1
|
Chen Y, Ali W, Men Y, Yan K, Li Z, Cai W, He Y, Qi J. Molecular insights into oocyte development and sperm storage in black rockfish (Sebastes schlegelii): Proteomic changes across reproductive stages. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 53:101368. [PMID: 39612540 DOI: 10.1016/j.cbd.2024.101368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/01/2024]
Abstract
Sperm storage in females is widespread among vertebrates and insects, and the expression of proteins in the female reproductive tract is influenced by the presence of sperm, allowing for adaptation to this phenomenon. Through histological observation, we confirmed that sperm were stored in the isthmic fossa outside the oocyte during the post-mating (POM) stage, and closer to the epithelial cells during the pre-fertilization (PRF) stage. In addition, we observed asynchronous ovarian development in black rockfish, where oocytes at various stages could be identified during the PRF phase. This study investigated the ovarian protein expression changes in black rockfish (Sebastes schlegelii) during key reproductive stages: pre-mating (PRM), POM, unmated control (POM-CT), and PRF. A total of 5012 proteins were identified, with notable fluctuations in protein expression observed at the PRF stage. Specifically, 140 proteins were upregulated and 615 downregulated when compared to the PRM stage, while 101 proteins were upregulated and 531 downregulated in comparison to the POM stage. The functional enrichment analysis of differentially expressed proteins (DEPs) revealed distinct pathways: POM vs. PRM showed involvement in vesicle sorting and hormone signaling; PRF vs. POM indicated pathways related to chromatin remodeling and gene expression regulation; and POM vs. POM-CT highlighted pathways associated with immune response. These findings suggested that these signaling pathways may play a crucial role in oocyte development and sperm storage. The majority of DEPs were localized in the nucleus, with key interactions involving proteins such as GSK3B and MED1. These findings enhance our understanding of the molecular mechanisms underlying oocyte maturation and sperm storage, providing insights relevant to reproductive biology and aquaculture practices.
Collapse
Affiliation(s)
- Ying Chen
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China
| | - Wajid Ali
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China
| | - Yu Men
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China
| | - Kai Yan
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China
| | - Zibin Li
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China
| | - Wenxiu Cai
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China
| | - Yan He
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China
| | - Jie Qi
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China.
| |
Collapse
|
2
|
Bogolyubova I, Salimov D, Bogolyubov D. Chromatin Configuration in Diplotene Mouse and Human Oocytes during the Period of Transcriptional Activity Extinction. Int J Mol Sci 2023; 24:11517. [PMID: 37511273 PMCID: PMC10380668 DOI: 10.3390/ijms241411517] [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: 06/28/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
In the oocyte nucleus, called the germinal vesicle (GV) at the prolonged diplotene stage of the meiotic prophase, chromatin undergoes a global rearrangement, which is often accompanied by the cessation of its transcriptional activity. In many mammals, including mice and humans, chromatin condenses around a special nuclear organelle called the atypical nucleolus or formerly nucleolus-like body. Chromatin configuration is an important indicator of the quality of GV oocytes and largely predicts their ability to resume meiosis and successful embryonic development. In mice, GV oocytes are traditionally divided into the NSN (non-surrounded nucleolus) and SN (surrounded nucleolus) based on the specific chromatin configuration. The NSN-SN transition is a key event in mouse oogenesis and the main prerequisite for the normal development of the embryo. As for humans, there is no single nomenclature for the chromatin configuration at the GV stage. This often leads to discrepancies and misunderstandings, the overcoming of which should expand the scope of the application of mouse oocytes as a model for developing new methods for assessing and improving the quality of human oocytes. As a first approximation and with a certain proviso, the mouse NSN/SN classification can be used for the primary characterization of human GV oocytes. The task of this review is to analyze and discuss the existing classifications of chromatin configuration in mouse and human GV oocytes with an emphasis on transcriptional activity extinction at the end of oocyte growth.
Collapse
Affiliation(s)
- Irina Bogolyubova
- Institute of Cytology of the Russian Academy of Sciences, 194064 St. Petersburg, Russia
| | - Daniil Salimov
- Clinical Institute of Reproductive Medicine, 620014 Yekaterinburg, Russia
| | - Dmitry Bogolyubov
- Institute of Cytology of the Russian Academy of Sciences, 194064 St. Petersburg, Russia
| |
Collapse
|
3
|
Ergun S, Gunes S, Hekim N, Esteves SC. In silico analysis of microRNA genes in azoospermia factor Y-chromosome microdeletions. Int Urol Nephrol 2022; 54:773-780. [DOI: 10.1007/s11255-022-03133-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 01/24/2022] [Indexed: 12/01/2022]
|
4
|
Akula SM, Bolin P, Cook PP. Cellular miR-150-5p may have a crucial role to play in the biology of SARS-CoV-2 infection by regulating nsp10 gene. RNA Biol 2021; 19:1-11. [PMID: 34904915 PMCID: PMC8786335 DOI: 10.1080/15476286.2021.2010959] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The role for circulating miRNAs as biomarkers of the COVID-19 disease remains uncertain. We analysed the circulating miRNA profile in twelve COVID-19 patients with moderate-severe disease. This analysis was conducted by performing next generation sequencing (NGS) followed by real-time polymerase chain reaction (RT-qPCR). Compared with healthy controls, we detected significant changes in the circulating miRNA profile of COVID-19 patients. The miRNAs that were significantly altered in all the COVID-19 patients were miR-150-5p, miR-375, miR-122-5p, miR-494-3p, miR-3197, miR-4690-5p, miR-1915-3p, and miR-3652. Infection assays performed using miRNA mimics in HEK-293 T cells determined miR-150-5p to have a crucial role in SARS-CoV-2 infection and this was based on the following data: (i) miR-150-5p mimic lowered in vitro SARS-CoV-2 infection; (ii) miR-150-5p inhibitor reversed the effects of miR-150-5p mimic on SARS-CoV-2 infection of cells; and (iii) a novel miRNA recognition element (MRE) was identified in the coding strand of SARS-CoV-2 nsp10, the expression of which could be inhibited by miR-150-5p mimic. Our findings identified crucial miRNA footprints in COVID-19 patients with moderate-severe disease. A combination of co-transfection and Western blotting experiments also determined the ability of miR-150-5p to inhibit SARS-CoV-2 infection via directly interacting with MRE in the coding strand of nsp10. Our investigation showed that a sharp decline in the miR-150-5p plasma levels in COVID-19 patients may support enhanced SARS-CoV-2 infection. Furthermore, this study provides insight into one possible mechanism by which COVID-19-induced changes to miR-150-5p levels may promote SARS-CoV-2 infection via modulating nsp10 expression.
Collapse
Affiliation(s)
- Shaw M Akula
- Department of Microbiology & Immunology (S.m. Akula), Department of Internal Medicine (P. Bolin, P.P.Cook), Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Paul Bolin
- Department of Microbiology & Immunology (S.m. Akula), Department of Internal Medicine (P. Bolin, P.P.Cook), Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Paul P Cook
- Department of Microbiology & Immunology (S.m. Akula), Department of Internal Medicine (P. Bolin, P.P.Cook), Brody School of Medicine at East Carolina University, Greenville, NC, USA
| |
Collapse
|
5
|
Stabile M, Colavito D, Del Giudice E, Rispoli AF, Ingenito MC, Naumova AK. A novel exomal ATRX mutation with preferential transmission to offspring: A case report and review of the literature for transmission ratio distortion in ATRX families. Mol Med Rep 2020; 22:4561-4566. [PMID: 33173999 PMCID: PMC7646821 DOI: 10.3892/mmr.2020.11574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 07/21/2020] [Indexed: 12/19/2022] Open
Abstract
The present case report describes an Italian family with three affected probands, who exhibited serious mental disability, which has not been associated with other anomalies, except with slight facial dysmorphism. Molecular multigenic analysis for intellectual disability identified a previously unreported variant, p.Ile1765Met (c.5295C>G) in the SNF domain of the ATRX protein (in exon 24). The identified mutation was found in a hemizygous state in all three affected probands and in a heterozygous state in the asymptomatic mother and the female sibling. With respect to the phenotypic similarities found in the patients with those described in previous studies, the consistency in the mode of inheritance and segregation of the mutation, the variant reported in the present case report may be considered as ‘likely pathogenic’. To investigate the hypothesis that the preferential transmission of the ATRX mutation observed in this family reflected a general trend, a meta-analysis into the segregation of ATRX mutations from published pedigrees, following allelic transmission from mothers who are heterozygous carriers to their offspring, was performed. A preferential transmission of the mutant allele to male offspring (58% of males inherited the mutant allele) was found; however, the bias was not statistically significant (P=0.29; χ2 test).
Collapse
Affiliation(s)
- Mariano Stabile
- Zygote Center, Center for Genetics, Prenatal Diagnosis, Fertility, I-84131 Salerno, Italy
| | - Davide Colavito
- Research & Innovation s.r.l. (R&I Genetics), I-35127 Padova, Italy
| | - Elda Del Giudice
- Research & Innovation s.r.l. (R&I Genetics), I-35127 Padova, Italy
| | - Anna F Rispoli
- Zygote Center, Center for Genetics, Prenatal Diagnosis, Fertility, I-84131 Salerno, Italy
| | - Marina C Ingenito
- Zygote Center, Center for Genetics, Prenatal Diagnosis, Fertility, I-84131 Salerno, Italy
| | - Anna K Naumova
- Departments of Obstetrics and Gynecology and Human Genetics, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Quebec H4A 3J1, Canada
| |
Collapse
|
6
|
Bogolyubova I, Bogolyubov D. Heterochromatin Morphodynamics in Late Oogenesis and Early Embryogenesis of Mammals. Cells 2020; 9:cells9061497. [PMID: 32575486 PMCID: PMC7348780 DOI: 10.3390/cells9061497] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/10/2020] [Accepted: 06/16/2020] [Indexed: 12/15/2022] Open
Abstract
During the period of oocyte growth, chromatin undergoes global rearrangements at both morphological and molecular levels. An intriguing feature of oogenesis in some mammalian species is the formation of a heterochromatin ring-shaped structure, called the karyosphere or surrounded "nucleolus", which is associated with the periphery of the nucleolus-like bodies (NLBs). Morphologically similar heterochromatin structures also form around the nucleolus-precursor bodies (NPBs) in zygotes and persist for several first cleavage divisions in blastomeres. Despite recent progress in our understanding the regulation of gene silencing/expression during early mammalian development, as well as the molecular mechanisms that underlie chromatin condensation and heterochromatin structure, the biological significance of the karyosphere and its counterparts in early embryos is still elusive. We pay attention to both the changes of heterochromatin morphology and to the molecular mechanisms that can affect the configuration and functional activity of chromatin. We briefly discuss how DNA methylation, post-translational histone modifications, alternative histone variants, and some chromatin-associated non-histone proteins may be involved in the formation of peculiar heterochromatin structures intimately associated with NLBs and NPBs, the unique nuclear bodies of oocytes and early embryos.
Collapse
|
7
|
O'Shea LC, Fair T, Hensey C. X-linked α-thalassemia with mental retardation is downstream of protein kinase A in the meiotic cell cycle signaling cascade in Xenopus oocytes and is dynamically regulated in response to DNA damage†. Biol Reprod 2020; 100:1238-1249. [PMID: 30649195 DOI: 10.1093/biolre/ioz001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 10/19/2018] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
X-linked α-thalassemia with mental retardation (ATRX) is a chromatin remodeling protein that belongs to the SWItch/sucrose non-fermentable (SWI2/SNF2) family of helicase/ATPases. During meiosis, ATRX is necessary for heterochromatin formation and maintenance of chromosome stability in order to ensure proper assembly of the metaphase II spindle. Previously, we established ATRX as a novel progesterone regulated protein during bovine meiotic maturation, in addition to being dynamically regulated in response to DNA damage in oocytes. In the present study, we utilize the Xenopus laevis model system to further elucidate the signaling pathways regulating ATRX expression within the oocyte. Here, we present an analysis of endogenous ATRX protein expression during oogenesis, oocyte meiotic maturation, and early embryonic development. ATRX expression is dynamically regulated as evidenced by loss of the protein in metaphase II of meiosis. The downstream activation of meiosis via protein kinase A inhibition resulted in a similar decrease in ATRX protein expression. We demonstrate that the ATRX protein is detected in ubiquitin immuno-precipitates from germinal vesicle oocyte extracts and experimentally demonstrate that proteosomal degradation is responsible for the decreased expression of ATRX during meiosis. ATRX expression is significantly increased in response to gamma-irradiation induced DNA damage in oocytes and embryos. This increased expression is independent of p53 protein expression in apoptotic embryos, as determined by the expression of active caspase-3. Thus, regulation of ATRX protein expression impacts on G2-M progression and ultimately has consequences for cell survival.
Collapse
Affiliation(s)
| | - Trudee Fair
- UCD School of Agriculture and Food Science, Dublin, Ireland
| | - Carmel Hensey
- UCD School of Bimolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
8
|
Bogolyubova IO, Sailau ZK, Bogolyubov DS. The dynamics of DAXX protein distribution in the nucleus of mouse early embryos. Acta Histochem 2019; 121:522-529. [PMID: 31029404 DOI: 10.1016/j.acthis.2019.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 02/02/2023]
Abstract
Nuclear distribution of Death-associated protein 6 (Daxx) was studied using fluorescent and electron microscopy in mouse embryos at different stages of development in vivo, from zygote to morula. Daxx was found in association with transcriptionally silent chromatin predominantly with a heterochromatin rim surrounding the nucleolus precursor bodies (NPBs) at all stages studied. At the zygote stage, Daxx was detected only at the periphery of NPBs both in male and female pronuclei. At the late two-cell stage, Daxx was localized not only in the heterochromatin rim at the periphery of NPBs but also in heterochromatin zones not associated with NPBs. At the morula stage, a diffuse distribution of Daxx prevailed. Scarce Daxx-positive zones were detected only in some embryos at the nucleolar periphery. Thus, Daxx is noticeably redistributed during mouse embryo cleavage, and the most conspicuous areas of Daxx concentration are observed at the end of two-cell stage. Daxx is found colocalized with the chromatin-remodeling protein ATRX exclusively in two-cell embryos, but the heterochromatin areas containing either Daxx or ATRX individually are also observed at this stage. However, most zones containing both Daxx and ATRX demonstrated a low FRET-efficiency. This suggest that two molecules are not approached sufficiently close for molecular interactions to occur. Our data suggests that Daxx may function without cooperation with ATRX at least at some stages of early mouse development.
Collapse
Affiliation(s)
- Irina O Bogolyubova
- Laboratory of Cell Morphology, Institute of Cytology of the Russian Academy of Science, 4 Tikhoretsky Ave., 194064 St. Petersburg, Russia.
| | - Zhuldyz K Sailau
- St. Petersburg State University, 7/9 Universitetskaya Emb., 199034 St. Petersburg, Russia
| | - Dmitry S Bogolyubov
- Laboratory of Cell Morphology, Institute of Cytology of the Russian Academy of Science, 4 Tikhoretsky Ave., 194064 St. Petersburg, Russia
| |
Collapse
|
9
|
Bogolyubov DS. Karyosphere (Karyosome): A Peculiar Structure of the Oocyte Nucleus. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 337:1-48. [PMID: 29551157 DOI: 10.1016/bs.ircmb.2017.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The karyosphere, aka the karyosome, is a meiosis-specific structure that represents a "knot" of condensed chromosomes joined together in a limited volume of the oocyte nucleus. The karyosphere is an evolutionarily conserved but morphologically rather "multifaceted" structure. It forms at the diplotene stage of meiotic prophase in many animals, from hydra and Drosophila to human. Karyosphere formation is generally linked with transcriptional silencing of the genome. It is believed that karyosphere/karyosome is a prerequisite for proper completion of meiotic divisions and further development. Here, a brief review on the karyosphere features in some invertebrates and vertebrates is provided. Special emphasis is made on terminology, since current discrepancies in this field may lead to confusions. In particular, it is proposed to distinguish the karyosphere with a capsule and the karyosome (a karyosphere devoid of a capsule). The "inverted" karyospheres are also considered, in which the chromosomes situate externally to an extrachromosomal structure (e.g., in human oocytes).
Collapse
Affiliation(s)
- Dmitry S Bogolyubov
- Institute of Cytology of the Russian Academy of Science, St. Petersburg, Russia.
| |
Collapse
|
10
|
Ilicheva N, Podgornaya O, Bogolyubov D, Pochukalina G. The karyosphere capsule in Rana temporaria oocytes contains structural and DNA-binding proteins. Nucleus 2018; 9:516-529. [PMID: 30272509 PMCID: PMC6244735 DOI: 10.1080/19491034.2018.1530935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 09/21/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022] Open
Abstract
During the last stages of oogenesis, oocyte chromosomes condense and come close together, forming the so-called karyosphere. Karyosphere formation is accompanied by an essential decrease in transcriptional activity. In the grass frog Rana temporaria, the karyosphere is surrounded by an extrachromosomal capsule that separates the chromosomes from the rest of the nucleoplasm. The karyosphere capsule (KC) of R. temporaria has been investigated in detail at the ultrastructural level, but its protein composition remained largely unknown. We demonstrate here that nuclear actin, especially F-actin, as well as lamins A/C and B are the most abundant proteins of the KC. Key proteins of nuclear pore complexes, such as Nup93 and Nup35, are also detectable in the KC. New antibodies recognizing the telomere-binding protein TRF2 allowed us to localize TRF2 in nuclear speckles. We also found that the R. temporaria KC contains some proteins involved in chromatin remodeling, including topoisomerase II and ATRX. Thus, we believe that KC isolates the chromosomes from the rest of the nucleoplasm during the final period of oocyte growth (late diplotene) and represents a specialized oocyte nuclear compartment to store a variety of factors involved in nuclear metabolism that can be used in future early development. Abbreviations: BrUTP: 5-bromouridine 5'-triphosphate; CytD: cytochalasin D; IGCs: interchromatin granule clasters; IgG: immunoglobulin G; KC: karyosphere capsule; Mw: molecular weight; NE: nuclear envelope; PBS: phosphate buffered saline; SDS-PAGE: sodium dodecyl sulfate polyacrylamide gel electrophoresis; Topo II: topoisomerase II.
Collapse
Affiliation(s)
- Nadya Ilicheva
- Laboratory of Cell Morphology, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Olga Podgornaya
- Laboratory of Cell Morphology, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
- Department of Cytology and Histology, Faculty of Biology, Saint Petersburg State University, St. Petersburg, Russia
- Laboratory of Biomedical Cell Technology, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Dmitry Bogolyubov
- Laboratory of Cell Morphology, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Galina Pochukalina
- Laboratory of Cell Morphology, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
11
|
Stepanova IS, Bogolyubov DS. Localization of the chromatin-remodeling protein ATRX in the oocyte nucleus of some insects. ACTA ACUST UNITED AC 2017. [DOI: 10.1134/s1990519x17050091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
12
|
O'Shea LC, Daly E, Hensey C, Fair T. ATRX is a novel progesterone-regulated protein and biomarker of low developmental potential in mammalian oocytes. Reproduction 2017; 153:671-682. [PMID: 28250240 DOI: 10.1530/rep-16-0443] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 02/03/2017] [Accepted: 02/28/2017] [Indexed: 01/09/2023]
Abstract
A multi-species meta-analysis of published transcriptomic data from models of oocyte competence identified the chromatin remodelling factor ATRX as a putative biomarker of oocyte competence. The objective of the current study was to test the hypothesis that ATRX protein expression by cumulus-oocyte complexes (COCs) reflects their intrinsic quality and developmental potential. In excess of 10,000 bovine COCs were utilised to test our hypothesis. COCs were in vitro matured (IVM) under conditions associated with reduced developmental potential: IVM in the presence or absence of (1) progesterone synthesis inhibitor (Trilostane); (2) nuclear progesterone receptor inhibitor (Aglepristone) or (3) an inducer of DNA damage (Staurosporine). ATRX protein expression and localisation were determined using immunocytochemistry and Western blot analysis. A proportion of COCs matured in the presence or absence of Trilostane was in vitro fertilised and cultured, and subsequent embryo development characteristics were analysed. In addition, ATRX expression was investigated in 40 human germinal vesicle-stage COCs. Our results showed that ATRX is expressed in human and bovine germinal vesicle oocytes and cumulus cells. In bovine, expression decreases after IVM. However, this decline is not observed in COCs matured under sub-optimal conditions. Blastocyst development rate and cell number are decreased, whereas the incidence of abnormal metaphase phase spindle and chromosome alignment are increased, after IVM in the presence of Trilostane (P < 0.05). In conclusion, localisation of ATRX to the cumulus cell nuclei and oocyte chromatin, after IVM, is associated with poor oocyte quality and low developmental potential. Furthermore, ATRX is dynamically regulated in response to progesterone signalling.
Collapse
Affiliation(s)
- Lynne C O'Shea
- School of Agriculture and Food Sciences .,School of Medicine
| | | | - Carmel Hensey
- School of Bimolecular and Biomedical ScienceUniversity College Dublin, Belfield, Dublin 4, Ireland
| | | |
Collapse
|
13
|
Nuclear distribution of the chromatin-remodeling protein ATRX in mouse early embryogenesis. Acta Histochem 2017; 119:18-25. [PMID: 27863708 DOI: 10.1016/j.acthis.2016.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/03/2016] [Accepted: 11/03/2016] [Indexed: 12/13/2022]
Abstract
The nucleus of mammalian embryos differs by transcriptional activity at different stages of early development. Here, we studied nuclear distribution of the chromatin-remodeling protein ATRX in pre-implantation mouse embryos. Immunofluorescent staining revealed the changes of ATRX nuclear distribution at the initial stages of early mouse development. At the stage of early zygote, a diffuse ATRX distribution pattern was prevalent. During the course of zygotic genome activation (ZGA), zones of increased ATRX concentration are observed, and they are most expressed in the nuclei of late 2-cell embryos. In the morula stage, the ATRX distribution becomes diffuse again. In zygotes, the patterns of ATRX distribution differ between male and female pronuclei. At all the stages, ATRX concentrates in the DAPI-positive areas of condensed chromatin. The level of colocalization between ATRX and heterochromatin was found the highest at the late 2-cell stage. When transcription was artificially suppressed, the pattern of intranuclear ATRX distribution was mostly determined by the mechanism of inhibitor action rather than the decreased level of transcriptional activity. Thus, the obvious changes of ATRX distribution occur and partially correlate with the main stages of ZGA during mouse early development, but these changes seem to be determined by other processes of structural and functional rearrangements of blastomere nuclei.
Collapse
|
14
|
The relationship between apoptosis, chromatin configuration, histone modification and competence of oocytes: A study using the mouse ovary-holding stress model. Sci Rep 2016; 6:28347. [PMID: 27321442 PMCID: PMC4913248 DOI: 10.1038/srep28347] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 06/01/2016] [Indexed: 12/31/2022] Open
Abstract
The epigenetic factors causing competence differences between SN (surrounded nucleolus) and NSN (non-surrounded nucleolus) oocytes, the significance for the increased histone acetylation and methylation in SN oocytes, and whether chromatin configuration or histone modification determines oocyte competence, are unclear. This study has addressed these issues by using the ovary-holding (OH) stress models where oocyte SN configuration was uncoupled from histone modifications and developmental potential. Prepubertal mouse ovaries containing high percentages of NSN oocytes were preserved at 37 or 39 °C for 1 or 2 h before examination for oocyte chromatin configuration, developmental competence, histone modification and apoptosis. Whereas 1-h OH at 37 °C caused a moderate apoptosis with increased oocyte competence, improved histone modification and a normal NSN-to-SN transition, harsher OH conditions induced a severe apoptosis with decreased oocyte competence, impaired histone modification and a pseudo (premature) NSN-to-SN transition. Observations on Fas/FasL expression and using the gld (generalized lymphoproliferative disorder) mice harboring FasL mutations indicated that OH triggered oocyte apoptosis with activation of the Fas signaling. It was concluded that OH stress caused oocyte apoptosis with activation of the Fas/FasL system and that oocyte competence was more closely correlated with histone modification than with chromatin configuration.
Collapse
|
15
|
An essential role for the intra-oocyte MAPK activity in the NSN-to-SN transition of germinal vesicle chromatin configuration in porcine oocytes. Sci Rep 2016; 6:23555. [PMID: 27009903 PMCID: PMC4806380 DOI: 10.1038/srep23555] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/08/2016] [Indexed: 02/07/2023] Open
Abstract
The mechanisms for the transition from non-surrounded nucleolus (NSN) to surrounded nucleolus (SN) chromatin configuration during oocyte growth/maturation are unclear. By manipulating enzyme activities and measuring important molecules using small-follicle pig oocytes with a high proportion of NSN configuration and an extended germinal vesicle stage in vitro, this study has the first time up-to-date established the essential role for intra-oocyte mitogen-activated protein kinase (MAPK) in the NSN-to-SN transition. Within the oocyte in 1–2 mm follicles, a cAMP decline activates MAPK, which prevents the NSN-to-SN transition by activating nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) while inhibiting histone deacetylase (HDAC). In cumulus cells of 1–2 mm follicles, a lower level of estradiol and oocyte-derived paracrine factor (ODPF) reduces natriuretic peptide receptor 2 (NPR2) while enhancing FSH and cAMP actions. FSH elevates cAMP levels, which decreases NPR2 while activating MAPK. MAPK closes the gap junctions, which, together with the NPR2 decrease, reduces cyclic guanosine monophosphate (cGMP) delivery leading to the cAMP decline within oocytes. In 3–6 mm follicles, a higher level of estradiol and ODPF and a FSH shortage initiate a reversion of the above events leading to MAPK inactivation and NSN-to-SN transition within oocytes.
Collapse
|
16
|
Trapphoff T, Heiligentag M, Dankert D, Demond H, Deutsch D, Fröhlich T, Arnold GJ, Grümmer R, Horsthemke B, Eichenlaub-Ritter U. Postovulatory aging affects dynamics of mRNA, expression and localization of maternal effect proteins, spindle integrity and pericentromeric proteins in mouse oocytes. Hum Reprod 2016; 31:133-49. [PMID: 26577303 PMCID: PMC5853592 DOI: 10.1093/humrep/dev279] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/06/2015] [Accepted: 10/13/2015] [Indexed: 12/20/2022] Open
Abstract
STUDY QUESTION Is the postovulatory aging-dependent differential decrease of mRNAs and polyadenylation of mRNAs coded by maternal effect genes associated with altered abundance and distribution of maternal effect and RNA-binding proteins (MSY2)? SUMMARY ANSWER Postovulatory aging results in differential reduction in abundance of maternal effect proteins, loss of RNA-binding proteins from specific cytoplasmic domains and critical alterations of pericentromeric proteins without globally affecting protein abundance. WHAT IS KNOWN ALREADY Oocyte postovulatory aging is associated with differential alteration in polyadenylation and reduction in abundance of mRNAs coded by selected maternal effect genes. RNA-binding and -processing proteins are involved in storage, polyadenylation and degradation of mRNAs thus regulating stage-specific recruitment of maternal mRNAs, while chromosomal proteins that are stage-specifically expressed at pericentromeres, contribute to control of chromosome segregation and regulation of gene expression in the zygote. STUDY DESIGN, SIZE, DURATION Germinal vesicle (GV) and metaphase II (MII) oocytes from sexually mature C57B1/6J female mice were investigated. Denuded in vivo or in vitro matured MII oocytes were postovulatory aged and analyzed by semiquantitative confocal microscopy for abundance and localization of polyadenylated RNAs, proteins of maternal effect genes (transcription activator BRG1 also known as ATP-dependent helicase SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4 (SMARCA4) and NOD-like receptor family pyrin domain containing 5 (NLRP5) also known as MATER), RNA-binding proteins (MSY2 also known as germ cell-specific Y-box-binding protein, YBX2), and post-transcriptionally modified histones (trimethylated histone H3K9 and acetylated histone H4K12), as well as pericentromeric ATRX (alpha thalassemia/mental retardation syndrome X-linked, also termed ATP-dependent helicase ATRX or X-linked nuclear protein (XNP)). For proteome analysis five replicates of 30 mouse oocytes were analyzed by selected reaction monitoring (SRM). MATERIAL AND METHODS GV and MII oocytes were obtained from large antral follicles or ampullae of sexually mature mice, respectively. Denuded MII oocytes were aged for 24 h post ovulation. For analysis of distribution and abundance of polyadenylated RNAs fixed oocytes were in situ hybridized to Cy5 labeled oligo(dT)20 nucleotides. Absolute quantification of protein concentration per oocyte of selected proteins was done by SRM proteome analysis. Relative abundance of ATRX was assessed by confocal laser scanning microscopy (CLSM) of whole mount formaldehyde fixed oocytes or after removal of zona and spreading. MSY2 protein distribution and abundance was studied in MII oocytes prior to, during and after exposure to nocodazole, or after aging for 2 h in presence of H2O2 or for 24 h in presence of a glutathione donor, glutathione ethylester (GEE). MAIN RESULTS AND ROLE OF CHANCE The significant reduction in abundance of proteins (P < 0.001) translated from maternal mRNAs was independent of polyadenylation status, while their protein localization was not significantly changed by aging. Most of other proteins quantified by SRM analysis did not significantly change in abundance upon aging except MSY2 and GTSF1. MSY2 was enriched in the subcortical RNP domain (SCRD) and in the spindle chromosome complex (SCC) in a distinct pattern, right and left to the chromosomes. There was a significant loss of MSY2 from the SCRD (P < 0.001) and the spindle after postovulatory aging. Microtubule de- and repolymerization caused reversible loss of MSY2 spindle-association whereas H2O2 stress did not significantly decrease MSY2 abundance. Aging in presence of GEE decreased significantly (P < 0.05) the aging-related overall and cytoplasmic loss of MSY2. Postovulatory aging increased significantly spindle abnormalities, unaligned chromosomes, and abundance of acetylated histone H4K12, and decreased pericentromeric trimethylated histone H3K9 (all P < 0.001). Spreading revealed a highly significant increase in pericentromeric ATRX (P < 0.001) upon ageing. Thus, the significantly reduced abundance of MSY2 protein, especially at the SCRD and the spindle may disturb the spatial control and timely recruitment, deadenylation and degradation of developmentally important RNAs. An autonomous program of degradation appears to exist which transiently and specifically induces the loss and displacement of transcripts and specific maternal proteins independent of fertilization in aging oocytes and thereby can critically affect chromosome segregation and gene expression in the embryo after fertilization. LIMITATION, REASONS FOR CAUTION We used the mouse oocyte to study processes associated with postovulatory aging, which may not entirely reflect processes in aging human oocytes. However, increases in spindle abnormalities, unaligned chromosomes and H4K12 acetylated histones, as well as in mRNA abundance and polyadenylation have been observed also in aged human oocytes suggesting conserved processes in aging. WIDER IMPLICATIONS OF THE FINDINGS Postovulatory aging precociously induces alterations in expression and epigenetic modifications of chromatin by ATRX and in histone pattern in MII oocytes that normally occur after fertilization, possibly contributing to disturbances in the oocyte-to-embryo transition (OET) and the zygotic gene activation (ZGA). These observations in mouse oocytes are also relevant to explain disturbances and reduced developmental potential of aged human oocytes and caution to prevent oocyte aging in vivo and in vitro. STUDY FUNDING/COMPETING INTERESTS The study has been supported by the German Research Foundation (DFG) (EI 199/7-1 | GR 1138/12-1 | HO 949/21-1 and FOR 1041). There is no competing interest.
Collapse
Affiliation(s)
- T Trapphoff
- Institute of Gene Technology/Microbiology, University of Bielefeld, Bielefeld, Germany
| | - M Heiligentag
- Institute of Gene Technology/Microbiology, University of Bielefeld, Bielefeld, Germany
| | - D Dankert
- Institute of Anatomy, University Hospital, University Duisburg-Essen, Essen, Germany
| | - H Demond
- Institute of Human Genetics, University Hospital, University Duisburg-Essen, Essen, Germany
| | - D Deutsch
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - T Fröhlich
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - G J Arnold
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - R Grümmer
- Institute of Anatomy, University Hospital, University Duisburg-Essen, Essen, Germany
| | - B Horsthemke
- Institute of Human Genetics, University Hospital, University Duisburg-Essen, Essen, Germany
| | - U Eichenlaub-Ritter
- Institute of Gene Technology/Microbiology, University of Bielefeld, Bielefeld, Germany
| |
Collapse
|
17
|
Wang Q, Wei H, Du J, Cao Y, Zhang N, Liu X, Liu X, Chen D, Ma W. H3 Thr3 phosphorylation is crucial for meiotic resumption and anaphase onset in oocyte meiosis. Cell Cycle 2015; 15:213-24. [PMID: 26636626 DOI: 10.1080/15384101.2015.1121330] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Haspin-catalyzed histone H3 threonine 3 (Thr3) phosphorylation facilitates chromosomal passenger complex (CPC) docking at centromeres, regulating indirectly chromosome behavior during somatic mitosis. It is not fully known about the expression and function of H3 with phosphorylated Thr3 (H3T3-P) during meiosis in oocytes. In this study, we investigated the expression and sub-cellular distribution of H3T3-P, as well as its function in mouse oocytes during meiotic division. Western blot analysis revealed that H3T3-P expression was only detected after germinal vesicle breakdown (GVBD), and gradually increased to peak level at metaphase I (MI), but sharply decreased at metaphase II (MII). Immunofluorescence showed H3T3-P was only brightly labeled on chromosomes after GVBD, with relatively high concentration across the whole chromosome axis from pro-metaphase I (pro-MI) to MI. Specially, H3T3-P distribution was exclusively limited to the local space between sister centromeres at MII stage. Haspin inhibitor, 5-iodotubercidin (5-ITu), dose- and time-dependently blocked H3T3-P expression in mouse oocytes. H3T3-P inhibition delayed the resumption of meiosis (GVBD) and chromatin condensation. Moreover, the loss of H3T3-P speeded up the meiotic transition to MII of pro-MI oocytes in spite of the presence of non-aligned chromosomes, even reversed MI-arrest induced with Nocodazole. The inhibition of H3T3-P expression distinguishably damaged MAD1 recruitment on centromeres, which indicates the spindle assembly checkpoint was impaired in function, logically explaining the premature onset of anaphase I. Therefore, Haspin-catalyzed histone H3 phosphorylation is essential for chromatin condensation and the following timely transition from meiosis I to meiosis II in mouse oocytes during meiotic division.
Collapse
Affiliation(s)
- Qian Wang
- a Department of Histology and Embryology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| | - Haojie Wei
- a Department of Histology and Embryology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| | - Juan Du
- a Department of Histology and Embryology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| | - Yan Cao
- a Department of Histology and Embryology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| | - Nana Zhang
- a Department of Histology and Embryology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| | - Xiaoyun Liu
- a Department of Histology and Embryology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| | - Xiaoyu Liu
- a Department of Histology and Embryology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| | - Dandan Chen
- a Department of Histology and Embryology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| | - Wei Ma
- a Department of Histology and Embryology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| |
Collapse
|
18
|
Wu XF, Yuan HJ, Li H, Gong S, Lin J, Miao YL, Wang TY, Tan JH. Restraint stress on female mice diminishes the developmental potential of oocytes: roles of chromatin configuration and histone modification in germinal vesicle stage oocytes. Biol Reprod 2014; 92:13. [PMID: 25411393 DOI: 10.1095/biolreprod.114.124396] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The mechanisms by which restraint stress impairs oocyte developmental potential are unclear. Factors causing differences between the developmental potential of oocytes with surrounded nucleolus (SN) and that of oocytes with nonsurrounded nucleolus (NSN) are not fully characterized. Furthermore, the relationship between increased histone acetylation and methylation and the increased developmental competence in SN oocytes is particularly worth exploring using a system where the SN configuration can be uncoupled (dissociated) from increased histone modifications. In this study, female mice were subjected to restraint for 24 or 48 h or for 23 days before being examined for oocyte chromatin configuration, histone modification, and development in vitro and in vivo. Results showed that restraint for 48 h or 23 days impaired NSN-to-SN transition, histone acetylation and methylation in SN oocytes, and oocyte developmental potential. However, whereas the percentage of stressed SN oocytes returned to normal after a 48-h postrestraint recovery, neither histone acetylation/methylation in SN oocytes nor developmental competence recovered following postrestraint recovery with equine chorionic gonadotropin (eCG) injection. Priming unstressed mice with eCG expedited oocyte histone modification to an early completion. Contrary to the levels of acetylated and methylated histones, the level of phosphorylated H3S10 increased significantly in the stressed SN oocytes. Together, the results suggest that 1) restraint stress impaired oocyte potential with disturbed histone modifications; 2) SN configuration was uncoupled from increased histone acetylation/methylation in the restraint-stressed oocytes; and 3) the developmental potential of SN oocytes is more closely correlated with epigenetic histone modification than with chromatin configuration.
Collapse
Affiliation(s)
- Xiu-Fen Wu
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City, PR China
| | - Hong-Jie Yuan
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City, PR China
| | - Hong Li
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City, PR China
| | - Shuai Gong
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City, PR China
| | - Juan Lin
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City, PR China
| | - Yi-Long Miao
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City, PR China
| | - Tian-Yang Wang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City, PR China
| | - Jing-He Tan
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City, PR China
| |
Collapse
|
19
|
Trapphoff T, Heiligentag M, El Hajj N, Haaf T, Eichenlaub-Ritter U. Chronic exposure to a low concentration of bisphenol A during follicle culture affects the epigenetic status of germinal vesicles and metaphase II oocytes. Fertil Steril 2013; 100:1758-67.e1. [DOI: 10.1016/j.fertnstert.2013.08.021] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 07/24/2013] [Accepted: 08/09/2013] [Indexed: 12/14/2022]
|