Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Sci ; 127(Pt 9): 2095-105, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24569880

RESUMO

Retinoic acid (RA) plays key roles in cell differentiation and growth arrest by activating nuclear RA receptors (RARs) (α, ß and γ), which are ligand-dependent transcription factors. RARs are also phosphorylated in response to RA. Here, we investigated the in vivo relevance of the phosphorylation of RARs during RA-induced neuronal differentiation of mouse embryonic stem cells (mESCs). Using ESCs where the genes encoding each RAR subtype had been inactivated, and stable rescue lines expressing RARs mutated in phospho-acceptor sites, we show that RA-induced neuronal differentiation involves RARγ2 and requires RARγ2 phosphorylation. By gene expression profiling, we found that the phosphorylated form of RARγ2 regulates a small subset of genes through binding an unusual RA response element consisting of two direct repeats with a seven-base-pair spacer. These new findings suggest an important role for RARγ phosphorylation during cell differentiation and pave the way for further investigations during embryonic development.


Assuntos
Receptores do Ácido Retinoico/metabolismo , Tretinoína/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Camundongos , Fosforilação , Receptor gama de Ácido Retinoico
2.
PLoS One ; 7(12): e53257, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23300903

RESUMO

It is currently admitted that Follicle-Stimulating Hormone (FSH) is physiologically involved in the development and function of fetal/neonatal Sertoli cells in the rat but not the mouse. However, FSH is produced by both species from late fetal life onwards. We thus reinvestigated the role of FSH in mouse testis development at day 0 (birth) 6, 8 and 10 post-partum (dpp) by using mice that lack functional FSH receptors (FSH-R(-/-)). At birth, the number and proliferative index of Sertoli cells were significantly lower in FSH-R(-/-) mice than in wild type neonates. Claudin 11 mRNA expression also was significantly reduced in FSH-R(-/-) testes at 0 and 8 dpp, whereas the mRNA levels of other Sertoli cell markers (Transferrin and Desert hedgehog) were comparable in FSH-R(-/-) and wild type testes. Conversely, AMH mRNA and protein levels were higher at birth, comparable at 6 dpp and then significantly lower in FSH-R(-/-) testes at 8-10 dpp in FSH-R(-/-) mice than in controls. Although the plasma concentration of LH and the number of Leydig cells were similar in FSH-R(-/-) and control (wild type), testosterone concentration and P450c17 mRNA expression were significantly increased in FSH-R(-/-) testes at birth. Conversely, at 10 dpp when adult Leydig cells appear, expression of the steroidogenic genes P450scc, P450c17 and StAR was lower in FSH-R(-/-) testes than in controls. In conclusion, our results show that 1) like in the rat, signaling via FSH-R controls Sertoli cell development and function during late fetal life in the mouse as well; 2) paracrine factors produced by Sertoli cells are involved in the FSH-R-dependent regulation of the functions of fetal Leydig cells in late fetal life; and 3) the role of FSH-R signaling changes during the prepubertal period.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Receptores do FSH/metabolismo , Transdução de Sinais/fisiologia , Testículo/fisiologia , Animais , Células Intersticiais do Testículo/fisiologia , Masculino , Camundongos , Camundongos Knockout , Receptores do FSH/genética , Células de Sertoli/fisiologia , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Testosterona/sangue
3.
Genesis ; 50(6): 482-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22121025

RESUMO

To facilitate the use of the new mutant resource developed in the mouse, we have generated Cre and FlpO deleter mice on a pure inbred C57BL/6N background. The new transgenic constructs were designed to drive either the Cre or FlpO recombinase, fused to a specific fluorescent marker, respectively the eGFP or the eYFP, and were inserted by homologous recombination in the neutral Rosa26 locus. They allow a rapid, cost-effective, and efficient identification of the carrier individuals through the coexpression of the fluorescent marker. The recombination efficiency of the two deleter lines, Gt(ROSA)26S or < tm1(ACTB-cre,-EGFP)Ics> and Gt(ROSA) 26S or < tm2(CAG-flpo, EYFP)Ics>, was carefully evaluated using five loxP-flanked or four FRT-flanked alleles located at different positions in the mouse genome. For each tested locus, we observed a 100% excision rate. The transgenic mice are easily distinguishable from wild type animals by their bright fluorescence that remains easily detectable until 10 days after birth. In the adult, fluorescence can still be detected in the unpigmented paws. Furthermore, they both display accumulation of the specific recombinase during oogenesis. These fluorescent 'Cre- and Flp- deleter' transgenic lines are valuable tools for the scientific community by their high and stable recombination efficiency, the simplicity of genotype identification and the maintenance of a pure genetic background when used to remove specific selection cassette or to induce complete loss-of-function allele.


Assuntos
DNA Nucleotidiltransferases/genética , Integrases/genética , Camundongos Transgênicos , Proteínas/genética , Animais , Proteínas de Bactérias/genética , Efeito Fundador , Marcação de Genes , Engenharia Genética , Genótipo , Proteínas de Fluorescência Verde/genética , Recombinação Homóloga , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos C57BL , RNA não Traduzido , Deleção de Sequência
4.
J Immunol ; 183(4): 2303-11, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19620299

RESUMO

Helios is a member of the Ikaros family of zinc finger transcription factors. It is expressed mainly in T cells, where it associates with Ikaros-containing complexes and has been proposed to act as a rate-limiting factor for Ikaros function. Overexpression of wild-type or dominant-negative Helios isoforms profoundly alters alphabeta T cell differentiation and activation, and endogenous Helios is expressed at strikingly high levels in regulatory T cells. Helios has also been implicated as a tumor suppressor in human T cell acute lymphoblastic leukemias. These studies suggest a central role for Helios in T cell development and homeostasis, but whether this protein is physiologically required in T cells is unclear. We report herein that inactivation of the Helios gene by homologous recombination does not impair the differentiation and effector cell function of alphabeta and gammadelta T cells, NKT cells, and regulatory T cells. These results suggest that Helios is not essential for T cells, and that its function can be compensated for by other members of the Ikaros family.


Assuntos
Diferenciação Celular/genética , Diferenciação Celular/imunologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Proliferação de Células , Células Cultivadas , Células Clonais , Proteínas de Ligação a DNA/fisiologia , Inativação Gênica/imunologia , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Família Multigênica/imunologia , Receptores de Antígenos de Linfócitos T/fisiologia , Subpopulações de Linfócitos T/metabolismo , Fatores de Transcrição/fisiologia
5.
Arterioscler Thromb Vasc Biol ; 28(5): 841-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18421008

RESUMO

OBJECTIVE: Identification of novel factors that contribute to myocardial repair and collateral vessel growth hold promise for treatment of heart diseases. We have shown that transient prokineticin receptor-1 (PKR1) gene transfer protects the heart against myocardial infarction in a mouse model. Here, we investigated the role of excessive PKR1 signaling in heart. METHODS AND RESULTS: Transgenic mice overexpressing PKR1 in cardiomyocytes displayed no spontaneous abnormalities in cardiomyocytes but showed an increased number of epicardial-derived progenitor cells (EPDCs), capillary density, and coronary arterioles. Coculturing EPDCs with H9c2 cardiomyoblasts overexpressing PKR1 promotes EPDC differentiation into endothelial and smooth muscle cells, mimicking our transgenic model. Overexpressing PKR1 in H9c2 cardiomyoblasts or in transgenic hearts upregulated prokineticin-2 levels. Exogenous prokineticin-2 induces significant outgrowth from neonatal and adult epicardial explants, promoting EPDC differentiation. These prokineticin-2 effects were abolished in cardiac explants from mice with PKR1-null mutation. Reduced capillary density and prokineticin-2 levels in PKR1-null mutant hearts supports the hypothesis of an autocrine/paracrine loop between PKR1 and prokineticin-2. CONCLUSIONS: Cardiomyocyte-PKR1 signaling upregulates its own ligand prokineticin-2 that acts as a paracrine factor, triggering EPDCs proliferation/differentiation. This study provides a novel insight for possible therapeutic strategies aiming at restoring pluripotency of adult EPDCs to promote neovasculogenesis by induction of cardiomyocyte PKR1 signaling.


Assuntos
Diferenciação Celular/fisiologia , Neovascularização Fisiológica/fisiologia , Pericárdio/patologia , Proteína Quinase C/metabolismo , Células-Tronco/patologia , Animais , Movimento Celular/fisiologia , Proliferação de Células , Técnicas de Cocultura , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Hormônios Gastrointestinais/genética , Hormônios Gastrointestinais/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Pericárdio/metabolismo , Proteína Quinase C/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Regulação para Cima
6.
Genesis ; 45(1): 38-43, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17216605

RESUMO

Nonsense-mediated mRNA decay (NMD) is a process of mRNA surveillance that degrades transcripts harboring a premature termination codon (PTC). Mammalian NMD was mostly studied in cultured cells so far and there was no direct evidence yet that NMD could operate in the brain. We introduced, by homologous recombination in mouse, a PTC in the mu opioid receptor gene (mor). mor transcript was severely downregulated in the brain of these knock-in mice. A systemic cycloheximide treatment significantly increased the level of the mutant mRNA, suggesting NMD involvement. To further corroborate this hypothesis, we generated a second knock-in mouse line where the PTC was placed at 10 instead of 96 nucleotides from the downstream splice junction. As predicted by the "termination codon position rule" established in vitro, mor transcript brain expression was rescued to wild-type level. These knock-in mouse lines will be valuable models to better understand and manipulate NMD in vivo.


Assuntos
Encéfalo/metabolismo , Códon sem Sentido , Estabilidade de RNA , RNA Mensageiro/metabolismo , Receptores Opioides mu/metabolismo , Animais , Cicloeximida/farmacologia , Regulação para Baixo , Camundongos , Mutação , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/genética , Receptores Opioides mu/genética
7.
Differentiation ; 74(9-10): 661-8, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17177861

RESUMO

Retinoic acid (RA), the active vitamin A derivative, is an important developmental signaling molecule in vertebrates. In this study, we have assessed whether minimal numbers and/or specific distributions of RA-producing cells can support normal mouse embryonic development. Retinaldehyde dehydrogenase 2 (RALDH2) is the main RA-synthesizing enzyme acting during development. We have generated an embryonic stem (ES) cell line homozygous for an Raldh2 gene disruption, and have analyzed chimeric embryos with various contributions of wild-type cells. Whereas embryos almost completely derived from Raldh2(-/-) cells phenocopy the corresponding germline null mutants, the presence of even small numbers (<10%) of wild-type cells can rescue most of the morphogenetic defects, including embryonic turning and axial elongation, and left-right looping of the heart tube. No consistent bias in the distribution of wild-type cells was observed in the phenotypically rescued Raldh2(-/-) chimeras. Analysis of an RA-sensitive transgene indicates that RA can diffuse from wild-type cells and elicit a widespread transcriptional response in Raldh2-deficient cells. Our results show that few wild-type RA-producing cells, even when present in apparent random distributions, can support early morphogenesis of the mouse embryo. However, the Raldh2(-/-) chimeric fetuses display lung abnormalities, persistent truncus arteriosus, and abnormal myocardial differentiation, showing that subsequent RA-dependent events cannot be fully rescued by the mosaic presence of wild-type cells.


Assuntos
Aldeído Oxirredutases/genética , Desenvolvimento Embrionário , Células-Tronco Embrionárias/enzimologia , Transdução de Sinais , Tretinoína/metabolismo , Anormalidades Múltiplas , Aldeído Oxirredutases/metabolismo , Animais , Blastocisto/metabolismo , Diferenciação Celular , Quimera/anormalidades , Quimera/embriologia , Quimera/metabolismo , Quimerismo , Desenvolvimento Embrionário/genética , Camundongos , Camundongos Transgênicos , Mutação , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Transgenes
8.
Hum Mol Genet ; 15(24): 3544-58, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17101632

RESUMO

Mutations of the spastin gene (Sp) are responsible for the most frequent autosomal dominant form of spastic paraplegia, a disease characterized by the degeneration of corticospinal tracts. We show that a deletion in the mouse Sp gene, generating a premature stop codon, is responsible for progressive axonal degeneration, restricted to the central nervous system, leading to a late and mild motor defect. The degenerative process is characterized by focal axonal swellings, associated with abnormal accumulation of organelles and cytoskeletal components. In culture, mutant cortical neurons showed normal viability and neurite density. However, they develop neurite swellings associated with focal impairment of retrograde transport. These defects occur near the growth cone, in a region characterized by the transition between stable microtubules rich in detyrosinated alpha-tubulin and dynamic microtubules composed almost exclusively of tyrosinated alpha-tubulin. Here, we show that the Sp mutation has a major impact on neurite maintenance and transport both in vivo and in vitro. These results highlight the link between spastin and microtubule dynamics in axons, but not in other neuronal compartments. In addition, it is the first description of a human neurodegenerative disease which involves this specialized region of the axon.


Assuntos
Adenosina Trifosfatases/genética , Axônios/metabolismo , Microtúbulos/metabolismo , Mutação , Adenosina Trifosfatases/fisiologia , Animais , Axônios/patologia , Axônios/ultraestrutura , Sequência de Bases , Comportamento Animal , Transporte Biológico , Western Blotting , Células Cultivadas , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Sistema Nervoso Central/ultraestrutura , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Éxons/genética , Deleção de Genes , Heterozigoto , Homozigoto , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Neurônios Motores/ultraestrutura , Neuritos/metabolismo , Neuritos/fisiologia , Estrutura Terciária de Proteína , Espastina
9.
Proc Natl Acad Sci U S A ; 103(25): 9691-6, 2006 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-16766653

RESUMO

The combination of fluorescent genetically encoded proteins with mouse engineering provides a fascinating means to study dynamic biological processes in mammals. At present, green fluorescent protein (GFP) mice were mainly developed to study gene expression patterns or cell morphology and migration. Here we used enhanced GFP (EGFP) to achieve functional imaging of a G protein-coupled receptor (GPCR) in vivo. We created mice where the delta-opioid receptor (DOR) is replaced by an active DOR-EGFP fusion. Confocal imaging revealed detailed receptor neuroanatomy throughout the nervous system of knock-in mice. Real-time imaging in primary neurons allowed dynamic visualization of drug-induced receptor trafficking. In DOR-EGFP animals, drug treatment triggered receptor endocytosis that correlated with the behavioral response. Mice with internalized receptors were insensitive to subsequent agonist administration, providing evidence that receptor sequestration limits drug efficacy in vivo. Direct receptor visualization in mice is a unique approach to receptor biology and drug design.


Assuntos
Proteínas de Fluorescência Verde/metabolismo , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Animais , Encéfalo/metabolismo , Endocitose , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Cinética , Camundongos , Camundongos Transgênicos , Transgenes/genética
10.
Proc Natl Acad Sci U S A ; 103(12): 4511-5, 2006 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-16537395

RESUMO

Deficiencies or mutations in the human pseudoautosomal SHOX gene are associated with a series of short-stature conditions, including Turner syndrome, Leri-Weill dyschondrosteosis, and Langer mesomelic dysplasia. Although this gene is absent from the mouse genome, the closely related paralogous gene Shox2 displays a similar expression pattern in developing limbs. Here, we report that the conditional inactivation of Shox2 in developing appendages leads to a strong phenotype, similar to the human conditions, although it affects a different proximodistal limb segment. Furthermore, using this mouse model, we establish the cellular etiology of these defects and show that Shox2 acts upstream the Runx2 gene, a key regulator of chondrogenesis.


Assuntos
Condrogênese/genética , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento , Transtornos do Crescimento/genética , Proteínas de Homeodomínio/metabolismo , Camundongos/genética , Animais , Desenvolvimento Ósseo/genética , Osso e Ossos/anormalidades , Condrócitos/química , Condrócitos/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/análise , Transtornos do Crescimento/patologia , Proteínas de Homeodomínio/genética , Humanos , Deformidades Congênitas dos Membros/genética , Deformidades Congênitas dos Membros/patologia , Camundongos Mutantes , Fenótipo , Deleção de Sequência , Síndrome
11.
Genesis ; 44(3): 155-8, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16496350

RESUMO

Retinoic acid, the active vitamin A derivative, has pleiotropic functions during vertebrate development and postnatal life. Retinaldehyde dehydrogenase 2 (RALDH2) acts as the main retinoic acid-synthesizing enzyme during development. Mouse Raldh2 germline null mutants are early embryonic lethal and exhibit complex abnormalities that include defective heart looping morphogenesis. To investigate later functions of this enzyme, we have engineered a "floxed" (loxP-flanked) allele allowing Cre-mediated somatic gene inactivations. Mice heterozygous or homozygous for the floxed Raldh2 allele are viable and fertile. We tested whether the novel Raldh2 allele behaves as a null mutation after Cre-mediated in vivo excision by crossing the conditional mutants with CMV-Cre transgenic mice. An embryonic lethal phenotype indistinguishable from that of germline mutants was obtained. The conditional allele described herein is a genetic tool for studying tissue-specific, RALDH2-dependent functions of retinoic acid during development and in adult life.


Assuntos
Aldeído Oxirredutases/genética , Marcação de Genes , Integrases/metabolismo , Tretinoína/metabolismo , Alelos , Animais , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/enzimologia , Feminino , Fibroblastos/citologia , Fibroblastos/enzimologia , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase , Recombinação Genética , Deleção de Sequência , Transfecção
12.
Curr Protoc Mol Biol ; Chapter 29: Unit 29A.1, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18265381

RESUMO

As the focus of human genetics shifts from Mendelian traits to complex diseases, a sophisticated genetic tool kit-with space for genetics (classical, molecular, statistical, and quantitative), metabolics, proteomics, bioinformatics, and mathematics-is required to elucidate their multifactorial traits and regulatory processes. Importantly, mouse resources optimized to study the actions of isolated genetic loci on a fixed background are insufficient on their own for studying intact polygenic networks and genetic interactions, and researchers must work in the context of experimental model systems that optimally mimic the genetic structure of human populations. The success of such phenogenomic approaches depend on the efficacy by which specific mutations (gene targeting) and variability (recombinant inbreeding) can be introduced into the mouse genome, and on the optimization of phenotyping analyses of the mutant mouse lines. This unit describes the basic genetic approaches used to in the study of mouse model systems.


Assuntos
Pesquisa em Genética , Genética Médica/métodos , Modelos Animais , Animais , Camundongos
13.
Genesis ; 41(3): 110-5, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15729686

RESUMO

Sef and Sprouty proteins function as feedback antagonists of fibroblast growth factor (Fgf) signaling in zebrafish embryos. To study the role of Sef in mice, we generated Sef homozygous mutant animals. These animals are viable and show normal expression of mid-hindbrain genes at embryonic days 8.5 and 9.5. To investigate the possibility of functional synergism between Sef and Sprouty proteins, we electroporated Sprouty2(Y55A), which functions in a dominant-negative manner in tissue culture cells into the mid-hindbrain region of wildtype and Sef mutant embryos. The expression pattern of Gbx2, a downstream target of Fgf signaling, was expanded or shifted in electroporated embryos, and this effect was significantly enhanced in the Sef mutant background. Altogether, our results demonstrate that Sef and Sproutys function synergistically to regulate Gbx2 expression in the anterior hindbrain.


Assuntos
Tronco Encefálico/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Proteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Proteínas/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Tronco Encefálico/embriologia , Clonagem Molecular , Eletroporação , Técnicas de Cultura Embrionária , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Fatores de Crescimento de Fibroblastos/fisiologia , Genótipo , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/genética , Camundongos , Camundongos Mutantes , Proteínas do Tecido Nervoso/biossíntese , Proteínas Serina-Treonina Quinases , Proteínas/genética , Transdução de Sinais/fisiologia , Proteínas de Peixe-Zebra
15.
Methods Mol Med ; 99: 269-99, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15131344

RESUMO

Gene targeting in mice by homologous recombination is a powerful approach to study the role of specific genes in vivo. This technology is now applied to pain-related genes to understand molecular mechanisms of nociceptive behaviors. In this chapter, we provide detailed methodological information for the construction of knockout animals, exemplified by the generation of mice lacking opioid receptor genes. We report our protocols for the production, maintenance, transfection, and selection of embryonic stem (ES) cells, as well as for blastocyst injection, which are generally applicable to any gene-targeting project. We also describe strategies for the construction of targeting vectors, as well as for ES cell and animal genotyping, in the context of mu, delta, and kappa opioid receptor genes. We finally provide a few examples of mouse phenotyping in pain behavioral assays.


Assuntos
Modelos Animais de Doenças , Inativação Gênica , Engenharia Genética/métodos , Camundongos Knockout/genética , Dor/genética , Animais , Camundongos , Camundongos Endogâmicos C57BL
16.
Blood ; 103(10): 3615-23, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-14739214

RESUMO

PU.1 is a hematopoietic-specific transcriptional activator that is absolutely required for the differentiation of B lymphocytes and myeloid-lineage cells. Although PU.1 is also expressed by early erythroid progenitor cells, its role in erythropoiesis, if any, is unknown. To investigate the relevance of PU.1 in erythropoiesis, we produced a line of PU.1-deficient mice carrying a green fluorescent protein reporter at this locus. We report here that PU.1 is tightly regulated during differentiation-it is expressed at low levels in erythroid progenitor cells and down-regulated upon terminal differentiation. Strikingly, PU.1-deficient fetal erythroid progenitors lose their self-renewal capacity and undergo proliferation arrest, premature differentiation, and apoptosis. In adult mice lacking one PU.1 allele, similar defects are detected following stress-induced erythropoiesis. These studies identify PU.1 as a novel and critical regulator of erythropoiesis and highlight the versatility of this transcription factor in promoting or preventing differentiation depending on the hematopoietic lineage.


Assuntos
Células Precursoras Eritroides/citologia , Eritropoese , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/fisiologia , Animais , Apoptose , Diferenciação Celular , Divisão Celular , Células Cultivadas , Eritroblastos/citologia , Feto , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde , Proteínas Luminescentes , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Proto-Oncogênicas/genética , Transativadores/genética
17.
Proc Natl Acad Sci U S A ; 100(24): 14457-62, 2003 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-14603033

RESUMO

Peroxisome proliferator-activated receptor gamma (PPAR gamma) is a nuclear receptor, which controls adipocyte differentiation. We targeted with homologous recombination the PPAR gamma 2-specific exon B, resulting in a white adipose tissue knockdown of PPAR gamma. Although homozygous (PPAR gamma hyp/hyp) mice are born with similar weight as the WT mice, the PPAR gamma hyp/hyp animals become growth retarded and develop severe lipodystrophy and hyperlipidemia. Almost half of these PPAR gamma hyp/hyp mice die before adulthood, whereas the surviving PPAR gamma hyp/hyp animals overcome the growth retardation, yet remain lipodystrophic. In contrast to most lipodystrophic models, the adult PPAR gamma hyp/hyp mice only have mild glucose intolerance and do not have a fatty liver. These metabolic consequences of the lipodystrophy are relatively benign because of the induction of a compensatory gene expression program in the muscle that enables efficient oxidation of excess lipids. The PPAR gamma hyp/hyp mice unequivocally demonstrate that PPAR gamma is the master regulator of adipogenesis in vivo and establish that lipid and glucose homeostasis can be relatively well maintained in the absence of white adipose tissue.


Assuntos
Tecido Adiposo/metabolismo , Lipodistrofia/metabolismo , Músculos/metabolismo , Receptores Citoplasmáticos e Nucleares/deficiência , Fatores de Transcrição/deficiência , Tecido Adiposo/patologia , Animais , Feminino , Marcação de Genes , Homozigoto , Lipodistrofia/genética , Lipodistrofia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
18.
J Clin Invest ; 112(4): 544-53, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12925695

RESUMO

We studied the immunological basis for the very potent encephalitogenicity of myelin/oligodendrocyte glycoprotein (MOG), a minor component of myelin in the CNS that is widely used to induce experimental autoimmune encephalomyelitis (EAE). For this purpose, we generated a mutant mouse lacking a functional mog gene. This MOG-deficient mouse presents no clinical or histological abnormalities, permitting us to directly assess the role of MOG as a target autoantigen in EAE. In contrast to WT mice, which developed severe EAE following immunization with whole myelin, MOG-deficient mice had a mild phenotype, demonstrating that the anti-MOG response is a major pathogenic component of the autoimmune response directed against myelin. Moreover, while MOG transcripts are expressed in lymphoid organs in minute amounts, both MOG-deficient and WT mice show similar T and B cell responses against the extracellular domain of MOG, including the immunodominant MOG 35-55 T cell epitope. Furthermore, no differences in the fine specificity of the T cell responses to overlapping peptides covering the complete mouse MOG sequence were observed between MOG+/+ and MOG-/- mice. In addition, upon adoptive transfer, MOG-specific T cells from WT mice and those from MOG-deficient mice are equally pathogenic. This total lack of immune tolerance to MOG in WT C57BL/6 mice may be responsible for the high pathogenicity of the anti-MOG immune response as well as the high susceptibility of most animal strains to MOG-induced EAE.


Assuntos
Tolerância Imunológica , Glicoproteína Associada a Mielina/genética , Glicoproteína Associada a Mielina/fisiologia , Animais , Linfócitos B/imunologia , Northern Blotting , Western Blotting , Encéfalo/metabolismo , Divisão Celular , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microscopia Eletrônica , Modelos Genéticos , Proteínas da Mielina , Bainha de Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito , Peptídeos/química , Fenótipo , Reação em Cadeia da Polimerase , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fatores de Tempo , Distribuição Tecidual
19.
EMBO J ; 22(9): 2255-63, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12727891

RESUMO

The DNA damage-dependent poly(ADP-ribose) polymerases, PARP-1 and PARP-2, homo- and heterodimerize and are both involved in the base excision repair (BER) pathway. Here, we report that mice carrying a targeted disruption of the PARP-2 gene are sensitive to ionizing radiation. Following alkylating agent treatment, parp-2(-/-)-derived mouse embryonic fibroblasts exhibit increased post-replicative genomic instability, G(2)/M accumulation and chromosome mis-segregation accompanying kinetochore defects. Moreover, parp-1(-/-)parp-2(-/-) double mutant mice are not viable and die at the onset of gastrulation, demonstrating that the expression of both PARP-1 and PARP-2 and/or DNA-dependent poly(ADP-ribosyl) ation is essential during early embryogenesis. Interestingly, specific female embryonic lethality is observed in parp-1(+/-)parp-2(-/-) mutants at E9.5. Meta phase analyses of E8.5 embryonic fibroblasts highlight a specific instability of the X chromosome in those females, but not in males. Together, these results support the notion that PARP-1 and PARP-2 possess both overlapping and non-redundant functions in the maintenance of genomic stability.


Assuntos
Apoptose/genética , Cromossomos , Desenvolvimento Embrionário e Fetal/fisiologia , Isoenzimas/fisiologia , Poli(ADP-Ribose) Polimerases/fisiologia , Animais , Sequência de Bases , Dano ao DNA , Primers do DNA , Genes Letais , Isoenzimas/genética , Metilnitrosoureia/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Dados de Sequência Molecular , Poli(ADP-Ribose) Polimerases/genética , Tolerância a Radiação
20.
Science ; 298(5597): 1395-401, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12376594

RESUMO

Humans expressing a defective form of the transcription factor AIRE (autoimmune regulator) develop multiorgan autoimmune disease. We used aire- deficient mice to test the hypothesis that this transcription factor regulates autoimmunity by promoting the ectopic expression of peripheral tissue- restricted antigens in medullary epithelial cells of the thymus. This hypothesis proved correct. The mutant animals exhibited a defined profile of autoimmune diseases that depended on the absence of aire in stromal cells of the thymus. Aire-deficient thymic medullary epithelial cells showed a specific reduction in ectopic transcription of genes encoding peripheral antigens. These findings highlight the importance of thymically imposed "central" tolerance in controlling autoimmunity.


Assuntos
Tolerância a Antígenos Próprios , Linfócitos T/imunologia , Timo/imunologia , Timo/metabolismo , Fatores de Transcrição/metabolismo , Envelhecimento , Animais , Autoanticorpos/análise , Autoanticorpos/sangue , Autoantígenos/biossíntese , Autoantígenos/genética , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Autoimunidade , Células Epiteliais/fisiologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Marcação de Genes , Humanos , Linfócitos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Poliendocrinopatias Autoimunes/genética , Poliendocrinopatias Autoimunes/imunologia , Poliendocrinopatias Autoimunes/metabolismo , Quimera por Radiação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/imunologia , Células Estromais/metabolismo , Timo/citologia , Fatores de Transcrição/genética , Proteína AIRE
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...