Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Mol Endocrinol ; 21(2): 388-400, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17158222

RESUMO

There is a growing interest in peroxisome proliferator-activated receptors (PPARs) as major players in the regulation of lipid and carbohydrate metabolism. Drugs targeting PPARs were in fact shown to have major relevance for the treatment of diseases associated with aging, such as arteriosclerosis and diabetes. However, a variety of toxic effects associated with PPAR ligand administration has been documented, including hepatocarcinogenesis, which may severely limit its therapeutic use. A better comprehension of the multiplicity of PPAR physiological functions is therefore mandatory for the development of novel, safer drugs. We here describe the generation of a novel transgenic mouse for the detection of the generalized activities of PPARs, the PPAR responsive element-Luc reporter mouse. In this model luciferase expression is under the control of a PPAR-inducible promoter in all target organs. By optical imaging and ex vivo analysis, we were able to demonstrate the remarkable gender specificity of the PPAR transcriptional activity in liver. In fact, in the liver of female PPAR responsive element-Luc, the PPAR reporter transgene is more than one order of magnitude less expressed, thus leading to the conclusion that the signaling in females is much less activated than in males. Diet or hormonal manipulations as demonstrated here by treatments with high-fat diet or gonad removal and hormone replacement do not influence this low activation. The extent of the gender difference in PPAR transcriptional activity and the ineffectiveness of hormone treatments or diet to significantly elevate liver PPAR activity in females led us to hypothesize that gender-specific epigenetic events occurring during development may affect PPAR signaling in the liver. This study sets the ground for understanding the differential susceptibility of the two genders to metabolic disorders; furthermore, the model generated provides a novel opportunity for the molecular characterization of PPAR activity in pathophysiological conditions.


Assuntos
Fígado/metabolismo , Luciferases/genética , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Elementos de Resposta , Animais , Gorduras na Dieta/administração & dosagem , Feminino , Regulação da Expressão Gênica , Genes Reporter , Humanos , Luciferases/biossíntese , Masculino , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos , Ovariectomia , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Receptores Ativados por Proliferador de Peroxissomo/genética , Regiões Promotoras Genéticas , Fatores Sexuais , Transdução de Sinais , Testosterona/farmacologia
2.
Mol Cell Biol ; 24(11): 5005-15, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15143191

RESUMO

Overexpression of the ped/pea-15 gene is a common feature of type 2 diabetes. In the present work, we show that transgenic mice ubiquitously overexpressing ped/pea-15 exhibited mildly elevated random-fed blood glucose levels and decreased glucose tolerance. Treatment with a 60% fat diet led ped/pea-15 transgenic mice to develop diabetes. Consistent with insulin resistance in these mice, insulin administration reduced glucose levels by only 35% after 45 min, compared to 70% in control mice. In vivo, insulin-stimulated glucose uptake was decreased by almost 50% in fat and muscle tissues of the ped/pea-15 transgenic mice, accompanied by protein kinase Calpha activation and block of insulin induction of protein kinase Czeta. These changes persisted in isolated adipocytes from the transgenic mice and were rescued by the protein kinase C inhibitor bisindolylmaleimide. In addition to insulin resistance, ped/pea-15 transgenic mice showed a 70% reduction in insulin response to glucose loading. Stable overexpression of ped/pea-15 in the glucose-responsive MIN6 beta-cell line also caused protein kinase Calpha activation and a marked decline in glucose-stimulated insulin secretion. Antisense block of endogenous ped/pea-15 increased glucose sensitivity by 2.5-fold in these cells. Thus, in vivo, overexpression of ped/pea-15 may lead to diabetes by impairing insulin secretion in addition to insulin action.


Assuntos
Diabetes Mellitus/genética , Glucose/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Insulina/metabolismo , Fosfoproteínas/genética , Animais , Proteínas Reguladoras de Apoptose , Diabetes Mellitus/etiologia , Diabetes Mellitus/metabolismo , Antígenos de Histocompatibilidade Classe I/biossíntese , Secreção de Insulina , Camundongos , Camundongos Transgênicos , Fosfoproteínas/biossíntese
3.
Gene ; 377: 150-8, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16787714

RESUMO

Activation-induced cytidine deaminase (AID), an enzyme with homology to members of the APOBEC family, is involved in somatic hypermutation (SHM) of immunoglobulin (Ig) genes, either by direct deamination of DNA or by an indirect action through its putative RNA editing activity. AID is able to mutate both Ig-like reporter constructs and selected non-Ig genes in normal B cells and in other cells when ectopically overexpressed in mammalian cells and transgenic mice. However, in spite of the fact that in these transgenic animals AID activity was driven by an ubiquitous promoter, only T lymphomas and lung adenomas occurred. In the present work, we constructed three sets of transgenic mice in which AID was under the control of lck, HTLV-I and MMTV promoters, respectively. The lck/AID mice developed thymic lymphomas with variable but high efficiency, while no tumor was detected in HTLV-I/AID mice after two years of monitoring. Four MMTV/AID founder mice died with an atypical clinical picture, although no mammary tumor was found. These findings suggest that additional factors, present in thymocytes but not in other tissues or in lymphoid cells at different stages of differentiation, are needed for AID to fully manifest its tumorigenic potential in mouse. Alternatively, the display of full AID mutagenic and transforming activity could be related to the existence of physiologic DSBs which occur in both thymocytes and switching B cells.


Assuntos
Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Animais , Sequência de Bases , Diferenciação Celular , Transformação Celular Neoplásica , DNA Complementar/genética , Proteínas de Ligação a DNA/genética , Feminino , Expressão Gênica , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Genes myc , Genes p53 , Vírus Linfotrópico T Tipo 1 Humano/genética , Rim/enzimologia , Rim/patologia , Fígado/enzimologia , Fígado/patologia , Linfonodos/enzimologia , Linfonodos/patologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/genética , Glândulas Mamárias Animais/enzimologia , Glândulas Mamárias Animais/patologia , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Camundongos Transgênicos , Mutação , Regiões Promotoras Genéticas , Linfócitos T/enzimologia , Linfócitos T/imunologia , Linfócitos T/patologia , Distribuição Tecidual
4.
Novartis Found Symp ; 273: 193-206; discussion 206-12, 261-4, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17120769

RESUMO

Mutations in the SLC26A2 cause a family of recessive chondrodysplasias that includes in order of decreasing severity achondrogenesis 1B, atelosteogenesis 2, diastrophic dysplasia and recessive multiple epiphyseal dysplasia. The gene encodes for a widely distributed sulfate/chloride antiporter of the cell membrane whose function is crucial for the uptake of inorganic sulfate that is needed for proteoglycan sulfation. To investigate the mechanisms leading to skeletal dysplasia, we generated a transgenic mouse with a mutation in Slc26a2 causing a partial loss of function of the sulfate transporter. Homozygous mutant mice were characterized by skeletal dysplasia with chondrocytes of irregular size, delay in the formation of the secondary ossification centre and osteoporosis of long bones. Impaired sulfate uptake was demonstrated in chondrocytes, osteoblasts and fibroblasts, but proteoglycan undersulfation was detected only in cartilage. The similarity with human diastrophic dysplasia makes this mouse a model to explore pathogenetic and therapeutic aspects of SLC26A2-related disorders.


Assuntos
Proteínas de Transporte de Ânions/metabolismo , Modelos Animais de Doenças , Saúde , Animais , Proteínas de Transporte de Ânions/química , Condrócitos/citologia , Sulfatos de Condroitina/metabolismo , Epífises/anormalidades , Camundongos , Camundongos Transgênicos , Transportadores de Sulfato , Sulfatos/metabolismo
5.
Oncogene ; 23(23): 4130-5, 2004 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-15007389

RESUMO

The tyrosinase (Tyr) gene encodes the enzyme tyrosinase that catalyses the conversion of L-tyrosine into DOPA (3,4-dihydroxyphenylalanine)-quinone. The albino mutation abrogates functional activity of tyrosinase resulting in deficiency of melanin pigment production in skin and retina. Tyr maps to a region in the central position of Chromosome 7 that contains a skin tumor-modifier locus. We rescued the albino mutation in transgenic mice to assess a possible role of Tyr gene in two-stage skin carcinogenesis. Transgenic expression of the functional Tyr(Cys) allele in albino mice (Tyr(Ser)) caused a reduction in skin papilloma multiplicity, in four independent experiments and at three dose levels of DMBA (9,10-dimethyl-1,2-benzanthracene). In vitro mechanistic studies demonstrated that transfection of the Tyr(Cys) allele in a human squamous cell carcinoma cell line (NCI-H520) increases tyrosinase enzyme activity and confers resistance to hydrogen peroxide-induced oxidative DNA damage. These results provide direct evidence that the Tyr gene can act as a skin cancer-modifier gene, whose mechanism of action may involve modulation of oxidative DNA damage.


Assuntos
Predisposição Genética para Doença , Monofenol Mono-Oxigenase/deficiência , Neoplasias Cutâneas/enzimologia , Albinismo/enzimologia , Albinismo/genética , Albinismo/metabolismo , Animais , Dano ao DNA , Camundongos , Camundongos Transgênicos , Monofenol Mono-Oxigenase/genética , Monofenol Mono-Oxigenase/metabolismo , Oxirredução , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo
7.
J Invest Dermatol ; 131(9): 1821-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21654836

RESUMO

The 14-3-3 protein family controls diverse biochemical processes through interaction with phosphorylated consensus sequences in protein targets. Its epithelial specific member, 14-3-3σ, also known as stratifin, is highly expressed in differentiated keratinocytes, and in vitro evidence indicates that 14-3-3σ downregulation leads to keratinocyte immortalization. To define the role of 14-3-3σ in skin homeostasis in vivo, we generated transgenic mice overexpressing 14-3-3σ in proliferating keratinocytes of the epidermis and hair follicle. Transgenic animals show decreased epidermal thickness and hair follicle density associated with reduced number of proliferating keratinocytes and decreased levels of keratins 14, 5, and 15. Primary keratinocytes isolated from transgenic mice manifest reduced proliferation and migration. Moreover, clonogenicity assessment and label-retaining analysis reveal a reduction in keratinocyte progenitor cell number in transgenic mice. Response to IGF-1 is strongly impaired in cultured transgenic keratinocytes compared with wild-type cells. Consistently, activation of phosphoinositol 3-kinase (PI3K), AKT, and Rac1, all IGF-1 downstream mediators, is reduced. Our results demonstrate that 14-3-3σ controls the in vivo epidermal proliferation-differentiation switch by reducing proliferative potential and forcing keratinocytes to exit the cell cycle, and that this effect associates with inhibition of the IGF-1 pathway.


Assuntos
Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Epiderme/fisiologia , Exonucleases/genética , Exonucleases/metabolismo , Folículo Piloso/fisiologia , Queratinócitos/fisiologia , Animais , Divisão Celular/fisiologia , Células Cultivadas , Células Clonais/citologia , Células Clonais/fisiologia , Células Epidérmicas , Exorribonucleases , Folículo Piloso/citologia , Fator de Crescimento Insulin-Like I/metabolismo , Queratinócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Fenótipo , Regiões Promotoras Genéticas/fisiologia , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia
8.
EMBO Rep ; 6(8): 755-61, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16007071

RESUMO

Although it has been clearly established that negative feedback loops have a fundamental role in the regulation of epidermal growth factor receptor (EGFR) signalling in flies, their role in the regulation of mammalian EGFR has been inferred only recently from in vitro studies. Here, we report on the forced expression of RALT/MIG-6, a negative feedback regulator of ErbB receptors, in mouse skin. A RALT transgene driven by the K14 promoter generated a dose-dependent phenotype resembling that caused by hypomorphic and antimorphic Egfr alleles-that is, wavy coat, curly whiskers and open eyes at birth. Ex vivo keratinocytes from K14-RALT mice showed reduced biochemical and biological responses when stimulated by ErbB ligands. Conversely, knockdown of RALT by RNA interference enhanced ErbB mitogenic signalling. Thus, RALT behaves as a suppressor of EGFR signalling in mouse skin.


Assuntos
Receptores ErbB/metabolismo , Cabelo/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Pele/metabolismo , Alelos , Animais , Western Blotting , Bromodesoxiuridina/farmacologia , Linhagem Celular , Proliferação de Células , Células Cultivadas , Relação Dose-Resposta a Droga , Fator de Crescimento Epidérmico/metabolismo , Heterozigoto , Humanos , Immunoblotting , Queratinócitos/citologia , Queratinócitos/metabolismo , Ligantes , Camundongos , Camundongos Transgênicos , Células NIH 3T3 , Proteínas Oncogênicas v-erbB/metabolismo , Fases de Leitura Aberta , Fenótipo , Fosforilação , Regiões Promotoras Genéticas , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Transgenes , Vibrissas
9.
Hum Mol Genet ; 14(6): 859-71, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15703192

RESUMO

Mutations in the diastrophic dysplasia sulfate transporter (DTDST or SLC26A2) cause a family of recessively inherited chondrodysplasias including, in order of decreasing severity, achondrogenesis 1B, atelosteogenesis 2, diastrophic dysplasia (DTD) and recessive multiple epiphyseal dysplasia. The gene encodes a widely distributed sulfate/chloride antiporter of the cell membrane whose function is crucial for the uptake of inorganic sulfate, which is needed for proteoglycan sulfation. To provide new insights in the pathogenetic mechanisms leading to skeletal and connective tissue dysplasia and to obtain an in vivo model for therapeutic approaches to DTD, we generated a Dtdst knock-in mouse with a partial loss of function of the sulfate transporter. In addition, the intronic neomycine cassette in the mutant allele contributed to the hypomorphic phenotype by inducing abnormal splicing. Homozygous mutant mice were characterized by growth retardation, skeletal dysplasia and joint contractures, thereby recapitulating essential aspects of the DTD phenotype in man. The skeletal phenotype included reduced toluidine blue staining of cartilage, chondrocytes of irregular size, delay in the formation of the secondary ossification center and osteoporosis of long bones. Impaired sulfate uptake was demonstrated in chondrocytes, osteoblasts and fibroblasts. In spite of the generalized nature of the sulfate uptake defect, significant proteoglycan undersulfation was detected only in cartilage. Chondrocyte proliferation and apoptosis studies suggested that reduced proliferation and/or lack of terminal chondrocyte differentiation might contribute to reduced bone growth. The similarity with human DTD makes this mouse strain a useful model to explore pathogenetic and therapeutic aspects of DTDST-related disorders.


Assuntos
Proteínas de Transporte/genética , Osteocondrodisplasias/genética , Osteocondrodisplasias/patologia , Fenótipo , Animais , Proteínas de Transporte de Ânions , Proteínas de Transporte/metabolismo , Modelos Animais de Doenças , Humanos , Proteínas de Membrana Transportadoras , Camundongos , Camundongos Transgênicos , Osteocondrodisplasias/metabolismo , Transportadores de Sulfato
10.
J Cell Sci ; 115(Pt 12): 2559-67, 2002 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12045226

RESUMO

Placenta growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family, comprising at least five cytokines specifically involved in the regulation of vascular and/or lymphatic endothelium differentiation. Several lines of evidence indicate a role for PlGF in monocyte chemotaxis and in potentiating the activity of VEGF, but the exact function of this cytokine is not fully understood. To define the biological role of PlGF in vivo, we have produced a transgenic mouse model overexpressing this factor in the skin by using a keratin 14 promoter cassette. Our data indicate that PlGF has strong angiogenic properties in both fetal and adult life. PlGF overexpression results in a substantial increase in the number, branching and size of dermal blood vessels as well as in enhanced vascular permeability. Indeed, intradermally injected recombinant PlGF was able to induce vessel permeability in wild-type mice. The analysis of vascular endothelial growth factor receptor 1/flt-1 and vascular endothelial growth factor receptor 2/flk-1 indicates that the two receptors are induced in the skin endothelium of transgenic mice suggesting that both are involved in mediating the effect of overexpressed PlGF.


Assuntos
Permeabilidade Capilar/genética , Endotélio Vascular/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/genética , Neovascularização Fisiológica/fisiologia , Proteínas da Gravidez/metabolismo , Pele/irrigação sanguínea , Pele/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Feminino , Queratina-14 , Queratinas/genética , Masculino , Camundongos , Camundongos Transgênicos , Modelos Animais , Cadeias Pesadas de Miosina , Miosina não Muscular Tipo IIB , Fator de Crescimento Placentário , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteínas da Gravidez/genética , Regiões Promotoras Genéticas/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , Proteínas Recombinantes de Fusão/genética , Pele/citologia , Regulação para Cima/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
11.
Hum Mol Genet ; 12(4): 399-413, 2003 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-12566387

RESUMO

We report here the creation of a constitutive knockout mouse for SURF1, a gene encoding one of the assembly proteins involved in the formation of cytochrome c oxidase (COX). Loss-of-function mutations of SURF1 cause Leigh syndrome associated with an isolated and generalized COX deficiency in humans. The murine phenotype is characterized by the following hallmarks: (1) high post-implantation embryonic lethality, affecting approximately 90% of the Surf1(-/-) individuals; (2) early-onset mortality of post-natal individuals; (3) highly significant deficit in muscle strength and motor performance; (4) profound and isolated defect of COX activity in skeletal muscle and liver, and, to a lesser extent, heart and brain; (5) morphological abnormalities of skeletal muscle, characterized by reduced histochemical reaction to COX and mitochondrial proliferation; (6) no obvious abnormalities in brain morphology, reflecting the virtual absence of overt neurological symptoms. These results indicate a function for murine Surf1 protein (Surf1p) specifically related to COX and recapitulate, at least in part, the human phenotype. This is the first mammalian model for a nuclear disease gene of a human mitochondrial disorder. Our model constitutes a useful tool to investigate the function of Surf1p, help understand the pathogenesis of Surf1p deficiency in vivo, and evaluate the efficacy of treatment.


Assuntos
Deficiência de Citocromo-c Oxidase/genética , Complexo IV da Cadeia de Transporte de Elétrons/genética , Doenças Mitocondriais/genética , Proteínas/genética , Alelos , Animais , Southern Blotting , Western Blotting , Encéfalo/enzimologia , Eletroforese em Gel de Poliacrilamida , Feminino , Vetores Genéticos , Humanos , Immunoblotting , Imuno-Histoquímica , Fígado/enzimologia , Masculino , Proteínas de Membrana , Camundongos , Camundongos Knockout , Proteínas Mitocondriais , Modelos Genéticos , Músculo Esquelético/enzimologia , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Distribuição Tecidual
12.
J Biol Chem ; 279(40): 41846-57, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15262984

RESUMO

The Sox2 transcription factor is expressed early in the stem cells of the blastocyst inner cell mass and, later, in neural stem cells. We previously identified a Sox2 5'-regulatory region directing transgene expression to the inner cell mass and, later, to neural stem cells and precursors of the forebrain. Here, we identify a core enhancer element able to specify transgene expression in forebrain neural precursors of mouse embryos, and we show that the same core element efficiently activates transcription in inner cell mass-derived embryonic stem (ES) cells. Mutation of POU factor binding sites, able to recognize the neural factors Brn1 and Brn2, shows that these sites contribute to transgene activity in neural cells. The same sites are also essential for activity in ES cells, where they bind different members of the POU family, including Oct4, as shown by gel shift assays and chromatin immunoprecipitation with anti-Oct4 antibodies. Our findings indicate a role for the same POU binding motifs in Sox2 transgene regulation in both ES and neural precursor cells. Oct4 might play a role in the regulation of Sox2 in ES (inner cell mass) cells and, possibly, at the transition between inner cell mass and neural cells, before recruitment of neural POU factors such as Brn1 and Brn2.


Assuntos
Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Nucleares/genética , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Região 5'-Flanqueadora , Animais , Sequência de Bases , Sítios de Ligação , Sequência Conservada , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos/citologia , Proteínas HMGB , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Fator 3 de Transcrição de Octâmero , Fatores de Transcrição SOXB1
13.
Blood ; 102(12): 3954-62, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12907433

RESUMO

The Kit (White) gene encodes the transmembrane receptor of stem cell factor/Kit ligand (KL) and is essential for the normal development/maintenance of pluripotent primordial germ cells (PGCs), hematopoietic stem cells (HSCs), melanoblasts, and some of their descendants. The molecular basis for the transcriptional regulation of Kit during development of these important cell types is unknown. We investigated Kit regulation in hematopoietic cells and PGCs. We identified 6 DNase I hypersensitive sites (HS1-HS6) within the promoter and first intron of the mouse Kit gene and developed mouse lines expressing transgenic green fluorescent protein (GFP) under the control of these regulatory elements. A construct driven by the Kit promoter and including all 6 HS sites is highly expressed during mouse development in Kit+ cells including PGCs and hematopoietic progenitors (erythroid blast-forming units and mixed colony-forming units). In contrast, the Kit promoter alone (comprising HS1) is sufficient to drive low-level GFP expression in PGCs, but unable to function in hematopoietic cells. Hematopoietic expression further requires the addition of the intronproximal HS2 fragment; HS2 also greatly potentiates the activity in PGCs. Thus, HS2 acts as an enhancer integrating transcriptional signals common to 2 developmentally unrelated stem cell/progenitor lineages. Optimal hematopoietic expression further requires HS3-HS6.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Genes Reguladores , Células Germinativas/citologia , Células-Tronco Hematopoéticas/citologia , Proteínas Proto-Oncogênicas c-kit/genética , Animais , Linhagem da Célula , Células Cultivadas , Desoxirribonuclease I , Embrião de Mamíferos , Proteínas de Fluorescência Verde , Hematopoese/genética , Proteínas Luminescentes/genética , Camundongos , Camundongos Transgênicos , Células-Tronco Multipotentes/citologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-kit/biossíntese , Distribuição Tecidual
14.
Hum Mol Genet ; 11(21): 2567-80, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12354782

RESUMO

Non-specific mental retardation (NSMR) is a common human disorder characterized by mental handicap as the only clinical symptom. Among the recently identified MR genes is GDI1, which encodes alpha Gdi, one of the proteins controlling the activity of the small GTPases of the Rab family in vesicle fusion and intracellular trafficking. We report the cognitive and behavioral characterization of mice carrying a deletion of Gdi1. The Gdi1-deficient mice are fertile and anatomically normal. They appear normal also in many tasks to assess spatial and episodic memory and emotional behavior. Gdi1-deficient mice are impaired in tasks requiring formation of short-term temporal associations, suggesting a defect in short-term memory. In addition, they show lowered aggression and altered social behavior. In mice, as in humans, lack of Gdi1 spares most central nervous system functions and preferentially impairs only a few forebrain functions required to form temporal associations. The general similarity to human mental retardation is striking, and suggests that the Gdi1 mutants may provide insights into the human defect and into the molecular mechanisms important for development of cognitive functions.


Assuntos
Inibidores de Dissociação do Nucleotídeo Guanina/genética , Memória , Comportamento Social , Animais , Encéfalo/citologia , Deleção de Genes , Hibridização In Situ , Deficiência Intelectual/genética , Camundongos , Camundongos Knockout , Proteínas rab de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA