Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Science ; 357(6352): 707-713, 2017 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-28663440

RESUMO

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.


Assuntos
Anormalidades Cardiovasculares/genética , Proteínas de Transporte/genética , Hormônios Placentários/genética , Placentação/genética , Pré-Eclâmpsia/genética , Animais , Apelina/genética , Apelina/metabolismo , Peso ao Nascer , Proteínas de Transporte/administração & dosagem , Proteínas de Transporte/metabolismo , Proteínas de Transporte/farmacologia , Feminino , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/genética , Hormônios Peptídicos , Placenta/irrigação sanguínea , Placenta/metabolismo , Gravidez , Proteinúria , Transdução de Sinais
2.
Stem Cell Reports ; 9(1): 1-4, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28700896

RESUMO

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.


Assuntos
Análise Citogenética/métodos , Epigênese Genética , Variação Genética , Células-Tronco Pluripotentes/metabolismo , Medicina Regenerativa , Humanos , Maine , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/transplante , Medicina Regenerativa/métodos , Medição de Risco
3.
Int J Dev Biol ; 60(10-11-12): 327-336, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28000904

RESUMO

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.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes , Medicina Regenerativa , História do Século XX , Humanos , Estados Unidos
4.
Development ; 143(11): 1993-9, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27246714

RESUMO

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.


Assuntos
Desenvolvimento Embrionário , beta Catenina/metabolismo , Alelos , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Adesão Celular , Deleção de Genes , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mutantes/metabolismo , Zigoto/metabolismo , beta Catenina/deficiência
5.
J Infect Dis ; 212 Suppl 1: S47-51, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-26116732

RESUMO

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.


Assuntos
Reprogramação Celular/genética , Retrovirus Endógenos/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Oócitos/virologia , RNA Interferente Pequeno/genética , Animais , Retrovirus Endógenos/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oogênese/genética , RNA Interferente Pequeno/metabolismo
6.
Stem Cells Dev ; 24(13): 1515-20, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25897827

RESUMO

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.


Assuntos
Hospedeiro Imunocomprometido , Teratocarcinoma/patologia , Teratoma/patologia , Animais , Proteína Quinase Ativada por DNA/genética , Proteínas de Ligação a DNA/genética , Subunidade gama Comum de Receptores de Interleucina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos NOD , Proteínas Nucleares/genética , Teratocarcinoma/imunologia , Teratoma/imunologia
7.
Artigo em Inglês | MEDLINE | ID: mdl-26763985

RESUMO

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.


Assuntos
Reprogramação Celular/genética , Epigênese Genética/genética , Proteína 28 com Motivo Tripartido/genética , Animais , Metilação de DNA/genética , Desenvolvimento Embrionário/genética , Feminino , Genoma , Impressão Genômica/genética , Células Germinativas , Humanos , Masculino , Mamíferos , Retroelementos/genética
9.
Genes Dev ; 28(8): 812-28, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24736841

RESUMO

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.


Assuntos
Blastocisto/metabolismo , Reprogramação Celular/genética , Metilação de DNA , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , 5-Metilcitosina/metabolismo , Animais , Embrião de Mamíferos , Células Germinativas/metabolismo , Humanos
10.
Stem Cell Reports ; 1(5): 379-86, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24286026

RESUMO

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.


Assuntos
Cromossomos Humanos Par 20/genética , Variações do Número de Cópias de DNA , Células-Tronco Embrionárias/metabolismo , Seleção Genética , Proteína bcl-X/metabolismo , Linhagem Celular , Amplificação de Genes , Loci Gênicos , Humanos , Mutação , Proteína bcl-X/genética
11.
Science ; 341(6150): 1110-2, 2013 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-24009393

RESUMO

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.


Assuntos
Blastocisto/metabolismo , Reprogramação Celular/genética , Quimerismo , Metilação de DNA , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Animais , Deleção de Genes , Loci Gênicos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Nucleares/genética , Proteínas Repressoras/genética , Análise de Célula Única , Proteína 28 com Motivo Tripartido
12.
Development ; 140(18): 3819-25, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23924633

RESUMO

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.


Assuntos
Estruturas Celulares/metabolismo , Inativação Gênica , Folículo Ovariano/metabolismo , Retroelementos/genética , Animais , Estruturas Celulares/ultraestrutura , Feminino , Regulação da Expressão Gênica , Células Germinativas/metabolismo , Infertilidade Feminina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Oogênese , Folículo Ovariano/ultraestrutura , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo
13.
Genes Dev ; 27(13): 1441-6, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23824537

RESUMO

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.


Assuntos
Massa Celular Interna do Blastocisto/citologia , Blastocisto/citologia , Diferenciação Celular , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular , Linhagem da Célula , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Teste de Complementação Genética , Via de Sinalização Hippo , Camundongos , Fosfoproteínas/metabolismo , Transdução de Sinais , Fatores de Tempo , Proteínas de Sinalização YAP
14.
PLoS One ; 8(7): e69764, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23894536

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Meiose , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/metabolismo , Oócitos/citologia , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Ciclo Celular/genética , Feminino , Regulação da Expressão Gênica , Ácido Hialurônico/metabolismo , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Modelos Moleculares , Dados de Sequência Molecular , Mães , Mutagênese Sítio-Dirigida , Mutação , Fatores de Regulação Miogênica/metabolismo , Oócitos/metabolismo , Folículo Ovariano/crescimento & desenvolvimento , Fosfoproteínas/genética , Ligação Proteica , Estrutura Terciária de Proteína , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
15.
Genes Dev ; 27(12): 1378-90, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23788624

RESUMO

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.


Assuntos
Massa Celular Interna do Blastocisto , Linhagem da Célula , Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio , Fator 3 de Transcrição de Octâmero , Fator de Transcrição STAT3 , Animais , Massa Celular Interna do Blastocisto/citologia , Massa Celular Interna do Blastocisto/metabolismo , Células Cultivadas , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Janus Quinase 1/genética , Janus Quinase 1/metabolismo , Fator Inibidor de Leucemia/genética , Fator Inibidor de Leucemia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Fosforilação , Células-Tronco Pluripotentes/fisiologia , Ligação Proteica , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
16.
Development ; 140(4): 853-60, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23318639

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Cromatina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Histonas/metabolismo , Mesoderma/fisiologia , Transdução de Sinais/fisiologia , Xenopus/embriologia , Animais , Western Blotting , Imunoprecipitação da Cromatina , Primers do DNA/genética , Gástrula/embriologia , Gástrula/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Morfolinos/genética , Proteína Nodal/metabolismo , Reação em Cadeia da Polimerase , Xenopus/metabolismo
17.
Development ; 139(20): 3722-31, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22991438

RESUMO

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.


Assuntos
Blastômeros/citologia , Blastômeros/fisiologia , Comunicação Celular , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Animais , Diferenciação Celular , Linhagem da Célula , Embrião de Mamíferos/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Gravidez
18.
Cytoskeleton (Hoboken) ; 69(12): 1040-6, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22887777

RESUMO

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.


Assuntos
Actinas/fisiologia , Divisão Celular/fisiologia , Zigoto/fisiologia , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/fisiologia , Actinas/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Gravidez , Fuso Acromático/metabolismo , Fuso Acromático/fisiologia , Zigoto/citologia , Zigoto/metabolismo
19.
Science ; 335(6075): 1499-502, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22442485

RESUMO

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.


Assuntos
Perda do Embrião , Embrião de Mamíferos/fisiologia , Epigênese Genética , Impressão Genômica , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Oócitos/fisiologia , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia , Animais , Blastocisto/fisiologia , Metilação de DNA , Regulação para Baixo , Desenvolvimento Embrionário , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , RNA Longo não Codificante , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Proteína 28 com Motivo Tripartido
20.
Methods Enzymol ; 477: 481-510, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20699156

RESUMO

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.


Assuntos
Blastocisto/metabolismo , Perfilação da Expressão Gênica/métodos , Oócitos/metabolismo , Animais , Feminino , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...