1
|
Hu C, Jiang Y, Ma C, Xu F, Cui C, Du X, Chen J, Zhu L, Yu S, He X, Yu W, Wang Y, Xu X. Decreased Cdk2 Activity Hindered Embryonic Development and Parthenogenesis Induction in Silkworm, Bombyx mori L. Int J Mol Sci 2025; 26:3341. [PMID: 40244186 PMCID: PMC11989892 DOI: 10.3390/ijms26073341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Cyclin-dependent protein kinase 2 (Cdk2), an important member of the serine/threonine-specific protein kinase family, plays a critical regulatory role in biological processes. Previous studies have demonstrated that Cdk2 is involved in the arrest and resumption of meiosis in mammalian oocytes. In this study, we explored the function of Cdk2 through parthenogenetic lines (PLs) and corresponding amphigonic lines (ALs) in a model lepidopteran insect silkworm, Bombyx mori L. Our findings revealed a positive correlation between Cdk2 activity and the parthenogenesis induction rate. The pharmacological inhibition of Cdk2 using the specific inhibitor AUZ454 not only significantly reduced the parthenogenesis induction rate but also caused developmental delays in embryos. These results demonstrate that Cdk2 is essential for parthenogenesis success and is a potential target gene for biological reproductive regulation.
Collapse
Affiliation(s)
- Chengjie Hu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Yonghou Jiang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Chenkai Ma
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Fang Xu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Chunguang Cui
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Xin Du
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Jine Chen
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Linbao Zhu
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Shaofang Yu
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Xingjian He
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Wei Yu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Yongqiang Wang
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Xia Xu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| |
Collapse
|
2
|
Puls O, Ruiz-Reynés D, Tavella F, Jin M, Kim Y, Gelens L, Yang Q. Spatial heterogeneity accelerates phase-to-trigger wave transitions in frog egg extracts. Nat Commun 2024; 15:10455. [PMID: 39622792 PMCID: PMC11612452 DOI: 10.1038/s41467-024-54752-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 11/19/2024] [Indexed: 12/06/2024] Open
Abstract
Cyclin-dependent kinase 1 (Cdk1) activity rises and falls throughout the cell cycle: a cell-autonomous process called mitotic oscillations. Mitotic oscillators can synchronize when spatially coupled, facilitating rapid, synchronous divisions in large early embryos of Drosophila (~0.5 mm) and Xenopus (~1.2 mm). Diffusion alone cannot achieve such long-range coordination. Instead, studies proposed mitotic waves-phase and trigger waves-as mechanisms of the coordination. How waves establish over time remains unclear. Using Xenopus laevis egg extracts and a Cdk1 Förster resonance energy transfer sensor, we observe a transition from phase to trigger wave dynamics in initially homogeneous cytosol. Spatial heterogeneity promotes this transition. Adding nuclei accelerates entrainment. The system transitions almost immediately when driven by metaphase-arrested extracts. Numerical simulations suggest phase waves appear transiently as trigger waves take time to entrain the system. Therefore, we show that both waves belong to a single biological process capable of coordinating the cell cycle over long distances.
Collapse
Affiliation(s)
- Owen Puls
- Department of Physics, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biophysics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Daniel Ruiz-Reynés
- Laboratory of Dynamics in Biological Systems, KU Leuven, Department of Cellular and Molecular Medicine, University of Leuven, B-3000, Leuven, Belgium
- IFISC (CSIC-UIB). Instituto de Física Interdisciplinar y Sistemas Complejos, E-07122, Palma de Mallorca, Spain
| | - Franco Tavella
- Department of Biophysics, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Minjun Jin
- Department of Biophysics, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yeonghoon Kim
- Department of Biophysics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lendert Gelens
- Laboratory of Dynamics in Biological Systems, KU Leuven, Department of Cellular and Molecular Medicine, University of Leuven, B-3000, Leuven, Belgium.
| | - Qiong Yang
- Department of Physics, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biophysics, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
3
|
Kouznetsova A, Valentiniene S, Liu JG, Kitajima TS, Brismar H, Höög C. Aurora B and Aurora C pools at two chromosomal regions collaboratively maintain chromosome alignment and prevent aneuploidy at the second meiotic division in mammalian oocytes. Front Cell Dev Biol 2024; 12:1470981. [PMID: 39355122 PMCID: PMC11442388 DOI: 10.3389/fcell.2024.1470981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/04/2024] [Indexed: 10/03/2024] Open
Abstract
Correct chromosome segregation is essential to preserve genetic integrity. The two protein kinases, Aurora B and its meiotic homolog Aurora C, regulate attachments between chromosomal kinetochores and microtubules, thereby contributing to the accuracy of the chromosome segregation process. Here we performed a detailed examination of the localization and activity of Aurora B/C kinases, their partner Incenp and the kinetochore target Hec1, during the second meiotic division in mouse oocytes. We found that a majority of Aurora B and C changed their localization from the outer kinetochore region of chromosomes at prometaphase II to an inner central region localized between sister centromeres at metaphase II. Depletion of the Aurora B/C pool at the inner central region using the haspin kinase inhibitor 5-iodotubercidin resulted in chromosome misalignments at the metaphase II stage. To further understand the role of the Aurora B/C pool at the central region, we examined the behaviour of single chromatids, that lack a central Aurora B/C pool but retain Aurora B/C at the outer kinetochores. We found that kinetochore-microtubule attachments at single chromatids were corrected at both prometaphase II and metaphase II stages, but that single chromatids compared to paired chromatids were more prone to misalignments following treatment of oocytes with the Aurora B/C inhibitory drugs AZD1152 and GSK1070916. We conclude that the Aurora B/C pool at the inner central region stabilizes chromosome alignment during metaphase II arrest, while Aurora B/C localized at the kinetochore assist in re-establishing chromosome positioning at the metaphase plate if alignment is lost. Collaboratively these two pools prevent missegregation and aneuploidy at the second meiotic division in mammalian oocytes.
Collapse
Affiliation(s)
- Anna Kouznetsova
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sonata Valentiniene
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jian-Guo Liu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Tomoya S. Kitajima
- Laboratory for Chromosome Segregation, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Hjalmar Brismar
- Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Stockholm, Sweden
| | - Christer Höög
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Puls O, Ruiz-Reynés D, Tavella F, Jin M, Kim Y, Gelens L, Yang Q. Mitotic waves in frog egg extracts: Transition from phase waves to trigger waves. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576267. [PMID: 38496576 PMCID: PMC10942321 DOI: 10.1101/2024.01.18.576267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Cyclin-dependent kinase 1 (Cdk1) activity rises and falls throughout the cell cycle, a cell-autonomous process known as mitotic oscillations. These oscillators can synchronize when spatially coupled, providing a crucial foundation for rapid synchronous divisions in large early embryos like Drosophila (~ 0.5 mm) and Xenopus (~ 1.2 mm). While diffusion alone cannot achieve such long-range coordination, recent studies have proposed two types of mitotic waves, phase and trigger waves, to explain the phenomena. How the waves establish over time for efficient spatial coordination remains unclear. Using Xenopus laevis egg extracts and a Cdk1 FRET sensor, we observe a transition from phase waves to a trigger wave regime in an initially homogeneous cytosol. Adding nuclei accelerates such transition. Moreover, the system transitions almost immediately to this regime when externally driven by metaphase-arrested extracts from the boundary. Employing computational modeling, we pinpoint how wave nature, including speed-period relation, depends on transient dynamics and oscillator properties, suggesting that phase waves appear transiently due to the time required for trigger waves to entrain the system and that spatial heterogeneity promotes entrainment. Therefore, we show that both waves belong to a single biological process capable of coordinating the cell cycle over long distances.
Collapse
Affiliation(s)
- Owen Puls
- Department of Physics, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel Ruiz-Reynés
- Laboratory of Dynamics in Biological Systems, KU Leuven, Department of Cellular and Molecular Medicine, University of Leuven, B-3000 Leuven, Belgium
- IFISC (CSIC-UIB). Instituto de Física Interdisciplinar y Sistemas Complejos, E-07122 Palma de Mallorca, Spain
| | - Franco Tavella
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Minjun Jin
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yeonghoon Kim
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lendert Gelens
- Laboratory of Dynamics in Biological Systems, KU Leuven, Department of Cellular and Molecular Medicine, University of Leuven, B-3000 Leuven, Belgium
| | - Qiong Yang
- Department of Physics, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
5
|
Bahrami N, Nazari A, Afshari Z, Aftabsavad S, Moini A, Noormohammadi Z. Gene expression and demographic analyses in women with the poor ovarian response: a computational approach. J Assist Reprod Genet 2023; 40:2627-2638. [PMID: 37642817 PMCID: PMC10643739 DOI: 10.1007/s10815-023-02919-4] [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: 06/12/2023] [Accepted: 08/17/2023] [Indexed: 08/31/2023] Open
Abstract
PURPOSE Poor response to ovarian stimulation (POR) typically is reflected as decreased follicular response and low estradiol (E2) levels following ovarian stimulation by FSH/HMG. Many genes are involved in oocyte maturation, and demographic features and lifestyle can affect the oocyte maturity and developmental competence. The present study was conducted to investigate the magnitude of gene expression and lifestyle habits in POR women as compared to healthy women, using different statistical and computational methods. METHODS Fifty women in the two groups were studied. The study groups included POR women (n = 25) with 1-9 released oocytes, and the control group (normal women, n = 25) with 9-15 released oocytes. Quantitative PCR was used to estimate the expression of FIGLA, ZAR1, WNT4, LHX8, APC, H1FOO, MOS, and DMC1 genes in granulosa cells. RESULTS The results showed no significant difference in the magnitude of the studied genes' expression and linear discriminant analysis did not differentiate the studied groups based on all the genes together. Redundancy analysis (RDA) and latent factor mixed model (LFMM) results produce no significant association between the genes' expression magnitude and the geographical variables of the patients' local habitat. Linear discriminant analysis (LDA) of the demographic features differentiated the two groups of women. CONCLUSION Our results indicate that demographic features may have an effect on sample gene expression levels.
Collapse
Affiliation(s)
- Nastaran Bahrami
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Arnoosh Nazari
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Afshari
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Somayeh Aftabsavad
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ashraf Moini
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
- Breast Disease Research Center (BDRC), Tehran University of Medical Science, Tehran, Iran
- Department of Obstetrics and Gynecology, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Noormohammadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
6
|
Tokmakov AA, Morichika Y, Teranishi R, Sato KI. Oxidative Stress-Induced Overactivation of Frog Eggs Triggers Calcium-Dependent Non-Apoptotic Cell Death. Antioxidants (Basel) 2022; 11:antiox11122433. [PMID: 36552641 PMCID: PMC9774297 DOI: 10.3390/antiox11122433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Excessive activation of frog eggs (overactivation) is a pathological process that renders eggs unfertilizable. Its physiological inducers are unknown. Previously, oxidative stress was shown to cause time- and dose-dependent overactivation of Xenopus laevis frog eggs. Here, we demonstrate that the oxidative stress-induced egg overactivation is a calcium-dependent phenomenon which can be attenuated in the presence of the selective calcium chelator BAPTA. Degradation of cyclin B2, which is known to be initiated by calcium transient in fertilized or parthenogenetically activated eggs, can also be observed in the overactivated eggs. Decline in mitochondrial membrane potential, ATP depletion and termination of protein synthesis manifest in the eggs within one hour of triggering overactivation. These intracellular events occur in the absence of caspase activation. Furthermore, plasma membrane integrity is compromised in the overactivated eggs, as evidenced by ATP leakage and egg swelling. In sum, our data demonstrate that oxidative stress-induced overactivation of frog eggs causes fast and dramatic disruption of cellular homeostasis, resulting in robust and expedited cell death by a calcium-dependent non-apoptotic mechanism.
Collapse
Affiliation(s)
- Alexander A. Tokmakov
- Institute of Advanced Technoogy, Faculty of Biology-Oriented Science and Technology, KinDai University, 930 Nishimitani, Kinokawa City 649-6493, Japan
- Correspondence:
| | - Yudai Morichika
- Laboratory of Cell Signaling and Development, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Ryuga Teranishi
- Laboratory of Cell Signaling and Development, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Ken-Ichi Sato
- Laboratory of Cell Signaling and Development, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| |
Collapse
|
7
|
Wassmann K. Separase Control and Cohesin Cleavage in Oocytes: Should I Stay or Should I Go? Cells 2022; 11:3399. [PMID: 36359795 PMCID: PMC9656630 DOI: 10.3390/cells11213399] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 10/19/2023] Open
Abstract
The key to gametogenesis is the proper execution of a specialized form of cell division named meiosis. Prior to the meiotic divisions, the recombination of maternal and paternal chromosomes creates new genetic combinations necessary for fitness and adaptation to an ever-changing environment. Two rounds of chromosome segregation -meiosis I and II- have to take place without intermediate S-phase and lead to the creation of haploid gametes harboring only half of the genetic material. Importantly, the segregation patterns of the two divisions are fundamentally different and require adaptation of the mitotic cell cycle machinery to the specificities of meiosis. Separase, the enzyme that cleaves Rec8, a subunit of the cohesin complex constituting the physical connection between sister chromatids, has to be activated twice: once in meiosis I and immediately afterwards, in meiosis II. Rec8 is cleaved on chromosome arms in meiosis I and in the centromere region in meiosis II. This step-wise cohesin removal is essential to generate gametes of the correct ploidy and thus, embryo viability. Hence, separase control and Rec8 cleavage must be perfectly controlled in time and space. Focusing on mammalian oocytes, this review lays out what we know and what we still ignore about this fascinating mechanism.
Collapse
Affiliation(s)
- Katja Wassmann
- Institut Jacques Monod, Université Paris Cité, CNRS, 75013 Paris, France
| |
Collapse
|
8
|
Ozturk S. Molecular determinants of the meiotic arrests in mammalian oocytes at different stages of maturation. Cell Cycle 2022; 21:547-571. [PMID: 35072590 PMCID: PMC8942507 DOI: 10.1080/15384101.2022.2026704] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/21/2021] [Accepted: 01/03/2022] [Indexed: 01/26/2023] Open
Abstract
Mammalian oocytes undergo two rounds of developmental arrest during maturation: at the diplotene of the first meiotic prophase and metaphase of the second meiosis. These arrests are strictly regulated by follicular cells temporally producing the secondary messengers, cAMP and cGMP, and other factors to regulate maturation promoting factor (composed of cyclin B1 and cyclin-dependent kinase 1) levels in the oocytes. Out of these normally appearing developmental arrests, permanent arrests may occur in the oocytes at germinal vesicle (GV), metaphase I (MI), or metaphase II (MII) stage. This issue may arise from absence or altered expression of the oocyte-related genes playing key roles in nuclear and cytoplasmic maturation. Additionally, the assisted reproductive technology (ART) applications such as ovarian stimulation and in vitro culture conditions both of which harbor various types of chemical agents may contribute to forming the permanent arrests. In this review, the molecular determinants of developmental and permanent arrests occurring in the mammalian oocytes are comprehensively evaluated in the light of current knowledge. As number of permanently arrested oocytes at different stages is increasing in ART centers, potential approaches for inducing permanent arrests to obtain competent oocytes are discussed.
Collapse
Affiliation(s)
- Saffet Ozturk
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| |
Collapse
|
9
|
Kinterová V, Kaňka J, Bartková A, Toralová T. SCF Ligases and Their Functions in Oogenesis and Embryogenesis-Summary of the Most Important Findings throughout the Animal Kingdom. Cells 2022; 11:234. [PMID: 35053348 PMCID: PMC8774150 DOI: 10.3390/cells11020234] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 12/10/2022] Open
Abstract
SCF-dependent proteolysis was first discovered via genetic screening of budding yeast almost 25 years ago. In recent years, more and more functions of SCF (Skp1-Cullin 1-F-box) ligases have been described, and we can expect the number of studies on this topic to increase. SCF ligases, which are E3 ubiquitin multi-protein enzymes, catalyse protein ubiquitination and thus allow protein degradation mediated by the 26S proteasome. They play a crucial role in the degradation of cell cycle regulators, regulation of the DNA repair and centrosome cycle and play an important role in several diseases. SCF ligases seem to be needed during all phases of development, from oocyte formation through fertilization, activation of the embryonic genome to embryo implantation. In this review, we summarize known data on SCF ligase-mediated degradation during oogenesis and embryogenesis. In particular, SCFβTrCP and SCFSEL-10/FBXW7 are among the most important and best researched ligases during early development. SCFβTrCP is crucial for the oogenesis of Xenopus and mouse and also in Xenopus and Drosophila embryogenesis. SCFSEL-10/FBXW7 participates in the degradation of several RNA-binding proteins and thereby affects the regulation of gene expression during the meiosis of C. elegans. Nevertheless, a large number of SCF ligases that are primarily involved in embryogenesis remain to be elucidated.
Collapse
Affiliation(s)
- Veronika Kinterová
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, 27721 Libechov, Czech Republic; (J.K.); (A.B.); (T.T.)
| | - Jiří Kaňka
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, 27721 Libechov, Czech Republic; (J.K.); (A.B.); (T.T.)
| | - Alexandra Bartková
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, 27721 Libechov, Czech Republic; (J.K.); (A.B.); (T.T.)
- Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, 949 01 Nitra, Slovakia
| | - Tereza Toralová
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, 27721 Libechov, Czech Republic; (J.K.); (A.B.); (T.T.)
| |
Collapse
|
10
|
Zeng Y, Shi J, Xu S, Shi R, Wu T, Li H, Xue X, Zhu Y, Chen B, Sang Q, Wang L. Bi-allelic mutations in MOS cause female infertility characterized by preimplantation embryonic arrest. Hum Reprod 2022; 37:612-620. [PMID: 34997960 DOI: 10.1093/humrep/deab281] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/18/2021] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION Are mutations in MOS (MOS proto-oncogene, serine/threonine kinase) involved in early embryonic arrest in infertile women? SUMMARY ANSWER We identified mutations in MOS that may cause human female infertility characterized by preimplantation embryonic arrest (PREMBA), and the effects of the mutations in human embryonic kidney 293T (HEK293T cells) and mouse oocytes provided evidence for a causal relation between MOS and female infertility. WHAT IS KNOWN ALREADY MOS, an activator of mitogen-activated protein kinase, mediates germinal vesicle breakdown and metaphase II arrest. Female MOS knockout mice are viable but sterile. Thus, MOS seems to be an important part of the mammalian cell cycle mechanism that regulates female meiosis. STUDY DESIGN, SIZE, DURATION Whole-exome sequencing, bioinformatics filtering analysis and genetic analysis were performed to identify two different biallelic mutations in MOS in two independent families. The infertile patients presenting with early embryonic arrest were recruited from October 2018 to June 2020. PARTICIPANTS/MATERIALS, SETTING, METHODS The female patients diagnosed with primary infertility were recruited from the reproduction centres of local hospitals. Genomic DNA from the affected individuals, their family members and healthy controls was extracted from peripheral blood. We performed whole-exome sequencing in patients diagnosed with PREMBA. Functional effects of the mutations were investigated in HEK293T cells by western blotting and in mouse oocytes by microinjection and immunofluorescence. MAIN RESULTS AND THE ROLE OF CHANCE We identified the homozygous missense mutation c.285C>A (p.(Asn95Lys)) and the compound heterozygous mutations c.467delG (p.(Gly156Alafs*18)) and c.956G>A (p.(Arg319His)) in MOS in two independent patients. The mutations c.285C>A (p.(Asn95Lys)) and c.467delG (p.(Gly156Alafs*18)) reduced the protein level of MOS, and all mutations reduced the ability of MOS to phosphorylate its downstream target, extracellular signal-regulated kinase1/2. In addition, the identified mutations reduced the capacity of exogenous human MOS to rescue the metaphase II exit phenotype, and the F-actin cytoskeleton of mouse oocytes was affected by the patient-derived mutations. LIMITATIONS, REASONS FOR CAUTION Owing to the lack of in vivo data from patient oocytes, the exact molecular mechanism affected by MOS mutations and leading to PREMBA is still unknown and should be further investigated using knock-out or knock-in mice. WIDER IMPLICATIONS OF THE FINDINGS We identified recessive mutations in MOS in two independent patients with the PREMBA phenotype. Our findings reveal the important role of MOS during human oocyte meiosis and embryonic development and suggest that mutations in MOS may be precise diagnostic markers for clinical genetic counselling. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Natural Science Foundation of China (81725006, 81822019, 81771581, 81971450, 81971382,82001538 and 82071642), the project supported by the Shanghai Municipal Science and Technology Major Project (2017SHZDZX01), the Project of the Shanghai Municipal Science and Technology Commission (19JC1411001), the Natural Science Foundation of Shanghai (19ZR1444500 and 21ZR1404800), the Shuguang Program of the Shanghai Education Development Foundation and the Shanghai Municipal Education Commission (18SG03), the Foundation of the Shanghai Health and Family Planning Commission (20154Y0162), the Capacity Building Planning Program for Shanghai Women and Children's Health Service and the collaborative innovation centre project construction for Shanghai Women and Children's Health. The authors have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Yang Zeng
- Institute of Pediatrics, Children's Hospital of Fudan University, the Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Juanzi Shi
- Reproductive Center, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Shiru Xu
- Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
| | - Rong Shi
- Reproductive Center, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Tonghua Wu
- Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
| | - Hongyan Li
- Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
| | - Xia Xue
- Reproductive Center, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Yuanchang Zhu
- Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Fudan University, Shanghai, China
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University, the Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, the Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Wang W, Wang W, Xu Y, Shi J, Fu J, Chen B, Mu J, Zhang Z, Zhao L, Lin J, Du J, Li Q, He L, Jin L, Sun X, Wang L, Sang Q. FBXO43 variants in patients with female infertility characterized by early embryonic arrest. Hum Reprod 2021; 36:2392-2402. [PMID: 34052850 DOI: 10.1093/humrep/deab131] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
STUDY QUESTION Can any new genetic factors responsible for early embryonic arrest in infertile patients be identified, together with the mechanism of pathogenic variants? SUMMARY ANSWER We identified three homozygous variants in the F-box protein 43 gene (FBXO43) in infertile patients and studies on the effects of the variants in HEK293T cells and mouse oocytes provided evidence for a causal relation between FBXO43 and female infertility. WHAT IS KNOWN ALREADY FBXO43, an inhibitor of the anaphase-promoting complex/cyclosome, mediates Metaphase II arrest as a component of the cytostatic factor in oocytes. Both male and female Fbxo43 knockout mice are viable but sterile. FBXO43, therefore, appears to be an essential component of the mammalian cell-cycle machinery that regulates both male and female meiosis. Until now, only one article has reported a homozygous FBXO43 variant associated with teratozoospermia, but the causal relationship was not established with functional evidence. STUDY DESIGN, SIZE, DURATION Whole-exome sequencing (WES) and homozygosity mapping were performed in 24 probands from consanguineous families who suffered from early embryonic arrest, and two different homozygous variants in FBXO43 were identified in two independent families. WES data from a further 950 infertile women with early embryonic arrest were screened for homozygous and compound heterozygous variants in FBXO43, and a third individual with an additional homozygous variant in FBXO43 was identified. The infertile patients presenting with early embryonic arrest were recruited from August 2016 to May 2020. PARTICIPANTS/MATERIALS, SETTING, METHODS The women diagnosed with primary infertility were recruited from the reproduction centers of local hospitals. Genomic DNA samples from the affected individuals, their family members, and healthy controls were extracted from peripheral blood. The FBXO43 variants were identified using WES, homozygosity mapping, in silico analysis, and variant screening. All of the variants were confirmed by Sanger sequencing, and the effects of the variants were investigated in human embryonic kidney (HEK) 293T cells by western blotting and in mouse oocytes by complementary RNA injection. MAIN RESULTS AND THE ROLE OF CHANCE We identified three homozygous variants in FBXO43 (NM_001029860.4)-namely, c.1490_1497dup (p.(Glu500Serfs*2)), c.1747C>T (p.(Gln583*)), and c.154delG (p.(Asp52Thrfs*30))-in three independent families. All of the homozygous variants reduced the protein level of FBXO43 and reduced the level of its downstream target Cyclin B1 in HEK293T cells. In addition, the variants reduced the ability of exogenous human FBXO43 to rescue the parthenogenetic activation phenotype in Fbxo43 knockdown mouse oocytes. LIMITATIONS, REASONS FOR CAUTION Owing to the lack of in vivo data from the oocytes of patients, the exact molecular mechanism remains unknown and should be further investigated using knock out or knock in mice. WIDER IMPLICATIONS OF THE FINDINGS Our study has identified three pathogenic variants in FBXO43 that are involved in human early embryonic arrest. These findings contribute to our understanding of the role of FBXO43 in human early embryonic development and provide a new genetic marker for female infertility. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Key Research and Development Program of China (2018YFC1003800, 2017YFC1001500, and 2016YFC1000600), the National Natural Science Foundation of China (81725006, 81822019, 81771581, 81971450, 81971382, and 82001552), the project supported by the Shanghai Municipal Science and Technology Major Project (2017SHZDZX01), the Project of the Shanghai Municipal Science and Technology Commission (19JC1411001), the Natural Science Foundation of Shanghai (19ZR1444500), the Shuguang Program of the Shanghai Education Development Foundation and the Shanghai Municipal Education Commission (18SG03), the Foundation of the Shanghai Health and Family Planning Commission (20154Y0162), the Capacity Building Planning Program for Shanghai Women and Children's Health Service, and the collaborative innovation center project construction for Shanghai Women and Children's Health. None of the authors have any competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Weijie Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Wenjing Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Yao Xu
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Juanzi Shi
- Reproductive Medicine Center, Shaanxi Maternal and Child Care Service Center, Xi'an, China
| | - Jing Fu
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Jian Mu
- Institute of Pediatrics, Children's Hospital of Fudan University, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Zhihua Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Lin Zhao
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Jing Lin
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jing Du
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Qiaoli Li
- Institute of Pediatrics, Children's Hospital of Fudan University, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Lin He
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaoxi Sun
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Gryaznova Y, Keating L, Touati SA, Cladière D, El Yakoubi W, Buffin E, Wassmann K. Kinetochore individualization in meiosis I is required for centromeric cohesin removal in meiosis II. EMBO J 2021; 40:e106797. [PMID: 33644892 PMCID: PMC8013791 DOI: 10.15252/embj.2020106797] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022] Open
Abstract
Partitioning of the genome in meiosis occurs through two highly specialized cell divisions, named meiosis I and meiosis II. Step-wise cohesin removal is required for chromosome segregation in meiosis I, and sister chromatid segregation in meiosis II. In meiosis I, mono-oriented sister kinetochores appear as fused together when examined by high-resolution confocal microscopy, whereas they are clearly separated in meiosis II, when attachments are bipolar. It has been proposed that bipolar tension applied by the spindle is responsible for the physical separation of sister kinetochores, removal of cohesin protection, and chromatid separation in meiosis II. We show here that this is not the case, and initial separation of sister kinetochores occurs already in anaphase I independently of bipolar spindle forces applied on sister kinetochores, in mouse oocytes. This kinetochore individualization depends on separase cleavage activity. Crucially, without kinetochore individualization in meiosis I, bivalents when present in meiosis II oocytes separate into chromosomes and not sister chromatids. This shows that whether centromeric cohesin is removed or not is determined by the kinetochore structure prior to meiosis II.
Collapse
Affiliation(s)
- Yulia Gryaznova
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Leonor Keating
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Sandra A Touati
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Damien Cladière
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Warif El Yakoubi
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
- Present address:
Cell and Developmental Biology CenterNational Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Eulalie Buffin
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Katja Wassmann
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| |
Collapse
|
13
|
Valencia C, Pérez FA, Matus C, Felmer R, Arias ME. Activation of bovine oocytes by protein synthesis inhibitors: new findings on the role of MPF/MAPKs†. Biol Reprod 2021; 104:1126-1138. [PMID: 33550378 DOI: 10.1093/biolre/ioab019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/16/2021] [Accepted: 02/02/2021] [Indexed: 11/13/2022] Open
Abstract
The present study evaluated the mechanism by which protein synthesis inhibitors activate bovine oocytes. The aim was to analyze the dynamics of MPF and MAPKs. MII oocytes were activated with ionomycin (Io), ionomycin+anisomycin (ANY) and ionomycin+cycloheximide (CHX) and by in vitro fertilization (IVF). The expression of cyclin B1, p-CDK1, p-ERK1/2, p-JNK, and p-P38 were evaluated by immunodetection and the kinase activity of ERK1/2 was measured by enzyme assay. Evaluations at 1, 4, and 15 hours postactivation (hpa) showed that the expression of cyclin B1 was not modified by the treatments. ANY inactivated MPF by p-CDK1Thr14-Tyr15 at 4 hpa (P < 0.05), CHX increased pre-MPF (p-CDK1Thr161 and p-CDK1Thr14-Tyr15) at 1 hpa and IVF increased p-CDK1Thr14-Tyr15 at 17 hours postfertilization (hpf) (P < 0.05). ANY and CHX reduced the levels of p-ERK1/2 at 4 hpa (P < 0.05) and its activity at 4 and 1 hpa, respectively (P < 0.05). Meanwhile, IVF increased p-ERK1/2 at 6 hpf (P < 0.05); however, its kinase activity decreased at 6 hpf (P < 0.05). p-JNK in ANY, CHX, and IVF oocytes decreased at 4 hpa (P < 0.05). p-P38 was only observed at 1 hpa, with no differences between treatments. In conclusion, activation of bovine oocytes by ANY, CHX, and IVF inactivates MPF by CDK1-dependent specific phosphorylation without cyclin B1 degradation. ANY or CHX promoted this inactivation, which seemed to be more delayed in the physiological activation (IVF). Both inhibitors modulated MPF activity via an ERK1/2-independent pathway, whereas IVF activated the bovine oocytes via an ERK1/2-dependent pathway. Finally, ANY does not activate the JNK and P38 kinase pathways.
Collapse
Affiliation(s)
- Cecilia Valencia
- Laboratory of Reproduction, Centre of Reproductive Biotechnology (CEBIOR-BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Felipe Alonso Pérez
- Laboratory of Reproduction, Centre of Reproductive Biotechnology (CEBIOR-BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Carola Matus
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Ricardo Felmer
- Laboratory of Reproduction, Centre of Reproductive Biotechnology (CEBIOR-BIOREN), Universidad de La Frontera, Temuco, Chile.,Department of Agricultural Sciences and Natural Resources, Faculty of Agriculture and Forestry, Universidad de La Frontera, Temuco, Chile
| | - María Elena Arias
- Laboratory of Reproduction, Centre of Reproductive Biotechnology (CEBIOR-BIOREN), Universidad de La Frontera, Temuco, Chile.,Department of Agricultural Production Faculty of Agriculture and Forestry, Universidad de La Frontera, Temuco, Chile
| |
Collapse
|
14
|
Tokmakov AA, Stefanov VE, Sato KI. Dissection of the Ovulatory Process Using ex vivo Approaches. Front Cell Dev Biol 2020; 8:605379. [PMID: 33363163 PMCID: PMC7755606 DOI: 10.3389/fcell.2020.605379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/19/2020] [Indexed: 12/23/2022] Open
Abstract
Ovulation is a unique physiological phenomenon that is essential for sexual reproduction. It refers to the entire process of ovarian follicle responses to hormonal stimulation resulting in the release of mature fertilization-competent oocytes from the follicles and ovaries. Remarkably, ovulation in different species can be reproduced out-of-body with high fidelity. Moreover, most of the molecular mechanisms and signaling pathways engaged in this process have been delineated using in vitro ovulation models. Here, we provide an overview of the major molecular and cytological events of ovulation observed in frogs, primarily in the African clawed frog Xenopus laevis, using mainly ex vivo approaches, with the focus on meiotic oocyte maturation and follicle rupture. For the purpose of comparison and generalization, we also refer extensively to ovulation in other biological species, most notoriously, in mammals.
Collapse
Affiliation(s)
| | - Vasily E Stefanov
- Department of Biochemistry, Saint Petersburg State University, Saint Petersburg, Russia
| | - Ken-Ichi Sato
- Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| |
Collapse
|
15
|
Li B, He X, Zhao Y, Bai D, Du M, Song L, Liu Z, Yin Z, Manglai D. Transcriptome profiling of developing testes and spermatogenesis in the Mongolian horse. BMC Genet 2020; 21:46. [PMID: 32345215 PMCID: PMC7187496 DOI: 10.1186/s12863-020-00843-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/13/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Horse testis development and spermatogenesis are complex physiological processes. METHODS To study these processes, three immature and three mature testes were collected from the Mongolian horse, and six libraries were established using high-throughput RNA sequencing technology (RNA-Seq) to screen for genes related to testis development and spermatogenesis. RESULTS A total of 16,237 upregulated genes and 8,641 downregulated genes were detected in the testis of the Mongolian horse. These genes play important roles in different developmental stages of spermatogenesis and testicular development. Five genes with alternative splicing events that may influence spermatogenesis and development of the testis were detected. GO (Gene ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analyses were performed for functional annotation of the differentially expressed genes. Pathways related to "spermatogenesis," male gamete generation," "spermatid development" and "oocyte meiosis" were significantly involved in different stages of testis development and spermatogenesis. CONCLUSION Genes, pathways and alternative splicing events were identified with inferred functions in the process of spermatogenesis in the Mongolian horse. The identification of these differentially expressed genetic signatures improves our understanding of horse testis development and spermatogenesis.
Collapse
Affiliation(s)
- Bei Li
- College of Animal Science, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
- lnner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Xiaolong He
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, 010031, China
| | - Yiping Zhao
- College of Animal Science, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
- lnner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Dongyi Bai
- College of Animal Science, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
- lnner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Ming Du
- College of Animal Science, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
- lnner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Lianjie Song
- College of Animal Science, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
- lnner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Zhuang Liu
- College of Animal Science, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
- lnner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Zhenchen Yin
- College of Animal Science, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
- lnner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Dugarjaviin Manglai
- College of Animal Science, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China.
- lnner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China.
- Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Equine Research Center, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| |
Collapse
|
16
|
Vadhan A, Wang YY, Yuan SSF, Lee YC, Hu SCS, Huang JY, Ishikawa T, Hou MF. EMI2 expression as a poor prognostic factor in patients with breast cancer. Kaohsiung J Med Sci 2020; 36:640-648. [PMID: 32253818 DOI: 10.1002/kjm2.12208] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 03/01/2020] [Indexed: 01/07/2023] Open
Abstract
Early mitotic inhibitor 2 (EMI2, gene symbol FBXO43), an APC/C inhibitor regulated by Plx1, is essential for cytostatic factor (CSF) activity. It belongs to subclass FBXO of the F-box proteins family. The aim of this study is to examine the clinicopathological significance of EMI2 in breast cancer. In this study, immunohistochemistry analysis was used to evaluate EMI2 expression in breast cancer tissues and then the association between EMI2 expression and clinicopathological factors was examined. Correlation of EMI2 with patient survival was analyzed by Kaplan-Meier survival curves. Among 192 patients analyzed, 105 (54.7%) had high expression of EMI2, and this was significantly associated with shortened disease free survival and overall survival in breast cancer patients. EMI2 expression was significantly associated with tumor grade (P = .006), tumor size (P < .001), and lymph node metastasis (P = .008). However, there was no significant correlation between EMI2 status and other biomarkers including ER, PR and Her2 status. Our results revealed that elevated EMI2 expression is a risk factor (hazard ratio = 3.93) for breast cancer and overexpression of EMI2 in breast cancer predicts higher risk of metastasis and worse survival. Therefore, EMI2 may be a potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Anupama Vadhan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yen-Yun Wang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shyng-Shiou F Yuan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yi-Chen Lee
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Stephen Chu-Sung Hu
- Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Jyun-Yuan Huang
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Takashi Ishikawa
- Department of Breast Oncology and Surgery, Tokyo Medical University, Tokyo, Japan
| | - Ming-Feng Hou
- Division of Breast Surgery, Department of Surgery, Center for Cancer Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
17
|
Wen L, Liu Q, Xu J, Liu X, Shi C, Yang Z, Zhang Y, Xu H, Liu J, Yang H, Huang H, Qiao J, Tang F, Chen ZJ. Recent advances in mammalian reproductive biology. SCIENCE CHINA. LIFE SCIENCES 2020; 63:18-58. [PMID: 31813094 DOI: 10.1007/s11427-019-1572-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/22/2019] [Indexed: 01/05/2023]
Abstract
Reproductive biology is a uniquely important topic since it is about germ cells, which are central for transmitting genetic information from generation to generation. In this review, we discuss recent advances in mammalian germ cell development, including preimplantation development, fetal germ cell development and postnatal development of oocytes and sperm. We also discuss the etiologies of female and male infertility and describe the emerging technologies for studying reproductive biology such as gene editing and single-cell technologies.
Collapse
Affiliation(s)
- Lu Wen
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology Third Hospital, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Qiang Liu
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology Third Hospital, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Jingjing Xu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Xixi Liu
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology Third Hospital, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Chaoyi Shi
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Zuwei Yang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Yili Zhang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Hong Xu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Jiang Liu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Hui Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Hefeng Huang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China.
| | - Jie Qiao
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology Third Hospital, College of Life Sciences, Peking University, Beijing, 100871, China.
| | - Fuchou Tang
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology Third Hospital, College of Life Sciences, Peking University, Beijing, 100871, China.
| | - Zi-Jiang Chen
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, 250021, China.
| |
Collapse
|
18
|
Kouznetsova A, Kitajima TS, Brismar H, Höög C. Post-metaphase correction of aberrant kinetochore-microtubule attachments in mammalian eggs. EMBO Rep 2019; 20:e47905. [PMID: 31290587 PMCID: PMC6680117 DOI: 10.15252/embr.201947905] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/24/2019] [Accepted: 06/07/2019] [Indexed: 12/29/2022] Open
Abstract
The accuracy of the two sequential meiotic divisions in oocytes is essential for creating a haploid gamete with a normal chromosomal content. Here, we have analysed the 3D dynamics of chromosomes during the second meiotic division in live mouse oocytes. We find that chromosomes form stable kinetochore-microtubule attachments at the end of prometaphase II stage that are retained until anaphase II onset. Remarkably, we observe that more than 20% of the kinetochore-microtubule attachments at the metaphase II stage are merotelic or lateral. However, < 1% of all chromosomes at onset of anaphase II are found to lag at the spindle equator and < 10% of the laggards missegregate and give rise to aneuploid gametes. Our results demonstrate that aberrant kinetochore-microtubule attachments are not corrected at the metaphase stage of the second meiotic division. Thus, the accuracy of the chromosome segregation process in mouse oocytes during meiosis II is ensured by an efficient correction process acting at the anaphase stage.
Collapse
Affiliation(s)
- Anna Kouznetsova
- Department of Cell and Molecular BiologyKarolinska InstitutetStockholmSweden
| | - Tomoya S Kitajima
- Laboratory for Chromosome SegregationRIKEN Center for Biosystems Dynamics ResearchKobeJapan
| | - Hjalmar Brismar
- Science for Life LaboratoryDepartment of Applied PhysicsRoyal Institute of TechnologySolnaSweden
| | - Christer Höög
- Department of Cell and Molecular BiologyKarolinska InstitutetStockholmSweden
| |
Collapse
|
19
|
Gibeaux R, Heald R. The Use of Cell-Free Xenopus Extracts to Investigate Cytoplasmic Events. Cold Spring Harb Protoc 2019; 2019:pdb.top097048. [PMID: 29980587 DOI: 10.1101/pdb.top097048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Experiments using cytoplasmic extracts prepared from Xenopus eggs have made important contributions to our understanding of the cell cycle, the cytoskeleton, and cytoplasmic membrane systems. Here we introduce the extract system and describe methods for visualizing and manipulating diverse cytoplasmic processes, and for assaying the functions, dynamics, and stability of individual factors. These in vitro approaches uniquely enable investigation of events at specific cell cycle states, including the assembly of actin- and microtubule-based structures, and the formation of the endoplasmic reticulum. Maternal stockpiles in extracts recapitulate diverse processes in the near absence of gene expression, and this biochemical system combined with microscopy empowers a wide range of mechanistic investigations.
Collapse
Affiliation(s)
- Romain Gibeaux
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720-3200
| | - Rebecca Heald
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720-3200
| |
Collapse
|
20
|
Reconstitution of Intracellular Calcium Signaling in Xenopus Egg Extracts. Methods Mol Biol 2019. [PMID: 30737685 DOI: 10.1007/978-1-4939-9009-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Generation of calcium signal in the cytoplasm of fertilized or parthenogenetically activated eggs has been extensively studied in the intact eggs of several biological species. Calcium transient was found to elicit a plethora of biochemical and cellular events in these cells. Remarkably, intracellular calcium signaling can also be reconstituted in cell-free environment. In this chapter, we describe the methods that allow reconstitution, detection, and quantification of the calcium signal in cell-free extracts of Xenopus oocytes and eggs.
Collapse
|
21
|
Carvacho I, Piesche M, Maier TJ, Machaca K. Ion Channel Function During Oocyte Maturation and Fertilization. Front Cell Dev Biol 2018; 6:63. [PMID: 29998105 PMCID: PMC6028574 DOI: 10.3389/fcell.2018.00063] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 06/04/2018] [Indexed: 12/20/2022] Open
Abstract
The proper maturation of both male and female gametes is essential for supporting fertilization and the early embryonic divisions. In the ovary, immature fully-grown oocytes that are arrested in prophase I of meiosis I are not able to support fertilization. Acquiring fertilization competence requires resumption of meiosis which encompasses the remodeling of multiple signaling pathways and the reorganization of cellular organelles. Collectively, this differentiation endows the egg with the ability to activate at fertilization and to promote the egg-to-embryo transition. Oocyte maturation is associated with changes in the electrical properties of the plasma membrane and alterations in the function and distribution of ion channels. Therefore, variations on the pattern of expression, distribution, and function of ion channels and transporters during oocyte maturation are fundamental to reproductive success. Ion channels and transporters are important in regulating membrane potential, but also in the case of calcium (Ca2+), they play a critical role in modulating intracellular signaling pathways. In the context of fertilization, Ca2+ has been shown to be the universal activator of development at fertilization, playing a central role in early events associated with egg activation and the egg-to-embryo transition. These early events include the block of polyspermy, the completion of meiosis and the transition to the embryonic mitotic divisions. In this review, we discuss the role of ion channels during oocyte maturation, fertilization and early embryonic development. We will describe how ion channel studies in Xenopus oocytes, an extensively studied model of oocyte maturation, translate into a greater understanding of the role of ion channels in mammalian oocyte physiology.
Collapse
Affiliation(s)
- Ingrid Carvacho
- Department of Biology and Chemistry, Faculty of Basic Sciences, Universidad Católica del Maule, Talca, Chile
| | - Matthias Piesche
- Biomedical Research Laboratories, Medicine Faculty, Universidad Católica del Maule, Talca, Chile
| | - Thorsten J. Maier
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe-University Hospital, Frankfurt, Germany
| | - Khaled Machaca
- Department of Physiology and Biophysics, Weill Cornell-Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| |
Collapse
|
22
|
Gopinathan L, Szmyd R, Low D, Diril MK, Chang HY, Coppola V, Liu K, Tessarollo L, Guccione E, van Pelt AMM, Kaldis P. Emi2 Is Essential for Mouse Spermatogenesis. Cell Rep 2018; 20:697-708. [PMID: 28723571 DOI: 10.1016/j.celrep.2017.06.033] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 05/12/2017] [Accepted: 06/12/2017] [Indexed: 12/01/2022] Open
Abstract
The meiotic functions of Emi2, an inhibitor of the APC/C complex, have been best characterized in oocytes where it mediates metaphase II arrest as a component of the cytostatic factor. We generated knockout mice to determine the in vivo functions of Emi2-in particular, its functions in the testis, where Emi2 is expressed at high levels. Male and female Emi2 knockout mice are viable but sterile, indicating that Emi2 is essential for meiosis but dispensable for embryonic development and mitotic cell divisions. We found that, besides regulating cell-cycle arrest in mouse eggs, Emi2 is essential for meiosis I progression in spermatocytes. In the absence of Emi2, spermatocytes arrest in early diplotene of prophase I. This arrest is associated with decreased Cdk1 activity and was partially rescued by a knockin mouse model of elevated Cdk1 activity. Additionally, we detected expression of Emi2 in spermatids and sperm, suggesting potential post-meiotic functions for Emi2.
Collapse
Affiliation(s)
- Lakshmi Gopinathan
- Institute of Molecular and Cell Biology (IMCB), A(∗)STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos #3-09, Singapore 138673, Republic of Singapore
| | - Radoslaw Szmyd
- Institute of Molecular and Cell Biology (IMCB), A(∗)STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos #3-09, Singapore 138673, Republic of Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore (NUS), Singapore 117456, Republic of Singapore
| | - Diana Low
- Institute of Molecular and Cell Biology (IMCB), A(∗)STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos #3-09, Singapore 138673, Republic of Singapore
| | - M Kasim Diril
- Institute of Molecular and Cell Biology (IMCB), A(∗)STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos #3-09, Singapore 138673, Republic of Singapore
| | - Heng-Yu Chang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Vincenzo Coppola
- Mouse Cancer Genetics Program, National Cancer Institute, NCI-Frederick, Building 560, 1050 Boyles Street, Frederick, MD 21702-1201, USA
| | - Kui Liu
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-405 30 Gothenburg, Sweden
| | - Lino Tessarollo
- Mouse Cancer Genetics Program, National Cancer Institute, NCI-Frederick, Building 560, 1050 Boyles Street, Frederick, MD 21702-1201, USA
| | - Ernesto Guccione
- Institute of Molecular and Cell Biology (IMCB), A(∗)STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos #3-09, Singapore 138673, Republic of Singapore; Department of Biochemistry, National University of Singapore (NUS), Singapore 117597, Republic of Singapore
| | - Ans M M van Pelt
- Center for Reproductive Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Philipp Kaldis
- Institute of Molecular and Cell Biology (IMCB), A(∗)STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos #3-09, Singapore 138673, Republic of Singapore; Department of Biochemistry, National University of Singapore (NUS), Singapore 117597, Republic of Singapore.
| |
Collapse
|
23
|
Kalous J, Tetkova A, Kubelka M, Susor A. Importance of ERK1/2 in Regulation of Protein Translation during Oocyte Meiosis. Int J Mol Sci 2018; 19:ijms19030698. [PMID: 29494492 PMCID: PMC5877559 DOI: 10.3390/ijms19030698] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/23/2018] [Accepted: 02/26/2018] [Indexed: 02/07/2023] Open
Abstract
Although the involvement of the extracellular signal-regulated kinases 1 and 2 (ERK1/2) pathway in the regulation of cytostatic factor (CSF) activity; as well as in microtubules organization during meiotic maturation of oocytes; has already been described in detail; rather less attention has been paid to the role of ERK1/2 in the regulation of mRNA translation. However; important data on the role of ERK1/2 in translation during oocyte meiosis have been documented. This review focuses on recent findings regarding the regulation of translation and the role of ERK1/2 in this process in the meiotic cycle of mammalian oocytes. The specific role of ERK1/2 in the regulation of mammalian target of rapamycin (mTOR); eukaryotic translation initiation factor 4E (eIF4E) and cytoplasmic polyadenylation element binding protein 1 (CPEB1) activity is addressed along with additional focus on the other key players involved in protein translation.
Collapse
Affiliation(s)
- Jaroslav Kalous
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Rumburska 89, 27721 Libechov, Czech Republic.
| | - Anna Tetkova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Rumburska 89, 27721 Libechov, Czech Republic.
- Department of Cell Biology, Faculty of Science, Charles University in Prague, Albertov 6, 12843 Prague 2, Czech Republic.
| | - Michal Kubelka
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Rumburska 89, 27721 Libechov, Czech Republic.
| | - Andrej Susor
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Rumburska 89, 27721 Libechov, Czech Republic.
| |
Collapse
|
24
|
Greaney J, Wei Z, Homer H. Regulation of chromosome segregation in oocytes and the cellular basis for female meiotic errors. Hum Reprod Update 2017; 24:135-161. [PMID: 29244163 DOI: 10.1093/humupd/dmx035] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 09/12/2017] [Accepted: 11/26/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Meiotic chromosome segregation in human oocytes is notoriously error-prone, especially with ageing. Such errors markedly reduce the reproductive chances of increasing numbers of women embarking on pregnancy later in life. However, understanding the basis for these errors is hampered by limited access to human oocytes. OBJECTIVE AND RATIONALE Important new discoveries have arisen from molecular analyses of human female recombination and aneuploidy along with high-resolution analyses of human oocyte maturation and mouse models. Here, we review these findings to provide a contemporary picture of the key players choreographing chromosome segregation in mammalian oocytes and the cellular basis for errors. SEARCH METHODS A search of PubMed was conducted using keywords including meiosis, oocytes, recombination, cohesion, cohesin complex, chromosome segregation, kinetochores, spindle, aneuploidy, meiotic cell cycle, spindle assembly checkpoint, anaphase-promoting complex, DNA damage, telomeres, mitochondria, female ageing and female fertility. We extracted papers focusing on mouse and human oocytes that best aligned with the themes of this review and that reported transformative and novel discoveries. OUTCOMES Meiosis incorporates two sequential rounds of chromosome segregation executed by a spindle whose component microtubules bind chromosomes via kinetochores. Cohesion mediated by the cohesin complex holds chromosomes together and should be resolved at the appropriate time, in a specific step-wise manner and in conjunction with meiotically programmed kinetochore behaviour. In women, the stage is set for meiotic error even before birth when female-specific crossover maturation inefficiency leads to the formation of at-risk recombination patterns. In adult life, multiple co-conspiring factors interact with at-risk crossovers to increase the likelihood of mis-segregation. Available evidence support that these factors include, but are not limited to, cohesion deterioration, uncoordinated sister kinetochore behaviour, erroneous microtubule attachments, spindle instability and structural chromosomal defects that impact centromeres and telomeres. Data from mice indicate that cohesin and centromere-specific histones are long-lived proteins in oocytes. Since these proteins are pivotal for chromosome segregation, but lack any obvious renewal pathway, their deterioration with age provides an appealing explanation for at least some of the problems in older oocytes. WIDER IMPLICATIONS Research in the mouse model has identified a number of candidate genes and pathways that are important for chromosome segregation in this species. However, many of these have not yet been investigated in human oocytes so it is uncertain at this stage to what extent they apply to women. The challenge for the future involves applying emerging knowledge of female meiotic molecular regulation towards improving clinical fertility management.
Collapse
Affiliation(s)
- Jessica Greaney
- Christopher Chen Oocyte Biology Research Laboratory, Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane & Women's Hospital Campus, Herston QLD 4029, Australia
| | - Zhe Wei
- Christopher Chen Oocyte Biology Research Laboratory, Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane & Women's Hospital Campus, Herston QLD 4029, Australia
| | - Hayden Homer
- Christopher Chen Oocyte Biology Research Laboratory, Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane & Women's Hospital Campus, Herston QLD 4029, Australia
| |
Collapse
|
25
|
Tokmakov AA, Sato KI, Stefanov VE. Postovulatory cell death: why eggs die via apoptosis in biological species with external fertilization. J Reprod Dev 2017; 64:1-6. [PMID: 29081453 PMCID: PMC5830352 DOI: 10.1262/jrd.2017-100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Spawned unfertilized eggs have been found to die by apoptosis in several species with external fertilization. However, there is no necessity for the externally laid eggs to degrade via this process, as apoptosis evolved as a mechanism to reduce the damaging effects of individual cell death on the whole organism. The recent observation of egg degradation in the genital tracts of some oviparous species provides a clue as to the physiological relevance of egg apoptosis in these animals. We hypothesize that egg apoptosis accompanies ovulation in species with external fertilization as a normal process to eliminate mature eggs retained in the genital tract after ovulation. Furthermore, apoptosis universally develops in ovulated eggs after spontaneous activation in the absence of fertilization. This paper provides an overview of egg apoptosis in several oviparous biological species, including frog, fish, sea urchin, and starfish.
Collapse
Affiliation(s)
| | - Ken-Ichi Sato
- Department of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Vasily E Stefanov
- Department of Biochemistry, St. Petersburg State University, St. Petersburg 199034, Russia
| |
Collapse
|
26
|
Pasternak M, Pfender S, Santhanam B, Schuh M. The BTG4 and CAF1 complex prevents the spontaneous activation of eggs by deadenylating maternal mRNAs. Open Biol 2017; 6:rsob.160184. [PMID: 27605379 PMCID: PMC5043581 DOI: 10.1098/rsob.160184] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/09/2016] [Indexed: 12/11/2022] Open
Abstract
Once every menstrual cycle, eggs are ovulated into the oviduct where they await fertilization. The ovulated eggs are arrested in metaphase of the second meiotic division, and only complete meiosis upon fertilization. It is crucial that the maintenance of metaphase arrest is tightly controlled, because the spontaneous activation of the egg would preclude the development of a viable embryo (Zhang et al. 2015 J. Genet. Genomics 42, 477-485. (doi:10.1016/j.jgg.2015.07.004); Combelles et al. 2011 Hum. Reprod. 26, 545-552. (doi:10.1093/humrep/deq363); Escrich et al. 2011 J. Assist. Reprod. Genet. 28, 111-117. (doi:10.1007/s10815-010-9493-5)). However, the mechanisms that control the meiotic arrest in mammalian eggs are only poorly understood. Here, we report that a complex of BTG4 and CAF1 safeguards metaphase II arrest in mammalian eggs by deadenylating maternal mRNAs. As a follow-up of our recent high content RNAi screen for meiotic genes (Pfender et al. 2015 Nature 524, 239-242. (doi:10.1038/nature14568)), we identified Btg4 as an essential regulator of metaphase II arrest. Btg4-depleted eggs progress into anaphase II spontaneously before fertilization. BTG4 prevents the progression into anaphase by ensuring that the anaphase-promoting complex/cyclosome (APC/C) is completely inhibited during the arrest. The inhibition of the APC/C relies on EMI2 (Tang et al. 2010 Mol. Biol. Cell 21, 2589-2597. (doi:10.1091/mbc.E09-08-0708); Ohe et al. 2010 Mol. Biol. Cell 21, 905-913. (doi:10.1091/mbc.E09-11-0974)), whose expression is perturbed in the absence of BTG4. BTG4 controls protein expression during metaphase II arrest by forming a complex with the CAF1 deadenylase and we hypothesize that this complex is recruited to the mRNA via interactions between BTG4 and poly(A)-binding proteins. The BTG4-CAF1 complex drives the shortening of the poly(A) tails of a large number of transcripts at the MI-MII transition, and this wave of deadenylation is essential for the arrest in metaphase II. These findings establish a BTG4-dependent pathway for controlling poly(A) tail length during meiosis and identify an unexpected role for mRNA deadenylation in preventing the spontaneous activation of eggs.
Collapse
Affiliation(s)
- Michał Pasternak
- Medical Research Council, Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Sybille Pfender
- Medical Research Council, Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Balaji Santhanam
- Medical Research Council, Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Melina Schuh
- Medical Research Council, Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| |
Collapse
|
27
|
Guo A, Huang S, Yu J, Wang H, Li H, Pei G, Shen L. Single-Cell Dynamic Analysis of Mitosis in Haploid Embryonic Stem Cells Shows the Prolonged Metaphase and Its Association with Self-diploidization. Stem Cell Reports 2017; 8:1124-1134. [PMID: 28457886 PMCID: PMC5425685 DOI: 10.1016/j.stemcr.2017.03.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 03/30/2017] [Accepted: 03/31/2017] [Indexed: 12/30/2022] Open
Abstract
The recent establishment of mammalian haploid embryonic stem cells (ESCs) provides new possibilities for genetic screening and for understanding genome evolution and function. However, the dynamics of mitosis in haploid ESCs is still unclear. Here, we report that the duration of mitosis in haploid ESCs, especially the metaphase, is significantly longer than that in diploid ESCs. Delaying mitosis by chemicals increased self-diploidization of haploid ESCs, while shortening mitosis stabilized haploid ESCs. Taken together, our study suggests that the delayed mitosis of haploid ESCs is associated with self-diploidization. Mitosis is prolonged in haploid ESCs, especially in metaphase Self-diploidization might be associated with the prolonged mitosis of haploid ESCs
Collapse
Affiliation(s)
- Ao Guo
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shichao Huang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jiali Yu
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Huihan Wang
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Haisen Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Gang Pei
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Li Shen
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
28
|
Levitan S, Sher N, Brekhman V, Ziv T, Lubzens E, Lotan T. The making of an embryo in a basal metazoan: Proteomic analysis in the sea anemoneNematostella vectensis. Proteomics 2015; 15:4096-104. [DOI: 10.1002/pmic.201500255] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 07/25/2015] [Accepted: 09/09/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Shimrit Levitan
- Marine Biology Department, The Leon H. Charney School of Marine Sciences; University of Haifa; Haifa Israel
| | - Noa Sher
- Bioinformatics Service Unit; University of Haifa; Haifa Israel
| | - Vera Brekhman
- Marine Biology Department, The Leon H. Charney School of Marine Sciences; University of Haifa; Haifa Israel
| | - Tamar Ziv
- Faculty of Biology; Technion - Israel Institute of Technology; Haifa Israel
| | - Esther Lubzens
- Faculty of Biology; Technion - Israel Institute of Technology; Haifa Israel
| | - Tamar Lotan
- Marine Biology Department, The Leon H. Charney School of Marine Sciences; University of Haifa; Haifa Israel
| |
Collapse
|
29
|
Herbert M, Kalleas D, Cooney D, Lamb M, Lister L. Meiosis and maternal aging: insights from aneuploid oocytes and trisomy births. Cold Spring Harb Perspect Biol 2015; 7:a017970. [PMID: 25833844 DOI: 10.1101/cshperspect.a017970] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In most organisms, genome haploidization requires reciprocal DNA exchanges (crossovers) between replicated parental homologs to form bivalent chromosomes. These are resolved to their four constituent chromatids during two meiotic divisions. In female mammals, bivalents are formed during fetal life and remain intact until shortly before ovulation. Extending this period beyond ∼35 years greatly increases the risk of aneuploidy in human oocytes, resulting in a dramatic increase in infertility, miscarriage, and birth defects, most notably trisomy 21. Bivalent chromosomes are stabilized by cohesion between sister chromatids, which is mediated by the cohesin complex. In mouse oocytes, cohesin becomes depleted from chromosomes during female aging. Consistent with this, premature loss of centromeric cohesion is a major source of aneuploidy in oocytes from older women. Here, we propose a mechanistic framework to reconcile data from genetic studies on human trisomy and oocytes with recent advances in our understanding of the molecular mechanisms of chromosome segregation during meiosis in model organisms.
Collapse
Affiliation(s)
- Mary Herbert
- Newcastle Fertility Centre, Centre for Life, Times Square, Newcastle upon Tyne NE1 4EP, United Kingdom Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Dimitrios Kalleas
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Daniel Cooney
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Mahdi Lamb
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Lisa Lister
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom
| |
Collapse
|
30
|
Zhang T, Zhou Y, Qi ST, Wang ZB, Qian WP, Ouyang YC, Shen W, Schatten H, Sun QY. Nuf2 is required for chromosome segregation during mouse oocyte meiotic maturation. Cell Cycle 2015; 14:2701-10. [PMID: 26054848 PMCID: PMC4613995 DOI: 10.1080/15384101.2015.1058677] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/07/2015] [Accepted: 05/16/2015] [Indexed: 10/23/2022] Open
Abstract
Nuf2 plays an important role in kinetochore-microtubule attachment and thus is involved in regulation of the spindle assembly checkpoint in mitosis. In this study, we examined the localization and function of Nuf2 during mouse oocyte meiotic maturation. Myc6-Nuf2 mRNA injection and immunofluorescent staining showed that Nuf2 localized to kinetochores from germinal vesicle breakdown to metaphase I stages, while it disappeared from the kinetochores at the anaphase I stage, but relocated to kinetochores at the MII stage. Overexpression of Nuf2 caused defective spindles, misaligned chromosomes, and activated spindle assembly checkpoint, and thus inhibited chromosome segregation and metaphase-anaphase transition in oocyte meiosis. Conversely, precocious polar body extrusion was observed in the presence of misaligned chromosomes and abnormal spindle formation in Nuf2 knock-down oocytes, causing aneuploidy. Our data suggest that Nuf2 is a critical regulator of meiotic cell cycle progression in mammalian oocytes.
Collapse
Affiliation(s)
- Teng Zhang
- Institute of Reproductive Sciences; College of Animal Science and Technology; Qingdao Agricultural University; Qingdao, China
- State Key Laboratory of Reproductive Biology; Institute of Zoology; Chinese Academy of Sciences; Beijing, China
| | - Yang Zhou
- State Key Laboratory of Reproductive Biology; Institute of Zoology; Chinese Academy of Sciences; Beijing, China
| | - Shu-Tao Qi
- State Key Laboratory of Reproductive Biology; Institute of Zoology; Chinese Academy of Sciences; Beijing, China
| | - Zhen-Bo Wang
- State Key Laboratory of Reproductive Biology; Institute of Zoology; Chinese Academy of Sciences; Beijing, China
| | - Wei-Ping Qian
- Department of Reproductive Medicine; Peking University Shenzhen Hospital; Medical Center of Peking University; Shenzhen, Guangdong, China
| | - Ying-Chun Ouyang
- State Key Laboratory of Reproductive Biology; Institute of Zoology; Chinese Academy of Sciences; Beijing, China
| | - Wei Shen
- Institute of Reproductive Sciences; College of Animal Science and Technology; Qingdao Agricultural University; Qingdao, China
| | - Heide Schatten
- Department of Veterinary Pathobiology; University of Missouri; Columbia, MO USA
| | - Qing-Yuan Sun
- Institute of Reproductive Sciences; College of Animal Science and Technology; Qingdao Agricultural University; Qingdao, China
- State Key Laboratory of Reproductive Biology; Institute of Zoology; Chinese Academy of Sciences; Beijing, China
| |
Collapse
|
31
|
Sato KI. Transmembrane signal transduction in oocyte maturation and fertilization: focusing on Xenopus laevis as a model animal. Int J Mol Sci 2014; 16:114-34. [PMID: 25546390 PMCID: PMC4307238 DOI: 10.3390/ijms16010114] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 12/15/2014] [Indexed: 11/16/2022] Open
Abstract
Fertilization is a cell biological phenomenon of crucial importance for the birth of new life in a variety of multicellular and sexual reproduction species such as algae, animal and plants. Fertilization involves a sequence of events, in which the female gamete "egg" and the male gamete "spermatozoon (sperm)" develop, acquire their functions, meet and fuse with each other, to initiate embryonic and zygotic development. Here, it will be briefly reviewed how oocyte cytoplasmic components are orchestrated to undergo hormone-induced oocyte maturation and sperm-induced activation of development. I then review how sperm-egg membrane interaction/fusion and activation of development in the fertilized egg are accomplished and regulated through egg coat- or egg plasma membrane-associated components, highlighting recent findings and future directions in the studies using Xenopus laevis as a model experimental animal.
Collapse
Affiliation(s)
- Ken-ichi Sato
- Laboratory of Cell Signaling and Development, Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kamigamo-motoyama, Kita-ku, Kyoto 603-8555, Japan.
| |
Collapse
|
32
|
Calcium signaling and meiotic exit at fertilization in Xenopus egg. Int J Mol Sci 2014; 15:18659-76. [PMID: 25322156 PMCID: PMC4227238 DOI: 10.3390/ijms151018659] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/01/2014] [Accepted: 10/09/2014] [Indexed: 11/16/2022] Open
Abstract
Calcium is a universal messenger that mediates egg activation at fertilization in all sexually reproducing species studied. However, signaling pathways leading to calcium generation and the mechanisms of calcium-induced exit from meiotic arrest vary substantially among species. Here, we review the pathways of calcium signaling and the mechanisms of meiotic exit at fertilization in the eggs of the established developmental model, African clawed frog, Xenopus laevis. We also discuss calcium involvement in the early fertilization-induced events in Xenopus egg, such as membrane depolarization, the increase in intracellular pH, cortical granule exocytosis, cortical contraction, contraction wave, cortical rotation, reformation of the nuclear envelope, sperm chromatin decondensation and sister chromatid segregation.
Collapse
|
33
|
The zinc-binding region (ZBR) fragment of Emi2 can inhibit APC/C by targeting its association with the coactivator Cdc20 and UBE2C-mediated ubiquitylation. FEBS Open Bio 2014; 4:689-703. [PMID: 25161877 PMCID: PMC4141206 DOI: 10.1016/j.fob.2014.06.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 06/16/2014] [Accepted: 06/30/2014] [Indexed: 01/21/2023] Open
Abstract
Overexpression of the ZBR fragment of Emi2, but not of Emi1, induces abnormal cell division. The Emi2 ZBR fragment impairs the association of the coactivator Cdc20 with APC/C. The Emi2 ZBR fragment inhibits ubiquitylation by the cullin-RING of APC/C and E2C. The Emi2 ZBR-specific residues for APC/C inhibitory activity have been identified.
Anaphase-promoting complex or cyclosome (APC/C) is a multisubunit ubiquitin ligase E3 that targets cell-cycle regulators. Cdc20 is required for full activation of APC/C in M phase, and mediates substrate recognition. In vertebrates, Emi2/Erp1/FBXO43 inhibits APC/C-Cdc20, and functions as a cytostatic factor that causes long-term M phase arrest of mature oocytes. In this study, we found that a fragment corresponding to the zinc-binding region (ZBR) domain of Emi2 inhibits cell-cycle progression, and impairs the association of Cdc20 with the APC/C core complex in HEK293T cells. Furthermore, we revealed that the ZBR fragment of Emi2 inhibits in vitro ubiquitin chain elongation catalyzed by the APC/C cullin-RING ligase module, the ANAPC2–ANAPC11 subcomplex, in combination with the ubiquitin chain-initiating E2, E2C/UBE2C/UbcH10. Structural analyses revealed that the Emi2 ZBR domain uses different faces for the two mechanisms. Thus, the double-faced ZBR domain of Emi2 antagonizes the APC/C function by inhibiting both the binding with the coactivator Cdc20 and ubiquitylation mediated by the cullin-RING ligase module and E2C. In addition, the tail region between the ZBR domain and the C-terminal RL residues [the post-ZBR (PZ) region] interacts with the cullin subunit, ANAPC2. In the case of the ZBR fragment of the somatic paralogue of Emi2, Emi1/FBXO5, these inhibitory activities against cell division and ubiquitylation were not observed. Finally, we identified two sets of key residues in the Emi2 ZBR domain that selectively exert each of the dual Emi2-specific modes of APC/C inhibition, by their mutation in the Emi2 ZBR domain and their transplantation into the Emi1 ZBR domain.
Collapse
|
34
|
Emi2 mediates meiotic MII arrest by competitively inhibiting the binding of Ube2S to the APC/C. Nat Commun 2014; 5:3667. [PMID: 24770399 DOI: 10.1038/ncomms4667] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 03/17/2014] [Indexed: 11/08/2022] Open
Abstract
In vertebrates, unfertilized eggs are arrested at metaphase of meiosis II by Emi2, a direct inhibitor of the APC/C ubiquitin ligase. Two different ubiquitin-conjugating enzymes, UbcH10 and Ube2S, work with the APC/C to target APC/C substrates for degradation. However, their possible roles and regulations in unfertilized/fertilized eggs are not known. Here we use Xenopus egg extracts to show that both UbcH10 and Ube2S are required for rapid cyclin B degradation at fertilization, when APC/C binding of Ube2S, but not of UbcH10, increases several fold, coincidently with (SCF(β-TrCP)-dependent) Emi2 degradation. Interestingly, before fertilization, Emi2 directly inhibits APC/C-Ube2S binding via the C-terminal tail, but on fertilization, its degradation allows the binding mediated by the Ube2S C-terminal tail. Significantly, Emi2 and Ube2S bind commonly to the APC/C catalytic subunit APC10 via their similar C-terminal tails. Thus, Emi2 competitively inhibits APC/C-Ube2S binding before fertilization, while its degradation on fertilization relieves the inhibition for APC/C activation.
Collapse
|
35
|
Duan RB, Zhang L, Chen DF, Yang F, Yang JS, Yang WJ. Two p90 ribosomal S6 kinase isoforms are involved in the regulation of mitotic and meiotic arrest in Artemia. J Biol Chem 2014; 289:16006-15. [PMID: 24755224 DOI: 10.1074/jbc.m114.553370] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
There are multiple isoforms of p90 ribosomal S6 kinase (RSK), which regulate diverse cellular functions such as cell growth, proliferation, maturation, and motility. However, the relationship between the structures and functions of RSK isoforms remains undetermined. Artemia is a useful model in which to study cell cycle arrest because these animals undergo prolonged diapauses, a state of obligate dormancy. A novel RSK isoform was identified in Artemia, which was termed Ar-Rsk2. This isoform was compared with an RSK isoform that we previously identified in Artemia, termed Ar-Rsk1. Ar-Rsk2 has an ERK-docking motif, whereas Ar-Rsk1 does not. Western blot analysis revealed that Ar-Rsk1 was activated by phosphorylation, which blocked meiosis in oocytes. Knockdown of Ar-Rsk1 reduced the level of phosphorylated cdc2 and thereby suppressed cytostatic factor activity. This indicates that Ar-Rsk1 regulates the cytostatic factor in meiosis. Expression of Ar-Rsk2 was down-regulated in Artemia cysts in which mitosis was arrested. Knockdown of Ar-Rsk2 resulted in decreased levels of cyclin D3 and phosphorylated histone H3, and the production of pseudo-diapause cysts. This indicates that Ar-Rsk2 regulates mitotic arrest. PLK and ERK RNAi showed that Ar-Rsk2, but not Ar-Rsk1, could be activated by PLK-ERK in Artemia. This is the first study to report that RSK isoforms with and without an ERK-docking motif regulate mitosis and meiosis, respectively. This study provides insight into the relationship between the structures and functions of RSK isoforms.
Collapse
Affiliation(s)
- Ru-Bing Duan
- From the Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education and College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Li Zhang
- From the Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education and College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Dian-Fu Chen
- From the Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education and College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Fan Yang
- From the Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education and College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jin-Shu Yang
- From the Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education and College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Wei-Jun Yang
- From the Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education and College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
36
|
Medvedev S, Stein P, Schultz RM. Specificity of calcium/calmodulin-dependent protein kinases in mouse egg activation. Cell Cycle 2014; 13:1482-8. [PMID: 24626179 DOI: 10.4161/cc.28432] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
CaMKIIγ, the predominant CaMKII isoform in mouse eggs, controls egg activation by regulating cell cycle resumption. In this study we further characterize the involvement and specificity of CaMKIIγ in mouse egg activation. Using exogenous expression of different cRNAs in Camk2g(-/-) eggs, we show that the other multifunctional CaM kinases, CaMKI, and CaMKIV, are not capable of substituting CaMKIIγ to initiate cell cycle resumption in response to a rise in intracellular Ca (2+). Exogenous expression of Camk2g or Camk2d results in activation of nearly 80% of Camk2g(-/-) MII eggs after stimulation with SrCl 2, which does not differ from the incidence of activation of wild-type eggs expressing exogenous Egfp. In contrast, none of the Camk2g(-/-) MII eggs expressing Camk1 or Camk4 activate in response to SrCl 2 treatment. Expression of a constitutively active form of Camk4 (ca-Camk4), but not Camk1, triggers egg activation. EMI2, an APC/C repressor, is a key component in regulating egg activation downstream of CaMKII in both Xenopus laevis and mouse. We show that exogenous expression of either Camk2g, Camk2d, or ca-Camk4, but not Camk1, Camk4, or a catalytically inactive mutant form of CaMKIIγ (kinase-dead) in Camk2g(-/-) mouse eggs leads to almost complete degradation (~90%) of exogenously expressed EMI2 followed by cell cycle resumption. Thus, degradation of EMI2 following its phosphorylation specifically by CaMKII is mechanistically linked to and promotes cell cycle resumption in MII eggs.
Collapse
Affiliation(s)
- Sergey Medvedev
- Department of Biology; University of Pennsylvania; Philadelphia, PA USA
| | - Paula Stein
- Department of Biology; University of Pennsylvania; Philadelphia, PA USA
| | - Richard M Schultz
- Department of Biology; University of Pennsylvania; Philadelphia, PA USA
| |
Collapse
|
37
|
Singh SA, Winter D, Kirchner M, Chauhan R, Ahmed S, Ozlu N, Tzur A, Steen JA, Steen H. Co-regulation proteomics reveals substrates and mechanisms of APC/C-dependent degradation. EMBO J 2014; 33:385-99. [PMID: 24510915 DOI: 10.1002/embj.201385876] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Using multiplexed quantitative proteomics, we analyzed cell cycle-dependent changes of the human proteome. We identified >4,400 proteins, each with a six-point abundance profile across the cell cycle. Hypothesizing that proteins with similar abundance profiles are co-regulated, we clustered the proteins with abundance profiles most similar to known Anaphase-Promoting Complex/Cyclosome (APC/C) substrates to identify additional putative APC/C substrates. This protein profile similarity screening (PPSS) analysis resulted in a shortlist enriched in kinases and kinesins. Biochemical studies on the kinesins confirmed KIFC1, KIF18A, KIF2C, and KIF4A as APC/C substrates. Furthermore, we showed that the APC/C(CDH1)-dependent degradation of KIFC1 regulates the bipolar spindle formation and proper cell division. A targeted quantitative proteomics experiment showed that KIFC1 degradation is modulated by a stabilizing CDK1-dependent phosphorylation site within the degradation motif of KIFC1. The regulation of KIFC1 (de-)phosphorylation and degradation provides insights into the fidelity and proper ordering of substrate degradation by the APC/C during mitosis.
Collapse
Affiliation(s)
- Sasha A Singh
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Clift D, Schuh M. Restarting life: fertilization and the transition from meiosis to mitosis. Nat Rev Mol Cell Biol 2013; 14:549-62. [PMID: 23942453 PMCID: PMC4021448 DOI: 10.1038/nrm3643] [Citation(s) in RCA: 194] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Fertilization triggers a complex cellular programme that transforms two highly specialized meiotic germ cells, the oocyte and the sperm, into a totipotent mitotic embryo. Linkages between sister chromatids are remodelled to support the switch from reductional meiotic to equational mitotic divisions; the centrosome, which is absent from the egg, is reintroduced; cell division shifts from being extremely asymmetric to symmetric; genomic imprinting is selectively erased and re-established; and protein expression shifts from translational control to transcriptional control. Recent work has started to reveal how this remarkable transition from meiosis to mitosis is achieved.
Collapse
Affiliation(s)
- Dean Clift
- Medical Research Council Laboratory of Molecular Biology (MRC LMB), Cambridge CB2 0QH, UK
| | | |
Collapse
|
39
|
Wassmann K. Sister chromatid segregation in meiosis II: deprotection through phosphorylation. Cell Cycle 2013; 12:1352-9. [PMID: 23574717 DOI: 10.4161/cc.24600] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Meiotic divisions (meiosis I and II) are specialized cell divisions to generate haploid gametes. The first meiotic division with the separation of chromosomes is named reductional division. The second division, which takes place immediately after meiosis I without intervening S-phase, is equational, with the separation of sister chromatids, similar to mitosis. This meiotic segregation pattern requires the two-step removal of the cohesin complex holding sister chromatids together: cohesin is removed from chromosome arms that have been subjected to homologous recombination in meiosis I and from the centromere region in meiosis II. Cohesin in the centromere region is protected from removal in meiosis I, but this protection has to be removed--deprotected--for sister chromatid segregation in meiosis II. Whereas the mechanisms of cohesin protection are quite well understood, the mechanisms of deprotection have been largely unknown until recently. In this review I summarize our current knowledge on cohesin deprotection.
Collapse
|
40
|
Bernhardt ML, Kong BY, Kim AM, O'Halloran TV, Woodruff TK. A zinc-dependent mechanism regulates meiotic progression in mammalian oocytes. Biol Reprod 2012; 86:114. [PMID: 22302686 DOI: 10.1095/biolreprod.111.097253] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Precise coordination of meiotic progression is a critical determinant of an egg's capacity to be fertilized successfully, and zinc has emerged as a key regulatory element in this process. An early manifestation of a regulatory role for this transition metal is the significant increase in total intracellular zinc. This accumulation is essential for meiotic progression beyond telophase I and the establishment of meiotic arrest at metaphase II. The subsequent developmental event, fertilization, induces a rapid expulsion of labile zinc that is a hallmark event in meiotic resumption. In the present study, we show that the zinc fluxes work, in part, by altering the activity of the cytostatic factor (CSF), the cellular activity required for the establishment and maintenance of metaphase II arrest in the mature, unfertilized egg. We propose a model in which zinc exerts concentration-dependent regulation of meiosis through the CSF component EMI2, a zinc-binding protein. Together, the data support the conclusion that zinc itself, through its interaction with EMI2, is a central component of the CSF.
Collapse
Affiliation(s)
- Miranda L Bernhardt
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | | | |
Collapse
|
41
|
Kronja I, Orr-Weaver TL. Translational regulation of the cell cycle: when, where, how and why? Philos Trans R Soc Lond B Biol Sci 2012; 366:3638-52. [PMID: 22084390 DOI: 10.1098/rstb.2011.0084] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Translational regulation contributes to the control of archetypal and specialized cell cycles, such as the meiotic and early embryonic cycles. Late meiosis and early embryogenesis unfold in the absence of transcription, so they particularly rely on translational repression and activation of stored maternal mRNAs. Here, we present examples of cell cycle regulators that are translationally controlled during different cell cycle and developmental transitions in model organisms ranging from yeast to mouse. Our focus also is on the RNA-binding proteins that affect cell cycle progression by recognizing special features in untranslated regions of mRNAs. Recent research highlights the significance of the cytoplasmic polyadenylation element-binding protein (CPEB). CPEB determines polyadenylation status, and consequently translational efficiency, of its target mRNAs in both transcriptionally active somatic cells as well as in transcriptionally silent mature Xenopus oocytes and early embryos. We discuss the role of CPEB in mediating the translational timing and in some cases spindle-localized translation of critical regulators of Xenopus oogenesis and early embryogenesis. We conclude by outlining potential directions and approaches that may provide further insights into the translational control of the cell cycle.
Collapse
Affiliation(s)
- Iva Kronja
- Whitehead Institute and Department of Biology, Massachusetts Institute of Technology, Nine Cambridge Center, Cambridge, MA 02142, USA
| | | |
Collapse
|
42
|
Chebotareva T, Taylor J, Mullins JJ, Wilmut I. Rat eggs cannot wait: Spontaneous exit from meiotic metaphase-II arrest. Mol Reprod Dev 2011; 78:795-807. [PMID: 21910153 DOI: 10.1002/mrd.21385] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 08/15/2011] [Indexed: 11/06/2022]
Abstract
Mammalian eggs await fertilisation while arrested at the second metaphase stage of meiotic division. A network of signalling pathways enables the establishment and maintenance of this metaphase-II arrest. In the absence of fertilisation, mammalian eggs can spontaneously exit metaphase II when parthenogenetically stimulated, or sometimes without any obvious stimulation. Ovulated rat eggs abortively release from metaphase-II arrest once removed from egg donors. Spontaneously activated rat eggs extrude the second polar body and proceed to the so-called metaphase III-'like' stage, with clumps of condensed chromatin scattered in the egg cytoplasm. It is still unclear what makes rat eggs susceptible to spontaneous activation; however, a vague picture of the signalling pathways involved in the process of spontaneous activation is beginning to emerge. Such cell cycle instability is one of the major reasons why it is more difficult to establish nuclear transfer in the rat. This review examines the known predisposing factors and biochemical mechanisms involved in spontaneous activation. The strategies used to prevent spontaneous metaphase-II release in rat eggs will also be discussed.
Collapse
Affiliation(s)
- Tatiana Chebotareva
- MRC Centre for Regenerative Medicine, Edinburgh University, Edinburgh, Scotland, UK.
| | | | | | | |
Collapse
|
43
|
Gotoh T, Villa LM, Capelluto DGS, Finkielstein CV. Regulatory pathways coordinating cell cycle progression in early Xenopus development. Results Probl Cell Differ 2011; 53:171-99. [PMID: 21630146 DOI: 10.1007/978-3-642-19065-0_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The African clawed frog, Xenopus laevis, is used extensively as a model organism for studying both cell development and cell cycle regulation. For over 20 years now, this model organism has contributed to answering fundamental questions concerning the mechanisms that underlie cell cycle transitions--the cellular components that synthesize, modify, repair, and degrade nucleic acids and proteins, the signaling pathways that allow cells to communicate, and the regulatory pathways that lead to selective expression of subsets of genes. In addition, the remarkable simplicity of the Xenopus early cell cycle allows for tractable manipulation and dissection of the basic components driving each transition. In this organism, early cell divisions are characterized by rapid cycles alternating phases of DNA synthesis and division. The post-blastula stages incorporate gap phases, lengthening progression, and allowing more time for DNA repair. Various cyclin/Cdk complexes are differentially expressed during the early cycles with orderly progression being driven by both the combined action of cyclin synthesis and degradation and the appropriate selection of specific substrates by their Cdk components. Like other multicellular organisms, chief developmental events in early Xenopus embryogenesis coincide with profound remodeling of the cell cycle, suggesting that cell proliferation and differentiation events are linked and coordinated through crosstalk mechanisms acting on signaling pathways involving the expression of cell cycle control genes.
Collapse
Affiliation(s)
- Tetsuya Gotoh
- Integrated Cellular Responses Laboratory, Department of Biological Sciences, Virginia Polytechnic Institute and State University, 1981 Kraft Drive, Blacksburg, VA 24061, USA
| | | | | | | |
Collapse
|
44
|
Abstract
Entry into, and passage through, the two meiotic divisions of the oocyte has to be highly coordinated to ensure proper segregation of chromosomes. This coordination ensures that the hallmark stops and starts of the meiotic process occur at the right time to prevent aneuploidy. The Anaphase-Promoting Complex is an activity mostly studied in the mitotic cell cycle division, where it has essential functions during mitosis. As detailed here the Anaphase-Promoting Complex also plays vital roles in controlling at least three meiotic events: maintenance of prophase I arrest, timely and faithful segregation of homologous chromosomes in meiosis I, and the meiotic arrest following ovulation.
Collapse
Affiliation(s)
- Keith T Jones
- University of Newcastle, 2308 Newcastle, NSW, Australia.
| |
Collapse
|
45
|
Kim KH, Kim EY, Kim Y, Kim E, Lee HS, Yoon SY, Lee KA. Gas6 downregulation impaired cytoplasmic maturation and pronuclear formation independent to the MPF activity. PLoS One 2011; 6:e23304. [PMID: 21850267 PMCID: PMC3151302 DOI: 10.1371/journal.pone.0023304] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Accepted: 07/14/2011] [Indexed: 11/18/2022] Open
Abstract
Previously, we found that the growth arrest-specific gene 6 (Gas6) is more highly expressed in germinal vesicle (GV) oocytes than in metaphase II (MII) oocytes using annealing control primer (ACP)-PCR technology. The current study was undertaken to investigate the role of Gas6 in oocyte maturation and fertilization using RNA interference (RNAi). Interestingly, despite the specific and marked decrease in Gas6 mRNA and protein expression in GVs after Gas6 RNAi, nuclear maturation including spindle structures and chromosome segregation was not affected. The only discernible effect induced by Gas6 RNAi was a change in maturation promoting factor (MPF) activity. After parthenogenetic activation, Gas6 RNAi-treated oocytes at the MII stage had not developed further and arrested at MII (90.0%). After stimulation with Sr(2+), Gas6-silenced MII oocytes had markedly reduced Ca(2+) oscillation and exhibited no exocytosis of cortical granules. In these oocytes, sperm penetration occurred during fertilization but not pronucleus (PN) formation. By roscovitine and colcemid treatment, we found that the Gas6 knockdown affected cytoplasmic maturation directly, independent to the changed MPF activity. These results strongly suggest that 1) the Gas6 signaling itself is important to the cytoplasmic maturation, but not nuclear maturation, and 2) the decreased Gas6 expression and decreased MPF activity separately or mutually influence sperm head decondensation and PN formation.
Collapse
Affiliation(s)
- Kyeoung-Hwa Kim
- Department of Biomedical Science, College of Life Science, Fertility Center, CHA Research Institute, CHA University, CHA General Hospital, Seoul, Korea
| | - Eun-Young Kim
- Department of Biomedical Science, College of Life Science, Fertility Center, CHA Research Institute, CHA University, CHA General Hospital, Seoul, Korea
| | - Yuna Kim
- Department of Biomedical Science, College of Life Science, Fertility Center, CHA Research Institute, CHA University, CHA General Hospital, Seoul, Korea
| | - Eunju Kim
- Department of Biomedical Science, College of Life Science, Fertility Center, CHA Research Institute, CHA University, CHA General Hospital, Seoul, Korea
| | - Hyun-Seo Lee
- Department of Biomedical Science, College of Life Science, Fertility Center, CHA Research Institute, CHA University, CHA General Hospital, Seoul, Korea
| | - Sook-Young Yoon
- Department of Biomedical Science, College of Life Science, Fertility Center, CHA Research Institute, CHA University, CHA General Hospital, Seoul, Korea
| | - Kyung-Ah Lee
- Department of Biomedical Science, College of Life Science, Fertility Center, CHA Research Institute, CHA University, CHA General Hospital, Seoul, Korea
- * E-mail:
| |
Collapse
|
46
|
McIsaac RS, Huang KC, Sengupta A, Wingreen NS. Does the potential for chaos constrain the embryonic cell-cycle oscillator? PLoS Comput Biol 2011; 7:e1002109. [PMID: 21779158 PMCID: PMC3136431 DOI: 10.1371/journal.pcbi.1002109] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2011] [Accepted: 05/14/2011] [Indexed: 11/24/2022] Open
Abstract
Although many of the core components of the embryonic cell-cycle network have been elucidated, the question of how embryos achieve robust, synchronous cellular divisions post-fertilization remains unexplored. What are the different schemes that could be implemented by the embryo to achieve synchronization? By extending a cell-cycle model previously developed for embryos of the frog Xenopus laevis to include the spatial dimensions of the embryo, we establish a novel role for the rapid, fertilization-initiated calcium wave that triggers cell-cycle oscillations. Specifically, in our simulations a fast calcium wave results in synchronized cell cycles, while a slow wave results in full-blown spatio-temporal chaos. We show that such chaos would ultimately lead to an unpredictable patchwork of cell divisions across the embryo. Given this potential for chaos, our results indicate a novel design principle whereby the fast calcium-wave trigger following embryo fertilization synchronizes cell divisions. Cell divisions across an embryo occur in rapid synchrony - like clockwork - starting within minutes of fertilization. How does an embryo achieve this remarkable uniformity? Simple diffusion is too slow: typical proteins diffuse with a rate of 10 µm2/s, requiring nearly 14 hours to traverse a 1 mm embryo. An exciting idea is that the embryo is an active medium, much like the heart where pulses of electrical activity result in organized contractions. However, just as the heart can have arrhythmias, our model predicts that oscillations in the embryo can become chaotic. What would be the biological consequences of this behavior? How do embryos avoid chaos? Our work provides potential answers to these questions: Chaos would lead to an unpredictable patchwork of cell divisions across the embryo - clearly a fatal defect in development. To avoid chaos then, we predict that cell-cycle oscillations need to be triggered throughout the embryo at almost precisely the same time. The threat that chaos will mar development therefore explains the mystery of why embryos universally employ a fast calcium wave to trigger cell-cycle oscillations. In this way, developing organisms get the synchronizing benefits of an active medium without suffering the destructive consequences of chaotic arrhythmias.
Collapse
Affiliation(s)
- R. Scott McIsaac
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Graduate Program in Quantitative and Computational Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Kerwyn Casey Huang
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
| | - Anirvan Sengupta
- Department of Physics & Astronomy, Rutgers University, Piscataway, New Jersey, United States of America
- BioMAPS Institute for Quantitative Biology, Rutgers University, Piscataway, New Jersey, United States of America
| | - Ned S. Wingreen
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
47
|
Eckerdt F, Yamamoto TM, Lewellyn AL, Maller JL. Identification of a polo-like kinase 4-dependent pathway for de novo centriole formation. Curr Biol 2011; 21:428-32. [PMID: 21353560 PMCID: PMC3093158 DOI: 10.1016/j.cub.2011.01.072] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 12/30/2010] [Accepted: 01/28/2011] [Indexed: 12/26/2022]
Abstract
Supernumerary centrosomes are a key cause of genomic instability in cancer cells. New centrioles can be generated by duplication with a mother centriole as a platform or, in the absence of preexisting centrioles, by formation de novo. Polo-like kinase 4 (Plk4) regulates both modes of centriole biogenesis, and Plk4 deregulation has been linked to tumor development. We show that Plx4, the Xenopus homolog of mammalian Plk4 and Drosophila Sak, induces de novo centriole formation in vivo in activated oocytes and in egg extracts, but not in immature or in vitro matured oocytes. Both kinase activity and the polo-box domain of Plx4 are required for de novo centriole biogenesis. Polarization microscopy in "cycling" egg extracts demonstrates that de novo centriole formation is independent of Cdk2 activity, a major difference compared to template-driven centrosome duplication that is linked to the nuclear cycle and requires cyclinA/E/Cdk2. Moreover, we show that the Mos-MAPK pathway blocks Plx4-dependent de novo centriole formation before fertilization, thereby ensuring paternal inheritance of the centrosome. The results define a new system for studying the biochemical and molecular basis of de novo centriole formation and centriole biogenesis in general.
Collapse
Affiliation(s)
- Frank Eckerdt
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045
| | - Tomomi M. Yamamoto
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045
| | - Andrea L. Lewellyn
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045
| | - James L. Maller
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045
| |
Collapse
|
48
|
Chang HY, Jennings PC, Stewart J, Verrills NM, Jones KT. Essential role of protein phosphatase 2A in metaphase II arrest and activation of mouse eggs shown by okadaic acid, dominant negative protein phosphatase 2A, and FTY720. J Biol Chem 2011; 286:14705-12. [PMID: 21383018 DOI: 10.1074/jbc.m110.193227] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vertebrate eggs arrest at second meiotic metaphase. The fertilizing sperm causes meiotic exit through Ca(2+)-mediated activation of the anaphase-promoting complex/cyclosome (APC/C). Although the loss in activity of the M-phase kinase CDK1 is known to be an essential downstream event of this process, the contribution of phosphatases to arrest and meiotic resumption is less apparent, especially in mammals. Therefore, we explored the role of protein phosphatase 2A (PP2A) in mouse eggs using pharmacological inhibition and activation as well as a functionally dominant-negative catalytic PP2A subunit (dn-PP2Ac-L199P) coupled with live cell imaging. We observed that PP2A inhibition using okadaic acid induced events normally observed at fertilization: degradation of the APC/C substrates cyclin B1 and securin resulting from loss of the APC/C inhibitor Emi2. Although sister chromatids separated, chromatin remained condensed, and polar body extrusion was blocked as a result of a rapid spindle disruption, which could be ameliorated by non-degradable cyclin B1, suggesting that spindle integrity was affected by CDK1 loss. Similar cell cycle effects to okadaic acid were also observed using dominant-negative PP2Ac. Preincubation of eggs with the PP2A activator FTY720 could block many of the actions of okadaic acid, including Emi2, cyclin B1, and securin degradation and sister chromatid separation. Therefore, in conclusion, we used okadaic acid, dn-PP2Ac-L199P, and FTY720 on mouse eggs to demonstrate that PP2A is needed to for both continued metaphase arrest and successful exit from meiosis.
Collapse
Affiliation(s)
- Heng-Yu Chang
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, 2308, Australia
| | | | | | | | | |
Collapse
|
49
|
McLean JR, Chaix D, Ohi MD, Gould KL. State of the APC/C: organization, function, and structure. Crit Rev Biochem Mol Biol 2011; 46:118-36. [PMID: 21261459 DOI: 10.3109/10409238.2010.541420] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The ubiquitin-proteasome protein degradation system is involved in many essential cellular processes including cell cycle regulation, cell differentiation, and the unfolded protein response. The anaphase-promoting complex/cyclosome (APC/C), an evolutionarily conserved E3 ubiquitin ligase, was discovered 15 years ago because of its pivotal role in cyclin degradation and mitotic progression. Since then, we have learned that the APC/C is a very large, complex E3 ligase composed of 13 subunits, yielding a molecular machine of approximately 1 MDa. The intricate regulation of the APC/C is mediated by the Cdc20 family of activators, pseudosubstrate inhibitors, protein kinases and phosphatases and the spindle assembly checkpoint. The large size, complexity, and dynamic nature of the APC/C represent significant obstacles toward high-resolution structural techniques; however, over the last decade, there have been a number of lower resolution APC/C structures determined using single particle electron microscopy. These structures, when combined with data generated from numerous genetic and biochemical studies, have begun to shed light on how APC/C activity is regulated. Here, we discuss the most recent developments in the APC/C field concerning structure, substrate recognition, and catalysis.
Collapse
Affiliation(s)
- Janel R McLean
- Howard Hughes Medical Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | | |
Collapse
|
50
|
Schindler K. Protein kinases and protein phosphatases that regulate meiotic maturation in mouse oocytes. Results Probl Cell Differ 2011; 53:309-341. [PMID: 21630151 DOI: 10.1007/978-3-642-19065-0_14] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Oocytes arrest at prophase of meiosis I (MI) and in vivo do not resume meiosis until they receive ovulatory cues. Meiotic resumption entails two rounds of chromosome segregation without an intervening round of DNA replication and an arrest at metaphase of meiosis II (MII); fertilization triggers exit from MII and entry into interphase. During meiotic resumption, there is a burst of protein phosphorylation and dephosphorylation that dramatically changes during the course of oocyte meiotic maturation. Many of these phosphorylation and dephosphorylation events are key to regulating meiotic cell cycle arrest and/or progression, chromosome dynamics, and meiotic spindle assembly and disassembly. This review, which is subdivided into sections based upon meiotic cell cycle stages, focuses on the major protein kinases and phosphatases that have defined requirements during meiosis in mouse oocytes and, when possible, connects these regulatory pathways.
Collapse
Affiliation(s)
- Karen Schindler
- Department of Biology, University of Pennsylvania, 433 S. University Avenue, Philadelphia, PA 19104, USA.
| |
Collapse
|