1
|
Iannitti R, Mascanzoni F, Colanzi A, Spano D. The role of Golgi complex proteins in cell division and consequences of their dysregulation. Front Cell Dev Biol 2025; 12:1513472. [PMID: 39839669 PMCID: PMC11747491 DOI: 10.3389/fcell.2024.1513472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
The GC (Golgi complex) plays a pivotal role in the trafficking and sorting of proteins and lipids until they reach their final destination. Additionally, the GC acts as a signalling hub to regulate a multitude of cellular processes, including cell polarity, motility, apoptosis, DNA repair and cell division. In light of these crucial roles, the GC has garnered increasing attention, particularly given the evidence that a dysregulation of GC-regulated signalling pathways may contribute to the onset of various pathological conditions. This review examines the functions of the GC and GC-localised proteins in regulating cell cycle progression, in both mitosis and meiosis. It reviews the involvement of GC-resident proteins in the formation and orientation of the spindle during cell division. In light of the roles played by the GC in controlling cell division, this review also addresses the involvement of the GC in cancer development. Furthermore, TCGA (The Cancer Genome Atlas) database has been queried in order to retrieve information on the genetic alterations and the correlation between the expression of GC-localised proteins and the survival of cancer patients. The data presented in this review highlight the relevance of the GC in regulating cell cycle progression, cellular differentiation and tumourigenesis.
Collapse
Affiliation(s)
| | | | | | - Daniela Spano
- Department of Biomedical Sciences (DSB), Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), Naples, Italy
| |
Collapse
|
2
|
Verlhac MH. Exploring the maternal inheritance transmitted by the oocyte to its progeny. C R Biol 2024; 347:45-52. [PMID: 38888193 DOI: 10.5802/crbiol.155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
Fertility is declining worldwide and many couples are turning towards assisted reproductive technologies (ART) to conceive babies. Organisms that propagate via sexual reproduction often come from the fusion between two gametes, an oocyte and a sperm, whose qualities seem to be decreasing in the human species. Interestingly, while the sperm mostly transmits its haploid genome, the oocyte transmits not only its haploid set of chromosomes but also its huge cytoplasm to its progeny. This is what can be defined as the maternal inheritance composed of chromosomes, organelles, lipids, metabolites, proteins and RNAs. To decipher the decline in oocyte quality, it is essential to explore the nature of the maternal inheritance, and therefore study the last stages of murine oogenesis, namely the end of oocyte growth followed by the two meiotic divisions. These divisions are extremely asymmetric in terms of the size of the daughter cells, allowing to preserve the maternal inheritance accumulated during oocyte growth within these huge cells to support early embryo development. Studies performed in Marie-Hélène Verlhac's lab have allowed to discover the unprecedented impact of original acto-myosin based mechanisms in the constitution as well as the preservation of this maternal inheritance and the consequences when these processes go awry.
Collapse
|
3
|
Rémillard-Labrosse G, Cohen S, Boucher É, Gagnon K, Vasilev F, Mihajlović AI, FitzHarris G. Oocyte and embryo culture under oil profoundly alters effective concentrations of small molecule inhibitors. Front Cell Dev Biol 2024; 12:1337937. [PMID: 38544820 PMCID: PMC10966923 DOI: 10.3389/fcell.2024.1337937] [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: 11/13/2023] [Accepted: 02/19/2024] [Indexed: 11/11/2024] Open
Abstract
Culture of oocytes and embryos in media under oil is a cornerstone of fertility treatment, and extensively employed in experimental investigation of early mammalian development. It has been noted anecdotally by some that certain small molecule inhibitors might lose activity in oil-covered culture systems, presumably by drug partitioning into the oil. Here we took a pseudo-pharmacological approach to appraise this formally using mouse oocytes and embryos. Using different culture dish designs with defined media:oil volume ratios, we show that the EC50 of the widely employed microtubule poison nocodazole shifts as a function of the media:oil ratio, such that nocodazole concentrations that prevent cell division in oil-free culture fail to in oil-covered media drops. Relatively subtle changes in culture dish design lead to measurable changes in EC50. This effect is not specific to one type of culture oil, and can be readily observed both in oocyte and embryo culture experiments. We subsequently applied a similar approach to a small panel of widely employed cell cycle-related inhibitors, finding that most lose activity in standard oil-covered oocyte/embryo culture systems. Our data suggest that loss of small molecule activity in oil-covered oocyte and embryo culture is a widespread phenomenon with potentially far-reaching implications for data reproducibility, and we recommend avoiding oil-covered culture for experiments employing inhibitors/drugs wherever possible.
Collapse
Affiliation(s)
| | - Sydney Cohen
- Centre de Recherche du Centre Hospitalier de I’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Éliane Boucher
- Centre de Recherche du Centre Hospitalier de I’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Kéryanne Gagnon
- Centre de Recherche du Centre Hospitalier de I’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Filip Vasilev
- Centre de Recherche du Centre Hospitalier de I’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Aleksandar I. Mihajlović
- Centre de Recherche du Centre Hospitalier de I’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Greg FitzHarris
- Centre de Recherche du Centre Hospitalier de I’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Obstetrics and Gynaecology, Université de Montréal, Montréal, QC, Canada
- Department of Pathology and Cell Biology, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
4
|
Yang Y, Zhao C, Chen B, Yu X, Zhou Y, Ni D, Zhang X, Zhang J, Ling X, Zhang Z, Huo R. Follicular fluid C3a-peptide promotes oocyte maturation through F-actin aggregation. BMC Biol 2023; 21:285. [PMID: 38066646 PMCID: PMC10709936 DOI: 10.1186/s12915-023-01760-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/03/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Immature cumulus-oocyte complexes are retrieved to obtain mature oocytes by in vitro maturation (IVM), a laboratory tool in reproductive medicine to obtain mature oocytes. Unfortunately, the efficiency of IVM is not satisfactory. To circumvent this problem, we therefore intended to commence with the composition of ovarian follicular fluid (FF), an important microenvironment influencing oocyte growth. It is well known that FF has a critical role in oocyte development and maturation. However, the components in human FF remain largely unknown, particularly with regard to small molecular peptides. RESULTS In current study, the follicular fluid derived from human mature and immature follicles were harvested. The peptide profiles of FF were further investigated by using combined ultrafiltration and LC-MS/MS. The differential peptides were preliminary determined by performing differentially expressed analysis. Human and mouse oocyte culture were used to verify the influence of differential peptides on oocyte development. Constructing plasmids, cell transfecting, Co-IP, PLA etc. were used to reveal the detail molecular mechanism. The results from differentially expressed peptide as well as cultured human and mouse oocytes analyses showed that highly conserved C3a-peptide, a cleavage product of complement C3a, definitely affected oocytes development. Intriguingly, C3a-peptide possessed a novel function that promoted F-actin aggregation and spindle migration, raised the percentage of oocytes at the MII stage, without increasing the chromosome aneuploidy ratio, especially in poor-quality oocytes. These effects of C3a-peptide were attenuated by C3aR morpholino inhibition, suggesting that C3a-peptide affected oocytes development by collaborating with its classical receptor, C3aR. Specially, we found that C3aR co-localized to the spindle with β-tubulin to recruit F-actin toward the spindle and subcortical region of the oocytes through specific binding to MYO10, a key regulator for actin organization, spindle morphogenesis and positioning in oocytes. CONCLUSIONS Our results provide a new perspective for improving IVM culture systems by applying FF components and also provide molecular insights into the physiological function of C3a-peptide, its interaction with C3aR, and their roles in enabling meiotic division of oocytes.
Collapse
Affiliation(s)
- Ye Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Chun Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China
| | - Beili Chen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Xiaoning Yu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Yuxi Zhou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China
| | - Danyu Ni
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China
| | - Xiaolan Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China
| | - Junqiang Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China
| | - Xiufeng Ling
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Reproduction, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu Province, 210004, China.
| | - Zhiguo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Anhui, China.
| | - Ran Huo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China.
- Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School,, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
5
|
Bourdais A, Dehapiot B, Halet G. MRCK activates mouse oocyte myosin II for spindle rotation and male pronucleus centration. J Cell Biol 2023; 222:e202211029. [PMID: 37651121 PMCID: PMC10470461 DOI: 10.1083/jcb.202211029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 06/24/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
Asymmetric meiotic divisions in oocytes rely on spindle positioning in close vicinity to the cortex. In metaphase II mouse oocytes, eccentric spindle positioning triggers cortical polarization, including the build-up of an actin cap surrounded by a ring of activated myosin II. While the role of the actin cap in promoting polar body formation is established, ring myosin II activation mechanisms and functions have remained elusive. Here, we show that ring myosin II activation requires myotonic dystrophy kinase-related Cdc42-binding kinase (MRCK), downstream of polarized Cdc42. MRCK inhibition resulted in spindle rotation defects during anaphase II, precluding polar body extrusion. Remarkably, disengagement of segregated chromatids from the anaphase spindle could rescue rotation. We further show that the MRCK/myosin II pathway is activated in the fertilization cone and is required for male pronucleus migration toward the center of the zygote. These findings provide novel insights into the mechanism of myosin II activation in oocytes and its role in orchestrating asymmetric division and pronucleus centration.
Collapse
Affiliation(s)
- Anne Bourdais
- University of Rennes, CNRS - UMR 6290, Institute of Genetics and Development of Rennes, Rennes, France
| | - Benoit Dehapiot
- University of Rennes, CNRS - UMR 6290, Institute of Genetics and Development of Rennes, Rennes, France
| | - Guillaume Halet
- University of Rennes, CNRS - UMR 6290, Institute of Genetics and Development of Rennes, Rennes, France
| |
Collapse
|
6
|
Yuen WS, Zhang QH, Bourdais A, Adhikari D, Halet G, Carroll J. Polo-like kinase 1 promotes Cdc42-induced actin polymerization for asymmetric division in oocytes. Open Biol 2023; 13:220326. [PMID: 36883283 PMCID: PMC9993042 DOI: 10.1098/rsob.220326] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
Polo-like kinase I (Plk1) is a highly conserved seronine/threonine kinase essential in meiosis and mitosis for spindle formation and cytokinesis. Here, through temporal application of Plk1 inhibitors, we identify a new role for Plk1 in the establishment of cortical polarity essential for highly asymmetric cell divisions of oocyte meiosis. Application of Plk1 inhibitors in late metaphase I abolishes pPlk1 from spindle poles and prevents the induction of actin polymerization at the cortex through inhibition of local recruitment of Cdc42 and Neuronal Wiskott-Aldrich Syndrome protein (N-WASP). By contrast, an already established polar actin cortex is insensitive to Plk1 inhibitors, but if the polar cortex is first depolymerized, Plk1 inhibitors completely prevent its restoration. Thus, Plk1 is essential for establishment but not maintenance of cortical actin polarity. These findings indicate that Plk1 regulates recruitment of Cdc42 and N-Wasp to coordinate cortical polarity and asymmetric cell division.
Collapse
Affiliation(s)
- Wai Shan Yuen
- Department of Anatomy and Developmental Biology and Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Qing Hua Zhang
- Department of Anatomy and Developmental Biology and Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Anne Bourdais
- University of Rennes, CNRS, IGDR - UMR 6290, F-35000 Rennes, France
| | - Deepak Adhikari
- Department of Anatomy and Developmental Biology and Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Guillaume Halet
- University of Rennes, CNRS, IGDR - UMR 6290, F-35000 Rennes, France
| | - John Carroll
- Department of Anatomy and Developmental Biology and Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
7
|
Kincade JN, Hlavacek A, Akera T, Balboula AZ. Initial spindle positioning at the oocyte center protects against incorrect kinetochore-microtubule attachment and aneuploidy in mice. SCIENCE ADVANCES 2023; 9:eadd7397. [PMID: 36800430 PMCID: PMC9937575 DOI: 10.1126/sciadv.add7397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 01/19/2023] [Indexed: 06/18/2023]
Abstract
Spindle positioning within the oocyte must be tightly regulated. In mice, the spindle is predominantly assembled at the oocyte center before its migration toward the cortex to achieve the highly asymmetric division, a characteristic of female meiosis. The significance of the initial central positioning of the spindle is largely unknown. We show that initial spindle positioning at the oocyte center is an insurance mechanism to avoid the premature exposure of the spindle to cortical CDC42 signaling, which perturbs proper kinetochore-microtubule attachments, leading to the formation of aneuploid gametes. These findings contribute to understanding why female gametes are notoriously associated with high rates of aneuploidy, the leading genetic cause of miscarriage and congenital abnormalities.
Collapse
Affiliation(s)
- Jessica N. Kincade
- Animal Sciences Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Avery Hlavacek
- Animal Sciences Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Takashi Akera
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ahmed Z. Balboula
- Animal Sciences Research Center, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
8
|
Feng J, Soto‐Moreno EJ, Prakash A, Balboula AZ, Qiao H. Adverse PFAS effects on mouse oocyte in vitro maturation are associated with carbon-chain length and inclusion of a sulfonate group. Cell Prolif 2023; 56:e13353. [PMID: 36305033 PMCID: PMC9890540 DOI: 10.1111/cpr.13353] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/04/2022] [Accepted: 10/09/2022] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVES Per- and polyfluoroalkyl substances (PFAS) are man-made chemicals that are widely used in various products. PFAS are characterized by their fluorinated carbon chains that make them hard to degrade and bioaccumulate in human and animals. Toxicological studies have shown PFAS toxic effects: cytotoxicity, immunotoxicity, neurotoxicity, and reproductive toxicity. However, it is still unclear how the structures of PFAS, such as carbon-chain length and functional groups, determine their reproductive toxicity. METHODS AND RESULTS By using a mouse-oocyte-in-vitro-maturation (IVM) system, we found the toxicity of two major categories of PFAS, perfluoroalkyl carboxylic acid (PFCA) and perfluoroalkyl sulfonic acid (PFSA), is elevated with increasing carbon-chain length and the inclusion of the sulfonate group. Specifically, at 600 μM, perfluorohexanesulfonic acid (PFHxS) and perfluorooctanesulfonic acid (PFOS) reduced the rates of both germinal-vesicle breakdown (GVBD) and polar-body extrusion (PBE) as well as enlarged polar bodies. However, the shorter PFSA, perfluorobutanesulfonic acid (PFBS), and all PFCA did not show similar adverse cytotoxicity. Further, we found that 600 μM PFHxS and PFOS exposure induced excess reactive oxygen species (ROS) and decreased mitochondrial membrane potential (MMP). Cytoskeleton analysis revealed that PFHxS and PFOS exposure induced chromosome misalignment, abnormal F-actin organization, elongated spindle formation, and symmetric division in the treated oocytes. These meiotic defects compromised oocyte developmental competence after parthenogenetic activation. CONCLUSIONS Our study provides new information on the structure-toxicity relationship of PFAS.
Collapse
Affiliation(s)
- Jianan Feng
- Department of Comparative BiosciencesUniversity of Illinois at Urbana‐ChampaignChampaignUrbanaUSA
| | | | - Aashna Prakash
- Department of Comparative BiosciencesUniversity of Illinois at Urbana‐ChampaignChampaignUrbanaUSA
| | - Ahmed Z. Balboula
- Division of Animal SciencesUniversity of MissouriMissouriColumbiaUSA
| | - Huanyu Qiao
- Department of Comparative BiosciencesUniversity of Illinois at Urbana‐ChampaignChampaignUrbanaUSA
- Carl R. Woese Institute for Genomic BiologyUniversity of Illinois at Urbana‐ChampaignChampaignUrbanaUSA
| |
Collapse
|
9
|
Zhuan Q, Li J, Zhou G, Du X, Liu H, Hou Y, Wan P, Fu X. Procyanidin B2 Protects Aged Oocytes Against Meiotic Defects Through Cortical Tension Modulation. Front Vet Sci 2022; 9:795050. [PMID: 35464357 PMCID: PMC9024290 DOI: 10.3389/fvets.2022.795050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/20/2022] [Indexed: 11/16/2022] Open
Abstract
Defects in meiotic process are the main factors responsible for the decreased developmental competence in aged oocytes. Our recent research indicated that natural antioxidant procyanidin B2 (PCB2) promoted maturation progress in oocytes from diabetic mice. However, the effect of PCB2 on aging-induced chromosome abnormalities and the underlying mechanism have not been explored. Here, we found that PCB2 recovered aging-caused developmental arrest during meiotic maturation, germinal vesicle breakdown (GVBD) rate was significantly higher in aged oocytes treated with PCB2 (P < 0.05). Furthermore, we discovered that cortical mechanics were altered during aging process, cortical tension-related proteins were aberrantly expressed in aged oocytes (P < 0.001). PCB2 supplementation efficaciously antagonized aging-induced decreased cortical tension (P < 0.001). Moreover, PCB2 restored spindle morphology (P < 0.01), maintained proper chromosome alignment (P < 0.05), and dramatically reduced reactive oxygen species (ROS) level (P < 0.05) in aged oocytes. Collectively, our results reveal that PCB2 supplementation is a feasible approach to protect oocytes from reproductive aging, contributing to the improvement of oocytes quality.
Collapse
Affiliation(s)
- Qingrui Zhuan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jun Li
- Department of Reproductive Medicine, Reproductive Medical Center, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guizhen Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xingzhu Du
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Hongyu Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yunpeng Hou
- State Key Laboratories of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Pengcheng Wan
- State Key Laboratory of Sheep Genetic Improvement and Healthy Breeding, Institute of Animal Husbandry and Veterinary Sciences, Xinjiang Academy of Agricultural and Reclamation Sciences, Shihhotze, China
| | - Xiangwei Fu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
- State Key Laboratory of Sheep Genetic Improvement and Healthy Breeding, Institute of Animal Husbandry and Veterinary Sciences, Xinjiang Academy of Agricultural and Reclamation Sciences, Shihhotze, China
- *Correspondence: Xiangwei Fu
| |
Collapse
|
10
|
Londoño-Vásquez D, Rodriguez-Lukey K, Behura SK, Balboula AZ. Microtubule organizing centers regulate spindle positioning in mouse oocytes. Dev Cell 2022; 57:197-211.e3. [PMID: 35030327 PMCID: PMC8792338 DOI: 10.1016/j.devcel.2021.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 10/18/2021] [Accepted: 12/10/2021] [Indexed: 01/26/2023]
Abstract
During female meiosis I (MI), spindle positioning must be tightly regulated to ensure the fidelity of the first asymmetric division and faithful chromosome segregation. Although the role of F-actin in regulating these critical processes has been studied extensively, little is known about whether microtubules (MTs) participate in regulating these processes. Using mouse oocytes as a model system, we characterize a subset of MT organizing centers that do not contribute directly to spindle assembly, termed mcMTOCs. Using laser ablation, STED super-resolution microscopy, and chemical manipulation, we show that mcMTOCs are required to regulate spindle positioning and faithful chromosome segregation during MI. We discuss how forces exerted by F-actin on the spindle are balanced by mcMTOC-nucleated MTs to anchor the spindle centrally and to regulate its timely migration. Our findings provide a model for asymmetric cell division, complementing the current F-actin-based models, and implicate mcMTOCs as a major player in regulating spindle positioning.
Collapse
Affiliation(s)
| | | | - Susanta K Behura
- Animal Sciences Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Ahmed Z Balboula
- Animal Sciences Research Center, University of Missouri, Columbia, MO 65211, USA; University of Cambridge, Department of Genetics, Downing Street, Cambridge, CB2 3EH, UK.
| |
Collapse
|
11
|
Bourdais A, Dehapiot B, Halet G. Cofilin regulates actin network homeostasis and microvilli length in mouse oocytes. J Cell Sci 2021; 134:273797. [PMID: 34841429 DOI: 10.1242/jcs.259237] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/18/2021] [Indexed: 11/20/2022] Open
Abstract
How multiple actin networks coexist in a common cytoplasm while competing for a shared pool of monomers is still an ongoing question. This is exemplified by meiotic maturation in the mouse oocyte, which relies on the dynamic remodeling of distinct cortical and cytoplasmic F-actin networks. Here, we show that the conserved actin-depolymerizing factor cofilin is activated in a switch-like manner upon meiosis resumption from prophase arrest. Interfering with cofilin activation during maturation resulted in widespread elongation of microvilli, while cytoplasmic F-actin was depleted, leading to defects in spindle migration and polar body extrusion. In contrast, cofilin inactivation in metaphase II-arrested oocytes resulted in a shutdown of F-actin dynamics, along with a dramatic overgrowth of the polarized actin cap. However, inhibition of the Arp2/3 complex to promote actin cap disassembly elicited ectopic microvilli outgrowth in the polarized cortex. These data establish cofilin as a key player in actin network homeostasis in oocytes and reveal that microvilli can act as a sink for monomers upon disassembly of a competing network.
Collapse
Affiliation(s)
- Anne Bourdais
- Institut Génétique et Développement de Rennes , CNRS IGDR UMR 6290, Université de Rennes 1, F-35000 Rennes, France
| | - Benoit Dehapiot
- Institut Génétique et Développement de Rennes , CNRS IGDR UMR 6290, Université de Rennes 1, F-35000 Rennes, France
| | - Guillaume Halet
- Institut Génétique et Développement de Rennes , CNRS IGDR UMR 6290, Université de Rennes 1, F-35000 Rennes, France
| |
Collapse
|
12
|
Verlhac MH. Preventing aneuploidy: The groom must wait until the bride is ready. J Cell Biol 2021; 220:e202108030. [PMID: 34505874 PMCID: PMC8438638 DOI: 10.1083/jcb.202108030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Fertilization often triggers the final step of haploidization of the female gamete genome. In this issue, Mori et al. (2021. J. Cell Biol.https://doi.org/10.1083/jcb.202012001) identify two successive actin-dependent mechanisms that delay fusion of maternal and paternal chromosomes, preventing inadvertent elimination of paternal chromosomes together with maternal ones.
Collapse
Affiliation(s)
- Marie-Hélène Verlhac
- Center for Interdisciplinary Research in Biology, Collège de France, UMR7241/U1050, Paris Sciences et Lettres Research University, Paris, France
| |
Collapse
|
13
|
Mori M, Yao T, Mishina T, Endoh H, Tanaka M, Yonezawa N, Shimamoto Y, Yonemura S, Yamagata K, Kitajima TS, Ikawa M. RanGTP and the actin cytoskeleton keep paternal and maternal chromosomes apart during fertilization. J Cell Biol 2021; 220:e202012001. [PMID: 34424312 PMCID: PMC8404465 DOI: 10.1083/jcb.202012001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 06/18/2021] [Accepted: 08/06/2021] [Indexed: 11/22/2022] Open
Abstract
Zygotes require two accurate sets of parental chromosomes, one each from the mother and the father, to undergo normal embryogenesis. However, upon egg-sperm fusion in vertebrates, the zygote has three sets of chromosomes, one from the sperm and two from the egg. The zygote therefore eliminates one set of maternal chromosomes (but not the paternal chromosomes) into the polar body through meiosis, but how the paternal chromosomes are protected from maternal meiosis has been unclear. Here we report that RanGTP and F-actin dynamics prevent egg-sperm fusion in proximity to maternal chromosomes. RanGTP prevents the localization of Juno and CD9, egg membrane proteins that mediate sperm fusion, at the cell surface in proximity to maternal chromosomes. Following egg-sperm fusion, F-actin keeps paternal chromosomes away from maternal chromosomes. Disruption of these mechanisms causes the elimination of paternal chromosomes during maternal meiosis. This study reveals a novel critical mechanism that prevents aneuploidy in zygotes.
Collapse
Affiliation(s)
- Masashi Mori
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Laboratory for Chromosome Segregation, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Tatsuma Yao
- Research and Development Center, Fuso Pharmaceutical Industries, Ltd., Osaka, Japan
- Graduate School of Biology-Oriented Science and Technology, Kindai University, Wakayama, Japan
| | - Tappei Mishina
- Laboratory for Chromosome Segregation, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Hiromi Endoh
- Laboratory for Ultrastructural Research, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Masahito Tanaka
- Physics and Cell Biology Laboratory, National Institute of Genetics & Department of Genetics, SOKENDAI University, Kanagawa, Japan
| | - Nao Yonezawa
- Graduate School of Biology-Oriented Science and Technology, Kindai University, Wakayama, Japan
| | - Yuta Shimamoto
- Physics and Cell Biology Laboratory, National Institute of Genetics & Department of Genetics, SOKENDAI University, Kanagawa, Japan
| | - Shigenobu Yonemura
- Laboratory for Ultrastructural Research, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Department of Cell Biology, Tokushima University Graduate School of Medical Science, Tokushima, Japan
| | - Kazuo Yamagata
- Graduate School of Biology-Oriented Science and Technology, Kindai University, Wakayama, Japan
| | - Tomoya S. Kitajima
- Laboratory for Chromosome Segregation, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| |
Collapse
|
14
|
Crozet F, Da Silva C, Verlhac MH, Terret ME. Myosin-X is dispensable for spindle morphogenesis and positioning in the mouse oocyte. Development 2021; 148:dev.199364. [PMID: 33722900 PMCID: PMC8077531 DOI: 10.1242/dev.199364] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/02/2021] [Indexed: 01/08/2023]
Abstract
Off-center spindle positioning in mammalian oocytes enables asymmetric divisions in size, which are important for subsequent embryogenesis. The migration of the meiosis I spindle from the oocyte center to its cortex is mediated by F-actin. Specifically, an F-actin cage surrounds the microtubule spindle and applies forces to it. To better understand how F-actin transmits forces to the spindle, we studied a potential direct link between F-actin and microtubules. For this, we tested the implication of myosin-X, a known F-actin and microtubule binder involved in spindle morphogenesis and/or positioning in somatic cells, amphibian oocytes and embryos. Using a mouse strain conditionally invalidated for myosin-X in oocytes and by live-cell imaging, we show that myosin-X is not localized on the spindle, and is dispensable for spindle and F-actin assembly. It is not required for force transmission as spindle migration and chromosome alignment occur normally. More broadly, myosin-X is dispensable for oocyte developmental potential and female fertility. We therefore exclude a role for myosin-X in transmitting F-actin-mediated forces to the spindle, opening new perspectives regarding this mechanism in mouse oocytes, which differ from most mitotic cells. Summary: Cortical spindle positioning in mammalian oocytes relies on the interplay between actin and the microtubule spindle. Myosin-X, an obvious candidate for linking these two cytoskeletal elements, is dispensable in mouse oocytes.
Collapse
Affiliation(s)
- Flora Crozet
- CIRB, Collège de France, UMR7241/U1050, 75005 Paris, France
| | | | | | | |
Collapse
|
15
|
Trebichalská Z, Kyjovská D, Kloudová S, Otevřel P, Hampl A, Holubcová Z. Cytoplasmic maturation in human oocytes: an ultrastructural study †. Biol Reprod 2020; 104:106-116. [PMID: 33404651 PMCID: PMC7786262 DOI: 10.1093/biolre/ioaa174] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/07/2020] [Accepted: 09/15/2020] [Indexed: 12/24/2022] Open
Abstract
Female fertility relies on successful egg development. Besides chromosome segregation, complex structural and biochemical changes in the cytoplasmic compartment are necessary to confer the female gamete the capacity to undergo normal fertilization and sustain embryonic development. Despite the profound impact on egg quality, morphological bases of cytoplasmic maturation remain largely unknown. Here, we report our findings from the ultrastructural analysis of 69 unfertilized human oocytes from 34 young and healthy egg donors. By comparison of samples fixed at three consecutive developmental stages, we explored how ooplasmic architecture changes during meiotic maturation in vitro. The morphometric image analysis supported observation that the major reorganization of cytoplasm occurs before polar body extrusion. The organelles initially concentrated around prophase nucleus were repositioned toward the periphery and evenly distributed throughout the ooplasm. As maturation progressed, distinct secretory apparatus appeared to transform into cortical granules that clustered underneath the oocyte's surface. The most prominent feature was the gradual formation of heterologous complexes composed of variable elements of endoplasmic reticulum and multiple mitochondria with primitive morphology. Based on the generated image dataset, we proposed a morphological map of cytoplasmic maturation, which may serve as a reference for future comparative studies. In conclusion, this work improves our understanding of human oocyte morphology, cytoplasmic maturation, and intracellular factors defining human egg quality. Although this analysis involved spare oocytes completing development in vitro, it provides essential insight into the enigmatic process by which human egg progenitors prepare for fertilization.
Collapse
Affiliation(s)
- Z Trebichalská
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - D Kyjovská
- Reprofit International, Clinic of Reproductive Medicine, Brno, Czech Republic
| | - S Kloudová
- Reprofit International, Clinic of Reproductive Medicine, Brno, Czech Republic
| | - P Otevřel
- Reprofit International, Clinic of Reproductive Medicine, Brno, Czech Republic
| | - A Hampl
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Z Holubcová
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Reprofit International, Clinic of Reproductive Medicine, Brno, Czech Republic
| |
Collapse
|
16
|
Ferrer-Vaquer A, Barragán M, Rodríguez A, Vassena R. Altered cytoplasmic maturation in rescued in vitro matured oocytes. Hum Reprod 2020; 34:1095-1105. [PMID: 31119269 DOI: 10.1093/humrep/dez052] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/26/2019] [Accepted: 03/28/2019] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION Do culture conditions affect cytoplasmic maturation in denuded immature non-GV oocytes? SUMMARY ANSWER The maturation rate of denuded non-GV oocytes is not affected by culture media, but in vitro maturation seems to alter the mitochondrial membrane potential, endoplasmic reticulum (ER) and actin cytoskeleton compared with in vivo maturation. WHAT IS KNOWN ALREADY In vitro maturation of denuded immature non-GV oocytes benefits cycles with poor in vivo MII oocyte collection, but maturation levels of non-GV oocytes are only scored by polar body extrusion. Since oocyte maturation involves nuclear as well as cytoplasmic maturation for full meiotic competence, further knowledge is needed about cytoplasmic maturation in in vitro culture. STUDY DESIGN, SIZE, DURATION This basic research study was carried out between January 2017 and September 2018. PARTICIPANTS/MATERIALS, SETTING, METHODS A total of 339 denuded immature non-GV oocytes were cultured in SAGE 1-Step (177) or G-2 PLUS (162) for 6-8 h after retrieval, and 72 in vivo matured MII oocytes were used as controls. Cultured immature non-GV oocytes were scored for polar body extrusion and analysed for mitochondrial membrane potential (ΔΨm), ER clusters, cortical granules number and distribution, spindle morphology and actin cytoskeleton organization. The obtained parameter values were compared to in vivo matured MII oocyte parameter values. MAIN RESULTS AND THE ROLE OF CHANCE The maturation rates of oocytes cultured in G-2 PLUS and SAGE 1-Step were similar (65% vs 64.2%; P = 0.91). The differences observed in cortical granule density were not statistically significant. Also spindle morphometric parameters were mostly similar between in vitro and in vivo matured MII oocytes. However, the number of ER clusters, the ΔΨm and the cortical actin thickness showed significant differences between in vivo MII oocytes and denuded immature non-GV oocytes cultured in vitro until meiosis completion. LIMITATIONS, REASONS FOR CAUTION Frozen-thawed oocytes together with fresh oocytes were used as controls. Due to technical limitations (fixation method and fluorochrome overlap), only one or two parameters could be studied per oocyte. Thus, a global view of the maturation status for each individual oocyte could not be obtained. WIDER IMPLICATIONS OF THE FINDINGS Characterization of in vitro matured oocytes at the cellular level will help us to understand the differences observed in the clinical outcomes reported with rescue IVM compared to in vivo MII oocytes and to improve the culture methods applied. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by intramural funding of Clinica Eugin and by the Torres Quevedo Program to A.F.-V. from the Spanish Ministry of Economy and Competitiveness. No competing interests are declared.
Collapse
|
17
|
Santella L, Limatola N, Chun JT. Cellular and molecular aspects of oocyte maturation and fertilization: a perspective from the actin cytoskeleton. ZOOLOGICAL LETTERS 2020; 6:5. [PMID: 32313685 PMCID: PMC7158055 DOI: 10.1186/s40851-020-00157-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/26/2020] [Indexed: 05/06/2023]
Abstract
ABSTRACT Much of the scientific knowledge on oocyte maturation, fertilization, and embryonic development has come from the experiments using gametes of marine organisms that reproduce by external fertilization. In particular, echinoderm eggs have enabled the study of structural and biochemical changes related to meiotic maturation and fertilization owing to the abundant availability of large and transparent oocytes and eggs. Thus, in vitro studies of oocyte maturation and sperm-induced egg activation in starfish are carried out under experimental conditions that resemble those occurring in nature. During the maturation process, immature oocytes of starfish are released from the prophase of the first meiotic division, and acquire the competence to be fertilized through a highly programmed sequence of morphological and physiological changes at the oocyte surface. In addition, the changes in the cortical and nuclear regions are essential for normal and monospermic fertilization. This review summarizes the current state of research on the cortical actin cytoskeleton in mediating structural and physiological changes during oocyte maturation and sperm and egg activation in starfish and sea urchin. The common denominator in these studies with echinoderms is that exquisite rearrangements of the egg cortical actin filaments play pivotal roles in gamete interactions, Ca2+ signaling, exocytosis of cortical granules, and control of monospermic fertilization. In this review, we also compare findings from studies using invertebrate eggs with what is known about the contributions made by the actin cytoskeleton in mammalian eggs. Since the cortical actin cytoskeleton affects microvillar morphology, movement, and positioning of organelles and vesicles, and the topography of the egg surface, these changes have impacts on the fertilization process, as has been suggested by recent morphological studies on starfish oocytes and eggs using scanning electron microscopy. Drawing the parallelism between vitelline layer of echinoderm eggs and the zona pellucida of mammalian eggs, we also discuss the importance of the egg surface in mediating monospermic fertilization. GRAPHICAL ABSTRACT
Collapse
Affiliation(s)
- Luigia Santella
- Department of Research Infrastructures for Marine Biological Resources, Stazione Zoologica Anton Dohrn, Villa Comunale, Napoli 80121, Italy
| | - Nunzia Limatola
- Department of Research Infrastructures for Marine Biological Resources, Stazione Zoologica Anton Dohrn, Villa Comunale, Napoli 80121, Italy
| | - Jong Tai Chun
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, Napoli 80121, Italy
| |
Collapse
|
18
|
Vandenberghe LTM, Heindryckx B, Smits K, Szymanska K, Ortiz-Escribano N, Ferrer-Buitrago M, Pavani K, Peelman L, Deforce D, De Sutter P, Van Soom A, De Schauwer C. Platelet-activating factor acetylhydrolase 1B3 (PAFAH1B3) is required for the formation of the meiotic spindle during in vitro oocyte maturation. Reprod Fertil Dev 2019; 30:1739-1750. [PMID: 30008286 DOI: 10.1071/rd18019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 06/06/2018] [Indexed: 11/23/2022] Open
Abstract
Platelet-activating factor (PAF) is a well-described autocrine growth factor involved in several reproductive processes and is tightly regulated by its hydrolysing enzyme, PAF acetylhydrolase 1B (PAFAH1B). This intracellular enzyme consists of three subunits: one regulatory, 1B1, and two catalytic, 1B2 and 1B3. PAFAH1B3 has remained uncharacterised until now. Here, we report that PAFAH1B3 is present during the different stages of the first meiotic division in bovine, murine and human oocytes. In these species, the PAFAH1B3 subunit was clearly present in the germinal vesicle, while at metaphase I and II, it localised primarily at the meiotic spindle structure. In cattle, manipulation of the microtubules of the spindle by nocodazole, taxol or cryopreservation revealed a close association with PAFAH1B3. On the other hand, disruption of the enzyme activity either by P11, a selective inhibitor of PAFAH1B3, or by PAFAH1B3 antibody microinjection, caused arrest at the MI stage with defective spindle morphology and consequent failure of first polar body extrusion. In conclusion, our results show that one of the catalytic subunits of PAFAH1B, namely PAFAH1B3, is present in bovine, murine and human oocytes and that it plays a functional role in spindle formation and meiotic progression during bovine oocyte maturation.
Collapse
Affiliation(s)
- L T M Vandenberghe
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - B Heindryckx
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - K Smits
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - K Szymanska
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium
| | - N Ortiz-Escribano
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - M Ferrer-Buitrago
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - K Pavani
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - L Peelman
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium
| | - D Deforce
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - P De Sutter
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - A Van Soom
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - C De Schauwer
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| |
Collapse
|
19
|
Zhang Y, Wan X, Wang HH, Pan MH, Pan ZN, Sun SC. RAB35 depletion affects spindle formation and actin-based spindle migration in mouse oocyte meiosis. ACTA ACUST UNITED AC 2019; 25:359-372. [DOI: 10.1093/molehr/gaz027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/28/2019] [Accepted: 05/17/2019] [Indexed: 12/16/2022]
Abstract
Abstract
Mammalian oocyte maturation involves a unique asymmetric cell division, in which meiotic spindle formation and actin filament-mediated spindle migration to the oocyte cortex are key processes. Here, we report that the vesicle trafficking regulator, RAB35 GTPase, is involved in regulating cytoskeleton dynamics in mouse oocytes. RAB35 GTPase mainly accumulated at the meiotic spindle periphery and cortex during oocyte meiosis. Depletion of RAB35 by morpholino microinjection led to aberrant polar body extrusion and asymmetric division defects in almost half the treated oocytes. We also found that RAB35 affected SIRT2 and αTAT for tubulin acetylation, which further modulated microtubule stability and meiotic spindle formation. Additionally, we found that RAB35 associated with RHOA in oocytes and modulated the ROCK–cofilin pathway for actin assembly, which further facilitated spindle migration for oocyte asymmetric division. Importantly, microinjection of Myc-Rab35 cRNA into RAB35-depleted oocytes could significantly rescue these defects. In summary, our results suggest that RAB35 GTPase has multiple roles in spindle stability and actin-mediated spindle migration in mouse oocyte meiosis.
Collapse
Affiliation(s)
- Yu Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xiang Wan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Hong-Hui Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Meng-Hao Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zhen-Nan Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
20
|
Xu M, Qian J, Si L, Qu X, Li J. The Effect of Epigenetic Changes on the Extrusion of the First Polar Body in Pig Oocytes During In Vitro Maturation. Cell Reprogram 2019; 21:129-140. [PMID: 31084435 DOI: 10.1089/cell.2018.0071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The present study was designed to investigate the comprehensive function of maternal factors of primordial germ cell 7 (PGC7) and POU5F1-POU class 5 homeobox 1 (OCT4), as well as the epigenetic modification roles on the mitosis for the extrusion of first polar body (PB1) in pig maturated oocytes. First, the common distribution of histone modifications, including H3K4me2, H3K27me3, H3K9me2, and H4K12ac and DNA methylation, were detected at the high level in the nucleus. However, only one part of the chromosome was higher methylated or acetylated when the mitosis happened to extrude the PB1. When the mitosis was inhibited by the cytochalasin B (CB) treatment, the expression of PGC7, OCT4, DNA methyltransferase1 (DNMT1), DNA methyltransferase3b (DNMT3b), tet methylcytosine dioxygenase 1 (TET1), tet methylcytosine dioxygenase 2 (TET2), and tet methylcytosine dioxygenase 3 (TET3) could be inhibited (p < 0.01), and no concentrated expression of the PGC7 and OCT4 was observed on the chromosome, but the levels of H3K9me2 and H4K12ac were higher. In addition, when the trichostatin A was performed on the in vitro maturation, the extrusion of the PB1 was inhibited too. And the histone methylation (H3K9me2 and H3K27me3) could be detected all the time with relative higher level and no demethylation could be observed. However, the expression of PGC7 and OCT4 was lower in the chromosome. It might indicate that the maternal factor of PGC7 and histone modification that included H4K12ac and H3K9me2 could regulate the extrusion of the PB1 and play an important role in the maturation of pig oocytes.
Collapse
Affiliation(s)
- Mingzhu Xu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, P.R. China
| | - Jialing Qian
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, P.R. China
| | - Linan Si
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, P.R. China
| | - Xiao Qu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, P.R. China
| | - Juan Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, P.R. China
| |
Collapse
|
21
|
Uraji J, Scheffler K, Schuh M. Functions of actin in mouse oocytes at a glance. J Cell Sci 2018; 131:131/22/jcs218099. [PMID: 30467138 DOI: 10.1242/jcs.218099] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Gametes undergo a specialized and reductional cell division termed meiosis. Female gametes (oocytes) undergo two rounds of meiosis; the first meiotic division produces the fertilizable egg, while the second meiotic division occurs upon fertilization. Both meiotic divisions are highly asymmetric, producing a large egg and small polar bodies. Actin takes over various essential function during oocyte meiosis, many of which commonly rely on microtubules in mitotic cells. Specifically, the actin network has been linked to long-range vesicle transport, nuclear positioning, spindle migration and anchorage, polar body extrusion and accurate chromosome segregation in mammalian oocytes. In this Cell Science at a Glance article and the accompanying poster, we summarize the many functions of the actin cytoskeleton in oocytes, with a focus on findings from the mouse model system.
Collapse
Affiliation(s)
- Julia Uraji
- Department of Meiosis, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Kathleen Scheffler
- Department of Meiosis, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Melina Schuh
- Department of Meiosis, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| |
Collapse
|
22
|
Abstract
Fertilizable eggs develop from diploid precursor cells termed oocytes. Once every menstrual cycle, an oocyte matures into a fertilizable egg in the ovary. To this end, the oocyte eliminates half of its chromosomes into a small cell termed a polar body. The egg is then released into the Fallopian tube, where it can be fertilized. Upon fertilization, the egg completes the second meiotic division, and the mitotic division of the embryo starts. This review highlights recent work that has shed light on the cytoskeletal structures that drive the meiotic divisions of the oocyte in mammals. In particular, we focus on how mammalian oocytes assemble a microtubule spindle in the absence of centrosomes, how they position the spindle in preparation for polar body extrusion, and how the spindle segregates the chromosomes. We primarily focus on mouse oocytes as a model system but also highlight recent insights from human oocytes.
Collapse
Affiliation(s)
- Binyam Mogessie
- Department of Meiosis, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany;
- Current affiliation: School of Biochemistry, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Kathleen Scheffler
- Department of Meiosis, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany;
| | - Melina Schuh
- Department of Meiosis, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany;
| |
Collapse
|
23
|
Wang JC, Lv H, Wu KL, Zhang YS, Luo HN, Chen ZJ. Discs large homologue 1 (Dlg1) coordinates mouse oocyte polarisation during maturation. Reprod Fertil Dev 2018; 29:1699-1707. [PMID: 27651179 DOI: 10.1071/rd15486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 08/19/2016] [Indexed: 11/23/2022] Open
Abstract
Mouse oocyte meiotic division requires the establishment of asymmetries in the oocyte before division, indicating the presence of polarity-establishing molecules. During mouse oocyte maturation proper orientation and positioning of the meiotic spindle at the oocyte cortex, as well as polarity in the oocyte cytoplasm and its oolemma, are necessary for the formation of functional haploid oocytes. Discs large homologue 1 (Dlg1) protein is a conserved protein that regulates cell polarity. In the present study, we found that Dlg1 was expressed at different stages of oocyte development. The localisation of Dlg1 during mouse oocyte maturation and its relationship with the cytoskeleton were analysed. Our data show that at the germinal vesicle stage, Dlg1 was present in the cytoplasm, prominently surrounding the germinal vesicle membrane. During maturation, Dlg1 became highly polarised by associating with the spindle and formed characteristic crescent-shaped accumulations under the cortex. Addition of nocodazole or cytochalasin B into the culture medium at different stages changed the localisation of Dlg1, indicating that the organisation of Dlg1 is a complex multi-step process and is dependent on microtubules and microfilaments. More importantly, we found that silencing of Dlg1 compromised the G2-M transition.
Collapse
Affiliation(s)
- Jun-Chao Wang
- Centre of Reproductive Medicine, Tianjin Central Hospital of Obstetrics and Gynaecology, 156 Nankai Sanma Road, Tianjin 300100, China
| | - Hong Lv
- Centre for Reproductive Medicine, Provincial Hospital Affiliated to Shandong University, Jinan 250001
| | - Ke-Liang Wu
- Centre for Reproductive Medicine, Provincial Hospital Affiliated to Shandong University, Jinan 250001
| | - Yun-Shan Zhang
- Centre of Reproductive Medicine, Tianjin Central Hospital of Obstetrics and Gynaecology, 156 Nankai Sanma Road, Tianjin 300100, China
| | - Hai-Ning Luo
- Centre of Reproductive Medicine, Tianjin Central Hospital of Obstetrics and Gynaecology, 156 Nankai Sanma Road, Tianjin 300100, China
| | - Zi-Jiang Chen
- Centre for Reproductive Medicine, Provincial Hospital Affiliated to Shandong University, Jinan 250001
| |
Collapse
|
24
|
Marlow FL. Recent advances in understanding oogenesis: interactions with the cytoskeleton, microtubule organization, and meiotic spindle assembly in oocytes. F1000Res 2018; 7. [PMID: 29755732 PMCID: PMC5911934 DOI: 10.12688/f1000research.13837.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/12/2018] [Indexed: 01/16/2023] Open
Abstract
Maternal control of development begins with production of the oocyte during oogenesis. All of the factors necessary to complete oocyte maturation, meiosis, fertilization, and early development are produced in the transcriptionally active early oocyte. Active transcription of the maternal genome is a mechanism to ensure that the oocyte and development of the early embryo begin with all of the factors needed for successful embryonic development. To achieve the maximum maternal store, only one functional cell is produced from the meiotic divisions that produce the oocyte. The oocyte receives the bulk of the maternal cytoplasm and thus is significantly larger than its sister cells, the tiny polar bodies, which receive a copy of the maternal genome but essentially none of the maternal cytoplasm. This asymmetric division is accomplished by an enormous cell that is depleted of centrosomes in early oogenesis; thus, meiotic divisions in oocytes are distinct from those of mitotic cells. Therefore, these cells must partition the chromosomes faithfully to ensure euploidy by using mechanisms that do not rely on a conventional centrosome-based mitotic spindle. Several mechanisms that contribute to assembly and maintenance of the meiotic spindle in oocytes have been identified; however, none is fully understood. In recent years, there have been many exciting and significant advances in oogenesis, contributed by studies using a myriad of systems. Regrettably, I cannot adequately cover all of the important advances here and so I apologize to those whose beautiful work has not been included. This review focuses on a few of the most recent studies, conducted by several groups, using invertebrate and vertebrate systems, that have provided mechanistic insight into how microtubule assembly and meiotic spindle morphogenesis are controlled in the absence of centrosomes.
Collapse
Affiliation(s)
- Florence L Marlow
- Department of Cell Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
25
|
Dai X, Lu Y, Zhang M, Miao Y, Zhou C, Cui Z, Xiong B. Melatonin improves the fertilization ability of post-ovulatory aged mouse oocytes by stabilizing ovastacin and Juno to promote sperm binding and fusion. Hum Reprod 2018; 32:598-606. [PMID: 28137755 DOI: 10.1093/humrep/dew362] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/01/2017] [Indexed: 12/14/2022] Open
Abstract
STUDY QUESTION What are the underlying mechanisms of the decline in the fertilization ability of post-ovulatory aged oocytes? SUMMARY ANSWER Melatonin improves the fertilization ability of post-ovulatory aged oocytes by reducing aging-induced reactive oxygen species (ROS) levels and inhibiting apoptosis and by maintaining the levels and localization of the fertilization proteins, ovastacin and Juno. WHAT IS KNOWN ALREADY Following ovulation, the quality of mammalian metaphase II oocytes irreversibly deteriorates over time with a concomitant loss of fertilization ability. Melatonin has been found to prevent post-ovulatory oocyte aging and extend the window for optimal fertilization in mice. STUDY DESIGN, SIZE, DURATION Mouse oocytes were randomly assigned to three groups and aged in vitro for 0, 6, 12 and 24 h, respectively. Increasing concentrations of melatonin (10-9 M, 10-7 M, 10-5 M and 10-3 M) were added to the 24 h aging group. PARTICIPANTS/MATERIALS, SETTING, METHODS Sperm binding assays, in-vitro fertilization, immunofluorescent staining and western blotting were performed to investigate key regulators and events during fertilization of post-ovulatory aged mouse oocytes. MAIN RESULTS AND THE ROLE OF CHANCE We found that the actin cap which promotes a cortical granule (CG) free domain is disrupted with a re-distribution of CGs in the subcortex of aged oocytes. Ovastacin, a CG metalloendoprotease, is mis-located and prematurely exocytosed in aged oocytes with subsequent cleavage of the zona pellucida protein ZP2. This disrupts the sperm recognition domain and dramatically reduces the number of sperm binding to the zona pellucida. The abundance of Juno, the sperm receptor on the oocyte membrane, also is reduced in aged oocytes. Exposure of aged oocytes to melatonin significantly elevates in-vitro fertilization rates potentially by rescuing the above age-associated defects of fertilization, and reducing ROS and inhibiting apoptosis. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION We explored the mechanisms of the decline in fertilization ability decline in aged mouse oocytes, in vitro but not in vivo. WIDER IMPLICATIONS OF THE FINDINGS Our findings may contribute to the development a more efficient method, involving melatonin, for improving IVF success rates. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by the National Natural Science Foundation (31571545) and the Natural Science Foundation of Jiangsu Province (BK20150677). The authors have no conflict of interest to disclose.
Collapse
Affiliation(s)
- Xiaoxin Dai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yajuan Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Mianqun Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yilong Miao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Changyin Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhaokang Cui
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Bo Xiong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
26
|
Panzica MT, McNally FJ. Mechanisms that prevent catastrophic interactions between paternal chromosomes and the oocyte meiotic spindle. Cell Cycle 2018; 17:529-534. [PMID: 29375006 DOI: 10.1080/15384101.2018.1431495] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Meiosis produces haploid gametes by accurately reducing chromosome ploidy through one round of DNA replication and two subsequent rounds of chromosome segregation and cell division. The cell divisions of female meiosis are highly asymmetric and give rise to a large egg and two very small polar bodies that do not contribute to development. These asymmetric divisions are driven by meiotic spindles that are small relative to the size of the egg and have one pole juxtaposed against the cell cortex to promote polar body extrusion. An additional unique feature of female meiosis is that fertilization occurs before extrusion of the second polar body in nearly all animal species. Thus sperm-derived chromosomes are present in the egg during female meiosis. Here, we explore the idea that the asymmetry of female meiosis spatially separates the sperm from the meiotic spindle to prevent detrimental interactions between the spindle and the paternal chromosomes.
Collapse
Affiliation(s)
- Michelle T Panzica
- a Department of Molecular and Cellular Biology , University of California , Davis , Davis , CA , USA
| | - Francis J McNally
- a Department of Molecular and Cellular Biology , University of California , Davis , Davis , CA , USA
| |
Collapse
|
27
|
Li L, Han L, Zhang J, Liu X, Ma R, Hou X, Ge J, Wang Q. Epsin2 promotes polarity establishment and meiotic division through activating Cdc42 in mouse oocyte. Oncotarget 2018; 7:50927-50936. [PMID: 27463009 PMCID: PMC5239448 DOI: 10.18632/oncotarget.10815] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/14/2016] [Indexed: 01/04/2023] Open
Abstract
Epsins are a conserved family of endocytic adaptors essential for diverse biological events. However, its role in oocytes remains completely unknown. Here, we report that specific depletion of Epsin2 in mouse oocytes significantly disrupts meiotic progression. Confocal microscopy reveals that Epsin2 knockdown results in the failure of actin cap formation and polar body extrusion during meiosis, indicative of the importance of Epsin2 in polarity establishment and cytokinesis. In addition, spindle defects and chromosome misalignment are readily observed in oocytes depleted of Epsin2. Moreover, we find that Epsin2 knockdown markedly decreases the activity of Cdc42 in oocytes and importantly, that the dominant-positive mutant of Cdc42 (Cdc42Q61L) is capable of partially rescuing the deficient phenotypes of Epsin2-knockdown oocytes. Together, our data identify Epsin2 as a novel player in regulating oocyte maturation, and demonstrate that Epsin2 promotes polarity establishment and meiotic division via activating Cdc42.
Collapse
Affiliation(s)
- Ling Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Longsen Han
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Jiaqi Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Xiaohui Liu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Rujun Ma
- Center of Reproductive Medicine, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xiaojing Hou
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Juan Ge
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
28
|
Severance AL, Latham KE. PLK1 regulates spindle association of phosphorylated eukaryotic translation initiation factor 4E-binding protein and spindle function in mouse oocytes. Am J Physiol Cell Physiol 2017; 313:C501-C515. [PMID: 28794108 PMCID: PMC5792166 DOI: 10.1152/ajpcell.00075.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 08/03/2017] [Accepted: 08/04/2017] [Indexed: 11/22/2022]
Abstract
Oocyte meiotic spindles are associated with spindle-enriched mRNAs, phosphorylated ribosome protein S6, and phosphorylated variants of the key translational regulator, eukaryotic translation initiation factor 4E-binding protein 1 (eIF4E-BP1), consistent with translational control of localized mRNAs by eIF4E-BP1 in facilitating spindle formation and stability. Using specific kinase inhibitors, we determined which kinases regulate phosphorylation status of eIF4E-BP1 associated with meiotic spindles in mouse oocytes and effects of kinase inhibition on chromosome congression and spindle formation. Neither ataxia telangiectasia-mutated kinase nor mechanistic target of rapamycin inhibition significantly affected phosphorylation status of spindle-associated eIF4E-BP1 at the phosphorylation sites examined. Spindle-associated phospho-eIF4E-BP1, spindle formation, and chromosome congression were strongly disrupted by polo-like kinase I (PLK1) inhibition at both metaphase I (MI) and MII. In addition, direct inhibition of eIF4E-BP1 via 4EGI led to spindle defects at MI, indicating a direct role for eIF4E-BP1 phosphorylation in meiotic spindle formation. PLK1 also regulated microtubule dynamics throughout the ooplasm, indicating likely coordination between spindle dynamics and broader ooplasm cytoskeletal dynamics. Because diverse upstream signaling pathways converge on PLK1, these results implicate PLK1 as a major regulatory nexus coupling endogenous and exogenous signals via eIF4E-BP1 to the regulation of spindle formation and stability.
Collapse
Affiliation(s)
- Ashley L Severance
- Reproductive and Developmental Sciences Program, Michigan State University , East Lansing, Michigan
- Genetics Graduate Program, Michigan State University , East Lansing, Michigan
| | - Keith E Latham
- Reproductive and Developmental Sciences Program, Michigan State University , East Lansing, Michigan
- Genetics Graduate Program, Michigan State University , East Lansing, Michigan
- Department of Animal Science, Michigan State University , East Lansing, Michigan ; and
- Department of Obstetrics, Gynecology, and Reproductive Biology, Michigan State University , East Lansing, Michigan
| |
Collapse
|
29
|
Dal Canto M, Guglielmo MC, Mignini Renzini M, Fadini R, Moutier C, Merola M, De Ponti E, Coticchio G. Dysmorphic patterns are associated with cytoskeletal alterations in human oocytes. Hum Reprod 2017; 32:750-757. [PMID: 28333241 DOI: 10.1093/humrep/dex041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/14/2017] [Indexed: 01/09/2023] Open
Abstract
Study question Are specific morphological anomalies in human mature oocytes, as revealed by transmitted light microscopy, associated with intrinsic damage to the meiotic spindle and actin cytoskeleton? Summary answer Aggregates of smooth endoplasmic reticulum (SER) and domains of centrally localized granular cytoplasm (GC) reflect intrinsic damage to the oocyte cytoskeleton, namely alterations in spindle size, chromosome misalignment and cortical actin disorganization. What is known already In preparation for ICSI, oocytes are often selected for use in treatment by morphological criteria, but the rationale and implications of this practice are controversial. Very little information is available on the relationship between oocyte morphology and intrinsic cellular characteristics, such as the actin cytoskeleton, meiotic spindle and chromosome alignment. Study design, size, duration A total of 170 metaphase II (MII) oocytes were donated by consenting IVF patients and analysed; 62 were classified as morphologically normal (control), 54 had SER clusters and 54 had centrally localized GC. Participants/materials, setting, methods Supernumerary oocytes were fixed within 3 h from recovery and stained for tubulin, chromatin and actin. Spindles were analysed for 1D and 2D characteristics by high-performance confocal microscopy. Chromosomes were classified as scattered or aligned and the conformation and intensity of cortical actin was evaluated. Main results and the role of chance In comparison with control oocytes, both SER and GC oocytes showed greater spindle length (P = 0.033 and 0.003, respectively) and GC oocytes also showed greater spindle width (P= 0.049) and area (P= 0.036). Control and SER oocytes had statistically comparable rates of chromosome displacement from the metaphase plate, unlike GC oocytes where chromosome displacement occurred at higher rate (P = 0.013). In situations where a complete Z-stack was reconstructed from a polar angle, chromosome disposition was classified as being normal when two sets of concentric arrays were visible. Based on these parameters, the proportions of oocytes with normal chromosomal arrangement or partial/total disarrangement was not statistically different between control and SER oocytes. Conversely, in GC oocytes, chromosome disarrangement was higher (P = 0.002). All control oocytes displayed a continuous meshwork of suboolemmal actin, which appeared as an uninterrupted ring in thin optical sections. In contrast, in SER and GC groups, integrity of suboolemmal actin was observed in only 66.7 and 42.9% of oocytes, respectively (P = 0.0001). Large scale data N/A. Limitations reason for caution Only two of several known oocyte dysmorphisms were investigated, while oocyte quality was assessed only by cytoskeletal criteria. Wider implications of the findings This study represents a significant step toward a more objective assessment of oocyte morphology, offering information that can assist embryologists to make a more aware and rationally founded decision on whether, and with what possible implications, oocytes with certain dysmorphic characters should be used for treatment or discarded. More generally, it also demonstrates that morphometric parameters of the cytoskeleton and chromosome organization can be used as biomarkers of oocyte quality. Study funding and competing interest(s) This study was funded by Biogenesi Reproductive Medicine Centre (Monza, Italy). All authors declare no conflict of interests.
Collapse
Affiliation(s)
| | | | - Mario Mignini Renzini
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi, Monza,Italy
| | - Rubens Fadini
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi, Monza,Italy
| | - Clarissa Moutier
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi, Monza,Italy
| | - Maria Merola
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi, Monza,Italy
| | - Elena De Ponti
- Department of Medical Physics, ASST Monza, Via Pergolesi, Monza, Italy
| | - Giovanni Coticchio
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi, Monza,Italy
| |
Collapse
|
30
|
He SW, Xu BH, Liu Y, Wang YL, Chen MH, Xu L, Liao BQ, Lui R, Li FP, Lin YH, Fu XP, Fu BB, Hong ZW, Liu YX, Qi ZQ, Wang HL. SKAP2 regulates Arp2/3 complex for actin-mediated asymmetric cytokinesis by interacting with WAVE2 in mouse oocytes. Cell Cycle 2017; 16:2272-2281. [PMID: 28933599 PMCID: PMC5788478 DOI: 10.1080/15384101.2017.1380126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
SKAP2 (Src kinase-associated phosphoprotein 2), a substrate of Src family kinases, has been suggested to be involved in actin-mediated cellular processes. However, little is known about its role in mouse oocyte maturation. In this study, we thus investigated the expression, localization, and functions of SKAP2 during mouse oocyte asymmetric division. SKAP2 protein expression was detected at all developmental stages in mouse oocytes. Immunofluorescent staining showed that SKAP2 was mainly distributed at the cortex of the oocytes during maturation. Treatment with cytochalasin B in oocytes confirmed that SKAP2 was co-localized with actin. Depletion of SKAP2 by injection with specific short interfering RNA caused failure of spindle migration, polar body extrusion, and cytokinesis defects. Meanwhile, the staining of actin filaments at the oocyte membrane and in the cytoplasm was significantly reduced after these treatments. SKAP2 depletion also disrupted actin cap and cortical granule-free domain formation, and arrested a large proportion of oocytes at the telophase stage. Moreover, Arp2/3 complex and WAVE2 expression was decreased after the depletion of SKAP2 activity. Our results indicate that SKAP2 regulates the Arp2/3 complex and is essential for actin-mediated asymmetric cytokinesis by interacting with WAVE2 in mouse oocytes.
Collapse
Affiliation(s)
- Shu-Wen He
- a Organ Transplantation Institute, Medical College, Xiamen University , Xiamen, Fujian , China.,b Fujian Key Laboratory of Organ and Tissue Regeneration , Xiamen, Fujian , China
| | - Bai-Hui Xu
- a Organ Transplantation Institute, Medical College, Xiamen University , Xiamen, Fujian , China.,g Department of Workshop 25, Shangdong new time Pharmaceutical Company Limited , Shangdong , China
| | - Yu Liu
- a Organ Transplantation Institute, Medical College, Xiamen University , Xiamen, Fujian , China.,b Fujian Key Laboratory of Organ and Tissue Regeneration , Xiamen, Fujian , China
| | - Ya-Long Wang
- a Organ Transplantation Institute, Medical College, Xiamen University , Xiamen, Fujian , China.,b Fujian Key Laboratory of Organ and Tissue Regeneration , Xiamen, Fujian , China
| | - Ming-Huang Chen
- a Organ Transplantation Institute, Medical College, Xiamen University , Xiamen, Fujian , China.,b Fujian Key Laboratory of Organ and Tissue Regeneration , Xiamen, Fujian , China.,c Department of Gynaecology and Obstetrics , Zhongshan Hospital , Xiamen, Fujian , China
| | - Lin Xu
- a Organ Transplantation Institute, Medical College, Xiamen University , Xiamen, Fujian , China.,b Fujian Key Laboratory of Organ and Tissue Regeneration , Xiamen, Fujian , China
| | - Bao-Qiong Liao
- a Organ Transplantation Institute, Medical College, Xiamen University , Xiamen, Fujian , China.,b Fujian Key Laboratory of Organ and Tissue Regeneration , Xiamen, Fujian , China
| | - Rui Lui
- c Department of Gynaecology and Obstetrics , Zhongshan Hospital , Xiamen, Fujian , China.,d Department of Gynaecology and Obstetrics , Zhongxin Hospital , Qingdao, Shangdong , China
| | - Fei-Ping Li
- b Fujian Key Laboratory of Organ and Tissue Regeneration , Xiamen, Fujian , China.,f Department of Life Science, Biological College, Southwest Forestry University , Kunming , China
| | - Yan-Hong Lin
- a Organ Transplantation Institute, Medical College, Xiamen University , Xiamen, Fujian , China.,e Department of Gynaecology and Obstetrics , the First Clinical Medical College, Fujian Medical University , Fuzhou , China
| | - Xian-Pei Fu
- a Organ Transplantation Institute, Medical College, Xiamen University , Xiamen, Fujian , China.,b Fujian Key Laboratory of Organ and Tissue Regeneration , Xiamen, Fujian , China
| | - Bin-Bin Fu
- a Organ Transplantation Institute, Medical College, Xiamen University , Xiamen, Fujian , China.,b Fujian Key Laboratory of Organ and Tissue Regeneration , Xiamen, Fujian , China
| | - Zi-Wei Hong
- a Organ Transplantation Institute, Medical College, Xiamen University , Xiamen, Fujian , China.,b Fujian Key Laboratory of Organ and Tissue Regeneration , Xiamen, Fujian , China
| | - Yu-Xin Liu
- a Organ Transplantation Institute, Medical College, Xiamen University , Xiamen, Fujian , China.,b Fujian Key Laboratory of Organ and Tissue Regeneration , Xiamen, Fujian , China
| | - Zhong-Quan Qi
- a Organ Transplantation Institute, Medical College, Xiamen University , Xiamen, Fujian , China.,b Fujian Key Laboratory of Organ and Tissue Regeneration , Xiamen, Fujian , China
| | - Hai-Long Wang
- a Organ Transplantation Institute, Medical College, Xiamen University , Xiamen, Fujian , China.,b Fujian Key Laboratory of Organ and Tissue Regeneration , Xiamen, Fujian , China
| |
Collapse
|
31
|
Pan MH, Wang F, Lu Y, Tang F, Duan X, Zhang Y, Xiong B, Sun SC. FHOD1 regulates cytoplasmic actin-based spindle migration for mouse oocyte asymmetric cell division. J Cell Physiol 2017; 233:2270-2278. [PMID: 28708292 DOI: 10.1002/jcp.26099] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 07/13/2017] [Indexed: 12/25/2022]
Abstract
FHOD1 is a member of Diaphanous-related formins (DRFs) which belongs to the Formin family. Previous studies have shown that the DFRs might affect several cellular functions such as morphogenesis, cytokinesis, cell polarity, and embryonic differentiation. However, there is no evidence showing the functions of FHOD1 during oocyte meiosis. This study is aimed at exploring the roles of FHOD1 during the mammalian oocyte maturation. Immunofluorescent staining showed that FHOD1 was restricted to the nucleus in germinal vesicle (GV) stage of the oocytes, after the GV breakdown FHOD1 was primarily located at two poles of the spindle at both metaphases I and II stages. Knockdown of FHOD1 by siRNA injection did not affect polar body extrusion but generated the large polar bodies. In addition, we observed the spindle migration failure in metaphase I oocytes, with a large number of meiotic spindles anchoring in the center of cytoplasm. The expression level of cytoplasmic actin but not cortex actin was significantly reduced, indicating that FHOD1 regulates cytoplasmic actin distribution for the spindle movement. Furthermore, we found that the disruption of ROCK (the Rho-dependent protein kinase) with inhibitor Y-27632 caused the decreased FHOD1 protein expression. Therefore, our data indicate that FHOD1 is regulated by ROCK for cytoplasm actin assembly and spindle migration during mouse oocyte meiosis.
Collapse
Affiliation(s)
- Meng-Hao Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Fei Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yujie Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Feng Tang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xing Duan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yu Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Bo Xiong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
32
|
Chaigne A, Terret ME, Verlhac MH. Asymmetries and Symmetries in the Mouse Oocyte and Zygote. Results Probl Cell Differ 2017; 61:285-299. [PMID: 28409310 DOI: 10.1007/978-3-319-53150-2_13] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mammalian oocytes grow periodically after puberty thanks to the dialogue with their niche in the follicle. This communication between somatic and germ cells promotes the accumulation, inside the oocyte, of maternal RNAs, proteins and other molecules that will sustain the two gamete divisions and early embryo development up to its implantation. In order to preserve their stock of maternal products, oocytes from all species divide twice minimizing the volume of their daughter cells to their own benefit. For this, they undergo asymmetric divisions in size where one main objective is to locate the division spindle with its chromosomes off-centred. In this chapter, we will review how this main objective is reached with an emphasis on the role of actin microfilaments in this process in mouse oocytes, the most studied example in mammals. This chapter is subdivided into three parts: I-General features of asymmetric divisions in mouse oocytes, II-Mechanism of chromosome positioning by actin in mouse oocytes and III-Switch from asymmetric to symmetric division at the oocyte-to-embryo transition.
Collapse
Affiliation(s)
- Agathe Chaigne
- MRC Laboratory for Molecular Cell Biology, UCL, London, WC1E 6BT, UK.,Institute for the Physics of Living Systems, UCL, London, WC1E 6BT, UK
| | | | | |
Collapse
|
33
|
Chen MH, Liu Y, Wang YL, Liu R, Xu BH, Zhang F, Li FP, Xu L, Lin YH, He SW, Liao BQ, Fu XP, Wang XX, Yang XJ, Wang HL. KIF2A regulates the spindle assembly and the metaphase I-anaphase I transition in mouse oocyte. Sci Rep 2016; 6:39337. [PMID: 27991556 PMCID: PMC5171862 DOI: 10.1038/srep39337] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/18/2016] [Indexed: 11/09/2022] Open
Abstract
KIF2A, a member of the kinesin-13 family, has been reported to play a role in spindle assembly in mitosis. However, its function in mammalian meiosis remains unknown. In this research, we examined the expression, localization and function of KIF2A during mouse oocyte meiosis. KIF2A was expressed in some key stages in mouse oocyte meiosis. Immunofluorescent staining showed that KIF2A distributed in the germinal vesicle at the germinal vesicle stage and as the spindle assembling after meiosis resumption, KIF2A gradually accumulated to the entire spindle. The treatment of oocytes with taxol and nocodazole demonstrated that KIF2A was co-localized with α-tubulin. Depletion of KIF2A by specific short interfering (si) RNA injection resulted in abnormal spindle assembly, failure of spindle migration, misaligned chromosomes and asymmetric cell division. Meanwhile, SKA1 expression level was decreased and the TACC3 localization was disrupted. Moreover, depletion of KIF2A disrupted the actin cap formation, arrested oocytes at metaphase I with spindle assembly checkpoint protein BubR1 activated and finally reduced the rate of the first polar body extrusion. Our data indicate that KIF2A regulates the spindle assembly, asymmetric cytokinesis and the metaphase I-anaphase I transition in mouse oocyte.
Collapse
Affiliation(s)
- Ming-Huang Chen
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen 361000, Fujian, China.,Department of Gynaecology and Obstetrics, Zhongshan Hospital, Xiamen University, Xiamen 361000, Fujian, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen 361000, Fujian, China
| | - Yu Liu
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen 361000, Fujian, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen 361000, Fujian, China
| | - Ya-Long Wang
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen 361000, Fujian, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen 361000, Fujian, China
| | - Rui Liu
- Department of Gynaecology and Obstetrics, Zhongshan Hospital, Xiamen University, Xiamen 361000, Fujian, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen 361000, Fujian, China.,Department of Gynaecology and Obstetrics, Zhongxin Hospital, Qingdao 266000, Shandong, China
| | - Bai-Hui Xu
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen 361000, Fujian, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen 361000, Fujian, China
| | - Fei Zhang
- Department of Gynaecology and Obstetrics, Zhongshan Hospital, Xiamen University, Xiamen 361000, Fujian, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen 361000, Fujian, China
| | - Fei-Ping Li
- Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen 361000, Fujian, China.,Biological College, Southwest Forestry University, Kunming 650000, Yunnan, China
| | - Lin Xu
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen 361000, Fujian, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen 361000, Fujian, China
| | - Yan-Hong Lin
- Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen 361000, Fujian, China.,Department of Gynaecology and Obstetrics, The First Clinical Medical College, Fujian Medical University, Fuzhou 350000, Fujian, China
| | - Shu-Wen He
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen 361000, Fujian, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen 361000, Fujian, China
| | - Bao-Qiong Liao
- Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen 361000, Fujian, China.,Department of Gynaecology and Obstetrics, Dongfang Hospital, Xiamen University, Fuzhou 350000, Fujian, China
| | - Xian-Pei Fu
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen 361000, Fujian, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen 361000, Fujian, China
| | - Xiao-Xue Wang
- Department of Gynaecology and Obstetrics, Zhongshan Hospital, Xiamen University, Xiamen 361000, Fujian, China
| | - Xiang-Jun Yang
- Department of Gynaecology and Obstetrics, Zhongshan Hospital, Xiamen University, Xiamen 361000, Fujian, China
| | - Hai-Long Wang
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen 361000, Fujian, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen 361000, Fujian, China
| |
Collapse
|
34
|
Van Blerkom J, Zimmermann S. Ganglioside-enriched microdomains define an oolemma that is functionally polarized with respect to fertilizability in the mouse. Reprod Biomed Online 2016; 33:458-475. [DOI: 10.1016/j.rbmo.2016.06.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/17/2016] [Accepted: 06/29/2016] [Indexed: 10/21/2022]
|
35
|
Nannas NJ, Higgins DM, Dawe RK. Anaphase asymmetry and dynamic repositioning of the division plane during maize meiosis. J Cell Sci 2016; 129:4014-4024. [PMID: 27609836 DOI: 10.1242/jcs.194860] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/05/2016] [Indexed: 01/12/2023] Open
Abstract
The success of an organism is contingent upon its ability to transmit genetic material through meiotic cell division. In plant meiosis I, the process begins in a large spherical cell without physical cues to guide the process. Yet, two microtubule-based structures, the spindle and phragmoplast, divide the chromosomes and the cell with extraordinary accuracy. Using a live-cell system and fluorescently labeled spindles and chromosomes, we found that the process self- corrects as meiosis proceeds. Metaphase spindles frequently initiate division off-center, and in these cases anaphase progression is asymmetric with the two masses of chromosomes traveling unequal distances on the spindle. The asymmetry is compensatory, such that the chromosomes on the side of the spindle that is farthest from the cell cortex travel a longer distance at a faster rate. The phragmoplast forms at an equidistant point between the telophase nuclei rather than at the original spindle mid-zone. This asymmetry in chromosome movement implies a structural difference between the two halves of a bipolar spindle and could allow meiotic cells to dynamically adapt to errors in metaphase and accurately divide the cell volume.
Collapse
Affiliation(s)
- Natalie J Nannas
- Department of Plant Biology, University of Georgia, Athens, GA 30605, USA
| | - David M Higgins
- Department of Plant Biology, University of Georgia, Athens, GA 30605, USA
| | - R Kelly Dawe
- Department of Plant Biology, University of Georgia, Athens, GA 30605, USA .,Department of Genetics, University of Georgia, Athens, GA 30605, USA
| |
Collapse
|
36
|
Tropomodulin-3 is essential in asymmetric division during mouse oocyte maturation. Sci Rep 2016; 6:29204. [PMID: 27374327 PMCID: PMC4931587 DOI: 10.1038/srep29204] [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: 01/21/2016] [Accepted: 06/16/2016] [Indexed: 11/08/2022] Open
Abstract
The dynamic polymerization and depolymerization of actin filaments is essential for various cellular processes such as cell migration, rotation, cytokinesis, and mammalian oocyte maturation. Tropomodulin 3 (Tmod3) binds to the slow-growing (pointed) ends of the actin filament, thereby protecting the filament from depolymerization. However, the roles of Tmod3 in mammalian oocyte maturation remain elusive. Tmod3 mRNA and protein is present at all stages of mouse oocyte maturation. Tmod3 protein is mainly localized in the cytoplasm and appears enriched near the chromosome during maturation. By knocking down or ectopically overexpressing Tmod3, we confirmed that Tmod3 regulate the level of the intracytoplasmic actin mesh and asymmetric spindle migration. Expression of N-terminal Tmod3 (correspond to 1–155 amino acids), which contains the tropomyosin-binding site, results in decreased density of the actin mesh, thereby demonstrating the importance of the interaction between tropomyosin and tropomodulin for the maintenance of the actin mesh. Taken together, these findings indicate that Tmod3 plays crucial roles in oocyte maturation, presumably by protecting the actin filament from depolymerization and thereby controlling the density of the cytoplasmic actin mesh.
Collapse
|
37
|
Mackenzie ACL, Kyle DD, McGinnis LA, Lee HJ, Aldana N, Robinson DN, Evans JP. Cortical mechanics and myosin-II abnormalities associated with post-ovulatory aging: implications for functional defects in aged eggs. Mol Hum Reprod 2016; 22:397-409. [PMID: 26921397 PMCID: PMC4884917 DOI: 10.1093/molehr/gaw019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 02/12/2016] [Accepted: 02/24/2016] [Indexed: 12/20/2022] Open
Abstract
STUDY HYPOTHESIS Cellular aging of the egg following ovulation, also known as post-ovulatory aging, is associated with aberrant cortical mechanics and actomyosin cytoskeleton functions. STUDY FINDING Post-ovulatory aging is associated with dysfunction of non-muscle myosin-II, and pharmacologically induced myosin-II dysfunction produces some of the same deficiencies observed in aged eggs. WHAT IS KNOWN ALREADY Reproductive success is reduced with delayed fertilization and when copulation or insemination occurs at increased times after ovulation. Post-ovulatory aged eggs have several abnormalities in the plasma membrane and cortex, including reduced egg membrane receptivity to sperm, aberrant sperm-induced cortical remodeling and formation of fertilization cones at the site of sperm entry, and reduced ability to establish a membrane block to prevent polyspermic fertilization. STUDY DESIGN, SAMPLES/MATERIALS, METHODS Ovulated mouse eggs were collected at 21-22 h post-human chorionic gonadotrophin (hCG) (aged eggs) or at 13-14 h post-hCG (young eggs), or young eggs were treated with the myosin light chain kinase (MLCK) inhibitor ML-7, to test the hypothesis that disruption of myosin-II function could mimic some of the effects of post-ovulatory aging. Eggs were subjected to various analyses. Cytoskeletal proteins in eggs and parthenogenesis were assessed using fluorescence microscopy, with further analysis of cytoskeletal proteins in immunoblotting experiments. Cortical tension was measured through micropipette aspiration assays. Egg membrane receptivity to sperm was assessed in in vitro fertilization (IVF) assays. Membrane topography was examined by low-vacuum scanning electron microscopy (SEM). MAIN RESULTS AND THE ROLE OF CHANCE Aged eggs have decreased levels and abnormal localizations of phosphorylated myosin-II regulatory light chain (pMRLC; P = 0.0062). Cortical tension, which is mediated in part by myosin-II, is reduced in aged mouse eggs when compared with young eggs, by ∼40% in the cortical region where the metaphase II spindle is sequestered and by ∼50% in the domain to which sperm bind and fuse (P < 0.0001). Aging-associated parthenogenesis is partly rescued by treating eggs with a zinc ionophore (P = 0.003), as is parthenogenesis induced by inhibition of mitogen-activated kinase (MAPK) 3/1 [also known as extracellular signal-regulated kinase (ERK)1/2] or MLCK. Inhibition of MLCK with ML-7 also results in effects that mimic those of post-ovulatory aging: fertilized ML-7-treated eggs show both impaired fertilization and increased extents of polyspermy, and ML-7-treated young eggs have several membrane abnormalities that are shared by post-ovulatory aged eggs. LIMITATIONS, REASONS FOR CAUTION These studies were done with mouse oocytes, and it remains to be fully determined how these findings from mouse oocytes would compare with other species. For studies using methods not amenable to analysis of large sample sizes and data are limited to what images one can capture (e.g. SEM), data should be interpreted conservatively. WIDER IMPLICATIONS OF THE FINDINGS These data provide insights into causes of reproductive failures at later post-copulatory times. LARGE SCALE DATA Not applicable. STUDY FUNDING AND COMPETING INTERESTS This project was supported by R01 HD037696 and R01 HD045671 from the NIH to J.P.E. Cortical tension studies were supported by R01 GM66817 to D.N.R. The authors declare there are no financial conflicts of interest.
Collapse
Affiliation(s)
- Amelia C L Mackenzie
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD 21205, USA
| | - Diane D Kyle
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD 21205, USA
| | - Lauren A McGinnis
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD 21205, USA
| | - Hyo J Lee
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD 21205, USA
| | - Nathalia Aldana
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD 21205, USA
| | - Douglas N Robinson
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Janice P Evans
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St, Baltimore, MD 21205, USA
| |
Collapse
|
38
|
Abstract
Calcineurin is required for oocyte exit from meiotic block in metaphase II (MII) stage in invertebrates and also in lower vertebrates. However, the role of calcineurin in mammalian oocyte activation is still unclear. The aim of this study was to determine whether calcineurin is involved in the processes regulating porcine oocyte activation. Indirect immunofluorescence demonstrated localization of both calcineurin subunits, CnA and CnB, especially in the cortex area of MII oocytes, in vitro fertilized and also parthenogenetically activated oocytes. After activation, the fluorescence intensity of the protein in the cortex area of oocytes remains unchanged; the protein calcineurin in the cytoplasm was recorded mainly around the pronuclei. Treatment of matured oocytes with calcineurin inhibitors, cyclosporin A (CsA) and hymenistatin I (HS-I), followed by activation with calcium ionophore A23187, significantly decreased the rate of activated oocytes compared to oocytes that were treated only with calcium ionophore (Ca-Io), (CsA+Ca-Io 25.0% v. Ca-Io 83.3%; HS-I+Ca-Io 32.5% v. Ca-Io 85.0%). Compared to the control, CsA treatment of matured oocytes followed by activation with Ca-Io did not affect the activity level of metaphase-promoting factor (MPF) and mitogen-activated protein kinase (MAPK) in activated oocytes evaluated by kinase activity assay. Simultaneous staining of calcineurin and cortical granule content in matured oocytes showed that calcineurin distributed in the cortical area of the oocyte has not been colocalized with cortical granules content. On the other hand, the calcineurin inhibition before parthenogenetic activation leads to a reduction of the cortical reaction level compared to oocytes that were not treated with CsA (complete exocytosis: CsA+Ca-Io 2.6% v. Ca-Io 83.9%; sum of cortical granule brightness: CsA + Ca-Io 0.69 v. Ca-Io 0.15). Our results showed that calcineurin is involved in the process of pig oocyte activation and cortical granule exocytosis; however this regulation seems to be MPF and MAPK independent.
Collapse
|
39
|
Namgoong S, Kim NH. Roles of actin binding proteins in mammalian oocyte maturation and beyond. Cell Cycle 2016; 15:1830-43. [PMID: 27152960 DOI: 10.1080/15384101.2016.1181239] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Actin nucleation factors, which promote the formation of new actin filaments, have emerged in the last decade as key regulatory factors controlling asymmetric division in mammalian oocytes. Actin nucleators such as formin-2, spire, and the ARP2/3 complex have been found to be important regulators of actin remodeling during oocyte maturation. Another class of actin-binding proteins including cofilin, tropomyosin, myosin motors, capping proteins, tropomodulin, and Ezrin-Radixin-Moesin proteins are thought to control actin cytoskeleton dynamics at various steps of oocyte maturation. In addition, actin dynamics controlling asymmetric-symmetric transitions after fertilization is a new area of investigation. Taken together, defining the mechanisms by which actin-binding proteins regulate actin cytoskeletons is crucial for understanding the basic biology of mammalian gamete formation and pre-implantation development.
Collapse
Affiliation(s)
- Suk Namgoong
- a Department of Animal Sciences , Chungbuk National University , Cheong-Ju , ChungChungBuk-do , Republic of Korea
| | - Nam-Hyung Kim
- a Department of Animal Sciences , Chungbuk National University , Cheong-Ju , ChungChungBuk-do , Republic of Korea
| |
Collapse
|
40
|
Abstract
Sexual reproduction is essential for many organisms to propagate themselves. It requires the formation of haploid female and male gametes: oocytes and sperms. These specialized cells are generated through meiosis, a particular type of cell division that produces cells with recombined genomes that differ from their parental origin. In this review, we highlight the end process of female meiosis, the divisions per se, and how they can give rise to a functional female gamete preparing itself for the ensuing zygotic development. In particular, we discuss why such an essential process in the propagation of species is so poorly controlled, producing a strong percentage of abnormal female gametes in the end. Eventually, we examine aspects related to the lack of centrosomes in female oocytes, the asymmetry in size of the mammalian oocyte upon division, and in mammals the direct consequences of these long-lived cells in the ovary.
Collapse
|
41
|
Chaigne A, Campillo C, Voituriez R, Gov NS, Sykes C, Verlhac MH, Terret ME. F-actin mechanics control spindle centring in the mouse zygote. Nat Commun 2016; 7:10253. [PMID: 26727405 PMCID: PMC4725770 DOI: 10.1038/ncomms10253] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 11/20/2015] [Indexed: 01/01/2023] Open
Abstract
Mitotic spindle position relies on interactions between astral microtubules nucleated by centrosomes and a rigid cortex. Some cells, such as mouse oocytes, do not possess centrosomes and astral microtubules. These cells rely only on actin and on a soft cortex to position their spindle off-centre and undergo asymmetric divisions. While the first mouse embryonic division also occurs in the absence of centrosomes, it is symmetric and not much is known on how the spindle is positioned at the exact cell centre. Using interdisciplinary approaches, we demonstrate that zygotic spindle positioning follows a three-step process: (1) coarse centring of pronuclei relying on the dynamics of an F-actin/Myosin-Vb meshwork; (2) fine centring of the metaphase plate depending on a high cortical tension; (3) passive maintenance at the cell centre. Altogether, we show that F-actin-dependent mechanics operate the switch between asymmetric to symmetric division required at the oocyte to embryo transition.
Collapse
Affiliation(s)
- Agathe Chaigne
- CIRB, Collège de France, and CNRS-UMR7241 and INSERM-U1050, Equipe labellisée Ligue contre le Cancer, Paris F-75005, France
| | - Clément Campillo
- Université Evry Val d'Essonne, LAMBE, Boulevard F Mitterrand, Evry 91025, France
| | | | - Nir S Gov
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Cécile Sykes
- CNRS-UMR168, Paris F-75248, France.,UPMC, 4 Place Jussieu, Paris F-75248, France.,Institut Curie, Centre de Recherche, Laboratoire Physico-Chimie, Paris F-75248, France
| | - Marie-Hélène Verlhac
- CIRB, Collège de France, and CNRS-UMR7241 and INSERM-U1050, Equipe labellisée Ligue contre le Cancer, Paris F-75005, France
| | - Marie-Emilie Terret
- CIRB, Collège de France, and CNRS-UMR7241 and INSERM-U1050, Equipe labellisée Ligue contre le Cancer, Paris F-75005, France
| |
Collapse
|
42
|
Duan X, Liu J, Zhu CC, Wang QC, Cui XS, Kim NH, Xiong B, Sun SC. RhoA-mediated MLC2 regulates actin dynamics for cytokinesis in meiosis. Cell Cycle 2015; 15:471-7. [PMID: 26701676 DOI: 10.1080/15384101.2015.1128590] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
During oocyte meiosis, the bipolar spindle forms in the central cytoplasm and then migrates to the cortex. Subsequently, the oocyte extrudes the polar body through two successive asymmetric divisions, which are regulated primarily by actin filaments. Myosin light chain2 (MLC2) phosphorylation plays pivotal roles in smooth muscle contraction, stress fiber formation, cell motility and cytokinesis. However, whether MLC2 phosphorylation participates in the oocyte polarization and asymmetric division has not been clarified. The present study investigated the expression and functions of MLC2 during mouse oocyte meiosis. Our result showed that p-MLC2 was localized in the oocyte cortex, with a thickened cap above the chromosomes. Meanwhile, p-MLC2 was also localized in the poles of spindle. Disruption of MLC2 activity by MLC2 knock down (KD) caused the failure of polar body extrusion. Immunofluorescent staining showed that a large proportion of oocytes arrested in telophase stage and failed to undergo cytokinesis after culturing for 12 hours. In the meantime, actin filament staining at oocyte membrane and cytoplasm were reduced in MLC2 KD oocytes. Finally, we found that the phosphorylation of MLC2 protein levels was decreased after disruption of RhoA activity. Above all, our data indicated that the RhoA-mediated MLC2 regulates the actin organization for cytokinesis during mouse oocyte maturation.
Collapse
Affiliation(s)
- Xing Duan
- a College of Animal Science and Technology, Nanjing Agricultural University , Nanjing , China
| | - Jun Liu
- a College of Animal Science and Technology, Nanjing Agricultural University , Nanjing , China
| | - Cheng-Cheng Zhu
- a College of Animal Science and Technology, Nanjing Agricultural University , Nanjing , China
| | - Qiao-Chu Wang
- a College of Animal Science and Technology, Nanjing Agricultural University , Nanjing , China
| | - Xiang-Shun Cui
- b Department of Animal Sciences , Chungbuk National University , Cheongju , Korea
| | - Nam-Hyung Kim
- b Department of Animal Sciences , Chungbuk National University , Cheongju , Korea
| | - Bo Xiong
- a College of Animal Science and Technology, Nanjing Agricultural University , Nanjing , China
| | - Shao-Chen Sun
- a College of Animal Science and Technology, Nanjing Agricultural University , Nanjing , China
| |
Collapse
|
43
|
Nguyen H, Ortega MA, Ko M, Marh J, Ward WS. ORC4 surrounds extruded chromatin in female meiosis. J Cell Biochem 2015; 116:778-86. [PMID: 25502171 DOI: 10.1002/jcb.25033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 12/09/2014] [Indexed: 12/19/2022]
Abstract
Six proteins, ORC1-6, make up the origin recognition complex (ORC) that initiates licensing of DNA replication origins. We have previously reported that subunit ORC2 is localized between the separating maternal chromosomes at anaphase II just after fertilization and is present in zygotic pronuclei at G1. Here, we found that ORC1, 3, and 5 all localize between the chromosomes at anaphase II, but could not be detected in zygotic G1. ORC6 localized to the periphery of the nucleoli at all zygotic stages. We identified an unexpected potential role for ORC4 in polar body formation. We found that in both female meiotic divisions, ORC4 surrounds the set of chromosomes, as a sphere-like structure, that will eventually be discarded in the polar bodies, but not the chromosomes that segregate into the oocyte. None of the other five ORC proteins are involved in this structure. In Zygotic G1, ORC4 surrounds the nuclei of the polar bodies, but was not detectable in the pronuclei. When the zygote entered mitosis ORC4 was only detected in the polar body. However, ORC4 appeared on both sets of separating chromosomes at telophase. At this point, the ORC4 that was in the polar body also migrated into the nuclei, suggesting that ORC4 or an associated protein is modified during the first embryonic cell cycle to allow it to bind DNA. Our results suggest that ORC4 may help identify the chromosomes that are destined to be expelled in the polar body, and may play a role in polar body extrusion. ORC4 surrounds the chromatin that will be extruded in the polar body in both female meiotic divisions, then makes a transition from the cytoplasm to the chromosomes at zygotic anaphase, suggesting multiple roles for this replication licensing protein.
Collapse
Affiliation(s)
- Hieu Nguyen
- Institute for Biogenesis Research Department of Anatomy, Biochemistry & Physiology, and the Department of Obstetrics and Gynecology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| | | | | | | | | |
Collapse
|
44
|
Jeon Y, Yoon JD, Cai L, Hwang SU, Kim E, Zheng Z, Jeung E, Lee E, Hyun SH. Zinc deficiency during in vitro maturation of porcine oocytes causes meiotic block and developmental failure. Mol Med Rep 2015; 12:5973-82. [PMID: 26238161 DOI: 10.3892/mmr.2015.4125] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 06/23/2015] [Indexed: 11/06/2022] Open
Abstract
The present study investigated the effects of zinc deficiency during in vitro maturation (IVM) of porcine oocytes. Zinc deficiency was induced by administering the membrane‑permeable zinc chelator N,N,N',N'‑tetrakis‑(2‑pyridylmethyl)‑ethylendiamine (TPEN). First, the effects of zinc deficiency during IVM on a TPEN‑treated group and a TPEN+zinc-treated group compared with a control group were assessed. The oocyte maturation rates and subsequent embryonic developmental competence of the TPEN+zinc‑treated oocytes were similar to those of the control oocytes (metaphase II [MII] rate, 93.0 and 92.7%, respectively, and blastocyst [BL] formation rate, 42.0 and 40.0%, respectively). These results were significantly different from those obtained for the TPEN‑treated oocytes (MII rate, 0.61%; BL formation rate, 0%). Although the TPEN‑treated oocytes were arrested at metaphase I (MI), the distribution of microtubules was normal. However, microfilament formation was abnormal in the TPEN‑treated oocytes. Furthermore, the effect of a temporary zinc deficiency during IVM on oocyte maturation and subsequent embryonic development was assessed. TPEN (10 µM) was added to the IVM medium for 0, 7, 15 or 22 h. The 0 h‑treated oocytes showed an 83.9% MII rate, while the 7 h‑treated oocytes had a significantly lower MII rate (44.8%). Most of the 15- and 22 h‑treated oocytes were arrested at MI (MI rate: 98.0 and 97.2%, respectively; MII rate, 0% in both groups). Reductions in the BL formation were dependent on the TPEN treatment duration (29.3, 9.2, 0, and 0% after 0, 7, 15 and 22 h, respectively). In conclusion, zinc is an essential element for successful oocyte maturation and embryonic development in pigs. Zinc deficiency caused a meiotic block and had lasting effects on early embryonic development.
Collapse
Affiliation(s)
- Yubyeol Jeon
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361‑763, Republic of Korea
| | - Junchul David Yoon
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361‑763, Republic of Korea
| | - Lian Cai
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361‑763, Republic of Korea
| | - Seon-Ung Hwang
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361‑763, Republic of Korea
| | - Eunhye Kim
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361‑763, Republic of Korea
| | - Zhong Zheng
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361‑763, Republic of Korea
| | - Euibae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361‑763, Republic of Korea
| | - Eunsong Lee
- Laboratory of Theriogenology, College of Veterinary Medicine, Kangwon National University, Chuncheon, Gangwon 200-710, Republic of Korea
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361‑763, Republic of Korea
| |
Collapse
|
45
|
Chen L, Ge ZJ, Wang ZB, Sun T, Ouyang YC, Sun QY, Sun YP. TGN38 is required for the metaphase I/anaphase I transition and asymmetric cell division during mouse oocyte meiotic maturation. Cell Cycle 2015; 13:2723-32. [PMID: 25486359 DOI: 10.4161/15384101.2015.945828] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The cellular functions of the trans-Golgi network protein TGN38 remain unknown. In this research, we studied the expression, localization and functions of TGN38 in the meiotic maturation of mouse oocytes. TGN38 was expressed at every stage of oocyte meiotic maturation and colocalized with γ-tubulin at metaphase I and metaphase II. The spindle microtubule disturbing agents nocodazole and taxol did not affect the colocalization of TGN38 and γ-tubulin. Depletion of TGN38 with specific siRNAs resulted in increased metaphase I arrest, accompanied with spindle assembly checkpoint activation and decreased first polar extrusion (PB1). In the oocytes that had extruded the PB1 after the depletion of TGN38, symmetric division occurred, leading to the production of 2 similarly sized cells. Moreover, the peripheral migration of metaphase I spindle and actin cap formation were impaired in TGN38-depleted oocytes. Our data suggest that TGN38 may regulate the metaphase I/anaphase I transition and asymmetric cell division in mouse oocytes.
Collapse
Affiliation(s)
- Lei Chen
- a Reproductive Medicine Center ; First Affiliated Hospital of Zhengzhou University ; Zhengzhou , Henan Province , China
| | | | | | | | | | | | | |
Collapse
|
46
|
Kim HC, Jo YJ, Kim NH, Namgoong S. Small molecule inhibitor of formin homology 2 domains (SMIFH2) reveals the roles of the formin family of proteins in spindle assembly and asymmetric division in mouse oocytes. PLoS One 2015; 10:e0123438. [PMID: 25837661 PMCID: PMC4383420 DOI: 10.1371/journal.pone.0123438] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/03/2015] [Indexed: 11/19/2022] Open
Abstract
Dynamic actin reorganization is the main driving force for spindle migration and asymmetric cell division in mammalian oocytes. It has been reported that various actin nucleators including Formin-2 are involved in the polarization of the spindle and in asymmetric cell division. In mammals, the formin family is comprised of 15 proteins. However, their individual roles in spindle migration and/or asymmetric division have not been elucidated yet. In this study, we employed a newly developed inhibitor for formin family proteins, small molecule inhibitor of formin homology 2 domains (SMIFH2), to assess the functions of the formin family in mouse oocyte maturation. Treatment with SMIFH2 during in vitro maturation of mouse oocytes inhibited maturation by decreasing cytoplasmic and cortical actin levels. In addition, treatment with SMIFH2, especially at higher concentrations (500 μM), impaired the proper formation of meiotic spindles, indicating that formins play a role in meiotic spindle formation. Knockdown of the mDia2 formins caused a similar decrease in oocyte maturation and abnormal spindle morphology, mimicking the phenotype of SMIFH2-treated cells. Collectively, these results suggested that besides Formin-2, the other proteins of the formin, including mDia family play a role in asymmetric division and meiotic spindle formation in mammalian oocytes.
Collapse
Affiliation(s)
- Hak-Cheol Kim
- Department of Animal Sciences, Chungbuk National University, Cheong-Ju, ChungBuk, Republic of Korea
| | - Yu-Jin Jo
- Department of Animal Sciences, Chungbuk National University, Cheong-Ju, ChungBuk, Republic of Korea
| | - Nam-Hyung Kim
- Department of Animal Sciences, Chungbuk National University, Cheong-Ju, ChungBuk, Republic of Korea
- * E-mail: (SN); (NHK)
| | - Suk Namgoong
- Department of Animal Sciences, Chungbuk National University, Cheong-Ju, ChungBuk, Republic of Korea
- * E-mail: (SN); (NHK)
| |
Collapse
|
47
|
Sharif B, Fadero T, Maddox AS. Anillin localization suggests distinct mechanisms of division plane specification in mouse oogenic meiosis I and II. Gene Expr Patterns 2015; 17:98-106. [PMID: 25818309 DOI: 10.1016/j.gep.2015.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 03/05/2015] [Accepted: 03/13/2015] [Indexed: 01/22/2023]
Abstract
Anillin is a conserved cytokinetic ring protein implicated in actomyosin cytoskeletal organization and cytoskeletal-membrane linkage. Here we explored anillin localization in the highly asymmetric divisions of the mouse oocyte that lead to the extrusion of two polar bodies. The purposes of polar body extrusion are to reduce the chromosome complement within the egg to haploid, and to retain the majority of the egg cytoplasm for embryonic development. Anillin's proposed roles in cytokinetic ring organization suggest that it plays important roles in achieving this asymmetric division. We report that during meiotic maturation, anillin mRNA is expressed and protein levels steadily rise. In meiosis I, anillin localizes to a cortical cap overlying metaphase I spindles, and a broad ring over anaphase spindles that are perpendicular to the cortex. Anillin is excluded from the cortex of the prospective first polar body, and highly enriched in the cytokinetic ring that severs the polar body from the oocyte. In meiosis II, anillin is enriched in a cortical stripe precisely coincident with and overlying the meiotic spindle midzone. These results suggest a model in which this cortical structure contributes to spindle re-alignment in meiosis II. Thus, localization of anillin as a conserved cytokinetic ring marker illustrates that the geometry of the cytokinetic ring is distinct between the two oogenic meiotic cytokineses in mammals.
Collapse
Affiliation(s)
- Bedra Sharif
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, Canada
| | - Tanner Fadero
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA
| | - Amy Shaub Maddox
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
48
|
Coticchio G, Dal Canto M, Mignini Renzini M, Guglielmo MC, Brambillasca F, Turchi D, Novara PV, Fadini R. Oocyte maturation: gamete-somatic cells interactions, meiotic resumption, cytoskeletal dynamics and cytoplasmic reorganization. Hum Reprod Update 2015; 21:427-54. [PMID: 25744083 DOI: 10.1093/humupd/dmv011] [Citation(s) in RCA: 346] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 02/11/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND In a growth phase occurring during most of folliculogenesis, the oocyte produces and accumulates molecules and organelles that are fundamental for the development of the preimplantation embryo. At ovulation, growth is followed by a phase of maturation that, although confined within a short temporal window, encompasses modifications of the oocyte chromosome complement and rearrangements of cytoplasmic components that are crucial for the achievement of developmental competence. Cumulus cells (CCs) are central to the process of maturation, providing the oocyte with metabolic support and regulatory cues. METHODS PubMed was used to search the MEDLINE database for peer-reviewed original articles and reviews concerning oocyte maturation in mammals. Searches were performed adopting 'oocyte' and 'maturation' as main terms, in association with other keywords expressing concepts relevant to the subject. The most relevant publications, i.e. those concerning major phenomena occurring during oocyte maturation in established experimental models and the human species, were assessed and discussed critically to offer a comprehensive description of the process of oocyte maturation. RESULTS By applying the above described search criteria, 6165 publications were identified, of which 543 were review articles. The number of publications increased steadily from 1974 (n = 7) to 2013 (n = 293). In 2014, from January to the time of submission of this manuscript, 140 original manuscripts and reviews were published. The studies selected for this review extend previous knowledge and shed new and astounding knowledge on oocyte maturation. It has long been known that resumption of meiosis and progression to the metaphase II stage is intrinsic to oocyte maturation, but novel findings have revealed that specific chromatin configurations are indicative of a propensity of the oocyte to resume the meiotic process and acquire developmental competence. Recently, genetic integrity has also been characterized as a factor with important implications for oocyte maturation and quality. Changes occurring in the cytoplasmic compartment are equally fundamental. Microtubules, actin filaments and chromatin not only interact to finalize chromosome segregation, but also crucially co-operate to establish cell asymmetry. This allows polar body extrusion to be accomplished with minimal loss of cytoplasm. The cytoskeleton also orchestrates the rearrangement of organelles in preparation for fertilization. For example, during maturation the distribution of the endoplasmic reticulum undergoes major modifications guided by microtubules and microfilaments to make the oocyte more competent in the generation of intracellular Ca(2+) oscillations that are pivotal for triggering egg activation. Cumulus cells are inherent to the process of oocyte maturation, emitting regulatory signals via direct cell-to-cell contacts and paracrine factors. In addition to nurturing the oocyte with key metabolites, CCs regulate meiotic resumption and modulate the function of the oocyte cytoskeleton. CONCLUSIONS Although the importance of oocyte maturation for the achievement of female meiosis has long been recognized, until recently much less was known of the significance of this process in relation to other fundamental developmental events. Studies on chromatin dynamics and integrity have extended our understanding of female meiosis. Concomitantly, cytoskeletal and organelle changes and the ancillary role of CCs have been better appreciated. This is expected to inspire novel concepts and advances in assisted reproduction technologies, such as the development of novel in vitro maturation systems and the identification of biomarkers of oocyte quality.
Collapse
Affiliation(s)
- Giovanni Coticchio
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| | - Mariabeatrice Dal Canto
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| | - Mario Mignini Renzini
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| | - Maria Cristina Guglielmo
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| | - Fausta Brambillasca
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| | - Diana Turchi
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| | - Paola Vittoria Novara
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| | - Rubens Fadini
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| |
Collapse
|
49
|
Ohtake J, Sakurai M, Hoshino Y, Tanemura K, Sato E. Expression of focal adhesion kinase in mouse cumulus-oocyte complexes, and effect of phosphorylation at Tyr397 on cumulus expansion. Mol Reprod Dev 2015; 82:218-31. [PMID: 25692763 DOI: 10.1002/mrd.22464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 01/16/2015] [Indexed: 01/22/2023]
Abstract
We investigated the expression of focal adhesion kinase (FAK) in mouse cumulus-oocyte complexes (COCs), as well as the role of FAK phosphorylation at Tyr397 during oocyte maturation. The effect of inhibiting FAK phosphorylation at Tyr397 during in vitro maturation (IVM) on subsequent fertilization and preimplantation embryo development was also examined. Western blotting analyses revealed that total and Tyr397-phosphorylated FAK were expressed in vivo in both cumulus cells and oocytes. Immunocytochemical studies localized this kinase throughout the cytoplasm of cumulus cells and oocytes; in particular, Tyr397-phosphorylated FAK tended to accumulate in regions where cumulus cells contact each other. Interestingly, the in vivo level of Tyr397 phosphorylation in cumulus cells was significantly lower after compared to before cumulus expansion. Addition of FAK inhibitor 14, which specifically blocks phosphorylation at Tyr397, stimulated oocyte meiotic maturation and cumulus expansion during IVM in the absence of follicle-stimulating hormone (FSH). Reverse-transcriptase PCR showed that the mRNA expression of hyaluronan synthase 2 (Has2), a marker of cumulus expansion, was significantly induced in cumulus cells. Subsequent in vitro fertilization and culture showed that more oocytes developed to the blastocyst stage when they were treated with FAK inhibitor 14 during IVM, although the blastocyst total cell number was lower than in oocytes stimulated with FSH. These results indicate that FAK is involved in the maturation of COCs; specifically, phosphorylation at Tyr397 may regulate cumulus expansion via the expression of Has2 mRNA in cumulus cells, which could affect the developmental competence of oocytes.
Collapse
Affiliation(s)
- Jun Ohtake
- Laboratory of Animal Reproduction, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | | | | | | | | |
Collapse
|
50
|
Zhang Y, Wang QC, Han J, Cao R, Cui XS, Kim NH, Rui R, Sun SC. Involvement of dynamin 2 in actin-based polar-body extrusion during porcine oocyte maturation. Mol Reprod Dev 2014; 81:725-34. [DOI: 10.1002/mrd.22341] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/08/2014] [Indexed: 11/07/2022]
Affiliation(s)
- Yu Zhang
- College of Animal Science and Technology; Nanjing Agricultural University; Nanjing People's Republic of China
- College of Veterinary Medicine; Nanjing Agricultural University; Nanjing People's Republic of China
| | - Qiao-Chu Wang
- College of Animal Science and Technology; Nanjing Agricultural University; Nanjing People's Republic of China
| | - Jun Han
- College of Animal Science and Technology; Nanjing Agricultural University; Nanjing People's Republic of China
| | - Rui Cao
- College of Animal Science and Technology; Nanjing Agricultural University; Nanjing People's Republic of China
| | - Xiang-Shun Cui
- Department of Animal Sciences; Chungbuk National University; Cheongju Chungbuk Korea
| | - Nam-Hyung Kim
- Department of Animal Sciences; Chungbuk National University; Cheongju Chungbuk Korea
| | - Rong Rui
- College of Veterinary Medicine; Nanjing Agricultural University; Nanjing People's Republic of China
| | - Shao-Chen Sun
- College of Animal Science and Technology; Nanjing Agricultural University; Nanjing People's Republic of China
| |
Collapse
|