Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Exp Neurol ; 171(2): 255-71, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11573978

RESUMEN

The cell-specific expression of both the oxytocin (OT) and vasopressin (VP) genes in magnocellular neurons (MCNs) of the hypothalamus has been proposed to be under the control of cis-elements in an intergenic region downstream of the VP gene. We examined this hypothesis using transgenic mice containing mouse genomic DNA-derived constructs linked to chloramphenicol acetyltransferase (CAT) reporters. VP gene expression was studied using constructs containing 3.8 kbp of the 5' flanking region and all the exons and introns in the mouse VP gene, which was fused at the end of exon 3 to a CAT reporter. The two VP-transgene constructs differed by the lengths of their VP gene 3' flanking regions (2.1 versus 3.6 kbp). A similar construct for the oxytocin CAT transgene was used which contained the full-length (3.6 kbp) downstream intergenic region between the mouse genes. All three transgenic constructs produced cell-specific expression of the CAT-reporter in the magnocellular neurons as determined by CAT-immunoreactivity. Oxytocin transgene expression was restricted to OT cells in two founders, and the two VP transgenes to VP cells in five founders. Electron microscopic immunocytochemistry showed that the CAT fusion proteins produced from the OT- and VP-transgenes were efficiently trafficked through the regulated secretory pathways in their respective magnocellular neurons, packaged into large dense core vesicles, and transported to nerve terminals in the posterior pituitary.


Asunto(s)
Cloranfenicol O-Acetiltransferasa/genética , Regulación de la Expresión Génica/fisiología , Ratones Transgénicos , Neurofisinas/genética , Oxitocina/genética , Núcleo Hipotalámico Paraventricular/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Núcleo Supraóptico/metabolismo , Vasopresinas/genética , Amígdala del Cerebelo/metabolismo , Animales , Cloranfenicol O-Acetiltransferasa/análisis , Exones , Genes Reporteros , Giro del Cíngulo/metabolismo , Ratones , Microscopía Inmunoelectrónica , Neurofisinas/análisis , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/ultraestructura , Proteínas Recombinantes de Fusión/análisis , Núcleo Supraóptico/citología , Núcleo Supraóptico/ultraestructura
2.
Dev Biol ; 237(2): 245-57, 2001 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-11543611

RESUMEN

SOX10 is a high-mobility-group transcription factor that plays a critical role in the development of neural crest-derived melanocytes. At E11.5, mouse embryos homozygous for the Sox10(Dom) mutation entirely lack neural crest-derived cells expressing the lineage marker KIT, MITF, or DCT. Moreover, neural crest cell cultures derived from homozygous embryos do not give rise to pigmented cells. In contrast, in Sox10(Dom) heterozygous embryos, melanoblasts expressing KIT and MITF do occur, albeit in reduced numbers, and pigmented cells eventually develop in nearly normal numbers both in culture and in vivo. Intriguingly, however, Sox10(Dom)/+ melanoblasts transiently lack Dct expression both in culture and in vivo, suggesting that during a critical developmental period SOX10 may serve as a transcriptional activator of Dct. Indeed, we found that SOX10 and DCT colocalized in early melanoblasts and that SOX10 is capable of transactivating the Dct promoter in vitro. Our data suggest that during early melanoblast development SOX10 acts as a critical transactivator of Dct, that MITF, on its own, is insufficient to stimulate Dct expression, and that delayed onset of Dct expression is not deleterious to the melanocyte lineage.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/fisiología , Oxidorreductasas Intramoleculares/metabolismo , Melanocitos/metabolismo , Cresta Neural/embriología , Factores de Transcripción , Transcripción Genética , Animales , Animales Modificados Genéticamente , Linaje de la Célula , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Galactósidos/metabolismo , Genotipo , Heterocigoto , Homocigoto , Inmunohistoquímica , Hibridación in Situ , Indoles/metabolismo , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Transcripción Asociado a Microftalmía , Mutación , Pigmentación/genética , Plásmidos/metabolismo , Factores de Transcripción SOXE , Factores de Tiempo , Transfección
3.
Development ; 127(24): 5379-89, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11076759

RESUMEN

Genetic and cell culture analyses have shown that the development of melanocytes from neural crest-derived precursor cells critically depends on the tyrosine kinase receptor KIT and the basic-helix-loop-helix-leucine zipper transcription factor MITF. KIT and MITF show complex interactions in that MITF is needed for the maintenance of Kit expression in melanoblasts and KIT signaling modulates MITF activity and stability in melanocyte cell lines. Using primary neural crest cell cultures from embryos homozygous for a Kit null allele marked by an inserted LacZ gene (Kit(W-LacZ)), we show that the onset of Mitf expression in melanoblasts does not require KIT. In fact, provided that the melanocyte growth factor endothelin-3 is present, a small number of MITF/beta-Gal-positive cells can be maintained for at least 2 weeks in Kit(W-LacZ)/Kit(W-LacZ) cultures. These cells express several pigment cell-specific genes that are thought or have been shown to be activated by MITF, including dautochrome tautomerase, pMel 17/Silver and tyrosinase-related protein-1, but lack expression of the MITF target gene tyrosinase, which encodes the rate-limiting enzyme in melanin synthesis. Consequently, the cells remain unpigmented. Addition of cholera toxin, which elevates cAMP levels and mimics part of the KIT signaling pathway, increases the number of MITF-positive cells in Kit(W-LacZ)/Kit(W-LacZ) cultures, leads to tyrosinase expression, and induces the differentiation of melanoblasts into mature, pigmented melanocytes. Even when added on day 5-6 of culture, cholera toxin still rescues tyrosinase expression and differentiation. The results thus demonstrate that the presence of MITF is not sufficient for tyrosinase expression in melanoblasts and that KIT signaling influences gene expression during melanocyte development in a gene-selective manner.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Melanocitos/citología , Melanocitos/metabolismo , Cresta Neural/citología , Cresta Neural/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factores de Transcripción , Animales , Secuencia de Bases , Células Cultivadas , AMP Cíclico/metabolismo , Cartilla de ADN/genética , Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Operón Lac , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor de Transcripción Asociado a Microftalmía , Monofenol Monooxigenasa/genética , Proteínas Proto-Oncogénicas c-kit/genética , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Transcripción Genética
4.
Hum Genet ; 107(1): 1-6, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10982026

RESUMEN

Waardenburg syndrome (WS) is associated with neural crest-derived melanocyte deficiency caused by mutations in either one of three transcription factors: MITF, PAX3, and SOX10. However, the hierarchical relationship of these transcription factors is largely unknown. We show that SOX10 is capable of transactivating the MITF promoter 100-fold, and that this transactivation is further stimulated by PAX3. Promoter deletion and mutational analyses indicate that SOX10 can activate MITF expression through binding to a region that is evolutionarily conserved between the mouse and human MITF promoters. A SOX10 mutant that models C-terminal truncations in WS can reduce wild-type SOX10 induction of MITF, suggesting these mutations may act in a dominant-negative fashion. Our data support a model in which the hypopigmentation in WS, of which these factors have been implicated, results from a disruption in function of the central melanocyte transcription factor MITF.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/genética , Factores de Transcripción/genética , Síndrome de Waardenburg/genética , Animales , Secuencia de Bases , Secuencia Conservada , Proteínas de Unión al ADN/biosíntesis , Evolución Molecular , Eliminación de Gen , Genes Dominantes , Genotipo , Células HeLa , Proteínas del Grupo de Alta Movilidad/biosíntesis , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Mutantes , Factor de Transcripción Asociado a Microftalmía , Datos de Secuencia Molecular , Mutagénesis , Mutación , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box , Fenotipo , Regiones Promotoras Genéticas , Factores de Transcripción SOXE , Homología de Secuencia de Ácido Nucleico , Factores de Tiempo , Transfección
5.
Development ; 127(16): 3581-91, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10903182

RESUMEN

During vertebrate eye development, the optic vesicle is partitioned into a domain at its distal tip that will give rise to the neuroretina, and another at its proximal base that will give rise to the pigmented epithelium. Both domains are initially bipotential, each capable of giving rise to either neuroretina or pigmented epithelium. The partitioning depends on extrinsic signals, notably fibroblast growth factors, which emanate from the overlying surface ectoderm and induce the adjacent neuroepithelium to assume the neuroretinal fate. Using explant cultures of mouse optic vesicles, we demonstrate that bipotentiality of the optic neuroepithelium is associated with the initial coexpression of the basic-helix-loop-helix-zipper transcription factor MITF, which is later needed solely in the pigmented epithelium, and a set of distinct transcription factors that become restricted to the neuroretina. Implantation of fibroblast growth factor-coated beads close to the base of the optic vesicle leads to a rapid downregulation of MITF and the development of an epithelium that, by morphology, gene expression, and lack of pigmentation, resembles the future neuroretina. Conversely, the removal of the surface ectoderm results in the maintenance of MITF in the distal optic epithelium, lack of expression of the neuroretinal-specific CHX10 transcription factor, and conversion of this epithelium into a pigmented monolayer. This phenomenon can be prevented by the application of fibroblast growth factor alone. In Mitf mutant embryos, parts of the future pigment epithelium become thickened, lose expression of a number of pigment epithelium transcription factors, gain expression of neuroretinal transcription factors, and eventually transdifferentiate into a laminated second retina. The results support the view that the bipotential optic neuroepithelium is characterized by overlapping gene expression patterns and that selective gene repression, brought about by local extrinsic signals, leads to the separation into discrete expression domains and, hence, to domain specification.


Asunto(s)
Proteínas de Unión al ADN/genética , Ojo/embriología , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Transducción de Señal , Factores de Transcripción/genética , Animales , Regulación hacia Abajo , Ectodermo , Secuencias Hélice-Asa-Hélice , Leucina Zippers , Mamíferos , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción Asociado a Microftalmía , Mutagénesis , Epitelio Pigmentado Ocular/embriología , Epitelio Pigmentado Ocular/metabolismo , Transcripción Genética
6.
Mamm Genome ; 9(8): 617-21, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9680380

RESUMEN

The rat gene encoding the microphthalmia-associated transcription factor (Mitf) was assigned to rat Chromosome (Chr) 4q34-q41, as well as the Gata2 and Mem1 genes. Rat Chr 4 is homologous to mouse Chr 6 and human Chr 3, which carry the Mitf (MITF) gene in these species (MMU 6, 40.0 cM, and HSA 3p14.1-p12.3). mib/mib rats, which are characterized by depigmentation, microphtalmy, osteopetrosis, and neurological disorders were shown to bear a deletion covering several kilobases of genomic DNA in the Mitf gene and to lack Mitf mRNA. The Mitf mutation in the mib/mib rats is thus very likely to be a Mitf null mutation, causing a phenotype similar to the one observed in the miVGA-9 mice, but including osteopetrosis as an additional feature.


Asunto(s)
Mapeo Cromosómico , Proteínas de Unión al ADN/genética , Microftalmía/genética , Factores de Transcripción/genética , Animales , Cromosomas Humanos Par 3 , Proteínas de Unión al ADN/biosíntesis , Marcadores Genéticos , Humanos , Células Híbridas , Ratones , Factor de Transcripción Asociado a Microftalmía , Reacción en Cadena de la Polimerasa , Ratas , Ratas Mutantes
7.
Hum Mol Genet ; 7(4): 703-8, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9499424

RESUMEN

Mutations in MITF (microphthalmia transcription factor) cause Waardenburg syndrome type 2 (WS2A) in humans, an autosomal dominant disorder consisting of deafness and hypopigmentation. Phenotypes vary significantly within WS2 pedigrees, and there is generally no correlation between the predicted biochemical properties of mutant MITF proteins and disease severity. We have identified a nonsense mutation in the Mitf gene of the anophthalmic white Wh) Syrian hamster that destabilizes its mRNA and prevents the encoded basic helix-loop-helix leucine zipper (bHLHzip) protein from dimerizing or binding DNA target sites. Although the resulting polypeptide does not act as a dominant-negative species in vitro , the Wh mutation is inherited as a semi-dominant trait. It thus more closely resembles WS2 than comparable Mitf alleles in laboratory mice and rats, which are expressed as purely recessive traits.


Asunto(s)
Anoftalmos/genética , Proteínas de Unión al ADN/genética , Factores de Transcripción , Síndrome de Waardenburg/genética , Secuencia de Aminoácidos , Animales , Northern Blotting , Cricetinae , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Haploidia , Inmunohistoquímica , Mesocricetus , Factor de Transcripción Asociado a Microftalmía , Datos de Secuencia Molecular , Mutación , Epitelio Pigmentado Ocular/metabolismo , Mutación Puntual , Reacción en Cadena de la Polimerasa , Síndrome de Waardenburg/patología
8.
Proc Natl Acad Sci U S A ; 95(6): 3188-92, 1998 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-9501238

RESUMEN

The gastrin-releasing peptide receptor (GRP-R) is one of three members of the mammalian bombesin subfamily of seven-transmembrane G protein-coupled receptors that mediate diverse biological responses including secretion, neuromodulation, chemotaxis, and growth. The X chromosome-linked GRP-R gene is expressed widely during embryonic development and predominantly in gastrointestinal, neuronal, and neuroendocrine systems in the adult. Surprisingly, gene-targeted mice lacking a functional GRP-R gene develop and reproduce normally and show no gross phenotypic abnormalities. However, peripheral administration of bombesin at dosages up to 32 nmol/kg to such mice had no effect on the suppression of glucose intake, whereas normal mice showed a dose-dependent suppression of glucose intake. These data suggest that selective agonists for the GRP-R may be useful in inducing satiety.


Asunto(s)
Bombesina/farmacología , Ingestión de Alimentos/efectos de los fármacos , Receptores de Bombesina/deficiencia , Saciedad/fisiología , Amilasas/metabolismo , Animales , Carbacol/farmacología , Relación Dosis-Respuesta a Droga , Ratones , Ratones Mutantes , Páncreas/efectos de los fármacos , Receptores de Bombesina/agonistas , Receptores de Bombesina/genética , Sincalida/farmacología
10.
Mech Dev ; 70(1-2): 155-66, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9510032

RESUMEN

The mouse microphthalmia (Mitf) gene encodes a basic-helix-loop-helix-zipper transcription factor whose mutations are associated with abnormalities in neuroepithelial and neural crest-derived melanocytes. In wild type embryos, Mitf expression in neuropithelium and neural crest precedes that of the melanoblast marker Dct, is then co-expressed with Dct, and gradually fades away except in cells in hair follicles. In embryos with severe Mitf mutations, neural crest-derived Mitf-expressing cells are rare, lack Dct expression, and soon become undetectable. In contrast, the neuroepithelial-derived Mitf-expressing cells of the retinal pigment layer are retained, express Dct, but not the melanogenic enzyme genes tyrosinase and Tyrp1, and remain unpigmented. The results show that melanocyte development critically depends on functional Mitf and that Mitf mutations affect the neural crest and the neuroepithelium in different ways.


Asunto(s)
Proteínas de Unión al ADN/genética , Secuencias Hélice-Asa-Hélice/genética , Melanocitos/citología , Melanocitos/metabolismo , Mutación , Factores de Transcripción/genética , Animales , Diferenciación Celular , Sordera/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Marcadores Genéticos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Factor de Transcripción Asociado a Microftalmía , Sistema Nervioso/citología , Sistema Nervioso/embriología , Cresta Neural/citología , Epitelio Pigmentado Ocular/embriología
11.
Biochem Cell Biol ; 76(6): 1093-9, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-10392719

RESUMEN

In both mice and humans, mutations in the genes encoding the endothelin B receptor and its ligand endothelin 3 lead to deficiencies in neural crest-derived melanocytes and enteric neurons. The discrete steps at which endothelins exert their functions in melanocyte development were examined in mouse neural crest cell cultures. Such cultures, kept in the presence of fetal calf serum, gave rise to cells expressing the early melanoblast marker Dct even in the absence of the phorbol ester tetradecanoyl phorbol acetate (TPA) or endothelins. However, these early Dct+ cells did not proliferate and pigmented cells never formed unless TPA or endothelins were added. In fact, endothelin 2 was as potent as TPA in promoting the generation of both Dct+ melanoblasts and pigmented cells, and endothelin 1 or endothelin 3 stimulated the generation of melanoblasts and of pigmented cells to an even greater extent. The inhibition of this stimulation by the selective endothelin B receptor antagonist BQ-788 (N-cis-2,6-dimethylpiperidinocarbonyl-L-alpha-methylleucyl-D -1-methoxycarbonyltryptophanyl-D-norleucine) suggested that the three endothelins all signal through the endothelin B receptor. This receptor was indeed expressed in Dct+ melanoblasts, in addition to cells lacking Dct expression. The results demonstrate that endothelins are potent stimulators of melanoblast proliferation and differentiation.


Asunto(s)
Endotelinas/fisiología , Melanocitos/fisiología , Cresta Neural/embriología , Transducción de Señal , Animales , Bromodesoxiuridina/metabolismo , División Celular , Supervivencia Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Endotelina-1/metabolismo , Endotelina-2/metabolismo , Endotelina-3/metabolismo , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Receptor de Endotelina B , Receptores de Endotelina/metabolismo , Acetato de Tetradecanoilforbol/metabolismo , Factores de Tiempo
12.
J Virol ; 71(12): 9792-5, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9371647

RESUMEN

Rat Mx2 and rat Mx3 are two alpha/beta interferon-inducible cytoplasmic GTPases that differ in three residues in the amino-terminal third, which also contains the tripartite GTP-binding domain, and that differ in five residues in the carboxy-terminal quarter, which also contains a dimerization domain. While Mx2 is active against vesicular stomatitis virus (VSV), Mx3 lacks antiviral activity. We mapped the functional difference between Mx2 and Mx3 protein to two critical residues in the carboxy-terminal parts of the molecules. An exchange of either residue 588 or 630 of Mx2 with the corresponding residues of Mx3 abolished anti-VSV activity, and the introduction of the two Mx2 residues on an Mx3 background partially restored anti-VSV activity. These results are consistent with the facts that Mx2 and Mx3 have similar intrinsic GTPase activities and that the GTPase domain of Mx3 can fully substitute for the GTPase domain of Mx2. Nevertheless, the amino-terminal third containing the GTP-binding domain is necessary for antiviral activity, since an amino-terminally truncated Mx2 protein is devoid of anti-VSV activity. Furthermore, Fab fragments of a monoclonal antibody known to neutralize antiviral activity block GTPase activity by binding an epitope in the carboxy-terminal half of Mx2 or Mx3 protein. The results are consistent with a two-domain model in which both the conserved amino-terminal half and the less-well-conserved carboxy-terminal half of Mx proteins carry functionally important domains.


Asunto(s)
Antivirales/inmunología , Epítopos/inmunología , GTP Fosfohidrolasas/inmunología , Proteínas de Unión al GTP , Proteínas/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Células 3T3 , Animales , Antivirales/genética , GTP Fosfohidrolasas/genética , Ratones , Proteínas de Resistencia a Mixovirus , Proteínas/genética , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología
13.
Proc Natl Acad Sci U S A ; 94(23): 12274-9, 1997 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-9356439

RESUMEN

We describe a method to design dominant-negative proteins (D-N) to the basic helix-loop-helix-leucine zipper (B-HLHZip) family of sequence-specific DNA binding transcription factors. The D-Ns specifically heterodimerize with the B-HLHZip dimerization domain of the transcription factors and abolish DNA binding in an equimolar competition. Thermal denaturation studies indicate that a heterodimer between a Myc B-HLHZip domain and a D-N consisting of a 12-amino acid sequence appended onto the Max dimerization domain (A-Max) is -6.3 kcal.mol-1 more stable than the Myc:Max heterodimer. One molar equivalent of A-Max can totally abolish the DNA binding activity of a Myc:Max heterodimer. This acidic extension also has been appended onto the dimerization domain of the B-HLHZip protein Mitf, a member of the transcription factor enhancer binding subfamily, to produce A-Mitf. The heterodimer between A-Mitf and the B-HLHZip domain of Mitf is -3.7 kcal.mol-1 more stable than the Mitf homodimer. Cell culture studies show that A-Mitf can inhibit Mitf-dependent transactivation both in acidic extension and in a dimerization-dependent manner. A-Max can inhibit Myc-dependent foci formation twice as well as the Max dimerization domain (HLHZip). This strategy of producing D-Ns may be applicable to other B-HLHZip or B-HLH proteins because it provides a method to inhibit the DNA binding of these transcription factors in a dimerization-specific manner.


Asunto(s)
Proteínas de Unión al ADN/química , Secuencias Hélice-Asa-Hélice , Leucina Zippers , Ingeniería de Proteínas , Secuencia de Aminoácidos , Proteínas de Unión al ADN/genética , Dimerización , Datos de Secuencia Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Factores de Transcripción/química , Factores de Transcripción/genética
14.
Development ; 124(12): 2377-86, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9199364

RESUMEN

The more than 20 different Mitf mutations in the mouse are all associated with deficiencies in neural crest-derived melanocytes that range from minor functional disturbances with some alleles to complete absence of mature melanocytes with others. In the trunk region of wild-type embryos, Mitf-expressing cells that coexpressed the melanoblast marker Dct and the tyrosine kinase receptor Kit were found in the dorsolateral neural crest migration pathway. In contrast, in embryos homozygous for an Mitf allele encoding a non-functional Mitf protein, Mitf-expressing cells were extremely rare, no Dct expression was ever found, and the number of Kit-expressing cells was much reduced. Wild-type neural crest cell cultures rapidly gave rise to cells that expressed Mitf and coexpressed Kit and Dct. With time in culture, Kit expression was increased, and pigmented, dendritic cells developed. Addition of the Kit ligand Mgf or endothelin 3 or a combination of these factors all rapidly increased the number of Dct-positive cells. Cultures from Mitf mutant embryos initially displayed Mitf-positive cells similar in numbers and Kit-expression as did wild-type cultures. However, Kit expression did not increase with time in culture and the mutant cells never responded to Mgf or endothelin 3, did not express Dct, and never showed pigment. In fact, even Mitf expression was rapidly lost. The results suggest that Mitf first plays a role in promoting the transition of precursor cells to melanoblasts and subsequently, by influencing Kit expression, melanoblast survival.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación del Desarrollo de la Expresión Génica , Oxidorreductasas Intramoleculares , Melanocitos/fisiología , Cresta Neural/citología , Cresta Neural/embriología , Factores de Transcripción , Secuencia de Aminoácidos , Animales , Diferenciación Celular/genética , Células Cultivadas , Proteínas de Unión al ADN/efectos de los fármacos , Embrión de Mamíferos/fisiología , Endotelina-3/farmacología , Isomerasas/genética , Melanocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Factor de Transcripción Asociado a Microftalmía , Datos de Secuencia Molecular , Mutación , Proteínas Proto-Oncogénicas c-kit/genética , Factor de Células Madre/farmacología
15.
Mol Cell Biol ; 16(3): 1203-11, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8622664

RESUMEN

In the DNA binding domain of microphthalmia-associated transcription factor (MITF), four mutations are reported: mi, Mi wh, mi ew, and mi or. MITFs encoded by the mi, Mi wh, mi ew, and Mi or mutant alleles (mi-MITF, Mi wh-MITF, Mi ew-MITF, and Mi or-MITF, respectively) interfered with the DNA binding of wild-type MITF, TFE3, and another basic helix-loop-helix leucine zipper protein in vitro. Polyclonal antibody against MITF was produced and used for investigating the subcellular localization of mutant MITFs. Immunocytochemistry and immunoblotting revealed that more than 99% of wild-type MITF and Mi wh-MITF located in nuclei of transfected NIH 3T3 and 293T cells. In contrast, mi-MITF predominantly located in the cytoplasm of cells transfected with the corresponding plasmid. When the immunoglobulin G (IgG)-conjugated peptides representing a part of the DNA binding domain containing mi and Mi wh mutations were microinjected into the cytoplasm of NRK49F cells, wild-type peptide and Mi wh-type peptide-IgG conjugate localized in nuclei but mi-type peptide-IgG conjugate was detectable only in the cytoplasm. It was also demonstrated that the nuclear translocation potential of Mi or-MITF was normal but that Mi ew-MITF was impaired as well as mi-MITF. In cotransfection assay, a strong dominant negative effect of Mi wh-MITF against wild-type MITF-dependent transactivation system on tyrosinase promoter was observed, but mi-MITF had a small effect. However, by the conjugation of simian virus 40 large-T-antigen-derived nuclear localization signal to mi-MITF, the dominant negative effect was enhanced. Furthermore, we demonstrated that the interaction between wild-type MITF and mi-MITF occurred in the cytoplasm and that mi-MITF had an inhibitory effect on nuclear localization potential of wild-type MITF.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Unión al ADN/genética , Células 3T3 , Secuencia de Aminoácidos , Animales , Proteínas de Unión al ADN/metabolismo , Leucina Zippers , Ratones , Ratones Mutantes , Factor de Transcripción Asociado a Microftalmía , Datos de Secuencia Molecular , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Eur J Immunol ; 24(12): 2966-73, 1994 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7805723

RESUMEN

T cell tolerance is usually established by clonal deletion of self-specific T cells in the thymus, or some times, in the periphery. Alternatively, tolerance may also be achieved by induction of clonal T cell unresponsiveness by a poorly understood mechanism called "anergy". We found that transgenic mice expressing a soluble form of vesicular stomatitis virus (VSV) glycoprotein (G) predominantly in liver and kidney exhibited normal B cell responses. VSV-G-specific T help-independent neutralizing IgM responses were within normal ranges, but no T help-dependent neutralizing IgG antibodies were generated upon immunization with recombinant VSV-G protein and recombinant vaccinia virus expressing VSV-G. This demonstrated absence of B cell tolerance but presence of T helper cell unresponsiveness. After adoptive transfer of transgenic spleen cells into thymectomized immuno-incompetent hosts, the unresponsive T helper cells regained function and switched the neutralizing IgM response to IgG, comparably to control T helper cells, within 7 days. Conversely, when naive non-transgenic spleen cells were transferred into transgenic mice, VSV-G-specific T helper cells became unresponsive within 3-4 days. These results suggest that VSV-G-specific T helper cells are rendered unresponsive within a few days in the VSV-G transgenic host also outside of the thymus and that this unresponsiveness was reversed by transfer into antigen-free recipients.


Asunto(s)
Tolerancia Inmunológica , Glicoproteínas de Membrana , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos B/inmunología , Secuencia de Bases , Cartilla de ADN/química , Inmunización Pasiva , Cooperación Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
18.
Nat Genet ; 8(3): 256-63, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7874168

RESUMEN

Mutations in the mouse microphthalmia (mi) gene affect the development of a number of cell types including melanocytes, osteoclasts and mast cells. Recently, mutations in the human mi gene (MITF) were found in patients with Waardenburg Syndrome type 2 (WS2), a dominantly inherited syndrome associated with hearing loss and pigmentary disturbances. We have characterized the molecular defects associated with eight murine mi mutations, which vary in both their mode of inheritance and in the cell types they affect. These molecular data, combined with the extensive body of genetic data accumulated for murine mi, shed light on the phenotypic and developmental consequences of mi mutations and offer a mouse model for WS2.


Asunto(s)
Alelos , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica , Microftalmía/genética , Mutación , Factores de Transcripción , Síndrome de Waardenburg , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas de Unión al ADN/química , Sordera/genética , Color del Ojo/genética , Genes Dominantes , Color del Cabello/genética , Secuencias Hélice-Asa-Hélice , Humanos , Leucina Zippers , Mastocitos/patología , Melanocitos/patología , Ratones , Ratones Endogámicos , Ratones Mutantes , Factor de Transcripción Asociado a Microftalmía , Modelos Moleculares , Datos de Secuencia Molecular , Cresta Neural/patología , Osteopetrosis/genética , Fenotipo , Conformación Proteica , Empalme del ARN , Anomalías Dentarias/genética , Síndrome de Waardenburg/clasificación , Síndrome de Waardenburg/genética
19.
Genes Dev ; 8(22): 2770-80, 1994 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-7958932

RESUMEN

The microphthalmia (mi) gene appears essential for pigment cell development and/or survival, based on its mutation in mi mice. It has also been linked to the human disorder Waardenburg Syndrome. The mi gene was recently cloned and predicts a basic/helix-loop-helix/leucine zipper (b-HLH-ZIP) factor with tissue-restricted expression. Here, we show that Mi protein binds DNA as a homo- or heterodimer with TFEB, TFE3, or TFEC, together constituting a new MiT family. Mi can also activate transcription through recognition of the M box, a highly conserved pigmentation gene promoter element, and may thereby determine tissue-specific expression of pigmentation enzymes. Six mi mutations shown recently to cluster in the b-HLH-ZIP region produce surprising and instructive effects on DNA recognition and oligomerization. An alternatively spliced exon located outside of the b-HLH-ZIP region is shown to significantly modulate DNA recognition by the basic domain. These findings suggest that Mi's critical roles in melanocyte survival and pigmentation are mediated by MiT family interactions and transcriptional activities.


Asunto(s)
Proteínas de Unión al ADN/farmacología , Melanocitos/fisiología , Factores de Transcripción , Empalme Alternativo , Animales , Secuencia de Bases , Regulación de la Expresión Génica , Genes Reporteros , Ratones , Factor de Transcripción Asociado a Microftalmía , Datos de Secuencia Molecular , Mutación
20.
J Neurochem ; 63(3): 822-33, 1994 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7519661

RESUMEN

Mutations in the myelin proteolipid protein (PLP) gene, such as that found in the jimpy mouse, result in an abnormal structure of the myelin, severe dysmyelination, and a reduction in the number of mature oligodendrocytes. To examine the functions of the two alternatively spliced isoforms of proteolipid protein, transgenic mice were generated that express either PLP or DM20 cDNAs placed under control of the PLP upstream regulatory region. The transgenes were bred into jimpy mice, and the effect of the transgenes on the dysmyelinating phenotype was analyzed. Neither the PLP transgene nor the DM20 transgene alone had an effect on myelination in the jimpy mice. Combining the two transgenes substantially increased the number of myelinated axons, suggesting that the two alternatively spliced products of the PLP locus perform distinct functions in oligodendrocytes. The enhanced myelination was not sufficient, however, for completely correcting the dysmyelinating phenotype of the jimpy mice, nor was it accompanied by the restoration of normal levels of myelin gene expression. The inability to rescue the jimpy phenotype is most likely attributable to a dominant negative action of the abnormal proteolipid proteins present in jimpy mice. These results demonstrate the complexity of proteolipid protein function in myelination.


Asunto(s)
Empalme Alternativo , Proteínas de la Mielina/genética , Vaina de Mielina/fisiología , Animales , Secuencia de Bases , Northern Blotting , Expresión Génica , Ratones , Ratones Jimpy , Ratones Transgénicos , Microscopía Electrónica , Datos de Secuencia Molecular , Mutación , Proteínas de la Mielina/fisiología , Proteína Proteolipídica de la Mielina , Vaina de Mielina/ultraestructura , Fenotipo , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA