801
|
Uysal F, Akkoyunlu G, Ozturk S. Dynamic expression of DNA methyltransferases (DNMTs) in oocytes and early embryos. Biochimie 2015; 116:103-13. [PMID: 26143007 DOI: 10.1016/j.biochi.2015.06.019] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/26/2015] [Indexed: 11/26/2022]
Abstract
Epigenetic mechanisms play critical roles in oogenesis and early embryo development in mammals. One of these epigenetic mechanisms, DNA methylation is accomplished through the activities of DNA methyltransferases (DNMTs), which are responsible for adding a methyl group to the fifth carbon atom of the cytosine residues within cytosine-phosphate-guanine (CpG) and non-CpG dinuclotide sites. Five DNMT enzymes have been identified in mammals including DNMT1, DNMT2, DNMT3A, DNMT3B, and DNMT3L. They function in two different methylation processes: maintenance and de novo. For maintenance methylation, DNMT1 preferentially transfers methyl groups to the hemi-methylated DNA strands following DNA replication. However, for de novo methylation activities both DNMT3A and DNMT3B function in the methylation of the unmodified cytosine residues. Although DNMT3L indirectly contributes to de novo methylation process, DNMT2 enables the methylation of the cytosine 38 in the anticodon loop of aspartic acid transfer RNA and does not methylate DNA. In this review article, we have evaluated and discussed the existing published studies to characterize the spatial and temporal expression patterns of the DNMTs in mouse, bovine and human oocytes and early embryos. We have also reviewed the effects of in vitro culture conditions (serum abundance and glucose concentration), aging, superovulation, vitrification, and somatic cell nuclear transfer technology on the dynamics of DNMTs.
Collapse
Affiliation(s)
- Fatma Uysal
- Department of Histology and Embryology, Akdeniz University, School of Medicine, Antalya, Turkey
| | - Gokhan Akkoyunlu
- Department of Histology and Embryology, Akdeniz University, School of Medicine, Antalya, Turkey
| | - Saffet Ozturk
- Department of Histology and Embryology, Akdeniz University, School of Medicine, Antalya, Turkey.
| |
Collapse
|
802
|
Zhao H, Yang L, Cui H. SIRT1 regulates autophagy and diploidization in parthenogenetic haploid embryonic stem cells. Biochem Biophys Res Commun 2015. [DOI: 10.1016/j.bbrc.2015.07.098] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
803
|
Yu C, Ji SY, Sha QQ, Sun QY, Fan HY. CRL4-DCAF1 ubiquitin E3 ligase directs protein phosphatase 2A degradation to control oocyte meiotic maturation. Nat Commun 2015; 6:8017. [PMID: 26281983 PMCID: PMC4557334 DOI: 10.1038/ncomms9017] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 07/07/2015] [Indexed: 01/10/2023] Open
Abstract
Oocyte meiosis is a specialized cell cycle that gives rise to fertilizable haploid gametes and is precisely controlled in various dimensions. We recently found that E3 ubiquitin ligase CRL4 is required for female fertility by regulating DNA hydroxymethylation to maintain oocyte survival and to promote zygotic genome reprogramming. However, not all phenotypes of CRL4-deleted oocytes could be explained by this mechanism. Here we show that CRL4 controls oocyte meiotic maturation by proteasomal degradation of protein phosphatase 2A scaffold subunit, PP2A-A. Oocyte-specific deletion of DDB1 or DCAF1 (also called VPRBP) results in delayed meiotic resumption and failure to complete meiosis I along with PP2A-A accumulation. DCAF1 directly binds to and results in the poly-ubiquitination of PP2A-A. Moreover, combined deletion of Ppp2r1a rescues the meiotic defects caused by DDB1/DCAF1 deficiency. These results provide in vivo evidence that CRL4-directed PP2A-A degradation is physiologically essential for regulating oocyte meiosis and female fertility. The E3 ubiquitin ligase CRL4 regulates oocyte survival through hydroxymethylation of genomic DNA. Here Yu et al. show that CRL4 is also required for oocytes to complete meiosis I by mediating the poly-ubiquitination and proteasomal degradation of the cell cycle regulator protein phosphatase 2A-A subunit.
Collapse
Affiliation(s)
- Chao Yu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Shu-Yan Ji
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Qian-Qian Sha
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Heng-Yu Fan
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
804
|
Maternal exercise before and during pregnancy does not impact offspring exercise or body composition in mice. J Negat Results Biomed 2015; 14:13. [PMID: 26235102 PMCID: PMC4522962 DOI: 10.1186/s12952-015-0032-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/01/2015] [Indexed: 01/16/2023] Open
Abstract
Background The genome, the environment, and their interactions simultaneously regulate complex traits such as body composition and voluntary exercise levels. One such environmental influence is the maternal milieu (i.e., in utero environment or maternal care). Variability in the maternal environment may directly impact the mother, and simultaneously has the potential to influence the physiology and/or behavior of offspring in utero, post birth, and into adulthood. Here, we utilized a murine model to examine the effects of the maternal environment in regard to voluntary exercise (absence of wheel running, wheel running prior to gestation, and wheel running prior to and throughout gestation) on offspring weight and body composition (% fat tissue and % lean tissue) throughout development (~3 to ~9 weeks of age). Additionally, we examined the effects of ~6 weeks of maternal exercise (prior to and during gestation) on offspring exercise levels at ~9 weeks of age. Results We observed no substantial effects of maternal exercise on subsequent male or female offspring body composition throughout development, or on the propensity of offspring to engage in voluntary wheel running. At the level of the individual, correlational analyses revealed some statistically significant relationships between maternal and offspring exercise levels, likely reflecting previously known heritability estimates for such traits. Conclusions The current results conflict with previous findings in human and mouse models demonstrating that maternal exercise has the potential to alter offspring phenotypes. We discuss our negative findings in the context of the timing of the maternal exercise and the level of biological organization of the examined phenotypes within the offspring. Electronic supplementary material The online version of this article (doi:10.1186/s12952-015-0032-x) contains supplementary material, which is available to authorized users.
Collapse
|
805
|
Environmental Impact on DNA Methylation in the Germline: State of the Art and Gaps of Knowledge. BIOMED RESEARCH INTERNATIONAL 2015; 2015:123484. [PMID: 26339587 PMCID: PMC4538313 DOI: 10.1155/2015/123484] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 05/03/2015] [Indexed: 12/19/2022]
Abstract
The epigenome consists of chemical changes in DNA and chromatin that without modifying the DNA sequence modulate gene expression and cellular phenotype. The epigenome is highly plastic and reacts to changing external conditions with modifications that can be inherited to daughter cells and across generations. Whereas this innate plasticity allows for adaptation to a changing environment, it also implies the potential of epigenetic derailment leading to so-called epimutations. DNA methylation is the most studied epigenetic mark. DNA methylation changes have been associated with cancer, infertility, cardiovascular, respiratory, metabolic, immunologic, and neurodegenerative pathologies. Experiments in rodents demonstrate that exposure to a variety of chemical stressors, occurring during the prenatal or the adult life, may induce DNA methylation changes in germ cells, which may be transmitted across generations with phenotypic consequences. An increasing number of human biomonitoring studies show environmentally related DNA methylation changes mainly in blood leukocytes, whereas very few data have been so far collected on possible epigenetic changes induced in the germline, even by the analysis of easily accessible sperm. In this paper, we review the state of the art on factors impinging on DNA methylation in the germline, highlight gaps of knowledge, and propose priorities for future studies.
Collapse
|
806
|
Zhang YL, Liu XM, Ji SY, Sha QQ, Zhang J, Fan HY. ERK1/2 activities are dispensable for oocyte growth but are required for meiotic maturation and pronuclear formation in mouse. J Genet Genomics 2015; 42:477-85. [PMID: 26408092 DOI: 10.1016/j.jgg.2015.07.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 07/18/2015] [Accepted: 07/23/2015] [Indexed: 01/06/2023]
Abstract
Previous studies revealed that extracellular regulated kinase-1 and -2 (ERK1/2) cascade plays pivotal roles in regulating oocyte meiotic cell cycle progression. However, most knowledge about the in vivo function of ERK1/2 in mammalian oocytes was indirectly obtained from analyzing the phenotypes of Mos knockout mice. In this study, we knocked out Erk1 and Erk2 in mouse oocytes as early as the primordial follicle stage using the well-characterized Gdf9-Cre mouse model, and for the first time directly investigated the in vivo function of ERK1/2 in mouse oocytes. In this novel mouse model, we observed that ERK1/2 activities in oocyte are dispensable for primordial follicle maintenance, activation and follicle growth. Different from the Mos null oocytes, the ERK1/2-deleted oocytes had well-assembled spindles at metaphase I (MI), extruded polar body-1 (PB1) with normal sizes, and did not undergo a full parthenogenetic activation characterized for pronuclear formation. However, the ovulated ERK1/2-deleted oocytes had poorly-assembled metaphase II (MII) spindles, spontaneously released polar body-2 (PB2), and were arrested at another metaphase called metaphase III (MIII). In addition, ERK1/2 deletion prevented male pronuclear formation after fertilization, and caused female infertility. In conclusion, these results indicate that ERK1/2 activities are required for not only MII-arrest maintenance, but also efficient pronuclear formation in mouse oocytes.
Collapse
Affiliation(s)
- Yin-Li Zhang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Xiao-Man Liu
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Shu-Yan Ji
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Qian-Qian Sha
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Jue Zhang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Heng-Yu Fan
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
807
|
Qiao Y, Wang X, Wang R, Li Y, Yu F, Yang X, Song L, Xu G, Chin YE, Jing N. AF9 promotes hESC neural differentiation through recruiting TET2 to neurodevelopmental gene loci for methylcytosine hydroxylation. Cell Discov 2015; 1:15017. [PMID: 27462416 PMCID: PMC4860857 DOI: 10.1038/celldisc.2015.17] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 05/25/2015] [Indexed: 01/23/2023] Open
Abstract
AF9 mutations have been implicated in human neurodevelopmental diseases and murine Af9 mediates histone methylation during cortical neuron generation. However, AF9 function and related mechanisms in human neurodevelopment remain unknown. Here we show that AF9 is necessary and sufficient for human embryonic stem cell (hESC) neural differentiation and neurodevelopmental gene activation. The 5-methylcytosine (5mC) dioxygenase TET2, which was identified in an AF9-associated protein complex, physically interacted with AF9. Both AF9 and TET2 co-localized in 5-hydroxymethylcytosine (5hmC)-positive hESC-derived neurons and were required for appropriate hESC neural differentiation. Upon binding to AAC-containing motifs, AF9 recruited TET2 to occupy the common neurodevelopmental gene loci to direct 5mC-to-5hmC conversion, which was followed by sequential activation of neural target genes and hESC neural commitment. These findings define an AF9-TET2 regulatory complex for modulating human neural development and reveal a novel mechanism by which the AF9 recognition specificity and TET2 hydroxylation activity cooperate to control neurodevelopmental gene activation.
Collapse
Affiliation(s)
- Yunbo Qiao
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , Shanghai, China
| | - Xiongjun Wang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Ran Wang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , Shanghai, China
| | - Yuanyuan Li
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , Shanghai, China
| | - Fang Yu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , Shanghai, China
| | - Xianfa Yang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Lu Song
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , Shanghai, China
| | - Guoliang Xu
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , Shanghai, China
| | - Y Eugene Chin
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , Shanghai, China
| |
Collapse
|
808
|
TET proteins in cancer: Current 'state of the art'. Crit Rev Oncol Hematol 2015; 96:425-36. [PMID: 26276226 DOI: 10.1016/j.critrevonc.2015.07.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 05/26/2015] [Accepted: 07/16/2015] [Indexed: 12/31/2022] Open
Abstract
Aberrations in DNA methylation patterns are observed from the early stages of carcinogenesis. However, the mechanisms that drive these changes remain elusive. The recent characterization of ten-eleven translocation (TET) enzymes as a source of newly modified cytosines (5-hydroxymethylcytosine, 5-formylcytosine and 5-carboxylcytosine) has shed new light on the DNA demethylation process. These cytosines are intermediates of an active DNA demethylation process and are epigenetic markers per se. In this review, we discuss the mechanism and function of TET proteins in biological processes as well as current knowledge regarding their expression and regulation in cancer.
Collapse
|
809
|
Herpin A, Englberger E, Zehner M, Wacker R, Gessler M, Schartl M. Defective autophagy through epg5 mutation results in failure to reduce germ plasm and mitochondria. FASEB J 2015; 29:4145-61. [PMID: 26183773 DOI: 10.1096/fj.14-265462] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 06/15/2015] [Indexed: 12/14/2022]
Abstract
Autophagy is an evolutionarily conserved catabolic process that transports cytoplasmic components to lysosomes for degradation. In addition to the canonical view of strict stress-response-induced autophagy, selectively programmed autophagy was recently reported in the context of gonad development of flies and worms, where autophagy seems to be necessary for clearance of germ plasm components. Similar functions have not been described in vertebrates. We used the medaka fish to study the role of autophagy in gonad formation and gametogenesis for the first time in a vertebrate organism for which the germ line is specified by germ plasm. Using a transgenic line deficient in the Ol-epg5 gene—a new critical component of the autophagy pathway—we show that such deficiency leads to an impaired autophagic flux, possibly attributed to compromised maturation or processing of the autophagosomes. Ol-epg5 deficiency correlates with selectively impaired spermatogenesis and low allele transmission rates of the mutant allele caused by failure of germ plasm and mitochondria clearance during the process of germ cell specification and in the adult gonads. The mouse epg-5 homolog is similarly expressed in the maturating and adult testes, suggesting an at least partially conserved function of this process during spermatogenesis in vertebrates.
Collapse
Affiliation(s)
- Amaury Herpin
- *Physiological Chemistry, Developmental Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany; Institut National de la Recherche Agronomique (INRA), Unité de Recherche 1037, Fish Physiology and Genomics, Rennes, France; and Comprehensive Cancer Centre, University Clinic Würzburg, Würzburg, Germany
| | - Eva Englberger
- *Physiological Chemistry, Developmental Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany; Institut National de la Recherche Agronomique (INRA), Unité de Recherche 1037, Fish Physiology and Genomics, Rennes, France; and Comprehensive Cancer Centre, University Clinic Würzburg, Würzburg, Germany
| | - Mario Zehner
- *Physiological Chemistry, Developmental Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany; Institut National de la Recherche Agronomique (INRA), Unité de Recherche 1037, Fish Physiology and Genomics, Rennes, France; and Comprehensive Cancer Centre, University Clinic Würzburg, Würzburg, Germany
| | - Robin Wacker
- *Physiological Chemistry, Developmental Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany; Institut National de la Recherche Agronomique (INRA), Unité de Recherche 1037, Fish Physiology and Genomics, Rennes, France; and Comprehensive Cancer Centre, University Clinic Würzburg, Würzburg, Germany
| | - Manfred Gessler
- *Physiological Chemistry, Developmental Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany; Institut National de la Recherche Agronomique (INRA), Unité de Recherche 1037, Fish Physiology and Genomics, Rennes, France; and Comprehensive Cancer Centre, University Clinic Würzburg, Würzburg, Germany
| | - Manfred Schartl
- *Physiological Chemistry, Developmental Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany; Institut National de la Recherche Agronomique (INRA), Unité de Recherche 1037, Fish Physiology and Genomics, Rennes, France; and Comprehensive Cancer Centre, University Clinic Würzburg, Würzburg, Germany
| |
Collapse
|
810
|
Abstract
SummaryAurora-A kinase (AURKA), a member of the serine/threonine protein kinase 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 information is known about AURKA in the early cleavage mouse embryo with respect to acentrosomal spindle assembly. In vitro experiments in which AURKA was inactivated with specific inhibitor MLN8237 during the early stages of embryogenesis documented gradual arrest in the cleavage ability of the mouse embryo. In the AURKA-inhibited 1-cell embryos, spindle formation and anaphase onset were delayed and chromosome segregation was defective. AURKA inhibition increased apoptosis during early embryonic development. In conclusion these data suggest that AURKA is essential for the correct chromosome segregation in the first mitosis as a prerequisite for normal later development after first cleavage.
Collapse
|
811
|
van Dijk SJ, Tellam RL, Morrison JL, Muhlhausler BS, Molloy PL. Recent developments on the role of epigenetics in obesity and metabolic disease. Clin Epigenetics 2015; 7:66. [PMID: 27408648 PMCID: PMC4940755 DOI: 10.1186/s13148-015-0101-5] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/29/2015] [Indexed: 12/20/2022] Open
Abstract
The increased prevalence of obesity and related comorbidities is a major public health problem. While genetic factors undoubtedly play a role in determining individual susceptibility to weight gain and obesity, the identified genetic variants only explain part of the variation. This has led to growing interest in understanding the potential role of epigenetics as a mediator of gene-environment interactions underlying the development of obesity and its associated comorbidities. Initial evidence in support of a role of epigenetics in obesity and type 2 diabetes mellitus (T2DM) was mainly provided by animal studies, which reported epigenetic changes in key metabolically important tissues following high-fat feeding and epigenetic differences between lean and obese animals and by human studies which showed epigenetic changes in obesity and T2DM candidate genes in obese/diabetic individuals. More recently, advances in epigenetic methodologies and the reduced cost of epigenome-wide association studies (EWAS) have led to a rapid expansion of studies in human populations. These studies have also reported epigenetic differences between obese/T2DM adults and healthy controls and epigenetic changes in association with nutritional, weight loss, and exercise interventions. There is also increasing evidence from both human and animal studies that the relationship between perinatal nutritional exposures and later risk of obesity and T2DM may be mediated by epigenetic changes in the offspring. The aim of this review is to summarize the most recent developments in this rapidly moving field, with a particular focus on human EWAS and studies investigating the impact of nutritional and lifestyle factors (both pre- and postnatal) on the epigenome and their relationship to metabolic health outcomes. The difficulties in distinguishing consequence from causality in these studies and the critical role of animal models for testing causal relationships and providing insight into underlying mechanisms are also addressed. In summary, the area of epigenetics and metabolic health has seen rapid developments in a short space of time. While the outcomes to date are promising, studies are ongoing, and the next decade promises to be a time of productive research into the complex interactions between the genome, epigenome, and environment as they relate to metabolic disease.
Collapse
Affiliation(s)
- Susan J van Dijk
- CSIRO Food and Nutrition Flagship, PO Box 52, North Ryde, NSW 1670 Australia
| | - Ross L Tellam
- CSIRO Agriculture Flagship, 306 Carmody Road, St Lucia, QLD 4067 Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, GPO Box 2471, Adelaide, SA 5001 Australia
| | - Beverly S Muhlhausler
- FOODplus Research Centre, Waite Campus, The University of Adelaide, PMB 1, Glen Osmond, SA 5064 Australia.,Women's and Children's Health Research Institute, 72 King William Road, North Adelaide, SA 5006 Australia
| | - Peter L Molloy
- CSIRO Food and Nutrition Flagship, PO Box 52, North Ryde, NSW 1670 Australia
| |
Collapse
|
812
|
Heinzmann J, Mattern F, Aldag P, Bernal-Ulloa SM, Schneider T, Haaf T, Niemann H. Extended in vitro maturation affects gene expression and DNA methylation in bovine oocytes. Mol Hum Reprod 2015; 21:770-82. [PMID: 26155800 DOI: 10.1093/molehr/gav040] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/22/2015] [Indexed: 12/31/2022] Open
Abstract
To mimic post-ovulatory ageing, we have extended the in vitro maturation (IVM) phase to 48 h and examined effects on (i) developmental potential, (ii) expression of a panel of developmentally important genes and (iii) gene-specific epigenetic marks. Results were compared with the 24 h IVM protocol (control) usually employed for bovine oocytes. Cleavage rates and blastocyst yields were significantly reduced in oocytes after extended IVM. No significant differences were observed in the methylation of entire alleles in oocytes for the genes bH19, bSNRPN, bZAR1, bOct4 and bDNMT3A. However, we found differentially methylated CpG sites in the bDNMT3Ls locus in oocytes after extended IVM and in embryos derived from them compared with controls. Moreover, embryos derived from the 48 h matured oocyte group were significantly less methylated at CpG5 and CpG7 compared with the 24 h group. CpG7 was significantly hypermethylated in embryos produced from the control oocytes, but not in oocytes matured for 48 h. Furthermore, methylation for CpG5-CpG8 of bDNMT3Ls was significantly lower in oocytes of the 24 h group compared with embryos derived therefrom, whereas no such difference was found for oocytes and embryos of the in vitro aged group. Expression of most of the selected genes was not affected by duration of IVM. However, transcript abundance for the imprinted gene bIGF2R was significantly reduced in oocytes analyzed after extended IVM compared with control oocytes. Transcript levels for bPRDX1, bDNMT3A and bBCLXL were significantly reduced in 4- to 8-cell embryos derived from in vitro aged oocytes. These results indicate that extended IVM leads to ageing-like alterations and demonstrate that epigenetic mechanisms are critically involved in ageing of bovine oocytes, which warrants further studies into epigenetic mechanisms involved in ageing of female germ cells, including humans.
Collapse
Affiliation(s)
- Julia Heinzmann
- Institute of Farm Animal Genetics (FLI), Mariensee, 31535 Neustadt, Germany
| | - Felix Mattern
- Institute of Human Genetics, Julius Maximilians University, 97070 Würzburg, Germany
| | - Patrick Aldag
- Institute of Farm Animal Genetics (FLI), Mariensee, 31535 Neustadt, Germany
| | | | - Tamara Schneider
- Institute of Human Genetics, Julius Maximilians University, 97070 Würzburg, Germany
| | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University, 97070 Würzburg, Germany
| | - Heiner Niemann
- Institute of Farm Animal Genetics (FLI), Mariensee, 31535 Neustadt, Germany
| |
Collapse
|
813
|
Djeddi A, Al Rawi S, Deuve JL, Perrois C, Liu YY, Russeau M, Sachse M, Galy V. Sperm-inherited organelle clearance in C. elegans relies on LC3-dependent autophagosome targeting to the pericentrosomal area. Development 2015; 142:1705-16. [PMID: 25922527 DOI: 10.1242/dev.117879] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Macroautophagic degradation of sperm-inherited organelles prevents paternal mitochondrial DNA transmission in C. elegans. The recruitment of autophagy markers around sperm mitochondria has also been observed in mouse and fly embryos but their role in degradation is debated. Both worm Atg8 ubiquitin-like proteins, LGG-1/GABARAP and LGG-2/LC3, are recruited around sperm organelles after fertilization. Whereas LGG-1 depletion affects autophagosome function, stabilizes the substrates and is lethal, we demonstrate that LGG-2 is dispensable for autophagosome formation but participates in their microtubule-dependent transport toward the pericentrosomal area prior to acidification. In the absence of LGG-2, autophagosomes and their substrates remain clustered at the cell cortex, away from the centrosomes and their associated lysosomes. Thus, the clearance of sperm organelles is delayed and their segregation between blastomeres prevented. This allowed us to reveal a role of the RAB-5/RAB-7 GTPases in autophagosome formation. In conclusion, the major contribution of LGG-2 in sperm-inherited organelle clearance resides in its capacity to mediate the retrograde transport of autophagosomes rather than their fusion with acidic compartments: a potential key function of LC3 in controlling the fate of sperm mitochondria in other species.
Collapse
Affiliation(s)
- Abderazak Djeddi
- Sorbonne Universités, UPMC, Univ Paris 06, Institut de Biologie Paris-Seine (IBPS), UMR7622, Paris F-75005, France CNRS, IBPS, UMR7622, Paris F-75005, France
| | - Sara Al Rawi
- Sorbonne Universités, UPMC, Univ Paris 06, Institut de Biologie Paris-Seine (IBPS), UMR7622, Paris F-75005, France CNRS, IBPS, UMR7622, Paris F-75005, France
| | - Jane Lynda Deuve
- Sorbonne Universités, UPMC, Univ Paris 06, Institut de Biologie Paris-Seine (IBPS), UMR7622, Paris F-75005, France CNRS, IBPS, UMR7622, Paris F-75005, France
| | - Charlene Perrois
- Sorbonne Universités, UPMC, Univ Paris 06, Institut de Biologie Paris-Seine (IBPS), UMR7622, Paris F-75005, France CNRS, IBPS, UMR7622, Paris F-75005, France
| | - Yu-Yu Liu
- Sorbonne Universités, UPMC, Univ Paris 06, Institut de Biologie Paris-Seine (IBPS), UMR7622, Paris F-75005, France CNRS, IBPS, UMR7622, Paris F-75005, France
| | - Marion Russeau
- Sorbonne Universités, UPMC, Univ Paris 06, Institut de Biologie Paris-Seine (IBPS), UMR7622, Paris F-75005, France CNRS, IBPS, UMR7622, Paris F-75005, France
| | - Martin Sachse
- PFMU, Imagopole, Institut Pasteur, Paris F-75015, France
| | - Vincent Galy
- Sorbonne Universités, UPMC, Univ Paris 06, Institut de Biologie Paris-Seine (IBPS), UMR7622, Paris F-75005, France CNRS, IBPS, UMR7622, Paris F-75005, France
| |
Collapse
|
814
|
Ong TP, Ozanne SE. Developmental programming of type 2 diabetes: early nutrition and epigenetic mechanisms. Curr Opin Clin Nutr Metab Care 2015; 18:354-60. [PMID: 26049632 DOI: 10.1097/mco.0000000000000177] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
PURPOSE OF REVIEW The environment experienced during critical windows of development can 'programme' long-term health and risk of metabolic diseases such as type 2 diabetes in the offspring. The purpose of this review is to discuss potential epigenetic mechanisms involved in the developmental programming of type 2 diabetes by early nutrition. RECENT FINDINGS Maternal and more recently paternal nutrition have been shown to play key roles in metabolic programming of the offspring. Although the exact mechanisms are still not clear, epigenetic processes have emerged as playing a plausible role. Epigenetic dysregulation is associated with several components that contribute to type 2 diabetes risk, including altered feeding behaviour, insulin secretion and insulin action. It may also contribute to transgenerational risk transmission. SUMMARY Epigenetic processes may represent a central underlying mechanism of developmental programming of type 2 diabetes. During embryonic and foetal development, extensive epigenetic remodelling takes place not only in somatic but also in primordial germ cells. Therefore, concerns have been raised that epigenetic dysregulation induced by a suboptimal early environment could programme altered phenotypes not only in the first generation but also in the subsequent ones. Characterizing these altered epigenetic marks has great implications for identifying individuals at an increased disease risk as well as potentially leading to novel preventive and treatment strategies.
Collapse
Affiliation(s)
- Thomas P Ong
- aUniversity of Cambridge Institute of Metabolic Science and MRC Metabolic Diseases Unit, Cambridge, UK bDepartment of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo and Food Research Center (FoRC), São Paulo, Brazil
| | | |
Collapse
|
815
|
Edwards N, Farookhi R, Clarke HJ. Identification of a β-galactosidase transgene that provides a live-cell marker of transcriptional activity in growing oocytes and embryos. Mol Hum Reprod 2015; 21:583-93. [PMID: 25882542 PMCID: PMC4487448 DOI: 10.1093/molehr/gav020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/27/2015] [Accepted: 04/09/2015] [Indexed: 01/14/2023] Open
Abstract
Identifying the events and molecular mechanisms that regulate oocyte growth has emerged as a key objective of research in human fertility, fuelled by evidence from human and animal studies indicating that disease and environmental factors can act on oocytes to affect the health of the resulting individual and by efforts to grow oocytes in vitro to enable fertility preservation of cancer survivors. Techniques that monitor the development of growing oocytes would be valuable tools to assess the progression of growth under different conditions. Most methods used to assess oocytes grown in vitro are indirect, however, relying on characteristics of the somatic compartment of the follicle, or compromise the oocyte, preventing its subsequent culture or fertilization. We investigated the utility of T-cell factor/lymphoid enhancer-binding factor (TCF/Lef)-LacZ transgene expression as a predictor of global transcriptional activity in oocytes and early embryos. Using a fluorescent β-galactosidase substrate combined with live-cell imaging, we show that TCF/Lef-LacZ transgene expression is detectable in growing oocytes, lost in fully grown oocytes and resumes in late two-cell embryos. Transgene expression is likely regulated by a Wnt-independent mechanism. Using chromatin analysis, LacZ expression and methods to monitor and inhibit transcription, we show that TCF/Lef-LacZ expression mirrors transcriptional activity in oocytes and preimplantation embryos. Oocytes and preimplantation embryos that undergo live-cell imaging for TCF/Lef-LacZ expression are able to continue development in vitro. TCF/Lef-LacZ reporter expression in living oocytes and early embryos is thus a sensitive and faithful marker of transcriptional activity that can be used to monitor and optimize conditions for oocyte growth.
Collapse
Affiliation(s)
- Nicole Edwards
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada Department of Physiology, McGill University, Montreal, QC, Canada Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Riaz Farookhi
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada Department of Physiology, McGill University, Montreal, QC, Canada Research Institute of the McGill University Health Centre, Montreal, QC, Canada Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Hugh J Clarke
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada Research Institute of the McGill University Health Centre, Montreal, QC, Canada Department of Experimental Medicine, McGill University, Montreal, QC, Canada Department of Biology, McGill University, Montreal, QC, Canada
| |
Collapse
|
816
|
Aslibekyan S, Demerath EW, Mendelson M, Zhi D, Guan W, Liang L, Sha J, Pankow JS, Liu C, Irvin MR, Fornage M, Hidalgo B, Lin LA, Thibeault KS, Bressler J, Tsai MY, Grove ML, Hopkins PN, Boerwinkle E, Borecki IB, Ordovas JM, Levy D, Tiwari HK, Absher DM, Arnett DK. Epigenome-wide study identifies novel methylation loci associated with body mass index and waist circumference. Obesity (Silver Spring) 2015; 23:1493-501. [PMID: 26110892 PMCID: PMC4482015 DOI: 10.1002/oby.21111] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 03/06/2015] [Accepted: 03/06/2015] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To conduct an epigenome-wide analysis of DNA methylation and obesity traits. METHODS DNA methylation was quantified in CD4+ T-cells using the Illumina Infinium HumanMethylation450 array in 991 participants of the Genetics of Lipid Lowering Drugs and Diet Network. Methylation at individual cytosine-phosphate-guanine (CpG) sites as a function of body mass index (BMI) and waist circumference (WC), adjusting for age, gender, study site, T-cell purity, smoking, and family structure, was modeled. RESULTS Epigenome-wide significant associations between eight CpG sites and BMI and five CpG sites and WC, successfully replicating the top hits in whole blood samples from the Framingham Heart Study (n = 2,377) and the Atherosclerosis Risk in Communities study (n = 2,097), were found. Top findings were in CPT1A (meta-analysis P = 2.7 × 10(-43) for BMI and 9.9 × 10(-23) for WC), PHGDH (meta-analysis P = 2.0 × 10(-15) for BMI and 4.0 × 10(-9) for WC), CD38 (meta-analysis P = 6.3 × 10(-11) for BMI and 1.6 × 10(-12) for WC), and long intergenic non-coding RNA 00263 (meta-analysis P = 2.2 × 10(-16) for BMI and 8.9 × 10(-14) for WC), regions with biologically plausible relationships to adiposity. CONCLUSIONS This large-scale epigenome-wide study discovered and replicated robust associations between DNA methylation at CpG loci and obesity indices, laying the groundwork for future diagnostic and/or therapeutic applications.
Collapse
Affiliation(s)
- Stella Aslibekyan
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ellen W Demerath
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael Mendelson
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
- Framingham Heart Study, Framingham, Massachusetts, USA
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Degui Zhi
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Liming Liang
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
- Framingham Heart Study, Framingham, Massachusetts, USA
- Department of Epidemiology, School of Public Health, Harvard University, Boston, Massachusetts, USA
- Department of Biostatistics, School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Jin Sha
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Chunyu Liu
- Framingham Heart Study, Framingham, Massachusetts, USA
- Department of Biostatistics, Boston University, Boston, Massachusetts, USA
| | - Marguerite R Irvin
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Texas, USA
| | - Bertha Hidalgo
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Li-An Lin
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Texas, USA
| | | | - Jan Bressler
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Texas, USA
| | - Michael Y Tsai
- Division of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Megan L Grove
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Texas, USA
| | - Paul N Hopkins
- School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Eric Boerwinkle
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Texas, USA
| | - Ingrid B Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University in St Louis, St Louis, Missouri, USA
| | - Jose M Ordovas
- Department of Epidemiology, Atherothrombosis and Imaging, Centro Nacional De Investigaciones Cardiovasculares, Madrid, Spain
- Instituto Madrileño De Estudios Avanzados Alimentacion, Madrid, Spain
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, USA
| | - Daniel Levy
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
- Framingham Heart Study, Framingham, Massachusetts, USA
| | - Hemant K Tiwari
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Devin M Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Donna K Arnett
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
817
|
Clarke HJ, Vieux KF. Epigenetic inheritance through the female germ-line: The known, the unknown, and the possible. Semin Cell Dev Biol 2015; 43:106-116. [DOI: 10.1016/j.semcdb.2015.07.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/04/2015] [Accepted: 07/06/2015] [Indexed: 02/06/2023]
|
818
|
Gui B, Yao Z, Huang Y, Mei L, Li Y, Liu D, Liu N, Xia Y, Liang D, Wu L. Morphometric analysis and developmental comparison of embryos from carriers with balanced chromosomal rearrangements in preimplantation genetic diagnosis cycles. Reprod Fertil Dev 2015; 28:1953-1963. [PMID: 26118930 DOI: 10.1071/rd15093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/28/2015] [Indexed: 11/23/2022] Open
Abstract
The morphological parameters of embryos from 22 carriers with balanced chromosomal rearrangements (CRs) were quantified and evaluated to determine their possible link to chromosomal composition. The morphometric characteristics of 168 embryos diagnosed by fluorescence in situ hybridisation were measured using an imaging tool and then analysed retrospectively. The mean zygotic diameter of normal-balanced embryos was significantly smaller compared with that of abnormal embryos (P=0.015). In addition, the reduction in total cytoplasmic volume for Day-3 embryos was significantly lower in normal or balanced embryos than in abnormal embryos (P=0.027). Moreover, the pronuclear volumes of embryos that failed to reach the blastocyst stage were significantly smaller compared with those of blastocysts (P=0.016). These findings indicate that morphometric characteristics are correlated with developmental outcomes as well as with chromosomal composition in embryos from balanced CR carriers. However, an effective indicator of developmental outcomes may not accurately reflect chromosomal composition. Combining morphometric and traditional qualitative assessment may increase the precision and standardisation of embryo evaluation as well as contributing to improved efficiency of preimplantation genetic diagnosis by selecting embryos with high developmental potential and preferentially testing embryos predicted to have a low risk of chromosomal imbalance.
Collapse
Affiliation(s)
- Baoheng Gui
- The State Key Laboratory of Medical Genetics of China, Central South University, No. 110, Xiangya Rd, Changsha, Hunan, 410008, P. R. China
| | - Zhongyuan Yao
- The State Key Laboratory of Medical Genetics of China, Central South University, No. 110, Xiangya Rd, Changsha, Hunan, 410008, P. R. China
| | - Yanru Huang
- The State Key Laboratory of Medical Genetics of China, Central South University, No. 110, Xiangya Rd, Changsha, Hunan, 410008, P. R. China
| | - Libin Mei
- The State Key Laboratory of Medical Genetics of China, Central South University, No. 110, Xiangya Rd, Changsha, Hunan, 410008, P. R. China
| | - Yanping Li
- The Reproductive Medical Center of Xiangya Hospital, Central South University, No. 87, Xiangya Rd, Changsha, Hunan, 410008, P. R. China
| | - Donge Liu
- The Reproductive Medical Center of Xiangya Hospital, Central South University, No. 87, Xiangya Rd, Changsha, Hunan, 410008, P. R. China
| | - Nenghui Liu
- The Reproductive Medical Center of Xiangya Hospital, Central South University, No. 87, Xiangya Rd, Changsha, Hunan, 410008, P. R. China
| | - Yan Xia
- The State Key Laboratory of Medical Genetics of China, Central South University, No. 110, Xiangya Rd, Changsha, Hunan, 410008, P. R. China
| | - Desheng Liang
- The State Key Laboratory of Medical Genetics of China, Central South University, No. 110, Xiangya Rd, Changsha, Hunan, 410008, P. R. China
| | - Lingqian Wu
- The State Key Laboratory of Medical Genetics of China, Central South University, No. 110, Xiangya Rd, Changsha, Hunan, 410008, P. R. China
| |
Collapse
|
819
|
Finger BJ, Harvey AJ, Green MP, Gardner DK. Combined parental obesity negatively impacts preimplantation mouse embryo development, kinetics, morphology and metabolism. Hum Reprod 2015; 30:2084-96. [PMID: 26089300 DOI: 10.1093/humrep/dev142] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 05/26/2015] [Indexed: 12/20/2022] Open
Abstract
STUDY QUESTION Does combined parental obesity, both an obese mother and father, have a greater effect on mouse preimplantation embryo development and quality than single-parent obesity? SUMMARY ANSWER Combined parental obesity causes a greater reduction in the blastocyst rate and a greater delay to the timing of key embryonic developmental events than single-parental obesity, as well as altering embryonic characteristics, such as zona pellucida width. WHAT IS KNOWN ALREADY Maternal or paternal obesity alone are known to have significant and detrimental impacts on preimplantation embryo development. Furthermore, these early embryonic perturbations can have long-term impacts on both offspring health and further generations. This is one of the first studies to examine the effects of having both an obese mother and an obese father. STUDY DESIGN, SIZE, DURATION A cross-sectional control versus treatment mouse study of diet-induced obesity was employed, in which 300 embryos per group were generated and studied from reciprocal matings: (i) control female and control male (Lean Parented Embryos); (ii) control female and obese male (Paternal Obese Parented Embryos); (iii) obese female and control male (Maternal Obese Parented Embryos) and (iv) obese female and obese male (Combined Obese Parented embryos). Assessments of the embryonic development rate, timing of development, morphological characteristics, metabolic gene expression, metabolism and cell lineage allocation were made at selected time points and analysed in relation to parental obesity status. PARTICIPANTS/MATERIALS, SETTING, METHODS Three-week-old C57BL6 male and female mice were fed control (7% total fat) or high fat (21% total fat) diets for a minimum of 8 weeks. Females were superovulated, mated, fertilized zygotes recovered and standard mouse in vitro embryo culture performed. Time-lapse monitoring was undertaken to compare developmental timings and morphological characteristics (embryonic area and zona pellucida width) for embryos from all four reciprocal matings. Differential staining identified cell lineage allocation. Real-time quantitative RT-PCR (qRT-PCR) and microfluorescence were used to measure gene expression and metabolism (glucose consumption and lactate production), respectively, in embryos from Lean Parented and Combined Obese Parented matings. This research was completed in a University research laboratory. MAIN RESULTS AND THE ROLE OF CHANCE Blastocyst rate was reduced in Combined Obese Parented embryos when compared with both Single Obese (11% decrease for Maternal Obese Parented, P < 0.05; 15% for Paternal Obese Parented, P < 0.05) and Lean Parented embryos (25% decrease, P < 0.01). Time-lapse analysis of developmental kinetics highlighted a delay of 1 h at the 2-3 cell division, extending to 6 h delay by the blastocyst stage for Combined Obese Parented embryos (P < 0.05). A reduction in the total cell number of Combined Obese Parented blastocysts was a further manifestation of this developmental delay (P < 0.05). Zona pellucida width was reduced in Combined Obese Parented embryos (P < 0.05). Glucose consumption was increased in Combined Obese Parented embryos (P < 0.05), which was associated with the up-regulation of Glucose transporter 1 expression (P < 0.05). LIMITATIONS AND REASON FOR CAUTION This study was completed in fertile C57BL/6 mice using a well-defined model of diet-induced obesity in which embryos were fertilized in vivo. Human obesity is complex, with many causes and co-morbidities, and therefore, the impact of combined obesity would require further investigation in human settings. WIDER IMPLICATIONS OF THE FINDINGS This study demonstrates that combined parental obesity has a detrimental impact on mouse embryo development, a finding consistent with previous studies on individual parent obesity. Of note, the effect of combined parental obesity upon embryo development markers was greater than that of individual parental obesity. Plausibly, human embryos will be similarly impacted. The reduction in the blastocyst rate and delayed time to developmental events confirms that embryos of obese parents differ from those of lean parents. Allowance for this should therefore be incorporated into clinical practice when selecting the best embryo for the transfer of an obese couple. STUDY FUNDING/COMPETING INTERESTS Funding was provided by University of Melbourne research monies. M.P.G. currently holds the position of Merck Serono Lecturer of Reproductive Biology. D.K.G. received research funds from Vitrolife AB Sweden. The other authors of this manuscript have nothing to declare and no conflicts of interest.
Collapse
Affiliation(s)
- Bethany J Finger
- School of BioSciences, University of Melbourne, Parkville, Melbourne, Victoria 3010, Australia
| | - Alexandra J Harvey
- School of BioSciences, University of Melbourne, Parkville, Melbourne, Victoria 3010, Australia
| | - Mark P Green
- School of BioSciences, University of Melbourne, Parkville, Melbourne, Victoria 3010, Australia
| | - David K Gardner
- School of BioSciences, University of Melbourne, Parkville, Melbourne, Victoria 3010, Australia
| |
Collapse
|
820
|
Abstract
SummaryThe aim of the present study was to determine the effects of increased dietary intake and high fat diet (HFD) in mice on artificial oocyte activation by using puromycin or roscovitine. Six-week-old mice were fed as either a control diet group, an increased dietary intake group or an HFD group for 4 weeks. Oocytes were obtained following superovulation and were divided into three treatment groups (no activation treatment, calcium ionophore and puromycin treatment, and calcium ionophore and roscovitine treatment) and were incubated for 4 h. Retrieved oocytes and numbers of oocytes activated as assessed by morphological changes were compared among the three treatment groups. The proportion of degenerated oocytes in HFD mice was significantly higher than that in control diet mice. The rates of activation in oocytes treated with roscovitine were 90.3% in control diet mice, 89.8% in increased dietary intake mice and 67.9% in HFD mice. The rate of activation in oocytes treated with roscovitine in HFD mice was significantly lower than the rates in control diet mice and increased dietary intake mice. The rates of activation in oocytes treated with puromycin were 90.6% in control diet mice, 94.0% in increased dietary intake mice and 71.4% in HFD mice, and the rate of activation in oocytes treated with puromycin in HFD mice was significantly lower than the rates in control diet mice and increased dietary intake mice. HFD-induced obesity deteriorated induction of oocyte activation by roscovitine or puromycin in mice.
Collapse
|
821
|
Dynamic changes of DNA epigenetic marks in mouse oocytes during natural and accelerated aging. Int J Biochem Cell Biol 2015; 67:121-7. [PMID: 25982203 DOI: 10.1016/j.biocel.2015.05.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 05/05/2015] [Accepted: 05/06/2015] [Indexed: 11/23/2022]
Abstract
Aging is a complex time-dependent biological process that takes place in every cell and organ, eventually leading to degenerative changes that affect normal biological functions. In the past decades, the number of older parents has increased significantly. While it is widely recognized that oocyte aging poses higher birth and reproductive risk, the exact molecular mechanisms remain largely elusive. DNA methylation of 5-cytosine (5mC) and histone modifications are among the key epigenetic mechanisms involved in critical developmental processes and have been linked to aging. However, the impact of oocyte aging on DNA demethylation pathways has not been examined. The recent discovery of Ten-Eleven-Translocation (TET) family proteins, thymine DNA glycosylase (TDG) and the demethylation intermediates 5hmC, 5fC and 5caC has provided novel clues to delineate the molecular mechanisms in DNA demethylation. In this study, we examined the cellular level of modified cytosines (5mC, 5hmC, 5fC and 5caC) and Tet/Tdg expression in oocytes obtained from natural and accelerated oocyte aging conditions. Here we show all the DNA demethylation marks are dynamically regulated in both aging conditions, which are associated with Tet3 over-expression and Tdg repression. Such an aberrant expression pattern was more profound in accelerated aging condition. The results suggest that DNA demethylation may be actively involved in oocyte aging and have implications for development of potential drug targets to rejuvenate aging oocytes. This article is part of a Directed Issue entitled: Epigenetics dynamics in development and disease.
Collapse
|
822
|
Pyle A, Hudson G, Wilson IJ, Coxhead J, Smertenko T, Herbert M, Santibanez-Koref M, Chinnery PF. Extreme-Depth Re-sequencing of Mitochondrial DNA Finds No Evidence of Paternal Transmission in Humans. PLoS Genet 2015; 11:e1005040. [PMID: 25973765 PMCID: PMC4431825 DOI: 10.1371/journal.pgen.1005040] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/28/2015] [Indexed: 11/18/2022] Open
Abstract
Recent reports have questioned the accepted dogma that mammalian mitochondrial DNA (mtDNA) is strictly maternally inherited. In humans, the argument hinges on detecting a signature of inter-molecular recombination in mtDNA sequences sampled at the population level, inferring a paternal source for the mixed haplotypes. However, interpreting these data is fraught with difficulty, and direct experimental evidence is lacking. Using extreme-high depth mtDNA re-sequencing up to ~1.2 million-fold coverage, we find no evidence that paternal mtDNA haplotypes are transmitted to offspring in humans, thus excluding a simple dilution mechanism for uniparental transmission of mtDNA present in all healthy individuals. Our findings indicate that an active mechanism eliminates paternal mtDNA which likely acts at the molecular level. Emerging evidence raises the possibility that human mitochondrial DNA (mtDNA) is not strictly maternally inherited, but it has not been technically possible to test this hypothesis directly. We identified trios with discordant mtDNA haplotypes, parent-offspring trios were validated using polymorphic microsatellites, and then used extreme-high depth mtDNA re-sequencing to look for paternally transmitted mtDNA. Despite having up to ~1.2 million-fold coverage of mtDNA, we find no evidence that paternal mtDNA haplotypes are transmitted to offspring in humans. Our findings exclude a simple dilution mechanism for uniparental transmission of mtDNA present in all healthy individuals.
Collapse
Affiliation(s)
- Angela Pyle
- Wellcome Trust Centre for Mitochondrial Research, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Gavin Hudson
- Wellcome Trust Centre for Mitochondrial Research, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Ian J. Wilson
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Jonathan Coxhead
- Wellcome Trust Centre for Mitochondrial Research, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Tania Smertenko
- Wellcome Trust Centre for Mitochondrial Research, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Mary Herbert
- Wellcome Trust Centre for Mitochondrial Research, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Mauro Santibanez-Koref
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Patrick F. Chinnery
- Wellcome Trust Centre for Mitochondrial Research, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- * E-mail:
| |
Collapse
|
823
|
Abstract
The early embryo and periconceptional period is a window during which environmental factors may cause permanent change in the pattern and characteristics of development leading to risk of adult onset disease. This has now been demonstrated across small and large animal models and also in the human. Most evidence of periconceptional 'programming' has emerged from maternal nutritional models but also other in vivo and in vitro conditions including assisted reproductive treatments, show consistent outcomes. This short review first reports on the range of environmental in vivo and in vitro periconceptional models and resulting long-term outcomes. Second, it uses the rodent maternal low protein diet model restricted to the preimplantation period and considers the stepwise maternal-embryonic dialogue that comprises the induction of programming. This dialogue leads to cellular and epigenetic responses by the embryo, mainly identified in the extra-embryonic cell lineages, and underpins an apparently permanent change in the growth trajectory during pregnancy and associates with increased cardiometabolic and behavioural disease in adulthood. We recognize the important advice of David Barker some years ago to investigate the sensitivity of the early embryo to developmental programming, an insight for which we are grateful.
Collapse
|
824
|
Affiliation(s)
- Valerio Carelli
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy
- Neurology Unit, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| |
Collapse
|
825
|
Wang S, Xia P, Rehm M, Fan Z. Autophagy and cell reprogramming. Cell Mol Life Sci 2015; 72:1699-713. [PMID: 25572296 PMCID: PMC11113636 DOI: 10.1007/s00018-014-1829-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 12/23/2014] [Accepted: 12/30/2014] [Indexed: 12/17/2022]
Abstract
Autophagy is an evolutionarily conserved process that degrades cytoplasmic components, thus contributing to cell survival and tissue homeostasis. Recent studies have demonstrated that autophagy maintains stem cells in relatively undifferentiated states (stemness) and also contributes to differentiation processes. Autophagy likewise plays a crucial role in somatic cell reprogramming, a finely regulated process that resets differentiated cells to a pluripotent state and that requires comprehensive alterations in transcriptional activities and epigenetic signatures. Autophagy assists in manifesting the functional consequences that arise from these alterations by modifying cellular protein expression profiles. The role of autophagy appears to be particularly relevant for early phases of cell reprogramming during the generation of induced pluripotent stems cells (iPSCs). In this review, we provide an overview of the core molecular machinery that constitutes the autophagic degradation system, describe the roles of autophagy in maintenance, self-renewal, and differentiation of stem cells, and discuss the autophagic process and its regulation during cell reprogramming.
Collapse
Affiliation(s)
- Shuo Wang
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Pengyan Xia
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Markus Rehm
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Zusen Fan
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| |
Collapse
|
826
|
Abstract
Because of their high-energy metabolism, neurons are strictly dependent on mitochondria, which generate cellular ATP through oxidative phosphorylation. The mitochondrial genome encodes for critical components of the oxidative phosphorylation pathway machinery, and therefore, mutations in mitochondrial DNA (mtDNA) cause energy production defects that frequently have severe neurological manifestations. Here, we review the principles of mitochondrial genetics and focus on prototypical mitochondrial diseases to illustrate how primary defects in mtDNA or secondary defects in mtDNA due to nuclear genome mutations can cause prominent neurological and multisystem features. In addition, we discuss the pathophysiological mechanisms underlying mitochondrial diseases, the cellular mechanisms that protect mitochondrial integrity, and the prospects for therapy.
Collapse
Affiliation(s)
- Valerio Carelli
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy; Neurology Unit, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
827
|
Zhang H, Baehrecke EH. Eaten alive: novel insights into autophagy from multicellular model systems. Trends Cell Biol 2015; 25:376-87. [PMID: 25862458 DOI: 10.1016/j.tcb.2015.03.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/12/2015] [Accepted: 03/11/2015] [Indexed: 11/29/2022]
Abstract
Autophagy delivers cytoplasmic material to lysosomes for degradation. First identified in yeast, the core genes that control this process are conserved in higher organisms. Studies of mammalian cell cultures have expanded our understanding of the core autophagy pathway, but cannot reveal the unique animal-specific mechanisms for the regulation and function of autophagy. Multicellular organisms have different types of cells that possess distinct composition, morphology, and organization of intracellular organelles. In addition, the autophagic machinery integrates signals from other cells and environmental conditions to maintain cell, tissue and organism homeostasis. Here, we highlight how studies of autophagy in flies and worms have identified novel core autophagy genes and mechanisms, and provided insight into the context-specific regulation and function of autophagy.
Collapse
Affiliation(s)
- Hong Zhang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Eric H Baehrecke
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
828
|
Huang XJ, Shen M, Wang L, Yu F, Wu W, Liu HL. Effects of tributyltin chloride on developing mouse oocytes and preimplantation embryos. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2015; 21:358-367. [PMID: 25898838 DOI: 10.1017/s1431927615000161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Tributyltin, an organotin, is ubiquitous in estuaries and freshwater systems. Previous reports suggest that tributyltin is an endocrine disruptor in many wildlife species and it inhibits aromatase in mammalian placental and granulosa-like tumor cell lines. However, no evidence showing the effects of tributyltin on oocytes or preimplantation embryonic developmental competence exists. Therefore, we investigated the role of tributyltin chloride (TBTCl) in the development of female oocytes and preimplantation embryos. Briefly, female ICR mice were gavaged with 0 (vehicle), 4, and 8 mg/kg of TBTCl each day for 18 days. The fluorescence intensity analysis showed that the 5-methylcytosine level decreased after TBTCl treatment, indicating that the general DNA methylation level decreased in the treated oocytes. Our results demonstrate that TBTCl treatment results in decreased mRNA levels of imprinted genes H19, Igf2r, and Peg3 during oocyte growth. The TBTCl-treated oocytes showed a significant increase in reactive oxygen species levels in germinal vesicle oocytes. In TBTCl-treated oocytes, there was no difference in GPx and Sod1 expression, but a decreased mRNA level of Cat occurred when compared with control. Moreover, the blastocysts with TBTCl exposure displayed higher apoptotic signals. These results suggest that TBTCl induces developmental defects in oocytes and preimplantation embryos.
Collapse
Affiliation(s)
- Xian-Ju Huang
- College of Animal Science and Technology,Nanjing Agricultural University,Weigang No. 1,Nanjing 210095,China
| | - Ming Shen
- College of Animal Science and Technology,Nanjing Agricultural University,Weigang No. 1,Nanjing 210095,China
| | - Lizhong Wang
- College of Animal Science and Technology,Nanjing Agricultural University,Weigang No. 1,Nanjing 210095,China
| | - Fengxiang Yu
- College of Animal Science and Technology,Nanjing Agricultural University,Weigang No. 1,Nanjing 210095,China
| | - Wangjun Wu
- College of Animal Science and Technology,Nanjing Agricultural University,Weigang No. 1,Nanjing 210095,China
| | - Hong-Lin Liu
- College of Animal Science and Technology,Nanjing Agricultural University,Weigang No. 1,Nanjing 210095,China
| |
Collapse
|
829
|
Velazquez MA. Impact of maternal malnutrition during the periconceptional period on mammalian preimplantation embryo development. Domest Anim Endocrinol 2015; 51:27-45. [PMID: 25498236 DOI: 10.1016/j.domaniend.2014.10.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 10/27/2014] [Accepted: 10/28/2014] [Indexed: 02/07/2023]
Abstract
During episodes of undernutrition and overnutrition the mammalian preimplantation embryo undergoes molecular and metabolic adaptations to cope with nutrient deficits or excesses. Maternal adaptations also take place to keep a nutritional microenvironment favorable for oocyte development and embryo formation. This maternal-embryo communication takes place via several nutritional mediators. Although adaptive responses to malnutrition by both the mother and the embryo may ensure blastocyst formation, the resultant quality of the embryo can be compromised, leading to early pregnancy failure. Still, studies have shown that, although early embryonic mortality can be induced during malnutrition, the preimplantation embryo possesses an enormous plasticity that allows it to implant and achieve a full-term pregnancy under nutritional stress, even in extreme cases of malnutrition. This developmental strategy, however, may come with a price, as shown by the adverse developmental programming induced by even subtle nutritional challenges exerted exclusively during folliculogenesis and the preimplantation period, resulting in offspring with a higher risk of developing deleterious phenotypes in adulthood. Overall, current evidence indicates that malnutrition during the periconceptional period can induce cellular and molecular alterations in preimplantation embryos with repercussions for fertility and postnatal health.
Collapse
Affiliation(s)
- M A Velazquez
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK.
| |
Collapse
|
830
|
Global DNA methylation was changed by a maternal high-lipid, high-energy diet during gestation and lactation in male adult mice liver. Br J Nutr 2015; 113:1032-9. [DOI: 10.1017/s0007114515000252] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An epigenetic mechanism has been suggested to explain the effects of the maternal diet on the development of disease in offspring. The present study aimed to observe the effects of a maternal high-lipid, high-energy (HLE) diet on the DNA methylation pattern of male offspring in mice. Female C57BL/6J mice were fed an HLE diet during gestation and lactation. The genomic DNA methylations at promoter sites of genes in the liver, mRNA and protein levels of selected genes related to lipid and glucose metabolism were measured by microarray, real-time PCR and Western blot. The results indicated that the percentage of methylated DNA in offspring from dams that were fed an HLE diet was significantly higher than that from dams that were fed a chow diet, and most of these genes were hypermethylated in promoter regions. The nuclear protein content and mRNA levels of hypermethylated genes, such as PPARγ and liver X receptor α (LXRα), were decreased significantly in offspring in the HLE group. The results suggested that the DNA methylation profile in adult offspring livers was changed by the maternal HLE diet during gestation and lactation.
Collapse
|
831
|
Lauand C, Niero EL, Dias VM, Machado-Santelli GM. Cell cycle synchronization and BrdU incorporation as a tool to study the possible selective elimination of ErbB1 gene in the micronuclei in A549 cells. ACTA ACUST UNITED AC 2015; 48:382-91. [PMID: 25760027 PMCID: PMC4445660 DOI: 10.1590/1414-431x20144262] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 12/10/2014] [Indexed: 11/22/2022]
Abstract
Lung cancer often exhibits molecular changes, such as the overexpression of the
ErbB1 gene that encodes epidermal growth factor receptor (EGFR).
ErbB1 amplification and mutation are associated with tumor
aggressiveness and low response to therapy. The aim of the present study was to
design a schedule to synchronize the cell cycle of A549 cell line (a non-small cell
lung cancer) and to analyze the possible association between the micronuclei (MNs)
and the extrusion of ErbB1 gene extra-copies. After double blocking,
by the process of fetal bovine serum deprivation and vincristine treatment, MNs
formation was monitored with 5-bromo-2-deoxyuridine (BrdU) incorporation, which is an
S-phase marker. Statistical analyses allowed us to infer that MNs may arise both in
mitosis as well as in interphase. The MNs were able to replicate their DNA and this
process seemed to be non-synchronous with the main cell nuclei. The presence of
ErbB1 gene in the MNs was evaluated by fluorescent in
situ hybridization (FISH). ErbB1 sequences were detected
in the MNs, but a relation between the MNs formation and extrusion of amplified
ErbB1could not be established. The present study sought to
elucidate the meaning of MNs formation and its association with the elimination of
oncogenes or other amplified sequences from the tumor cells.
Collapse
Affiliation(s)
- C Lauand
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - E L Niero
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - V M Dias
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - G M Machado-Santelli
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
832
|
Ney PA. Mitochondrial autophagy: Origins, significance, and role of BNIP3 and NIX. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2775-83. [PMID: 25753537 DOI: 10.1016/j.bbamcr.2015.02.022] [Citation(s) in RCA: 240] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 01/24/2015] [Accepted: 02/26/2015] [Indexed: 12/24/2022]
Abstract
Mitochondrial autophagy (mitophagy) is a core cellular activity. In this review, we consider mitophagy and related cellular processes and discuss their significance for human disease. Strong parallels exist between mitophagy and xenophagy employed in host defense. These mechanisms converge on receptors in the innate immune system in clinically relevant scenarios. Mitophagy is part of a cellular quality control mechanism, which is implicated in degenerative disease, especially neurodegenerative disease. Furthermore, mitophagy is an aspect of cellular remodeling, which is employed during development. BNIP3 and NIX are related multi-functional outer mitochondrial membrane proteins. BNIP3 regulates mitophagy during hypoxia, whereas NIX is required for mitophagy during development of the erythroid lineage. Recent advances in the field of BNIP3- and NIX-mediated mitophagy are discussed.
Collapse
Affiliation(s)
- Paul A Ney
- Department of Cell & Molecular Biology, Lindsley F. Kimball Research Institute, New York Blood Center, 310 East 67 Street, New York, NY 10065-6275, USA.
| |
Collapse
|
833
|
Coticchio G, Dal Canto M, Mignini Renzini M, Guglielmo MC, Brambillasca F, Turchi D, Novara PV, Fadini R. Oocyte maturation: gamete-somatic cells interactions, meiotic resumption, cytoskeletal dynamics and cytoplasmic reorganization. Hum Reprod Update 2015; 21:427-54. [PMID: 25744083 DOI: 10.1093/humupd/dmv011] [Citation(s) in RCA: 348] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 02/11/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND In a growth phase occurring during most of folliculogenesis, the oocyte produces and accumulates molecules and organelles that are fundamental for the development of the preimplantation embryo. At ovulation, growth is followed by a phase of maturation that, although confined within a short temporal window, encompasses modifications of the oocyte chromosome complement and rearrangements of cytoplasmic components that are crucial for the achievement of developmental competence. Cumulus cells (CCs) are central to the process of maturation, providing the oocyte with metabolic support and regulatory cues. METHODS PubMed was used to search the MEDLINE database for peer-reviewed original articles and reviews concerning oocyte maturation in mammals. Searches were performed adopting 'oocyte' and 'maturation' as main terms, in association with other keywords expressing concepts relevant to the subject. The most relevant publications, i.e. those concerning major phenomena occurring during oocyte maturation in established experimental models and the human species, were assessed and discussed critically to offer a comprehensive description of the process of oocyte maturation. RESULTS By applying the above described search criteria, 6165 publications were identified, of which 543 were review articles. The number of publications increased steadily from 1974 (n = 7) to 2013 (n = 293). In 2014, from January to the time of submission of this manuscript, 140 original manuscripts and reviews were published. The studies selected for this review extend previous knowledge and shed new and astounding knowledge on oocyte maturation. It has long been known that resumption of meiosis and progression to the metaphase II stage is intrinsic to oocyte maturation, but novel findings have revealed that specific chromatin configurations are indicative of a propensity of the oocyte to resume the meiotic process and acquire developmental competence. Recently, genetic integrity has also been characterized as a factor with important implications for oocyte maturation and quality. Changes occurring in the cytoplasmic compartment are equally fundamental. Microtubules, actin filaments and chromatin not only interact to finalize chromosome segregation, but also crucially co-operate to establish cell asymmetry. This allows polar body extrusion to be accomplished with minimal loss of cytoplasm. The cytoskeleton also orchestrates the rearrangement of organelles in preparation for fertilization. For example, during maturation the distribution of the endoplasmic reticulum undergoes major modifications guided by microtubules and microfilaments to make the oocyte more competent in the generation of intracellular Ca(2+) oscillations that are pivotal for triggering egg activation. Cumulus cells are inherent to the process of oocyte maturation, emitting regulatory signals via direct cell-to-cell contacts and paracrine factors. In addition to nurturing the oocyte with key metabolites, CCs regulate meiotic resumption and modulate the function of the oocyte cytoskeleton. CONCLUSIONS Although the importance of oocyte maturation for the achievement of female meiosis has long been recognized, until recently much less was known of the significance of this process in relation to other fundamental developmental events. Studies on chromatin dynamics and integrity have extended our understanding of female meiosis. Concomitantly, cytoskeletal and organelle changes and the ancillary role of CCs have been better appreciated. This is expected to inspire novel concepts and advances in assisted reproduction technologies, such as the development of novel in vitro maturation systems and the identification of biomarkers of oocyte quality.
Collapse
Affiliation(s)
- Giovanni Coticchio
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| | - Mariabeatrice Dal Canto
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| | - Mario Mignini Renzini
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| | - Maria Cristina Guglielmo
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| | - Fausta Brambillasca
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| | - Diana Turchi
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| | - Paola Vittoria Novara
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| | - Rubens Fadini
- Biogenesi Reproductive Medicine Centre, Istituti Clinici Zucchi, Via Zucchi 24, 20900 Monza, Italy
| |
Collapse
|
834
|
Weckert E. The potential of future light sources to explore the structure and function of matter. IUCRJ 2015; 2:230-45. [PMID: 25866660 PMCID: PMC4392416 DOI: 10.1107/s2052252514024269] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 11/03/2014] [Indexed: 05/15/2023]
Abstract
Structural studies in general, and crystallography in particular, have benefited and still do benefit dramatically from the use of synchrotron radiation. Low-emittance storage rings of the third generation provide focused beams down to the micrometre range that are sufficiently intense for the investigation of weakly scattering crystals down to the size of several micrometres. Even though the coherent fraction of these sources is below 1%, a number of new imaging techniques have been developed to exploit the partially coherent radiation. However, many techniques in nanoscience are limited by this rather small coherent fraction. On the one hand, this restriction limits the ability to study the structure and dynamics of non-crystalline materials by methods that depend on the coherence properties of the beam, like coherent diffractive imaging and X-ray correlation spectroscopy. On the other hand, the flux in an ultra-small diffraction-limited focus is limited as well for the same reason. Meanwhile, new storage rings with more advanced lattice designs are under construction or under consideration, which will have significantly smaller emittances. These sources are targeted towards the diffraction limit in the X-ray regime and will provide roughly one to two orders of magnitude higher spectral brightness and coherence. They will be especially suited to experiments exploiting the coherence properties of the beams and to ultra-small focal spot sizes in the regime of several nanometres. Although the length of individual X-ray pulses at a storage-ring source is of the order of 100 ps, which is sufficiently short to track structural changes of larger groups, faster processes as they occur during vision or photosynthesis, for example, are not accessible in all details under these conditions. Linear accelerator (linac) driven free-electron laser (FEL) sources with extremely short and intense pulses of very high coherence circumvent some of the limitations of present-day storage-ring sources. It has been demonstrated that their individual pulses are short enough to outrun radiation damage for single-pulse exposures. These ultra-short pulses also enable time-resolved studies 1000 times faster than at standard storage-ring sources. Developments are ongoing at various places for a totally new type of X-ray source combining a linac with a storage ring. These energy-recovery linacs promise to provide pulses almost as short as a FEL, with brilliances and multi-user capabilities comparable with a diffraction-limited storage ring. Altogether, these new X-ray source developments will provide smaller and more intense X-ray beams with a considerably higher coherent fraction, enabling a broad spectrum of new techniques for studying the structure of crystalline and non-crystalline states of matter at atomic length scales. In addition, the short X-ray pulses of FELs will enable the study of fast atomic dynamics and non-equilibrium states of matter.
Collapse
|
835
|
The effect of maternal body condition on in vivo production of zygotes and behavior of delivered offspring in mice. Theriogenology 2015; 83:577-89. [DOI: 10.1016/j.theriogenology.2014.10.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 10/21/2014] [Accepted: 10/25/2014] [Indexed: 11/18/2022]
|
836
|
Ge ZJ, Schatten H, Zhang CL, Sun QY. Oocyte ageing and epigenetics. Reproduction 2015; 149:R103-R114. [PMID: 25391845 PMCID: PMC4397590 DOI: 10.1530/rep-14-0242] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 11/07/2014] [Accepted: 11/12/2014] [Indexed: 12/13/2022]
Abstract
It has become a current social trend for women to delay childbearing. However, the quality of oocytes from older females is compromised and the pregnancy rate of older women is lower. With the increased rate of delayed childbearing, it is becoming more and more crucial to understand the mechanisms underlying the compromised quality of oocytes from older women, including mitochondrial dysfunctions, aneuploidy and epigenetic changes. Establishing proper epigenetic modifications during oogenesis and early embryo development is an important aspect in reproduction. The reprogramming process may be influenced by external and internal factors that result in improper epigenetic changes in germ cells. Furthermore, germ cell epigenetic changes might be inherited by the next generations. In this review, we briefly summarise the effects of ageing on oocyte quality. We focus on discussing the relationship between ageing and epigenetic modifications, highlighting the epigenetic changes in oocytes from advanced-age females and in post-ovulatory aged oocytes as well as the possible underlying mechanisms.
Collapse
Affiliation(s)
- Zhao-Jia Ge
- Reproductive Medicine CenterHenan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province 450003, People's Republic of ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, #1 Beichen West Road, Chaoyang District, Beijing 100101, People's Republic of ChinaReproductive Medicine CenterPeople's Hospital of Zhengzhou University, Zhengzhou, Henan Province 450003, People's Republic of ChinaDepartment of Veterinary PathobiologyUniversity of Missouri, Columbia, Missouri 65211, USA Reproductive Medicine CenterHenan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province 450003, People's Republic of ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, #1 Beichen West Road, Chaoyang District, Beijing 100101, People's Republic of ChinaReproductive Medicine CenterPeople's Hospital of Zhengzhou University, Zhengzhou, Henan Province 450003, People's Republic of ChinaDepartment of Veterinary PathobiologyUniversity of Missouri, Columbia, Missouri 65211, USA Reproductive Medicine CenterHenan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province 450003, People's Republic of ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, #1 Beichen West Road, Chaoyang District, Beijing 100101, People's Republic of ChinaReproductive Medicine CenterPeople's Hospital of Zhengzhou University, Zhengzhou, Henan Province 450003, People's Republic of ChinaDepartment of Veterinary PathobiologyUniversity of Missouri, Columbia, Missouri 65211, USA
| | - Heide Schatten
- Reproductive Medicine CenterHenan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province 450003, People's Republic of ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, #1 Beichen West Road, Chaoyang District, Beijing 100101, People's Republic of ChinaReproductive Medicine CenterPeople's Hospital of Zhengzhou University, Zhengzhou, Henan Province 450003, People's Republic of ChinaDepartment of Veterinary PathobiologyUniversity of Missouri, Columbia, Missouri 65211, USA
| | - Cui-Lian Zhang
- Reproductive Medicine CenterHenan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province 450003, People's Republic of ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, #1 Beichen West Road, Chaoyang District, Beijing 100101, People's Republic of ChinaReproductive Medicine CenterPeople's Hospital of Zhengzhou University, Zhengzhou, Henan Province 450003, People's Republic of ChinaDepartment of Veterinary PathobiologyUniversity of Missouri, Columbia, Missouri 65211, USA Reproductive Medicine CenterHenan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province 450003, People's Republic of ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, #1 Beichen West Road, Chaoyang District, Beijing 100101, People's Republic of ChinaReproductive Medicine CenterPeople's Hospital of Zhengzhou University, Zhengzhou, Henan Province 450003, People's Republic of ChinaDepartment of Veterinary PathobiologyUniversity of Missouri, Columbia, Missouri 65211, USA
| | - Qing-Yuan Sun
- Reproductive Medicine CenterHenan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province 450003, People's Republic of ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, #1 Beichen West Road, Chaoyang District, Beijing 100101, People's Republic of ChinaReproductive Medicine CenterPeople's Hospital of Zhengzhou University, Zhengzhou, Henan Province 450003, People's Republic of ChinaDepartment of Veterinary PathobiologyUniversity of Missouri, Columbia, Missouri 65211, USA
| |
Collapse
|
837
|
Endoplasmic reticulum stress signaling in mammalian oocytes and embryos: life in balance. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 316:227-65. [PMID: 25805126 DOI: 10.1016/bs.ircmb.2015.01.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mammalian oocytes and embryos are exquisitely sensitive to a wide range of insults related to physical stress, chemical exposure, and exposures to adverse maternal nutrition or health status. Although cells manifest specific responses to various stressors, many of these stressors intersect at the endoplasmic reticulum (ER), where disruptions in protein folding and production of reactive oxygen species initiate downstream signaling events. These signals modulate mRNA translation and gene transcription, leading to recovery, activation of autophagy, or with severe and prolonged stress, apoptosis. ER stress signaling has recently come to the fore as a major contributor to embryo demise. Accordingly, agents that modulate or inhibit ER stress signaling have yielded beneficial effects on embryo survival and long-term developmental potential. We review here the mechanisms of ER stress signaling, their connections to mammalian oocytes and embryos, and the promising indications that interventions in this pathway may provide new opportunities for improving mammalian reproduction and health.
Collapse
|
838
|
Wagner-Vogel G, Lämmer F, Kämper J, Basse CW. Uniparental mitochondrial DNA inheritance is not affected in Ustilago maydis Δatg11 mutants blocked in mitophagy. BMC Microbiol 2015; 15:23. [PMID: 25652096 PMCID: PMC4326477 DOI: 10.1186/s12866-015-0358-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/22/2015] [Indexed: 11/18/2022] Open
Abstract
Background Maternal or uniparental inheritance (UPI) of mitochondria is generally observed in sexual eukaryotes, however, the underlying mechanisms are diverse and largely unknown. Recently, based on the use of mutants blocked in autophagy, it has been demonstrated that autophagy is required for strict maternal inheritance in the nematode Caenorhabditis elegans. Uniparental mitochondrial DNA (mtDNA) inheritance has been well documented for numerous fungal species, and in particular, has been shown to be genetically governed by the mating-type loci in the isogamous species Cryptococcus neoformans, Phycomyces blakesleeanus and Ustilago maydis. Previously, we have shown that the a2 mating-type locus gene lga2 is decisive for UPI during sexual development of U. maydis. In axenic culture, conditional overexpression of lga2 triggers efficient loss of mtDNA as well as mitophagy. To assess a functional relationship, we have investigated UPI in U. maydis Δatg11 mutants, which are blocked in mitophagy. Results This study has revealed that Δatg11 mutants are not affected in pathogenic development and this has allowed us to analyse UPI under comparable developmental conditions between mating-compatible wild-type and mutant strain combinations. Explicitly, we have examined two independent strain combinations that gave rise to different efficiencies of UPI. We demonstrate that in both cases UPI is atg11-independent, providing evidence that mitophagy is not critical for UPI in U. maydis, even under conditions of strict UPI. Conclusions Until now, analysis of a role of mitophagy in UPI has not been reported for microbial species. Our study suggests that selective autophagy does not contribute to UPI in U. maydis, but is rather a consequence of selective mtDNA elimination in response to mitochondrial damage. Electronic supplementary material The online version of this article (doi:10.1186/s12866-015-0358-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gaby Wagner-Vogel
- Department of Genetics, Institute for Applied Biosciences of the Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| | - Frauke Lämmer
- Department of Genetics, Institute for Applied Biosciences of the Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| | - Jörg Kämper
- Department of Genetics, Institute for Applied Biosciences of the Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| | - Christoph W Basse
- Department of Genetics, Institute for Applied Biosciences of the Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| |
Collapse
|
839
|
Abstract
There is now considerable epidemiological and experimental evidence indicating that early-life environmental conditions, including nutrition, affect subsequent development in later life. These conditions induce highly integrated responses in endocrine-related homeostasis, resulting in persistent changes in the developmental trajectory producing an altered adult phenotype. Early-life events trigger processes that prepare the individual for particular circumstances that are anticipated in the postnatal environment. However, where the intrauterine and postnatal environments differ markedly, such modifications to the developmental trajectory may prove maladaptive in later life. Reproductive maturation and function are similarly influenced by early-life events. This should not be surprising, because the primordial follicle pool is established early in life and is thus vulnerable to early-life events. Results of clinical and experimental studies have indicated that early-life adversity is associated with a decline in ovarian follicular reserve, changes in ovulation rates, and altered age at onset of puberty. However, the underlying mechanisms regulating the relationship between the early-life developmental environment and postnatal reproductive development and function are unclear. This review examines the evidence linking early-life nutrition and effects on the female reproductive system, bringing together clinical observations in humans and experimental data from targeted animal models.
Collapse
Affiliation(s)
- K A Chan
- Departments of Biochemistry and Biomedical SciencesPediatricsObstetrics and GynecologyMcMaster University, 1280 Main Street West HSC 4H30A, Hamilton, Ontario, Canada L8S 4K1
| | - M W Tsoulis
- Departments of Biochemistry and Biomedical SciencesPediatricsObstetrics and GynecologyMcMaster University, 1280 Main Street West HSC 4H30A, Hamilton, Ontario, Canada L8S 4K1
| | - D M Sloboda
- Departments of Biochemistry and Biomedical SciencesPediatricsObstetrics and GynecologyMcMaster University, 1280 Main Street West HSC 4H30A, Hamilton, Ontario, Canada L8S 4K1 Departments of Biochemistry and Biomedical SciencesPediatricsObstetrics and GynecologyMcMaster University, 1280 Main Street West HSC 4H30A, Hamilton, Ontario, Canada L8S 4K1 Departments of Biochemistry and Biomedical SciencesPediatricsObstetrics and GynecologyMcMaster University, 1280 Main Street West HSC 4H30A, Hamilton, Ontario, Canada L8S 4K1
| |
Collapse
|
840
|
Lane M, Zander-Fox DL, Robker RL, McPherson NO. Peri-conception parental obesity, reproductive health, and transgenerational impacts. Trends Endocrinol Metab 2015; 26:84-90. [PMID: 25523615 DOI: 10.1016/j.tem.2014.11.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 11/16/2014] [Accepted: 11/20/2014] [Indexed: 12/20/2022]
Abstract
Maternal over-nutrition during pregnancy is a risk factor for pregnancy complications and is increasingly associated with adverse childhood outcomes such as increased propensity for obesity and metabolic disease. However, there is emerging evidence that parental lifestyle factors prior to and at conception have a powerful impact on the health of the offspring for more than one generation. Maternal and paternal obesity prior to conception alters the molecular composition of both oocytes and sperm, which can partly escape epigenetic reprogramming at fertilization, altering the developmental trajectory of the resultant embryo, ultimately increasing the incidence of obesity and metabolic disorders in offspring. Understanding the molecular underpinning of these changes may help create interventions to reduce the risk of disease in future generations.
Collapse
Affiliation(s)
- Michelle Lane
- Robinson Research Institute, School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, South Australia; Monash IVF Group, Richmond, Victoria, Australia; Freemasons Centre for Men's Health, University of Adelaide, Adelaide, South Australia.
| | - Deirdre L Zander-Fox
- Robinson Research Institute, School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, South Australia; Repromed, Dulwich, South Australia
| | - Rebecca L Robker
- Robinson Research Institute, School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, South Australia
| | - Nicole O McPherson
- Robinson Research Institute, School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, South Australia; Repromed, Dulwich, South Australia; Freemasons Centre for Men's Health, University of Adelaide, Adelaide, South Australia
| |
Collapse
|
841
|
Zhou LQ, Dean J. Reprogramming the genome to totipotency in mouse embryos. Trends Cell Biol 2015; 25:82-91. [PMID: 25448353 PMCID: PMC4312727 DOI: 10.1016/j.tcb.2014.09.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/17/2014] [Accepted: 09/23/2014] [Indexed: 02/03/2023]
Abstract
Despite investigative interest, the artificial derivation of pluripotent stem cells remains inefficient and incomplete reprogramming hinders its potential as a reliable tool in regenerative medicine. By contrast, fusion of terminally differentiated gametes at fertilization activates efficient epigenetic reprogramming to ensure totipotency of early embryos. Understanding the epigenetic mechanisms required for the transition from the fertilized egg to the embryo can improve efforts to reprogram differentiated cells to pluripotent/totipotent cells for therapeutic use. We review recent discoveries that are providing insight into the molecular mechanisms required for epigenetic reprogramming to totipotency in vivo.
Collapse
Affiliation(s)
- Li-quan Zhou
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jurrien Dean
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
842
|
Brenseke B, Bahamonde J, Talanian M, Kornfeind E, Daly J, Cobb G, Zhang J, Prater MR, Davis GC, Good DJ. Mitigating or exacerbating effects of maternal-fetal programming of female mice through the food choice environment. Endocrinology 2015; 156:182-92. [PMID: 25386832 PMCID: PMC4272389 DOI: 10.1210/en.2014-1523] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Humans live, eat, and become overweight/obese in complex surroundings where there are many available food choices. Prenatal exposure to poor food choices predisposes offspring to increased negative health risks, including obesity. Many animal experiments have analyzed intergenerational body weight parameters in an environment without food choices, which may not be directly translatable to the human food environment. In this study, offspring from mothers with a defined high-fat diet (HFD) or low-fat diet (LFD) were arbitrarily assigned to either an exclusively LFD or HFD or to a diet where they have a choice between LFD and HFD (choice diet). Offspring displayed negative outcomes of increased body weight, body fat, serum leptin, and blood glucose levels when given the choice diet compared with offspring on the LFD. Conversely, improved energy expenditure was found for offspring given the choice diet compared with offspring from HFD dams given LFD. In addition, maternal diet-specific influences on offspring metabolic parameters were identified, especially in offspring from HFD dams, including positive outcomes of reduced leptin in LFD offspring, reduced corticosterone and cholesterol levels in HFD offspring, and increased exercise levels in choice offspring, as well as the negative outcome of increased calorie intake in LFD offspring from HFD dams. This defined model can now be used as the basis for future studies to characterize the cycle of inter- and intragenerational obesity and whether more realistic diet environments, especially those including choice, can mitigate phenotype.
Collapse
Affiliation(s)
- Bonnie Brenseke
- Departments of Biomedical Sciences and Pathobiology (B.B., J.B., E.K., M.R.P.), Agricultural and Applied Economics (G.C.D.), and Human Nutrition, Foods and Exercise (M.T., J.D., G.C., J.Z., D.J.G.),Virginia Tech, Blacksburg, Virginia 24061; and Department of Biomedical Sciences (M.R.P.), Edward Via College of Osteopathic Medicine, Blacksburg, Virginia 24060
| | | | | | | | | | | | | | | | | | | |
Collapse
|
843
|
Paganelli L, Caillaud MC, Quentin M, Damiani I, Govetto B, Lecomte P, Karpov PA, Abad P, Chabouté ME, Favery B. Three BUB1 and BUBR1/MAD3-related spindle assembly checkpoint proteins are required for accurate mitosis in Arabidopsis. THE NEW PHYTOLOGIST 2015; 205:202-15. [PMID: 25262777 DOI: 10.1111/nph.13073] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 07/27/2014] [Indexed: 05/13/2023]
Abstract
The spindle assembly checkpoint (SAC) is a refined surveillance mechanism which ensures that chromosomes undergoing mitosis do not segregate until they are properly attached to the spindle microtubules (MT). The SAC has been extensively studied in metazoans and yeast, but little is known about its role in plants. We identified proteins interacting with a MT-associated protein MAP65-3, which plays a critical role in organising mitotic MT arrays, and carried out a functional analysis of previously and newly identified SAC components. We show that Arabidopsis SAC proteins BUB3.1, MAD2, BUBR1/MAD3s and BRK1 interact with each other and with MAP65-3. We found that two BUBR1/MAD3s interacted specifically at centromeres. When stably expressed in Arabidopsis, BRK1 localised to the kinetochores during all stages of the mitotic cell cycle. Early in mitosis, BUB3.1 and BUBR1/MAD3.1 localise to the mitotic spindle, where MAP65-3 organises spindle MTs. A double-knockout mad3.1 mad3.2 mutant presented spindle MT abnormalities, chromosome misalignments on the metaphase plate and the production of lagging chromosomes and micronuclei during mitosis. We conclude that BRK1 and BUBR1/MAD3-related proteins play a key role in ensuring faithful chromosome segregation during mitosis and that their interaction with MAP65-3 may be important for the regulation of MT-chromosome attachment.
Collapse
Affiliation(s)
- Laetitia Paganelli
- UMR 1355, Institut Sophia Agrobiotech, INRA, 400 route des Chappes, F-06903, Sophia-Antipolis, France; UMR 7254, CNRS, 400 route des Chappes, F-06903, Sophia-Antipolis, France; UMR 1355, Université de Nice Sophia-Antipolis, 400 route des Chappes, F-06903, Sophia-Antipolis, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
844
|
Zhang T, Zhou Y, Qi ST, Wang ZB, Qian WP, Ouyang YC, Shen W, Schatten H, Sun QY. Nuf2 is required for chromosome segregation during mouse oocyte meiotic maturation. Cell Cycle 2015; 14:2701-10. [PMID: 26054848 PMCID: PMC4613995 DOI: 10.1080/15384101.2015.1058677] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/07/2015] [Accepted: 05/16/2015] [Indexed: 10/23/2022] Open
Abstract
Nuf2 plays an important role in kinetochore-microtubule attachment and thus is involved in regulation of the spindle assembly checkpoint in mitosis. In this study, we examined the localization and function of Nuf2 during mouse oocyte meiotic maturation. Myc6-Nuf2 mRNA injection and immunofluorescent staining showed that Nuf2 localized to kinetochores from germinal vesicle breakdown to metaphase I stages, while it disappeared from the kinetochores at the anaphase I stage, but relocated to kinetochores at the MII stage. Overexpression of Nuf2 caused defective spindles, misaligned chromosomes, and activated spindle assembly checkpoint, and thus inhibited chromosome segregation and metaphase-anaphase transition in oocyte meiosis. Conversely, precocious polar body extrusion was observed in the presence of misaligned chromosomes and abnormal spindle formation in Nuf2 knock-down oocytes, causing aneuploidy. Our data suggest that Nuf2 is a critical regulator of meiotic cell cycle progression in mammalian oocytes.
Collapse
Affiliation(s)
- Teng Zhang
- Institute of Reproductive Sciences; College of Animal Science and Technology; Qingdao Agricultural University; Qingdao, China
- State Key Laboratory of Reproductive Biology; Institute of Zoology; Chinese Academy of Sciences; Beijing, China
| | - Yang Zhou
- State Key Laboratory of Reproductive Biology; Institute of Zoology; Chinese Academy of Sciences; Beijing, China
| | - Shu-Tao Qi
- State Key Laboratory of Reproductive Biology; Institute of Zoology; Chinese Academy of Sciences; Beijing, China
| | - Zhen-Bo Wang
- State Key Laboratory of Reproductive Biology; Institute of Zoology; Chinese Academy of Sciences; Beijing, China
| | - Wei-Ping Qian
- Department of Reproductive Medicine; Peking University Shenzhen Hospital; Medical Center of Peking University; Shenzhen, Guangdong, China
| | - Ying-Chun Ouyang
- State Key Laboratory of Reproductive Biology; Institute of Zoology; Chinese Academy of Sciences; Beijing, China
| | - Wei Shen
- Institute of Reproductive Sciences; College of Animal Science and Technology; Qingdao Agricultural University; Qingdao, China
| | - Heide Schatten
- Department of Veterinary Pathobiology; University of Missouri; Columbia, MO USA
| | - Qing-Yuan Sun
- Institute of Reproductive Sciences; College of Animal Science and Technology; Qingdao Agricultural University; Qingdao, China
- State Key Laboratory of Reproductive Biology; Institute of Zoology; Chinese Academy of Sciences; Beijing, China
| |
Collapse
|
845
|
Autophagy and ubiquitin-mediated proteolysis may not be involved in the degradation of spermatozoon mitochondria in mouse and porcine early embryos. ZYGOTE 2014; 24:31-41. [PMID: 25513816 DOI: 10.1017/s0967199414000689] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The mitochondrial genome is maternally inherited in animals, despite the fact that paternal mitochondria enter oocytes during fertilization. Autophagy and ubiquitin-mediated degradation are responsible for the elimination of paternal mitochondria in Caenorhabditis elegans; however, the involvement of these two processes in the degradation of paternal mitochondria in mammals is not well understood. We investigated the localization patterns of light chain 3 (LC3) and ubiquitin in mouse and porcine embryos during preimplantation development. We found that LC3 and ubiquitin localized to the spermatozoon midpiece at 3 h post-fertilization, and that both proteins were colocalized with paternal mitochondria and removed upon fertilization during the 4-cell stage in mouse and the zygote stage in porcine embryos. Sporadic paternal mitochondria were present beyond the morula stage in the mouse, and paternal mitochondria were restricted to one blastomere of 4-cell embryos. An autophagy inhibitor, 3-methyladenine (3-MA), did not affect the distribution of paternal mitochondria compared with the positive control, while an autophagy inducer, rapamycin, accelerated the removal of paternal mitochondria compared with the control. After the intracytoplasmic injection of intact spermatozoon into mouse oocytes, LC3 and ubiquitin localized to the spermatozoon midpiece, but remnants of undegraded paternal mitochondria were retained until the blastocyst stage. Our results show that paternal mitochondria colocalize with autophagy receptors and ubiquitin and are removed after in vitro fertilization, but some remnants of sperm mitochondrial sheath may persist up to morula stage after intracytoplasmic spermatozoon injection (ICSI).
Collapse
|
846
|
Moraes CT, Bacman SR, Williams SL. Manipulating mitochondrial genomes in the clinic: playing by different rules. Trends Cell Biol 2014; 24:209-11. [PMID: 24679453 DOI: 10.1016/j.tcb.2014.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 02/10/2014] [Accepted: 02/11/2014] [Indexed: 11/19/2022]
Abstract
Recently, several publications have surfaced describing methods to manipulate mitochondrial genomes in tissues and embryos. With them, a somewhat sensationalistic uproar about the generation of children with 'three parents' has dominated the discussion in the lay media. It is important that society understands the singularities of mitochondrial genetics to judge these procedures in a rational light, so that this ongoing discussion does not preclude the helping of patients and families harboring mutated mitochondrial genomes.
Collapse
Affiliation(s)
- Carlos T Moraes
- University of Miami Miller School of Medicine, Department of Neurology and Cell Biology, 1420 NW 9th Avenue, Rm 229, Miami, FL 33136, USA.
| | - Sandra R Bacman
- University of Miami Miller School of Medicine, Department of Neurology and Cell Biology, 1420 NW 9th Avenue, Rm 229, Miami, FL 33136, USA
| | - Sion L Williams
- University of Miami Miller School of Medicine, Department of Neurology and Cell Biology, 1420 NW 9th Avenue, Rm 229, Miami, FL 33136, USA
| |
Collapse
|
847
|
Schatten H, Sun QY, Prather R. The impact of mitochondrial function/dysfunction on IVF and new treatment possibilities for infertility. Reprod Biol Endocrinol 2014; 12:111. [PMID: 25421171 PMCID: PMC4297407 DOI: 10.1186/1477-7827-12-111] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 09/04/2014] [Indexed: 11/12/2022] Open
Abstract
Mitochondria play vital roles in oocyte functions and they are critical indicators of oocyte quality which is important for fertilization and development into viable offspring. Quality-compromised oocytes are correlated with infertility, developmental disorders, reduced blastocyst cell number and embryo loss in which mitochondrial dysfunctions play a significant role. Increasingly, women affected by metabolic disorders such as diabetes or obesity and oocyte aging are seeking treatment in IVF clinics to overcome the effects of adverse metabolic conditions on mitochondrial functions and new treatments have become available to restore oocyte quality. The past decade has seen enormous advances in potential therapies to restore oocyte quality and includes dietary components and transfer of mitochondria from cells with mitochondrial integrity into mitochondria-impaired oocytes. New technologies have opened up new possibilities for therapeutic advances which will increase the success rates for IVF of oocytes from women with compromised oocyte quality.
Collapse
Affiliation(s)
- Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO USA
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100080 Beijing, China
| | - Randall Prather
- National Swine Resource and Research Center, University of Missouri, 65211 Columbia, USA
- Division of Animal Science, University of Missouri, 65211 Columbia, USA
| |
Collapse
|
848
|
Burgstaller JP, Johnston IG, Poulton J. Mitochondrial DNA disease and developmental implications for reproductive strategies. Mol Hum Reprod 2014; 21:11-22. [PMID: 25425607 PMCID: PMC4275042 DOI: 10.1093/molehr/gau090] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mitochondrial diseases are potentially severe, incurable diseases resulting from dysfunctional mitochondria. Several important mitochondrial diseases are caused by mutations in mitochondrial DNA (mtDNA), the genetic material contained within mitochondria, which is maternally inherited. Classical and modern therapeutic approaches exist to address the inheritance of mtDNA disease, but are potentially complicated by the fact that cellular mtDNA populations evolve according to poorly-understood dynamics during development and organismal lifetimes. We review these therapeutic approaches and models of mtDNA dynamics during development, and discuss the implications of recent results from these models for modern mtDNA therapies. We particularly highlight mtDNA segregation—differences in proliferative rates between different mtDNA haplotypes—as a potential and underexplored issue in such therapies. However, straightforward strategies exist to combat this and other potential therapeutic problems. In particular, we describe haplotype matching as an approach with the power to potentially ameliorate any expected issues from mtDNA incompatibility.
Collapse
Affiliation(s)
- Joerg Patrick Burgstaller
- Biotechnology in Animal Production, Department for Agrobiotechnology, IFA Tulln, 3430 Tulln, Austria Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Iain G Johnston
- Department of Mathematics, Imperial College London, London SW7 2AZ, UK
| | - Joanna Poulton
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
849
|
Wei Y, Schatten H, Sun QY. Environmental epigenetic inheritance through gametes and implications for human reproduction. Hum Reprod Update 2014; 21:194-208. [PMID: 25416302 DOI: 10.1093/humupd/dmu061] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Traditional studies focused on DNA as the heritable information carrier that passes the phenotype from parents to offspring. However, increasing evidence suggests that information, that is independent of the DNA sequence, termed epigenetic information, can be inherited between generations. Recently, in our lab, we found that prediabetes in fathers increases the susceptibility to diabetes in offspring through gametic cytosine methylation changes. Paternal prediabetes changed overall methylation patterns in sperm, and a large portion of differentially methylated loci can be transmitted to pancreatic islets of offspring up to the second generation. In this review, we survey the extensive examples of environmentally induced epigenetic inheritance in various species, ranging from Caenorhabditis elegans to humans. We focus mainly on elucidating the molecular basis of environmental epigenetic inheritance through gametes, which is an emerging theme and has important implications for explaining the prevalence of obesity, type 2 diabetes and other chronic non-genetic diseases, which is also important for understanding the influence of environmental exposures on reproductive and overall health in offspring. METHODS For this review, we included relevant data and information obtained through a PubMed database search for all English language articles published up to August 2014 which included the term 'environmental epigenetic inheritance' and 'transgenerational epigenetic inheritance'. We focused on research papers using animal models including Drosophila, C. elegans, mouse and rat. Human data were also included. RESULTS Evidence from animal models suggests that environmental epigenetic inheritance through gametes exists in various species. Extensive molecular evidence suggests that epigenetic information carriers including DNA methylation, non-coding RNAs and chromatin proteins in gametes play important roles in the transmission of phenotypes from parents to offspring. CONCLUSIONS Given the large number of experimental evidence from various organisms, it is clear that parental environmental alterations can affect the phenotypes of offspring through gametic epigenetic alterations. This more recent thinking based on new data may have implications in explaining the prevalence of obesity, type 2 diabetes and other chronic non-genetic diseases. This also implies that, in the near future, epigenetic factors which are heritable should be regarded important in determining the risk of certain diseases. Moreover, identification of epigenetic markers in gametes (polar body or sperm) may hold great promise for predicting susceptibility to and preventing certain non-genetic diseases in offspring, as well as providing indications on parental environmental exposures.
Collapse
Affiliation(s)
- Yanchang Wei
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, 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, China
| |
Collapse
|
850
|
Erpapazoglou Z, Walker O, Haguenauer-Tsapis R. Versatile roles of k63-linked ubiquitin chains in trafficking. Cells 2014; 3:1027-88. [PMID: 25396681 PMCID: PMC4276913 DOI: 10.3390/cells3041027] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/14/2014] [Accepted: 10/21/2014] [Indexed: 12/11/2022] Open
Abstract
Modification by Lys63-linked ubiquitin (UbK63) chains is the second most abundant form of ubiquitylation. In addition to their role in DNA repair or kinase activation, UbK63 chains interfere with multiple steps of intracellular trafficking. UbK63 chains decorate many plasma membrane proteins, providing a signal that is often, but not always, required for their internalization. In yeast, plants, worms and mammals, this same modification appears to be critical for efficient sorting to multivesicular bodies and subsequent lysosomal degradation. UbK63 chains are also one of the modifications involved in various forms of autophagy (mitophagy, xenophagy, or aggrephagy). Here, in the context of trafficking, we report recent structural studies investigating UbK63 chains assembly by various E2/E3 pairs, disassembly by deubiquitylases, and specifically recognition as sorting signals by receptors carrying Ub-binding domains, often acting in tandem. In addition, we address emerging and unanticipated roles of UbK63 chains in various recycling pathways that function by activating nucleators required for actin polymerization, as well as in the transient recruitment of signaling molecules at the plasma or ER membrane. In this review, we describe recent advances that converge to elucidate the mechanisms underlying the wealth of trafficking functions of UbK63 chains.
Collapse
Affiliation(s)
- Zoi Erpapazoglou
- Institut Jacques Monod-CNRS, UMR 7592, Université-Paris Diderot, Sorbonne Paris Cité, F-75205 Paris, France.
| | - Olivier Walker
- Institut des Sciences Analytiques, UMR5280, Université de Lyon/Université Lyon 1, 69100 Villeurbanne, France.
| | - Rosine Haguenauer-Tsapis
- Institut Jacques Monod-CNRS, UMR 7592, Université-Paris Diderot, Sorbonne Paris Cité, F-75205 Paris, France.
| |
Collapse
|