1
|
Ni KD, Wei CG, Zhu JQ, Mu CK, Wang CL, Hou CC. Transcriptome analysis of different stages of testis development in Portunus trituberculatus. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101453. [PMID: 40010143 DOI: 10.1016/j.cbd.2025.101453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/28/2025] [Accepted: 02/19/2025] [Indexed: 02/28/2025]
Abstract
The swimming crab (Portunus trituberculatus) is an important marine economic species, however its artificial breeding yield is relatively low. Currently, the main challenge faced by the swimming crab seed industry is the reliance on wild populations for seed cultivation, which results in unstable yield and quality, affecting the healthy development of the crab farming industry to some extent. The quality of germplasm resources depends on the quality of gametes, and the quality of sperm depends on the orderly genetic regulation process of spermatogenesis. Therefore, elucidating the genetic regulatory mechanisms of spermatogenesis is of great significance for improving the germplasm resources of P. trituberculatus. To gain a deeper understanding of this process, we conducted a comparative transcriptome study on the testis of the swimming crab at different developmental stages. This study aims to identify key genes that regulate testicular development. We performed paraffin section identification on the testicular tissue of male crabs and conducted transcriptome analysis on the testicular tissue at five different developmental stages and somatic cells. Through differential expression analysis, we screened a total of 31,788 differentially expressed genes (DEGs) from stages I to VI. Through Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, we found that these DEGs were significantly enriched in 15 pathways, including important functional pathways such as the adrenergic signaling pathway, HIF-1 signaling pathway, and TGF-β signaling pathway. GO analysis results showed that calcium ion homeostasis and cell skeleton-related activities were significantly enriched in stage II. Further protein-protein interaction network analysis revealed 68 hub genes, including 13 eukaryotic initiation factors, 6 Ras superfamily members, and 6 genes related to cell division. In addition, genes such as Actin, Myosin, and Nup50 consistently showed high expression at all developmental stages, while genes related to calcium ion homeostasis, such as CaM, significantly increased in expression during stage II. Hsp90 and apoptosis-related genes had higher expression in stage IV, while Smad4 had higher expression in stage V. These results suggest that stage II of the swimming crab sperm development may be a critical period for spermatogenesis, and stage IV may be an important period for regulating sperm quality and quantity. This study not only provides a foundation for further research on the molecular mechanisms of testicular development and spermatogenesis in the swimming crab but also offers theoretical support for improving breeding yield, which has significant practical application value.
Collapse
Affiliation(s)
- Kai-Di Ni
- Key Laboratory of Aquacultural Biotechnology, and Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Chao-Guang Wei
- Key Laboratory of Aquacultural Biotechnology, and Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Jun-Quan Zhu
- Key Laboratory of Aquacultural Biotechnology, and Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Chang-Kao Mu
- Key Laboratory of Aquacultural Biotechnology, and Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Chun-Lin Wang
- Key Laboratory of Aquacultural Biotechnology, and Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Cong-Cong Hou
- Key Laboratory of Aquacultural Biotechnology, and Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
2
|
Thomalla JM, Wolfner MF. No transcription, no problem: Protein phosphorylation changes and the transition from oocyte to embryo. Curr Top Dev Biol 2025; 162:165-205. [PMID: 40180509 DOI: 10.1016/bs.ctdb.2025.01.001] [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] [Indexed: 04/05/2025]
Abstract
Although mature oocytes are arrested in a differentiated state, they are provisioned with maternally-derived macromolecules that will start embryogenesis. The transition to embryogenesis, called 'egg activation', occurs without new transcription, even though it includes major cell changes like completing stalled meiosis, translating stored mRNAs, cytoskeletal remodeling, and changes to nuclear architecture. In most animals, egg activation is triggered by a rise in free calcium in the egg's cytoplasm, but we are only now beginning to understand how this induces the egg to transition to totipotency and proliferation. Here, we discuss the model that calcium-dependent protein kinases and phosphatases modify the phosphorylation landscape of the maternal proteome to activate the egg. We review recent phosphoproteomic mass spectrometry analyses that revealed broad phospho-regulation during egg activation, both in number of phospho-events and classes of regulated proteins. Our interspecies comparisons of these proteins pinpoints orthologs and protein families that are phospho-regulated in activating eggs, many of which function in hallmark events of egg activation, and others whose regulation and activity warrant further study. Finally, we discuss key phospho-regulating enzymes that may act apically or as intermediates in the phosphorylation cascades during egg activation. Knowing the regulators, targets, and effects of phospho-regulation that cause an egg to initiate embryogenesis is crucial at both fundamental and applied levels for understanding female fertility, embryo development, and cell-state transitions.
Collapse
Affiliation(s)
- Jonathon M Thomalla
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, United States; Department of Biomedical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, NY, United States
| | - Mariana F Wolfner
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, United States.
| |
Collapse
|
3
|
Swann K. The characteristics of the calcium signals that activate mammalian eggs at fertilization. Curr Top Dev Biol 2024; 162:317-350. [PMID: 40180513 DOI: 10.1016/bs.ctdb.2024.12.002] [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] [Indexed: 04/05/2025]
Abstract
Gamete membrane fusion in mammals brings the paternal genome into the cytoplasm of the egg. It also enables signals to pass from the sperm into the egg to trigger the completion of meiosis and the start of embryo development. The essential signal to activate development in all mammals studied, consists of a series of transient increases in the cytosolic Ca2+ concentration driven by cycles of InsP3 production. This review focusses on the characteristics of these sperm-induced Ca2+ signals. I consider how some specific features of sperm-derived phospholipase C-zeta (PLCζ), along with the known properties of the type 1 InsP3 receptor, provide a basis for understanding the mechanisms of the dynamic changes in Ca2+ observed in fertilizing eggs. I describe how the PLCζ targeting of cytoplasmic vesicles in the egg cytoplasm, that contain PI(4,5)P2, is necessary to explain the rapid waves associated with the rising phase of each Ca2+ transient. I also discuss the importance of the repetitive Ca2+ rises for egg activation and the way mitochondrial ATP production may modulate Ca2+ release in eggs. Finally, I consider the role that a sperm-induced ATP increase may play in the egg activation process.
Collapse
Affiliation(s)
- Karl Swann
- School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, Wales, United Kingdom.
| |
Collapse
|
4
|
Machaty Z. The signal that stimulates mammalian embryo development. Front Cell Dev Biol 2024; 12:1474009. [PMID: 39355121 PMCID: PMC11442298 DOI: 10.3389/fcell.2024.1474009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/05/2024] [Indexed: 10/03/2024] Open
Abstract
Embryo development is stimulated by calcium (Ca2+) signals that are generated in the egg cytoplasm by the fertilizing sperm. Eggs are formed via oogenesis. They go through a cell division known as meiosis, during which their diploid chromosome number is halved and new genetic combinations are created by crossing over. During formation the eggs also acquire cellular components that are necessary to produce the Ca2+ signal and also, to support development of the newly formed embryo. Ionized calcium is a universal second messenger used by cells in a plethora of biological processes and the eggs develop a "toolkit", a set of molecules needed for signaling. Meiosis stops twice and these arrests are controlled by a complex interaction of regulatory proteins. The first meiotic arrest lasts until after puberty, when a luteinizing hormone surge stimulates meiotic resumption. The cell cycle proceeds to stop again in the middle of the second meiotic division, right before ovulation. The union of the female and male gametes takes place in the oviduct. Following gamete fusion, the sperm triggers the release of Ca2+ from the egg's intracellular stores which in mammals is followed by repetitive Ca2+ spikes known as Ca2+ oscillations in the cytosol that last for several hours. Downstream sensor proteins help decoding the signal and stimulate other molecules whose actions are required for proper development including those that help to prevent the fusion of additional sperm cells to the egg and those that assist in the release from the second meiotic arrest, completion of meiosis and entering the first mitotic cell division. Here I review the major steps of egg formation, discuss the signaling toolkit that is essential to generate the Ca2+ signal and describe the steps of the signal transduction mechanism that activates the egg's developmental program and turns it into an embryo.
Collapse
Affiliation(s)
- Zoltan Machaty
- Department of Animal Sciences Purdue University West Lafayette, West Lafayette, IN, United States
| |
Collapse
|
5
|
Chen C, Huang Z, Dong S, Ding M, Li J, Wang M, Zeng X, Zhang X, Sun X. Calcium signaling in oocyte quality and functionality and its application. Front Endocrinol (Lausanne) 2024; 15:1411000. [PMID: 39220364 PMCID: PMC11361953 DOI: 10.3389/fendo.2024.1411000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Calcium (Ca2+) is a second messenger for many signal pathways, and changes in intracellular Ca2+ concentration ([Ca2+]i) are an important signaling mechanism in the oocyte maturation, activation, fertilization, function regulation of granulosa and cumulus cells and offspring development. Ca2+ oscillations occur during oocyte maturation and fertilization, which are maintained by Ca2+ stores and extracellular Ca2+ ([Ca2+]e). Abnormalities in Ca2+ signaling can affect the release of the first polar body, the first meiotic division, and chromosome and spindle morphology. Well-studied aspects of Ca2+ signaling in the oocyte are oocyte activation and fertilization. Oocyte activation, driven by sperm-specific phospholipase PLCζ, is initiated by concerted intracellular patterns of Ca2+ release, termed Ca2+ oscillations. Ca2+ oscillations persist for a long time during fertilization and are coordinately engaged by a variety of Ca2+ channels, pumps, regulatory proteins and their partners. Calcium signaling also regulates granulosa and cumulus cells' function, which further affects oocyte maturation and fertilization outcome. Clinically, there are several physical and chemical options for treating fertilization failure through oocyte activation. Additionally, various exogenous compounds or drugs can cause ovarian dysfunction and female infertility by inducing abnormal Ca2+ signaling or Ca2+ dyshomeostasis in oocytes and granulosa cells. Therefore, the reproductive health risks caused by adverse stresses should arouse our attention. This review will systematically summarize the latest research progress on the aforementioned aspects and propose further research directions on calcium signaling in female reproduction.
Collapse
Affiliation(s)
- Chen Chen
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Zefan Huang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Shijue Dong
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Mengqian Ding
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Jinran Li
- Center for Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Miaomiao Wang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Xuhui Zeng
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Xiaoning Zhang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Xiaoli Sun
- Center for Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| |
Collapse
|
6
|
Tong KY, Liu WW, Sun LW, Liu DY, Xiang YZ, Li C, Chai LW, Chen K, Huang GN, Li JY. Novel PLCZ1 mutation caused polyspermy during in vitro fertilization. Asian J Androl 2024; 26:389-395. [PMID: 38445955 PMCID: PMC11280200 DOI: 10.4103/aja202376] [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: 02/01/2023] [Accepted: 12/22/2023] [Indexed: 03/07/2024] Open
Abstract
Failure of oocyte activation, including polyspermy and defects in pronuclear (PN) formation, triggers early embryonic developmental arrest. Many studies have shown that phospholipase C zeta 1 ( PLCZ1 ) mutations cause failure of PN formation following intracytoplasmic sperm injection (ICSI); however, whether PLCZ1 mutation is associated with polyspermy during in vitro fertilization (IVF) remains unknown. Whole-exome sequencing (WES) was performed to identify candidate mutations in couples with primary infertility. Sanger sequencing was used to validate the mutations. Multiple PLCZ1 -mutated sperm were injected into human and mouse oocytes to explore whether PN formation was induced. Assisted oocyte activation (AOA) after ICSI was performed to overcome the failure of oocyte activation. We identified three PLCZ1 mutations in three patients who experienced polyspermy during IVF cycles, including a novel missense mutation c.1154C>T, p.R385Q. PN formation failure was observed during the ICSI cycle. However, injection of multiple PLCZ1- mutated sperm induced PN formation, suggesting that the Ca 2+ oscillations induced by the sperm exceeded the necessary threshold for PN formation. AOA after ICSI enabled normal fertilization, and all patients achieved successful pregnancies. These findings expand the mutational spectrum of PLCZ1 and suggest an important role for PLCZ1 in terms of blocking polyspermy. Furthermore, this study may benefit genetic diagnoses in cases of abnormal fertilization and provide potential appropriate therapeutic measures for these patients with sperm-derived polyspermy.
Collapse
Affiliation(s)
- Ke-Ya Tong
- Chongqing Key Laboratory of Human Embryo Engineering, Center for Reproductive Medicine, Women and Children’s Hospital of Chongqing Medical University, Chongqing 400012, China
- Chongqing Clinical Research Center for Reproductive Medicine, Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400012, China
| | - Wei-Wei Liu
- Chongqing Key Laboratory of Human Embryo Engineering, Center for Reproductive Medicine, Women and Children’s Hospital of Chongqing Medical University, Chongqing 400012, China
- Chongqing Clinical Research Center for Reproductive Medicine, Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400012, China
| | - Li-Wei Sun
- Chongqing Key Laboratory of Human Embryo Engineering, Center for Reproductive Medicine, Women and Children’s Hospital of Chongqing Medical University, Chongqing 400012, China
- Chongqing Clinical Research Center for Reproductive Medicine, Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400012, China
| | - Dong-Yun Liu
- Chongqing Key Laboratory of Human Embryo Engineering, Center for Reproductive Medicine, Women and Children’s Hospital of Chongqing Medical University, Chongqing 400012, China
- Chongqing Clinical Research Center for Reproductive Medicine, Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400012, China
| | - Ye-Zhou Xiang
- Chongqing Key Laboratory of Human Embryo Engineering, Center for Reproductive Medicine, Women and Children’s Hospital of Chongqing Medical University, Chongqing 400012, China
- Chongqing Clinical Research Center for Reproductive Medicine, Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400012, China
| | - Chong Li
- Chongqing Key Laboratory of Human Embryo Engineering, Center for Reproductive Medicine, Women and Children’s Hospital of Chongqing Medical University, Chongqing 400012, China
- Chongqing Clinical Research Center for Reproductive Medicine, Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400012, China
| | - Lu-Wei Chai
- Chongqing Key Laboratory of Human Embryo Engineering, Center for Reproductive Medicine, Women and Children’s Hospital of Chongqing Medical University, Chongqing 400012, China
- Chongqing Clinical Research Center for Reproductive Medicine, Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400012, China
| | - Ke Chen
- Chongqing Key Laboratory of Human Embryo Engineering, Center for Reproductive Medicine, Women and Children’s Hospital of Chongqing Medical University, Chongqing 400012, China
- Chongqing Clinical Research Center for Reproductive Medicine, Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400012, China
| | - Guo-Ning Huang
- Chongqing Key Laboratory of Human Embryo Engineering, Center for Reproductive Medicine, Women and Children’s Hospital of Chongqing Medical University, Chongqing 400012, China
- Chongqing Clinical Research Center for Reproductive Medicine, Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400012, China
| | - Jing-Yu Li
- Chongqing Key Laboratory of Human Embryo Engineering, Center for Reproductive Medicine, Women and Children’s Hospital of Chongqing Medical University, Chongqing 400012, China
- Chongqing Clinical Research Center for Reproductive Medicine, Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400012, China
| |
Collapse
|
7
|
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
|
8
|
Rojas J, Hinostroza F, Vergara S, Pinto-Borguero I, Aguilera F, Fuentes R, Carvacho I. Knockin' on Egg's Door: Maternal Control of Egg Activation That Influences Cortical Granule Exocytosis in Animal Species. Front Cell Dev Biol 2021; 9:704867. [PMID: 34540828 PMCID: PMC8446563 DOI: 10.3389/fcell.2021.704867] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/16/2021] [Indexed: 12/23/2022] Open
Abstract
Fertilization by multiple sperm leads to lethal chromosomal number abnormalities, failed embryo development, and miscarriage. In some vertebrate and invertebrate eggs, the so-called cortical reaction contributes to their activation and prevents polyspermy during fertilization. This process involves biogenesis, redistribution, and subsequent accumulation of cortical granules (CGs) at the female gamete cortex during oogenesis. CGs are oocyte- and egg-specific secretory vesicles whose content is discharged during fertilization to block polyspermy. Here, we summarize the molecular mechanisms controlling critical aspects of CG biology prior to and after the gametes interaction. This allows to block polyspermy and provide protection to the developing embryo. We also examine how CGs form and are spatially redistributed during oogenesis. During egg activation, CG exocytosis (CGE) and content release are triggered by increases in intracellular calcium and relies on the function of maternally-loaded proteins. We also discuss how mutations in these factors impact CG dynamics, providing unprecedented models to investigate the genetic program executing fertilization. We further explore the phylogenetic distribution of maternal proteins and signaling pathways contributing to CGE and egg activation. We conclude that many important biological questions and genotype–phenotype relationships during fertilization remain unresolved, and therefore, novel molecular players of CG biology need to be discovered. Future functional and image-based studies are expected to elucidate the identity of genetic candidates and components of the molecular machinery involved in the egg activation. This, will open new therapeutic avenues for treating infertility in humans.
Collapse
Affiliation(s)
- Japhet Rojas
- Laboratorio Fisiología de la Reproducción, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile.,Escuela de Ingeniería en Biotecnología, Facultad de Ciencias Agrarias y Forestales, Universidad Católica del Maule, Talca, Chile
| | - Fernando Hinostroza
- Laboratorio Fisiología de la Reproducción, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile.,Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile.,Centro de Investigación en Neuropsicología y Neurociencias Cognitivas, Facultad de Ciencias de la Salud, Universidad Católica del Maule, Talca, Chile
| | - Sebastián Vergara
- Laboratorio Fisiología de la Reproducción, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile.,Escuela de Ingeniería en Biotecnología, Facultad de Ciencias Agrarias y Forestales, Universidad Católica del Maule, Talca, Chile
| | - Ingrid Pinto-Borguero
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Felipe Aguilera
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Ricardo Fuentes
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Ingrid Carvacho
- Laboratorio Fisiología de la Reproducción, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile
| |
Collapse
|
9
|
Dehapiot B, Clément R, Bourdais A, Carrière V, Huet S, Halet G. RhoA- and Cdc42-induced antagonistic forces underlie symmetry breaking and spindle rotation in mouse oocytes. PLoS Biol 2021; 19:e3001376. [PMID: 34491981 PMCID: PMC8448345 DOI: 10.1371/journal.pbio.3001376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 09/17/2021] [Accepted: 07/30/2021] [Indexed: 11/25/2022] Open
Abstract
Mammalian oocyte meiotic divisions are highly asymmetric and produce a large haploid gamete and 2 small polar bodies. This relies on the ability of the cell to break symmetry and position its spindle close to the cortex before anaphase occurs. In metaphase II–arrested mouse oocytes, the spindle is actively maintained close and parallel to the cortex, until fertilization triggers sister chromatid segregation and the rotation of the spindle. The latter must indeed reorient perpendicular to the cortex to enable cytokinesis ring closure at the base of the polar body. However, the mechanisms underlying symmetry breaking and spindle rotation have remained elusive. In this study, we show that spindle rotation results from 2 antagonistic forces. First, an inward contraction of the cytokinesis furrow dependent on RhoA signaling, and second, an outward attraction exerted on both sets of chromatids by a Ran/Cdc42-dependent polarization of the actomyosin cortex. By combining live segmentation and tracking with numerical modeling, we demonstrate that this configuration becomes unstable as the ingression progresses. This leads to spontaneous symmetry breaking, which implies that neither the rotation direction nor the set of chromatids that eventually gets discarded are biologically predetermined. Mammalian oocyte meiotic divisions are highly asymmetric and produce a large haploid gamete and two small polar bodies, but the mechanisms underlying the required symmetry breaking and spindle rotation have remained elusive. This study shows that spindle rotation in activated mouse oocytes relies on spontaneous symmetry breaking resulting from an unstable configuration generated by cleavage furrow ingression and cortical chromosome attraction.
Collapse
Affiliation(s)
- Benoit Dehapiot
- Aix Marseille Université, CNRS, IBDM-UMR7288, Turing Center for Living Systems, Marseille, France
- Univ Rennes, CNRS, IGDR—UMR 6290, Rennes, France
- * E-mail: (BD); (GH)
| | - Raphaël Clément
- Aix Marseille Université, CNRS, IBDM-UMR7288, Turing Center for Living Systems, Marseille, France
| | | | | | | | - Guillaume Halet
- Univ Rennes, CNRS, IGDR—UMR 6290, Rennes, France
- * E-mail: (BD); (GH)
| |
Collapse
|
10
|
Lee HC, Edmonds ME, Duncan FE, O'Halloran TV, Woodruff TK. Zinc exocytosis is sensitive to myosin light chain kinase inhibition in mouse and human eggs. Mol Hum Reprod 2021; 26:228-239. [PMID: 32119740 DOI: 10.1093/molehr/gaaa017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 02/07/2020] [Indexed: 12/20/2022] Open
Abstract
Zinc dynamics are essential for oocyte meiotic maturation, egg activation, and preimplantation embryo development. During fertilisation and egg activation, the egg releases billions of zinc atoms (Zn2+) in an exocytotic event termed the 'zinc spark'. We hypothesised that this zinc transport and exocytosis is dependent upon the intracellular trafficking of cortical granules (CG) which requires myosin-actin-dependent motors. Treatment of mature mouse and human eggs with ML-7, a myosin light chain kinase inhibitor (MLCK), resulted in an 80% reduction in zinc spark intensity compared to untreated controls when activated with ionomycin. Moreover, CG migration towards the plasma membrane was significantly decreased in ML-7-treated eggs compared with controls when activated parthenogenetically with ionomycin. In sperm-induced fertilisation via intracytoplasmic sperm injection (ICSI), ML-7-treated mouse eggs exhibited decreased labile zinc intensity and cortical CG staining. Collectively, these data demonstrate that ML-7 treatment impairs zinc release from both murine and human eggs after activation, demonstrating that zinc exocytosis requires myosin light chain kinase activity. Further, these results provide additional support that zinc is likely stored and released from CGs. These data underscore the importance of intracellular zinc trafficking as a crucial component of egg maturation necessary for egg activation and early embryo development.
Collapse
Affiliation(s)
- Hoi Chang Lee
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Maxwell E Edmonds
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Thomas V O'Halloran
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA.,Department of Chemistry, Northwestern University, Evanston, IL, USA
| | - Teresa K Woodruff
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| |
Collapse
|
11
|
Converse A, Thomas P. The zinc transporter ZIP9 (Slc39a9) regulates zinc dynamics essential to egg activation in zebrafish. Sci Rep 2020; 10:15673. [PMID: 32973303 PMCID: PMC7518430 DOI: 10.1038/s41598-020-72515-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 09/02/2020] [Indexed: 01/22/2023] Open
Abstract
The zinc transporter ZIP9 (SLC39A9) was recently characterized as a membrane androgen receptor in various teleost and mammalian cell models. ZIP9 shows the highest expression in ovaries of teleosts, a tissue in which both androgen signaling and zinc dynamics have significant roles. To examine the role of ZIP9 in ovarian physiology, we generated a ZIP9-mutant zebrafish strain using a CRISPR/Cas9 system. zip9-/- females showed significant reductions in fecundity, embryo viability, and growth of their offspring compared to wildtype (WT) fish. Furthermore, a high proportion of zip9-/- eggs failed to undergo normal chorion elevation during activation. In WT eggs, zinc was detected in cortically-localized vesicles which underwent exocytosis upon activation. zip9-/- eggs showed abnormal cortical vesicle development and had a significantly depressed activation-induced zinc release compared to WT eggs. Moreover, pharmacologically sustained elevation of zinc in WT eggs prior to activation resulted in abnormal chorion elevation similar to that observed in zip9-/- eggs. These results indicate that ZIP9 is essential for proper zinc modulation during zebrafish egg activation and presents the first evidence of zinc modulation during egg activation in a non-mammalian species.
Collapse
Affiliation(s)
- Aubrey Converse
- Marine Science Institute, The University of Texas at Austin, 750 Channelview Dr., Port Aransas, TX, 78373, USA.
| | - Peter Thomas
- Marine Science Institute, The University of Texas at Austin, 750 Channelview Dr., Port Aransas, TX, 78373, USA
| |
Collapse
|
12
|
Calcium Oscillatory Patterns and Oocyte Activation During Fertilization: a Possible Mechanism for Total Fertilization Failure (TFF) in Human In Vitro Fertilization? Reprod Sci 2020; 28:639-648. [PMID: 32813196 DOI: 10.1007/s43032-020-00293-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/06/2020] [Indexed: 10/23/2022]
Abstract
This paper reviews the effects of calcium oscillatory patterns in oocytes and early embryo development. Total fertilization failure (TFF) is the failure of fertilization in all oocytes in a human IVF cycle, even after treatment with intracytoplasmic sperm injection (ICSI). It is not well understood and currently attributed to oocyte activation deficiency. Calcium signaling is important in oocyte activation events. Calcium oscillations, in particular, have been reported in animal and human oocytes after fertilization. Abnormal calcium oscillations after fertilization may be the principal mechanism for TFF. While studies also establish strong associations between abnormal calcium oscillatory patterns and suboptimal developmental outcomes, critical basic parameters and their mechanism of action have yet to be identified. Empirical use of artificial oocyte activation (AOA) methods has shown initial success in helping patients overcome TFF. The AOA methods attempt to raise calcium levels after fertilization, but the efficacy and safety of these AOA methods are still in early stages of addressing TFF. Additional information about calcium oscillatory patterns and the effects of AOA in human ART may allow the prevention of TFF or allow treatment of TFF patients effectively and safely.
Collapse
|
13
|
Wang H, Li Y, Yang J, Duan X, Kalab P, Sun SX, Li R. Symmetry breaking in hydrodynamic forces drives meiotic spindle rotation in mammalian oocytes. SCIENCE ADVANCES 2020; 6:eaaz5004. [PMID: 32284983 PMCID: PMC7124937 DOI: 10.1126/sciadv.aaz5004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/14/2020] [Indexed: 06/11/2023]
Abstract
Patterned cell divisions require a precisely oriented spindle that segregates chromosomes and determines the cytokinetic plane. In this study, we investigated how the meiotic spindle orients through an obligatory rotation during meiotic division in mouse oocytes. We show that spindle rotation occurs at the completion of chromosome segregation, whereby the separated chromosome clusters each define a cortical actomyosin domain that produces cytoplasmic streaming, resulting in hydrodynamic forces on the spindle. These forces are initially balanced but become unbalanced to drive spindle rotation. This force imbalance is associated with spontaneous symmetry breaking in the distribution of the Arp2/3 complex and myosin-II on the cortex, brought about by feedback loops comprising Ran guanosine triphosphatase signaling, Arp2/3 complex activity, and myosin-II contractility. The torque produced by the unbalanced hydrodynamic forces, coupled with a pivot point at the spindle midzone cortical contract, constitutes a unique mechanical system for meiotic spindle rotation.
Collapse
Affiliation(s)
- HaiYang Wang
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Yizeng Li
- Department of Mechanical Engineering, Kennesaw State University, Marietta, GA 30060, USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jing Yang
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- School of Physical Science and Technology, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Xing Duan
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Petr Kalab
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sean X. Sun
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Rong Li
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
14
|
Dyballa S, Miñana R, Rubio-Brotons M, Cornet C, Pederzani T, Escaramis G, Garcia-Serna R, Mestres J, Terriente J. Comparison of Zebrafish Larvae and hiPSC Cardiomyocytes for Predicting Drug-Induced Cardiotoxicity in Humans. Toxicol Sci 2019; 171:283-295. [PMID: 31359052 PMCID: PMC6760275 DOI: 10.1093/toxsci/kfz165] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular drug toxicity is responsible for 17% of drug withdrawals in clinical phases, half of post-marketed drug withdrawals and remains an important adverse effect of several marketed drugs. Early assessment of drug-induced cardiovascular toxicity is mandatory and typically done in cellular systems and mammals. Current in vitro screening methods allow high-throughput but are biologically reductionist. The use of mammal models, which allow a better translatability for predicting clinical outputs, is low-throughput, highly expensive, and ethically controversial. Given the analogies between the human and the zebrafish cardiovascular systems, we propose the use of zebrafish larvae during early drug discovery phases as a balanced model between biological translatability and screening throughput for addressing potential liabilities. To this end, we have developed a high-throughput screening platform that enables fully automatized in vivo image acquisition and analysis to extract a plethora of relevant cardiovascular parameters: heart rate, arrhythmia, AV blockage, ejection fraction, and blood flow, among others. We have used this platform to address the predictive power of zebrafish larvae for detecting potential cardiovascular liabilities in humans. We tested a chemical library of 92 compounds with known clinical cardiotoxicity profiles. The cross-comparison with clinical data and data acquired from human induced pluripotent stem cell cardiomyocytes calcium imaging showed that zebrafish larvae allow a more reliable prediction of cardiotoxicity than cellular systems. Interestingly, our analysis with zebrafish yields similar predictive performance as previous validation meta-studies performed with dogs, the standard regulatory preclinical model for predicting cardiotoxic liabilities prior to clinical phases.
Collapse
Affiliation(s)
- Sylvia Dyballa
- ZeClinics SL, IGTP (German Trias and Pujol Institute), Badalona 08916, Spain
| | - Rafael Miñana
- ZeClinics SL, IGTP (German Trias and Pujol Institute), Badalona 08916, Spain
| | - Maria Rubio-Brotons
- ZeClinics SL, IGTP (German Trias and Pujol Institute), Badalona 08916, Spain
| | - Carles Cornet
- ZeClinics SL, IGTP (German Trias and Pujol Institute), Badalona 08916, Spain
| | - Tiziana Pederzani
- ZeClinics SL, IGTP (German Trias and Pujol Institute), Badalona 08916, Spain
| | - Georgia Escaramis
- CIBER Epidemiology and Public Health
- Department of Biomedicine, Faculty of Life Science and Health, University of Barcelona 08036, Barcelona, Spain
- Research Group on Statistics, Econometrics and Health (GRECS), UdG, Girona 17071, Spain
| | | | - Jordi Mestres
- Chemotargets SL, Parc Científic de Barcelona, Barcelona 08028, Spain
- Systems Pharmacology, Research Program on Biomedical Informatics (GRIB), IMIM Hospital del Mar Medical Research Institute, Barcelona 08002, Spain
- University Pompeu Fabra, PRBB (Barcelona Biomedical Research Park), Barcelona 08002, Spain
| | - Javier Terriente
- ZeClinics SL, IGTP (German Trias and Pujol Institute), Badalona 08916, Spain
| |
Collapse
|
15
|
Duan X, Sun SC. Actin cytoskeleton dynamics in mammalian oocyte meiosis†. Biol Reprod 2018; 100:15-24. [DOI: 10.1093/biolre/ioy163] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/11/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Xing Duan
- 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
|
16
|
Martin JH, Bromfield EG, Aitken RJ, Nixon B. Biochemical alterations in the oocyte in support of early embryonic development. Cell Mol Life Sci 2017; 74:469-485. [PMID: 27604868 PMCID: PMC11107538 DOI: 10.1007/s00018-016-2356-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/28/2016] [Accepted: 09/01/2016] [Indexed: 01/01/2023]
Abstract
Notwithstanding the enormous reproductive potential encapsulated within a mature mammalian oocyte, these cells present only a limited window for fertilization before defaulting to an apoptotic cascade known as post-ovulatory oocyte aging. The only cell with the capacity to rescue this potential is the fertilizing spermatozoon. Indeed, the union of these cells sets in train a remarkable series of events that endows the oocyte with the capacity to divide and differentiate into the trillions of cells that comprise a new individual. Traditional paradigms hold that, beyond the initial stimulation of fluctuating calcium (Ca2+) required for oocyte activation, the fertilizing spermatozoon plays limited additional roles in the early embryo. While this model has now been drawn into question in view of the recent discovery that spermatozoa deliver developmentally important classes of small noncoding RNAs and other epigenetic modulators to oocytes during fertilization, it is nevertheless apparent that the primary responsibility for oocyte activation rests with a modest store of maternally derived proteins and mRNA accumulated during oogenesis. It is, therefore, not surprising that widespread post-translational modifications, in particular phosphorylation, hold a central role in endowing these proteins with sufficient functional diversity to initiate embryonic development. Indeed, proteins targeted for such modifications have been linked to oocyte activation, recruitment of maternal mRNAs, DNA repair and resumption of the cell cycle. This review, therefore, seeks to explore the intimate relationship between Ca2+ release and the suite of molecular modifications that sweep through the oocyte to ensure the successful union of the parental germlines and ensure embryogenic fidelity.
Collapse
Affiliation(s)
- Jacinta H Martin
- Discipline of Biological Sciences and Priority Research Center for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.
| | - Elizabeth G Bromfield
- Discipline of Biological Sciences and Priority Research Center for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - R John Aitken
- Discipline of Biological Sciences and Priority Research Center for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Brett Nixon
- Discipline of Biological Sciences and Priority Research Center for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| |
Collapse
|
17
|
Bury L, Coelho PA, Glover DM. From Meiosis to Mitosis: The Astonishing Flexibility of Cell Division Mechanisms in Early Mammalian Development. Curr Top Dev Biol 2016; 120:125-71. [PMID: 27475851 DOI: 10.1016/bs.ctdb.2016.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The execution of female meiosis and the establishment of the zygote is arguably the most critical stage of mammalian development. The egg can be arrested in the prophase of meiosis I for decades, and when it is activated, the spindle is assembled de novo. This spindle must function with the highest of fidelity and yet its assembly is unusually achieved in the absence of conventional centrosomes and with minimal influence of chromatin. Moreover, its dramatic asymmetric positioning is achieved through remarkable properties of the actin cytoskeleton to ensure elimination of the polar bodies. The second meiotic arrest marks a uniquely prolonged metaphase eventually interrupted by egg activation at fertilization to complete meiosis and mark a period of preparation of the male and female pronuclear genomes not only for their entry into the mitotic cleavage divisions but also for the imminent prospect of their zygotic expression.
Collapse
Affiliation(s)
- L Bury
- University of Cambridge, Cambridge, United Kingdom.
| | - P A Coelho
- University of Cambridge, Cambridge, United Kingdom
| | - D M Glover
- University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
18
|
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
|
19
|
Sanders JR, Swann K. Molecular triggers of egg activation at fertilization in mammals. Reproduction 2016; 152:R41-50. [PMID: 27165049 DOI: 10.1530/rep-16-0123] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/09/2016] [Indexed: 01/15/2023]
Abstract
In mammals, the sperm activates the development of the egg by triggering a series of oscillations in the cytosolic-free Ca(2+) concentration (Ca(2+) i). The sperm triggers these cytosolic Ca(2+i) oscillations after sperm-egg membrane fusion, as well as after intracytoplasmic sperm injection (ICSI). These Ca(2+) i oscillations are triggered by a protein located inside the sperm. The identity of the sperm protein has been debated over many years, but all the repeatable data now suggest that it is phospholipase Czeta (PLCζ). The main downstream target of Ca(2+) i oscillations is calmodulin-dependent protein kinase II (CAMKII (CAMK2A)), which phosphorylates EMI2 and WEE1B to inactivate the M-phase promoting factor protein kinase activity (MPF) and this ultimately triggers meiotic resumption. A later decline in the activity of mitogen-activated protein kinase (MAPK) then leads to the completion of activation which is marked by the formation of pronuclei and entry into interphase of the first cell cycle. The early cytosolic Ca(2+) increases also trigger exocytosis via a mechanism that does not involve CAMKII. We discuss some recent developments in our understanding of these triggers for egg activation within the framework of cytosolic Ca(2+) signaling.
Collapse
Affiliation(s)
| | - Karl Swann
- School of BiosciencesCardiff University, Cardiff, UK
| |
Collapse
|
20
|
Li YR, Yang WX. Myosin superfamily: The multi-functional and irreplaceable factors in spermatogenesis and testicular tumors. Gene 2015; 576:195-207. [PMID: 26478466 DOI: 10.1016/j.gene.2015.10.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 09/21/2015] [Accepted: 10/12/2015] [Indexed: 10/22/2022]
Abstract
Spermatogenesis is a fundamental process in sexual development and reproduction, in which the diploid spermatogonia transform into haploid mature spermatozoa. This process is under the regulation of multiple factors and pathway. Myosin has been implicated in various aspects during spermatogenesis. Myosins constitute a diverse superfamily of actin-based molecular motors that translocate along microfilament in an ATP-dependent manner, and six kinds of myosins have been proved that function during spermatogenesis. In mitosis and meiosis, myosins play an important role in spindle assembly and positioning, karyokinesis and cytokinesis. During spermiogenesis, myosins participate in acrosomal formation, nuclear morphogenesis, mitochondrial translocation and spermatid individualization. In this review, we summarize current understanding of the functions of myosin in spermatogenesis and some reproductive system diseases such as testicular tumors and prostate cancer, and discuss the roles of possible upstream molecules which regulate myosin in these processes.
Collapse
Affiliation(s)
- Yan-Ruide Li
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
21
|
McGinnis LA, Lee HJ, Robinson DN, Evans JP. MAPK3/1 (ERK1/2) and Myosin Light Chain Kinase in Mammalian Eggs Affect Myosin-II Function and Regulate the Metaphase II State in a Calcium- and Zinc-Dependent Manner. Biol Reprod 2015; 92:146. [PMID: 25904014 DOI: 10.1095/biolreprod.114.127027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 04/16/2015] [Indexed: 12/25/2022] Open
Abstract
Vertebrate eggs are arrested at metaphase of meiosis II, a state classically known as cytostatic factor arrest. Maintenance of this arrest until the time of fertilization and then fertilization-induced exit from metaphase II are crucial for reproductive success. Another key aspect of this meiotic arrest and exit is regulation of the metaphase II spindle, which must be appropriately localized adjacent to the egg cortex during metaphase II and then progress into successful asymmetric cytokinesis to produce the second polar body. This study examined the mitogen-activated protein kinases MAPK3 and MAPK1 (also known as ERK1/2) as regulators of these two related aspects of mammalian egg biology, specifically testing whether this MAPK pathway affected myosin-II function and whether myosin-II perturbation would produce some of the same effects as MAPK pathway perturbation. Inhibition of the MEK1/2-MAPK pathway with U0126 leads to reduced levels of phosphorylated myosin-regulatory light chain (pMRLC) and causes a reduction in cortical tension, effects that are mimicked by treatment with the myosin light chain kinase (MLCK) inhibitor ML-7. These data indicate that one mechanism by which the MAPK pathway acts in eggs is by affecting myosin-II function. We further show that MAPK or MLCK inhibition induces loss of normal cortical spindle localization or parthenogenetic egg activation. This parthenogenesis is dependent on cytosolic and extracellular calcium and can be rescued by hyperloading eggs with zinc, suggesting that these effects of inhibition of MLCK or the MAPK pathway are linked with dysregulation of ion homeostasis.
Collapse
Affiliation(s)
- Lauren A McGinnis
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Hyo J Lee
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Douglas N Robinson
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Janice P Evans
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
22
|
Liang QX, Zhang QH, Qi ST, Wang ZW, Hu MW, Ma XS, Zhu MS, Schatten H, Wang ZB, Sun QY. Deletion of Mylk1 in Oocytes Causes Delayed Morula-to-Blastocyst Transition and Reduced Fertility Without Affecting Folliculogenesis and Oocyte Maturation in Mice1. Biol Reprod 2015; 92:97. [DOI: 10.1095/biolreprod.114.122127] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 03/06/2015] [Indexed: 01/10/2023] Open
|
23
|
Abstract
Accurate positioning of spindles is essential for asymmetric mitotic and meiotic cell divisions that are crucial for animal development and oocyte maturation, respectively. The predominant model for spindle positioning, termed "cortical pulling," involves attachment of the microtubule-based motor cytoplasmic dynein to the cortex, where it exerts a pulling force on microtubules that extend from the spindle poles to the cell cortex, thereby displacing the spindle. Recent studies have addressed important details of the cortical pulling mechanism and have revealed alternative mechanisms that may be used when microtubules do not extend from the spindle to the cortex.
Collapse
Affiliation(s)
- Francis J McNally
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
24
|
Maddox AS, Azoury J, Dumont J. Polar body cytokinesis. Cytoskeleton (Hoboken) 2012; 69:855-68. [PMID: 22927361 DOI: 10.1002/cm.21064] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Accepted: 08/20/2012] [Indexed: 02/04/2023]
Abstract
Polar body cytokinesis is the physical separation of a small polar body from a larger oocyte or ovum. This maternal meiotic division shares many similarities with mitotic and spermatogenic cytokinesis, but there are several distinctions, which will be discussed in this review. We synthesize results from many different model species, including those popular for their genetics and several that are more obscure in modern cell biology. The site of polar body division is determined before anaphase, by the eccentric, cortically associated meiotic spindle. Depending on the species, either the actin or microtubule cytoskeleton is required for spindle anchoring. Chromatin is necessary and sufficient to elicit differentiation of the associated cortex, via Ran-based signaling. The midzone of the anaphase spindle serves as a hub for regulatory complexes that elicit Rho activation, and ultimately actomyosin contractile ring assembly and contraction. Polar body cytokinesis uniquely requires another Rho family GTPase, Cdc42, for dynamic reorganization of the polar cortex. This is perhaps due to the considerable asymmetry of this division, wherein the polar body and the oocyte/ovum have distinct fates and very different sizes. Thus, maternal meiotic cytokinesis appears to occur via simultaneous polar relaxation and equatorial contraction, since the polar body is extruded from the spherical oocyte through the nascent contractile ring. As such, polar body cytokinesis is an interesting and important variation on the theme of cell division.
Collapse
Affiliation(s)
- Amy Shaub Maddox
- Institut de recherche en immunology et en cancerologie (IRIC), Université de Montréal, Montréal, Quebec, Canada.
| | | | | |
Collapse
|
25
|
Miao YL, Williams CJ. Calcium signaling in mammalian egg activation and embryo development: the influence of subcellular localization. Mol Reprod Dev 2012; 79:742-56. [PMID: 22888043 DOI: 10.1002/mrd.22078] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Accepted: 07/27/2012] [Indexed: 11/07/2022]
Abstract
Calcium (Ca(2+) ) signals drive the fundamental events surrounding fertilization and the activation of development in all species examined to date. Initial studies of Ca(2+) signaling at fertilization in marine animals were tightly linked to new discoveries of bioluminescent proteins and their use as fluorescent Ca(2+) sensors. Since that time, there has been rapid progress in our understanding of the key functions for Ca(2+) in many cell types and of the impact of cellular localization on Ca(2+) signaling pathways. In this review, which focuses on mammalian egg activation, we consider how Ca(2+) is regulated and stored at different stages of oocyte development and examine the functions of molecules that serve as both regulators of Ca(2+) release and effectors of Ca(2+) signals. We then summarize studies exploring how Ca(2+) directs downstream effectors mediating both egg activation and later signaling events required for successful preimplantation embryo development. Throughout this review, we focus attention on how localization of Ca(2+) signals influences downstream signaling events, and attempt to highlight gaps in our knowledge that are ripe for future research.
Collapse
Affiliation(s)
- Yi-Liang Miao
- Reproductive Medicine Group, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | |
Collapse
|
26
|
Kimura K, Kimura A. Rab6 is required for the exocytosis of cortical granules and the recruitment of separase to the granules during the oocyte-to-embryo transition in Caenorhabditis elegans. J Cell Sci 2012; 125:5897-905. [PMID: 22992455 DOI: 10.1242/jcs.116400] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Remodeling of the embryo surface after fertilization is mediated by the exocytosis of cortical granules derived from the Golgi complex. This process is essential for oocyte-to-embryo transition in many species. However, how the fertilization signal reaches the cortical granules for their timely exocytosis is largely unknown. In Caenorhabditis elegans, the recruitment of separase, a downstream effector of the fertilization signal, to the cortical granules is essential for exocytosis because separase is required for membrane fusion. However, the molecule that recruits separase to the cortical granules remains unidentified. In this study, we found that Rab6, a Golgi-associated GTPase, is essential to recruit separase to the cortical granules in C. elegans embryos. Knockdown of the rab-6.1 gene, a Rab6 homolog in C. elegans, resulted in failure of the membrane fusion step of cortical granule exocytosis. Using a transgenic strain that expresses GFP-fused RAB-6.1, we found that RAB-6.1 temporarily co-localized with separase on the cortical granules for a few minutes and then was dispersed in the cytoplasm concomitantly with membrane fusion. We found that RAB-6.1, as well as cyclin-dependent kinase (CDK)-1 and anaphase promoting complex/cyclosome (APC/C), was required to recruit separase to the cortical granules. RAB-6.1 was not required for the chromosome segregation process, unlike CDK-1, APC/C and SEP-1. The results indicate that RAB-6.1 is required specifically for the membrane fusion step of exocytosis and for the recruitment of separase to the granules. Thus, RAB-6.1 is an important molecule for the timely exocytosis of the cortical granules during oocyte-to-embryo transition.
Collapse
Affiliation(s)
- Kenji Kimura
- Cell Architecture Laboratory, Structural Biology Center, National Institute of Genetics, Mishima 411-8540, Japan
| | | |
Collapse
|
27
|
Swift LM, Asfour H, Posnack NG, Arutunyan A, Kay MW, Sarvazyan N. Properties of blebbistatin for cardiac optical mapping and other imaging applications. Pflugers Arch 2012; 464:503-12. [PMID: 22990759 DOI: 10.1007/s00424-012-1147-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 08/20/2012] [Indexed: 11/27/2022]
Abstract
Blebbistatin is a recently discovered myosin II inhibitor. It is rapidly becoming a compound of choice to reduce motion artifacts during cardiac optical mapping, as well as to study cell motility and cell invasion. Although blebbistatin has a number of advantages over other electromechanical uncouplers, many of its properties have yet to be addressed. Here we describe several methodological issues associated with the use of blebbistatin, including its spectral properties, reversibility, and its effect on tissue metabolic state. We show that if precautions are not taken, perfusion with blebbistatin may result in blebbistatin precipitate that accumulates in the vasculature. Although such precipitate is fluorescent, it is not detectable within wavelength bands that are typically used for transmembrane voltage fluorescence imaging (i.e., emission wavelengths >600 nm). Therefore, blockage of the microcirculation by blebbistatin may cause data misinterpretation in studies that use voltage-sensitive dyes. Blebbistatin may also impact imaging of green fluorophores due to the spectral shift it causes in endogenous tissue fluorescence. 3D excitation-emission matrices of blebbistatin in precipitate form and in various solutions (DMSO, water, and 1 % aqueous albumin) revealed significant changes in the fluorescence of this molecule in different environments. Finally, we examined the reversibility of blebbistatin's uncoupling effect on cardiac contraction. Our findings provide important new information about the properties of this myosin II inhibitor, which will aid in the proper design and interpretation of studies that use this compound.
Collapse
Affiliation(s)
- Luther M Swift
- Department of Pharmacology and Physiology, The George Washington University Medical Center, 2300 Eye Street, Washington, DC 20037, USA
| | | | | | | | | | | |
Collapse
|
28
|
Actin cytoskeleton in cell polarity and asymmetric division during mouse oocyte maturation. Cytoskeleton (Hoboken) 2012; 69:727-37. [DOI: 10.1002/cm.21048] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 06/18/2012] [Accepted: 06/21/2012] [Indexed: 12/22/2022]
|
29
|
Wang Q, Racowsky C, Deng M. Mechanism of the chromosome-induced polar body extrusion in mouse eggs. Cell Div 2011; 6:17. [PMID: 21867530 PMCID: PMC3179692 DOI: 10.1186/1747-1028-6-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 08/25/2011] [Indexed: 12/11/2022] Open
Abstract
Background An oocyte undergoes two rounds of asymmetric division to generate a haploid gamete and two small polar bodies designed for apoptosis. Chromosomes play important roles in specifying the asymmetric meiotic divisions in the oocytes but the underlying mechanism is poorly understood. Results Chromosomes independently induce spindle formation and cortical actomyosin assembly into special cap and ring structures in the cortex of the oocyte. The spindle and the cortical cap/ring interact to generate mechanical forces, leading to polar body extrusion. Two distinct force-driven membrane changes were observed during 2nd polar body extrusion: a protrusion of the cortical cap and a membrane invagination induced by an anaphase spindle midzone. The cortical cap protrusion and invagination help rotate the spindle perpendicularly so that the spindle midzone can induce bilateral furrows at the shoulder of the protruding cap, leading to an abscission of the polar body. It is interesting to note that while the mitotic spindle midzone induces bilateral furrowing, leading to efficient symmetric division in the zygote, the meiotic spindle midzone induced cytokinetic furrowing only locally. Conclusions Distinct forces driving cortical cap protrusion and membrane invagination are involved in spindle rotation and polar body extrusion during meiosis II in mouse oocytes.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Obstetrics and Gynecology and Reproductive Biology, 75 Francis Street, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
30
|
Initial diameter of the polar body contractile ring is minimized by the centralspindlin complex. Dev Biol 2011; 359:137-148. [PMID: 21889938 DOI: 10.1016/j.ydbio.2011.08.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 08/02/2011] [Accepted: 08/17/2011] [Indexed: 11/20/2022]
Abstract
Polar body formation is an essential step in forming haploid eggs from diploid oocytes. This process involves completion of a highly asymmetric cytokinesis that results in a large egg and two small polar bodies. Unlike mitotic contractile rings, polar body contractile rings assemble over one spindle pole so that the spindle must move through the contractile ring before cytokinesis. During time-lapse imaging of C. elegans meiosis, the contractile ring moved downward along the length of the spindle and completed scission at the midpoint of the spindle, even when spindle length or rate of ring movement was increased. Patches of myosin heavy chain and dynamic furrowing of the plasma membrane over the entire embryo suggested that global cortical contraction forces the meiotic spindle and overlying membrane out through the contractile ring center. Consistent with this model, depletion of myosin phosphatase increased the velocity of ring movement along the length of the spindle. Global dynamic furrowing, which was restricted to anaphase I and II, was dependent on myosin II, the anaphase promoting complex and separase, but did not require cortical contact by the spindle. Large cortical patches of myosin during metaphase I and II indicated that myosin was already in the active form before activation of separase. To identify the signal at the midpoint of the anaphase spindle that induces scission, we depleted two proteins that mark the exact midpoint of the spindle during late anaphase, CYK-4 and ZEN-4. Depletion of either protein resulted in the unexpected phenotype of initial ingression of a polar body ring with twice the diameter of wild type. This phenotype revealed a novel mechanism for minimizing polar body size. Proteins at the spindle midpoint are required for initial ring ingression to occur close to the membrane-proximal spindle pole.
Collapse
|
31
|
Ellefson ML, McNally FJ. CDK-1 inhibits meiotic spindle shortening and dynein-dependent spindle rotation in C. elegans. ACTA ACUST UNITED AC 2011; 193:1229-44. [PMID: 21690306 PMCID: PMC3216336 DOI: 10.1083/jcb.201104008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Before chromosome expulsion into polar bodies during female meiosis, the APC inhibits CDK-1 to allow dynein-driven spindle rotation. In animals, the female meiotic spindle is positioned at the egg cortex in a perpendicular orientation to facilitate the disposal of half of the chromosomes into a polar body. In Caenorhabditis elegans, the metaphase spindle lies parallel to the cortex, dynein is dispersed on the spindle, and the dynein activators ASPM-1 and LIN-5 are concentrated at spindle poles. Anaphase-promoting complex (APC) activation results in dynein accumulation at spindle poles and dynein-dependent rotation of one spindle pole to the cortex, resulting in perpendicular orientation. To test whether the APC initiates spindle rotation through cyclin B–CDK-1 inactivation, separase activation, or degradation of an unknown dynein inhibitor, CDK-1 was inhibited with purvalanol A in metaphase-I–arrested, APC-depleted embryos. CDK-1 inhibition resulted in the accumulation of dynein at spindle poles and dynein-dependent spindle rotation without chromosome separation. These results suggest that CDK-1 blocks rotation by inhibiting dynein association with microtubules and with LIN-5–ASPM-1 at meiotic spindle poles and that the APC promotes spindle rotation by inhibiting CDK-1.
Collapse
Affiliation(s)
- Marina L Ellefson
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | | |
Collapse
|
32
|
Matsumura F, Yamakita Y, Yamashiro S. Myosin light chain kinases and phosphatase in mitosis and cytokinesis. Arch Biochem Biophys 2011; 510:76-82. [PMID: 21396909 PMCID: PMC3114266 DOI: 10.1016/j.abb.2011.03.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 03/02/2011] [Accepted: 03/03/2011] [Indexed: 01/23/2023]
Abstract
At mitosis, cells undergo drastic alterations in morphology and cytoskeletal organization including cell rounding during prophase, mitotic spindle assembly during prometaphase and metaphase, chromatid segregation in anaphase, and cytokinesis during telophase. It is well established that myosin II is a motor responsible for cytokinesis. Recent reports have indicated that myosin II is also involved in spindle assembly and karyokinesis. In this review, we summarize current understanding of the functions of myosin II in mitosis and cytokinesis of higher eukaryotes, and discuss the roles of possible upstream molecules that control myosin II in these mitotic events.
Collapse
Affiliation(s)
- Fumio Matsumura
- Department of Molecular Biology & Biochemistry, Rutgers, Piscataway, NJ 08854, USA.
| | | | | |
Collapse
|
33
|
Ca2+ signaling during mammalian fertilization: requirements, players, and adaptations. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a006767. [PMID: 21441584 DOI: 10.1101/cshperspect.a006767] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Changes in the intracellular concentration of calcium ([Ca(2+)](i)) represent a vital signaling mechanism enabling communication among cells and between cells and the environment. The initiation of embryo development depends on a [Ca(2+)](i) increase(s) in the egg, which is generally induced during fertilization. The [Ca(2+)](i) increase signals egg activation, which is the first stage in embryo development, and that consist of biochemical and structural changes that transform eggs into zygotes. The spatiotemporal patterns of [Ca(2+)](i) at fertilization show variability, most likely reflecting adaptations to fertilizing conditions and to the duration of embryonic cell cycles. In mammals, the focus of this review, the fertilization [Ca(2+)](i) signal displays unique properties in that it is initiated after gamete fusion by release of a sperm-derived factor and by periodic and extended [Ca(2+)](i) responses. Here, we will discuss the events of egg activation regulated by increases in [Ca(2+)](i), the possible downstream targets that effect these egg activation events, and the property and identity of molecules both in sperm and eggs that underpin the initiation and persistence of the [Ca(2+)](i) responses in these species.
Collapse
|
34
|
Abstract
Fertilization is a complex process comprised of numerous steps. During fertilization, two highly specialized and differentiated cells (sperm and egg) fuse and subsequently trigger the development of an embryo from a quiescent, arrested oocyte. Molecular interactions between the sperm and egg are necessary for regulating the developmental potential of an oocyte, and precise coordination and regulation of gene expression and protein function are critical for proper embryonic development. The nematode Caenorhabditis elegans has emerged as a valuable model system for identifying genes involved in fertilization and the oocyte-to-embryo transition as well as for understanding the molecular mechanisms that govern these processes. In this review, we will address current knowledge of the molecular underpinnings of gamete interactions during fertilization and the oocyte-to-embryo transition in C. elegans. We will also compare our knowledge of these processes in C. elegans to what is known about similar processes in mammalian, specifically mouse, model systems.
Collapse
Affiliation(s)
- Matthew R Marcello
- Waksman Institute and Department of Genetics Rutgers University, Piscataway, NJ 08854, USA.
| | | |
Collapse
|
35
|
Larson SM, Lee HJ, Hung PH, Matthews LM, Robinson DN, Evans JP. Cortical mechanics and meiosis II completion in mammalian oocytes are mediated by myosin-II and Ezrin-Radixin-Moesin (ERM) proteins. Mol Biol Cell 2010; 21:3182-92. [PMID: 20660156 PMCID: PMC2938384 DOI: 10.1091/mbc.e10-01-0066] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Revised: 07/01/2010] [Accepted: 07/12/2010] [Indexed: 11/11/2022] Open
Abstract
Cell division is inherently mechanical, with cell mechanics being a critical determinant governing the cell shape changes that accompany progression through the cell cycle. The mechanical properties of symmetrically dividing mitotic cells have been well characterized, whereas the contribution of cellular mechanics to the strikingly asymmetric divisions of female meiosis is very poorly understood. Progression of the mammalian oocyte through meiosis involves remodeling of the cortex and proper orientation of the meiotic spindle, and thus we hypothesized that cortical tension and stiffness would change through meiotic maturation and fertilization to facilitate and/or direct cellular remodeling. This work shows that tension in mouse oocytes drops about sixfold during meiotic maturation from prophase I to metaphase II and then increases ∼1.6-fold upon fertilization. The metaphase II egg is polarized, with tension differing ∼2.5-fold between the cortex over the meiotic spindle and the opposite cortex, suggesting that meiotic maturation is accompanied by assembly of a cortical domain with stiffer mechanics as part of the process to achieve asymmetric cytokinesis. We further demonstrate that actin, myosin-II, and the ERM (Ezrin/Radixin/Moesin) family of proteins are enriched in complementary cortical domains and mediate cellular mechanics in mammalian eggs. Manipulation of actin, myosin-II, and ERM function alters tension levels and also is associated with dramatic spindle abnormalities with completion of meiosis II after fertilization. Thus, myosin-II and ERM proteins modulate mechanical properties in oocytes, contributing to cell polarity and to completion of meiosis.
Collapse
Affiliation(s)
- Stephanie M. Larson
- *Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, and
| | - Hyo J. Lee
- *Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, and
| | - Pei-hsuan Hung
- *Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, and
| | - Lauren M. Matthews
- *Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, and
| | - Douglas N. Robinson
- Department of Cell Biology, and
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| | - Janice P. Evans
- *Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, and
| |
Collapse
|
36
|
Fabritius AS, Ellefson ML, McNally FJ. Nuclear and spindle positioning during oocyte meiosis. Curr Opin Cell Biol 2010; 23:78-84. [PMID: 20708397 DOI: 10.1016/j.ceb.2010.07.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Revised: 07/16/2010] [Accepted: 07/19/2010] [Indexed: 10/19/2022]
Abstract
Female meiosis is unique in that an asymmetrically positioned meiotic spindle expels chromosomes into tiny, non-developing polar bodies. The extrusion of chromosomes into polar bodies is always mediated by meiotic spindles that are attached to the oocyte cortex by one pole. The asymmetric, cortical positioning of the oocyte meiotic spindle preserves the volume and contents of the oocyte. Recent work in C. elegans and mouse has provided mechanistic details of spindle positioning in oocytes.
Collapse
Affiliation(s)
- Amy S Fabritius
- Department of Molecular and Cellular Biology, University of California-Davis, Davis, CA 95616, United States
| | | | | |
Collapse
|
37
|
The gamma isoform of CaM kinase II controls mouse egg activation by regulating cell cycle resumption. Proc Natl Acad Sci U S A 2009; 107:81-6. [PMID: 19966304 DOI: 10.1073/pnas.0912658106] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Fertilization triggers a rise in intracellular Ca(2+) concentration ([Ca(2+)](i)) in the egg that initiates a series of events known as egg activation. These events include cortical granule exocytosis that establishes a block to polyspermy, resumption of meiosis, and recruitment of maternal mRNAs into polysomes for translation. Several calcium-dependent proteins, including calcium/calmodulin-dependent protein kinase II (CaMKII), have been implicated in egg activation. However, the precise role of CaMKII in mediating specific events of egg activation and the identity of the isoform(s) present in mouse eggs have not been unequivocally established. Through targeted deletion of the gamma isoform of CaMKII, we find that CaMKIIgamma is the predominant CaMKII isoform in mouse eggs and that it is essential for egg activation. Although CaMKIIgamma(-/-) eggs exhibit a normal pattern of Ca(2+) oscillations after insemination and undergo cortical granule exocytosis, they fail to resume meiosis or to recruit maternal mRNAs. Surprisingly, we find that the recruitment of maternal mRNAs does not directly depend on CaMKII, but requires elevated [Ca(2+)](i) and metaphase II exit. We conclude that CaMKIIgamma specifically controls mouse egg activation by regulating cell cycle resumption.
Collapse
|
38
|
Chang HY, Minahan K, Merriman JA, Jones KT. Calmodulin-dependent protein kinase gamma 3 (CamKIIgamma3) mediates the cell cycle resumption of metaphase II eggs in mouse. Development 2009; 136:4077-81. [PMID: 19906843 DOI: 10.1242/dev.042143] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Mature mammalian eggs are ovulated arrested at meiotic metaphase II. Sperm break this arrest by an oscillatory Ca(2+) signal that is necessary and sufficient for the two immediate events of egg activation: cell cycle resumption and cortical granule release. Previous work has suggested that cell cycle resumption, but not cortical granule release, is mediated by calmodulin-dependent protein kinase II (CamKII). Here we find that mouse eggs contain detectable levels of only one CamKII isoform, gamma 3. Antisense morpholino knockdown of CamKIIgamma3 during oocyte maturation produces metaphase II eggs that are insensitive to parthenogenetic activation by Ca(2+) stimulation and insemination. The effect is specific to this morpholino, as a 5-base-mismatch morpholino is without effect, and is rescued by CamKIIgamma3 or constitutively active CamKII cRNAs. Although CamKII-morpholino-treated eggs fail to exit metaphase II arrest, cortical granule exocytosis is not blocked. Therefore, CamKIIgamma3 plays a necessary and sufficient role in transducing the oscillatory Ca(2+) signal into cell cycle resumption, but not into cortical granule release.
Collapse
Affiliation(s)
- Heng-Yu Chang
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | | | | | | |
Collapse
|
39
|
Deng M, Li R. Sperm chromatin-induced ectopic polar body extrusion in mouse eggs after ICSI and delayed egg activation. PLoS One 2009; 4:e7171. [PMID: 19787051 PMCID: PMC2746308 DOI: 10.1371/journal.pone.0007171] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Accepted: 08/26/2009] [Indexed: 12/05/2022] Open
Abstract
Meiotic chromosomes in an oocyte are not only a maternal genome carrier but also provide a positional signal to induce cortical polarization and define asymmetric meiotic division of the oocyte, resulting in polar body extrusion and haploidization of the maternal genome. The meiotic chromosomes play dual function in determination of meiosis: 1) organizing a bipolar spindle formation and 2) inducing cortical polarization and assembly of a distinct cortical cytoskeleton structure in the overlying cortex for polar body extrusion. At fertilization, a sperm brings exogenous paternal chromatin into the egg, which induces ectopic cortical polarization at the sperm entry site and leads to a cone formation, known as fertilization cone. Here we show that the sperm chromatin-induced fertilization cone formation is an abortive polar body extrusion due to lack of spindle induction by the sperm chromatin during fertilization. If experimentally manipulating the fertilization process to allow sperm chromatin to induce both cortical polarization and spindle formation, the fertilization cone can be converted into polar body extrusion. This suggests that sperm chromatin is also able to induce polar body extrusion, like its maternal counterpart. The usually observed cone formation instead of ectopic polar body extrusion induced by sperm chromatin during fertilization is due to special sperm chromatin compaction which restrains it from rapid spindle induction and therefore provides a protective mechanism to prevent a possible paternal genome loss during ectopic polar body extrusion.
Collapse
Affiliation(s)
- Manqi Deng
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- * E-mail: (MD); (RL)
| | - Rong Li
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- * E-mail: (MD); (RL)
| |
Collapse
|
40
|
Sosnik J, Miranda PV, Spiridonov NA, Yoon SY, Fissore RA, Johnson GR, Visconti PE. Tssk6 is required for Izumo relocalization and gamete fusion in the mouse. J Cell Sci 2009; 122:2741-9. [PMID: 19596796 DOI: 10.1242/jcs.047225] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
One of the most important processes in fertilization is the fusion of egg and sperm; however, the molecular mechanisms involved in this process are not well understood. So far, using genetic approaches, only two proteins have been demonstrated to be necessary for this process: Izumo in sperm and CD9 in the egg. Here we demonstrate that sperm produced by Tssk6 (Sstk)-null mice present defects that prevent the successful fertilization of eggs in vitro and the fusion to zona-pellucida-free eggs. Tssk6 is a member of the testis-specific serine kinase family of proteins and is expressed postmeiotically in male germ cells. In order for fusion to occur, during the process known as acrosome reaction Izumo needs to relocate from the anterior head to other regions, including the postacrosomal compartment. Tssk6-null sperm fails to relocate Izumo during the acrosome reaction. Agents that interfere with actin dynamics blocked the acrosome-reaction-associated translocation of Izumo that is required for fusion in wild-type sperm. Additionally, actin polymerization was compromised in Tssk6-null sperm. Taken together, our results indicate that Tssk6 is involved in sperm-egg fusion through the regulation of actin polymerization and changes in Izumo localization.
Collapse
Affiliation(s)
- Julian Sosnik
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Ai JS, Wang Q, Yin S, Shi LH, Xiong B, OuYang YC, Hou Y, Chen DY, Schatten H, Sun QY. Regulation of peripheral spindle movement and spindle rotation during mouse oocyte meiosis: new perspectives. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2008; 14:349-356. [PMID: 18598570 DOI: 10.1017/s1431927608080343] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Spindle movement, including spindle migration during first meiosis and spindle rotation during second meiosis, is essential for asymmetric divisions in mouse oocytes. Previous studies by others and us have shown that microfilaments are required for both spindle migration and rotation. In the present study, we aimed to further investigate the mechanism controlling spindle movement during mouse oocyte meiosis. By employing drug treatment and immunofluorescence microscopy, we showed that dynamic microtubule assembly was involved in both spindle migration and rotation. Furthermore, we found that the calcium/CaM/CaMKII pathway was important for regulating spindle rotation.
Collapse
Affiliation(s)
- Jun-Shu Ai
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Tsaadon L, Kaplan-Kraicer R, Shalgi R. Myristoylated alanine-rich C kinase substrate, but not Ca2+/calmodulin-dependent protein kinase II, is the mediator in cortical granules exocytosis. Reproduction 2008; 135:613-24. [DOI: 10.1530/rep-07-0554] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Sperm–egg fusion induces cortical granules exocytosis (CGE), a process that ensures the block to polyspermy. CGE can be induced independently by either a rise in intracellular calcium concentration or protein kinase C (PKC) activation. We have previously shown that myristoylated alanine-rich C kinase substrate (MARCKS) cross-links filamentous actin (F-actin) and regulates its reorganization. This activity is reduced either by PKC-induced MARCKS phosphorylation (PKC pathway) or by its direct binding to calmodulin (CaM; CaM pathway), both inducing MARCKS translocation, F-actin reorganization, and CGE. Currently, we examine the involvement of Ca2+/CaM-dependent protein kinase II (CaMKII) and MARCKS in promoting CGE and show that PKC pathway can compensate for lack of Ca2+/CaM pathway. Microinjecting eggs with either overexpressed protein or complementary RNA of constitutively active αCaMKII triggered resumption of second meiotic division, but induced CGE of an insignificant magnitude compared with CGE induced by wt αCaMKII. Microinjecting eggs with mutant-unphosphorylatable MARCKS reduced the intensity of 12-O-tetradecanoylphorbol 13-acetate or ionomycin-induced CGE by 50%, indicating that phosphorylation of MARCKS by novel and/or conventional PKCs (n/cPKCs) is a pivotal event associated with CGE. Moreover, we were able to demonstrate cPKCs involvement in ionomycin-induced MARCKS translocation and CGE. These results led us to propose that MARCKS, rather than CaMKII, as a key mediator of CGE.
Collapse
|
43
|
Lucas-Lopez C, Allingham JS, Lebl T, Lawson CPAT, Brenk R, Sellers JR, Rayment I, Westwood NJ. The small molecule tool (S)-(-)-blebbistatin: novel insights of relevance to myosin inhibitor design. Org Biomol Chem 2008; 6:2076-84. [PMID: 18528569 DOI: 10.1039/b801223g] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The small molecule blebbistatin is now a front line tool in the study of myosin function. Chemical modification of the tricyclic core of blebbistatin could deliver the next generation of myosin inhibitors and to help address this we report here on the impact of structural changes in the methyl-substituted aromatic ring of blebbistatin on its biological activity. Chemical methods for the preparation of isomeric methyl-containing analogues are reported and a series of co-crystal structures are used to rationalise the observed variations in their biological activity. These studies further support the view that the previously identified binding mode of blebbistatin to Dictyostelium discoideum myosin II is of relevance to its mode of action. A discussion of the role that these observations have on planning the synthesis of focused libraries of blebbistatin analogues is also provided including an assessment of possibilities by computational methods. These studies are ultimately directed at the development of novel myosin inhibitors with improved affinity and different selectivity profiles from blebbistatin itself.
Collapse
Affiliation(s)
- Cristina Lucas-Lopez
- School of Chemistry and the Centre for Biomolecular Sciences, University of St Andrews, North Haugh, St Andrews, Fife, UK
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Galat V, Zhou Y, Taborn G, Garton R, Iannaccone P. Overcoming MIII arrest from spontaneous activation in cultured rat oocytes. CLONING AND STEM CELLS 2008; 9:303-14. [PMID: 17907941 DOI: 10.1089/clo.2006.0059] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The rat oocyte spontaneously activates under a wide variety of conditions. This process progresses to MIII arrest that is not responsive to parthenogenetic activation and development. Insofar as activation involves extrusion of the second polar body (PBII), we set out to determine if preventing this step by inhibiting microfilaments would change the course of spontaneous activation (SA). In particular, how long does the effect of SA persist while retaining reversibility of PBII extrusion once inhibitors are removed? We wanted to determine if the eggs would be responsive to parthenogenetic activation and capable of resuming development once a permanent inhibition is achieved. We set out to determine whether SA would depend on the ovular age of oocytes. Inhibiting of PBII extrusion was achieved by affecting microtubules with demecolcine or nocodazole or actin filaments with cytochalasin B (CB) and cytochalasin D (CD). We found that all oocytes undergo SA and progression to MIII; however, the rapidity of spontaneous activation is a function of the ovular age of the oocyte. The resumption of the meiosis period changes dramatically from 20 to 180 min with decreasing ovular age. We established that suppression of PB formation can be effectively achieved in oocytes of younger ovular age, and that inhibition of PB extrusion became irreversible after 3.5 h of treatment. We established that drug-treated oocytes could undergo subsequent reactivation and in vitro development to blastocysts. The rate of in vitro development of cytochalasin-treated group was comparable to parthenogenetic controls, while nocodazole and demecolcine produced oocytes that developed at lower frequencies. Thus, the application of the microfilament inhibiting drugs helps to overcome the negative effect of SA that results in MIII arrest. Here we also show optimized parthenogenetic stimulation that resulted in development to the blastocyst stage at frequency comparable to development of fertilized embryos.
Collapse
Affiliation(s)
- Vasiliy Galat
- Department of Pediatrics, Northwestern University Feinberg School of Medicine and the Developmental Biology Program of Children's Memorial Research Center, Chicago, Illinois 60614, USA
| | | | | | | | | |
Collapse
|
45
|
Involvement of calcium/calmodulin-dependent protein kinase kinase in meiotic maturation of pig oocytes. Anim Reprod Sci 2008; 111:17-30. [PMID: 18367350 DOI: 10.1016/j.anireprosci.2008.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Revised: 02/04/2008] [Accepted: 02/06/2008] [Indexed: 11/20/2022]
Abstract
Calcium (Ca(2+))/calmodulin-dependent protein kinase kinase (CaMKK) is a novel member of Ca(2+)/calmodulin-dependent protein kinase (CaMK) family, whose physiological roles in regulating meiotic cell cycle needs to be determined. We showed by Western blot that CaMKK was expressed in pig oocytes at various maturation stages. Confocal microscopy was employed to observe CaMKK distribution. In oocytes at the germinal vesicle (GV) or prometaphase I (pro-MI) stage, CaMKK was distributed in the nucleus, around the condensed chromatin and the cortex of the cell. At metaphase I (MI) stage, CaMKK was concentrated in the cortex of the cell. After transition to anaphase I or telophase I stage, CaMKK was detected around the separating chromosomes and in the cortex of the cell. At metaphase II (MII) stage, CaMKK was localized to the cortex of the cell, with a thicker area near the first polar body (PB1). Treatment of pig cumulus-enclosed oocytes with STO-609, a membrane-permeable CaMKK inhibitor, resulted in the delay/inhibition of the meiotic resumption and the inhibition of first polar body emission. The correlation between CaMKK and microfilaments during meiotic maturation of pig oocytes was then studied. CaMKK and microfilaments were colocalized from MI to MII during porcine oocyte maturation. After oocytes were treated with STO-609, microfilaments were depolymerized, while in oocytes exposed to cytochalasin B (CB), a microfilament polymerization inhibitor, CaMKK became diffused evenly throughout the cell. These data suggest that CaMKK is involved in regulating the meiotic cell cycle probably by interacting with microfilaments in pig oocytes.
Collapse
|
46
|
Ducibella T, Fissore R. The roles of Ca2+, downstream protein kinases, and oscillatory signaling in regulating fertilization and the activation of development. Dev Biol 2008; 315:257-79. [PMID: 18255053 DOI: 10.1016/j.ydbio.2007.12.012] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Revised: 12/12/2007] [Accepted: 12/13/2007] [Indexed: 12/12/2022]
Abstract
Reviews in Developmental Biology have covered the pathways that generate the all-important intracellular calcium (Ca(2+)) signal at fertilization [Miyazaki, S., Shirakawa, H., Nakada, K., Honda, Y., 1993a. Essential role of the inositol 1,4,5-trisphosphate receptor/Ca(2+) release channel in Ca(2+) waves and Ca(2+) oscillations at fertilization of mammalian eggs. Dev. Biol. 158, 62-78; Runft, L., Jaffe, L., Mehlmann, L., 2002. Egg activation at fertilization: where it all begins. Dev. Biol. 245, 237-254] and the different temporal responses of Ca(2+) in many organisms [Stricker, S., 1999. Comparative biology of calcium signaling during fertilization and egg activation in animals. Dev. Biol. 211, 157-176]. Those reviews raise the importance of identifying how Ca(2+) causes the events of egg activation (EEA) and to what extent these temporal Ca(2+) responses encode developmental information. This review covers recent studies that have analyzed how these Ca(2+) signals are interpreted by specific proteins, and how these proteins regulate various EEA responsible for the onset of development. Many of these proteins are protein kinases (CaMKII, PKC, MPF, MAPK, MLCK) whose activity is directly or indirectly regulated by Ca(2+), and whose amount increases during late oocyte maturation. We cover biochemical progress in defining the signaling pathways between Ca(2+) and the EEA, as well as discuss how oscillatory or multiple Ca(2+) signals are likely to have specific advantages biochemically and/or developmentally. These emerging concepts are put into historical context, emphasizing that key contributions have come from many organisms. The intricate interdependence of Ca(2+), Ca(2+)-dependent proteins, and the EEA raise many new questions for future investigations that will provide insight into the extent to which fertilization-associated signaling has long-range implications for development. In addition, answers to these questions should be beneficial to establishing parameters of egg quality for human and animal IVF, as well as improving egg activation protocols for somatic cell nuclear transfer to generate stem cells and save endangered species.
Collapse
Affiliation(s)
- Tom Ducibella
- Department of OB/GYN, Tufts-New England Medical Center, Boston, MA 02111, USA.
| | | |
Collapse
|
47
|
AI JS, WANG Q, LI M, SHI LH, OLA SI, XIONG B, YIN S, CHEN DY, SUN QY. Roles of Microtubules and Microfilaments in Spindle Movements During Rat Oocyte Meiosis. J Reprod Dev 2008; 54:391-6. [DOI: 10.1262/jrd.20034] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Jun-Shu AI
- State Key Laboratory of Reproductive Biology, Institute of Zoology
- Graduate School, Chinese Academy of Sciences
| | - Qiang WANG
- State Key Laboratory of Reproductive Biology, Institute of Zoology
- Graduate School, Chinese Academy of Sciences
| | - Mo LI
- State Key Laboratory of Reproductive Biology, Institute of Zoology
- Graduate School, Chinese Academy of Sciences
| | - Li-Hong SHI
- State Key Laboratory of Reproductive Biology, Institute of Zoology
- Graduate School, Chinese Academy of Sciences
| | - Safiriyu Idowu OLA
- State Key Laboratory of Reproductive Biology, Institute of Zoology
- Department of Animal Science, Obafemi Awolowo University
| | - Bo XIONG
- State Key Laboratory of Reproductive Biology, Institute of Zoology
- Graduate School, Chinese Academy of Sciences
| | - Shen YIN
- State Key Laboratory of Reproductive Biology, Institute of Zoology
- Graduate School, Chinese Academy of Sciences
| | - Da-Yuan CHEN
- State Key Laboratory of Reproductive Biology, Institute of Zoology
| | - Qing-Yuan SUN
- State Key Laboratory of Reproductive Biology, Institute of Zoology
| |
Collapse
|
48
|
Horner VL, Wolfner MF. Transitioning from egg to embryo: Triggers and mechanisms of egg activation. Dev Dyn 2008; 237:527-44. [DOI: 10.1002/dvdy.21454] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
49
|
Abstract
1. Mammalian eggs are arrested at metaphase of their second meiotic division when ovulated and remain arrested until fertilized. The sperm delivers into the egg phospholipase C (PLC) zeta, which triggers a series of Ca(2+) spikes lasting several hours. The Ca(2+) spikes provide the necessary and sufficient trigger for all the events of fertilization, including exit from metaphase II arrest and extrusion of cortical granules that block the entry of other sperm. 2. The oscillatory Ca(2+) signal switches on calmodulin-dependent protein kinase II (CaMKII), which phosphorylates the egg-specific protein Emi2, earmarking it for degradation. To remain metaphase II arrested, eggs must maintain high levels of maturation-promoting factor (MPF) activity, a heterodimer of CDK1 and cyclin B1. Emi2 prevents loss of MPF by blocking cyclin B1 degradation, a process that is achieved by inhibiting the activity of the anaphase-promoting complex/cyclosome. However, CaMKII is not the primary initiator in the extrusion of cortical granules. 3. Ca(2+) spiking is also observed in mitosis of one-cell embryos, probably because PLCzeta contains a nuclear localization signal and so is released into the cytoplasm following nuclear envelope breakdown. The function of these mitotic Ca(2+) spikes remains obscure, although they are not absolutely required for passage through mitosis. 4. Intriguingly, the pattern of Ca(2+) spikes observed at fertilization has an effect on both pre- and postimplantation development in a manner that is independent of their ability to activate eggs. This suggests that the Ca(2+) spikes set in train at fertilization are having effects on processes initiated in the newly fertilized egg but whose influences are only observed several cell divisions later. The nature of the signals remains little explored, but their importance is clear and so warrants further investigation.
Collapse
Affiliation(s)
- Keith T Jones
- Institute for Cell and Molecular Biosciences, The Medical School, Framlington Place, University of Newcastle, Newcastle, UK.
| |
Collapse
|
50
|
Gardner AJ, Knott JG, Jones KT, Evans JP. CaMKII can participate in but is not sufficient for the establishment of the membrane block to polyspermy in mouse eggs. J Cell Physiol 2007; 212:275-80. [PMID: 17455234 DOI: 10.1002/jcp.21046] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Fertilization triggers initiation of development and establishment of blocks on the egg coat and plasma membrane to prevent fertilization by multiple sperm (polyspermy). The mechanism(s) by which mammalian eggs establish the membrane block to polyspermy is largely unknown. Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) appears to be the key regulator of several egg activation events (completion of meiosis, progression to embryonic interphase, recruitment of maternal mRNAs). Since sperm-induced increases in cytosolic Ca(2+) play a role in establishment of the membrane block to polyspermy in mouse eggs, we hypothesized that CaMKII was a Ca(2+)-dependent effector leading to this change in egg membrane function. To test this hypothesis, we modulated CaMKII activity in two ways: activating eggs parthenogenetically by introducing constitutively active CaMKIIalpha (CA-CaMKII) into unfertilized eggs, and inhibiting endogenous CaMKII in fertilized eggs with myristoylated autocamtide 2-related inhibitory peptide (myrAIP). We find that eggs treated with myrAIP establish a less effective membrane block to polyspermy than do control eggs, but that CA-CaMKII is not sufficient for membrane block establishment, despite the fact that CA-CaMKII-activated eggs undergo other egg activation events. This suggests that: (1) CaMKII activity contributes to the membrane block, but this not faithfully mimicked by CA-CaMKII and furthermore, other pathways, in addition to those activated by Ca(2+) and CaMKII, also participate in membrane block establishment; (2) CA-CaMKII has a range of effects as a parthenogenetic trigger of egg activation (high levels of cell cycle resumption, modest levels of cortical granule exocytosis, and no membrane block establishment).
Collapse
Affiliation(s)
- Allison J Gardner
- Department of Biochemistry and Molecular Biology, Division of Reproductive Biology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | | | | | | |
Collapse
|