Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nature ; 589(7840): 110-115, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33239785

RESUMO

In mammals, telomere protection is mediated by the essential protein TRF2, which binds chromosome ends and ensures genome integrity1,2. TRF2 depletion results in end-to-end chromosome fusions in all cell types that have been tested so far. Here we find that TRF2 is dispensable for the proliferation and survival of mouse embryonic stem (ES) cells. Trf2-/- (also known as Terf2) ES cells do not exhibit telomere fusions and can be expanded indefinitely. In response to the deletion of TRF2, ES cells exhibit a muted DNA damage response that is characterized by the recruitment of γH2AX-but not 53BP1-to telomeres. To define the mechanisms that control this unique DNA damage response in ES cells, we performed a CRISPR-Cas9-knockout screen. We found a strong dependency of TRF2-null ES cells on the telomere-associated protein POT1B and on the chromatin remodelling factor BRD2. Co-depletion of POT1B or BRD2 with TRF2 restores a canonical DNA damage response at telomeres, resulting in frequent telomere fusions. We found that TRF2 depletion in ES cells activates a totipotent-like two-cell-stage transcriptional program that includes high levels of ZSCAN4. We show that the upregulation of ZSCAN4 contributes to telomere protection in the absence of TRF2. Together, our results uncover a unique response to telomere deprotection during early development.


Assuntos
Células-Tronco Pluripotentes/metabolismo , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/deficiência , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Pluripotentes/citologia , Proteína 2 de Ligação a Repetições Teloméricas/genética , Células-Tronco Totipotentes/citologia , Células-Tronco Totipotentes/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo
2.
Dev Cell ; 29(3): 321-9, 2014 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-24768164

RESUMO

The mechanistic target of rapamycin complex 1 (mTORC1) integrates cues from growth factors and nutrients to control metabolism. In contrast to the growth factor input, genetic disruption of nutrient-dependent activation of mTORC1 in mammals remains unexplored. We engineered mice lacking RagA and RagB genes, which encode the GTPases responsible for mTORC1 activation by nutrients. RagB has limited expression, and its loss shows no effects on mammalian physiology. RagA deficiency leads to E10.5 embryonic death, loss of mTORC1 activity, and severe growth defects. Primary cells derived from these mice exhibit no regulation of mTORC1 by nutrients and maintain high sensitivity to growth factors. Deletion of RagA in adult mice is lethal. Upon RagA loss, a myeloid population expands in peripheral tissues. RagA-specific deletion in liver increases cellular responses to growth factors. These results show the essentiality of nutrient sensing for mTORC1 activity in mice and its suppression of PI3K/Akt signaling.


Assuntos
Embrião de Mamíferos/embriologia , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Linhagem Celular , Hepatócitos/metabolismo , Fígado/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Proteínas Monoméricas de Ligação ao GTP/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética
3.
J Immunol ; 190(2): 695-702, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23241879

RESUMO

Recognition of nucleic acids by TLR9 requires its trafficking from the endoplasmic reticulum to endolysosomal compartments and its subsequent proteolytic processing. Both processes depend on interactions of TLR9 with the polytopic endoplasmic reticulum-resident protein UNC93B1. To examine the intracellular behavior of TLR9 in primary APCs, we generated transgenic mice expressing a TLR9-GFP fusion. The TLR9-GFP transgene is functional and is proteolytically processed in resting bone marrow-derived macrophages (BMDMs), dendritic cells, and B cells. Inhibition of cleavage impairs TLR9-dependent responses in all primary APCs analyzed. The kinetics of TLR9-GFP processing in BMDMs and B cells differs: in B cells, proteolysis occurs at a faster rate, consistent with an almost exclusive localization to endolysosomes at the resting state. In contrast to the joint requirement for cathepsins L and S for TLR9 cleavage in macrophages, TLR9-GFP cleavage depends on cathepsin L activity in B cells. As expected, in BMDMs and B cells from UNC93B1 (3d) mutant mice, cleavage of TLR9-GFP is essentially blocked, and the expression level of UNC93B1 appears tightly correlated with TLR9-GFP cleavage. We conclude that proteolysis is a universal requirement for TLR9 activation in the primary cell types tested, however the cathepsin requirement, rate of cleavage, and intracellular behavior of TLR9 varies. The observed differences in trafficking indicate the possibility of distinct modes of endosomal content sampling to facilitate initiation of TLR-driven responses in APCs.


Assuntos
Células Apresentadoras de Antígenos/metabolismo , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Animais , Linfócitos B/metabolismo , Células da Medula Óssea/metabolismo , Linhagem Celular , Retículo Endoplasmático/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lisossomos/metabolismo , Macrófagos/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Transgênicos , Estabilidade Proteica , Transporte Proteico , Proteólise , Transdução de Sinais , Transgenes
4.
Cell Rep ; 1(5): 461-71, 2012 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-22832272

RESUMO

To study the CD8(+) T cell response against a mouse γ-herpes virus, we generated K(b)-MHV-68-ORF8(604-612)RAG(-/-) CD8(+) T cell receptor transnuclear (TN) mice as a source of virus-specific CD8(+) T cells. K(b)-ORF8-Tet(+) CD8(+) T cells, expanded in the course of a resolving MHV-68 infection, served as a source of nucleus donors. Various in vivo and ex vivo assay criteria demonstrated the fine specificity and functionality of TN cells. TN cells proliferated extensively in response to viral infection, helped control viral burden, and exhibited a phenotype similar to that of endogenous K(b)-ORF8-Tet(+) cells. When compared to OT-1 cells, TN cells displayed distinct properties in response to lymphopenia and cognate antigen stimulation, which may be attributable to the affinity of the TCR expressed by the TN cells. The availability of MHV-68-specific CD8(+) TCR TN mice provides a new tool for investigating aspects of host-pathogen interactions unique to γ-herpes viruses.


Assuntos
Linfócitos T CD8-Positivos/patologia , Epitopos/metabolismo , Glicoproteínas/metabolismo , Antígenos H-2/metabolismo , Infecções por Herpesviridae/fisiopatologia , Receptores de Antígenos de Linfócitos T/metabolismo , Rhadinovirus/metabolismo , Proteínas Virais/metabolismo , Animais , Linfócitos T CD8-Positivos/metabolismo , Núcleo Celular/metabolismo , Proliferação de Células , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/prevenção & controle , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Fenótipo , Infecções Tumorais por Vírus/metabolismo , Infecções Tumorais por Vírus/fisiopatologia , Infecções Tumorais por Vírus/prevenção & controle , Carga Viral/fisiologia
5.
PLoS Genet ; 7(5): e1002054, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21573140

RESUMO

MicroRNAs (miRNAs) post-transcriptionally regulate the expression of thousands of distinct mRNAs. While some regulatory interactions help to maintain basal cellular functions, others are likely relevant in more specific settings, such as response to stress. Here we describe such a role for the mir-290-295 cluster, the dominant miRNA cluster in mouse embryonic stem cells (mESCs). Examination of a target list generated from bioinformatic prediction, as well as expression data following miRNA loss, revealed strong enrichment for apoptotic regulators, two of which we validated directly: Caspase 2, the most highly conserved mammalian caspase, and Ei24, a p53 transcriptional target. Consistent with these predictions, mESCs lacking miRNAs were more likely to initiate apoptosis following genotoxic exposure to gamma irradiation or doxorubicin. Knockdown of either candidate partially rescued this pro-apoptotic phenotype, as did transfection of members of the mir-290-295 cluster. These findings were recapitulated in a specific mir-290-295 deletion line, confirming that they reflect miRNA functions at physiological levels. In contrast to the basal regulatory roles previously identified, the pro-survival phenotype shown here may be most relevant to stressful gestations, where pro-oxidant metabolic states induce DNA damage. Similarly, this cluster may mediate chemotherapeutic resistance in a neoplastic context, making it a useful clinical target.


Assuntos
Células-Tronco Embrionárias/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Apoptose/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Sequência de Bases , Caspase 2/genética , Caspase 2/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Doxorrubicina/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos da radiação , Raios gama , Deleção de Genes , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transdução de Sinais
6.
Stem Cells ; 29(6): 992-1000, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21563275

RESUMO

Pluripotent cells can be derived from different types of somatic cells by nuclear reprogramming through the ectopic expression of four transcription factors, Oct3/4, Sox2, Klf4, and c-Myc. However, it is unclear whether postmitotic neurons are susceptible to direct reprogramming. Here, we show that postnatal cortical neurons, the vast majority of which are postmitotic, are amenable to epigenetic reprogramming. However, ectopic expression of the four canonical reprogramming factors is not sufficient to reprogram postnatal neurons. Efficient reprogramming was only achieved after forced cell proliferation by p53 suppression. Additionally, overexpression of repressor element-1 silencing transcription, a suppressor of neuronal gene activity, increased reprogramming efficiencies in combination with the reprogramming factors. Our findings indicate that terminally differentiated postnatal neurons are able to acquire the pluripotent state by direct epigenetic reprogramming, and this process is made more efficient through the suppression of lineage specific gene expression.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Neurônios/citologia , Proteínas Repressoras/metabolismo , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Blastocisto/citologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Células Cultivadas , Técnicas de Cocultura , Transferência Embrionária , Fibroblastos/citologia , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Fator 4 Semelhante a Kruppel , Antígenos CD15/metabolismo , Camundongos , Proteína Homeobox Nanog , Neurônios/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Regiões Promotoras Genéticas , Teratoma/patologia , Quimeras de Transplante
7.
PLoS Biol ; 8(3): e1000605, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21468303

RESUMO

Ubiquitin-dependent processes control much of cellular physiology. We show that expression of a highly active, Epstein-Barr virus-derived deubiquitylating enzyme (EBV-DUB) blocks proteasomal degradation of cytosolic and ER-derived proteins by preemptive removal of ubiquitin from proteasome substrates, a treatment less toxic than the use of proteasome inhibitors. Recognition of misfolded proteins in the ER lumen, their dislocation to the cytosol, and degradation are usually tightly coupled but can be uncoupled by the EBV-DUB: a misfolded glycoprotein that originates in the ER accumulates in association with cytosolic chaperones as a deglycosylated intermediate. Our data underscore the necessity of a DUB activity for completion of the dislocation reaction and provide a new means of inhibition of proteasomal proteolysis with reduced cytotoxicity.


Assuntos
Herpesvirus Humano 4/enzimologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais , Ubiquitina/metabolismo , Proteínas Virais/metabolismo , Biocatálise , Linhagem Celular , Retículo Endoplasmático/metabolismo , Glicoproteínas/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Dobramento de Proteína , Processamento de Proteína Pós-Traducional , Transporte Proteico , Especificidade por Substrato
8.
Cell ; 144(5): 782-95, 2011 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-21376238

RESUMO

During development and regeneration, proliferation of tissue-specific stem cells is tightly controlled to produce organs of a predetermined size. The molecular determinants of this process remain poorly understood. Here, we investigate the function of Yap1, the transcriptional effector of the Hippo signaling pathway, in skin biology. Using gain- and loss-of-function studies, we show that Yap1 is a critical modulator of epidermal stem cell proliferation and tissue expansion. Yap1 mediates this effect through interaction with TEAD transcription factors. Additionally, our studies reveal that α-catenin, a molecule previously implicated in tumor suppression and cell density sensing in the skin, is an upstream negative regulator of Yap1. α-catenin controls Yap1 activity and phosphorylation by modulating its interaction with 14-3-3 and the PP2A phosphatase. Together, these data identify Yap1 as a determinant of the proliferative capacity of epidermal stem cells and as an important effector of a "crowd control" molecular circuitry in mammalian skin.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proliferação de Células , Células Epidérmicas , Fosfoproteínas/metabolismo , alfa Catenina/metabolismo , Proteínas 14-3-3/metabolismo , Animais , Proteínas de Ciclo Celular , Linhagem Celular , Epiderme/metabolismo , Camundongos , Proteínas de Sinalização YAP
9.
J Vis Exp ; (43)2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20972401

RESUMO

Lymphocytes, such as T cells, undergo genetic V(D)J recombination, to generate a receptor with a certain specificity. Mice transgenic for a rearranged antigen-specific T cell receptor (TCR) have been an indispensable tool to study T cell development and function. However, such TCRs are usually isolated from the relevant T cells after long-term culture often following repeated antigen stimulation, which unavoidably selects for T cells with high affinity. Random genomic integration of the TCR α- and ß-chain and expression from non-endogenous promoters can lead to variations in expression level and kinetics. Epigenetic reprogramming via somatic cell nuclear transfer provides a tool to generate embryonic stem cells and mice from any cell of interest. Consequently, when SCNT is applied to T cells of known specificity, these genetic V(D)J rearrangements are transferred to the SCNT-embryonic stem cells (ESCs) and the mice derived from them, while epigenetic marks are reset. We have demonstrated that T cells with pre-defined specificities against Toxoplasma gondii can be used to generate mouse models that express the specific TCR from their endogenous loci, without experimentally introduced genetic modification. The relative ease and speed with which such transnuclear models can be obtained holds promise for the construction of other disease models.


Assuntos
Células-Tronco Embrionárias/imunologia , Técnicas de Transferência Nuclear , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Toxoplasma/imunologia , Animais , Epitopos de Linfócito T/imunologia , Feminino , Camundongos
10.
Science ; 328(5975): 243-8, 2010 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-20378817

RESUMO

Mice that are transgenic for rearranged antigen-specific T cell receptors (TCRs) are essential tools to study T cell development and function. Such TCRs are usually isolated from the relevant T cells after long-term culture, often after repeated antigen stimulation, which unavoidably skews the T cell population used. Random genomic integration of the TCR alpha and beta chain and expression from nonendogenous promoters represent additional drawbacks of transgenics. Using epigenetic reprogramming via somatic cell nuclear transfer, we demonstrated that T cells with predefined specificities against Toxoplasma gondii can be used to generate mouse models that express the TCR from their endogenous loci, without experimentally introduced genetic modification. The relative ease and speed with which such transnuclear models can be obtained holds promise for the construction of other disease models.


Assuntos
Antígenos de Protozoários/imunologia , Camundongos Transgênicos , Técnicas de Transferência Nuclear , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Toxoplasma/imunologia , Toxoplasmose Animal/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Células-Tronco Embrionárias , Epitopos de Linfócito T , Feminino , Genes Codificadores da Cadeia alfa de Receptores de Linfócitos T , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/genética
11.
Nat Methods ; 6(2): 147-52, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19122668

RESUMO

Cell fusion has been used for many different purposes, including generation of hybridomas and reprogramming of somatic cells. The fusion step is the key event in initiation of these procedures. Standard fusion techniques, however, provide poor and random cell contact, leading to low yields. We present here a microfluidic device to trap and properly pair thousands of cells. Using this device, we paired different cell types, including fibroblasts, mouse embryonic stem cells and myeloma cells, achieving pairing efficiencies up to 70%. The device is compatible with both chemical and electrical fusion protocols. We observed that electrical fusion was more efficient than chemical fusion, with membrane reorganization efficiencies of up to 89%. We achieved greater than 50% properly paired and fused cells over the entire device, fivefold greater than with a commercial electrofusion chamber and observed reprogramming in hybrids between mouse embryonic stem cells and mouse embryonic fibroblasts.


Assuntos
Fusão Celular/métodos , Técnicas Analíticas Microfluídicas/instrumentação , Animais , Linhagem Celular Tumoral , Eletroporação/métodos , Células-Tronco Embrionárias , Processamento de Imagem Assistida por Computador/métodos , Camundongos , Técnicas Analíticas Microfluídicas/métodos , Células NIH 3T3 , Polietilenoglicóis/farmacologia
12.
Stem Cells ; 26(6): 1628-35, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18369099

RESUMO

Successful hematopoietic stem cell (HSC) transplantation is often limited by the numbers of HSCs, and robust methods to expand HSCs ex vivo are needed. We previously showed that angiopoietin-like proteins (Angptls), a group of growth factors isolated from a fetal liver HSC-supportive cell population, improved ex vivo expansion of HSCs. Here, we demonstrate that insulin-like growth factor-binding protein 2 (IGFBP2), secreted by a tumorigenic cell line, also enhanced ex vivo expansion of mouse HSCs. On the basis of these findings, we established a completely defined, serum-free culture system for mouse HSCs, containing SCF, thrombopoietin, fibroblast growth factor 1, Angptl3, and IGFBP2. As measured by competitive repopulation analyses, there was a 48-fold increase in numbers of long-term repopulating mouse HSCs after 21 days of culture. This is the first demonstration that IGFBP2 stimulates expansion or proliferation of murine stem cells. Our finding also suggests that certain cancer cells synthesize proteins that can stimulate HSC expansion. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Células-Tronco Hematopoéticas/citologia , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/farmacologia , Expansão de Tecido/métodos , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Meios de Cultivo Condicionados , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Rim , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
13.
Cell Stem Cell ; 1(3): 346-52, 2007 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-18371368

RESUMO

Parthenogenesis and somatic cell nuclear transfer (SCNT) are two methods for deriving embryonic stem (ES) cells that are genetically matched to the oocyte donor or somatic cell donor, respectively. Using genome-wide single nucleotide polymorphism (SNP) analysis, we demonstrate distinct signatures of genetic recombination that distinguish parthenogenetic ES cells from those generated by SCNT. We applied SNP analysis to the human ES cell line SCNT-hES-1, previously claimed to have been derived by SCNT, and present evidence that it represents a human parthenogenetic ES cell line. Genome-wide SNP analysis represents a means to validate the genetic provenance of an ES cell line.


Assuntos
Células-Tronco Embrionárias/metabolismo , Técnicas de Transferência Nuclear , Partenogênese , Recombinação Genética/genética , Animais , Linhagem Celular , Análise Citogenética , Metilação de DNA , Genoma Humano/genética , Heterozigoto , Homozigoto , Humanos , Camundongos , Polimorfismo de Nucleotídeo Único/genética , Análise de Sequência de DNA , Manejo de Espécimes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA