed: 08/07/2024] Open
Abstract
Extracellular vesicles (EVs), particles enriched in bioactive molecules like proteins, nucleic acids, and lipids, are crucial mediators of intercellular communication and play key roles in various physiological and pathological processes. EVs have been shown to be involved in ovarian follicular function and to be altered in two prevalent gynecological disorders; polycystic ovarian syndrome (PCOS) and endometriosis.Ovarian follicles are complex microenvironments where folliculogenesis takes place with well-orchestrated interactions between granulosa cells, oocytes, and their surrounding stromal cells. Recent research unveiled the presence of EVs, including exosomes and microvesicles, in the follicular fluid (FFEVs), which constitutes part of the developing oocyte's microenvironment. In the context of PCOS, a multifaceted endocrine, reproductive, and metabolic disorder, studies have explored the dysregulation of these FFEVs and their cargo. Nine PCOS studies were included in this review and two miRNAs were commonly reported in two different studies, miR-379 and miR-200, both known to play a role in female reproduction. Studies have also demonstrated the potential use of EVs as diagnostic tools and treatment options.Endometriosis, another prevalent gynecological disorder characterized by ectopic growth of endometrial-like tissue, has also been linked to aberrant EV signaling. EVs in the peritoneal fluid of women with endometriosis carry molecules that modulate the immune response and promote the establishment and maintenance of endometriosis lesions. EVs derived from endometriosis lesions, serum and peritoneal fluid obtained from patients with endometriosis showed no commonly reported biomolecules between the eleven reviewed studies. Importantly, circulating EVs have been shown to be potential biomarkers, also reflecting the severity of the pathology.Understanding the interplay of EVs within human ovarian follicles may provide valuable insights into the pathophysiology of both PCOS and endometriosis. Targeting EV-mediated communication may open avenues for novel diagnostic and therapeutic approaches for these common gynecological disorders. More research is essential to unravel the mechanisms underlying EV involvement in folliculogenesis and its dysregulation in PCOS and endometriosis, ultimately leading to more effective and personalized interventions.
Collapse
Affiliation(s)
- Cyntia Duval
- CReATe Fertility Center, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | | | - Benjamin K Tsang
- Inflammation and Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Departments of Obstetrics and Gynecology & Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Clifford L Librach
- CReATe Fertility Center, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada
- Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
5
|
Elkrewi M, Vicoso B. Single-nucleus atlas of the Artemia female reproductive system suggests germline repression of the Z chromosome. PLoS Genet 2024; 20:e1011376. [PMID: 39213449 PMCID: PMC11392275 DOI: 10.1371/journal.pgen.1011376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 09/12/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Our understanding of the molecular pathways that regulate oogenesis and define cellular identity in the Arthropod female reproductive system and the extent of their conservation is currently very limited. This is due to the focus on model systems, including Drosophila and Daphnia, which do not reflect the observed diversity of morphologies, reproductive modes, and sex chromosome systems. We use single-nucleus RNA and ATAC sequencing to produce a comprehensive single nucleus atlas of the adult Artemia franciscana female reproductive system. We map our data to the Fly Cell Atlas single-nucleus dataset of the Drosophila melanogaster ovary, shedding light on the conserved regulatory programs between the two distantly related Arthropod species. We identify the major cell types known to be present in the Artemia ovary, including germ cells, follicle cells, and ovarian muscle cells. Additionally, we use the germ cells to explore gene regulation and expression of the Z chromosome during meiosis, highlighting its unique regulatory dynamics and allowing us to explore the presence of meiotic sex chromosome silencing in this group.
Collapse
Affiliation(s)
- Marwan Elkrewi
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Beatriz Vicoso
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| |
Collapse
|
6
|
Nazou E, Potiris A, Mavrogianni D, Drakaki E, Vogiatzis AA, Sarli V, Vrantza T, Zikopoulos A, Louis K, Skentou C, Panagopoulos P, Drakakis P, Stavros S. Oocyte Maturation and miRNAs: Studying a Complicate Interaction to Reveal Possible Biomarkers for Female Infertility. Diseases 2024; 12:121. [PMID: 38920553 PMCID: PMC11202488 DOI: 10.3390/diseases12060121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/25/2024] [Accepted: 05/31/2024] [Indexed: 06/27/2024] Open
Abstract
Cellular metabolism, apoptosis, fertilization, and proliferation of granulosa cells belong to a battery of processes where microRNAs can be detected and associated with infertility. The aim of the present review is to focus on mammalian oocyte maturation events and the association between oocyte growth and miRNA expression. PubMed/Medline, Google Scholar and Scopus databases were searched, and 33 studies were included. Regarding the correlation among miRNA expression and the regulation of granulosa cells and cumulus cells, the most important miRNAs were let-7b, let-7c and miR-21. Additionally, the loss of Dicer, an enzyme involved in miRNA biogenesis, is probably a crucial factor in oogenesis, oocyte maturation and embryogenesis. Furthermore, miRNAs interfere with different cellular mechanisms like apoptosis, steroidogenesis, genome integrity, angiogenesis, antioxidative response and, consequently, oocyte maturation. Hence, it is of major importance to clarify the role and mechanism of each miRNA as understanding its action may develop new tools and establish new diagnostic and treatment approaches for infertility and ovarian disorders.
Collapse
Affiliation(s)
- Eleni Nazou
- Third Department of Obstetrics and Gynecology, University General Hospital “ATTIKON”, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (E.N.); (A.-A.V.); (V.S.); (T.V.); (A.Z.); (K.L.); (P.P.); (P.D.); (S.S.)
| | - Anastasios Potiris
- Third Department of Obstetrics and Gynecology, University General Hospital “ATTIKON”, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (E.N.); (A.-A.V.); (V.S.); (T.V.); (A.Z.); (K.L.); (P.P.); (P.D.); (S.S.)
| | - Despoina Mavrogianni
- First Department of Obstetrics and Gynecology, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 115 28 Athens, Greece; (D.M.); (E.D.)
| | - Eirini Drakaki
- First Department of Obstetrics and Gynecology, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 115 28 Athens, Greece; (D.M.); (E.D.)
| | - Aris-Anargyros Vogiatzis
- Third Department of Obstetrics and Gynecology, University General Hospital “ATTIKON”, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (E.N.); (A.-A.V.); (V.S.); (T.V.); (A.Z.); (K.L.); (P.P.); (P.D.); (S.S.)
| | - Vaia Sarli
- Third Department of Obstetrics and Gynecology, University General Hospital “ATTIKON”, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (E.N.); (A.-A.V.); (V.S.); (T.V.); (A.Z.); (K.L.); (P.P.); (P.D.); (S.S.)
| | - Tereza Vrantza
- Third Department of Obstetrics and Gynecology, University General Hospital “ATTIKON”, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (E.N.); (A.-A.V.); (V.S.); (T.V.); (A.Z.); (K.L.); (P.P.); (P.D.); (S.S.)
| | - Athanasios Zikopoulos
- Third Department of Obstetrics and Gynecology, University General Hospital “ATTIKON”, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (E.N.); (A.-A.V.); (V.S.); (T.V.); (A.Z.); (K.L.); (P.P.); (P.D.); (S.S.)
| | - Konstantinos Louis
- Third Department of Obstetrics and Gynecology, University General Hospital “ATTIKON”, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (E.N.); (A.-A.V.); (V.S.); (T.V.); (A.Z.); (K.L.); (P.P.); (P.D.); (S.S.)
| | - Chara Skentou
- Department of Obstetrics and Gynecology, Medical School, University of Ioannina, 45110 Ioannina, Greece;
| | - Periklis Panagopoulos
- Third Department of Obstetrics and Gynecology, University General Hospital “ATTIKON”, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (E.N.); (A.-A.V.); (V.S.); (T.V.); (A.Z.); (K.L.); (P.P.); (P.D.); (S.S.)
| | - Peter Drakakis
- Third Department of Obstetrics and Gynecology, University General Hospital “ATTIKON”, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (E.N.); (A.-A.V.); (V.S.); (T.V.); (A.Z.); (K.L.); (P.P.); (P.D.); (S.S.)
| | - Sofoklis Stavros
- Third Department of Obstetrics and Gynecology, University General Hospital “ATTIKON”, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (E.N.); (A.-A.V.); (V.S.); (T.V.); (A.Z.); (K.L.); (P.P.); (P.D.); (S.S.)
| |
Collapse
|
7
|
Kunitomi C, Romero M, Daldello EM, Schindler K, Conti M. Multiple intersecting pathways are involved in CPEB1 phosphorylation and regulation of translation during mouse oocyte meiosis. Development 2024; 151:dev202712. [PMID: 38785133 PMCID: PMC11190569 DOI: 10.1242/dev.202712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024]
Abstract
The RNA-binding protein cytoplasmic polyadenylation element binding 1 (CPEB1) plays a fundamental role in regulating mRNA translation in oocytes. However, the specifics of how and which protein kinase cascades modulate CPEB1 activity are still controversial. Using genetic and pharmacological tools, and detailed time courses, we have re-evaluated the relationship between CPEB1 phosphorylation and translation activation during mouse oocyte maturation. We show that both the CDK1/MAPK and AURKA/PLK1 pathways converge on CPEB1 phosphorylation during prometaphase of meiosis I. Only inactivation of the CDK1/MAPK pathway disrupts translation, whereas inactivation of either pathway alone leads to CPEB1 stabilization. However, CPEB1 stabilization induced by inactivation of the AURKA/PLK1 pathway does not affect translation, indicating that destabilization and/or degradation is not linked to translational activation. The accumulation of endogenous CCNB1 protein closely recapitulates the translation data that use an exogenous template. These findings support the overarching hypothesis that the activation of translation during prometaphase in mouse oocytes relies on a CDK1/MAPK-dependent CPEB1 phosphorylation, and that translational activation precedes CPEB1 destabilization.
Collapse
Affiliation(s)
- Chisato Kunitomi
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | - Mayra Romero
- Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Human Genetics Institute of New Jersey, Piscataway, NJ 08854, USA
| | - Enrico Maria Daldello
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine, LBD - IBPS, F-75005 Paris, France
| | - Karen Schindler
- Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Human Genetics Institute of New Jersey, Piscataway, NJ 08854, USA
| | - Marco Conti
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
8
|
Tu S, Yu G, Ge F, Xu R, Jin Z, Xie X, Zhu D. Comparative transcriptomic characterization of the ovary in the spawning process of the mud crab Scylla paramamosain. Dev Growth Differ 2024; 66:274-284. [PMID: 38501505 DOI: 10.1111/dgd.12921] [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: 06/18/2023] [Revised: 02/11/2024] [Accepted: 02/28/2024] [Indexed: 03/20/2024]
Abstract
Oviposition is induced upon mating in most insects. Spawning is a physiological process that is fundamental for the reproduction of Scylla paramamosain. However, the molecular mechanisms underlying the spawning process in this species are poorly understood. Herein, comprehensive ovary transcriptomic analysis was conducted at the germinal vesicle breakdown stage (GVBD), spawning stage, 0.5 h post-spawning stage, and 24 h post-spawning stage of S. paramamosain for gene discovery. A total of 67,230 unigenes were generated, and 27,975 (41.61%) unigenes were annotated. Meanwhile, the differentially expressed genes (DEGs) between the different groups were identified, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis was subsequently conducted. These results suggested that octopamine (OA) and tyramine (TA) could induce oviposition, while dopamine (DA) and serotonin (5-hydroxytryptamine [5-HT]) inhibit oviposition. The 20-hydroxyecdysone (20E) and methyl farnesoate (MF) signal pathways might be positively associated with oviposition. Furthermore, numerous transcripts that encode neuropeptides and their G-protein-coupled receptors (GPCRs), such as CNMamide, RYamide, ecdysis-triggering hormone (ETH), GPA2/GPB5 receptor, and Moody receptor, appear to be differentially expressed during the spawning process. Eleven unigenes were selected for qRT-PCR and the pattern was found to be consistent with the transcriptome expression pattern. Our work is the first spawning-related investigation of S. paramamosain focusing on the ovary at the whole transcriptome level. These findings assist in improving our understanding of spawning regulation in S. paramamosain and provide information for oviposition studies in other crustaceans.
Collapse
Affiliation(s)
- Shisheng Tu
- School of Marine Science, Ningbo University, Ningbo, China
| | - Guohong Yu
- School of Marine Science, Ningbo University, Ningbo, China
| | - Fuqiang Ge
- School of Marine Science, Ningbo University, Ningbo, China
| | - Rui Xu
- School of Marine Science, Ningbo University, Ningbo, China
| | - Zhongwen Jin
- Ningbo Ocean and Fisheries Research Institute of Zhejiang Province, Ningbo, China
| | - Xi Xie
- School of Marine Science, Ningbo University, Ningbo, China
| | - Dongfa Zhu
- School of Marine Science, Ningbo University, Ningbo, China
| |
Collapse
|
9
|
Zhu Z, He M, Zhang T, Zhao T, Qin S, Gao M, Wang W, Zheng W, Chen Z, Liu L, Hao M, Zhou B, Zhang H, Wang J, Wang F, Xia G, Wang C. LSD1 promotes the FSH responsive follicle formation by regulating autophagy and repressing Wt1 in the granulosa cells. Sci Bull (Beijing) 2024; 69:1122-1136. [PMID: 38302330 DOI: 10.1016/j.scib.2024.01.015] [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: 08/08/2023] [Revised: 12/08/2023] [Accepted: 01/05/2024] [Indexed: 02/03/2024]
Abstract
In a growing follicle, the survival and maturation of the oocyte largely depend on support from somatic cells to facilitate FSH-induced mutual signaling and chemical communication. Although apoptosis and autophagy in somatic cells are involved in the process of FSH-induced follicular development, the underlying mechanisms require substantial study. According to our study, along with FSH-induced antral follicles (AFs) formation, both lysine-specific demethylase 1 (LSD1) protein levels and autophagy increased simultaneously in granulosa cells (GCs) in a time-dependent manner, we therefore evaluated the importance of LSD1 upon facilitating the formation of AFs correlated to autophagy in GCs. Conditional knockout of Lsd1 in GCs resulted in significantly decreased AF number and subfertility in females, accompanied by marked suppression of the autophagy in GCs. On the one hand, depletion of Lsd1 resulted in accumulation of Wilms tumor 1 homolog (WT1), at both the protein and mRNA levels. WT1 prevented the expression of FSH receptor (Fshr) in GCs and thus reduced the responsiveness of the secondary follicles to FSH induction. On the other hand, depletion of LSD1 resulted in suppressed level of autophagy by upregulation of ATG16L2 in GCs. We finally approved that LSD1 contributed to these sequential activities in GCs through its H3K4me2 demethylase activity. Therefore, the importance of LSD1 in GCs is attributable to its roles in both accelerating autophagy and suppressing WT1 expression to ensure the responsiveness of GCs to FSH during AFs formation.
Collapse
Affiliation(s)
- Zijian Zhu
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Meina He
- College of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
| | - Tuo Zhang
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
| | - Ting Zhao
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Shaogang Qin
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Meng Gao
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Wenji Wang
- School of Life Sciences, Taizhou University, Taizhou 318000, China
| | - Wenying Zheng
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Ziqi Chen
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Longping Liu
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ming Hao
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Bo Zhou
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Hua Zhang
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jianbin Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Fengchao Wang
- Transgenic Animal Center, National Institute of Biological Sciences, Beijing 102206, China.
| | - Guoliang Xia
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan 750021, China.
| | - Chao Wang
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
10
|
Chen SY, Wang TE, Lee WY, Yang YY, Lai HC, Matsuda F, Kosek H, Chen YT, Li SH, Tsai PS. Cre-LoxP and tamoxifen-induced deletion of ovarian quiescin sulfhydryl oxidase 2 showed disruption of ovulatory activity in mice. J Ovarian Res 2024; 17:66. [PMID: 38504307 PMCID: PMC10949576 DOI: 10.1186/s13048-024-01388-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 03/08/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Quiescin sulfhydryl oxidase 2 (QSOX2) is a flavin adenine dinucleotide-dependent sulfhydryl oxidase that is known to be involved in protein folding, cell growth regulation, and redox state modification through oxidative activities. Earlier studies demonstrated the tissue and cellular localization of QSOX2 in the male reproductive tract, as well as the highly-regulated mechanism of QSOX2 protein synthesis and expression through the coordinated action of testosterone and epididymal-enriched amino acid, glutamate. However, the presence and the functions of QSOX2 in female reproduction are unknown. In this study, we applied the Cre-loxP gene manipulation system to generate the heterozygous and homozygous Qsox2 knockout mice and examined its effects on ovarian function. RESULTS We demonstrated that QSOX2 was detected in the follicle-supporting cells (granulosa and cumulus cells) of ovarian follicles of all stages but was absent in the corpus luteum, suggesting its supportive role in folliculogenesis. In comparison with reproductive organogenesis in wild-type mice, there was no difference in testicular and epididymal structure in male Qsox2 knockout; however, Qsox2 knockout disrupted the regular ovulation process in female mice as a drastic decrease in the formation of the corpus luteum was detected, and no pregnancy was achieved when mating males with homozygous Qsox2 knockout females. RNAseq analyses further revealed that Qsox2 knockout altered critical signaling pathways and genes that are responsible for maintaining ovarian functions. CONCLUSION Our data demonstrated for the first time that Qsox2 is critical for ovarian function in mice.
Collapse
Affiliation(s)
- Shih-Yun Chen
- Department of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | - Tse-En Wang
- Graduate Institute of Veterinary Medicine, National Taiwan University, 10617, Taipei, Taiwan
| | - Wei-Yun Lee
- Graduate Institute of Veterinary Medicine, National Taiwan University, 10617, Taipei, Taiwan
| | - Ya-Yi Yang
- Graduate Institute of Veterinary Medicine, National Taiwan University, 10617, Taipei, Taiwan
| | - Hong-Chun Lai
- Department of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | - Fuko Matsuda
- Department of Veterinary Medical Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Haruhiko Kosek
- Center for Integrative Medical Sciences (IMS), RIEKN, Yokohama, Kanagawa, 230-0045, Japan
| | - You-Tzung Chen
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, 10617, Taipei, Taiwan
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, 10055, Taipei, Taiwan
| | - Sheng-Hsiang Li
- Department of Medical Research, MacKay Memorial Hospital, 25160, Tamsui, Taiwan
| | - Pei-Shiue Tsai
- Department of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan.
- Graduate Institute of Veterinary Medicine, National Taiwan University, 10617, Taipei, Taiwan.
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, 10617, Taipei, Taiwan.
| |
Collapse
|
11
|
Conti M, Kunitomi C. A genome-wide perspective of the maternal mRNA translation program during oocyte development. Semin Cell Dev Biol 2024; 154:88-98. [PMID: 36894378 PMCID: PMC11250054 DOI: 10.1016/j.semcdb.2023.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 02/01/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
Transcriptional and post-transcriptional regulations control gene expression in most cells. However, critical transitions during the development of the female gamete relies exclusively on regulation of mRNA translation in the absence of de novo mRNA synthesis. Specific temporal patterns of maternal mRNA translation are essential for the oocyte progression through meiosis, for generation of a haploid gamete ready for fertilization and for embryo development. In this review, we will discuss how mRNAs are translated during oocyte growth and maturation using mostly a genome-wide perspective. This broad view on how translation is regulated reveals multiple divergent translational control mechanisms required to coordinate protein synthesis with progression through the meiotic cell cycle and with development of a totipotent zygote.
Collapse
Affiliation(s)
- Marco Conti
- Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, and Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA.
| | - Chisato Kunitomi
- Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, and Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
12
|
Sharma A, Dubey PK, Kumar P, Tiwari KN, Tripathi A. Identification and molecular characterization of genes modulating progression of an oocyte from M-I to M-II in rat ovary. Am J Reprod Immunol 2024; 91:e13825. [PMID: 38389407 DOI: 10.1111/aji.13825] [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: 10/04/2023] [Revised: 01/02/2024] [Accepted: 02/05/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND To achieve oocyte competence for successful fertilization, bidirectional communication between oocyte and granulosa cells is crucial. The acquisition of meiotic competency in oocyte is facilitated by various regulatory genes however, expression pattern of these genes is not well documented during meiotic transition from Metaphase-I to Metaphase-II stage. Therefore, the present research analyzed the expression pattern of regulatory genes that are involved in the transition from M-I to M-II stages in rat oocyte. METHODS The analysis of the data was conducted by applying an array of bioinformatic tools. The investigation of gene group interactions was carried out by employing the STRING database, which relies on co-expression information. The gene ontology (GO) analysis was performed utilizing the comparative GO database. Functional annotation for GO and pathway enrichment analysis were performed for genes involved in networking. The GO obtained through computational simulations was subsequently validated using quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis. RESULTS The findings of our study suggest that there is a distinct gene expression pattern in both the oocyte and granulosa cells. This pattern indicates that oocyte-secreted factors, such as BMP15 and GDF9, play a crucial role in regulating the progression of the meiotic cell cycle from the M-I to M-II stages. We have also examined the level of mRNA expression of genes including CYP11A1, CYP19A1, and STAR, which are crucial for the steroidogenesis. CONCLUSIONS It is fascinating to observe that the oscillatory pattern of specific key genes may hold significance in the process of in vitro oocyte maturation, specifically during the transition from the M-I to M-II stage. It might be useful for determining biomarker genes and potential pathways that play a role in attaining oocyte competency, thereby aiding in the assessment of oocyte quality for the purpose of achieving successful fertilization.
Collapse
Affiliation(s)
- Alka Sharma
- Zoology Section, MMV, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Pawan K Dubey
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Pradeep Kumar
- Department of Botany, MMV, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Kavindra Nath Tiwari
- Department of Botany, MMV, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Anima Tripathi
- Zoology Section, MMV, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
13
|
Kunitomi C, Romero M, Daldello EM, Schindler K, Conti M. Multiple intersecting pathways are involved in the phosphorylation of CPEB1 to activate translation during mouse oocyte meiosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.17.575938. [PMID: 38293116 PMCID: PMC10827138 DOI: 10.1101/2024.01.17.575938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The RNA-binding protein cytoplasmic polyadenylation element binding 1 (CPEB1) plays a fundamental role in the regulation of mRNA translation in oocytes. However, the nature of protein kinase cascades modulating the activity of CPEB1 is still a matter of controversy. Using genetic and pharmacological tools and detailed time courses, here we have reevaluated the relationship between CPEB1 phosphorylation and the activation of translation during mouse oocyte maturation. We show that both the CDK1/MAPK and AURKA/PLK1 pathways converge on the phosphorylation of CPEB1 during prometaphase. Only inactivation of the CDK1/MAPK pathway disrupts translation, while inactivation of either pathway leads to CPEB1 stabilization. However, stabilization of CPEB1 induced by inactivation of the AURKA/PLK1 does not affect translation, indicating that destabilization/degradation can be dissociated from translational activation. The accumulation of the endogenous CCNB1 protein closely recapitulates the translation data. These findings support the overarching hypothesis that the activation of translation in prometaphase in mouse oocytes relies on a CDK1-dependent CPEB1 phosphorylation, and this translational activation precedes CPEB1 destabilization.
Collapse
Affiliation(s)
- Chisato Kunitomi
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | - Mayra Romero
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Human Genetics Institute of New Jersey
| | - Enrico Maria Daldello
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine, LBD - IBPS, F-75005 Paris, France
| | - Karen Schindler
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Human Genetics Institute of New Jersey
| | - Marco Conti
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
14
|
Berg C, Sieber M, Sun J. Finishing the egg. Genetics 2024; 226:iyad183. [PMID: 38000906 PMCID: PMC10763546 DOI: 10.1093/genetics/iyad183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/27/2023] [Indexed: 11/26/2023] Open
Abstract
Gamete development is a fundamental process that is highly conserved from early eukaryotes to mammals. As germ cells develop, they must coordinate a dynamic series of cellular processes that support growth, cell specification, patterning, the loading of maternal factors (RNAs, proteins, and nutrients), differentiation of structures to enable fertilization and ensure embryonic survival, and other processes that make a functional oocyte. To achieve these goals, germ cells integrate a complex milieu of environmental and developmental signals to produce fertilizable eggs. Over the past 50 years, Drosophila oogenesis has risen to the forefront as a system to interrogate the sophisticated mechanisms that drive oocyte development. Studies in Drosophila have defined mechanisms in germ cells that control meiosis, protect genome integrity, facilitate mRNA trafficking, and support the maternal loading of nutrients. Work in this system has provided key insights into the mechanisms that establish egg chamber polarity and patterning as well as the mechanisms that drive ovulation and egg activation. Using the power of Drosophila genetics, the field has begun to define the molecular mechanisms that coordinate environmental stresses and nutrient availability with oocyte development. Importantly, the majority of these reproductive mechanisms are highly conserved throughout evolution, and many play critical roles in the development of somatic tissues as well. In this chapter, we summarize the recent progress in several key areas that impact egg chamber development and ovulation. First, we discuss the mechanisms that drive nutrient storage and trafficking during oocyte maturation and vitellogenesis. Second, we examine the processes that regulate follicle cell patterning and how that patterning impacts the construction of the egg shell and the establishment of embryonic polarity. Finally, we examine regulatory factors that control ovulation, egg activation, and successful fertilization.
Collapse
Affiliation(s)
- Celeste Berg
- Department of Genome Sciences, University of Washington, Seattle, WA 98195-5065 USA
| | - Matthew Sieber
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | - Jianjun Sun
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269 USA
| |
Collapse
|
15
|
Gilchrist RB, Ho TM, De Vos M, Sanchez F, Romero S, Ledger WL, Anckaert E, Vuong LN, Smitz J. A fresh start for IVM: capacitating the oocyte for development using pre-IVM. Hum Reprod Update 2024; 30:3-25. [PMID: 37639630 DOI: 10.1093/humupd/dmad023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/08/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND While oocyte IVM is practiced sporadically it has not achieved widespread clinical practice globally. However, recently there have been some seminal advances in our understanding of basic aspects of oocyte biology and ovulation from animal studies that have led to novel approaches to IVM. A significant recent advance in IVM technology is the use of biphasic IVM approaches. These involve the collection of immature oocytes from small antral follicles from minimally stimulated patients/animals (without hCG-priming) and an ∼24 h pre-culture of oocytes in an advanced culture system ('pre-IVM') prior to IVM, followed by routine IVF procedures. If safe and efficacious, this novel procedure may stand to make a significant impact on human ART practices. OBJECTIVE AND RATIONALE The objectives of this review are to examine the major scientific advances in ovarian biology with a unique focus on the development of pre-IVM methodologies, to provide an insight into biphasic IVM procedures, and to report on outcomes from animal and clinical human data, including safety data. The potential future impact of biphasic IVM on ART practice is discussed. SEARCH METHODS Peer review original and review articles were selected from PubMed and Web of Science searches for this narrative review. Searches were performed using the following keywords: oocyte IVM, pre-IVM, biphasic IVM, CAPA-IVM, hCG-triggered/primed IVM, natural cycle IVF/M, ex-vivo IVM, OTO-IVM, oocyte maturation, meiotic competence, oocyte developmental competence, oocyte capacitation, follicle size, cumulus cell (CC), granulosa cell, COC, gap-junction communication, trans-zonal process, cAMP and IVM, cGMP and IVM, CNP and IVM, EGF-like peptide and IVM, minimal stimulation ART, PCOS. OUTCOMES Minimizing gonadotrophin use means IVM oocytes will be collected from small antral (pre-dominant) follicles containing oocytes that are still developing. Standard IVM yields suboptimal clinical outcomes using such oocytes, whereas pre-IVM aims to continue the oocyte's development ex vivo, prior to IVM. Pre-IVM achieves this by eliciting profound cellular changes in the oocyte's CCs, which continue to meet the oocyte's developmental needs during the pre-IVM phase. The literature contains 25 years of animal research on various pre-IVM and biphasic IVM procedures, which serves as a large knowledge base for new approaches to human IVM. A pre-IVM procedure based on c-type natriuretic peptide (named 'capacitation-IVM' (CAPA-IVM)) has undergone pre-clinical human safety and efficacy trials and its adoption into clinical practice resulted in healthy live birth rates not different from conventional IVF. WIDER IMPLICATIONS Over many decades, improvements in clinical IVM have been gradual and incremental but there has likely been a turning of the tide in the past few years, with landmark discoveries in animal oocyte biology finally making their way into clinical practice leading to improved outcomes for patients. Demonstration of favorable clinical results with CAPA-IVM, as the first clinically tested biphasic IVM system, has led to renewed interest in IVM as an alternative, low-intervention, low-cost, safe, patient-friendly ART approach, and especially for patients with PCOS. The same new approach is being used as part of fertility preservation in patients with cancer and holds promise for social oocyte freezing.
Collapse
Affiliation(s)
- Robert B Gilchrist
- Fertility & Research Centre, Discipline of Women's Health, School of Clinical Medicine, University of New South Wales Sydney, NSW, Australia
| | - Tuong M Ho
- IVFMD, My Duc Hospital, Ho Chi Minh City, Vietnam
| | - Michel De Vos
- Brussels IVF, UZ Brussel, Brussels, Belgium
- Follicle Biology Laboratory, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Flor Sanchez
- Centro de Estudios e Investigaciones en Biología y Medicina Reproductiva, Lima, Peru
| | - Sergio Romero
- Laboratory of Reproductive Biology and Fertility Preservation, Cayetano Heredia University (UPCH), Lima, Peru
- Centro de Fertilidad y Reproducción Asistida, Lima, Peru
| | - William L Ledger
- Fertility & Research Centre, Discipline of Women's Health, School of Clinical Medicine, University of New South Wales Sydney, NSW, Australia
- City Fertility, Global CHA IVF Partners, Sydney, NSW, Australia
| | - Ellen Anckaert
- Follicle Biology Laboratory, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lan N Vuong
- Department of Obstetrics and Gynaecology, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Johan Smitz
- Follicle Biology Laboratory, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
16
|
Dong R, Abazarikia A, Luan Y, Yu SY, Kim SY. Molecular Mechanisms Determining Mammalian Oocyte Quality with the Treatment of Cancer Therapy. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2024; 238:97-119. [PMID: 39030356 DOI: 10.1007/978-3-031-55163-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Cancer is a global public health issue and remains one of the leading causes of death in the United States (Siegel et al. CA Cancer J Clin. 72:7-33, 2022). It is estimated in the US in 2022, about 935,000 new cases of cancer will be diagnosed in women, and the probability of developing invasive cancer is 5.8% for females younger than 50 years old (Siegel et al. CA Cancer J Clin. 72:7-33, 2022). However, advances in screening programs, diagnostic methods, and therapeutic options have greatly increased the five-year survival rate in reproductive-age women with a variety of cancers. Given the clinical consequences of gonadotoxic cancer therapies, young, female cancer survivors may face compromised fertility, premature ovarian insufficiency, early-onset menopause, and endocrine dysregulation (Bedoschi et al. Future Oncol. 12:2333-44, 2016). Gonadotoxic side effects may include decreased oocyte quality within surviving follicles, loss of ovarian follicles, and impaired ovarian function. In reproductive-age women, oocyte quality is an important element for successful clinical pregnancies and healthy offspring as poor-quality oocytes may be a cause of infertility (McClam et al. Biol Reprod. 106:328-37, 2022; Marteil et al. Reprod Biol. 9:203-24, 2009; Krisher. J Anim Sci. 82: E14-E23, 2004). Thus, it is critical to determine the quantity and quality of surviving follicles in the ovary after cancer treatment and to assess oocyte quality within those surviving follicles as these are markers for determining the capacity for ovarian function restoration and future fertility, especially for young cancer survivors (Xu et al. Nat Med. 17:1562-3, 2011). The long-term effects of cancer therapeutics on oocyte quality are influenced by factors including, but not limited to, individual patient characteristics (e.g. age, health history, comorbidities, etc.), disease type, or treatment regimen (Marci et al. Reprod Biol Endocrinol. 16:1-112, 2018). These effects may translate clinically into an impaired production of viable oocytes and compromised fertility (Garutti et al. ESMO Open. 6:100276, 2021).
Collapse
Affiliation(s)
- Rosemary Dong
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- , Omaha, USA
| | - Amirhossein Abazarikia
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- , Omaha, USA
| | - Yi Luan
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- , Omaha, USA
| | - Seok-Yeong Yu
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- , Omaha, USA
| | - So-Youn Kim
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
- , Omaha, USA.
| |
Collapse
|
17
|
Uju CN, Unniappan S. Growth factors and female reproduction in vertebrates. Mol Cell Endocrinol 2024; 579:112091. [PMID: 37863469 DOI: 10.1016/j.mce.2023.112091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/05/2023] [Accepted: 10/13/2023] [Indexed: 10/22/2023]
Abstract
Female reproductive efficiency is influenced by the outcomes of various processes, including folliculogenesis, apoptosis, response to gonadotropin signaling, oocyte maturation, and ovulation. The role of hormones in regulating these processes and other reproductive activities has been well established. It is becoming increasingly evident that in addition to well-characterized hormones, growth factors play vital roles in regulating some of these reproductive activities. Growth factors and their receptors are widely distributed in vertebrate ovaries at different stages of ovarian development, indicating their involvement in intraovarian reproductive functions. In the ovary, cell surface receptors allow growth factors to regulate intraovarian reproductive activities. Understanding these actions in the reproductive axis would provide a tool to target growth factors and/or their receptors to yield desirable reproductive outcomes. These include enrichment of in vitro maturation and fertilization culture media, and management of infertility. This review discusses some widely characterized growth factors belonging to the TGF, EGF, IGF, FGF, and BDNF family of peptides and their role in female reproduction in vertebrates, with a focus on mammals.
Collapse
Affiliation(s)
- Chinelo N Uju
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5B4, Canada
| | - Suraj Unniappan
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5B4, Canada.
| |
Collapse
|
18
|
McKinley E, Speckhart SL, Keane JA, Oliver MA, Rhoads ML, Edwards JL, Biase FH, Ealy AD. Influences of Supplementing Selective Members of the Interleukin-6 Cytokine Family on Bovine Oocyte Competency. Animals (Basel) 2023; 14:44. [PMID: 38200775 PMCID: PMC10778514 DOI: 10.3390/ani14010044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/11/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
This work explored whether supplementing selective members of the interleukin-6 (IL6) cytokine family during in vitro bovine oocyte maturation affects maturation success, cumulus-oocyte complex (COC) gene expression, fertilization success, and embryo development potential. Human recombinant proteins for IL6, IL11, and leukemia inhibitory factor (LIF) were supplemented to COCs during the maturation period, then fertilization and embryo culture commenced without further cytokine supplementation. The first study determined that none of these cytokines influenced the rate that oocytes achieved arrest at meiosis II. The second study identified that LIF and IL11 supplementation increases AREG transcript abundance. Supplementation with IL6 supplementation did not affect AREG abundance but reduced HAS2 transcript abundance. Several other transcriptional markers of oocyte competency were not affected by any of the cytokines. The third study determined that supplementing these cytokines during maturation did not influence fertilization success, but either LIF or IL11 supplementation increased blastocyst development. No effect of IL6 supplementation on subsequent blastocyst development was detected. The fourth experiment explored whether each cytokine treatment affects the post-thaw survivability of cryopreserved IVP blastocysts. None of the cytokines supplemented during oocyte maturation produced any positive effects on post-thaw blastocyst re-expansion and hatching. In conclusion, these outcomes implicate IL11 and LIF as potentially useful supplements for improving bovine oocyte competency.
Collapse
Affiliation(s)
- Endya McKinley
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (E.M.); (S.L.S.); (J.A.K.); (M.A.O.); (M.L.R.); (F.H.B.)
| | - Savannah L. Speckhart
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (E.M.); (S.L.S.); (J.A.K.); (M.A.O.); (M.L.R.); (F.H.B.)
| | - Jessica A. Keane
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (E.M.); (S.L.S.); (J.A.K.); (M.A.O.); (M.L.R.); (F.H.B.)
| | - Mary A. Oliver
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (E.M.); (S.L.S.); (J.A.K.); (M.A.O.); (M.L.R.); (F.H.B.)
| | - Michelle L. Rhoads
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (E.M.); (S.L.S.); (J.A.K.); (M.A.O.); (M.L.R.); (F.H.B.)
| | - J. Lannett Edwards
- Department of Animal Science, The University of Tennessee, Knoxville, TN 37996, USA;
| | - Fernando H. Biase
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (E.M.); (S.L.S.); (J.A.K.); (M.A.O.); (M.L.R.); (F.H.B.)
| | - Alan D. Ealy
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (E.M.); (S.L.S.); (J.A.K.); (M.A.O.); (M.L.R.); (F.H.B.)
| |
Collapse
|
19
|
Xu D, Song S, Wang F, Li Y, Li Z, Yao H, Zhao Y, Zhao Z. Single-cell transcriptomic atlas of goat ovarian aging. J Anim Sci Biotechnol 2023; 14:151. [PMID: 38053167 DOI: 10.1186/s40104-023-00948-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/10/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND The ovaries are one of the first organs that undergo degenerative changes earlier in the aging process, and ovarian aging is shown by a decrease in the number and quality of oocytes. However, little is known about the molecular mechanisms of female age-related fertility decline in different types of ovarian cells during aging, especially in goats. Therefore, the aim of this study was to reveal the mechanisms driving ovarian aging in goats at single-cell resolution. RESULTS For the first time, we surveyed the single-cell transcriptomic landscape of over 27,000 ovarian cells from newborn, young and aging goats, and identified nine ovarian cell types with distinct gene-expression signatures. Functional enrichment analysis showed that ovarian cell types were involved in their own unique biological processes, such as Wnt beta-catenin signalling was enriched in germ cells, whereas ovarian steroidogenesis was enriched in granulosa cells (GCs). Further analysis showed that ovarian aging was linked to GCs-specific changes in the antioxidant system, oxidative phosphorylation, and apoptosis. Subsequently, we identified a series of dynamic genes, such as AMH, CRABP2, THBS1 and TIMP1, which determined the fate of GCs. Additionally, FOXO1, SOX4, and HIF1A were identified as significant regulons that instructed the differentiation of GCs in a distinct manner during ovarian aging. CONCLUSIONS This study revealed a comprehensive aging-associated transcriptomic atlas characterizing the cell type-specific mechanisms during ovarian aging at the single-cell level and offers new diagnostic biomarkers and potential therapeutic targets for age-related goat ovarian diseases.
Collapse
Affiliation(s)
- Dejun Xu
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Shuaifei Song
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Fuguo Wang
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Yawen Li
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Ziyuan Li
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Hui Yao
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Yongju Zhao
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Zhongquan Zhao
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
20
|
Fair T, Lonergan P. The oocyte: the key player in the success of assisted reproduction technologies. Reprod Fertil Dev 2023; 36:133-148. [PMID: 38064189 DOI: 10.1071/rd23164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
The ovulation of a mature oocyte at metaphase II of meiosis, with optimal potential to undergo fertilisation by a sperm cell, complete meiosis and sustain the switch to mitotic division, and support early embryo development, involves a protracted and disrupted/delayed series of processes. Many of these are targeted for exploitation in vivo , or recapitulation in vitro , by the livestock industry. Reproductive technologies, including AI, multiple ovulation embryo transfer, ovum pick-up, in vitro embryo production, and oestrus and ovulation synchronisation, offer practitioners and producers the opportunity to produce offspring from genetically valuable dams in much greater numbers than they would normally have in their lifetime, while in vitro oocyte and follicle culture are important platforms for researchers to interrogate the physiological mechanisms driving fertility. The majority of these technologies target the ovarian follicle and the oocyte within; thus, the quality and capability of the recovered oocyte determine the success of the reproductive intervention. Molecular and microscopical technologies have grown exponentially, providing powerful platforms to interrogate the molecular mechanisms which are integral to or affected by ART. The development of the bovine oocyte from its differentiation in the ovary to ovulation is described in the light of its relevance to key aspects of individual interventions, while highlighting the historical timeline.
Collapse
Affiliation(s)
- Trudee Fair
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, Ireland
| | - Pat Lonergan
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
21
|
Pei Z, Deng K, Xu C, Zhang S. The molecular regulatory mechanisms of meiotic arrest and resumption in Oocyte development and maturation. Reprod Biol Endocrinol 2023; 21:90. [PMID: 37784186 PMCID: PMC10544615 DOI: 10.1186/s12958-023-01143-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/20/2023] [Indexed: 10/04/2023] Open
Abstract
In human female primordial germ cells, the transition from mitosis to meiosis begins from the fetal stage. In germ cells, meiosis is arrested at the diplotene stage of prophase in meiosis I (MI) after synapsis and recombination of homologous chromosomes, which cannot be segregated. Within the follicle, the maintenance of oocyte meiotic arrest is primarily attributed to high cytoplasmic concentrations of cyclic adenosine monophosphate (cAMP). Depending on the specific species, oocytes can remain arrested for extended periods of time, ranging from months to even years. During estrus phase in animals or the menstrual cycle in humans, the resumption of meiosis occurs in certain oocytes due to a surge of luteinizing hormone (LH) levels. Any factor interfering with this process may lead to impaired oocyte maturation, which in turn affects female reproductive function. Nevertheless, the precise molecular mechanisms underlying this phenomenon has not been systematically summarized yet. To provide a comprehensive understanding of the recently uncovered regulatory network involved in oocyte development and maturation, the progress of the cellular and molecular mechanisms of oocyte nuclear maturation including meiosis arrest and meiosis resumption is summarized. Additionally, the advancements in understanding the molecular cytoplasmic events occurring in oocytes, such as maternal mRNA degradation, posttranslational regulation, and organelle distribution associated with the quality of oocyte maturation, are reviewed. Therefore, understanding the pathways regulating oocyte meiotic arrest and resumption will provide detailed insight into female reproductive system and provide a theoretical basis for further research and potential approaches for novel disease treatments.
Collapse
Affiliation(s)
- Zhenle Pei
- Shanghai Ji Ai Genetics & IVF Institute, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Ke Deng
- Shanghai Ji Ai Genetics & IVF Institute, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Congjian Xu
- Shanghai Ji Ai Genetics & IVF Institute, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China.
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, 200032, China.
| | - Shuo Zhang
- Shanghai Ji Ai Genetics & IVF Institute, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China.
| |
Collapse
|
22
|
Xu Y, Wang E, Liu T, Wang S, Wu F, Zhao X, Wang A. Whole exome sequencing identifies a novel homozygous missense mutation of LHCGR gene in primary infertile women with empty follicle syndrome. J Obstet Gynaecol Res 2023; 49:2436-2445. [PMID: 37462066 DOI: 10.1111/jog.15747] [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: 02/21/2023] [Accepted: 07/05/2023] [Indexed: 10/03/2023]
Abstract
AIM The genetic basis of empty follicle syndrome (EFS) is largely unknown, and the aim of this study was to investigate the genetic causes of EFS in primary infertile women. METHODS Four affected women diagnosed with anovulation were recruited, and whole exome sequencing (WES) was requested for the genetic diagnosis of the cases. One hundred healthy controls were verified by Sanger sequencing. RESULTS A novel homozygous variant of the LHCGR gene (NM_000233:c.1847C>A) was revealed in one affected individual by WES. Trios analysis of the mutation revealed an autosomal recessive pattern. This LHCGR variant was absent in 100 healthy controls and predicted to be highly damaging to the function of LHCGR. CONCLUSIONS The novel variant extends the mutational spectrum of the LHCGR gene associated with female sterility, which promotes the prognostic value of testing for LHCGR mutations in infertile women with EFS.
Collapse
Affiliation(s)
- Yang Xu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Department of Reproductive Medicine, Linyi People's Hospital, Shandong University, Linyi, China
| | - Enhua Wang
- Teaching and Research Office of Medical Genetics, Shandong Medical College, Linyi, China
| | - Tianfeng Liu
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, China
| | - Surong Wang
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, China
| | - Fengxia Wu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Xiangyu Zhao
- Department of Medical Genetics, Linyi People's Hospital, Shandong University, Linyi, China
| | - Ancong Wang
- Department of Reproductive Medicine, Linyi People's Hospital, Shandong University, Linyi, China
| |
Collapse
|
23
|
Song X, Jiang H, Lv P, Cui K, Liu Q, Yin S, Liu H, Li Z. Transcriptome analyses reveal transcriptional profiles of horse oocytes before and after in vitro maturation. Reprod Domest Anim 2023; 58:1468-1479. [PMID: 37650336 DOI: 10.1111/rda.14462] [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: 06/15/2023] [Revised: 08/06/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023]
Abstract
Oocyte in vitro maturation is necessary for the study and application of animal-assisted reproduction technology in animal reproduction and breeding. The comprehensive transcriptional profile of equine oocyte maturated in vitro has not been fully mined yet, which makes many key transcriptional events still unidentified. Here, Smart-seq2 was performed to analyse the gene expression pattern and the underlying regulatory mechanism of horse germinal vesicle (GV) and in vitro metaphase II (MII) oocytes. The results showed that 6402 genes (2640 up-regulated and 3762 down-regulated in MII samples compared to GV) and 4021 lncRNA transcripts (1210 up-regulated and 2811 down-regulated in MII samples compared to GV) were differentially expressed in GV and MII oocytes. Further, GO and KEGG analysis found that differentially expressed mRNAs and lncRNAs were mainly enriched in the pathways related to energy and lipid metabolism. In addition, LGALS3 was found a key gene in mediating the regulation of oocyte meiosis recovery and fertilization ability. This study provides novel knowledge about gene expression and energy metabolism during equine oocyte maturation and a reference for the further study and application of assisted reproductive technology in horse reproduction and breeding.
Collapse
Affiliation(s)
- Xinhui Song
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Hancai Jiang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Peiru Lv
- Henan Chuangyuan Biotechnology Co. Ltd, Zhengzhou, China
| | - Kuiqing Cui
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Qingyou Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Shan Yin
- Henan Chuangyuan Biotechnology Co. Ltd, Zhengzhou, China
| | - Hongbo Liu
- Henan Chuangyuan Biotechnology Co. Ltd, Zhengzhou, China
| | - Zhipeng Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| |
Collapse
|
24
|
Telfer EE, Grosbois J, Odey YL, Rosario R, Anderson RA. Making a good egg: human oocyte health, aging, and in vitro development. Physiol Rev 2023; 103:2623-2677. [PMID: 37171807 PMCID: PMC10625843 DOI: 10.1152/physrev.00032.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023] Open
Abstract
Mammalian eggs (oocytes) are formed during fetal life and establish associations with somatic cells to form primordial follicles that create a store of germ cells (the primordial pool). The size of this pool is influenced by key events during the formation of germ cells and by factors that influence the subsequent activation of follicle growth. These regulatory pathways must ensure that the reserve of oocytes within primordial follicles in humans lasts for up to 50 years, yet only approximately 0.1% will ever be ovulated with the rest undergoing degeneration. This review outlines the mechanisms and regulatory pathways that govern the processes of oocyte and follicle formation and later growth, within the ovarian stroma, through to ovulation with particular reference to human oocytes/follicles. In addition, the effects of aging on female reproductive capacity through changes in oocyte number and quality are emphasized, with both the cellular mechanisms and clinical implications discussed. Finally, the details of current developments in culture systems that support all stages of follicle growth to generate mature oocytes in vitro and emerging prospects for making new oocytes from stem cells are outlined.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Johanne Grosbois
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Yvonne L Odey
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roseanne Rosario
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
25
|
Chen N, Zhang Y, Tian Y, Wu S, Gao F, Yuan X. Deciphering Cellular Heterogeneity and Communication Patterns in Porcine Antral Follicles by Single-Cell RNA Sequencing. Animals (Basel) 2023; 13:3019. [PMID: 37835625 PMCID: PMC10571650 DOI: 10.3390/ani13193019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/17/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
The antral follicle stage is a critical period in mammalian oocyte maturation, marked by complex interactions between oocyte development and neighboring granulosa cells. Understanding the heterogeneity and communication patterns of granulosa cells within antral follicles is crucial for deciphering their roles in follicle development and oocyte maturation. Here, we employed single-cell RNA-sequencing to explore the molecular and cellular characteristics of porcine antral follicles. Our analysis revealed distinct subpopulations within mural and cumulus granulosa cells, indicating diverse cellular states and functions within the follicles. Functional enrichment analysis unveiled the involvement of specific subpopulations in steroid biosynthesis, cumulus expansion, and cellular communication. Moreover, comparing mature and less mature follicles highlighted differences in cell distribution and functions, indicating developmental-specific variations. Our findings shed light on the intricate cellular heterogeneity and communication network within porcine antral follicles, providing valuable insights into the regulation of follicle development and oocyte maturation in pigs. These results hold promise for improving pig reproductive efficiency and advancing human reproductive medicine.
Collapse
Affiliation(s)
- Na Chen
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
| | - Yong Zhang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
| | - Yuhan Tian
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510000, China
| | - Shumei Wu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510000, China
| | - Fei Gao
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark
| | - Xiaolong Yuan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510000, China
| |
Collapse
|
26
|
Maugrion E, Shedova EN, Uzbekov R, Teixeira-Gomes AP, Labas V, Tomas D, Banliat C, Singina GN, Uzbekova S. Extracellular Vesicles Contribute to the Difference in Lipid Composition between Ovarian Follicles of Different Size Revealed by Mass Spectrometry Imaging. Metabolites 2023; 13:1001. [PMID: 37755281 PMCID: PMC10538054 DOI: 10.3390/metabo13091001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/01/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023] Open
Abstract
Follicular fluid (FF) ensures a safe environment for oocyte growth and maturation inside the ovarian follicle in mammals. In each cycle, the large dominant follicle (LF) contains the oocyte designated to be ovulated, whereas the small subordinate follicles (SFs) of the same wave will die through atresia. In cows, the oocytes from the SF, being 2 mm in size, are suitable for in vitro reproduction biotechnologies, and their competence in developing an embryo depends on the size of the follicles. FF contains proteins, metabolites, fatty acids, and a multitude of extracellular vesicles (ffEVs) of different origins, which may influence oocyte competence through bidirectional exchanges of specific molecular cargo between follicular cells and enclosed oocytes. FF composition evolves along with follicle growth, and the abundance of different lipids varies between the LF and SF. Here, significant differences in FF lipid content between the LFs and SFs within the same ovary were demonstrated by MALD-TOF mass spectrometry imaging on bovine ovarian sections. We then aimed to enlighten the lipid composition of FF, and MALDI-TOF lipid profiling was performed on cellular, vesicular, and liquid fractions of FF. Differential analyses on the abundance of detected lipid features revealed specific enrichment of phospholipids in different ffEV types, such as microvesicles (MVs) and exosomes (Exo), compared to depleted FF. MALDI-TOF lipid profiling on MVs and Exo from the LF and SF samples (n = 24) revealed that more than 40% of detected features were differentially abundant between the groups of MVs and Exo from the different follicles (p < 0.01, fold change > 2). Glycerophospholipid and sphingolipid features were more abundant in ffEVs from the SFs, whereas different lysophospholipids, including phosphatidylinositols, were more abundant in the LFs. As determined by functional analysis, the specific lipid composition of ffEVs suggested the involvement of vesicular lipids in cell signaling pathways and largely contributed to the differentiation of the dominant and subordinate follicles.
Collapse
Affiliation(s)
- Emilie Maugrion
- CNRS, INRAE, University of Tours, PRC, 37380 Nouzilly, France (A.-P.T.-G.); (V.L.); (D.T.)
- PIXANIM, INRAE, University of Tours, CHU of Tours, 37380 Nouzilly, France
| | | | - Rustem Uzbekov
- Laboratory of Cell Biology and Electron Microscopy, Medical Faculty, University of Tours, 37032 Tours, France
- Faculty of Bioengineering and Bioinformatics, Moscow State University, 119992 Moscow, Russia
| | - Ana-Paula Teixeira-Gomes
- CNRS, INRAE, University of Tours, PRC, 37380 Nouzilly, France (A.-P.T.-G.); (V.L.); (D.T.)
- PIXANIM, INRAE, University of Tours, CHU of Tours, 37380 Nouzilly, France
| | - Valerie Labas
- CNRS, INRAE, University of Tours, PRC, 37380 Nouzilly, France (A.-P.T.-G.); (V.L.); (D.T.)
- PIXANIM, INRAE, University of Tours, CHU of Tours, 37380 Nouzilly, France
| | - Daniel Tomas
- CNRS, INRAE, University of Tours, PRC, 37380 Nouzilly, France (A.-P.T.-G.); (V.L.); (D.T.)
- PIXANIM, INRAE, University of Tours, CHU of Tours, 37380 Nouzilly, France
| | - Charles Banliat
- PIXANIM, INRAE, University of Tours, CHU of Tours, 37380 Nouzilly, France
- Ecole Supérieure d’Agricultures (ESA), 49007 Angers, France
| | - Galina N. Singina
- L.K. Ernst Federal Research Center for Animal Husbandry, 142132 Podolsk, Russia
| | - Svetlana Uzbekova
- CNRS, INRAE, University of Tours, PRC, 37380 Nouzilly, France (A.-P.T.-G.); (V.L.); (D.T.)
| |
Collapse
|
27
|
Buratini J, Dellaqua TT, de Lima PF, Renzini MM, Canto MD, Price CA. Oocyte secreted factors control genes regulating FSH signaling and the maturation cascade in cumulus cells: the oocyte is not in a hurry. J Assist Reprod Genet 2023; 40:1961-1971. [PMID: 37204638 PMCID: PMC10371970 DOI: 10.1007/s10815-023-02822-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/29/2023] [Indexed: 05/20/2023] Open
Abstract
PURPOSE To assess the effects of the oocyte on mRNA abundance of FSHR, AMH and major genes of the maturation cascade (AREG, EREG, ADAM17, EGFR, PTGS2, TNFAIP6, PTX3, and HAS2) in bovine cumulus cells. METHODS (1) Intact cumulus-oocyte complexes, (2) microsurgically oocytectomized cumulus-oolema complexes (OOX), and (3) OOX + denuded oocytes (OOX+DO) were subjected to in vitro maturation (IVM) stimulated with FSH for 22 h or with AREG for 4 and 22 h. After IVM, cumulus cells were separated and relative mRNA abundance was measured by RT-qPCR. RESULTS After 22 h of FSH-stimulated IVM, oocytectomy increased FSHR mRNA levels (p=0.005) while decreasing those of AMH (p=0.0004). In parallel, oocytectomy increased mRNA abundance of AREG, EREG, ADAM17, PTGS2, TNFAIP6, and PTX3, while decreasing that of HAS2 (p<0.02). All these effects were abrogated in OOX+DO. Oocytectomy also reduced EGFR mRNA levels (p=0.009), which was not reverted in OOX+DO. The stimulatory effect of oocytectomy on AREG mRNA abundance (p=0.01) and its neutralization in OOX+DO was again observed after 4 h of AREG-stimulated IVM. After 22 h of AREG-stimulated IVM, oocytectomy and addition of DOs to OOX caused the same effects on gene expression observed after 22 h of FSH-stimulated IVM, except for ADAM17 (p<0.025). CONCLUSION These findings suggest that oocyte-secreted factors inhibit FSH signaling and the expression of major genes of the maturation cascade in cumulus cells. These may be important actions of the oocyte favoring its communication with cumulus cells and preventing premature activation of the maturation cascade.
Collapse
Affiliation(s)
- Jose Buratini
- Biogenesi, Reproductive Medicine Centre, Monza, Italy
- Clinica EUGIN, Milan, Italy
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP Brazil
| | - Thaisy Tino Dellaqua
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP Brazil
| | - Paula Fernanda de Lima
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP Brazil
| | | | | | - Christopher A. Price
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Canada
| |
Collapse
|
28
|
Ponce-Barajas P, Colazo MG, Behrouzi A, Ree TO, Kastelic JP, Ambrose DJ. Morphologic, Steroidogenic, and Transcriptomic Assessment of the Corpus Luteum in Holstein Cows after Spontaneous or Hormone-Induced Ovulation. Animals (Basel) 2023; 13:2283. [PMID: 37508059 PMCID: PMC10376098 DOI: 10.3390/ani13142283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
There is evidence that replacing the gonadotropin-releasing hormone (GnRH) with porcine luteinizing hormone (pLH) to synchronize ovulation prior to artificial insemination (AI) increased pregnancy per AI in dairy cows without affecting blood progesterone (P4) concentrations. Whether morphologic, steroidogenic, and transcriptomic differences exist among corpora lutea (CL) formed after ovulation induced by GnRH and pLH is unclear. Our main objective, therefore, was to compare CL characteristics between GnRH- and pLH-induced CL. In 24 non-lactating Holstein cows, ovulations were spontaneous (Spont-Ov) or induced with 100 µg GnRH, 25 mg pLH, or 1 mg estradiol benzoate (EB), with CL excised 12 d after ovulation. In pLH- versus GnRH-treated cows, the duration of elevated LH (above baseline) was prolonged (10 versus 6 h, respectively, p < 0.01), but CL dimensions, pixel intensity of CL images, proportions of steroidogenic and non-steroidogenic luteal cells, and mean plasma LH did not significantly differ. Post-ovulation mean plasma P4 (ng/mL) did not differ among Spont-Ov (3.0) pLH (3.1) or GnRH (3.0) cows but were lower in EB cows (2.0). In vitro P4 concentration was greater in luteal explants of pLH-treated cows than in all other groups (combined means, 16.0 vs. 12.3 µg/mL, p < 0.02). Relative abundance of mRNA for oxytocin receptor (OXTR) was 2-fold higher (p < 0.01) in CL of pLH vs. GnRH cows and highest in Spont-Ov CL. In summary, pLH-treated cows had a longer LH peak, and greatest luteal tissue concentrations and in vitro production of P4. We inferred that increased P4 concentrations at the ovarian-uterine level in pLH-treated cows could have promoted embryo development and increased pregnancy per AI.
Collapse
Affiliation(s)
- Patricio Ponce-Barajas
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Marcos G Colazo
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Amir Behrouzi
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Todd O Ree
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - John P Kastelic
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Divakar J Ambrose
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 1C9, Canada
| |
Collapse
|
29
|
Nie H, Xu Y, Zhang Y, Wen Y, Zhan J, Xia Y, Zhou Y, Wang R, Wu X. The effects of endogenous FSH and its receptor on oogenesis and folliculogenesis in female Alligator sinensis. BMC ZOOL 2023; 8:8. [PMID: 37403129 DOI: 10.1186/s40850-023-00170-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 06/26/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND The precise mechanisms of hormone action responsible for the full course of events modulating folliculogenesis in crocodilian have not been determined, although histological features have been identified. RESULTS The Alligator sinensis ovarian morphological characteristics observed at 1, 15, 30, 60, 90, and 300 days post hatching(dph) revealed that the dynamic changes in germ cells varied in different meiotic and developmental stages, confirming that the processes of folliculogenesis were protracted and asynchronous. The presence of endogenous follicle-stimulating hormone(FSH) mRNA and protein expression within the cerebrum at 1 dph, in parallel with the increase in germ cells within the germ cell nests(Nest) from 1 dph to 15 dph, suggested that endocrine regulation of the pituitary-gonad axis is an early event in oogonia division. Furthermore, the endogenous expression of FSH showed a trend of negative feedback augmentation accompanied by the exhaustion of maternal yolk E2 observed at 15 dph. Such significant elevation of endogenous FSH levels was observed to be related to pivotal events in the transition from mitosis to meiosis, as reflected by the proportion of oogonia during premeiosis interphase, with endogenous FSH levels reaching a peak at the earliest time step of 1 dph. In addition, the simultaneous upregulation of premeiotic marker STRA8 mRNA expression and the increase in endogenous FSH further verified the above speculation. The strongly FSHr-positive label in the oocytes within Pre-previtellogenic follicles was synchronized with the significant elevation of ovarian cAMP detected at 300 dph, which suggested that diplotene arrest maintenance during early vitellogenesis might be FSH dependent. In addition, preferential selection in asynchronous meiotic initiation has been supposed to act on somatic supportive cells and not directly on germ cells via regulation of FSH that in turn affects downstream estrogen levels. This suggestion was verified by the reciprocal stimulating effect of FSH and E2 on the accelerated meiotic marker SYCP3 and by the inhibited cell apoptosis demonstrated in ovarian cell culture in vitro. CONCLUSION The corresponding results contribute an expansion of the understanding of physiological processes and shed some light on the specific factors responsible for gonadotropin function in the early folliculogenesis of crocodilians.
Collapse
Affiliation(s)
- Haitao Nie
- Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China
| | - Yunlu Xu
- Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China
| | - Yuqian Zhang
- Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China
| | - Yue Wen
- Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China
| | - Jixiang Zhan
- Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China
| | - Yong Xia
- Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China
| | - Yongkang Zhou
- Alligator Research Center of Anhui Province, Xuanzhou, 242000, People's Republic of China
| | - Renping Wang
- Alligator Research Center of Anhui Province, Xuanzhou, 242000, People's Republic of China
| | - Xiaobing Wu
- Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China.
| |
Collapse
|
30
|
Zhang FL, Li WD, Zhang G, Zhang M, Liu ZJ, Zhu KX, Liu QC, Zhang SE, Shen W, Zhang XF. Identification of unique transcriptomic signatures through integrated multispecies comparative analysis and WGCNA in bovine oocyte development. BMC Genomics 2023; 24:265. [PMID: 37202739 DOI: 10.1186/s12864-023-09362-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 05/06/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Cattle (Bos taurus) are a major large livestock, however, compared with other species, the transcriptional specificity of bovine oocyte development has not been emphasised. RESULTS To reveal the unique transcriptional signatures of bovine oocyte development, we used integrated multispecies comparative analysis and weighted gene co-expression network analysis (WGCNA) to perform bioinformatic analysis of the germinal follicle (GV) and second meiosis (MII) gene expression profile from cattle, sheep, pigs and mice. We found that the expression levels of most genes were down-regulated from GV to MII in all species. Next, the multispecies comparative analysis showed more genes involved in the regulation of cAMP signalling during bovine oocyte development. Moreover, the green module identified by WGCNA was closely related to bovine oocyte development. Finally, integrated multispecies comparative analysis and WGCNA picked up 61 bovine-specific signature genes that participate in metabolic regulation and steroid hormone biosynthesis. CONCLUSION In a short, this study provides new insights into the regulation of cattle oocyte development from a cross-species comparison.
Collapse
Affiliation(s)
- Fa-Li Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266100, China
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Wei-Dong Li
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong, China
| | - Geng Zhang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Min Zhang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Zhao-Jun Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ke-Xin Zhu
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Qing-Chun Liu
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Shu-Er Zhang
- Animal Husbandry General Station of Shandong Province, Jinan, 250010, China
| | - Wei Shen
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Xi-Feng Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266100, China.
- Qingdao Longming Cattle Industry Co., Ltd, Qingdao, China.
| |
Collapse
|
31
|
Marcial Lopez A, Ratner LD, Martinez CS, Di Giorgio N, Poutanen M, Huhtaniemi IT, Rulli SB. Persistently expressed human chorionic gonadotropin induces premature luteinization and progressive alterations on the reproductive axis in female mice. Gen Comp Endocrinol 2023; 336:114247. [PMID: 36858273 DOI: 10.1016/j.ygcen.2023.114247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023]
Abstract
The hypothalamic-pituitary-gonadal axis plays a fundamental role in the endocrine regulation of the reproductive function in mammals. Any change in the function of the participating hormones or their receptors can lead to alterations in sexual differentiation, the onset of puberty, infertility, cancer development, and other dysfunctions. In this study, we analyzed the influence of persistently elevated levels of the human chorionic gonadotropin hormone (hCG), a powerful agonist of pituitary luteinizing hormone (LH), on the reproductive axis of female mice. As a consequence of chronic hCG hypersecretion through a global expression of the hCGbeta-subunit in transgenic (TG) female mice, a series of events perturbed the prepubertal to juvenile transition. The imbalance in gonadotropin action was first manifested by precocious puberty and alterations in gonadal hormone production, with the consequent ovarian function disruption and infertility in adulthood. The expansion of cumulus cells in vivo and in vitro, ovulatory capacity, and gene expression of ovulation-related marker genes after hormone stimulation were normal in 3-week-old TG females. However, the expression of genes related to steroidogenesis and luteinization such as Lhcgr, Prlr, and the steroidogenic enzymes Cyp11a1, Cyp17a1, and Cyp19a1 were significantly elevated in the TG females. This study demonstrates that the excessive secretion of hCG in concert with high prolactin, induced premature luteinization, and enhanced ovarian steroidogenesis, as was shown by the up-regulation of luteal cell markers and progesterone synthesis in the TG mice. Furthermore, progressively impaired reproductive function of the TG females occurred from the peripubertal stage to adulthood, thus culminating in infertility.
Collapse
Affiliation(s)
- Agustina Marcial Lopez
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490 (1428), Buenos Aires, Argentina
| | - Laura D Ratner
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490 (1428), Buenos Aires, Argentina
| | - Carolina S Martinez
- Laboratorio de Bio-nanotecnología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Grupo vinculado GBEyB, IMBICE-CONICET-CICPBA, Bernal, Argentina
| | - Noelia Di Giorgio
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490 (1428), Buenos Aires, Argentina
| | - Matti Poutanen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland; Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland
| | - Ilpo T Huhtaniemi
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland; Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London W12 0NN, UK
| | - Susana B Rulli
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490 (1428), Buenos Aires, Argentina.
| |
Collapse
|
32
|
Zheng W, Zhang T, Zhao T, Zhu Z, Qin S, Yan H, He M, Zhou B, Xia G, Zhang H, Wang C. cAMP controls the balance between dormancy and activation of primordial follicles in mouse ovaries. PNAS NEXUS 2023; 2:pgad055. [PMID: 36938502 PMCID: PMC10019762 DOI: 10.1093/pnasnexus/pgad055] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/22/2023] [Accepted: 02/09/2023] [Indexed: 02/25/2023]
Abstract
In mammalian ovaries, the balance between dormancy and activation of primordial follicles determines the female fecundity and endocrine homeostasis. Recently, several functional molecules and pathways have been reported to be involved in the activation of primordial follicles. However, the homeostasis regulatory mechanisms of primordial follicle activation are still scant. Our previous study has proved that a relatively higher concentration of cyclic AMP (cAMP) is required for primordial follicle formation. Here, we identified that cAMP also plays a vital role in the balance between dormancy and activation of primordial follicles. Our results showed that the concentration of cAMP remained stable in neonatal mouse ovaries, which is due to ADCY3, the synthetase of cAMP, and PDE3A, the hydrolytic enzyme of cAMP, were synchronously increased during the activation of primordial follicles in mouse ovaries. Once the concentration of cAMP in neonatal ovaries was either elevated or reduced in vitro, the activation of primordial follicles was either accelerated or decelerated accordingly. In addition, a higher concentration of cAMP in the ovaries of puberty mice improved primordial follicle activation in vivo. Finally, cAMP promoted primordial follicle activation via canonical mTORC1-PI3K signaling cascades and PKA signaling. In conclusion, our findings reveal that the concentration of cAMP acts as a key regulator in balancing the dormancy and activation of primordial follicles in the mouse ovary.
Collapse
Affiliation(s)
| | | | - Ting Zhao
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Zijian Zhu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Shaogang Qin
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Hao Yan
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Meina He
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou Province 550025, China
| | - Bo Zhou
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Guoliang Xia
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan 750021, China
| | - Hua Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Chao Wang
- To whom correspondence should be addressed:
| |
Collapse
|
33
|
Lin B, Zhou X, Jiang D, Shen X, Ouyang H, Li W, Xu D, Fang L, Tian Y, Li X, Huang Y. Comparative transcriptomic analysis reveals candidate genes for seasonal breeding in the male Lion-Head goose. Br Poult Sci 2023; 64:157-163. [PMID: 36440984 DOI: 10.1080/00071668.2022.2152651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
1. Due to seasonal breeding, geese breeds from Southern China have low egg yield. The genetic makeup underlying performance of local breeds is largely unknown, and few studies have investigated this problem. This study integrated 21 newly generated and 50 publicly existing RNA-seq libraries, representing the hypothalamus, pituitary and testis, to identify candidate genes and importantly related pathways associated with seasonal breeding in male Lion-Head geese.2. In total, 19, 119 and 302 differentially expressed genes (DEGs) were detected in the hypothalamus, pituitary and testis, respectively, of male Lion-Head geese between non-breeding and breeding periods. These genes were significantly involved in the neuropeptide signalling pathway, gland development, neuroactive ligand-receptor interaction, JAK-STAT signalling pathway, cAMP signalling pathway, PI3K-Akt signalling pathway and Foxo signalling pathway.3. By integrating another 50 RNA-seq samples 4, 18 and 40 promising DEGs were confirmed in hypothalamus, pituitary and testis, respectively.4. HOX genes were identified as having important roles in the development of testis between non-breeding and breeding periods of male Lion-Head geese.
Collapse
Affiliation(s)
- B Lin
- Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, P. R. China
| | - X Zhou
- Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, P. R. China
| | - D Jiang
- Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, P. R. China
| | - X Shen
- Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, P. R. China
| | - H Ouyang
- Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, P. R. China
| | - W Li
- Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, P. R. China
| | - D Xu
- Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, P. R. China
| | - L Fang
- MRC Human Genetics Unit at Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Y Tian
- Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, P. R. China
| | - X Li
- Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, P. R. China
| | - Y Huang
- Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
34
|
Fang L, Sun YP, Cheng JC. The role of amphiregulin in ovarian function and disease. Cell Mol Life Sci 2023; 80:60. [PMID: 36749397 PMCID: PMC11071807 DOI: 10.1007/s00018-023-04709-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/17/2023] [Accepted: 01/24/2023] [Indexed: 02/08/2023]
Abstract
Amphiregulin (AREG) is an epidermal growth factor (EGF)-like growth factor that binds exclusively to the EGF receptor (EGFR). Treatment with luteinizing hormone (LH) and/or human chorionic gonadotropin dramatically induces the expression of AREG in the granulosa cells of the preovulatory follicle. In addition, AREG is the most abundant EGFR ligand in human follicular fluid. Therefore, AREG is considered a predominant propagator that mediates LH surge-regulated ovarian functions in an autocrine and/or paracrine manner. In addition to the well-characterized stimulatory effect of LH on AREG expression, recent studies discovered that several local factors and epigenetic modifications participate in the regulation of ovarian AREG expression. Moreover, aberrant expression of AREG has recently been reported to contribute to the pathogenesis of several ovarian diseases, such as ovarian hyperstimulation syndrome, polycystic ovary syndrome, and epithelial ovarian cancer. Furthermore, increasing evidence has elucidated new applications of AREG in assisted reproductive technology. Collectively, these studies highlight the importance of AREG in female reproductive health and disease. Understanding the normal and pathological roles of AREG and elucidating the molecular and cellular mechanisms of AREG regulation of ovarian functions will inform innovative approaches for fertility regulation and the prevention and treatment of ovarian diseases. Therefore, this review summarizes the functional roles of AREG in ovarian function and disease.
Collapse
Affiliation(s)
- Lanlan Fang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 40, Daxue Road, Zhengzhou, 450052, Henan, China
| | - Ying-Pu Sun
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 40, Daxue Road, Zhengzhou, 450052, Henan, China
| | - Jung-Chien Cheng
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 40, Daxue Road, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
35
|
Xie J, Xu X, Liu S. Intercellular communication in the cumulus-oocyte complex during folliculogenesis: A review. Front Cell Dev Biol 2023; 11:1087612. [PMID: 36743407 PMCID: PMC9893509 DOI: 10.3389/fcell.2023.1087612] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/10/2023] [Indexed: 01/20/2023] Open
Abstract
During folliculogenesis, the oocyte and surrounding cumulus cells form an ensemble called the cumulus-oocyte complex (COC). Due to their interdependence, research on the COC has been a hot issue in the past few decades. A growing body of literature has revealed that intercellular communication is critical in determining oocyte quality and ovulation. This review provides an update on the current knowledge of COC intercellular communication, morphology, and functions. Transzonal projections (TZPs) and gap junctions are the most described structures of the COC. They provide basic metabolic and nutrient support, and abundant molecules for signaling pathways and regulations. Oocyte-secreted factors (OSFs) such as growth differentiation factor 9 and bone morphogenetic protein 15 have been linked with follicular homeostasis, suggesting that the communications are bidirectional. Using advanced techniques, new evidence has highlighted the existence of other structures that participate in intercellular communication. Extracellular vesicles can carry transcripts and signaling molecules. Microvilli on the oocyte can induce the formation of TZPs and secrete OSFs. Cell membrane fusion between the oocyte and cumulus cells can lead to sharing of cytoplasm, in a way making the COC a true whole. These findings give us new insights into related reproductive diseases like polycystic ovary syndrome and primary ovarian insufficiency and how to improve the outcomes of assisted reproduction.
Collapse
Affiliation(s)
- Jun Xie
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao Xu
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Suying Liu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China,*Correspondence: Suying Liu,
| |
Collapse
|
36
|
Pei J, Xiong L, Guo S, Wang X, La Y, Chu M, Liang C, Yan P, Guo X. Single-Cell Transcriptomics Analysis Reveals a Cell Atlas and Cell Communication in Yak Ovary. Int J Mol Sci 2023; 24:ijms24031839. [PMID: 36768166 PMCID: PMC9915757 DOI: 10.3390/ijms24031839] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/15/2022] [Accepted: 01/15/2023] [Indexed: 01/19/2023] Open
Abstract
Yaks (Bos grunniens) are the only bovine species that adapt well to the harsh high-altitude environment in the Qinghai-Tibetan plateau. However, the reproductive adaptation to the climate of the high elevation remains to be elucidated. Cell composition and molecular characteristics are the foundation of normal ovary function which determines reproductive performance. So, delineating ovarian characteristics at a cellular molecular level is conducive to elucidating the mechanism underlying the reproductive adaption of yaks. Here, the single-cell RNA-sequencing (scRNA-seq) was employed to depict an atlas containing different cell types with specific molecular signatures in the yak ovary. The cell types were identified on the basis of their specifically expressed genes and biological functions. As a result, a cellular atlas of yak ovary was established successfully containing theca cells, stromal cells, endothelial cells, smooth muscle cells, natural killer cells, macrophages, and proliferating cells. A cell-to-cell communication network between the distinct cell types was constructed. The theca cells were clustered into five subtypes based on their biological functions. Further, CYP11A1 was confirmed as a marker gene for the theca cells by immunofluorescence staining. Our work reveals an ovarian atlas at the cellular molecular level and contributes to providing insights into reproductive adaption in yaks.
Collapse
Affiliation(s)
- Jie Pei
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Lin Xiong
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Shaoke Guo
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Xingdong Wang
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Yongfu La
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Min Chu
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Chunnian Liang
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Ping Yan
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Xian Guo
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
- Correspondence:
| |
Collapse
|
37
|
Extracellular vesicles-encapsulated microRNA in mammalian reproduction: A review. Theriogenology 2023; 196:174-185. [PMID: 36423512 DOI: 10.1016/j.theriogenology.2022.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/08/2022] [Accepted: 11/12/2022] [Indexed: 11/16/2022]
Abstract
Extracellular vesicles (EVs) are nanoscale cell-derived lipid vesicles that participate in cell-cell communication by delivering cargo, including mRNAs, proteins and non-coding RNAs, to recipient cells. MicroRNA (miRNA), a non-coding RNA typically 22 nucleotides long, is crucial for nearly all developmental and pathophysiological processes in mammals by regulating recipient cells gene expression. Infertility is a worldwide health issue that affects 10-15% of couples during their reproductive years. Although assisted reproductive technology (ART) gives infertility couples hope, the failure of ART is mainly unknown. It is well accepted that EVs-encapsulated miRNAs have a role in different reproductive processes, implying that these EVs-encapsulated miRNAs could optimize ART, improve reproductive rate, and treat infertility. As a result, in this review, we describe the present understanding of EVs-encapsulated miRNAs in reproduction regulation.
Collapse
|
38
|
Luciano AM, Franciosi F, Dey P, Ladron De Guevara M, Monferini N, Bonumallu SKN, Musmeci G, Fagali Franchi F, Garcia Barros R, Colombo M, Lodde V. Progress toward species-tailored prematuration approaches in carnivores. Theriogenology 2023; 196:202-213. [PMID: 36423514 DOI: 10.1016/j.theriogenology.2022.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/05/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
In the past four decades, the bovine model has been highly informative and inspiring to assisted reproductive technologies (ART) in other species. Most of the recent advances in ART have come from studies in cattle, particularly those unveiling the importance of several processes that must be recapitulated in vitro to ensure the proper development of the oocyte. The maintenance of structural and functional communications between the cumulus cells and the oocyte and a well-orchestrated chromatin remodeling with the gradual silencing of transcriptional activity represent essential processes for the progressive acquisition of oocyte developmental competence. These markers are now considered the milestones of physiological approaches to increase the efficiency of reproductive technologies. Different in vitro approaches have been proposed. In particular, the so-called "pre-IVM" or "prematuration" is a culture step performed before in vitro maturation (IVM) to support the completion of the oocyte differentiation process. Although these attempts only partially improved the embryo quality and yield, they currently represent a proof of principle that oocytes retrieved from an ovary or an ovarian batch shouldn't be treated as a whole and that tailored approaches can be developed for culturing competent oocytes in several species, including humans. An advancement in ART's efficiency would be desirable in carnivores, where the success is still limited. Since the progress in reproductive medicine has often come from comparative studies, this review highlights aspects that have been critical in other species and how they may be extended to carnivores.
Collapse
Affiliation(s)
- Alberto Maria Luciano
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900, Lodi, Italy.
| | - Federica Franciosi
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900, Lodi, Italy
| | - Pritha Dey
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900, Lodi, Italy
| | - Magdalena Ladron De Guevara
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900, Lodi, Italy
| | - Noemi Monferini
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900, Lodi, Italy
| | - Sai Kamal Nag Bonumallu
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900, Lodi, Italy
| | - Giulia Musmeci
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900, Lodi, Italy
| | - Fernanda Fagali Franchi
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900, Lodi, Italy
| | - Rodrigo Garcia Barros
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900, Lodi, Italy
| | - Martina Colombo
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900, Lodi, Italy
| | - Valentina Lodde
- Reproductive and Developmental Biology Laboratory, Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900, Lodi, Italy
| |
Collapse
|
39
|
Bypassing Mendel's First Law: Transmission Ratio Distortion in Mammals. Int J Mol Sci 2023; 24:ijms24021600. [PMID: 36675116 PMCID: PMC9863905 DOI: 10.3390/ijms24021600] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Mendel's law of segregation states that the two alleles at a diploid locus should be transmitted equally to the progeny. A genetic segregation distortion, also referred to as transmission ratio distortion (TRD), is a statistically significant deviation from this rule. TRD has been observed in several mammal species and may be due to different biological mechanisms occurring at diverse time points ranging from gamete formation to lethality at post-natal stages. In this review, we describe examples of TRD and their possible mechanisms in mammals based on current knowledge. We first focus on the differences between TRD in male and female gametogenesis in the house mouse, in which some of the most well studied TRD systems have been characterized. We then describe known TRD in other mammals, with a special focus on the farmed species and in the peculiar common shrew species. Finally, we discuss TRD in human diseases. Thus far, to our knowledge, this is the first time that such description is proposed. This review will help better comprehend the processes involved in TRD. A better understanding of these molecular mechanisms will imply a better comprehension of their impact on fertility and on genome evolution. In turn, this should allow for better genetic counseling and lead to better care for human families.
Collapse
|
40
|
Pei J, Xiong L, Guo S, Wang X, Bao P, Wu X, Yan P, Guo X. A single-cell transcriptomic atlas characterizes cell types and their molecular features in yak ovarian cortex. FASEB J 2023; 37:e22718. [PMID: 36527406 DOI: 10.1096/fj.202201176rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/21/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
The ovary as one of the most dynamic organs produces steroids to orchestrate female secondary sexual characteristics, harbors ovarian reserve for oocytes, releases mature oocytes for fertilization, and maintains pregnancy. Yak (Bos grunniens) is the only bovid animal that can adapt to the harsh climatic conditions on the Qinghai-Tibetan Plateau (altitudes of over 3000 m above sea level). However, the cellular atlas is composed of oocytes and other somatic cells, and their individual molecular characteristics remain to be elucidated in the yak ovary. Here, single-cell RNA sequencing (scRNA-seq) was performed to delineate the molecular signature of various cell types in the yak ovarian cortex. A cellular atlas of yak ovarian cortex was constructed successfully on the basis of the differentially expressed genes (DEGs) from the distinct cell types and their functional enrichment analysis, comprising endothelial cells, nature kill cells, stromal cells, smooth muscle cells, oocytes, macrophages, epithelial cells, and granulosa cells. Meanwhile, the signature genes were determined based on their expression specificity in each cell type. A cell-to-cell communication network was built in light of the differentially overexpressed ligand and receptor genes from each cell type. Further, the oocytes were subdivided into four subtypes based on their individual DEGs and the functional enrichment of the DEGs. FST and TOP2A were identified as maker genes for oocytes by immunostaining in the yak ovarian cortex. The cellular atlas reveals the biological characteristics of the ovarian cortex at the cellular molecular level and provides insights into female reproductive biology via cellular communications in the yak.
Collapse
Affiliation(s)
- Jie Pei
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Lin Xiong
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Shaoke Guo
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xingdong Wang
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Pengjia Bao
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Xiaoyun Wu
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Ping Yan
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| | - Xian Guo
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, China
| |
Collapse
|
41
|
Hou X, Zeb A, Dil S, Zhou J, Zhang H, Shi B, Muhammad Z, Khan I, Zaman Q, Shah WA, Jiang X, Wu L, Ma H, Shi Q. A homozygous KASH5 frameshift mutation causes diminished ovarian reserve, recurrent miscarriage, and non-obstructive azoospermia in humans. Front Endocrinol (Lausanne) 2023; 14:1128362. [PMID: 36864840 PMCID: PMC9971600 DOI: 10.3389/fendo.2023.1128362] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
The meiosis-specific LINC complex, composed of the KASH5 and SUN1 proteins, tethers the moving chromosomes to the nuclear envelope to facilitate homolog pairing and is essential for gametogenesis. Here, we applied whole-exome sequencing for a consanguineous family with five siblings suffering from reproductive failure, and identified a homozygous frameshift mutation in KASH5 (c.1270_1273del, p.Arg424Thrfs*20). This mutation leads to the absence of KASH5 protein expression in testes and non-obstructive azoospermia (NOA) due to meiotic arrest before the pachytene stage in the affected brother. The four sisters displayed diminished ovarian reserve (DOR), with one sister never being pregnant but still having dominant follicle at 35 years old and three sisters suffering from at least 3 miscarriages occurring within the third month of gestation. The truncated KASH5 mutant protein, when expressed in cultured cells, displays a similar localization encircling the nucleus and a weakened interaction with SUN1, as compared with the full-length KASH5 proteins, which provides a potential explanation for the phenotypes in the affected females. This study reported sexual dimorphism for influence of the KASH5 mutation on human germ cell development, and extends the clinical manifestations associated with KASH5 mutations, providing genetic basis for the molecular diagnosis of NOA, DOR, and recurrent miscarriage.
Collapse
Affiliation(s)
- Xiaoning Hou
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Aurang Zeb
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Sobia Dil
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Jianteng Zhou
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Huan Zhang
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Baolu Shi
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Zubair Muhammad
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Ihsan Khan
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Qamar Zaman
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Wasim Akbar Shah
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Xiaohua Jiang
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Limin Wu
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Hui Ma
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
- *Correspondence: Qinghua Shi, ; Hui Ma,
| | - Qinghua Shi
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
- *Correspondence: Qinghua Shi, ; Hui Ma,
| |
Collapse
|
42
|
Cheng R, Zhang Z, Zhan C, Qin T, Wang L, Zhang X. Environmentally relevant concentrations of selenite trigger reproductive toxicity by affecting oocyte development and promoting larval apoptosis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 316:120648. [PMID: 36375579 DOI: 10.1016/j.envpol.2022.120648] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 06/16/2023]
Abstract
As a trace element, selenium (Se) has been widely added to food to maintain the physiological homeostasis of the organism. The adverse effects of Se on the reproduction of zebrafish have been investigated, however, the effects of Se on the maturation and apoptosis of zebrafish oocytes remain unclear. In this study, zebrafish embryos (2 h post fertilization, hpf) were exposed to 0, 12.5, 25, 50, and 100 μg Se/L for 120 days. The results demonstrated that exposure to selenite decreased the gonad-somatic index (GSI) and cumulative production of eggs, inhibited oocyte maturation (OM), and increased oocyte apoptosis in females. Exposure to selenite decreased the contents of sex hormones (E2) in the serum and increased the levels of reactive oxygen species (ROS) and cyclic adenosine monophosphate (cAMP) in the ovary. Furthermore, exposure to selenite downregulated the transcription level of genes on the HPG axis, decreased the phosphorylation level of CyclinB and the protein content of cAMP-dependent protein kinase (Pka), and upregulated the expression of genes (eif2s1a and chop) and proteins (Grp78, Chop) related to endoplasmic reticulum stress (ERS) and apoptosis. Moreover, maternal exposure to selenite resulted in the apoptosis of offspring and upregulated the content of ROS and the transcription level of genes related to ERS and apoptosis.
Collapse
Affiliation(s)
- Rui Cheng
- College of Fisheries, Huazhong Agricultural University; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan, 430070, China; Key Laboratory of Ecological Impacts of Hydraulic-Projects and Restoration of Aquatic Ecosystem of Ministry of Water Resources, Institute of Hydroecology, MWR & CAS, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Zhiming Zhang
- Key Laboratory of Ecological Impacts of Hydraulic-Projects and Restoration of Aquatic Ecosystem of Ministry of Water Resources, Institute of Hydroecology, MWR & CAS, Wuhan, 430070, China
| | - Chunhua Zhan
- College of Fisheries, Huazhong Agricultural University; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan, 430070, China
| | - Tianlong Qin
- College of Fisheries, Huazhong Agricultural University; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan, 430070, China
| | - Li Wang
- College of Fisheries, Huazhong Agricultural University; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Xuezhen Zhang
- College of Fisheries, Huazhong Agricultural University; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China.
| |
Collapse
|
43
|
Min X, Zhu Y, Hu Y, Yang M, Yu H, Xiong Y, Fu W, Li J, Matsuda F, Xiong X. Analysis of PPP1R11 expression in granulosa cells during developmental follicles of yak and its effects on cell function. Reprod Domest Anim 2023; 58:129-140. [PMID: 36178063 DOI: 10.1111/rda.14272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/22/2022] [Accepted: 09/24/2022] [Indexed: 01/07/2023]
Abstract
The aims of this study were to analyse the protein phosphatase 1 regulatory subunit 11 (PPP1R11) expression and cellular localization in yak follicles and investigate its effects on cell proliferation, apoptosis and oestrogen secretion in granulosa cells (GCs). Ten healthy and non-pregnant female yaks (4-year-old) were used as experimental animals. The mRNA relative expression level of PPP1R11 in GCs from small (<3.0 mm), medium (3.0-5.9 mm) and large (6.0-9.0 mm) follicles was detected by RT-qPCR, and the cellular localization of PPP1R11 protein was detected by immunohistochemistry staining (IHC). After isolation, culture and identification of yak GCs in vitro, si-PPP1R11 and si-NC (negative control) were transfected into GCs. RT-qPCR and immunofluorescence staining were used to evaluate the interference efficiency, and ELISA was performed to detect oestrogen concentration. Then, EdU staining and TUNEL staining were conducted to analyse cell proliferation and apoptosis. In addition, the oestrogen synthesis, proliferation- and apoptosis-related genes were detected by RT-qPCR after knockdown PPP1R11. The results showed that PPP1R11 is mainly located in ovarian GCs, and the expression levels of PPP1R11 in GCs from large follicles were significantly higher than that from medium and small follicles. Transfection of si-PPP1R11 into GCs could significantly inhibit the expression of PPP1R11. Interestingly, the oestrogen secretion ability and the expression level of oestrogen pathway-related genes (STAR, CYP11A1, CYP19A1 and HSD17B1) were also significantly downregulated. Moreover, the proportion of positive cells was decreased, and cellular proliferation-related genes (PCNA, CCNB1 and CDC25A) were significantly downregulated after knockdown PPP1R11. However, the proportion of apoptotic cells was increased, and apoptosis-related genes (BAX, CASP3 and P53) were significantly upregulated. Taken together, this study was the first revealed the expression and cellular localization of PPP1R11 in yak follicles. Interference PPP1R11 could reduce oestrogen secretion, inhibit proliferation and promote apoptosis in GCs, which provided a basis for further studies on the regulatory mechanism of PPP1R11 in follicle development.
Collapse
Affiliation(s)
- Xingyu Min
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Yanjin Zhu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Yulei Hu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Manzhen Yang
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu, China
| | - Hailing Yu
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu, China
| | - Yan Xiong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu, China.,Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu, China
| | - Wei Fu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Jian Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu, China.,Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu, China
| | - Fuko Matsuda
- Laboratory of Theriogenology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Xianrong Xiong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Southwest Minzu University, Chengdu, China
| |
Collapse
|
44
|
Gupta A, Trigun SK. Cilostamide, a phosphodiesterase 3A inhibitor, sustains meiotic arrest of rat oocytes by modulating cyclic adenosine monophosphate level and the key regulators of maturation promoting factor. J Cell Biochem 2022; 123:2030-2043. [PMID: 36125973 DOI: 10.1002/jcb.30328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/24/2022] [Accepted: 09/05/2022] [Indexed: 12/24/2022]
Abstract
Cilostamide, a phosphodiesterase 3A (Pde3A) inhibitor, is known to increase intraoocyte cyclic adenosine monophosphate (cAMP) level which is involved in sustaining meiotic arrest of the oocytes. To explore the mechanisms involved in the cilostamide-mediated meiotic arrest of the oocytes, the present study describes the effects of cilostamide on cAMP level and related factors involved in maturation of the oocytes at its different meiotic stages; diplotene, metaphase I (MI) and metaphase II (MII). The oocytes from these three stages were collected from rat ovary and incubated with 10 µM cilostamide for 3 h in CO2 incubator. The levels of cAMP, cyclic guanosine monophosphate (cGMP) and the key players of maintaining meiotic arrest during oocyte maturation; Emi2, Apc, Cyclin B1, and Cdk1, were analyzed in diplotene, MI and MII stages. Pde3A was found to be expressed at all three stages but with the lowest level in MI oocyte. As compared to the control sets, the cAMP concentration was found to be highest in MII whereas cGMP was highest in the diplotene stage of cilostamide-treated group. The treated group showed declined reactive oxygen species level as compared with the control counterparts. Relatively increased levels of the Emi2, Cyclin B1, and phosphorylated thr161 of Cdk1 versus declined levels of phosphorylated thr14/tyr15 of Cdk1 in diplotene and MII stage oocytes are known to be involved in maintaining meiotic arrest and all these factors were found to undergo similar pattern of change due to the treatment with cilostamide. The findings thus suggest that cilostamide treatment promotes meiotic arrest by Pde3A inhibition led increase of both cAMP and cGMP level vis-a-vis modulation of the related regulatory factors such as Emi2, CyclinB1, and phosphorylated status of Cdk1 in diplotene and MII stage oocytes. Such a mechanism of meiotic arrest could allow the oocyte to prepare itself for meiotic maturation and thereby to improve oocyte quality.
Collapse
Affiliation(s)
- Anumegha Gupta
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Surendra Kumar Trigun
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
45
|
Dellaqua TT, Vígaro RA, Janini LCZ, Dal Canto M, Renzini MM, Lodde V, Luciano AM, Buratini J. Neuregulin 1 (NRG1) modulates oocyte nuclear maturation during IVM and improves post-IVF embryo development. Theriogenology 2022; 195:209-216. [DOI: 10.1016/j.theriogenology.2022.10.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/31/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
46
|
Gap junctions mediate discrete regulatory steps during fly spermatogenesis. PLoS Genet 2022; 18:e1010417. [PMID: 36174062 PMCID: PMC9578636 DOI: 10.1371/journal.pgen.1010417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 10/18/2022] [Accepted: 09/07/2022] [Indexed: 11/19/2022] Open
Abstract
Gametogenesis requires coordinated signaling between germ cells and somatic cells. We previously showed that Gap junction (GJ)-mediated soma-germline communication is essential for fly spermatogenesis. Specifically, the GJ protein Innexin4/Zero population growth (Zpg) is necessary for somatic and germline stem cell maintenance and differentiation. It remains unknown how GJ-mediated signals regulate spermatogenesis or whether the function of these signals is restricted to the earliest stages of spermatogenesis. Here we carried out comprehensive structure/function analysis of Zpg using insights obtained from the protein structure of innexins to design mutations aimed at selectively perturbing different regulatory regions as well as the channel pore of Zpg. We identify the roles of various regulatory sites in Zpg in the assembly and maintenance of GJs at the plasma membrane. Moreover, mutations designed to selectively disrupt, based on size and charge, the passage of cargos through the Zpg channel pore, blocked different stages of spermatogenesis. Mutations were identified that progressed through early germline and soma development, but exhibited defects in entry to meiosis or sperm individualisation, resulting in reduced fertility or sterility. Our work shows that specific signals that pass through GJs regulate the transition between different stages of gametogenesis.
Collapse
|
47
|
Zhao L, Pan Y, Wang M, Wang J, Wang Y, Han X, Wang J, Zhang T, Zhao T, He H, Cui Y, Yu S. Integrated analysis of the expression profiles of the lncRNA-miRNA-mRNA ceRNA network in granulosa and cumulus cells from yak ovaries. BMC Genomics 2022; 23:633. [PMID: 36057545 PMCID: PMC9441039 DOI: 10.1186/s12864-022-08848-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/16/2022] [Indexed: 11/10/2022] Open
Abstract
Background Growing oocytes acquire the ability to mature through two-way communication between gametes and surrounding somatic cumulus cells (CCs). Granulosa cells (GCs) support oocyte growth, regulate meiosis progression, and modulate global oocyte transcription activity. However, the proliferation and differentiation of the yak ovary in GCs and CCs remain unclear. To characterize the important roles of long non-coding RNA, (lncRNA), microRNA (miRNA), and messenger RNA (mRNA), whole-transcriptome analysis was performed. Real-time quantitative fluorescence PCR was performed to verify the selected RNA sequences. Results Important gene ontology terms and Kyoto Encyclopedia of Genes and Genomes pathways related to differentiation and oocyte development were identified for the target genes of differentially expressed lncRNAs, miRNAs, and mRNAs. In total,6223 mRNAs (2197 upregulated, 4026 downregulated), 643 lncRNAs (204 upregulated, 479 downregulated), and 559 miRNAs (311 upregulated, 248 downregulated) were significantly altered between the two groups. Target genes involved in cell adhesion, cell differentiation, regulation of developmental processes, cell proliferation, embryo development, signal transduction, apoptosis, and aromatic compound biosynthetic processes were significantly enriched. These RNAs were involved in ECM-receptor interaction, MAPK signaling, Hippo signaling, PI3K-Akt signaling, cell cycle, cell adhesion, leukocyte trans-endothelial migration, and actin cytoskeleton regulation. Conclusions A comprehensive analysis of the co-expression network of competing endogenous RNAs (ceRNAs) will facilitate the understanding of the process of granulosa cell proliferation and differentiation and offer a theoretical basis for the development of oocytes. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08848-3.
Collapse
Affiliation(s)
- Ling Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Yangyang Pan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Meng Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Junqian Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Yaying Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xiaohong Han
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jinglei Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Tongxiang Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Tian Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Honghong He
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Yan Cui
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Sijiu Yu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China. .,Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Lanzhou, 730070, China.
| |
Collapse
|
48
|
Read CC, Edwards JL, Schrick FN, Rhinehart JD, Payton RR, Campagna SR, Castro HF, Klabnik JL, Moorey SE. Preovulatory serum estradiol concentration is positively associated with oocyte ATP and follicular fluid metabolite abundance in lactating beef cattle. J Anim Sci 2022; 100:6620784. [PMID: 35772749 PMCID: PMC9246671 DOI: 10.1093/jas/skac136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/08/2022] [Indexed: 12/13/2022] Open
Abstract
Cattle induced to ovulate a small, physiologically immature preovulatory follicle had reduced oocyte developmental competence that resulted in decreased embryo cleavage and day 7 embryo quality compared with animals induced to ovulate a more advanced follicle. RNA-sequencing was performed on oocytes and their corresponding cumulus cells approximately 23 h after gonadotropin-releasing hormone (GnRH) administration to induce the preovulatory gonadotropin surge suggested reduced capacity for glucose metabolism and oxidative phosphorylation in the cumulus cells and oocytes from follicles ≤11.7 mm, respectively. We hypothesized that induced ovulation of a small, physiologically immature preovulatory follicle results in a suboptimal follicular microenvironment and reduced oocyte metabolic capacity. We performed a study with the objective to determine the impact of preovulatory follicle diameter and serum estradiol concentration at GnRH administration on oocyte metabolic competence and follicular fluid metabolome profiles. We synchronized the development of a preovulatory follicle and collected the follicle contents via transvaginal aspiration approximately 19 h after GnRH administration in lactating beef cows (n = 319). We determined ATP levels and mitochondrial DNA (mtDNA) copy number in 110 oocytes and performed ultra-high-performance liquid chromatography–high resolution mass spectrometry metabolomic studies on 45 follicular fluid samples. Intraoocyte ATP and the amount of ATP produced per mtDNA copy number were associated with serum estradiol concentration at GnRH and time from GnRH administration to follicle aspiration (P < 0.05). mtDNA copy number was not related to follicle diameter at GnRH, serum estradiol concentration at GnRH, or any potential covariates (P > 0.10). We detected 90 metabolites in the aspirated follicular fluid. We identified 22 metabolites associated with serum estradiol concentration at GnRH and 63 metabolites associated with follicular fluid progesterone concentration at the time of follicle aspiration (FDR < 0.10). Pathway enrichment analysis of significant metabolites suggested altered proteinogenesis, citric acid cycle, and pyrimidine metabolism in follicles of reduced estrogenic capacity pre-gonadotropin surge or reduced progesterone production by the time of follicle aspiration.
Collapse
Affiliation(s)
- Casey C Read
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - J Lannett Edwards
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - F Neal Schrick
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Justin D Rhinehart
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Rebecca R Payton
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Shawn R Campagna
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, USA
| | - Hector F Castro
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, USA
| | - Jessica L Klabnik
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Sarah E Moorey
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
49
|
Moorey SE, Hessock EA, Edwards JL. Preovulatory follicle contributions to oocyte competence in cattle: importance of the ever-evolving intrafollicular environment leading up to the luteinizing hormone surge. J Anim Sci 2022; 100:skac153. [PMID: 35772757 PMCID: PMC9246662 DOI: 10.1093/jas/skac153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/25/2022] [Indexed: 12/13/2022] Open
Abstract
The preovulatory intrafollicular environment plays a major role in determining oocyte competence. The basis of this review is to highlight the importance of the preovulatory follicle's physiological status prior to the preovulatory luteinizing hormone (LH) surge and onset of oocyte maturation to promote an optimal follicular microenvironment and optimal oocyte developmental competence in cattle. While the underlying mechanisms remain unclear, and are likely multifactorial, the preovulatory follicle's physiological status prior to the preovulatory LH surge is highly influential on the oocyte's capacity to undergo postfertilization embryo development. Changes in the intrafollicular environment of the preovulatory follicle including steroid hormone production, metabolome profiles, and proteome profiles likely support the oocyte's developmental and metabolic competency. This review focuses on the relationship between bovine oocyte developmental competency and antral follicle progression to the preovulatory phase, the role of the preovulatory follicle in improving oocyte developmental competence in cattle, and the importance of the ever-evolving preovulatory intrafollicular environment for optimal fertility.
Collapse
Affiliation(s)
- Sarah E Moorey
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Emma A Hessock
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - J Lannett Edwards
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
50
|
Sohel MMH, Hoelker M, Schellander K, Tesfaye D. The extent of the abundance of exosomal and non-exosomal extracellular miRNAs in the bovine follicular fluid. Reprod Domest Anim 2022; 57:1208-1217. [PMID: 35765751 DOI: 10.1111/rda.14195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/30/2022] [Accepted: 06/17/2022] [Indexed: 11/27/2022]
Abstract
Follicular fluid (FF) plays an important role during follicular development and it contains several bioactive molecules including extracellular microRNAs (ECmiRNAs) that may mediate cell-cell communication during follicular development. Yet, the distribution patterns of ECmiRNAs in FF is not well characterized. This study aims to investigate the distribution of ECmiRNAs in two major fractions, namely exosomal and non-exosomal, of bovine follicular fluid (bFF). Exosomal and non-exosomal fractions from bFF were separated using Exoquick™ exosomes precipitation kit. miRNA expression was evaluated using the human miRCURY LNA™ Universal RT miRNA PCR array system. Transmission electron microscopy and immunoblotting revealed that the isolated vesicles were exosomes. The real-time PCR-based expression analysis revealed that 516 miRNAs were detected in the exosomal fraction of bFF, while 393 miRNAs were detected in the non-exosomal fraction. Among the detected miRNAs, a total of 370 miRNAs were detected in both fractions, while 145 miRNAs and 23 miRNAs were solely detected in exosomal and non-exosomal fractions, respectively. Exploratory pathway analysis showed that the genes targeted by exosomal and non-exosomal miRNAs to be involved in MAPK, Wnt, FoxO, TGF-beta, Oxytocin, ErbB, PI3K-Akt, Neurotrophin signalling pathways which are believed to be involved in follicular development, cell proliferation, and meiotic resumption. The results of our study demonstrated that besides the exosomal fraction, non-exosomal fractions can carry a significant amount of miRNAs in bFF where the exosomal fraction carries a significantly higher number of detectable miRNAs.
Collapse
Affiliation(s)
- Md Mahmodul Hasan Sohel
- Department of Life Sciences, School of Environment and Life Sciences, Independent University, Bangladesh.,Animal Breeding and Husbandry Group, Institute of Animal Science, University of Bonn, Bonn, Germany.,Department of Genetics, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Michael Hoelker
- Animal Breeding and Husbandry Group, Institute of Animal Science, University of Bonn, Bonn, Germany
| | - Karl Schellander
- Animal Breeding and Husbandry Group, Institute of Animal Science, University of Bonn, Bonn, Germany
| | - Dawit Tesfaye
- Animal Breeding and Husbandry Group, Institute of Animal Science, University of Bonn, Bonn, Germany.,Department of Biomedical Sciences, Animal Reproduction and Biotechnology Lab, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|