Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 9(1): 1-4, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28700896

ABSTRACT

Pluripotent stem cells may acquire genetic and epigenetic variants during culture following their derivation. At a conference organized by the International Stem Cell Initiative, and held at The Jackson Laboratory, Bar Harbor, Maine, October 2016, participants discussed how the appearance of such variants can be monitored and minimized and, crucially, how their significance for the safety of therapeutic applications of these cells can be assessed. A strong recommendation from the meeting was that an international advisory group should be set up to review the genetic and epigenetic changes observed in human pluripotent stem cell lines and establish a framework for evaluating the risks that they may pose for clinical use.


Subject(s)
Cytogenetic Analysis/methods , Epigenesis, Genetic , Genetic Variation , Pluripotent Stem Cells/metabolism , Regenerative Medicine , Humans , Maine , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/transplantation , Regenerative Medicine/methods , Risk Assessment
2.
Science ; 357(6352): 707-713, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28663440

ABSTRACT

Preeclampsia (PE) is a gestational hypertensive syndrome affecting between 5 and 8% of all pregnancies. Although PE is the leading cause of fetal and maternal morbidity and mortality, its molecular etiology is still unclear. Here, we show that ELABELA (ELA), an endogenous ligand of the apelin receptor (APLNR, or APJ), is a circulating hormone secreted by the placenta. Elabela but not Apelin knockout pregnant mice exhibit PE-like symptoms, including proteinuria and elevated blood pressure due to defective placental angiogenesis. In mice, infusion of exogenous ELA normalizes hypertension, proteinuria, and birth weight. ELA, which is abundant in human placentas, increases the invasiveness of trophoblast-like cells, suggesting that it enhances placental development to prevent PE. The ELA-APLNR signaling axis may offer a new paradigm for the treatment of common pregnancy-related complications, including PE.


Subject(s)
Cardiovascular Abnormalities/genetics , Carrier Proteins/genetics , Placental Hormones/genetics , Placentation/genetics , Pre-Eclampsia/genetics , Animals , Apelin/genetics , Apelin/metabolism , Birth Weight , Carrier Proteins/administration & dosage , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Female , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , Peptide Hormones , Placenta/blood supply , Placenta/metabolism , Pregnancy , Proteinuria , Signal Transduction
3.
Int J Dev Biol ; 60(10-11-12): 327-336, 2016.
Article in English | MEDLINE | ID: mdl-28000904

ABSTRACT

In 2016, a symposium was convened in Leroy C. Stevens' honor, in association with a meeting of the International Stem Cell Initiative (ISCI). ISCI, funded internationally, is composed of a group of ~100 scientists from many countries, under the leadership of Peter Andrews, who have worked together to characterize a significant number of human pluripotent stem cell lines, to monitor their genetic stability and their differentiation into mature cell types and tissues in vitro and in vivo. Those at the ISCI meeting puzzled through one of the thorniest problems in the therapeutic use of the differentiated derivatives of pluripotent stem cells for human therapy; namely, pluripotent stem cells can differentiate into any cell type in the adult organism, but they also have the capacity for unlimited self-renewal, hence if mutated they may have tumorigenic potential. The meeting considered how these cells might become genetically or epigenetically abnormal and how the safety of these cells for human therapeutic uses could be assessed and assured. The symposium was an opportunity to pay tribute to Leroy Stevens and to the basic science origins of this newest aspect of regenerative medicine. It was a time to reflect on the past and on how it can influence the future of our field.


Subject(s)
Cell Differentiation , Pluripotent Stem Cells , Regenerative Medicine , History, 20th Century , Humans , United States
4.
Development ; 143(11): 1993-9, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27246714

ABSTRACT

ß-catenin (CTNNB1) is integral to cell adhesion and to the canonical Wnt signaling pathway. The effects of maternal and zygotic CTNNB1 on embryogenesis have each been separately assessed, whereas the effect of its total absence has not. As the 'traditional' conditional Ctnnb1 knockout alleles give rise to truncated CTNNB1 fragments, we designed a new knockout allele incapable of CTNNB1 production. Mouse embryos lacking intact maternal/zygotic CTNNB1 from two knockout strains were examined in detail. Preimplantation embryos are formed, yet abnormalities in their size and shape were found throughout pre- and early postimplantation development. In the absence of the zona pellucida, embryos lacking CTNNB1 undergo fission and these separated blastomeres can become small trophoblastic vesicles, which in turn induce decidual reactions. Comparing the severity of this defective adhesion phenotype in embryos bearing the null allele with those carrying the 'traditional' knockout allele suggests a hypomorphic effect of the truncated CTNNB1 protein fragment, an important observation with possible impact on previous and future studies.


Subject(s)
Embryonic Development , beta Catenin/metabolism , Alleles , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Adhesion , Gene Deletion , Mice, Inbred C57BL , Mice, Knockout , Mutant Proteins/metabolism , Zygote/metabolism , beta Catenin/deficiency
5.
J Infect Dis ; 212 Suppl 1: S47-51, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26116732

ABSTRACT

Complete genomic reprogramming happens twice during the life of a genome, once during the formation of gametes in their parents and once after their union at fertilization. For that matter complete genomic reprogramming happens twice in the same parental cell, the oocyte, when it is forming and after it matures and receives the paternal gamete. Control of these processes in this unique single cell is epigenetic, and our understanding of it is based on information gleaned from imprinting, X chromosome inactivation, and activation and silencing of endogenous retroviruses (ERV). Activation of ERVs is attributed to demethylation of chromatin and DNA, whereas silencing requires methylation, attributed to the nuage proteins, which engage the Piwi-interacting RNA pathway and other posttranscriptional mechanisms. This reprogramming process has evolved throughout speciation because in mammals, but not fish, flies and worms, nuage-component muations affect male and female gametogenesis differentially. Transcription of ERVs is derepressed in both sexes in nuage-mutant mice, but whereas males are sterile, females are fertile. Using a proteomic approach we now report molecular interactions between nuage proteins and components of the oocyte cytoplasmic lattice and speculate how this interaction could preserve ERV/host chimeric gene products affecting female fertility.


Subject(s)
Cellular Reprogramming/genetics , Endogenous Retroviruses/genetics , Gene Expression Regulation, Developmental/genetics , Oocytes/virology , RNA, Small Interfering/genetics , Animals , Endogenous Retroviruses/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Oogenesis/genetics , RNA, Small Interfering/metabolism
6.
Stem Cells Dev ; 24(13): 1515-20, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25897827

ABSTRACT

The embryonic portion of 7-day-old mouse embryos transplanted to extrauterine sites of syngeneic adult animals gives rise to teratoid tumors, which may be either benign [teratomas (T)] or malignant [teratocarcinomas (TC)]. The incidence of embryo-derived TC varies from one mouse strain to another, indicating that some strains are TC-permissive whereas others are relatively TC-nonpermissive. Embryos of a TC-permissive mouse strain (DBA/2J) and a TC-nonpermissive one (C57BL/6J) were transplanted into NOD.Cg-Prkdc(scid) Il2rg(tm1Wjl)/SzJ (NSG) mice to determine their tumorigenic potential in the absence of functional adaptive and innate immune responses in the hosts. C57BL/6J embryos transplanted to NSG mice gave rise to TC in 31% of cases, whereas the incidence of TC produced from DBA/2J transplanted embryos was 71%. The NSG embryos transplanted to syngeneic hosts gave rise to TC in 67% of cases, allowing the classification of NSG as a TC-permissive strain. A previously reported correlation between teratocarcinoma and splenomegaly was also observed in the NSG mice. The capacity of these tumors to differentiate into the cells and tissues of the normal embryo is mapped through a detailed histological analysis. These data suggest that teratocarcinogenesis, in the absence of host innate and adaptive immunity, is largely determined by the genetic background of the embryo.


Subject(s)
Immunocompromised Host , Teratocarcinoma/pathology , Teratoma/pathology , Animals , DNA-Activated Protein Kinase/genetics , DNA-Binding Proteins/genetics , Interleukin Receptor Common gamma Subunit/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred NOD , Nuclear Proteins/genetics , Teratocarcinoma/immunology , Teratoma/immunology
7.
Article in English | MEDLINE | ID: mdl-26763985

ABSTRACT

The genetic information is largely identical across most cell types in a given organism but the epigenome, which controls expression of the genome, is cell type- and context-dependent. Although most mature mammalian cells appear to have a stable, heritable epigenome, a dynamic intricate process reshapes it as these cells transition from soma to germline and back again. During normal embryogenesis, primordial germ cells, of somatic origin, are set aside to become gametes. In doing so their genome is reprogrammed-that is, the epigenome of specific regions is replaced in a sex-specific fashion as they terminally differentiate into oocytes or spermatocytes in the gonads. Upon union of these gametes, reprogramming of the new organism's epigenome is initiated, which eventually leads, through pluripotent cells, to the cell lineages required for proper embryonic development to a sexually mature adult. This never-ending cycle of birth and rebirth is accomplished through methylation and demethylation of specific genomic sites within the gametes and pluripotent cells of an organism. This enigmatic process of natural epigenomic reprogramming is now being dissected in vivo, focusing on specific genomic regions-that is, imprinted genes and retrotransposons, where TRIM28 molecular complexes appear to guide the transition from gamete to embryo.


Subject(s)
Cellular Reprogramming/genetics , Epigenesis, Genetic/genetics , Tripartite Motif-Containing Protein 28/genetics , Animals , DNA Methylation/genetics , Embryonic Development/genetics , Female , Genome , Genomic Imprinting/genetics , Germ Cells , Humans , Male , Mammals , Retroelements/genetics
9.
Genes Dev ; 28(8): 812-28, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24736841

ABSTRACT

Methylation of DNA is an essential epigenetic control mechanism in mammals. During embryonic development, cells are directed toward their future lineages, and DNA methylation poses a fundamental epigenetic barrier that guides and restricts differentiation and prevents regression into an undifferentiated state. DNA methylation also plays an important role in sex chromosome dosage compensation, the repression of retrotransposons that threaten genome integrity, the maintenance of genome stability, and the coordinated expression of imprinted genes. However, DNA methylation marks must be globally removed to allow for sexual reproduction and the adoption of the specialized, hypomethylated epigenome of the primordial germ cell and the preimplantation embryo. Recent technological advances in genome-wide DNA methylation analysis and the functional description of novel enzymatic DNA demethylation pathways have provided significant insights into the molecular processes that prepare the mammalian embryo for normal development.


Subject(s)
Blastocyst/metabolism , Cellular Reprogramming/genetics , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Developmental , 5-Methylcytosine/metabolism , Animals , Embryo, Mammalian , Germ Cells/metabolism , Humans
10.
Stem Cell Reports ; 1(5): 379-86, 2013.
Article in English | MEDLINE | ID: mdl-24286026

ABSTRACT

Human embryonic stem cells (hESCs) regularly acquire nonrandom genomic aberrations during culture, raising concerns about their safe therapeutic application. The International Stem Cell Initiative identified a copy number variant (CNV) amplification of chromosome 20q11.21 in 25% of hESC lines displaying a normal karyotype. By comparing four cell lines paired for the presence or absence of this CNV, we show that those containing this amplicon have higher population doubling rates, attributable to enhanced cell survival through resistance to apoptosis. Of the three genes encoded within the minimal amplicon and expressed in hESCs, only overexpression of BCL2L1 (BCL-XL isoform) provides control cells with growth characteristics similar to those of CNV-containing cells, whereas inhibition of BCL-XL suppresses the growth advantage of CNV cells, establishing BCL2L1 as a driver mutation. Amplification of the 20q11.21 region is also detectable in human embryonal carcinoma cell lines and some teratocarcinomas, linking this mutation with malignant transformation.


Subject(s)
Chromosomes, Human, Pair 20/genetics , DNA Copy Number Variations , Embryonic Stem Cells/metabolism , Selection, Genetic , bcl-X Protein/metabolism , Cell Line , Gene Amplification , Genetic Loci , Humans , Mutation , bcl-X Protein/genetics
11.
Science ; 341(6150): 1110-2, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-24009393

ABSTRACT

Epigenetic alterations are increasingly recognized as causes of human cancers and disease. These aberrations are likely to arise during genomic reprogramming in mammalian preimplantation embryos, when their epigenomes are most vulnerable. However, this process is only partially understood because of the experimental inaccessibility of early-stage embryos. Here, we introduce a methodologic advance, probing single cells for various DNA-methylation errors at multiple loci, to reveal failed maintenance of epigenetic mark results in chimeric mice, which display unpredictable phenotypes leading to developmental arrest. Yet we show that mouse pronuclear transfer can be used to ameliorate such reprogramming defects. This study not only details the epigenetic reprogramming dynamics in early mammalian embryos but also suggests diagnostic and potential future therapeutic applications.


Subject(s)
Blastocyst/metabolism , Cellular Reprogramming/genetics , Chimerism , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Animals , Gene Deletion , Genetic Loci , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Proteins/genetics , Repressor Proteins/genetics , Single-Cell Analysis , Tripartite Motif-Containing Protein 28
12.
Development ; 140(18): 3819-25, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23924633

ABSTRACT

Mobilization of endogenous retrotransposons can destabilize the genome, an imminent danger during epigenetic reprogramming of cells in the germline. The P-element-induced wimpy testis (PIWI)-interacting RNA (piRNA) pathway is known to silence retrotransposons in the mouse testes. Several piRNA pathway components localize to the unique, germline structure known as the nuage. In this study, we surveyed mouse ovaries and found, for the first time, transient appearance of nuage-like structures in oocytes of primordial follicles. Mouse vasa homolog (MVH), Piwi-like 2 (PIWIL2/MILI) and tudor domain-containing 9 (TDRD9) are present in these structures, whereas aggregates of germ cell protein with ankyrin repeats, sterile alpha motif and leucine zipper (GASZ) localize separately in the cytoplasm. Retrotransposons are silenced in primordial ovarian follicles, and de-repressed upon reduction of piRNA expression in Mvh, Mili or Gasz mutants. However, these null-mutant females, unlike their male counterparts, are fertile, uncoupling retrotransposon activation from sterility.


Subject(s)
Cellular Structures/metabolism , Gene Silencing , Ovarian Follicle/metabolism , Retroelements/genetics , Animals , Cellular Structures/ultrastructure , Female , Gene Expression Regulation , Germ Cells/metabolism , Infertility, Female/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Oogenesis , Ovarian Follicle/ultrastructure , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism
13.
Genes Dev ; 27(13): 1441-6, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23824537

ABSTRACT

Cellular localization of the Yes-associated protein (YAP) is dependent on large tumor suppressor (LATS) kinase activity and initiates lineage specification in the preimplantation embryo. We temporally reduced LATS activity to disrupt this early event, allowing its reactivation at later stages. This interference resulted in an irreversible lineage misspecification and aberrant polarization of the inner cell mass (ICM). Complementation experiments revealed that neither epiblast nor primitive endoderm can be established from these ICMs. We therefore conclude that precisely timed YAP localization in early morulae is essential to prevent trophectoderm marker expression in, and lineage specification of, the ICM.


Subject(s)
Blastocyst Inner Cell Mass/cytology , Blastocyst/cytology , Cell Differentiation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Lineage , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genetic Complementation Test , Hippo Signaling Pathway , Mice , Phosphoproteins/metabolism , Signal Transduction , Time Factors , YAP-Signaling Proteins
14.
PLoS One ; 8(7): e69764, 2013.
Article in English | MEDLINE | ID: mdl-23894536

ABSTRACT

Mammalian oocytes are arrested at prophase I of meiosis, and resume meiosis prior to ovulation. Coordination of meiotic arrest and resumption is partly dependent on the post-transcriptional regulation of maternal transcripts. Here, we report that, SPINDLIN1 (SPIN1), a maternal protein containing Tudor-like domains, interacts with a known mRNA-binding protein SERBP1, and is involved in regulating maternal transcripts to control meiotic resumption. Mouse oocytes deficient for Spin1 undergo normal folliculogenesis, but are defective in resuming meiosis. SPIN1, via its Tudor-like domain, forms a ribonucleoprotein complex with SERBP1, and regulating mRNA stability and/or translation. The mRNA for the cAMP-degrading enzyme, PDE3A, is reduced in Spin1 mutant oocytes, possibly contributing to meiotic arrest. Our study demonstrates that Spin1 regulates maternal transcripts post-transcriptionally and is involved in meiotic resumption.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Meiosis , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Oocytes/cytology , Phosphoproteins/chemistry , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Cell Cycle Proteins/genetics , Female , Gene Expression Regulation , Hyaluronic Acid/metabolism , Male , Mice , Microtubule-Associated Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mothers , Mutagenesis, Site-Directed , Mutation , Myogenic Regulatory Factors/metabolism , Oocytes/metabolism , Ovarian Follicle/growth & development , Phosphoproteins/genetics , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
Genes Dev ; 27(12): 1378-90, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23788624

ABSTRACT

Although it is known that OCT4-NANOG are required for maintenance of pluripotent cells in vitro, the upstream signals that regulate this circuit during early development in vivo have not been identified. Here we demonstrate, for the first time, signal transducers and activators of transcription 3 (STAT3)-dependent regulation of the OCT4-NANOG circuitry necessary to maintain the pluripotent inner cell mass (ICM), the source of in vitro-derived embryonic stem cells (ESCs). We show that STAT3 is highly expressed in mouse oocytes and becomes phosphorylated and translocates to the nucleus in the four-cell and later stage embryos. Using leukemia inhibitory factor (Lif)-null embryos, we found that STAT3 phosphorylation is dependent on LIF in four-cell stage embryos. In blastocysts, interleukin 6 (IL-6) acts in an autocrine fashion to ensure STAT3 phosphorylation, mediated by janus kinase 1 (JAK1), a LIF- and IL-6-dependent kinase. Using genetically engineered mouse strains to eliminate Stat3 in oocytes and embryos, we firmly establish that STAT3 is essential for maintenance of ICM lineages but not for ICM and trophectoderm formation. Indeed, STAT3 directly binds to the Oct4 and Nanog distal enhancers, modulating their expression to maintain pluripotency of mouse embryonic and induced pluripotent stem cells. These results provide a novel genetic model of cell fate determination operating through STAT3 in the preimplantation embryo and pluripotent stem cells in vivo.


Subject(s)
Blastocyst Inner Cell Mass , Cell Lineage , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins , Octamer Transcription Factor-3 , STAT3 Transcription Factor , Animals , Blastocyst Inner Cell Mass/cytology , Blastocyst Inner Cell Mass/metabolism , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phosphorylation , Pluripotent Stem Cells/physiology , Protein Binding , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
16.
Development ; 140(4): 853-60, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23318639

ABSTRACT

Cellular differentiation during embryogenesis involves complex gene regulation to enable the activation and repression of genes. Here, we show that mesodermal competence is inhibited in Xenopus embryos depleted of histones H3 and H3.3, which fail to respond to Nodal/Activin signaling and exhibit concomitant loss of mesodermal gene expression. We find that transcriptional activation in gastrula embryos does not correlate with promoter deposition of H3.3. Instead, gastrulation defects in H3.3/H3-deficient embryos are partially rescued with concurrent depletion of the linker histone H1A. In addition, we show that linker histone H1-induced premature loss of mesodermal competence in animal cap explants can be abrogated with the overexpression of nucleosomal H3.3/H3. Our findings establish a chromatin-mediated regulatory mechanism in which a threshold level of H3 is required to prevent H1-induced gene repression, and thus facilitate mesodermal differentiation in response to inductive signaling.


Subject(s)
Cell Differentiation/physiology , Chromatin/metabolism , Gene Expression Regulation, Developmental/physiology , Histones/metabolism , Mesoderm/physiology , Signal Transduction/physiology , Xenopus/embryology , Animals , Blotting, Western , Chromatin Immunoprecipitation , DNA Primers/genetics , Gastrula/embryology , Gastrula/metabolism , Gene Expression Regulation, Developmental/genetics , Morpholinos/genetics , Nodal Protein/metabolism , Polymerase Chain Reaction , Xenopus/metabolism
17.
Development ; 139(20): 3722-31, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22991438

ABSTRACT

The inside-outside model has been invoked to explain cell-fate specification of the pre-implantation mammalian embryo. Here, we investigate whether cell-cell interaction can influence the fate specification of embryonic blastomeres by sequentially separating the blastomeres in two-cell stage mouse embryos and continuing separation after each cell division throughout pre-implantation development. This procedure eliminates information provided by cell-cell interaction and cell positioning. Gene expression profiles, polarity protein localization and functional tests of these separated blastomeres reveal that cell interactions, through cell position, influence the fate of the blastomere. Blastomeres, in the absence of cell contact and inner-outer positional information, have a unique pattern of gene expression that is characteristic of neither inner cell mass nor trophectoderm, but overall they have a tendency towards a 'trophectoderm-like' gene expression pattern and preferentially contribute to the trophectoderm lineage.


Subject(s)
Blastomeres/cytology , Blastomeres/physiology , Cell Communication , Embryo, Mammalian/cytology , Embryonic Development , Animals , Cell Differentiation , Cell Lineage , Embryo, Mammalian/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Pregnancy
18.
Cytoskeleton (Hoboken) ; 69(12): 1040-6, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22887777

ABSTRACT

Positioning of the cleavage plane is regulated to ensure proper animal development. Most animal cells rely on the astral microtubules to position the mitotic spindle, which in turn specifies the cleavage plane. The mouse zygote lacks discernible astral microtubules but still divides symmetrically. Here, we demonstrate a cloud-like accumulation of F-actin surrounds the spindle in zygotes and when this actin network is disassembled, the spindle assumes an off-center position, and the resulting zygote divides asymmetrically into two unequal size blastomeres. Interestingly, when the spindle is micromanipulated to the subcortical region, the zygote without the actin network is unable to reposition the spindle and cleavage plane at the cell center. This study reveals that an actin network maintains the central spindle position in anastral mitosis, and ensures the first embryonic mitosis is symmetrical. © 2012 Wiley Periodicals, Inc.


Subject(s)
Actins/physiology , Cell Division/physiology , Zygote/physiology , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/physiology , Actins/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Pregnancy , Spindle Apparatus/metabolism , Spindle Apparatus/physiology , Zygote/cytology , Zygote/metabolism
19.
Science ; 335(6075): 1499-502, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22442485

ABSTRACT

Phenotypic variability in genetic disease is usually attributed to genetic background variation or environmental influence. Here, we show that deletion of a single gene, Trim28 (Kap1 or Tif1ß), from the maternal germ line alone, on an otherwise identical genetic background, results in severe phenotypic and epigenetic variability that leads to embryonic lethality. We identify early and minute epigenetic variations in blastomeres of the preimplantation embryo of these animals, suggesting that the embryonic lethality may result from the misregulation of genomic imprinting in mice lacking maternal Trim28. Our results reveal the long-range effects of a maternal gene deletion on epigenetic memory and illustrate the delicate equilibrium of maternal and zygotic factors during nuclear reprogramming.


Subject(s)
Embryo Loss , Embryo, Mammalian/physiology , Epigenesis, Genetic , Genomic Imprinting , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Oocytes/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Animals , Blastocyst/physiology , DNA Methylation , Down-Regulation , Embryonic Development , Female , Gene Expression Regulation, Developmental , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Phenotype , RNA, Long Noncoding , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Tripartite Motif-Containing Protein 28
20.
Methods Enzymol ; 477: 481-510, 2010.
Article in English | MEDLINE | ID: mdl-20699156

ABSTRACT

During its growth phase, a mouse oocyte accumulates RNA that is the sole template for new protein synthesis in the transcriptionally silent interval between growth completion and transcriptional activation of the embryonic genome. Over this transcriptionally silent interval, almost half the quantity of RNA accumulated in the full-grown oocyte is degraded, while stable messages undergo major transcript-specific polyadenylation fluctuations associated with timely translation of new proteins. These processes, in the background of substantial RNA degradation, create unique pitfalls for transcriptome analysis. Three particular challenges are discussed herein. (1) Systematic errors of relative quantification occur if standard approaches are used, wherein samples are normalized to a constant quantity of RNA, or when computational analyses are normalized to an apparent "constant" endogenous to the sample. We show that use of a fixed quantity of exogenous RNA per oocyte or embryo alleviates this problem. (2) Comparison of large-scale expression analyses from widely disparate platforms highlights how the differing protocols produce correspondingly different lists of genes with significant changes in transcript abundance. Only with careful attention to the differences among experiments can such discrepancies be understood. (3) The complete assessment of changes in expression requires correspondingly comprehensive assessment of the role of isoform-specific changes.


Subject(s)
Blastocyst/metabolism , Gene Expression Profiling/methods , Oocytes/metabolism , Animals , Female , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...