851
|
Gu L, Liu H, Gu X, Boots C, Moley KH, Wang Q. Metabolic control of oocyte development: linking maternal nutrition and reproductive outcomes. Cell Mol Life Sci 2014; 72:251-71. [PMID: 25280482 DOI: 10.1007/s00018-014-1739-4] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 09/12/2014] [Accepted: 09/22/2014] [Indexed: 02/01/2023]
Abstract
Obesity, diabetes, and related metabolic disorders are major health issues worldwide. As the epidemic of metabolic disorders continues, the associated medical co-morbidities, including the detrimental impact on reproduction, increase as well. Emerging evidence suggests that the effects of maternal nutrition on reproductive outcomes are likely to be mediated, at least in part, by oocyte metabolism. Well-balanced and timed energy metabolism is critical for optimal development of oocytes. To date, much of our understanding of oocyte metabolism comes from the effects of extrinsic nutrients on oocyte maturation. In contrast, intrinsic regulation of oocyte development by metabolic enzymes, intracellular mediators, and transport systems is less characterized. Specifically, decreased acid transport proteins levels, increased glucose/lipid content and elevated reactive oxygen species in oocytes have been implicated in meiotic defects, organelle dysfunction and epigenetic alteration. Therefore, metabolic disturbances in oocytes may contribute to the diminished reproductive potential experienced by women with metabolic disorders. In-depth research is needed to further explore the underlying mechanisms. This review also discusses several approaches for metabolic analysis. Metabolomic profiling of oocytes, the surrounding granulosa cells, and follicular fluid will uncover the metabolic networks regulating oocyte development, potentially leading to the identification of oocyte quality markers and prevention of reproductive disease and poor outcomes in offspring.
Collapse
Affiliation(s)
- Ling Gu
- College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, Jiangsu, China,
| | | | | | | | | | | |
Collapse
|
852
|
Burgio E, Lopomo A, Migliore L. Obesity and diabetes: from genetics to epigenetics. Mol Biol Rep 2014; 42:799-818. [DOI: 10.1007/s11033-014-3751-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
853
|
Mishra P, Chan DC. Mitochondrial dynamics and inheritance during cell division, development and disease. Nat Rev Mol Cell Biol 2014; 15:634-46. [PMID: 25237825 DOI: 10.1038/nrm3877] [Citation(s) in RCA: 772] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During cell division, it is critical to properly partition functional sets of organelles to each daughter cell. The partitioning of mitochondria shares some common features with that of other organelles, particularly in the use of interactions with cytoskeletal elements to facilitate delivery to the daughter cells. However, mitochondria have unique features - including their own genome and a maternal mode of germline transmission - that place additional demands on this process. Consequently, mechanisms have evolved to regulate mitochondrial segregation during cell division, oogenesis, fertilization and tissue development, as well as to ensure the integrity of these organelles and their DNA, including fusion-fission dynamics, organelle transport, mitophagy and genetic selection of functional genomes. Defects in these processes can lead to cell and tissue pathologies.
Collapse
Affiliation(s)
- Prashant Mishra
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - David C Chan
- 1] Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA. [2] Howard Hughes Medical Institute, California Institute of Technology, Pasadena, California 91125, USA
| |
Collapse
|
854
|
Global genomic and transcriptomic analysis of human pancreatic islets reveals novel genes influencing glucose metabolism. Proc Natl Acad Sci U S A 2014; 111:13924-9. [PMID: 25201977 DOI: 10.1073/pnas.1402665111] [Citation(s) in RCA: 352] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Genetic variation can modulate gene expression, and thereby phenotypic variation and susceptibility to complex diseases such as type 2 diabetes (T2D). Here we harnessed the potential of DNA and RNA sequencing in human pancreatic islets from 89 deceased donors to identify genes of potential importance in the pathogenesis of T2D. We present a catalog of genetic variants regulating gene expression (eQTL) and exon use (sQTL), including many long noncoding RNAs, which are enriched in known T2D-associated loci. Of 35 eQTL genes, whose expression differed between normoglycemic and hyperglycemic individuals, siRNA of tetraspanin 33 (TSPAN33), 5'-nucleotidase, ecto (NT5E), transmembrane emp24 protein transport domain containing 6 (TMED6), and p21 protein activated kinase 7 (PAK7) in INS1 cells resulted in reduced glucose-stimulated insulin secretion. In addition, we provide a genome-wide catalog of allelic expression imbalance, which is also enriched in known T2D-associated loci. Notably, allelic imbalance in paternally expressed gene 3 (PEG3) was associated with its promoter methylation and T2D status. Finally, RNA editing events were less common in islets than previously suggested in other tissues. Taken together, this study provides new insights into the complexity of gene regulation in human pancreatic islets and better understanding of how genetic variation can influence glucose metabolism.
Collapse
|
855
|
El Hajj N, Schneider E, Lehnen H, Haaf T. Epigenetics and life-long consequences of an adverse nutritional and diabetic intrauterine environment. Reproduction 2014; 148:R111-20. [PMID: 25187623 PMCID: PMC4241689 DOI: 10.1530/rep-14-0334] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The phenomenon that adverse environmental exposures in early life are associated with increased susceptibilities for many adult, particularly metabolic diseases, is now referred to as ‘developmental origins of health and disease (DOHAD)’ or ‘Barker’ hypothesis. Fetal overnutrition and undernutrition have similar long-lasting effects on the setting of the neuroendocrine control systems, energy homeostasis, and metabolism, leading to life-long increased morbidity. There are sensitive time windows during early development, where environmental cues can program persistent epigenetic modifications which are generally assumed to mediate these gene–environment interactions. Most of our current knowledge on fetal programing comes from animal models and epidemiological studies in humans, in particular the Dutch famine birth cohort. In industrialized countries, there is more concern about adverse long-term consequences of fetal overnutrition, i.e. by exposure to gestational diabetes mellitus and/or maternal obesity which affect 10–20% of pregnancies. Epigenetic changes due to maternal diabetes/obesity may predispose the offspring to develop metabolic disease later in life and, thus, transmit the adverse environmental exposure to the next generation. This vicious cycle could contribute significantly to the worldwide metabolic disease epidemics. In this review article, we focus on the epigenetics of an adverse intrauterine environment, in particular gestational diabetes, and its implications for the prevention of complex disease.
Collapse
Affiliation(s)
- Nady El Hajj
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| | - Eberhard Schneider
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| | - Harald Lehnen
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| | - Thomas Haaf
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| |
Collapse
|
856
|
Ma JY, Zhang T, Shen W, Schatten H, Sun QY. Molecules and mechanisms controlling the active DNA demethylation of the mammalian zygotic genome. Protein Cell 2014; 5:827-36. [PMID: 25152302 PMCID: PMC4225482 DOI: 10.1007/s13238-014-0095-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 07/21/2014] [Indexed: 12/02/2022] Open
Abstract
The active DNA demethylation in early embryos is essential for subsequent development. Although the zygotic genome is globally demethylated, the DNA methylation of imprinted regions, part of repeat sequences and some gamete-specific regions are maintained. Recent evidence has shown that multiple proteins and biological pathways participate in the regulation of active DNA demethylation, such as TET proteins, DNA repair pathways and DNA methyltransferases. Here we review the recent understanding regarding proteins associated with active DNA demethylation and the regulatory networks controlling the active DNA demethylation in early embryos.
Collapse
Affiliation(s)
- Jun-Yu Ma
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, China
| | | | | | | | | |
Collapse
|
857
|
Majnik A, Gunn V, Fu Q, Lane RH. Epigenetics: an accessible mechanism through which to track and respond to an obesogenic environment. Expert Rev Endocrinol Metab 2014; 9:605-614. [PMID: 30736198 DOI: 10.1586/17446651.2014.949241] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Obesity and its consequences impact everyone. Obesity occurs because of an interaction between an obesogenic environment and genetics. In order to confront obesity, we must understand the contribution of each of these components. Environmental influences on obesity include our extrinsic environment, such as food deserts, as well as our intrinsic environment, like perinatal exposures. Epigenetics provides a biological mechanism to reveal the accumulation of extrinsic and intrinsic environmental exposures from fetal life to adulthood. Human and animal studies demonstrate changes in epigenetic modifications which are associated with an obesogenic environment. Furthermore, evidence exists in humans and animal models that suggest environmental epigenetics may serve as a biomarker or a target for intervention. To successfully target obesity, we must intervene on an environmental as well as genetic level. Combating food deserts for example will help to change the extrinsic environment, while targeting epigenetic modification remains a goal for changing our biology.
Collapse
Affiliation(s)
- Amber Majnik
- a Medical College of Wisconsin, 8701 Watertown Plank Rd, TBRC-CRI C2485, Milwaukee WI 53226, USA
| | - Veronica Gunn
- b Children's Hospital of Wisconsin, Children's Corporate Center, Suite 525, PO Box 1997, Milwaukee, WI 53201-1997, USA
| | - Qi Fu
- a Medical College of Wisconsin, 8701 Watertown Plank Rd, TBRC-CRI C2485, Milwaukee WI 53226, USA
| | - Robert H Lane
- b Children's Hospital of Wisconsin, Children's Corporate Center, Suite 525, PO Box 1997, Milwaukee, WI 53201-1997, USA
| |
Collapse
|
858
|
Stricker SA. Calcium signaling and endoplasmic reticulum dynamics during fertilization in marine protostome worms belonging to the phylum Nemertea. Biochem Biophys Res Commun 2014; 450:1182-7. [DOI: 10.1016/j.bbrc.2014.03.156] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 03/31/2014] [Indexed: 10/25/2022]
|
859
|
O'Doherty AM, O'Gorman A, al Naib A, Brennan L, Daly E, Duffy P, Fair T. Negative energy balance affects imprint stability in oocytes recovered from postpartum dairy cows. Genomics 2014; 104:177-85. [PMID: 25084396 DOI: 10.1016/j.ygeno.2014.07.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 12/27/2022]
Abstract
Ovarian follicle development in post-partum, high-producing dairy cows, occurs in a compromised endogenous metabolic environment (referred to as negative energy balance, NEB). Key events that occur during oocyte/follicle growth, such as the vital process of genomic imprinting, may be detrimentally affected by this altered ovarian environment. Imprinting is crucial for placental function and regulation of fetal growth, therefore failure to establish and maintain imprints during oocyte growth may contribute to early embryonic loss. Using ovum pick-up (OPU), oocytes and follicular fluid samples were recovered from cows between days 20 and 115 post-calving, encompassing the NEB period. In a complimentary study, cumulus oocyte complexes were in vitro matured under high non-esterified fatty acid (NEFA) concentrations and in the presence of the methyl-donor S-adenosylmethionine (SAM). Pyrosequencing revealed the loss of methylation at several imprinted loci in the OPU derived oocytes. The loss of DNA methylation was observed at the PLAGL1 locus in oocytes, following in vitro maturation (IVM) in the presence of elevated NEFAs and SAM. Finally, metabolomic analysis of postpartum follicular fluid samples revealed significant differences in several branched chain amino acids, with fatty acid profiles bearing similarities to those characteristic of lactating dairy cows. These results provide the first evidence that (1) the postpartum ovarian environment may affect maternal imprint acquisition and (2) elevated NEFAs during IVM can lead to the loss of imprinted gene methylation in bovine oocytes.
Collapse
Affiliation(s)
- Alan M O'Doherty
- School of Agriculture & Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Aoife O'Gorman
- School of Agriculture & Food Science, University College Dublin, Belfield, Dublin 4, Ireland; Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Abdullah al Naib
- Lyons Research Farm, University College Dublin, Belfield, Dublin 4, Ireland
| | - Lorraine Brennan
- School of Agriculture & Food Science, University College Dublin, Belfield, Dublin 4, Ireland; Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Edward Daly
- Lyons Research Farm, University College Dublin, Belfield, Dublin 4, Ireland
| | - Pat Duffy
- Lyons Research Farm, University College Dublin, Belfield, Dublin 4, Ireland
| | - Trudee Fair
- School of Agriculture & Food Science, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
860
|
Politi Y, Gal L, Kalifa Y, Ravid L, Elazar Z, Arama E. Paternal mitochondrial destruction after fertilization is mediated by a common endocytic and autophagic pathway in Drosophila. Dev Cell 2014; 29:305-20. [PMID: 24823375 DOI: 10.1016/j.devcel.2014.04.005] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 01/16/2014] [Accepted: 04/02/2014] [Indexed: 12/12/2022]
Abstract
Almost all animals contain mitochondria of maternal origin only, but the exact mechanisms underlying this phenomenon are still vague. We investigated the fate of Drosophila paternal mitochondria after fertilization. We demonstrate that the sperm mitochondrial derivative (MD) is rapidly eliminated in a stereotypical process dubbed paternal mitochondrial destruction (PMD). PMD is initiated by a network of vesicles resembling multivesicular bodies and displaying common features of the endocytic and autophagic pathways. These vesicles associate with the sperm tail and mediate the disintegration of its plasma membrane. Subsequently, the MD separates from the axoneme and breaks into smaller fragments, which are then sequestered by autophagosomes for degradation in lysosomes. We further provide evidence for the involvement of the ubiquitin pathway and the autophagy receptor p62 in this process. Finally, we show that the ubiquitin ligase Parkin is not involved in PMD, implying a divergence from the autophagic pathway of damaged mitochondria.
Collapse
Affiliation(s)
- Yoav Politi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Liron Gal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yossi Kalifa
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Liat Ravid
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Zvulun Elazar
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Eli Arama
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
861
|
Chaffin CL, Latham KE, Mtango NR, Midic U, VandeVoort CA. Dietary sugar in healthy female primates perturbs oocyte maturation and in vitro preimplantation embryo development. Endocrinology 2014; 155:2688-95. [PMID: 24731100 PMCID: PMC4060180 DOI: 10.1210/en.2014-1104] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The consumption of refined sugars continues to pose a significant health risk. However, nearly nothing is known about the effects of sugar intake by healthy women on the oocyte or embryo. Using rhesus monkeys, we show that low-dose sucrose intake over a 6-month period has an impact on the oocyte with subsequent effects on the early embryo. The ability of oocytes to resume meiosis was significantly impaired, although the differentiation of the somatic component of the ovarian follicle into progesterone-producing cells was not altered. Although the small subset of oocytes that did mature were able to be fertilized in vitro and develop into preimplantation blastocysts, there were >1100 changes in blastocyst gene expression. Because sucrose treatment ended before fertilization, the effects of sugar intake by healthy primates are concluded to be epigenetic modifications to the immature oocyte that are manifest in the preimplantation embryo.
Collapse
Affiliation(s)
- Charles L Chaffin
- Department of Obstetrics, Gynecology, and Reproductive Sciences (C.L.C.), University of Maryland School of Medicine, Baltimore, Maryland 21210; Department of Animal Science (K.E.L., U.M.), Michigan State University, East Lansing, Michigan 48824; The Fels Institute for Cancer Research and Molecular Biology and Department of Biochemistry (N.R.M.), Temple University School of Medicine, Philadelphia, Pennsylvania 19140; and California National Primate Research Center and Department of Obstetrics and Gynecology (C.A.V.), University of California, Davis, California 95616
| | | | | | | | | |
Collapse
|
862
|
Lu CL, Wang TR, Yan LY, Xia X, Zhu XH, Li R, Zhao HC, Yan J, Yin TL, Jin HY, Zhang Y, Zhang WX, Feng HL, Qiao J. Gonadotropin-mediated dynamic alterations during bovine oocyte maturation in vitro. Biol Reprod 2014; 91:44. [PMID: 24943039 DOI: 10.1095/biolreprod.114.117945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Gonadotropins have been widely used in human-assisted reproduction and animal science for the past four decades. However, the effects of gonadotropins on oocyte maturation at the molecular and biochemical levels are poorly understood. To determine the effects of gonadotropins (recombinant follicle stimulating hormone and urinary human menopausal gonadotropin) on oocyte maturation, we used the bovine oocyte in vitro maturation model. First, we studied the effects of increasing gonadotropin concentrations on nuclear maturation and mitochondrial function in oocytes. Gonadotropins at concentrations of 0.075 and 0.75 IU/ml improved nuclear maturation and increased inner mitochondrial membrane potential and ATP levels; however, there were no beneficial effects at concentrations of 7.5 and 75 IU/ml. Second, we studied the effects of increasing gonadotropin concentrations on the status of methylation in matured (MII) oocytes. Aberrant methylation and demethylation of H19, SNRPN, and PEG3 genes were observed in MII oocytes at all concentrations except 0.075 IU/ml. The expression of genes that function in spindle formation, cell cycle control, and methylation was also downregulated by high gonadotropin concentrations. In conclusion, we established the optimal gonadotropin concentration (i.e., 0.075 IU/ml) to be used for bovine oocyte in vitro maturation studies. These results may provide a guide for clinical stimulation protocols and help to reduce the risks associated with gonadotropin administration during in vitro fertilization treatment.
Collapse
Affiliation(s)
- Cui-Ling Lu
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Tian-Ren Wang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Li-Ying Yan
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Xi Xia
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Xiao-Hui Zhu
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Rong Li
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Hong-Cui Zhao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Jie Yan
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Tai-Lang Yin
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Hong-Yan Jin
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Wen-Xin Zhang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Huai-Liang Feng
- Department of Obstetrics and Gynecology, New York Hospital Queens-affiliated Weill Medical College of Cornell University, New York, New York
| | - Jie Qiao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| |
Collapse
|
863
|
Regulation of mitochondrial genome inheritance by autophagy and ubiquitin-proteasome system: implications for health, fitness, and fertility. BIOMED RESEARCH INTERNATIONAL 2014; 2014:981867. [PMID: 25028670 PMCID: PMC4083708 DOI: 10.1155/2014/981867] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 04/01/2014] [Accepted: 04/07/2014] [Indexed: 01/07/2023]
Abstract
Mitochondria, the energy-generating organelles, play a role in numerous cellular functions including adenosine triphosphate (ATP) production, cellular homeostasis, and apoptosis. Maternal inheritance of mitochondria and mitochondrial DNA (mtDNA) is universally observed in humans and most animals. In general, high levels of mitochondrial heteroplasmy might contribute to a detrimental effect on fitness and disease resistance. Therefore, a disposal of the sperm-derived mitochondria inside fertilized oocytes assures normal preimplantation embryo development. Here we summarize the current research and knowledge concerning the role of autophagic pathway and ubiquitin-proteasome-dependent proteolysis in sperm mitophagy in mammals, including humans. Current data indicate that sperm mitophagy inside the fertilized oocyte could occur along multiple degradation routes converging on autophagic clearance of paternal mitochondria. The influence of assisted reproductive therapies (ART) such as intracytoplasmic sperm injection (ICSI), mitochondrial replacement (MR), and assisted fertilization of oocytes from patients of advanced reproductive age on mitochondrial function, inheritance, and fitness and for the development and health of ART babies will be of particular interest to clinical audiences. Altogether, the study of sperm mitophagy after fertilization has implications in the timing of evolution and developmental and reproductive biology and in human health, fitness, and management of mitochondrial disease.
Collapse
|
864
|
Hu MW, Wang ZB, Jiang ZZ, Qi ST, Huang L, Liang QX, Schatten H, Sun QY. Scaffold subunit Aalpha of PP2A is essential for female meiosis and fertility in mice. Biol Reprod 2014; 91:19. [PMID: 24899574 DOI: 10.1095/biolreprod.114.120220] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Ppp2r1a encodes the scaffold subunit Aalpha of protein phosphatase 2A (PP2A), which is an important and ubiquitously expressed serine threonine phosphatase family and plays a critical role in many fundamental cellular processes. To identify the physiological role of PP2A in female germ cell meiosis, we selectively disrupted Ppp2r1a expression in oocytes by using the Cre-Loxp conditional knockout system. Here we report for the first time that oocyte-specific deletion of Ppp2r1a led to severe female subfertility without affecting follicle survival, growth, and ovulation. PP2A-Aalpha was essential for regulating oocyte meiotic maturation because depletion of PP2A-Aalpha facilitated germinal vesicle breakdown, causing elongation of the MII spindle and precocious separation of sister chromatids. The resulting eggs had high risk of aneuploidy, though they could be fertilized, leading to defective embryonic development and thus subfertility. Our findings provide strong evidence that PP2A-Aalpha within the oocyte plays an indispensable role in oocyte meiotic maturation, though it is dispensable for folliculogenesis in the mouse ovary.
Collapse
Affiliation(s)
- Meng-Wen Hu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China University of Chinese Academy of Sciences, Beijing, China
| | - Zhen-Bo Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zong-Zhe Jiang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China University of Chinese Academy of Sciences, Beijing, China
| | - Shu-Tao Qi
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lin Huang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qiu-Xia Liang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China University of Chinese Academy of Sciences, Beijing, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
865
|
Ge ZJ, Zhang CL, Schatten H, Sun QY. Maternal diabetes mellitus and the origin of non-communicable diseases in offspring: the role of epigenetics. Biol Reprod 2014; 90:139. [PMID: 24829025 DOI: 10.1095/biolreprod.114.118141] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025] Open
Abstract
Offspring of diabetic mothers are susceptible to the onset of metabolic syndromes, such as type 2 diabetes and obesity at adulthood, and this trend can be inherited between generations. Genetics cannot fully explain how the noncommunicable disease in offspring of diabetic mothers is caused and inherited by the next generations. Many studies have confirmed that epigenetics may be crucial for the detrimental effects on offspring exposed to the hyperglycemic environment. Although the adverse effects on epigenetics in offspring of diabetic mothers may be the result of the poor intrauterine environment, epigenetic modifications in oocytes of diabetic mothers are also affected. Therefore, the present review is focused on the epigenetic alterations in oocytes and embryos of diabetic mothers. Furthermore, we also discuss initial mechanistic insight on maternal diabetes mellitus causing alterations of epigenetic modifications.
Collapse
Affiliation(s)
- Zhao-Jia Ge
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China Reproductive Medicine Center, Henan Provincial People's Hospital, Zhengzhou, Henan Province, P.R. China Reproductive Medicine Center, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, P.R. China
| | - Cui-Lian Zhang
- Reproductive Medicine Center, Henan Provincial People's Hospital, Zhengzhou, Henan Province, P.R. China Reproductive Medicine Center, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, P.R. China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China
| |
Collapse
|
866
|
Role of autophagy in embryogenesis. Curr Opin Genet Dev 2014; 27:60-6. [PMID: 24861852 DOI: 10.1016/j.gde.2014.03.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/21/2014] [Accepted: 03/21/2014] [Indexed: 01/08/2023]
Abstract
Eukaryotes have evolved multiple mechanisms for inactivating macromolecules in order to maintain their functionality. Autophagy-the process of self-eating-leads to the degradation of cytoplasmic components for the dynamic remodeling of subcellular compartments, turnover and recycling of macromolecules, and regulation of cellular activity through the control of specific intracellular signaling pathways. This fundamental process is also implicated in systemic response to starvation and immune challenges, as well as anti-tumorigenesis and anti-senescence. Recent studies have also highlighted an important role for autophagy in embryonic development. In this review, we discuss the emerging evidence for the varied functions of autophagy at different stages of development, with an emphasis on the early events of embryogenesis.
Collapse
|
867
|
Molecular insights into NF2/Merlin tumor suppressor function. FEBS Lett 2014; 588:2743-52. [PMID: 24726726 DOI: 10.1016/j.febslet.2014.04.001] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/01/2014] [Accepted: 04/02/2014] [Indexed: 02/07/2023]
Abstract
The FERM domain protein Merlin, encoded by the NF2 tumor suppressor gene, regulates cell proliferation in response to adhesive signaling. The growth inhibitory function of Merlin is induced by intercellular adhesion and inactivated by joint integrin/receptor tyrosine kinase signaling. Merlin contributes to the formation of cell junctions in polarized tissues, activates anti-mitogenic signaling at tight-junctions, and inhibits oncogenic gene expression. Thus, inactivation of Merlin causes uncontrolled mitogenic signaling and tumorigenesis. Merlin's predominant tumor suppressive functions are attributable to its control of oncogenic gene expression through regulation of Hippo signaling. Notably, Merlin translocates to the nucleus where it directly inhibits the CRL4(DCAF1) E3 ubiquitin ligase, thereby suppressing inhibition of the Lats kinases. A dichotomy in NF2 function has emerged whereby Merlin acts at the cell cortex to organize cell junctions and propagate anti-mitogenic signaling, whereas it inhibits oncogenic gene expression through the inhibition of CRL4(DCAF1) and activation of Hippo signaling. The biochemical events underlying Merlin's normal function and tumor suppressive activity will be discussed in this Review, with emphasis on recent discoveries that have greatly influenced our understanding of Merlin biology.
Collapse
|
868
|
Ge ZJ, Liang QX, Hou Y, Han ZM, Schatten H, Sun QY, Zhang CL. Maternal obesity and diabetes may cause DNA methylation alteration in the spermatozoa of offspring in mice. Reprod Biol Endocrinol 2014; 12:29. [PMID: 24721882 PMCID: PMC3984639 DOI: 10.1186/1477-7827-12-29] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 04/03/2014] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The adverse effects on offspring of diabetic and/or obese mothers can be passed to the next generation. However, the mechanisms behind this are still unclear. Epigenetics may play a key role during this process. METHODS To confirm the hypothesis, we investigated the DNA methylation of several imprinted genes in spermatozoa of offspring from diabetic and/or obese mothers utilizing streptozotocin (STZ)- and high-fat-diet (HFD)-induced mouse models. RESULTS We found that the DNA methylation of Peg3 was significantly increased in spermatozoa of offspring of obese mothers compared to that in spermatozoa of offspring of normal mothers. The DNA methylation of H19 was significantly higher in spermatozoa of offspring of diabetic mothers than that in spermatozoa of offspring of non-diabetic mothers. CONCLUSIONS These results indicate that pre-gestational diabetes and/or obesity can alter DNA methylation in offspring spermatozoa.
Collapse
Affiliation(s)
- Zhao-Jia Ge
- Reproductive Medicine Center, Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, P.R. China
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China
- Reproductive Medicine Center, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan province, P.R. China
| | - Qiu-Xia Liang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Yi Hou
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Zhi-Ming Han
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, 65211 Columbia, MO, USA
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Cui-Lian Zhang
- Reproductive Medicine Center, Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, P.R. China
- Reproductive Medicine Center, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan province, P.R. China
| |
Collapse
|
869
|
Liew SH, Vaithiyanathan K, Cook M, Bouillet P, Scott CL, Kerr JB, Strasser A, Findlay JK, Hutt KJ. Loss of the proapoptotic BH3-only protein BCL-2 modifying factor prolongs the fertile life span in female mice. Biol Reprod 2014; 90:77. [PMID: 24571986 DOI: 10.1095/biolreprod.113.116947] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The duration of the female fertile life span is influenced by the number of oocytes stored in the ovary as primordial follicles. Cell death, both during ovarian development in the embryo and in the postnatal ovary, plays a critical role in determining how many primordial follicles are established and maintained within the ovary. However, the roles of individual apoptotic regulators in mediating cell death within the ovary have not yet been characterized. In this study, gene targeted mice were used to investigate the role of BCL-2-modifying factor (BMF), a proapoptotic protein belonging to the BH3-only subgroup of the BCL-2 family, in determining the number of primordial follicles maintained in the adult ovary and the length of the fertile life span. Stereological analysis of ovaries showed that Bmf(-/-) mice had significantly more primordial follicles than wild-type (WT) control animals at Postnatal Days 100, 200, 300, and 400 but not at Day 20. No differences were observed between WT and Bmf(-/-) mice in the number of ova shed following ovulatory stimulation with exogenous gonadotropins. Bmf(-/-) females were fertile and produced the same number pups/litter as WT females, but Bmf(-/-) females produced litters more frequently and consequently more offspring than WT females over a 6-mo period. Furthermore, the fertile life span of Bmf(-/-) females was significantly extended compared to WT females. Our findings support an important role for BMF in determining the number of primordial follicles maintained in the ovary throughout adult reproductive life and thus indicate that the length of female fertility may be extended by increasing the number of primordial follicles maintained within the ovary through inhibition of BMF.
Collapse
Affiliation(s)
- Seng H Liew
- MIMR-PHI Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
870
|
Proteomes of animal oocytes: what can we learn for human oocytes in the in vitro fertilization programme? BIOMED RESEARCH INTERNATIONAL 2014; 2014:856907. [PMID: 24804254 PMCID: PMC3996292 DOI: 10.1155/2014/856907] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 02/13/2014] [Indexed: 12/18/2022]
Abstract
Oocytes are crucial cells for mammalian reproduction, yet the molecular principles underlying oocyte development are only partially understood. Therefore, contemporary proteomic approaches have been used increasingly to provide new insights into oocyte quality and maturation in various species such as mouse, pig, and cow. Especially, animal studies have helped in elucidating the molecular status of oocytes during in vitro maturation and other procedures of assisted reproduction. The aim of this review is to summarize the literature on mammalian oocyte proteome and secretome research in the light of natural and assisted reproduction and on lessons to be learned for human oocytes, which have so far remained inaccessible for proteome analysis.
Collapse
|
871
|
Green DR, Levine B. To be or not to be? How selective autophagy and cell death govern cell fate. Cell 2014; 157:65-75. [PMID: 24679527 PMCID: PMC4020175 DOI: 10.1016/j.cell.2014.02.049] [Citation(s) in RCA: 554] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/25/2014] [Accepted: 02/25/2014] [Indexed: 12/15/2022]
Abstract
The health of metazoan organisms requires an effective response to organellar and cellular damage either by repair of such damage and/or by elimination of the damaged parts of the cells or the damaged cell in its entirety. Here, we consider the progress that has been made in the last few decades in determining the fates of damaged organelles and damaged cells through discrete, but genetically overlapping, pathways involving the selective autophagy and cell death machinery. We further discuss the ways in which the autophagy machinery may impact the clearance and consequences of dying cells for host physiology. Failure in the proper removal of damaged organelles and/or damaged cells by selective autophagy and cell death processes is likely to contribute to developmental abnormalities, cancer, aging, inflammation, and other diseases.
Collapse
Affiliation(s)
- Douglas R Green
- Department of Immunology, St. Jude's Children's Research Hospital, Memphis, TN 38205, USA.
| | - Beth Levine
- Center for Autophagy Research, Department of Internal Medicine, Department of Microbiology and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
872
|
Escobar-Henriques M, Langer T. Dynamic survey of mitochondria by ubiquitin. EMBO Rep 2014; 15:231-43. [PMID: 24569520 PMCID: PMC3989689 DOI: 10.1002/embr.201338225] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 01/16/2014] [Accepted: 01/20/2014] [Indexed: 12/27/2022] Open
Abstract
Ubiquitin is a post-translational modifier with proteolytic and non-proteolytic roles in many biological processes. At mitochondria, it performs regulatory homeostatic functions and contributes to mitochondrial quality control. Ubiquitin is essential for mitochondrial fusion, regulates mitochondria-ER contacts, and participates in maternal mtDNA inheritance. Under stress, mitochondrial dysfunction induces ubiquitin-dependent responses that involve mitochondrial proteome remodeling and culminate in organelle removal by mitophagy. In addition, many ubiquitin-dependent mechanisms have been shown to regulate innate immune responses and xenophagy. Here, we review the emerging roles of ubiquitin at mitochondria.
Collapse
Affiliation(s)
- Mafalda Escobar-Henriques
- Institute for Genetics, Centre for Molecular Medicine (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of CologneCologne, Germany
| | - Thomas Langer
- Institute for Genetics, Centre for Molecular Medicine (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of CologneCologne, Germany
- Max-Planck-Institute for the Biology of AgingCologne, Germany
| |
Collapse
|
873
|
Ge ZJ, Luo SM, Lin F, Liang QX, Huang L, Wei YC, Hou Y, Han ZM, Schatten H, Sun QY. DNA methylation in oocytes and liver of female mice and their offspring: effects of high-fat-diet-induced obesity. ENVIRONMENTAL HEALTH PERSPECTIVES 2014; 122:159-164. [PMID: 24316659 PMCID: PMC3915265 DOI: 10.1289/ehp.1307047] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 12/05/2013] [Indexed: 05/25/2023]
Abstract
BACKGROUND Maternal obesity has adverse effects on oocyte quality, embryo development, and the health of the offspring. OBJECTIVES To understand the underlying mechanisms responsible for the negative effects of maternal obesity, we investigated the DNA methylation status of several imprinted genes and metabolism-related genes. METHODS Using a high-fat-diet (HFD)-induced mouse model of obesity, we analyzed the DNA methylation of several imprinted genes and metabolism-related genes in oocytes from control and obese dams and in oocytes and liver from their offspring. Analysis was performed using combined bisulfite restriction analysis (COBRA) and bisulfite sequencing. RESULTS DNA methylation of imprinted genes in oocytes was not altered in either obese dams or their offspring; however, DNA methylation of metabolism-related genes was changed. In oocytes of obese mice, the DNA methylation level of the leptin (Lep) promoter was significantly increased and that of the Ppar-α promoter was reduced. Increased methylation of Lep and decreased methylation of Ppar-α was also observed in the liver of female offspring from dams fed the high-fat diet (OHFD). mRNA expression of Lep and Ppar-α was also significantly altered in the liver of these OHFD. In OHFD oocytes, the DNA methylation level of Ppar-α promoter was increased. CONCLUSIONS Our results indicate that DNA methylation patterns of several metabolism-related genes are changed not only in oocytes of obese mice but also in oocytes and liver of their offspring. These data may contribute to the understanding of adverse effects of maternal obesity on reproduction and health of the offspring.
Collapse
Affiliation(s)
- Zhao-Jia Ge
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
874
|
Frank LA, Sutton-McDowall ML, Gilchrist RB, Thompson JG. The effect of peri-conception hyperglycaemia and the involvement of the hexosamine biosynthesis pathway in mediating oocyte and embryo developmental competence. Mol Reprod Dev 2014; 81:391-408. [DOI: 10.1002/mrd.22299] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 12/31/2013] [Indexed: 12/12/2022]
Affiliation(s)
- Laura A. Frank
- The Robinson Institute, The Research Centre for Reproductive Health, School of Paediatrics and Reproductive Health, The University of Adelaide; Adelaide South Australia Australia
| | - Melanie L. Sutton-McDowall
- The Robinson Institute, The Research Centre for Reproductive Health, School of Paediatrics and Reproductive Health, The University of Adelaide; Adelaide South Australia Australia
| | - Robert B. Gilchrist
- The Robinson Institute, The Research Centre for Reproductive Health, School of Paediatrics and Reproductive Health, The University of Adelaide; Adelaide South Australia Australia
| | - Jeremy G. Thompson
- The Robinson Institute, The Research Centre for Reproductive Health, School of Paediatrics and Reproductive Health, The University of Adelaide; Adelaide South Australia Australia
| |
Collapse
|
875
|
Taylor EM, Jones AD, Henagan TM. A Review of Mitochondrial-derived Fatty Acids in Epigenetic Regulation of Obesity and Type 2 Diabetes. ACTA ACUST UNITED AC 2014; 2:1-4. [PMID: 25364776 DOI: 10.15226/jnhfs.2014.00127] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Type 2 diabetes, the leading metabolic disease, is characterized by insulin resistance and is associated with obesity. The onset of type 2 diabetes is largely due to environmental inputs, such as high dietary fat content and decreased levels of exercise. Insulin resistance resulting from high fat diet is associated with skeletal muscle mitochondrial dysfunction, leading to alterations in lipid accumulation and specific species of intracellular fatty acids; whereas, exercise training augments insulin resistance while improving skeletal muscle mitochondrial function and producing beneficial fatty acid profiles. Additionally, high fat diets and exercise alter epigenetic modifications, including DNA methylation and histone acetylation, to produce differences in metabolic gene expression that are associated with insulin resistance and sensitivity, respectively. Recent evidence suggests that short chain fatty acids that act as histone deacetylase inhibitors prevent and ameliorate obesity and insulin resistance. Here, we discuss the potential of mitochondrial-derived fatty acids, especially short chain fatty acids, to epigenetically regulate obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Erin M Taylor
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| | - Aarin D Jones
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| | - Tara M Henagan
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
876
|
Cecconi S, Rossi G, Deldar H, Cellini V, Patacchiola F, Carta G, Macchiarelli G, Canipari R. Post-ovulatory ageing of mouse oocytes affects the distribution of specific spindle-associated proteins and Akt expression levels. Reprod Fertil Dev 2014; 26:562-9. [PMID: 23622715 DOI: 10.1071/rd13010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 03/20/2013] [Indexed: 12/16/2023] Open
Abstract
The aim of this study has been to determine the effects of in vivo post-ovulatory ageing (POA) on the distribution of spindle-associated proteins, histone H3/H4 post-translational modifications and on v-akt murine thymoma viral oncogene homolog 1 (Akt) expression levels. To this end, oocytes were retrieved 13, 29 and 33h after human chorionic gonadotrophin (hCG) treatment. The presence and distribution at the meiotic spindle of acetylated tubulin, γ-tubulin, polo kinase-1 and Ser473/Thr308 phosphorylated Akt (pAkt) as well as histone H3 and H4 acetylation and phosphorylation levels were assayed via immunofluorescence. Akt expression levels were determined via reverse transcription-polymerase chain reaction and western blotting analyses. Spindles from oocytes recovered 13h and 29h after hCG treatment showed similar levels of acetylated tubulin but ageing induced: (1) translocation of γ-tubulin from spindle poles to microtubules, (2) absence of Thr308- and Ser473-pAkt in 76% and 30% of oocytes, respectively, and (3) a significant reduction in phosphorylation levels of serine 10 on histone 3. At 29h, a significant decrease in Akt mRNA, but not in pAkt or Akt protein levels, was recorded. By contrast, protein content significantly decreased 33h after hCG. We conclude that POA impairs oocyte viability and fertilisability by altering the expression levels and spindle distribution of proteins that are implicated in cell survival and chromosome segregation. Together, these events could play a role in oocyte apoptosis.
Collapse
Affiliation(s)
- Sandra Cecconi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Piazza S. Tommasi, Coppito, 67100 L'Aquila, Italy
| | - Gianna Rossi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Piazza S. Tommasi, Coppito, 67100 L'Aquila, Italy
| | - Hamid Deldar
- Department of Animal Science, College of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Resources University, P.O. Box 578, Sari, Iran
| | - Valerio Cellini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Piazza S. Tommasi, Coppito, 67100 L'Aquila, Italy
| | - Felice Patacchiola
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Piazza S. Tommasi, Coppito, 67100 L'Aquila, Italy
| | - Gaspare Carta
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Piazza S. Tommasi, Coppito, 67100 L'Aquila, Italy
| | - Guido Macchiarelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Piazza S. Tommasi, Coppito, 67100 L'Aquila, Italy
| | - Rita Canipari
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Section of Histology and Embryology, School of Pharmacy and Medicine, 'Sapienza' University of Rome, V.le Regina Elena 324, 00161 Rome, Italy
| |
Collapse
|
877
|
Yang P, Zhang H. You are what you eat: multifaceted functions of autophagy during C. elegans development. Cell Res 2014; 24:80-91. [PMID: 24296782 PMCID: PMC3879703 DOI: 10.1038/cr.2013.154] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Autophagy involves the sequestration of a portion of the cytosolic contents in an enclosed double-membrane autophagosomal structure and its subsequent delivery to lysosomes for degradation. Autophagy activity functions in multiple biological processes during Caenorhabditis elegans development. The basal level of autophagy in embryos removes aggregate-prone proteins, paternal mitochondria and spermatid-specific membranous organelles (MOs). Autophagy also contributes to the efficient removal of embryonic apoptotic cell corpses by promoting phagosome maturation. During larval development, autophagy modulates miRNA-mediated gene silencing by selectively degrading AIN-1, a component of miRNA-induced silencing complex, and thus participates in the specification of multiple cell fates controlled by miRNAs. During development of the hermaphrodite germline, autophagy acts coordinately with the core apoptotic machinery to execute genotoxic stress-induced germline cell death and also cell death when caspase activity is partially compromised. Autophagy is also involved in the utilization of lipid droplets in the aging process in adult animals. Studies in C. elegans provide valuable insights into the physiological functions of autophagy in the development of multicellular organisms.
Collapse
Affiliation(s)
- Peiguo Yang
- State Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China
| | - Hong Zhang
- State Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China
| |
Collapse
|
878
|
Ge ZJ, Liang QX, Luo SM, Wei YC, Han ZM, Schatten H, Sun QY, Zhang CL. Diabetic uterus environment may play a key role in alterations of DNA methylation of several imprinted genes at mid-gestation in mice. Reprod Biol Endocrinol 2013; 11:119. [PMID: 24378208 PMCID: PMC3896855 DOI: 10.1186/1477-7827-11-119] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/26/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Maternal diabetes mellitus not only has severe deleterious effects on fetal development, but also it affects transmission to the next generation. However, the underlying mechanisms for these effects are still not clear. METHODS We investigated the methylation patterns and expressions of the imprinted genes Peg3, Snrpn, and H19 in mid-gestational placental tissues and on the whole fetus utilizing the streptozotocin (STZ)-induced hyperglycemic mouse model for quantitative analysis of methylation by PCR and quantitative real-time PCR. The protein expression of Peg3 was evaluated by Western blot. RESULTS We found that the expression of H19 was significantly increased, while the expression of Peg3 was significantly decreased in dpc10.5 placentas of diabetic mice. We further found that the methylation level of Peg3 was increased and that of H19 was reduced in dpc10.5 placentas of diabetic mice. When pronuclear embryos of normal females were transferred to normal/diabetic (NN/ND) pseudopregnant females, the methylation and expression of Peg3 in placentas was also clearly altered in the ND group compared to the NN group. However, when the pronuclear embryos of diabetic female were transferred to normal pesudopregnant female mice (DN), the methylation and expression of Peg3 and H19 in dpc10.5 placentas was similar between the two groups. CONCLUSIONS We suggest that the effects of maternal diabetes on imprinted genes may primarily be caused by the adverse uterus environment.
Collapse
Affiliation(s)
- Zhao-Jia Ge
- Reproductive Medicine Center, Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, P. R. China
- Reproductive Medicine Center, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, P. R. China
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Qiu-Xia Liang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Shi-Ming Luo
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Yan-Chang Wei
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Zhi-Ming Han
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Cui-Lian Zhang
- Reproductive Medicine Center, Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, P. R. China
| |
Collapse
|
879
|
Wong M, Gertz B, Chestnut BA, Martin LJ. Mitochondrial DNMT3A and DNA methylation in skeletal muscle and CNS of transgenic mouse models of ALS. Front Cell Neurosci 2013; 7:279. [PMID: 24399935 PMCID: PMC3872319 DOI: 10.3389/fncel.2013.00279] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 12/12/2013] [Indexed: 12/13/2022] Open
Abstract
Cytosine methylation is an epigenetic modification of DNA catalyzed by DNA methyltransferases. Cytosine methylation of mitochondrial DNA (mtDNA) is believed to have relative underrepresentation; however, possible tissue and cell differences in mtDNA methylation and relationships to neurodegenerative disease have not been examined. We show by immunoblotting that DNA methyltransferase 3A (Dnmt3a) isoform is present in pure mitochondria of adult mouse CNS, skeletal muscle, and testes, and adult human cerebral cortex. Dnmt1 was not detected in adult mouse CNS or skeletal muscle mitochondria but appeared bound to the outer mitochondrial membrane. Immunofluorescence confirmed the mitochondrial localization of Dnmt3a and showed 5-methylcytosine (5mC) immunoreactivity in mitochondria of neurons and skeletal muscle myofibers. DNA pyrosequencing of two loci (D-loop and 16S rRNA gene) and twelve cytosine-phosphate-guanine (CpG) sites in mtDNA directly showed a tissue differential presence of 5mC. Because mitochondria have been implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS), but the disease mechanisms are uncertain, we evaluated mitochondrial Dnmt3a and 5mC levels in human superoxide dismutase-1 (SOD1) transgenic mouse models of ALS. Mitochondrial Dnmt3a protein levels were reduced significantly in skeletal muscle and spinal cord at presymptomatic or early disease. Immunofluorescence showed that 5mC immunoreactivity was present in mitochondria of neurons and skeletal myofibers, and 5mC immunoreactivity became aggregated in motor neurons of ALS mice. DNA pyrosequencing revealed significant abnormalities in 16S rRNA gene methylation in ALS mice. Immunofluorescence showed that 5mC immunoreactivity can be sequestered into autophagosomes and that mitophagy was increased and mitochondrial content was decreased in skeletal muscle in ALS mice. This study reveals a tissue-preferential mitochondrial localization of Dnmt3a and presence of cytosine methylation in mtDNA of nervous tissue and skeletal muscle and demonstrates that mtDNA methylation patterns and mitochondrial Dnmt3a levels are abnormal in skeletal muscle and spinal cord of presymptomatic ALS mice, and these abnormalities occur in parallel with loss of myofiber mitochondria.
Collapse
Affiliation(s)
- Margaret Wong
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Barry Gertz
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Department of Pathology, Pathobiology Graduate Program, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Barry A Chestnut
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Department of Pathology, Pathobiology Graduate Program, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Lee J Martin
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Department of Pathology, Pathobiology Graduate Program, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Department of Neuroscience, Johns Hopkins University School of Medicine Baltimore, MD, USA
| |
Collapse
|
880
|
Nakamura N. Ubiquitination regulates the morphogenesis and function of sperm organelles. Cells 2013; 2:732-50. [PMID: 24709878 PMCID: PMC3972651 DOI: 10.3390/cells2040732] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 11/12/2013] [Accepted: 11/29/2013] [Indexed: 11/29/2022] Open
Abstract
It is now understood that protein ubiquitination has diverse cellular functions in eukaryotes. The molecular mechanism and physiological significance of ubiquitin-mediated processes have been extensively studied in yeast, Drosophila and mammalian somatic cells. Moreover, an increasing number of studies have emphasized the importance of ubiquitination in spermatogenesis and fertilization. The dysfunction of various ubiquitin systems results in impaired sperm development with abnormal organelle morphology and function, which in turn is highly associated with male infertility. This review will focus on the emerging roles of ubiquitination in biogenesis, function and stability of sperm organelles in mammals.
Collapse
Affiliation(s)
- Nobuhiro Nakamura
- Department of Biological Sciences, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan.
| |
Collapse
|
881
|
BUB1 and BUBR1 inhibition decreases proliferation and colony formation, and enhances radiation sensitivity in pediatric glioblastoma cells. Childs Nerv Syst 2013; 29:2241-8. [PMID: 23728478 DOI: 10.1007/s00381-013-2175-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 05/16/2013] [Indexed: 01/13/2023]
Abstract
PURPOSE Glioblastoma (GBM) is a very aggressive and lethal brain tumor with poor prognosis. Despite new treatment strategies, patients' median survival is still lower than 1 year in most cases. The expression of the BUB gene family has demonstrated to be altered in a variety of solid tumors, pointing to a role as putative therapeutic target. The purpose of this study was to determine BUB1, BUB3, and BUBR1 gene expression profiles in glioblastoma and to analyze the effects of BUB1 and BUBR1 inhibition combined or not with Temozolomide and radiation in the pediatric SF188 GBM cell line. METHODS For gene expression analysis, 8 cell lines and 18 tumor samples were used. The effect of BUB1 and BUBR1 inhibition was evaluated using siRNA. Apoptosis, cell proliferation, cell cycle kinetics, micronuclei formation, and clonogenic capacity were analyzed after BUB1 and BUBR1 inhibition. Additionally, combinatorial effects of gene inhibition and radiation or Temozolomide (TMZ) treatment were evaluated through proliferation and clonogenic capacity assays. RESULTS We report the upregulation of BUB1 and BUBR1 expression and the downregulation of BUB3 in GBM samples and cell lines when compared to white matter samples (p < 0.05). Decreased cell proliferation and colony formation after BUB1 and BUBR1 inhibition were observed, along with increased micronuclei formation. Combinations with TMZ also caused cell cycle arrest and increased apoptosis. Moreover, our results demonstrate that BUB1 and BUBR1 inhibition sensitized SF188 cells to γ-irradiation as shown by decreased growth and abrogation of colony formation capacity. CONCLUSION BUB1 and BUBR1 inhibition decreases proliferation and shows radiosensitizing effects on pediatric GBM cells, which could improve treatment strategies for this devastating tumor. Collectively, these findings highlight the potentials of BUB1 and BUBR1 as putative therapeutic targets for glioblastoma treatment.
Collapse
|
882
|
Qian J, Winkler C, Bollen M. 4D-networking by mitotic phosphatases. Curr Opin Cell Biol 2013; 25:697-703. [DOI: 10.1016/j.ceb.2013.06.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 06/14/2013] [Accepted: 06/18/2013] [Indexed: 01/21/2023]
|
883
|
Luo SM, Schatten H, Sun QY. Sperm Mitochondria in Reproduction: Good or Bad and Where Do They Go? J Genet Genomics 2013; 40:549-56. [DOI: 10.1016/j.jgg.2013.08.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 08/12/2013] [Accepted: 08/27/2013] [Indexed: 11/24/2022]
|
884
|
Soubry A, Murphy SK, Wang F, Huang Z, Vidal AC, Fuemmeler BF, Kurtzberg J, Murtha A, Jirtle RL, Schildkraut JM, Hoyo C. Newborns of obese parents have altered DNA methylation patterns at imprinted genes. Int J Obes (Lond) 2013; 39:650-7. [PMID: 24158121 PMCID: PMC4048324 DOI: 10.1038/ijo.2013.193] [Citation(s) in RCA: 222] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 09/25/2013] [Accepted: 10/06/2013] [Indexed: 12/14/2022]
Abstract
Background: Several epidemiologic studies have demonstrated associations between periconceptional environmental exposures and health status of the offspring in later life. Although these environmentally related effects have been attributed to epigenetic changes, such as DNA methylation shifts at imprinted genes, little is known about the potential effects of maternal and paternal preconceptional overnutrition or obesity. Objective: We examined parental preconceptional obesity in relation to DNA methylation profiles at multiple human imprinted genes important in normal growth and development, such as: maternally expressed gene 3 (MEG3), mesoderm-specific transcript (MEST), paternally expressed gene 3 (PEG3), pleiomorphic adenoma gene-like 1 (PLAGL1), epsilon sarcoglycan and paternally expressed gene 10 (SGCE/PEG10) and neuronatin (NNAT). Methods: We measured methylation percentages at the differentially methylated regions (DMRs) by bisulfite pyrosequencing in DNA extracted from umbilical cord blood leukocytes of 92 newborns. Preconceptional obesity, defined as BMI ⩾30 kg m−2, was ascertained through standardized questionnaires. Results: After adjusting for potential confounders and cluster effects, paternal obesity was significantly associated with lower methylation levels at the MEST (β=−2.57; s.e.=0.95; P=0.008), PEG3 (β=−1.71; s.e.=0.61; P=0.005) and NNAT (β=−3.59; s.e.=1.76; P=0.04) DMRs. Changes related to maternal obesity detected at other loci were as follows: β-coefficient was +2.58 (s.e.=1.00; P=0.01) at the PLAGL1 DMR and −3.42 (s.e.=1.69; P=0.04) at the MEG3 DMR. Conclusion: We found altered methylation outcomes at multiple imprint regulatory regions in children born to obese parents, compared with children born to non-obese parents. In spite of the small sample size, our data suggest a preconceptional influence of parental life-style or overnutrition on the (re)programming of imprint marks during gametogenesis and early development. More specifically, the significant and independent association between paternal obesity and the offspring's methylation status suggests the susceptibility of the developing sperm for environmental insults. The acquired imprint instability may be carried onto the next generation and increase the risk for chronic diseases in adulthood.
Collapse
Affiliation(s)
- A Soubry
- 1] Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA [2] Epidemiology Research Group, Department of Public Health and Primary Care, Faculty of Medicine, KULeuven, Leuven, Belgium
| | - S K Murphy
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke University Medical Center, Durham, NC, USA
| | - F Wang
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Z Huang
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke University Medical Center, Durham, NC, USA
| | - A C Vidal
- Department of Obstetrics and Gynecology, Division of Clinical and Epidemiologic Research and Cancer Prevention, Duke University Medical Center, Durham, NC, USA
| | - B F Fuemmeler
- Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA
| | - J Kurtzberg
- Carolinas Cord Blood Bank, Robertson Cell and Translational Therapy Program, Duke Translational Research Institute, Duke University Medical Center, Durham, NC, USA
| | - A Murtha
- Department of Obstetrics and Gynecology, Division of Maternal and Fetal Medicine, Duke University Medical Center, Durham, NC, USA
| | - R L Jirtle
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, USA
| | - J M Schildkraut
- 1] Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA [2] Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA
| | - C Hoyo
- Department of Obstetrics and Gynecology, Division of Clinical and Epidemiologic Research and Cancer Prevention, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
885
|
Ajduk A, Zernicka-Goetz M. Quality control of embryo development. Mol Aspects Med 2013; 34:903-18. [DOI: 10.1016/j.mam.2013.03.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 03/05/2013] [Accepted: 03/19/2013] [Indexed: 11/28/2022]
|
886
|
Affiliation(s)
- Jonathan M. G. Higgins
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mary Herbert
- Newcastle Fertility Centre and Institute for Aging and Health, International Centre for Life, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
887
|
Moshkin YM, Doyen CM, Kan TW, Chalkley GE, Sap K, Bezstarosti K, Demmers JA, Ozgur Z, van Ijcken WFJ, Verrijzer CP. Histone chaperone NAP1 mediates sister chromatid resolution by counteracting protein phosphatase 2A. PLoS Genet 2013; 9:e1003719. [PMID: 24086141 PMCID: PMC3784504 DOI: 10.1371/journal.pgen.1003719] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 06/26/2013] [Indexed: 12/27/2022] Open
Abstract
Chromosome duplication and transmission into daughter cells requires the precisely orchestrated binding and release of cohesin. We found that the Drosophila histone chaperone NAP1 is required for cohesin release and sister chromatid resolution during mitosis. Genome-wide surveys revealed that NAP1 and cohesin co-localize at multiple genomic loci. Proteomic and biochemical analysis established that NAP1 associates with the full cohesin complex, but it also forms a separate complex with the cohesin subunit stromalin (SA). NAP1 binding to cohesin is cell-cycle regulated and increases during G2/M phase. This causes the dissociation of protein phosphatase 2A (PP2A) from cohesin, increased phosphorylation of SA and cohesin removal in early mitosis. PP2A depletion led to a loss of centromeric cohesion. The distinct mitotic phenotypes caused by the loss of either PP2A or NAP1, were both rescued by their concomitant depletion. We conclude that the balanced antagonism between NAP1 and PP2A controls cohesin dissociation during mitosis.
Collapse
Affiliation(s)
- Yuri M. Moshkin
- Department of Biochemistry and Centre for Biomedical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Cecile M. Doyen
- Department of Biochemistry and Centre for Biomedical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tsung-Wai Kan
- Department of Biochemistry and Centre for Biomedical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Gillian E. Chalkley
- Department of Biochemistry and Centre for Biomedical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Karen Sap
- Proteomics Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Karel Bezstarosti
- Proteomics Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jeroen A. Demmers
- Proteomics Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Zeliha Ozgur
- Genomics Centre, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - C. Peter Verrijzer
- Department of Biochemistry and Centre for Biomedical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
888
|
Liang XW, Cui XS, Sun SC, Jin YX, Heo YT, Namgoong S, Kim NH. Superovulation induces defective methylation in line-1 retrotransposon elements in blastocyst. Reprod Biol Endocrinol 2013; 11:69. [PMID: 23866265 PMCID: PMC3723434 DOI: 10.1186/1477-7827-11-69] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 07/15/2013] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Series of epigenetic events happen during preimplantation development. Therefore assistant reproduction techniques (ART) have the potential to disrupt epigenetic regulation during embryo development. The purpose of this study was to investigate whether defects in methylation patterns in blastocyst due to superovulation originate from abnormal expression of Dnmts. METHODS Low- (6 IU) and high- (10 IU) dosage of PMSG was used to stimulate the female mice. The metaphase II(MII) oocytes, zygotes and blastocyst stage embryos were collected. Global methylation and methylation at H3K9 in zygote, and methylation at repeated sequence Line 1 and IAP in blastocysts were assayed. In addition, expression of Dnmts was examined in oocytes and zygotes. RESULTS Global DNA methylation and methylation at H3K9 in zygotes derived from females after low- or high-dosage hormone treatment were unaltered compared to that in controls. Moreover, DNA methylation at IAP in blastocysts was also unaffected, regardless of hormone dosage. In contrast, methylation at Line1 decreased when high-dose hormone was administered. Unexpectedly, expression of Dnmt3a, Dnmt3b, Dnmt3L as well as maintenance Dnmt1o in oocytes and zygotes was not disrupted. CONCLUSIONS The results suggest that defects in embryonic methylation patterns do not originate from the disruption of Dnmt expression.
Collapse
Affiliation(s)
- Xing-Wei Liang
- Department of Animal Sciences, Chungbuk National University, Cheongju 361-763, South Korea
| | - Xiang-Shun Cui
- Department of Animal Sciences, Chungbuk National University, Cheongju 361-763, South Korea
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yong-Xun Jin
- Department of Animal Sciences, Chungbuk National University, Cheongju 361-763, South Korea
| | - Young Tae Heo
- Department of Animal Sciences, Chungbuk National University, Cheongju 361-763, South Korea
| | - Suk Namgoong
- Department of Animal Sciences, Chungbuk National University, Cheongju 361-763, South Korea
| | - Nam-Hyung Kim
- Department of Animal Sciences, Chungbuk National University, Cheongju 361-763, South Korea
| |
Collapse
|
889
|
Baran V, Solc P, Kovarikova V, Rehak P, Sutovsky P. Polo-like kinase 1 is essential for the first mitotic division in the mouse embryo. Mol Reprod Dev 2013; 80:522-34. [PMID: 23649868 DOI: 10.1002/mrd.22188] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 04/29/2013] [Indexed: 11/11/2022]
Abstract
Polo-like kinase 1 (PLK1), a member of the serine/threonine protein kinases family, is involved in multiple steps of mitotic progression. It regulates centrosome maturation, mitotic spindle formation, and cytokinesis. While studied extensively in somatic cells, little is known about PLK1 activities in the mammalian preimplantation embryo. We examined the role of PLK1 in the one-cell mouse embryo. Western blotting showed that the PLK1 protein content increased significantly during the S-phase of the one-cell stage and declined during the first mitotic division. Activation of PLK1 preceded nuclear envelope breakdown (NEBD) in both pronuclei at the entry to first embryo mitosis. Immunofluorescence revealed the presence of phosphorylated, active PLK1 (pThr(210) -PLK1) in both male and female pronuclei, and in the microtubule-organizing centers (MTOCs) shortly before NEBD. During the first mitotic metaphase, pThr(210) -PLK1 accumulated at the spindle poles and was also associated with condensed chromosomes. Inhibition of PLK1 activity with a specific PLK1 inhibitor, BI 2536, at the one-cell stage induced the formation of a bipolar spindle that displayed disordered microtubular arrangements and dislocated, condensed chromosomes. Although such embryos entered mitosis, they did not complete mitosis and arrested at metaphase. Time-lapse recording revealed progressive misalignment of condensed chromosomes during first mitotic metaphase. These data indicate that PLK1 activity is not essential for entry into first mitosis, but is required for the events leading up to metaphase-anaphase transition in the one-cell mouse embryo.
Collapse
Affiliation(s)
- V Baran
- Institute of Animal Physiology, Slovak Academy of Sciences, Kosice, Slovakia.
| | | | | | | | | |
Collapse
|
890
|
Qiao J, Wang ZB, Feng HL, Miao YL, Wang Q, Yu Y, Wei YC, Yan J, Wang WH, Shen W, Sun SC, Schatten H, Sun QY. The root of reduced fertility in aged women and possible therapentic options: current status and future perspects. Mol Aspects Med 2013; 38:54-85. [PMID: 23796757 DOI: 10.1016/j.mam.2013.06.001] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 06/06/2013] [Indexed: 12/21/2022]
Abstract
It is well known that maternal ageing not only causes increased spontaneous abortion and reduced fertility, but it is also a high genetic disease risk. Although assisted reproductive technologies (ARTs) have been widely used to treat infertility, the overall success is still low. The main reasons for age-related changes include reduced follicle number, compromised oocyte quality especially aneuploidy, altered reproductive endocrinology, and increased reproductive tract defect. Various approaches for improving or treating infertility in aged women including controlled ovarian hyperstimulation with intrauterine insemination (IUI), IVF/ICSI-ET, ovarian reserve testing, preimplantation genetic diagnosis and screening (PGD/PGS), oocyte selection and donation, oocyte and ovary tissue cryopreservation before ageing, miscarriage prevention, and caloric restriction are summarized in this review. Future potential reproductive techniques for infertile older women including oocyte and zygote micromanipulations, derivation of oocytes from germ stem cells, ES cells, and iPS cells, as well as through bone marrow transplantation are discussed.
Collapse
Affiliation(s)
- Jie Qiao
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, People's Republic of China
| | - Zhen-Bo Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Huai-Liang Feng
- Department of Laboratory Medicine, and Obstetrics and Gynecology, New York Hospital Queens, Weill Medical College of Cornell University, New York, NY, USA
| | - Yi-Liang Miao
- Reproductive Medicine Group, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Qiang Wang
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 660 South Euclid Ave., St. Louis, MO 63110, USA
| | - Yang Yu
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, People's Republic of China
| | - Yan-Chang Wei
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Jie Yan
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, People's Republic of China
| | - Wei-Hua Wang
- Houston Fertility Institute, Tomball Regional Hospital, Tomball, TX 77375, USA
| | - Wei Shen
- Laboratory of Germ Cell Biology, Department of Animal Science, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Shao-Chen Sun
- Department of Animal Science, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China.
| |
Collapse
|
891
|
Bembenek JN, Verbrugghe KJC, Khanikar J, Csankovszki G, Chan RC. Condensin and the spindle midzone prevent cytokinesis failure induced by chromatin bridges in C. elegans embryos. Curr Biol 2013; 23:937-46. [PMID: 23684975 DOI: 10.1016/j.cub.2013.04.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 03/12/2013] [Accepted: 04/09/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND During cell division, chromosomes must clear the path of the cleavage furrow before the onset of cytokinesis. The abscission checkpoint in mammalian cells stabilizes the cleavage furrow in the presence of a chromatin obstruction. This provides time to resolve the obstruction before the cleavage furrow regresses or breaks the chromosomes, preventing aneuploidy or DNA damage. Two unanswered questions in the proposed mechanistic pathway of the abscission checkpoint concern factors involved in (1) resolving the obstructions and (2) coordinating obstruction resolution with the delay in cytokinesis. RESULTS We found that the one-cell and two-cell C. elegans embryos suppress furrow regression following depletion of essential chromosome-segregation factors: topoisomerase II(TOP-2), CENP-A(HCP-3), cohesin, and to a lesser degree, condensin. Chromatin obstructions activated Aurora B(AIR-2) at the spindle midzone, which is needed for the abscission checkpoint in other systems. Condensin I, but not condensin II, localizes to the spindle midzone in anaphase and to the midbody during normal cytokinesis. Interestingly, condensin I is enriched on chromatin bridges and near the midzone/midbody in an AIR-2-dependent manner. Disruption of AIR-2, the spindle midzone, or condensin leads to cytokinesis failure in a chromatin-obstruction-dependent manner. Examination of the condensin-deficient embryos uncovered defects in both the resolution of the chromatin obstructions and the maintenance of the stable cleavage furrow. CONCLUSIONS We postulate that condensin I is recruited by Aurora B(AIR-2) to aid in the resolution of chromatin obstructions and also helps generate a signal to maintain the delay in cytokinesis.
Collapse
Affiliation(s)
- Joshua N Bembenek
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | |
Collapse
|
892
|
Wang L, Lu A, Zhou HX, Sun R, Zhao J, Zhou CJ, Shen JP, Wu SN, Liang CG. Casein kinase 1 alpha regulates chromosome congression and separation during mouse oocyte meiotic maturation and early embryo development. PLoS One 2013; 8:e63173. [PMID: 23690993 PMCID: PMC3655170 DOI: 10.1371/journal.pone.0063173] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 03/28/2013] [Indexed: 11/18/2022] Open
Abstract
Casein kinase I alpha (CK1α) is a member of serine/threonine protein kinase, generally present in all eukaryotes. In mammals, CK1α regulates the transition from interphase to metaphase in mitosis. However, little is known about its role in meiosis. Here we examined Ck1α mRNA and protein expression, as well as its subcellular localization in mouse oocytes from germinal vesicle to the late 1-cell stage. Our results showed that the expression level of CK1α was increased in metaphase. Immunostaining results showed that CK1α colocalized with condensed chromosomes during oocyte meiotic maturation and early embryo development. We used the loss-of-function approach by employing CK1α specific morpholino injection to block the function of CK1α. This functional blocking leads to failure of polar body 1 (PB1) extrusion, chromosome misalignment and MII plate incrassation. We further found that D4476, a specific and efficient CK1 inhibitor, decreased the rate of PB1 extrusion. Moreover, D4476 resulted in giant polar body extrusion, oocyte pro-MI arrest, chromosome congression failure and impairment of embryo developmental potential. In addition, we employed pyrvinium pamoate (PP), an allosteric activator of CK1α, to enhance CK1α activity in oocytes. Supplementation of PP induced oocyte meiotic maturation failure, severe congression abnormalities and misalignment of chromosomes. Taken together, our study for the first time demonstrates that CK1α is required for chromosome alignment and segregation during oocyte meiotic maturation and early embryo development.
Collapse
Affiliation(s)
- Lu Wang
- The Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China
| | - Angeleem Lu
- The Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China
| | - Hong-Xia Zhou
- The Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China
| | - Ran Sun
- The Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China
| | - Jie Zhao
- The Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China
| | - Cheng-Jie Zhou
- The Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China
| | - Jiang-Peng Shen
- The Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China
| | - Sha-Na Wu
- The Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China
| | - Cheng-Guang Liang
- The Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China
- * E-mail:
| |
Collapse
|
893
|
Ge ZJ, Liang XW, Guo L, Liang QX, Luo SM, Wang YP, Wei YC, Han ZM, Schatten H, Sun QY. Maternal diabetes causes alterations of DNA methylation statuses of some imprinted genes in murine oocytes. Biol Reprod 2013; 88:117. [PMID: 23515675 DOI: 10.1095/biolreprod.112.105981] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Maternal diabetes has adverse effects not only on oocyte quality but also on embryo development. However, it is still unknown whether the DNA imprinting in oocytes is altered by diabetes. By using streptozotocin (STZ)-induced and nonobese diabetic (NOD) mouse models we investigated the effect of maternal diabetes on DNA methylation of imprinted genes in oocytes. Mice which were judged as being diabetic 4 days after STZ injection were used for experiments. In superovulated oocytes of diabetic mice, the methylation pattern of Peg3 differential methylation regions (DMR) was affected in a time-dependent manner, and evident demethylation was observed on Day 35 after STZ injection. The expression level of DNA methyltransferases (DNMTs) was also decreased in a time-dependent manner in diabetic oocytes. However, the methylation patterns of H19 and Snrpn DMRs were not significantly altered by maternal diabetes, although there were some changes in Snrpn. In NOD mice, the methylation pattern of Peg3 was similar to that of STZ-induced mice. Embryo development was adversely affected by maternal diabetes; however, no evident imprinting abnormality was observed in oocytes from female offspring derived from a diabetic mother. These results indicate that maternal diabetes has adverse effects on DNA methylation of maternally imprinted gene Peg3 in oocytes of a diabetic female in a time-dependent manner, but methylation in offspring's oocytes is normal.
Collapse
Affiliation(s)
- Zhao-Jia Ge
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
894
|
Wassmann K. Sister chromatid segregation in meiosis II: deprotection through phosphorylation. Cell Cycle 2013; 12:1352-9. [PMID: 23574717 DOI: 10.4161/cc.24600] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Meiotic divisions (meiosis I and II) are specialized cell divisions to generate haploid gametes. The first meiotic division with the separation of chromosomes is named reductional division. The second division, which takes place immediately after meiosis I without intervening S-phase, is equational, with the separation of sister chromatids, similar to mitosis. This meiotic segregation pattern requires the two-step removal of the cohesin complex holding sister chromatids together: cohesin is removed from chromosome arms that have been subjected to homologous recombination in meiosis I and from the centromere region in meiosis II. Cohesin in the centromere region is protected from removal in meiosis I, but this protection has to be removed--deprotected--for sister chromatid segregation in meiosis II. Whereas the mechanisms of cohesin protection are quite well understood, the mechanisms of deprotection have been largely unknown until recently. In this review I summarize our current knowledge on cohesin deprotection.
Collapse
|
895
|
Mantikou E, Wong KM, Repping S, Mastenbroek S. Molecular origin of mitotic aneuploidies in preimplantation embryos. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1921-30. [DOI: 10.1016/j.bbadis.2012.06.013] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 06/14/2012] [Accepted: 06/26/2012] [Indexed: 01/06/2023]
|
896
|
Yang SW, Gao C, Chen L, Song YL, Zhu JL, Qi ST, Jiang ZZ, Wang ZW, Lin F, Huang H, Xing FQ, Sun QY. Nek9 regulates spindle organization and cell cycle progression during mouse oocyte meiosis and its location in early embryo mitosis. Cell Cycle 2012; 11:4366-77. [PMID: 23159858 DOI: 10.4161/cc.22690] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Nek9 (also known as Nercc1), a member of the NIMA (never in mitosis A) family of protein kinases, regulates spindle formation, chromosome alignment and segregation in mitosis. Here, we showed that Nek9 protein was expressed from germinal vesicle (GV) to metaphase II (MII) stages in mouse oocytes with no detectable changes. Confocal microscopy identified that Nek9 was localized to the spindle poles at the metaphase stages and associated with the midbody at anaphase or telophase stage in both meiotic oocytes and the first mitotic embyros. Depletion of Nek9 by specific morpholino injection resulted in severely defective spindles and misaligned chromosomes with significant pro-MI/MI arrest and failure of first polar body (PB1) extrusion. Knockdown of Nek9 also impaired the spindle-pole localization of γ-tubulin and resulted in retention of the spindle assembly checkpoint protein Bub3 at the kinetochores even after 10 h of culture. Live-cell imaging analysis also confirmed that knockdown of Nek9 resulted in oocyte arrest at the pro-MI/MI stage with abnormal spindles, misaligned chromosomes and failed polar body emission. Taken together, our results suggest that Nek9 may act as a MTOC-associated protein regulating microtubule nucleation, spindle organization and, thus, cell cycle progression during mouse oocyte meiotic maturation, fertilization and early embryo cleavage.
Collapse
Affiliation(s)
- Shang-Wu Yang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
897
|
Schuyler SC, Wu YF, Kuan VJW. The Mad1-Mad2 balancing act--a damaged spindle checkpoint in chromosome instability and cancer. J Cell Sci 2012; 125:4197-206. [PMID: 23093575 DOI: 10.1242/jcs.107037] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cancer cells are commonly aneuploid. The spindle checkpoint ensures accurate chromosome segregation by controlling cell cycle progression in response to aberrant microtubule-kinetochore attachment. Damage to the checkpoint, which is a partial loss or gain of checkpoint function, leads to aneuploidy during tumorigenesis. One form of damage is a change in levels of the checkpoint proteins mitotic arrest deficient 1 and 2 (Mad1 and Mad2), or in the Mad1:Mad2 ratio. Changes in Mad1 and Mad2 levels occur in human cancers, where their expression is regulated by the tumor suppressors p53 and retinoblastoma 1 (RB1). By employing a standard assay, namely the addition of a mitotic poison at mitotic entry, it has been shown that checkpoint function is normal in many cancer cells. However, in several experimental systems, it has been observed that this standard assay does not always reveal checkpoint aberrations induced by changes in Mad1 or Mad2, where excess Mad1 relative to Mad2 can lead to premature anaphase entry, and excess Mad2 can lead to a delay in entering anaphase. This Commentary highlights how changes in the levels of Mad1 and Mad2 result in a damaged spindle checkpoint, and explores how these changes cause chromosome instability that can lead to aneuploidy during tumorigenesis.
Collapse
Affiliation(s)
- Scott C Schuyler
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, 333 Taiwan, Republic of China.
| | | | | |
Collapse
|
898
|
Liang X, Ma J, Schatten H, Sun Q. Epigenetic changes associated with oocyte aging. SCIENCE CHINA-LIFE SCIENCES 2012; 55:670-6. [DOI: 10.1007/s11427-012-4354-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 06/19/2012] [Indexed: 11/29/2022]
|
899
|
Nath S, Moghe M, Chowdhury A, Godbole K, Godbole G, Doiphode M, Roychoudhury S. Is germline transmission of MAD2 gene deletion associated with human fetal loss? Mol Hum Reprod 2012; 18:554-62. [PMID: 22869558 DOI: 10.1093/molehr/gas031] [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
The spindle assembly checkpoint (SAC) monitors proper attachment of spindles to the kinetochore during mitotic and meiotic cell divisions and thus prevents aneuploidy. Chromosomal aneuploidy has been found to be associated with pregnancy loss and birth defects. Mad2 is one of the critical molecules of SAC. Deregulated Mad2 expression has been found to be associated with defective SAC-mediated abnormal meiotic progression in cell studies using animal models. Whether mutation in MAD2L1 is associated with the loss of Mad2 expression in aborted human fetuses is unknown. In this study, a correlation between aneuploidy and MAD2 defect was examined in primary fibroblast cultures obtained from abortuses. We report three trisomic abortuses with undetectable Mad2 expression. Further, quantitative real-time PCR revealed copy number deletion of MAD2 gene in these fetuses. Analysis of parental DNA samples available from two families revealed copy number loss of the same gene, suggesting Mendelian inheritance of MAD2 deletion. This germline transmission of exonic deletion of MAD2 is possibly associated with its loss of expression resulting in abnormal SAC function, subsequent aneuploidy and pregnancy loss.
Collapse
Affiliation(s)
- Somsubhra Nath
- Cancer Biology and Inflammatory Disorder Division, CSIR, Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700032, India
| | | | | | | | | | | | | |
Collapse
|
900
|
Sun SC, Gao WW, Xu YN, Jin YX, Wang QL, Yin XJ, Cui XS, Kim NH. Degradation of actin nucleators affects cortical polarity of aged mouse oocytes. Fertil Steril 2012; 97:984-90. [PMID: 22306711 DOI: 10.1016/j.fertnstert.2012.01.101] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 01/12/2012] [Accepted: 01/12/2012] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To investigate the molecular mechanism of mouse oocyte polarity loss during aging. DESIGN Experimental study. SETTING Academic basic research laboratory. ANIMAL(S) Mice. INTERVENTION(S) Oocytes were collected 16 hours after injection of hCG and cultured in M16 medium for an additional 14 hours with or without caffeine. MAIN OUTCOME MEASURE(S) Expression and localizations of actin nucleators actin-related protein 2/3 complex, JMY, and WAVE2 were examined by immunofluorescence staining, and their messenger RNA levels were examined by real-time reverse transcription-polymerase chain reaction. RESULT(S) The protein and messenger RNA levels of actin-related protein 2/3 complex, JMY, and WAVE2 were decreased in aged oocytes, but the levels were normal in caffeine-treated aged oocytes. CONCLUSION(S) Our data indicated that the loss of oocyte polarity may be due to the degradation of actin nucleators in aged oocytes.
Collapse
Affiliation(s)
- Shao-Chen Sun
- Department of Animal Sciences, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | | | | | | | | | | | | | | |
Collapse
|