1
|
Bafleh WS, Abdulsamad HMR, Al-Qaraghuli SM, El Khatib RY, Elbahrawi RT, Abdukadir AM, Alsawae SM, Dimassi Z, Hamdan H, Kashir J. Applications of advances in mRNA-based platforms as therapeutics and diagnostics in reproductive technologies. Front Cell Dev Biol 2023; 11:1198848. [PMID: 37305677 PMCID: PMC10250609 DOI: 10.3389/fcell.2023.1198848] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/18/2023] [Indexed: 06/13/2023] Open
Abstract
The recent COVID-19 pandemic led to many drastic changes in not only society, law, economics, but also in science and medicine, marking for the first time when drug regulatory authorities cleared for use mRNA-based vaccines in the fight against this outbreak. However, while indeed representing a novel application of such technology in the context of vaccination medicine, introducing RNA into cells to produce resultant molecules (proteins, antibodies, etc.) is not a novel principle. It has been common practice to introduce/inject mRNA into oocytes and embryos to inhibit, induce, and identify several factors in a research context, while such aspects have also been proposed as potential therapeutic and diagnostic applications to combat infertility in humans. Herein, we describe key areas where mRNA-based platforms have thus far represented potential areas of clinical applications, describing the advantages and limitations of such applications. Finally, we also discuss how recent advances in mRNA-based platforms, driven by the recent pandemic, may stand to benefit the treatment of infertility in humans. We also present brief future directions as to how we could utilise recent and current advancements to enhance RNA therapeutics within reproductive biology, specifically with relation to oocyte and embryo delivery.
Collapse
Affiliation(s)
- Wjdan S. Bafleh
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Haia M. R. Abdulsamad
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Sally M. Al-Qaraghuli
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Riwa Y. El Khatib
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Rawdah Taha Elbahrawi
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Azhar Mohamud Abdukadir
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | | | - Zakia Dimassi
- Department of Pediatrics, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Hamdan Hamdan
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
- Healthcare Engineering Innovation Center (HEIC), Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Junaid Kashir
- Department of Biology, College of Arts and Science, Khalifa University, Abu Dhabi, United Arab Emirates
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Kashir J, Ganesh D, Jones C, Coward K. OUP accepted manuscript. Hum Reprod Open 2022; 2022:hoac003. [PMID: 35261925 PMCID: PMC8894871 DOI: 10.1093/hropen/hoac003] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/16/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Oocyte activation deficiency (OAD) is attributed to the majority of cases underlying failure of ICSI cycles, the standard treatment for male factor infertility. Oocyte activation encompasses a series of concerted events, triggered by sperm-specific phospholipase C zeta (PLCζ), which elicits increases in free cytoplasmic calcium (Ca2+) in spatially and temporally specific oscillations. Defects in this specific pattern of Ca2+ release are directly attributable to most cases of OAD. Ca2+ release can be clinically mediated via assisted oocyte activation (AOA), a combination of mechanical, electrical and/or chemical stimuli which artificially promote an increase in the levels of intra-cytoplasmic Ca2+. However, concerns regarding safety and efficacy underlie potential risks that must be addressed before such methods can be safely widely used. OBJECTIVE AND RATIONALE Recent advances in current AOA techniques warrant a review of the safety and efficacy of these practices, to determine the extent to which AOA may be implemented in the clinic. Importantly, the primary challenges to obtaining data on the safety and efficacy of AOA must be determined. Such questions require urgent attention before widespread clinical utilization of such protocols can be advocated. SEARCH METHODS A literature review was performed using databases including PubMed, Web of Science, Medline, etc. using AOA, OAD, calcium ionophores, ICSI, PLCζ, oocyte activation, failed fertilization and fertilization failure as keywords. Relevant articles published until June 2019 were analysed and included in the review, with an emphasis on studies assessing large-scale efficacy and safety. OUTCOMES Contradictory studies on the safety and efficacy of AOA do not yet allow for the establishment of AOA as standard practice in the clinic. Heterogeneity in study methodology, inconsistent sample inclusion criteria, non-standardized outcome assessments, restricted sample size and animal model limitations render AOA strictly experimental. The main scientific concern impeding AOA utilization in the clinic is the non-physiological method of Ca2+ release mediated by most AOA agents, coupled with a lack of holistic understanding regarding the physiological mechanism(s) underlying Ca2+ release at oocyte activation. LIMITATIONS, REASONS FOR CAUTION The number of studies with clinical relevance using AOA remains significantly low. A much wider range of studies examining outcomes using multiple AOA agents are required. WIDER IMPLICATIONS In addition to addressing the five main challenges of studies assessing AOA safety and efficacy, more standardized, large-scale, multi-centre studies of AOA, as well as long-term follow-up studies of children born from AOA, would provide evidence for establishing AOA as a treatment for infertility. The delivery of an activating agent that can more accurately recapitulate physiological fertilization, such as recombinant PLCζ, is a promising prospect for the future of AOA. Further to PLCζ, many other avenues of physiological oocyte activation also require urgent investigation to assess other potential physiological avenues of AOA. STUDY FUNDING/COMPETING INTERESTS D.G. was supported by Stanford University’s Bing Overseas Study Program. J.K. was supported by a Healthcare Research Fellowship Award (HF-14-16) made by Health and Care Research Wales (HCRW), alongside a National Science, Technology, and Innovation plan (NSTIP) project grant (15-MED4186-20) awarded by the King Abdulaziz City for Science and Technology (KACST). The authors have no competing interests to declare.
Collapse
Affiliation(s)
| | | | - Celine Jones
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Level 3, Women’s Centre, John Radcliffe Hospital, Oxford, UK
| | - Kevin Coward
- Correspondence address. Nuffield Department of Women’s & Reproductive Health, University of Oxford, Level 3, Women’s Centre, John Radcliffe Hospital, Oxford, OS3 9DU, UK. E-mail: https://orcid.org/0000-0003-3577-4041
| |
Collapse
|
3
|
Mitochondrial fission promotes radiation-induced increase in intracellular Ca 2+ level leading to mitotic catastrophe in mouse breast cancer EMT6 cells. Biochem Biophys Res Commun 2019; 522:144-150. [PMID: 31757415 DOI: 10.1016/j.bbrc.2019.11.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 11/04/2019] [Indexed: 01/11/2023]
Abstract
Mitochondrial dynamics are crucial for cellular survival in response to various stresses. Previously, we reported that Drp1 promoted mitochondrial fission after x-irradiation and its inhibition resulted in reduced cellular radiosensitivity and mitotic catastrophe. However, the mechanisms of radiation-induced mitotic catastrophe related to mitochondrial fission remain unclear. In this study, we investigated the involvement of cellular ATP production, ROS generation, and Ca2+ levels in mitotic catastrophe in EMT6 cells. Knockdown of Drp1 and Fis1, which are mitochondrial fission regulators, resulted in elongated mitochondria and significantly attenuated cellular radiosensitivity. Reduced mitochondrial fission mainly decreased mitotic catastrophe rather than necrosis and apoptosis after irradiation. Cellular ATP contents in Drp1 and Fis1 knockdown cells were similar to those in control cells. N-acetylcysteine and 2-glucopyranoside ascorbic acid have no effect on mitotic catastrophe after irradiation. The cellular [Ca2+]i level increased after irradiation, which was completely suppressed by Drp1 and Fis1 inhibition. Furthermore, BAPTA-AM significantly reduced radiation-induced mitotic catastrophe, indicating that cellular Ca2+ is a key mediator of mitotic catastrophe induction after irradiation. These results suggest that mitochondrial fission is associated with radiation-induced mitotic catastrophe via cytosolic Ca2+ regulation.
Collapse
|
4
|
Li R, Leblanc J, He K, Liu XJ. Spindle function in Xenopus oocytes involves possible nanodomain calcium signaling. Mol Biol Cell 2016; 27:3273-3283. [PMID: 27582389 PMCID: PMC5170860 DOI: 10.1091/mbc.e16-05-0338] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/25/2016] [Indexed: 11/11/2022] Open
Abstract
Injection of dibromo-BAPTA caused immediate collapse of meiotic spindles in frog oocytes. In contrast, EGTA had no effect on the spindle or polar body emission. The disruption of spindle integrity by the fast but not slow calcium chelators suggests that meiotic spindle function in the oocytes involves nanodomain calcium signaling. Intracellular calcium transients are a universal phenomenon at fertilization and are required for egg activation, but the exact role of Ca2+ in second-polar-body emission remains unknown. On the other hand, similar calcium transients have not been demonstrated during oocyte maturation, and yet, manipulating intracellular calcium levels interferes with first-polar-body emission in mice and frogs. To determine the precise role of calcium signaling in polar body formation, we used live-cell imaging coupled with temporally precise intracellular calcium buffering. We found that BAPTA-based calcium chelators cause immediate depolymerization of spindle microtubules in meiosis I and meiosis II. Surprisingly, EGTA at similar or higher intracellular concentrations had no effect on spindle function or polar body emission. Using two calcium probes containing permutated GFP and the calcium sensor calmodulin (Lck-GCaMP3 and GCaMP3), we demonstrated enrichment of the probes at the spindle but failed to detect calcium increase during oocyte maturation at the spindle or elsewhere. Finally, endogenous calmodulin was found to colocalize with spindle microtubules throughout all stages of meiosis. Our results—most important, the different sensitivities of the spindle to BAPTA and EGTA—suggest that meiotic spindle function in frog oocytes requires highly localized, or nanodomain, calcium signaling.
Collapse
Affiliation(s)
- Ruizhen Li
- Ottawa Hospital Research Institute, Ottawa Hospital-General Campus, Ottawa, ON K1H 8L6, Canada
| | - Julie Leblanc
- Ottawa Hospital Research Institute, Ottawa Hospital-General Campus, Ottawa, ON K1H 8L6, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Kevin He
- Ottawa Hospital Research Institute, Ottawa Hospital-General Campus, Ottawa, ON K1H 8L6, Canada
| | - X Johné Liu
- Ottawa Hospital Research Institute, Ottawa Hospital-General Campus, Ottawa, ON K1H 8L6, Canada .,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada.,Department of Obstetrics and Gynaecology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
5
|
Jia JL, Han YH, Kim HC, Ahn M, Kwon JW, Luo Y, Gunasekaran P, Lee SJ, Lee KS, Kyu Bang J, Kim NH, Namgoong S. Structural basis for recognition of Emi2 by Polo-like kinase 1 and development of peptidomimetics blocking oocyte maturation and fertilization. Sci Rep 2015; 5:14626. [PMID: 26459104 PMCID: PMC4602232 DOI: 10.1038/srep14626] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/01/2015] [Indexed: 11/08/2022] Open
Abstract
In a mammalian oocyte, completion of meiosis is suspended until fertilization by a sperm, and the cell cycle is arrested by a biochemical activity called cytostatic factor (CSF). Emi2 is one of the CSFs, and it maintains the protein level of maturation promoting factor (MPF) by inhibiting ubiquitin ligase anaphase promoting complex/cyclosome (APC/C). Degradation of Emi2 via ubiquitin-mediated proteolysis after fertilization requires phosphorylation by Polo-like kinase 1 (Plk1). Therefore, recognition and phosphorylation of Emi2 by Plk1 are crucial steps for cell cycle resumption, but the binding mode of Emi2 and Plk1 is poorly understood. Using biochemical assays and X-ray crystallography, we found that two phosphorylated threonines (Thr(152) and Thr(176)) in Emi2 are each responsible for the recruitment of one Plk1 molecule by binding to its C-terminal polo box domain (PBD). We also found that meiotic maturation and meiosis resumption via parthenogenetic activation were impaired when Emi2 interaction with Plk1-PBD was blocked by a peptidomimetic called 103-8. Because of the inherent promiscuity of kinase inhibitors, our results suggest that targeting PBD of Plk1 may be an effective strategy for the development of novel and specific contraceptive agents that block oocyte maturation and/or fertilization.
Collapse
Affiliation(s)
- Jia-Lin Jia
- Department of Animal Sciences, Chungbuk National University, Republic of Korea
| | - Young-Hyun Han
- Department of Animal Sciences, Chungbuk National University, Republic of Korea
| | - Hak-Cheol Kim
- Department of Animal Sciences, Chungbuk National University, Republic of Korea
| | - Mija Ahn
- Division of Magnetic Resonance, Korea Basic Science Institute, Ochang, Korea
| | - Jeong-Woo Kwon
- Department of Animal Sciences, Chungbuk National University, Republic of Korea
| | - Yibo Luo
- Department of Animal Sciences, Chungbuk National University, Republic of Korea
| | | | - Soo-Jae Lee
- College of Pharmacy, Chungbuk National University, Republic of Korea
| | - Kyung S. Lee
- National Cancer Institute, National Institute of Health, Rockville, Maryland, United States
| | - Jeong Kyu Bang
- Division of Magnetic Resonance, Korea Basic Science Institute, Ochang, Korea
| | - Nam-Hyung Kim
- Department of Animal Sciences, Chungbuk National University, Republic of Korea
| | - Suk Namgoong
- Department of Animal Sciences, Chungbuk National University, Republic of Korea
| |
Collapse
|
6
|
Premkumar KV, Chaube SK. Nitric oxide signals postovulatory aging-induced abortive spontaneous egg activation in rats. Redox Rep 2015; 20:184-92. [PMID: 25780809 DOI: 10.1179/1351000215y.0000000003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE The aim of this study was to determine whether an increase of intracellular nitric oxide (NO) level signals postovulatory aging-induced abortive spontaneous egg activation (SEA) in rats. METHODS Freshly ovulated eggs (arrested at metaphase-II stage; M-II) were cultured in vitro for 3 hours to induce postovulatory egg aging. The morphological changes, inducible nitric oxide synthase (iNOS) expression, NO, cytosolic free Ca(2+), 3',5' cyclic guanosine monophosphate (cGMP), cell division cycle 25B (Cdc25B) and Wee1 levels, specific phosphorylation (pThr-14/Tyr-15) as well as total cyclin-dependent kinases-1 (Cdk1) (PSTAIRE) levels were analyzed. RESULTS Postovulatory aging induced generation of NO possibly through an iNOS-mediated pathway. The increase in NO level was associated with augmented cytosolic free Ca(2+) as well as cGMP levels in aged eggs. A significant increase in Wee1 level and decrease of Cdc25B level were observed in aged eggs. An accumulation of phosphorylated Cdk1 (pThr-14/Tyr-15) level was observed in aged eggs, while total Cdk1 (PSTAIR) level remained unchanged. CONCLUSION Our study demonstrates that generation of NO through an iNOS-mediated pathway increases cytosolic free Ca2+and cGMP levels. High levels of these signal molecules trigger the accumulation of phosphorylated Cdk1 in aged eggs. Thus, NO signals the accumulation of phosphorylated Cdk1 and induces postovulatory aging-induced abortive SEA in the rat.
Collapse
|
7
|
Abstract
Mammalian oocytes are particularly error prone in segregating their chromosomes during their two meiotic divisions. This results in the creation of an embryo that has inherited the wrong number of chromosomes: it is aneuploid. The incidence of aneuploidy rises significantly with maternal age and so there is much interest in understanding this association and the underlying causes of aneuploidy. The spindle assembly checkpoint, a surveillance mechanism that operates in all cells to prevent chromosome mis-segregation, and the cohesive ties that hold those chromosomes together, have thus both been the subject of intensive investigation in oocytes. It is possible that a lowered sensitivity of the spindle assembly checkpoint to certain types of chromosome attachment error may endow oocytes with an innate susceptibility to aneuploidy, which is made worse by an age-related loss in the factors that hold the chromosomes together.
Collapse
Affiliation(s)
- Keith T Jones
- Centre for Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | | |
Collapse
|
8
|
Kashir J, Deguchi R, Jones C, Coward K, Stricker SA. Comparative biology of sperm factors and fertilization-induced calcium signals across the animal kingdom. Mol Reprod Dev 2013; 80:787-815. [PMID: 23900730 DOI: 10.1002/mrd.22222] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 07/23/2013] [Indexed: 11/08/2022]
Abstract
Fertilization causes mature oocytes or eggs to increase their concentrations of intracellular calcium ions (Ca²⁺) in all animals that have been examined, and such Ca²⁺ elevations, in turn, provide key activating signals that are required for non-parthenogenetic development. Several lines of evidence indicate that the Ca²⁺ transients produced during fertilization in mammals and other taxa are triggered by soluble factors that sperm deliver into oocytes after gamete fusion. Thus, for a broad-based analysis of Ca²⁺ dynamics during fertilization in animals, this article begins by summarizing data on soluble sperm factors in non-mammalian species, and subsequently reviews various topics related to a sperm-specific phospholipase C, called PLCζ, which is believed to be the predominant activator of mammalian oocytes. After characterizing initiation processes that involve sperm factors or alternative triggering mechanisms, the spatiotemporal patterns of Ca²⁺ signals in fertilized oocytes or eggs are compared in a taxon-by-taxon manner, and broadly classified as either a single major transient or a series of repetitive oscillations. Both solitary and oscillatory types of fertilization-induced Ca²⁺ signals are typically propagated as global waves that depend on Ca²⁺ release from the endoplasmic reticulum in response to increased concentrations of inositol 1,4,5-trisphosphate (IP₃). Thus, for taxa where relevant data are available, upstream pathways that elevate intraoocytic IP3 levels during fertilization are described, while other less-common modes of producing Ca²⁺ transients are also examined. In addition, the importance of fertilization-induced Ca²⁺ signals for activating development is underscored by noting some major downstream effects of these signals in various animals.
Collapse
Affiliation(s)
- Junaid Kashir
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Level 3, Women's Centre, John Radcliffe Hospital, Headington, Oxford, UK
| | | | | | | | | |
Collapse
|
9
|
Holt JE, Lane SIR, Jennings P, García-Higuera I, Moreno S, Jones KT. APC(FZR1) prevents nondisjunction in mouse oocytes by controlling meiotic spindle assembly timing. Mol Biol Cell 2012; 23:3970-81. [PMID: 22918942 PMCID: PMC3469513 DOI: 10.1091/mbc.e12-05-0352] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The APC activator FZR1 has a role in controlling the timing of meiosis I spindle assembly. Oocytes lacking FZR1 undergo accelerated meiosis I, associated with earlier spindle assembly checkpoint satisfaction and APCCDC20 activity, resulting in high rates of aneuploidy. FZR1 is an anaphase-promoting complex (APC) activator best known for its role in the mitotic cell cycle at M-phase exit, in G1, and in maintaining genome integrity. Previous studies also established that it prevents meiotic resumption, equivalent to the G2/M transition. Here we report that mouse oocytes lacking FZR1 undergo passage through meiosis I that is accelerated by ∼1 h, and this is due to an earlier onset of spindle assembly checkpoint (SAC) satisfaction and APCCDC20 activity. However, loss of FZR1 did not compromise SAC functionality; instead, earlier SAC satisfaction was achieved because the bipolar meiotic spindle was assembled more quickly in the absence of FZR1. This novel regulation of spindle assembly by FZR1 led to premature bivalent attachment to microtubules and loss of kinetochore-bound MAD2. Bivalents, however, were observed to congress poorly, leading to nondisjunction rates of 25%. We conclude that in mouse oocytes FZR1 controls the timing of assembly of the bipolar spindle and in so doing the timing of SAC satisfaction and APCCDC20 activity. This study implicates FZR1 as a major regulator of prometaphase whose activity helps to prevent chromosome nondisjunction.
Collapse
Affiliation(s)
- Janet E Holt
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.
| | | | | | | | | | | |
Collapse
|
10
|
Liu XJ. Polar body emission. Cytoskeleton (Hoboken) 2012; 69:670-85. [PMID: 22730245 DOI: 10.1002/cm.21041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 05/15/2012] [Accepted: 05/21/2012] [Indexed: 12/13/2022]
Abstract
Generation of a haploid female germ cell, the egg, consists of two rounds of asymmetric cell division (meiosis I and meiosis II), yielding two diminutive and nonviable polar bodies and a large haploid egg. Animal eggs are also unique in the lack of centrioles and therefore form meiotic spindles without the pre-existence of the two dominant microtubule organizing centers (centrosomes) found in mitosis. Meiotic spindle assembly is further complicated by the unique requirement of sister chromatid mono-oriented in meiosis I. Nonetheless, the eggs appear to adopt many of the same proteins and mechanisms described in mitosis, with necessary modifications to accommodate their special needs. Unraveling these special modifications will not only help understanding animal reproduction, but should also enhance our understanding of cell division in general.
Collapse
Affiliation(s)
- X Johné Liu
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa Hospital Civic Campus, 1053 Carling Avenue, Ottawa, K1Y 4E9, Canada.
| |
Collapse
|
11
|
Calcium Oscillations, Oocyte Activation, and Phospholipase C zeta. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:1095-121. [DOI: 10.1007/978-94-007-2888-2_50] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
12
|
Martín-Romero FJ, López-Guerrero AM, Álvarez IS, Pozo-Guisado E. Role of Store-Operated Calcium Entry During Meiotic Progression and Fertilization of Mammalian Oocytes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 295:291-328. [DOI: 10.1016/b978-0-12-394306-4.00014-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
13
|
Roscovitine in combination with calcium ionophore induces oocyte activation through reduction of M-phase promoting factor activity in mice. ZYGOTE 2011; 20:321-5. [DOI: 10.1017/s0967199411000591] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SummaryThe aim of the present study was to determine oocyte activation and change in M-phase promoting factor (MPF) activity induced by treatment with calcium ionophore and roscovitine in comparison with those induced by treatment with roscovitine alone and treatment with calcium ionophore and puromycin in mice. Freshly ovulated oocytes obtained from 6–8-week-old mice were divided into five groups (no activation treatment; 5 μM calcium ionophore A23187; 50 μM roscovitine; 5 μM calcium ionophore and 10 μg/ml puromycin; and 5 μM calcium ionophore and 50 μM roscovitine) and were incubated for 6 h. Oocyte activation, assessed by morphological changes, and changes in MPF activity in the five groups at 0, 2, 4 and 6 h of incubation were examined. Activated oocytes were defined as oocytes with at least one pronucleus. Oocytes treated with roscovitine alone were not activated during the 6-h incubation period. All of the oocytes in the calcium ionophore with puromycin group and in the calcium ionophore with roscovitine group were activated. The percentage activity of MPF in oocytes treated with roscovitine alone was decreased after 2 h and increased after 4 h of incubation. The percentage activity of MPF in oocytes treated with calcium ionophore and roscovitine was significantly decreased with suppression of MPF activity being maintained for 6 h, and this change was similar to that in oocytes treated with calcium ionophore and puromycin. Roscovitine with calcium ionophore is effective for induction of oocyte activation through suppression of MPF activity in mice.
Collapse
|
14
|
Liang SL, Zhao QJ, Li XC, Jin YP, Wang YP, Su XH, Guan WJ, Ma YH. Dynamic analysis of Ca²+ level during bovine oocytes maturation and early embryonic development. J Vet Sci 2011; 12:133-42. [PMID: 21586872 PMCID: PMC3104167 DOI: 10.4142/jvs.2011.12.2.133] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Mammalian oocyte maturation and early embryo development processes are Ca2+-dependent. In this study, we used confocal microscopy to investigate the distribution pattern of Ca2+ and its dynamic changes in the processes of bovine oocytes maturation, in vitro fertilization (IVF), parthenogenetic activation (PA) and somatic cell nuclear transfer (SCNT) embryo development. During the germinal vesicle (GV) and GV breakdown stage, Ca2+ was distributed in the cortical ooplasm and throughout the oocytes from the MI to MII stage. In IVF embryos, Ca2+ was distributed in the cortical ooplasm before the formation of the pronucleus. In 4-8 cell embryos and morulas, Ca2+ was present throughout the blastomere. In PA embryos, Ca2+ was distributed throughout the blastomere at 48 h, similar to in the 4-cell and 8-cell phase and the morula. At 6 h after activation, there was almost no distribution of Ca2+ in the SCNT embryos. However, Ca2+ was distributed in the donor nucleus at 10 h and it was distributed throughout the blastomere in the 2-8 cell embryos. In this study, Ca2+ showed significant fluctuations with regularity of IVF and SCNT groups, but PA did not. Systematic investigation of the Ca2+ location and distribution changes during oocyte maturation and early embryo development processes should facilitate a better understanding of the mechanisms involved in oocyte maturation, reconstructed embryo activation and development, ultimately improving the reconstructed embryo development rate.
Collapse
Affiliation(s)
- Su Li Liang
- College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling 712100, China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
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
|
16
|
Brahmaraju M, Bhagya KP, Titus S, Sebastian A, Devi AN, Laloraya M, Kumar PG. AIRE1A might be involved in cyclin B2 degradation in testicular lysates. Biochem Cell Biol 2011; 89:411-22. [PMID: 21819345 DOI: 10.1139/o11-029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The autoimmune regulator gene Aire shows predominant expression in thymus and other immunologically relevant tissues, and is assigned the major function of programming autoreactive T-cell deletion. However, the expression of this gene in tissues outside the immune system raises a question about its possible function beyond the T-cell deletion dogma. We detected Aire in mouse testis, and the expression of AIRE protein was remarkably high in postmeiotic germ cells. Sequencing results indicate that testis expressed Aire variant 1a. AIRE could be detected in spermatozoa, with heavy localization on the principal acrosomal domains. Mouse oocytes stained negatively for AIRE before fertilization, but stained positively for AIRE 30 min after fertilization. In the zygote, the levels of AIRE correlated negatively with cyclin B2 levels. Goat testicular lysates spiked with recombinant human AIRE exhibited augmented cyclin B2 degradation in the presence of protease inhibitors, which was inhibited by MG-132, indicating the operation of proteasomal pathways. Thus, this study identifies a correlation between the presence of AIRE and proteasomal breakdown of cyclin B2, which leads us to speculate that cyclin B2 could be a target of AIRE's E3-ubiquitin ligase activity.
Collapse
Affiliation(s)
- M Brahmaraju
- Division of Molecular Reproduction, Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Poojappura, Trivandrum, Kerala, India
| | | | | | | | | | | | | |
Collapse
|
17
|
Zheng LP, Huang J, Zhang DL, Xu LQ, Li F, Wu L, Liu ZY, Zheng YH. c-erbB2 and c-myb induce mouse oocyte maturation involving activation of maturation promoting factor. DNA Cell Biol 2011; 31:164-70. [PMID: 21793718 DOI: 10.1089/dna.2011.1219] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Proto-oncogenes are involved in cell growth, proliferation, and differentiation. In the present study, we investigated the roles and mediating pathways of proto-oncogenes c-erbB(2) and c-myb in mouse oocyte maturation by RT-PCR, real-time quantitative PCR, western blot, and recombinant proto-oncogene protein microinjection. Results showed that both c-erbB(2) and c-myb antisense oligodeoxynucleotides (c-erbB(2) ASODN and c-myb ASODN) inhibited germinal vesicle breakdown and the first polar body extrusion in a dose-dependent manner. However, microinjection of recombinant c-erbB(2) or c-myb protein into germinal vesicle stage oocytes stimulated oocyte meiotic maturation. In addition, the expression of c-erbB(2) and c-myb mRNA was detected in oocytes; and c-erbB(2) ASODN and c-myb ASODN inhibited c-erbB(2) mRNA and c-myb mRNA expression, respectively. Maturation promoting factor (MPF) inhibitor roscovitine did not affect the expression of c-erbB(2) mRNA and c-myb mRNA, but blocked the effects of recombinant c-erbB(2) and c-myb protein-induced oocyte maturation. Further, cyclin B1 protein expression in oocytes was remarkably inhibited by c-erbB(2) ASODN, c-myb ASODN, and roscovitine. Nonsense tat ODN had no effect on the expression of c-erbB(2), c-myb, and cyclin B1. These results suggest that c-erbB(2) and c-myb may induce oocyte maturation through mediating a pathway involving the activation of MPF.
Collapse
Affiliation(s)
- Li-Ping Zheng
- Medical Experimental Teaching Department, Nanchang University, Nanchang, China
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Lane SIR, Chang HY, Jennings PC, Jones KT. The Aurora kinase inhibitor ZM447439 accelerates first meiosis in mouse oocytes by overriding the spindle assembly checkpoint. Reproduction 2010; 140:521-30. [DOI: 10.1530/rep-10-0223] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Previous studies have established that when maturing mouse oocytes are continuously incubated with the Aurora inhibitor ZM447439, meiotic maturation is blocked. In this study, we observe that by altering the time of addition of the inhibitor, oocyte maturation can actually be accelerated by 1 h as measured by the timing of polar body extrusion. ZM447439 also had the ability to overcome a spindle assembly checkpoint (SAC) arrest caused by nocodazole and so rescue polar body extrusion. Consistent with the ability of the SAC to inhibit cyclin B1 degradation by blocking activation of the anaphase-promoting complex, we could also observe a rescue in cyclin B1 degradation when ZM447439 was added to nocodazole-treated oocytes. The acceleration of the first meiotic division by ZM447439, which has not been achieved previously, and its effects on the SAC are all consistent with the proposed mitotic role of Aurora B in activating the SAC. We hypothesize that Aurora kinase activity controls the SAC in meiosis I, despite differences to the mitotic cell cycle division in spindle architecture brought about by the meiotic mono-orientation of sister kinetochores.
Collapse
|
19
|
Beall S, Brenner C, Segars J. Oocyte maturation failure: a syndrome of bad eggs. Fertil Steril 2010; 94:2507-13. [PMID: 20378111 DOI: 10.1016/j.fertnstert.2010.02.037] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Revised: 02/15/2010] [Accepted: 02/16/2010] [Indexed: 11/29/2022]
Abstract
To show that disruption of meiotic competence results in cell cycle arrest, and the production of immature oocytes that are not capable of fertilization. Through an extensive review of animal studies and clinical case reports, we define the syndrome of oocyte maturation failure as a distinct oocyte disorder, present a classification system based on clinical parameters, and discuss the potential molecular origins for the disease.
Collapse
Affiliation(s)
- Stephanie Beall
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | |
Collapse
|
20
|
Vichera G, Alfonso J, Duque CC, Silvestre MA, Pereyra-Bonnet F, Fernández-Martín R, Salamone D. Chemical Activation with a Combination of Ionomycin and Dehydroleucodine for Production of Parthenogenetic, ICSI and Cloned Bovine Embryos. Reprod Domest Anim 2009; 45:e306-12. [DOI: 10.1111/j.1439-0531.2009.01563.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
21
|
Feng D, Zhou Y, Ling B, Gao T, Shi Y, Wei H, Tian Z. Effects of the conditioned medium of mesenchymal stem cells on mouse oocyte activation and development. Braz J Med Biol Res 2009; 42:506-14. [DOI: 10.1590/s0100-879x2009000600006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Accepted: 03/11/2009] [Indexed: 11/22/2022] Open
Affiliation(s)
| | | | - B. Ling
- Anhui Medical University; Anhui Medical University, China
| | - T. Gao
- Anhui Medical University, China
| | | | - H.M. Wei
- University of Science and Technology of China, China
| | - Z.G. Tian
- University of Science and Technology of China, China
| |
Collapse
|
22
|
Calcium regulation of mitochondria motility and morphology. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2008; 1787:1363-73. [PMID: 19138660 DOI: 10.1016/j.bbabio.2008.12.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 12/10/2008] [Accepted: 12/10/2008] [Indexed: 12/21/2022]
Abstract
In the Fifties, electron microscopy studies on neuronal cells showed that mitochondria typically cluster at synaptic terminals, thereby introducing the concept that proper mitochondria trafficking and partitioning inside the cell could provide functional support to the execution of key physiological processes. Today, the notion that a central event in the life of every eukaryotic cell is to configure, maintain, and reorganize the mitochondrial network at sites of high energy demand in response to environmental and cellular cues is well established, and the challenge ahead is to define the underlying molecular mechanisms and regulatory pathways. Recent pioneering studies have further contributed to place mitochondria at the center of the cell biology by showing that the machinery governing remodeling of mitochondria shape and structure regulates the functional output of the organelle as the powerhouse of the cell, the gateway to programmed cell death, and the platform for Ca(2+) signaling. Thus, a raising issue is to identify the cues integrating mitochondria trafficking and dynamics into cell physiology and metabolism. Given the versatile function of calcium as a second messenger and of the role of mitochondria as a major calcium store, evidences are emerging linking Ca(2+) transients to the modulation of mitochondrial activities. This review focuses on calcium as a switch controlling mitochondria motility and morphology in steady state, stressed, and pathological conditions.
Collapse
|
23
|
Yamamuro T, Kano K, Naito K. Functions of FZR1 and CDC20, Activators of the Anaphase-Promoting Complex, During Meiotic Maturation of Swine Oocytes1. Biol Reprod 2008; 79:1202-9. [DOI: 10.1095/biolreprod.108.070326] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
24
|
Ajduk A, Ciemerych MA, Nixon V, Swann K, Maleszewski M. Fertilization differently affects the levels of cyclin B1 and M-phase promoting factor activity in maturing and metaphase II mouse oocytes. Reproduction 2008; 136:741-52. [DOI: 10.1530/rep-08-0271] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fertilization affects levels of cyclin B1 and M-phase promoting factor (MPF) activity in maturing and metaphase II mouse oocytes in two distinct ways. In metaphase II oocytes, it leads to a Ca2+-dependent, continuous degradation of cyclin B1 and inactivation of cyclin dependent kinase (CDC2A)–cyclin B1 complex (MPF). In this paper, we show that neither mono- nor polyspermic fertilization of prometaphase I and metaphase I oocytes triggered degradation of cyclin B1. However, polyspermic fertilization of prometaphase I oocytes led to a transient decrease in MPF activity that lasted for 2 h. The inactivation of MPF in polyspermic prometaphase I oocytes did not depend on the fertilization-induced increase in the cytoplasmic concentration of free Ca2+ions, but was caused, at least in part, by dephosphorylation of CDC2A at threonine 161 (Thr161). We found that polyspermic fertilization did not affect glutathione levels in prometaphase I oocytes, and concluded that the decrease in MPF activity and dephosphorylation of CDC2A at Thr161 in polyspermic prometaphase I oocytes were not caused by a change in the redox status of the cell induced by an introduction of excessive amount of sperm protamines. Instead, we propose that inactivation of MPF activity in polyspermic maturing oocytes is caused by a change in nucleo-cytoplasmic ratio that leads to a ‘titration’ of kinases and phosphatases responsible for keeping MPF in an active state. This idea is supported by the finding that oocytes fused with thymocytes rather than spermatozoa also showed a transient decrease in MPF activity.
Collapse
|
25
|
Mori H, Mizobe Y, Inoue S, Uenohara A, Takeda M, Yoshida M, Miyoshi K. Effects of cycloheximide on parthenogenetic development of pig oocytes activated by ultrasound treatment. J Reprod Dev 2008; 54:364-9. [PMID: 18635923 DOI: 10.1262/jrd.20064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The present study was carried out to examine the parthenogenetic development of pig oocytes treated with different concentrations of cycloheximide for different durations following activation by ultrasound stimulation. When oocytes were treated with 10 microg/ml cycloheximide for different durations, the blastocyst formation rate of oocytes treated for 5 h was significantly (P<0.05) higher than those of oocytes treated for 0-2 h. The blastocyst formation rate of oocytes treated with 10 microg/ml cycloheximide for 5 h was significantly (P<0.05) higher than those of oocytes treated with 0-5 or 15-20 microg/ml cycloheximide for the same duration. When oocytes were treated with different concentrations of cycloheximide for 2 h, however, the blastocyst formation rate of oocytes treated with 40 microg/ml cycloheximide was significantly (P<0.05) higher than those of oocytes treated with 0-10 or 50 microg/ml cycloheximide. The blastocyst formation rate of oocytes treated with 10 microg/ml cycloheximide for 5 h was not significantly different from that of oocytes treated with 40 microg/ml cycloheximide for 2 h. These treatments did not affect the activation status of oocytes compared with controls that were not treated with cycloheximide. The results of the present study showed that cycloheximide improves the parthenogenetic development of pig oocytes activated by ultrasound stimulation.
Collapse
Affiliation(s)
- Hironori Mori
- Laboratory of Animal Reproduction, Faculty of Agriculture, Kagoshima University, Kagoshima, Japan
| | | | | | | | | | | | | |
Collapse
|
26
|
Nabti I, Reis A, Levasseur M, Stemmann O, Jones KT. Securin and not CDK1/cyclin B1 regulates sister chromatid disjunction during meiosis II in mouse eggs. Dev Biol 2008; 321:379-86. [PMID: 18639540 DOI: 10.1016/j.ydbio.2008.06.036] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 06/23/2008] [Accepted: 06/25/2008] [Indexed: 11/17/2022]
Abstract
Mammalian eggs remain arrested at metaphase of the second meiotic division (metII) for an indeterminate time before fertilization. During this period, which can last several hours, the continued attachment of sister chromatids is thought to be achieved by inhibition of the protease separase. Separase is known to be inhibited by binding either securin or Maturation (M-Phase)-Promoting Factor, a heterodimer of CDK1/cyclin B1. However, the relative contribution of securin and CDK/cyclin B1 to sister chromatid attachment during metII arrest has not been assessed. Although there are conditions in which either CDK1/cyclinB1 activity or securin can prevent sister chromatid disjunction, principally by overexpression of non-degradable cyclin B1 or securin, we find here that separase activity is primarily regulated by securin and not CDK1/cyclin B1. Thus the CDK1 inhibitor roscovitine and an antibody we designed to block the interaction of CDK1/cyclin B1 with separase, both failed to induce sister disjunction. In contrast, securin morpholino knockdown specifically induced loss of sister attachment, that could be restored by securin cRNA rescue. During metII arrest separase appears primarily regulated by securin binding, not CDK1/cyclin B1.
Collapse
Affiliation(s)
- Ibtissem Nabti
- Institute for Cell and Molecular Biosciences, The Medical School, Framlington Place, University of Newcastle, Newcastle, NE2 4HH, UK
| | | | | | | | | |
Collapse
|
27
|
Nánássy L, Lee K, Jávor A, Macháty Z. Effects of activation methods and culture conditions on development of parthenogenetic porcine embryos. Anim Reprod Sci 2008; 104:264-74. [PMID: 17320316 DOI: 10.1016/j.anireprosci.2007.01.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2006] [Accepted: 01/26/2007] [Indexed: 11/18/2022]
Abstract
The effects of different activation methods and culture conditions on early development of porcine parthenotes were examined. Three different activation methods were tested: (1) electroporation; (2) electroporation followed by incubation in the presence of butyrolactone I, an inhibitor of cdc2 and cdk2 kinases; and (3) electroporation followed by a treatment with cycloheximide, a blocker of protein synthesis. The activated oocytes were cultured in two different media, NCSU-23 and PZM-3 under 5% CO2 in air. In a separate experiment, the effects of high (approximately 20%) or low (5%) O2 tension on early embryo development were also evaluated. The average pronuclear formation was less (p<0.05) in the electroporated oocytes (83.9+/-1.7%) compared with those activated by electroporation and butyrolactone I or electroporation plus cycloheximide (92.8+/-0.8 and 93.0+/-1.0%). In PZM-3 medium, the average frequencies of blastocyst formation (59.7+/-3.6%) and hatching (10.6+/-1.3%) were greater than those in NCSU-23 medium (39.9+/-3.1% blastocyst formation, p<0.05; and 0.2+/-0.2% hatching; p<0.001). Furthermore, the average nuclear number was also greater (p<0.001) in blastocysts developed in PZM-3 (50.2+/-1.3) than in those developed in NCSU-23 (35.3+/-1.1). Blastocyst formation was similar (p>0.10) among the three activation procedures when parthenotes were cultured in NCSU-23, while in PZM-3 more (p<0.05) parthenotes produced by electroporation plus butyrolactone or electroporation plus cycloheximide developed into blastocysts compared to electroporation alone (64.9+/-5.2 and 68.6+/-3.5% compared with 45.6+/-4.7%). Incidences of apoptotic nuclei were similar (p>0.10) among all treatments. No difference in development was found between parthenotes that developed under high versus low O2 tension (p>0.10). These results demonstrate that activation methods targeting the calcium signaling pathway at several points trigger embryonic development more efficiently than electroporation alone. The data also imply that the PZM-3 medium provides for enhanced culture conditions for the early development of parthenogenetic porcine embryos than NCSU-23.
Collapse
Affiliation(s)
- László Nánássy
- Department of Animal Breeding Science, Center of Agricultural Sciences, University of Debrecen, Böszörményi Street 138, Debrecen, Hungary
| | | | | | | |
Collapse
|
28
|
Martín-Romero FJ, Ortíz-de-Galisteo JR, Lara-Laranjeira J, Domínguez-Arroyo JA, González-Carrera E, Alvarez IS. Store-operated calcium entry in human oocytes and sensitivity to oxidative stress. Biol Reprod 2007; 78:307-15. [PMID: 18003943 DOI: 10.1095/biolreprod.107.064527] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Calcium signaling is a cellular event that plays a key role at many steps of fertilization and early development. However, little is known regarding the contribution of extracellular Ca(2+) influx into the cell to this signaling in gametes and early embryos. To better know the significance of calcium entry on oocyte physiology, we have evaluated the mechanism of store-operated calcium entry (SOCE) in human metaphase II (MII) oocytes and its sensitivity to oxidative stress, one of the major factors implicated in the outcome of in vitro fertilization (IVF) techniques. We show that depletion of intracellular Ca(2+) stores through inhibition of sarco(endo)plasmic Ca(2+)-ATPase with thapsigargin triggers Ca(2+) entry in resting human oocytes. Ba(2+) and Mn(2+) influx was also stimulated following inhibition, and Ca(2+) entry was sensitive to pharmacological inhibition because the SOCE blocker 2-aminoethoxydiphenylborate (2-APB) reduced calcium and barium entry. These results support the conclusion that there is a plasma membrane mechanism responsible for the capacitative divalent cation entry in human oocytes. Moreover, the Ca(2+) entry mechanism described in MII oocytes was found to be highly sensitive to oxidative stress. Hydrogen peroxide, at micromolar concentrations that could mimic culture conditions in IVF, elicited an increase of [Ca(2+)](i) that was dependent on the presence of extracellular Ca(2+). This rise was preventable by 2-APB, indicating that it was mainly due to the enhanced influx through store-operated calcium channels. In sum, our results demonstrate the occurrence of SOCE in human MII oocytes and the modification of this pathway due to oxidative stress, with possible consequences in IVF.
Collapse
Affiliation(s)
- Francisco Javier Martín-Romero
- Departamento de Bioquímicay Biología Molecular, Reproduction and Development Group (REDES), Universidad de Extremadura, Badajoz-06071, Spain.
| | | | | | | | | | | |
Collapse
|
29
|
Gardner AJ, Williams CJ, Evans JP. Establishment of the mammalian membrane block to polyspermy: evidence for calcium-dependent and -independent regulation. Reproduction 2007; 133:383-93. [PMID: 17307906 DOI: 10.1530/rep-06-0304] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
One crucial result of egg activation is the establishment of blocks on the zona pellucida and the egg plasma membrane to prevent fertilization by additional sperm. The mechanism(s) by which a mammalian egg regulates the establishment of the membrane block to polyspermy is largely unknown. Since Ca(2+) signaling regulates several egg activation events, this study investigates how sperm-induced Ca(2+) transients affect the membrane block to polyspermy, building on our previous work (Biology of Reproduction 67:1342). We demonstrate that mouse eggs that experience only one sperm-induced Ca(2+) transient establish a membrane block that is less effective, than in eggs that experience normal sperm-induced Ca(2+) transients but that is more effective than in eggs with completely suppressed [Ca(2+)](cyt) increases. Sperm-induced increases in [Ca(2+)](cyt) regulate the timing of membrane block establishment, as this block is established more slowly in eggs that experience one or no sperm-induced Ca(2+) transients. Finally, our studies produce the intriguing discovery that there is also a Ca(2+)-independent event that is associated with fertilization in the pathway leading to membrane block establishment. Taken together, these data indicate that Ca(2+) plays a role in facilitating membrane block establishment by regulating the timing with which this change in egg membrane function occurs, and also that the membrane block differs from other post-fertilization egg activation responses as Ca(2+) is not the only stimulus. The membrane block to polyspermy in mammalian eggs is likely to be the culmination of multiple post-fertilization events that together modify the egg membrane's receptivity to sperm.
Collapse
Affiliation(s)
- Allison J Gardner
- Division of Reproductive Biology, Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Room W3606, 615 N. Wolfe St., Maryland, USA
| | | | | |
Collapse
|
30
|
Ito J, Kato M, Hochi S, Hirabayashi M. Effect of Enucleation on Inactivation of Cytostatic Factor Activity in Matured Rat Oocytes. CLONING AND STEM CELLS 2007; 9:257-66. [PMID: 17579558 DOI: 10.1089/clo.2006.0042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In mammals, matured oocytes are arrested at the MII stage until fertilization, which is regulated by cytostaticfactor (CSF) activity. Maturation-promoting factor (MPF) and the mitogen-activated protein kinase (MAPK) pathway are known as candidates for CSF. Despite of the results that nuclear and perinuclear materials were dispensable for activation of MPF and MAPK in other species, our previous study in rats demonstrated that MPF activity was rapidly decreased after enucleation. We showed here for the first time that nuclear and perinuclear materials were indispensable for CSF activity in matured rat oocytes. In both cytoplasm-removed and enucleated oocytes, high activity of p34(cdc2) kinase was observed immediately after manipulation, but the activity of enucleated oocytes was dramatically reduced within 1 h. Cyclin B level was also decreased, corresponding with inactivation of p34(cdc2) kinase. In enucleated oocytes, the Mos level was dramatically decreased, and both MEK and MAPK dephosphorylation were also induced. A combined treatment with a proteasome inhibitor, MG132, and a protein phosphatase inhibitor, okadaic acid, dramatically improved both levels of p-MAPK and cyclin B in these enucleated oocytes. These data suggest that nuclear and perinuclear materials of matured rat oocytes suppress proteasome and protein phosphatase activation, which is indispensable for stability of CSF.
Collapse
Affiliation(s)
- Junya Ito
- Section of Molecular Genetics, Center for Brain Experiment, National Institute for Physiological Sciences, Okazaki, Aichi, Japan.
| | | | | | | |
Collapse
|
31
|
Malcuit C, Fissore RA. Activation of fertilized and nuclear transfer eggs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 591:117-31. [PMID: 17176559 DOI: 10.1007/978-0-387-37754-4_9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
In all animal species, initiation of embryonic development occurs shortly after the joining together of the gametes from each of the sexes. The first of these steps, referred to as "egg activation", is a series of molecular events that results in the syngamy of the two haploid genomes and the beginning of cellular divisions for the new diploid embryo. For many years it has been known that the incoming sperm drives this process, as an unfertilized egg will remain dormant until it can no longer sustain normal metabolic processes. Until recently, it was also believed that the sperm was the only cell capable of creating a viable embryo and offspring. Recent advances in cell biology have allowed researchers to not only understand the molecular mechanisms of egg activation, but to exploit the use of pharmacological agents to bypass sperm-induced egg activation for the creation of animals by somatic cell nuclear transfer. This chapter will focus on the molecular events of egg activation in mammals as they take place during fertilization, and will discuss how these mechanisms are successfully bypassed in processes such as somatic cell nuclear transfer.
Collapse
Affiliation(s)
- Christopher Malcuit
- Department of Veterinary and Animal Sciences, Paige Laboratory, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | | |
Collapse
|
32
|
Villemure M, Chen HY, Kurokawa M, Fissore RA, Taketo T. The presence of X- and Y-chromosomes in oocytes leads to impairment in the progression of the second meiotic division. Dev Biol 2007; 301:1-13. [PMID: 17123505 DOI: 10.1016/j.ydbio.2006.10.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2006] [Revised: 10/19/2006] [Accepted: 10/24/2006] [Indexed: 11/28/2022]
Abstract
The oocytes of B6.Y(TIR) sex-reversed female mice can be fertilized but the resultant embryos die at early cleavage stages. In the present study, we examined chromosome segregation at meiotic divisions in the oocytes of XY female mice, compared to those of XX littermates. The timing and frequency of oocyte maturation in culture were comparable between the oocytes from both types of females. At the first meiotic division, the X- and Y-chromosomes segregated independently and were retained in oocytes at equal frequencies. However, more oocytes retained the correct number of chromosomes than anticipated from random segregation. The oocytes that had reached MII-stage were activated by fertilization or incubation with SrCl(2). As expected, the majority of oocytes from XX females completed the second meiotic division and reached the 2-cell stage in 24 h. By contrast, more than half of oocytes from XY females initially remained at the MII-stage while the rest precociously entered interphase after SrCl(2) activation; very few oocytes were seen at the second anaphase or telophase and they often showed impairment of sister-chromatid separation. Eventually the majority of oocytes entered interphase and formed pronuclei, but very few reached the 2-cell stage. Similar results were obtained after fertilization. We conclude that the XY chromosomal composition in oocyte leads to impairment in the progression of the second meiotic division.
Collapse
Affiliation(s)
- M Villemure
- Urology Research Laboratory, Department of Surgery, McGill University, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
33
|
Rogers NT, Halet G, Piao Y, Carroll J, Ko MSH, Swann K. The absence of a Ca(2+) signal during mouse egg activation can affect parthenogenetic preimplantation development, gene expression patterns, and blastocyst quality. Reproduction 2006; 132:45-57. [PMID: 16816332 DOI: 10.1530/rep.1.01059] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A series of Ca(2+) oscillations during mammalian fertilization is necessary and sufficient to stimulate meiotic resumption and pronuclear formation. It is not known how effectively development continues in the absence of the initial Ca(2+) signal. We have triggered parthenogenetic egg activation with cycloheximide that causes no Ca(2+) increase, with ethanol that causes a single large Ca(2+) increase, or with Sr(2+) that causes Ca(2+) oscillations. Eggs were co-treated with cytochalasin D to make them diploid and they formed pronuclei and two-cell embryos at high rates with each activation treatment. However, far fewer of the embryos that were activated by cycloheximide reached the blastocyst stagecompared tothose activated by Sr(2+) orethanol. Any cycloheximide-activated embryos that reached the blastocyst stage had a smaller inner cell mass number and a greater rate of apoptosis than Sr(2+)-activated embryos. The poor development of cycloheximide-activated embryos was due to the lack of Ca(2+) increase because they developed to blastocyst stages at high rates when co-treated with Sr(2+) or ethanol. Embryos activated by either Sr(2+) or cycloheximide showed similar signs of initial embryonic genome activation (EGA) when measured using a reporter gene. However, microarray analysis of gene expression at the eight-cell stage showed that activation by Sr(2+) leads to a distinct pattern of gene expression from that seen with embryos activated by cycloheximide. These data suggest that activation of mouse eggs in the absence of a Ca(2+) signal does not affect initial parthenogenetic events, but can influence later gene expression and development.
Collapse
Affiliation(s)
- N T Rogers
- Department of Anatomy and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | | | | | | | | | | |
Collapse
|
34
|
Mehlmann LM, Kalinowski RR, Ross LF, Parlow AF, Hewlett EL, Jaffe LA. Meiotic resumption in response to luteinizing hormone is independent of a Gi family G protein or calcium in the mouse oocyte. Dev Biol 2006; 299:345-55. [PMID: 16949564 PMCID: PMC1864934 DOI: 10.1016/j.ydbio.2006.07.039] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2006] [Revised: 07/17/2006] [Accepted: 07/28/2006] [Indexed: 11/29/2022]
Abstract
The signaling pathway by which luteinizing hormone (LH) acts on the somatic cells of vertebrate ovarian follicles to stimulate meiotic resumption in the oocyte requires a decrease in cAMP in the oocyte, but how cAMP is decreased is unknown. Activation of Gi family G proteins can lower cAMP by inhibiting adenylate cyclase or stimulating a cyclic nucleotide phosphodiesterase, but we show here that inhibition of this class of G proteins by injection of pertussis toxin into follicle-enclosed mouse oocytes does not prevent meiotic resumption in response to LH. Likewise, elevation of Ca2+ can lower cAMP through its action on Ca2+-sensitive adenylate cyclases or phosphodiesterases, but inhibition of a Ca2+ rise by injection of EGTA into follicle-enclosed mouse oocytes does not inhibit the LH response. Thus, neither of these well-known mechanisms of cAMP regulation can account for LH signaling to the oocyte in the mouse ovary.
Collapse
Affiliation(s)
- Lisa M. Mehlmann
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06032
| | - Rebecca R. Kalinowski
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06032
| | - Lavinia F. Ross
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06032
| | - Albert F. Parlow
- National Hormone and Peptide Program, Harbor-UCLA Medical Center, Torrance, CA 90509
| | - Erik L. Hewlett
- Division of Infectious Diseases and International Health, and Departments of Medicine and Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Laurinda A. Jaffe
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06032
| |
Collapse
|
35
|
Madgwick S, Hansen DV, Levasseur M, Jackson PK, Jones KT. Mouse Emi2 is required to enter meiosis II by reestablishing cyclin B1 during interkinesis. ACTA ACUST UNITED AC 2006; 174:791-801. [PMID: 16966421 PMCID: PMC2064334 DOI: 10.1083/jcb.200604140] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
During interkinesis, a metaphase II (MetII) spindle is built immediately after the completion of meiosis I. Oocytes then remain MetII arrested until fertilization. In mouse, we find that early mitotic inhibitor 2 (Emi2), which is an anaphase-promoting complex inhibitor, is involved in both the establishment and the maintenance of MetII arrest. In MetII oocytes, Emi2 needs to be degraded for oocytes to exit meiosis, and such degradation, as visualized by fluorescent protein tagging, occurred tens of minutes ahead of cyclin B1. Emi2 antisense morpholino knockdown during oocyte maturation did not affect polar body (PB) extrusion. However, in interkinesis the central spindle microtubules from meiosis I persisted for a short time, and a MetII spindle failed to assemble. The chromatin in the oocyte quickly decondensed and a nucleus formed. All of these effects were caused by the essential role of Emi2 in stabilizing cyclin B1 after the first PB extrusion because in Emi2 knockdown oocytes a MetII spindle was recovered by Emi2 rescue or by expression of nondegradable cyclin B1 after meiosis I.
Collapse
Affiliation(s)
- Suzanne Madgwick
- Institute for Cell and Molecular Biosciences, The Medical School, University of Newcastle, Newcastle NE2 4HH, England, UK.
| | | | | | | | | |
Collapse
|
36
|
Abstract
A dramatic increase in intracellular Ca(2+) concentration ([Ca(2+)](i)) occurs in eggs at fertilization common to all animal species examined to date, and this serves as a pivotal signal for egg activation characterized by resumption of meiotic cell division and formation of the pronuclei. In mammalian eggs, repetitive [Ca(2+)](i) rises (Ca(2+) oscillations) each of which accompanies a propagating wave across the egg occur due to release of Ca(2+) from the endoplasmic reticulum mainly through type 1 inositol 1,4,5-trisphosphate (IP(3)) receptor. Ca(2+) oscillations are induced by a cytosolic sperm factor driven into the egg cytoplasm upon sperm-egg fusion. A current strong candidate of the sperm factor is a novel sperm-specific isozyme of phospholipase C (IP(3)-producing enzyme), PLCzeta. Recent extensive research has reveled characteristics of PLCzeta such as the Ca(2+) oscillation-inducing activity after injection of PLCzeta-encoding RNA or recombinant PLCzeta into mouse eggs, extremely high Ca(2+)-sensitivity of the enzymatic activity in vitro, and nuclear translocation ability possibly related to cell-cycle-dependent regulation of Ca(2+) oscillations. [Ca(2+)](i) rises cause successive activation of calmodulin-dependent kinase II and E3 ubiquitin ligase, lead to proteolysis of ubiquitinated cyclin B1 and inactivation of metaphase-promoting factor (Cdk1/cyclin B1 complex), and result in the release of eggs from meiotic arrest.
Collapse
Affiliation(s)
- Shunichi Miyazaki
- Department of Physiology, Tokyo Women's Medical University School of Medicine, Japan.
| | | |
Collapse
|
37
|
Polanski Z, Hoffmann S, Tsurumi C. Oocyte nucleus controls progression through meiotic maturation. Dev Biol 2006; 281:184-95. [PMID: 15893972 DOI: 10.1016/j.ydbio.2005.02.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2004] [Revised: 02/21/2005] [Accepted: 02/23/2005] [Indexed: 11/26/2022]
Abstract
We analyzed progression through the meiotic maturation in oocytes manipulated to replace the prophase oocyte nucleus with the nucleus from a cumulus cell, a pachytene spermatocyte or the pronucleus from a fertilized egg. Removal of the oocyte nucleus led to a significant reduction in histone H1 kinase activity. Replacement of the oocyte nucleus by a pronucleus followed by culture resulted in premature pseudomeiotic division and occasional abnormal cytokinesis; however, histone H1 kinase activity was rescued, microtubules formed a bipolar spindle, and chromosomes were condensed. In addition to the anomalies observed after pronuclear transfer, those after transfer of the nucleus from a cumulus cell or spermatocyte included a dramatically impaired ability to form the bipolar spindle or to condense chromosomes, and histone H1 kinase activity was not rescued. Expression of a cyclin B-YFP in enucleated oocytes receiving the cumulus cell nucleus rescued histone H1 kinase activity, but spindle formation and chromosome condensation remained impaired, indicating a pleiotropic effect of oocyte nucleus removal. However, when the cumulus cell nucleus was first transformed into pronuclei (transfer into a metaphase II oocyte followed by activation), such pronuclei supported maturation after transfer into the oocyte in a manner similar to that of normal pronuclei. These results show that the oocyte nucleus contains specific components required for the control of progression through the meiotic maturation and that some of these components are also present in pronuclei.
Collapse
Affiliation(s)
- Zbigniew Polanski
- Department of Developmental Biology, Max-Planck-Institute of Immunobiology, Stuebeweg 51, D-79108 Freiburg, Germany.
| | | | | |
Collapse
|
38
|
Abstract
Fertilization in all species studied to date induces an increase in the intracellular concentration of free calcium ions ([Ca2+]i) within the egg. In mammals, this [Ca2+]i signal is delivered in the form of long-lasting [Ca2+]i oscillations that begin shortly after fusion of the gametes and persist beyond the time of completion of meiosis. While not fully elucidated, recent evidence supports the notion that the sperm delivers into the ooplasm a trigger of oscillations, the so-called sperm factor (SF). The recent discovery that mammalian sperm harbor a specific phospholipase C (PLC), PLCzeta has consolidated this view. The fertilizing sperm, and presumably PLCzeta promote Ca2+ release in eggs via the production of inositol 1,4,5-trisphosphate (IP3), which binds and gates its receptor, the type-1 IP3 receptor, located on the endoplasmic reticulum, the Ca2+ store of the cell. Repetitive Ca2+ release in this manner results in a positive cumulative effect on downstream signaling molecules that are responsible for the completion of all the events comprising egg activation. This review will discuss recent advances in our understanding of how [Ca2+]i oscillations are initiated and regulated in mammals, highlight areas of discrepancies, and emphasize the need to better characterize the downstream molecular cascades that are dependent on [Ca2+]i oscillations and that may impact embryo development.
Collapse
Affiliation(s)
- Christopher Malcuit
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | | | | |
Collapse
|
39
|
Hoffmann S, Tsurumi C, Kubiak JZ, Polanski Z. Germinal vesicle material drives meiotic cell cycle of mouse oocyte through the 3′UTR-dependent control of cyclin B1 synthesis. Dev Biol 2006; 292:46-54. [PMID: 16490186 DOI: 10.1016/j.ydbio.2005.12.052] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2005] [Revised: 12/23/2005] [Accepted: 12/23/2005] [Indexed: 11/27/2022]
Abstract
We compared the profile of histone H1 kinase activity, reflecting Maturation Promoting Factor (MPF) activity in oocytes bisected at the germinal vesicle (GV) stage and allowed to mature as separate oocyte halves in vitro. Whereas the oocyte halves containing the nucleus exhibited the same profile of increased kinase activity as that typical for intact oocytes, the anuclear halves revealed strong inhibition of the increase in this activity soon after germinal vesicle breakdown (GVBD). In contrast, the profile of MAP kinase activity did not differ significantly between anuclear and nucleus-containing oocyte halves throughout maturation. Of the two MPF components, CDK1 and cyclin B1, the amount of the latter was significantly reduced in anuclear halves, a reduction due to low-level synthesis and not to enhanced degradation. Expression of three reporter luciferase RNAs constructed, respectively, to contain cyclin B1-specific 3'UTR, the globin-specific 3'UTR, or no 3'UTR sequence was enhanced in nuclear halves, with significantly greater enhancement for the construct containing cyclin B1-specific 3'UTR as compared to the two other RNAs. We conclude that the profile of activity of MPF during mouse oocyte maturation is controlled by an unknown GV-associated factor(s) acting via 3'UTR-dependent control of cyclin B1 synthesis. These results require the revision of the hitherto prevailing view that the control of MPF activity during mouse oocyte maturation is independent of GV-derived material.
Collapse
Affiliation(s)
- Steffen Hoffmann
- Department of Developmental Biology, Max Planck Institute of Immunobiology, Stuebeweg 51, 79-108 Freiburg, Germany
| | | | | | | |
Collapse
|
40
|
Tomashov-Matar R, Tchetchik D, Eldar A, Kaplan-Kraicer R, Oron Y, Shalgi R. Strontium-induced rat egg activation. Reproduction 2006; 130:467-74. [PMID: 16183864 DOI: 10.1530/rep.1.00746] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Parthenogenetic agents that evoke cytosolic calcium concentration ([Ca2+]i) oscillations similar to those evoked by sperm, mimic fertilization more faithfully than agents that trigger a single [Ca2+]i transient. Strontium chloride (SrCl2) binds to and activates the Ca2+-binding site on the inositol 1,4,5-trisphosphate receptor and evokes [Ca2+]i oscillations. Although SrCl2 has been reported to activate mouse eggs, little is known regarding the pattern of the [Ca2+]i oscillations it evokes in rat eggs and their effect on the early events of egg activation: cortical granule exocytosis (CGE) and completion of meiosis (CM). In the current study we investigated the effect of various concentrations of SrCl2 (2, 4 or 6 mM) on [Ca2+]i, by monitoring [Ca2+]i oscillations in fura-2-loaded rat eggs. Treatment with 2 mM SrCl2 was optimal for inducing the first [Ca2+]i transient, which was similar in duration to that triggered by sperm. However, the frequency and duration of the subsequent [Ca2+]i oscillations were lower and longer in SrCl2-activated than in sperm-activated eggs. The degree of CGE was identical in eggs activated by either sperm or SrCl2, as assessed by semi-quantitative immunohistochemistry combined with confocal microscopy. Evoking 1, 2 or 10 [Ca2+]i oscillations (8, 15 or 60 min in SrCl2 respectively) had no effect on the intensity of fluorescent CGE reporter dyes, while 60-min exposure to SrCl2 caused a delay in CM. Our results demonstrate that SrCl2 is an effective parthenogenetic agent that mimics rat egg activation by sperm, as judged by the generation of [Ca2+]i oscillations, CGE and CM.
Collapse
Affiliation(s)
- R Tomashov-Matar
- Cell and Developmental Biology and Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv 9978 Tel-Aviv, Israel
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
It was discovered about 30 years ago that a dramatic increase in intracellular calcium ion concentration ([Ca(2+)](i)) occurs at fertilization and that this increase acts as the pivotal signal for egg activation. Later, the Ca(2+) signal at fertilization turned out to be ubiquitous among animal species. Extensive advance has been brought during these 30 years in research on spatiotemporal aspects and signaling mechanisms of the [Ca(2+)](i) increase, sperm factors that induce the Ca(2+) response, and cell cycle resumption caused by the [Ca(2+)](i) rise. I provide a historical account of these advances in mammals, sea urchins, and a few other models.
Collapse
Affiliation(s)
- Shunichi Miyazaki
- Department of Physiology, Tokyo Women's Medical University School of Medicine, Tokyo 162-8666, Japan.
| |
Collapse
|
42
|
Eliyahu E, Shtraizent N, Tsaadon A, Shalgi R. Association between myristoylated alanin-rich C kinase substrate (MARCKS) translocation and cortical granule exocytosis in rat eggs. Reproduction 2006; 131:221-31. [PMID: 16452716 DOI: 10.1530/rep.1.00794] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cortical granule exocytosis (CGE), following egg activation, is a secretory process that blocks polyspermy and enables successful embryonic development. CGE can be triggered independently by either a rise in intracellular calcium concentration ([Ca2+]i) or activation of protein kinase C (PKC). The present study investigates the signal transduction pathways leading to CGE through activation of PKC or stimulation of a rise in [Ca2+]i. Using Western blot analysis, co-immunoprecipitation and immunohistochemistry, combined with various inhibitors or activators, we investigated the link between myristoylated alanin-rich C kinase substrate (MARCKS) translocation and CGE. We were able to demonstrate translocation of MARCKS from the plasma membrane to the cortex, in fertilized as well as in parthenogenetically activated eggs. MARCKS phosphorylation was demonstrated upon PKC activation, whereas a PKC inhibitor (myrPKCψ) prevented both MARCKS translocation and CGE in 12-O-tetradecanoyl phorbol-13-acetate (TPA)-activated eggs. We have further shown that upon egg activation the amount of phosphorylated MARCKS (p-MARCKS) and the amount of calmodulin bound to MARCKS were increased. MARCKS translocation in ionomycin activated eggs was also inhibited by the calmodulin inhibitor N-(6-aminohexyl)-5-chloro-1-napthalenesulfonamide hydrochloride (W7). These results complement other studies showing MARCKS requirement for exocytosis and imply that upon fertilization, MARCKS translocation is followed by CGE. These findings present a significant contribution to our understanding of CGE in mammalian eggs in particular, as well as cellular exocytosis in general.
Collapse
Affiliation(s)
- Efrat Eliyahu
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | | | | | | |
Collapse
|
43
|
Abstract
Mammalian eggs arrest at metaphase of the second meiotic division (MetII). Sperm break this arrest by inducing a series of Ca2+spikes that last for several hours. During this time cell cycle resumption is induced, sister chromatids undergo anaphase and the second polar body is extruded. This is followed by decondensation of the chromatin and the formation of pronuclei. Ca2+spiking is both the necessary and solely sufficient sperm signal to induce full egg activation. How MetII arrest is established, how the Ca2+spiking is induced and how the signal is transduced into cell cycle resumption are the topics of this review. Although the roles of most components of the signal transduction pathway remain to be fully investigated, here I present a model in which a sperm-specific phospholipase C (PLCζ) generates Ca2+spikes to activate calmodulin-dependent protein kinase II and so switch on the Anaphase-Promoting Complex/Cyclosome (APC/C). APC/C activation leads to securin and cyclin B1 degradation and in so doing allows sister chromatids to be segregated and to decondense.
Collapse
Affiliation(s)
- Keith T Jones
- Institute for Cell and Molecular Biosciences, The Medical School, Framlington Place, University of Newcastle, Newcastle, NE2 4HH, UK.
| |
Collapse
|
44
|
FitzHarris G, Larman M, Richards C, Carroll J. An increase in [Ca2+]i is sufficient but not necessary for driving mitosis in early mouse embryos. J Cell Sci 2005; 118:4563-75. [PMID: 16179613 DOI: 10.1242/jcs.02586] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
An increase in intracellular Ca2+ concentration ([Ca2+]i) has been shown to drive sea-urchin embryos and some fibroblasts through nuclear-envelope breakdown (NEBD) and the metaphase-to-anaphase transition. Mitotic Ca2+ transients can be pan-cellular global events or localized to the perinuclear region. It is not known whether Ca2+ is a universal regulator of mitosis or whether its role is confined to specific cell types. To test the hypothesis that Ca2+ is a universal regulator of mitosis, we have investigated the role of Ca2+ in mitosis in one-cell mouse embryos. Fertilized embryos generate Ca2+ transients during the first mitotic division. Imposing a Ca2+ transient by photorelease of inositol (1,4,5)-trisphosphate [Ins(1,4,5)P3] resulted in acceleration of mitosis entry, suggesting that a [Ca2+]i increase is capable of triggering mitosis. Mitotic Ca2+ transients were inhibited using three independent approaches: injection of intracellular Ca2+ buffers; downregulation of Ins(1,4,5)P3 receptors; and removal of extracellular Ca2+. None of the interventions had any effects on the timing of NEBD or cytokinesis. The possibility that NEBD is driven by localized perinuclear Ca2+ transients was examined using two-photon microscopy but no Ca2+-dependent increases in fluorescence were found to precede NEBD. Finally, the second mitotic division took place in the absence of any detectable [Ca2+]i increase. Thus, although an induced [Ca2+]i increase can accelerate mitosis entry, neither cytosolic nor perinuclear [Ca2+] increases appear to be necessary for progression through mitosis in mouse embryos.
Collapse
MESH Headings
- Animals
- Calcium/metabolism
- Calcium Channels/metabolism
- Calcium Signaling/physiology
- Chelating Agents/chemistry
- Chelating Agents/metabolism
- Embryo, Mammalian/cytology
- Embryo, Mammalian/physiology
- Female
- Fertilization/physiology
- Fluorescent Dyes/chemistry
- Fluorescent Dyes/metabolism
- Fura-2/chemistry
- Fura-2/metabolism
- Inositol 1,4,5-Trisphosphate/chemistry
- Inositol 1,4,5-Trisphosphate/metabolism
- Inositol 1,4,5-Trisphosphate Receptors
- Mice
- Microinjections
- Microscopy, Fluorescence
- Mitosis/physiology
- Receptors, Cytoplasmic and Nuclear/metabolism
Collapse
Affiliation(s)
- Greg FitzHarris
- Department of Physiology, University College London, Gower Street, London WC1E 6BT, UK
| | | | | | | |
Collapse
|
45
|
Madgwick S, Levasseur M, Jones KT. Calmodulin-dependent protein kinase II, and not protein kinase C, is sufficient for triggering cell-cycle resumption in mammalian eggs. J Cell Sci 2005; 118:3849-59. [PMID: 16091425 DOI: 10.1242/jcs.02506] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Mouse eggs arrest at metaphase II following ovulation and are only triggered to complete meiosis when fertilized. Sperm break the cell-cycle arrest by a long-lasting series of Ca2+ spikes that lead to an activation of the anaphase-promoting complex/cyclosome. The signal transduction pathway is not fully resolved but both protein kinase C (PKC) and calmodulin-dependent protein kinase II (CamKII) activities increase at fertilization and previous pharmacological studies have implicated both in cell-cycle resumption. We have used a combination of pharmacological inhibitors and constitutively active cRNA constructs of PKCα and CamKIIα microinjected into mouse eggs to show that it is CamKII and not PKC that is the sufficient trigger for cell-cycle resumption from metaphase II arrest.
Constitutively active PKC constructs had no effect on the resumption of meiosis but caused an immediate and persistent elevation in intracellular Ca2+ when store-operated Ca2+ entry was stimulated. With respect to resumption of meiosis, the effects of constitutively active CamKII on eggs were the same as sperm. Eggs underwent second polar body extrusion and pronucleus formation with normal timings; while both securin and cyclin B1 destruction, visualised by coupling to fluorescent protein tags, were complete by the time of polar body extrusion. Induction of a spindle checkpoint by overexpression of Mad2 or by spindle poisons blocked CamKII-induced resumption of meiosis, but the Ca2+ chelator BAPTA did not. Furthermore direct measurement of Ca2+ levels showed that CamKII did not induce exit from metaphase II arrest by raising Ca2+. Therefore, we conclude that PKCs may play an important role in maintaining Ca2+ spiking at fertilization by promoting store-operated Ca2+ entry, while CamKII transduces cell-cycle resumption, and lies downstream of sperm-induced Ca2+ release but upstream of a spindle checkpoint. These data, combined with the knowledge that CamKII activity increase at fertilization, suggest that mouse eggs undergo cell-cycle resumption through stimulation of CamKII.
Collapse
Affiliation(s)
- Suzanne Madgwick
- Institute for Cell and Molecular Biosciences, The Medical School, Framlington Place, University of Newcastle, Newcastle, NE2 4HH, UK
| | | | | |
Collapse
|
46
|
Coward K, Ponting CP, Chang HY, Hibbitt O, Savolainen P, Jones KT, Parrington J. Phospholipase Cζ, the trigger of egg activation in mammals, is present in a non-mammalian species. Reproduction 2005; 130:157-63. [PMID: 16049153 DOI: 10.1530/rep.1.00707] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The activation of the egg to begin development into an embryo is triggered by a sperm-induced increase in intracellular egg Ca2+. There has been much controversy about how the sperm induces this fundamental developmental event, but recent studies suggest that, in mammals, egg activation is triggered by a testis-specific phospholipase C: PLCζ. Since the discovery of PLCζ, it has been unclear whether its role in triggering egg activation is common to all vertebrates, or is confined to mammals. Here, we demonstrate for the first time that PLCζ is present in a non-mammalian vertebrate. Using genomic and cDNA databases, we have identified the cDNA encoding a PLCζ orthologue in the domestic chicken that, like the mammalian isoforms, is a testis-specific gene. The chicken PLCζ cDNA is 2152 bp in size and encodes an open reading frame of 639 amino acids. When injected into mouse oocytes, chicken PLCζ cRNA triggers Ca2+oscillations, indicating that it has functional properties similar to those of mammalian PLCζ. Our findings suggest that PLCζ may have a universal role in triggering egg activation in vertebrates.
Collapse
Affiliation(s)
- K Coward
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| | | | | | | | | | | | | |
Collapse
|
47
|
Sone Y, Ito M, Shirakawa H, Shikano T, Takeuchi H, Kinoshita K, Miyazaki S. Nuclear translocation of phospholipase C-zeta, an egg-activating factor, during early embryonic development. Biochem Biophys Res Commun 2005; 330:690-4. [PMID: 15809052 DOI: 10.1016/j.bbrc.2005.03.032] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2005] [Indexed: 11/28/2022]
Abstract
Phospholipase C-zeta (PLCzeta), a strong candidate of the egg-activating sperm factor, causes intracellular Ca2+ oscillations and egg activation, and is subsequently accumulated into the pronucleus (PN), when expressed in mouse eggs by injection of RNA encoding PLCzeta. Changes in the localization of expressed PLCzeta were investigated by tagging with a fluorescent protein. PLCzeta began to translocate into the PN formed at 5-6 h after RNA injection and increased there. Observation in the same embryo revealed that PLCzeta in the PN dispersed to the cytoplasm upon nuclear envelope breakdown and translocated again into the nucleus after cleavage. The dynamics was found in the second mitosis as well. When RNA was injected into fertilization-originated 1-cell embryos or blastomere(s) of 2-8-cell embryos, the nuclear localization of expressed PLCzeta was recognized in every embryo up to blastocyst. Thus, PLCzeta exhibited alternative cytoplasm/nucleus localization during development. This supports the view that the sperm factor could control cell cycle-dependent generation of Ca2+ oscillations in early embryogenesis.
Collapse
Affiliation(s)
- Yoshie Sone
- Department of Physiology, Tokyo Women's Medical University School of Medicine, Tokyo 162-8666, Japan
| | | | | | | | | | | | | |
Collapse
|
48
|
Mehlmann LM, Jaffe LA. SH2 domain-mediated activation of an SRC family kinase is not required to initiate Ca2+ release at fertilization in mouse eggs. Reproduction 2005; 129:557-64. [PMID: 15855619 DOI: 10.1530/rep.1.00638] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
SRC family kinases (SFKs) function in initiating Ca2+release at fertilization in several species in the vertebrate evolutionary line, but whether they play a similar role in mammalian fertilization has been uncertain. We investigated this question by first determining which SFK proteins are expressed in mouse eggs, and then measuring Ca2+release at fertilization in the presence of dominant negative inhibitors. FYN and YES proteins were found in mouse eggs, but other SFKs were not detected; based on this, we injected mouse eggs with a mixture of FYN and YES Src homology 2 (SH2) domains. These SH2 domains were effective inhibitors of Ca2+release at fertilization in starfish eggs, but did not inhibit Ca2+release at fertilization in mouse eggs. Thus the mechanism by which sperm initiate Ca2+release in mouse eggs does not depend on SH2 domain-mediated activation of an SFK. We also tested the small molecule SFK inhibitor SU6656, and found that it became compartmentalized in the egg cytoplasm, thus suggesting caution in the use of this inhibitor. Our findings indicate that although the initiation of Ca2+release at fertilization of mammalian eggs occurs by a pathway that has many similarities to that in evolutionarily earlier animal groups, the requirement for SH2 domain-mediated activation of an SFK is not conserved.
Collapse
Affiliation(s)
- Lisa M Mehlmann
- Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, Connecticut 06032, USA.
| | | |
Collapse
|
49
|
Madgwick S, Nixon VL, Chang HY, Herbert M, Levasseur M, Jones KT. Maintenance of sister chromatid attachment in mouse eggs through maturation-promoting factor activity. Dev Biol 2004; 275:68-81. [PMID: 15464573 DOI: 10.1016/j.ydbio.2004.07.024] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2004] [Revised: 07/26/2004] [Accepted: 07/27/2004] [Indexed: 11/30/2022]
Abstract
Mammalian eggs naturally arrest at metaphase of the second meiotic division, until sperm triggers a series of Ca(2+) spikes that result in activation of the anaphase-promoting complex/cyclosome (APC/C). APC/C activation at metaphase targets destruction-box containing substrates, such as cyclin B1 and securin, for degradation, and as such eggs complete the second meiotic division. Cyclin B1 degradation reduces maturation (M-phase)-promoting factor (MPF) activity and securin degradation allows sister chromatid separation. Here we examined the second meiotic division in mouse eggs following expression of a cyclin B1 construct with an N-terminal 90 amino acid deletion (Delta 90 cyclin B1) that was visualized by coupling to EGFP. This cyclin construct was not an APC/C substrate, and so following fertilization, sperm were incapable of stimulating Delta 90 cyclin B1 degradation. In these eggs, chromatin remained condensed and no pronuclei formed. As a consequence of the lack of pronucleus formation, sperm-triggered Ca(2+) spiking continued indefinitely, consistent with a current model in which the sperm-activating factor is localized to the nucleus. Because Ca(2+) spiking was not inhibited by Delta 90 cyclin B1, the degradation timing of securin, visualized by coupling it to EGFP, was unaffected. However, despite rapid securin degradation, sister chromatids remained attached. This was a direct consequence of MPF activity because separation was induced following application of the MPF inhibitor roscovitine. Similar observations regarding the ability of MPF to prevent sister chromatid separation have recently been made in Xenopus egg extracts and in HeLa cells. The results presented here show this mechanism can also occur in intact mammalian eggs and further that this mechanism appears conserved among vertebrates. We present a model in which metaphase II arrest is maintained primarily by MPF levels only.
Collapse
Affiliation(s)
- Suzanne Madgwick
- Cell and Developmental Physiology Research Group, Institute of Cell and Molecular Biosciences, The Medical School, Framlington Place, University of Newcastle, NE2 4HH, UK
| | | | | | | | | | | |
Collapse
|
50
|
Chang HY, Levasseur M, Jones KT. Degradation of APCcdc20 and APCcdh1 substrates during the second meiotic division in mouse eggs. J Cell Sci 2004; 117:6289-96. [PMID: 15561765 DOI: 10.1242/jcs.01567] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Metaphase II-arrested mouse eggs are stimulated to complete meiosis by sperm-induced Ca2+ spiking. The Ca2+ signal causes activation of the E3 ligase anaphase-promoting complex/cyclosome (APC), leading to the destruction of key proteins necessary for meiotic exit. We show, using western blots of mouse eggs, the presence of both APC activators cdc20 and cdh1, which target D-box and D-box/KEN-box substrates, respectively, for proteolysis. We decided to examine the temporal activation of APCcdc20 and APCcdh1 by coupling APC substrates to GFP and examining their destruction in real-time following release from second meiotic division arrest. D-box substrates were degraded quickly after the initiation of sperm-induced Ca2+ spiking, such that their degradation was complete by the time of second polar body extrusion. By contrast, KEN-box-containing substrates were degraded when CDK1 activity was low, during the period between polar body extrusion and pronucleus formation. This observation of apparent APCcdh1 activity in meiosis II based on destruction of exogenous GFP-coupled substrates was then confirmed by observing destruction of endogenous APCcdh1 substrates. These data are consistent with a model of initial APCcdc20 activation on sperm-induced activation, followed by APCcdh1 activation after second polar body extrusion. Interestingly, therefore, we propose that mammalian eggs undergo meiosis II with both APCcdc20 and APCcdh1, whereas eggs of other species so far described have APCcdc20 activity only.
Collapse
Affiliation(s)
- Heng-Yu Chang
- Cell and Developmental Physiology Research Group, Institute for Cell and Molecular Biosciences, The Medical School, Framlington Place, University of Newcastle, Newcastle, NE2 4HH, UK
| | | | | |
Collapse
|