Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Hum Mol Genet ; 30(24): 2456-2468, 2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34296265

RESUMO

The rare, fatal neurodegenerative disorder Niemann-Pick disease type C1 (NPC1) arises from lysosomal accumulation of unesterified cholesterol and glycosphingolipids. These subcellular pathologies lead to phenotypes of hepatosplenomegaly, neurological degeneration and premature death. The timing and severity of NPC1 clinical presentation is extremely heterogeneous. This study analyzed RNA-Seq data from 42 NPC1 patient-derived, primary fibroblast cell lines to determine transcriptional changes induced by treatment with 2-hydroxypropyl-ß-cyclodextrin (HPßCD), a compound currently under investigation in clinical trials. A total of 485 HPßCD-responsive genes were identified. Pathway enrichment analysis of these genes showed significant involvement in cholesterol and lipid biosynthesis. Furthermore, immunohistochemistry of the cerebellum as well as measurements of plasma from Npc1m1N null mice treated with HPßCD and adeno-associated virus gene therapy suggests that one of the identified genes, GPNMB, may serve as a useful biomarker of treatment response in NPC1 disease. Overall, this large NPC1 patient-derived dataset provides a comprehensive foundation for understanding the genomic response to HPßCD treatment.


Assuntos
Doença de Niemann-Pick Tipo C , 2-Hidroxipropil-beta-Ciclodextrina , Animais , Biomarcadores , Modelos Animais de Doenças , Proteínas do Olho/genética , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Doença de Niemann-Pick Tipo C/tratamento farmacológico , Doença de Niemann-Pick Tipo C/genética , Doença de Niemann-Pick Tipo C/patologia , Transcriptoma
2.
PLoS Biol ; 16(5): e2003648, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29723194

RESUMO

Melanocyte stem cells (McSCs) and mouse models of hair graying serve as useful systems to uncover mechanisms involved in stem cell self-renewal and the maintenance of regenerating tissues. Interested in assessing genetic variants that influence McSC maintenance, we found previously that heterozygosity for the melanogenesis associated transcription factor, Mitf, exacerbates McSC differentiation and hair graying in mice that are predisposed for this phenotype. Based on transcriptome and molecular analyses of Mitfmi-vga9/+ mice, we report a novel role for MITF in the regulation of systemic innate immune gene expression. We also demonstrate that the viral mimic poly(I:C) is sufficient to expose genetic susceptibility to hair graying. These observations point to a critical suppressor of innate immunity, the consequences of innate immune dysregulation on pigmentation, both of which may have implications in the autoimmune, depigmenting disease, vitiligo.


Assuntos
Células-Tronco Adultas , Cor de Cabelo/imunologia , Imunidade Inata , Melanócitos , Fator de Transcrição Associado à Microftalmia/fisiologia , Animais , Feminino , Regulação da Expressão Gênica , Predisposição Genética para Doença , Cor de Cabelo/genética , Interferon Tipo I/metabolismo , Camundongos , Camundongos Transgênicos , Poli I-C
3.
Hum Mol Genet ; 26(1): 52-64, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27798114

RESUMO

Niemann-Pick disease, type C1 (NPC1) is a heritable lysosomal storage disease characterized by a progressive neurological degeneration that causes disability and premature death. A murine model of NPC1 disease (Npc1-/-) displays a rapidly progressing form of NPC1 disease which is characterized by weight loss, ataxia, increased cholesterol storage, loss of cerebellar Purkinje neurons and early lethality. To test the potential efficacy of gene therapy for NPC1, we constructed adeno-associated virus serotype 9 (AAV9) vectors to deliver the NPC1 gene under the transcriptional control of the neuronal-specific (CamKII) or a ubiquitous (EF1a) promoter. The Npc1-/- mice that received a single dose of AAV9-CamKII-NPC1 as neonates (2.6 × 1011GC) or at weaning (1.3 × 1012GC), and the mice that received a single dose of AAV9-EF1a-NPC1 at weaning (1.2 × 1012GC), exhibited an increased life span, characterized by delayed weight loss and diminished motor decline. Cholesterol storage and Purkinje neuron loss were also reduced in the central nervous system of AAV9 treated Npc1-/- mice. Treatment with AAV9-EF1a-NPC1, as compared to AAV9-CamKII-NPC1, resulted in significantly increased survival (mean survival increased from 69 days to 166 and 97 days, respectively) and growth, and reduced hepatic-cholesterol accumulation. Our results provide the first demonstration that gene therapy may represent a therapeutic option for NPC1 patients and suggest that extraneuronal NPC1 expression can further augment the lifespan of the Npc1-/- mice after systemic AAV gene delivery.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos/administração & dosagem , Longevidade/genética , Doença de Niemann-Pick Tipo C/terapia , Proteínas/genética , Animais , Cerebelo/metabolismo , Cerebelo/patologia , Colesterol/metabolismo , Modelos Animais de Doenças , Feminino , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Proteína C1 de Niemann-Pick , Doença de Niemann-Pick Tipo C/genética , Células de Purkinje/metabolismo , Células de Purkinje/patologia
4.
Pigment Cell Melanoma Res ; 37(2): 259-264, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37874775

RESUMO

MFSD12 functions as a transmembrane protein required for import of cysteine into melanosomes and lysosomes. The MFSD12 locus has been associated with phenotypic variation in skin color across African, Latin American, and East Asian populations. The frequency of a particular MFSD12 coding variant, rs2240751 (MAF = 0.08), has been reported to correlate with solar radiation and occur at highest frequency in Peruvian (PEL MAF = 0.48) and Han Chinese (CHB MAF = 0.40) populations, suggesting it could be causative for associated phenotypic variation in skin color. We have generated a mouse knock-in allele, Mfsd12Y182H , to model the human missense p.Tyr182His human variant. We demonstrate that the variant transcript is stably expressed and that agouti mice homozygote for the variant allele are viable with an altered coat color. This in vivo data confirms that the MFSD12 p.Tyr182His variant functions as a hypomorphic allele sufficient to alter mammalian pigmentation.


Assuntos
Proteínas de Membrana , Pigmentação da Pele , Animais , Camundongos , Proteína Agouti Sinalizadora/genética , Alelos , Cor de Cabelo/genética , Homozigoto , Proteínas de Membrana/genética , Mutação de Sentido Incorreto/genética , Pigmentação da Pele/genética
5.
Nat Genet ; 34(1): 108-12, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12704387

RESUMO

To gain insight into melanoma pathogenesis, we characterized an insertional mouse mutant, TG3, that is predisposed to develop multiple melanomas. Physical mapping identified multiple tandem insertions of the transgene into intron 3 of Grm1 (encoding metabotropic glutamate receptor 1) with concomitant deletion of 70 kb of intronic sequence. To assess whether this insertional mutagenesis event results in alteration of transcriptional regulation, we analyzed Grm1 and two flanking genes for aberrant expression in melanomas from TG3 mice. We observed aberrant expression of only Grm1. Although we did not detect its expression in normal mouse melanocytes, Grm1 was ectopically expressed in the melanomas from TG3 mice. To confirm the involvement of Grm1 in melanocytic neoplasia, we created an additional transgenic line with Grm1 expression driven by the dopachrome tautomerase promoter. Similar to the original TG3, the Tg(Grm1)EPv line was susceptible to melanoma. In contrast to human melanoma, these transgenic mice had a generalized hyperproliferation of melanocytes with limited transformation to fully malignant metastasis. We detected expression of GRM1 in a number of human melanoma biopsies and cell lines but not in benign nevi and melanocytes. This study provides compelling evidence for the importance of metabotropic glutamate signaling in melanocytic neoplasia.


Assuntos
Melanoma/genética , Melanoma/metabolismo , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Animais , DNA Complementar/genética , DNA de Neoplasias/genética , Regulação Neoplásica da Expressão Gênica , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Mutagênese Insercional , Transdução de Sinais , Neoplasias Cutâneas/patologia , Transfecção
6.
PLoS Genet ; 4(9): e1000177, 2008 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-18773073

RESUMO

The neural crest (NC) is a population of embryonic stem cells that gives rise to numerous cell types, including the glia and neurons of the peripheral and enteric nervous systems and the melanocytes of the skin and hair. Mutations in genes and genetic pathways regulating NC development lead to a wide spectrum of human developmental disorders collectively called neurocristopathies. To identify molecular pathways regulating NC development and to understand how alterations in these processes lead to disease, we established an N-ethyl-N-nitrosourea (ENU) mutagenesis screen utilizing a mouse model sensitized for NC defects, Sox10(LacZ/+). Out of 71 pedigrees analyzed, we identified and mapped four heritable loci, called modifier of Sox10 expression pattern 1-4 (msp1-4), which show altered NC patterning. In homozygous msp1 embryos, Sox10(LacZ) expression is absent in cranial ganglia, cranial nerves, and the sympathetic chain; however, the development of other Sox10-expressing cells appears unaffected by the mutation. Linkage analysis, sequencing, and complementation testing confirmed that msp1 is a new allele of the receptor tyrosine kinase Erbb3, Erbb3(msp1), that carries a single amino acid substitution in the extracellular region of the protein. The ENU-induced mutation does not alter protein expression, however, it is sufficient to impair ERBB3 signaling such that the embryonic defects observed in msp1 resemble those of Erbb3 null alleles. Biochemical analysis of the mutant protein showed that ERBB3 is expressed on the cell surface, but its ligand-induced phosphorylation is dramatically reduced by the msp1 mutation. These findings highlight the importance of the mutated residue for ERBB3 receptor function and activation. This study underscores the utility of using an ENU mutagenesis to identify genetic pathways regulating NC development and to dissect the roles of discrete protein domains, both of which contribute to a better understanding of gene function in a cellular and developmental setting.


Assuntos
Substituição de Aminoácidos , Proteínas de Ligação a DNA/genética , Proteínas de Grupo de Alta Mobilidade/genética , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo , Fatores de Transcrição/genética , Alelos , Sequência de Aminoácidos , Aminoácidos/genética , Aminoácidos/metabolismo , Animais , Sequência de Bases , Proteínas de Ligação a DNA/metabolismo , Etilnitrosoureia/farmacologia , Expressão Gênica , Proteínas de Grupo de Alta Mobilidade/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutação , Crista Neural/citologia , Crista Neural/embriologia , Crista Neural/metabolismo , Fenótipo , Fosforilação , RNA Mensageiro/metabolismo , Fatores de Transcrição SOXE , Fatores de Transcrição/metabolismo
7.
Life Sci Alliance ; 4(10)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34407999

RESUMO

Niemann-Pick C1 disease (NPC1) is a rare, fatal neurodegenerative disease caused by mutations in NPC1, which encodes the lysosomal cholesterol transport protein NPC1. Disease pathology involves lysosomal accumulation of cholesterol and lipids, leading to neurological and visceral complications. Targeting the central nervous system (CNS) from systemic circulation complicates treatment of neurological diseases with gene transfer techniques. Selected and engineered capsids, for example, adeno-associated virus (AAV)-PHP.B facilitate peripheral-to-CNS transfer and hence greater CNS transduction than parental predecessors. We report that systemic delivery to Npc1 m1N/m1N mice using an AAV-PHP.B vector ubiquitously expressing NPC1 led to greater disease amelioration than an otherwise identical AAV9 vector. In addition, viral copy number and biodistribution of GFP-expressing reporters showed that AAV-PHP.B achieved more efficient, albeit variable, CNS transduction than AAV9 in Npc1 m1N/m1N mice. This variability was associated with segregation of two alleles of the putative AAV-PHP.B receptor Ly6a in Npc1 m1N/m1N mice. Our data suggest that robust improvements in NPC1 disease phenotypes occur even with modest CNS transduction and that improved neurotrophic capsids have the potential for superior NPC1 AAV gene therapy vectors.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Doença de Niemann-Pick Tipo C/genética , Doença de Niemann-Pick Tipo C/terapia , Transdução Genética , Animais , Modelos Animais de Doenças , Feminino , Expressão Gênica , Genes Reporter , Vetores Genéticos/administração & dosagem , Masculino , Camundongos , Camundongos Transgênicos , Proteína C1 de Niemann-Pick/genética , Fenótipo , Distribuição Tecidual , Transgenes , Resultado do Tratamento
8.
Hum Mol Genet ; 17(14): 2118-31, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18397875

RESUMO

Haploinsufficiency for the transcription factor SOX10 is associated with the pigmentary deficiencies of Waardenburg syndrome (WS) and is modeled in Sox10 haploinsufficient mice (Sox10(LacZ/+)). As genetic background affects WS severity in both humans and mice, we established an N-ethyl-N-nitrosourea (ENU) mutagenesis screen to identify modifiers that increase the phenotypic severity of Sox10(LacZ/+) mice. Analysis of 230 pedigrees identified three modifiers, named modifier of Sox10 neurocristopathies (Mos1, Mos2 and Mos3). Linkage analysis confirmed their locations on mouse chromosomes 13, 4 and 3, respectively, within regions distinct from previously identified WS loci. Positional candidate analysis of Mos1 identified a truncation mutation in a hedgehog(HH)-signaling mediator, GLI-Kruppel family member 3 (Gli3). Complementation tests using a second allele of Gli3 (Gli3(Xt-J)) confirmed that a null mutation of Gli3 causes the increased hypopigmentation in Sox10(LacZ/+);Gli3(Mos1/)(+) double heterozygotes. Early melanoblast markers (Mitf, Sox10, Dct, and Si) are reduced in Gli3(Mos1/)(Mos1) embryos, indicating that loss of GLI3 signaling disrupts melanoblast specification. In contrast, mice expressing only the GLI3 repressor have normal melanoblast specification, indicating that the full-length GLI3 activator is not required for specification of neural crest to the melanocyte lineage. This study demonstrates the feasibility of sensitized screens to identify disease modifier loci and implicates GLI3 and other HH signaling components as modifiers of human neurocristopathies.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Grupo de Alta Mobilidade/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Mutagênese , Proteínas do Tecido Nervoso/metabolismo , Crista Neural/fisiopatologia , Fatores de Transcrição/metabolismo , Síndrome de Waardenburg/metabolismo , Síndrome de Waardenburg/fisiopatologia , Animais , Sequência de Bases , Diferenciação Celular , Mapeamento Cromossômico , Códon sem Sentido , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/fisiopatologia , Etilnitrosoureia/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Teste de Complementação Genética , Proteínas de Grupo de Alta Mobilidade/genética , Humanos , Fatores de Transcrição Kruppel-Like/genética , Masculino , Melanócitos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutagênicos/farmacologia , Proteínas do Tecido Nervoso/genética , Crista Neural/efeitos dos fármacos , Crista Neural/embriologia , Crista Neural/metabolismo , Pigmentação , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição SOXE , Fatores de Transcrição/genética , Síndrome de Waardenburg/embriologia , Síndrome de Waardenburg/genética , Proteína Gli3 com Dedos de Zinco
9.
Dis Model Mech ; 13(3)2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-31996359

RESUMO

Niemann-Pick disease type C1 (NPC1) is a rare, fatal neurodegenerative disorder characterized by lysosomal accumulation of unesterified cholesterol and glycosphingolipids. These subcellular pathologies lead to phenotypes of hepatosplenomegaly, neurological degeneration and premature death. NPC1 is extremely heterogeneous in the timing of clinical presentation and is associated with a wide spectrum of causative NPC1 mutations. To study the genetic architecture of NPC1, we have generated a new NPC1 mouse model, Npc1em1PavNpc1em1Pav/em1Pav mutants showed notably reduced NPC1 protein compared to controls and displayed the pathological and biochemical hallmarks of NPC1. Interestingly, Npc1em1Pav/em1Pav mutants on a C57BL/6J genetic background showed more severe visceral pathology and a significantly shorter lifespan compared to Npc1em1Pav/em1Pav mutants on a BALB/cJ background, suggesting that strain-specific modifiers contribute to disease severity and survival. QTL analysis for lifespan of 202 backcross N2 mutants on a mixed C57BL/6J and BALB/cJ background detected significant linkage to markers on chromosomes 1 and 7. The discovery of these modifier regions demonstrates that mouse models are powerful tools for analyzing the genetics underlying rare human diseases, which can be used to improve understanding of the variability in NPC1 phenotypes and advance options for patient diagnosis and therapy.This article has an associated First Person interview with the first author of the paper.


Assuntos
Patrimônio Genético , Longevidade , Doença de Niemann-Pick Tipo C/patologia , Índice de Gravidade de Doença , Alelos , Animais , Sequência de Bases , Cromossomos de Mamíferos/genética , Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lisossomos/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Degeneração Neural/patologia , Proteína C1 de Niemann-Pick , Fenótipo , Locos de Características Quantitativas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sobrevida , Vísceras/patologia , Redução de Peso
10.
PLoS Genet ; 1(3): e38, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16184191

RESUMO

Pancreatic insufficiency (PI) when left untreated results in a state of malnutrition due to an inability to absorb nutrients. Frequently, PI is diagnosed as part of a larger clinical presentation in cystic fibrosis or Shwachman-Diamond syndrome. In this study, a mouse model for isolated exocrine PI was identified in a mouse line generated by a transgene insertion. The trait is inherited in an autosomal recessive pattern, and homozygous animals are growth retarded, have abnormal immunity, and have reduced life span. Mice with the disease locus, named pequeño (pq), exhibit progressive apoptosis of pancreatic acinar cells with severe exocrine acinar cell loss by 8 wk of age, while the islets and ductal tissue persist. The mutation in pq/pq mice results from a random transgene insertion. Molecular characterization of the transgene insertion site by fluorescent in situ hybridization and genomic deletion mapping identified an approximately 210-kb deletion on Chromosome 3, deleting two genes. One of these genes, Serpini2, encodes a protein that is a member of the serpin family of protease inhibitors. Reintroduction of only the Serpini2 gene by bacterial artificial chromosome transgenic complementation corrected the acinar cell defect as well as body weight and immune phenotypes, showing that deletion of Serpini2 causes the pequeño phenotype. Dietary supplementation of pancreatic enzymes also corrected body size, body weight, and immunodeficiency, and increased the life span of Serpini2-deficient mice, despite continued acinar cell loss. To our knowledge, this study describes the first characterized genetic animal model for isolated PI. Genetic complementation of the transgene insertion mutant demonstrates that Serpini2 deficiency directly results in the acinar cell apoptosis, malabsorption, and malnutrition observed in pq/pq mice. The rescue of growth retardation, immunodeficiency, and mortality by either Serpini2 bacterial artificial chromosome transgenic expression or by pancreatic enzyme supplementation demonstrates that these phenotypes are secondary to malnutrition in pq/pq mice.


Assuntos
Apoptose/genética , Pancreatopatias/genética , Serpinas/deficiência , Serpinas/genética , Animais , Transtornos do Crescimento/genética , Hibridização in Situ Fluorescente , Desnutrição/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Regiões Promotoras Genéticas
11.
Mech Dev ; 123(2): 124-34, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16412618

RESUMO

Mutations in the transcription factor Sox10 or Endothelin Receptor B (Ednrb) result in Waardenburg Syndrome Type IV (WS-IV), which presents with deficiencies of neural crest derived melanocytes (hypopigmentation) and enteric ganglia (hypoganglionosis). As Sox10 and Ednrb are expressed in mouse migratory neural crest cells and melanoblasts, we investigated the possibility that SOX10 and EDNRB function through a hierarchical relationship during melanocyte development. However, our results support a distinct rather than hierarchical relationship. First, SOX10 expression continues in Ednrb null melanoblasts, demonstrating that SOX10 expression is not dependent on EDNRB function. Second, Ednrb expression persists in E10.5 Sox10null embryos, demonstrating that Ednrb is not dependent on SOX10 for expression in migratory neural crest cells. Third, over-expression of SOX10 in melanoblasts of mice that harbor null or hypomorphic Ednrb alleles does not rescue hypopigmentation, suggesting that SOX10 overexpression can neither complement a lack of EDNRB function nor increase Ednrb expression. Fourth, mice that are double heterozygous for loss-of-function mutations in Sox10 and Ednrb do not demonstrate synergistically increased hypopigmentation compared to mice that are single heterozygotes for either mutation alone, suggesting a lack of direct genetic interaction between these genes. Our results suggest that SOX10 does not directly activate Ednrb transcription in the melanocyte lineage. Given that SOX10 directly activates Ednrb in the enteric nervous system, our results suggest that SOX10 may differentially activate target genes based on the particular cellular context.


Assuntos
Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Grupo de Alta Mobilidade/metabolismo , Melanócitos/fisiologia , Proteínas de Neoplasias/metabolismo , Crista Neural/embriologia , Receptor de Endotelina B/metabolismo , Fatores de Transcrição/metabolismo , Animais , Heterozigoto , Proteínas de Grupo de Alta Mobilidade/genética , Hipopigmentação/genética , Hipopigmentação/metabolismo , Oxirredutases Intramoleculares/genética , Melanócitos/citologia , Camundongos , Camundongos Mutantes , Mutação , Proteínas de Neoplasias/genética , Crista Neural/citologia , Crista Neural/metabolismo , Regiões Promotoras Genéticas , Receptor de Endotelina B/genética , Fatores de Transcrição SOXE , Fatores de Transcrição/genética
12.
Pigment Cell Res ; 18(3): 167-80, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15892713

RESUMO

Summary WNT1 and WNT3a have been described as having redundant roles in promoting the development of neural crest-derived melanocytes (NC-Ms). We used cell lineage restricted retroviral infections to examine the effects of WNT signaling on defined cell types in neural crest cultures. RCAS retroviral infections were targeted to melanoblasts (NC-M precursor cells) derived from transgenic mice that express the virus receptor, TVA, under the control of a melanoblast promoter (DCT). As expected, over 90% of DCT-TVA+ cells expressed early melanoblast markers MITF and KIT. However, by following the fate of infected cells in standard culture conditions, we find that only 5% of descendents were NC-Ms. The majority of the descendents were not NC-Ms, but expressed smooth muscle cell markers, demonstrating that mammalian melanoblasts are not committed to the NC-M lineage. RCAS infection of DCT-TVA+ cells demonstrated that overexpression of canonical WNT signaling genes (betaCAT, WNT3a or WNT1) can increase NC-M numbers in an endothelin dependent manner. However, WNT1 and WNT3a have different modes of action with respect to melanoblast fate. Intrinsic over-expression of betaCAT or WNT3a can increase NC-M numbers by biasing the fate of DCT-TVA+ cells to NC-Ms. In contrast, the DCT-TVA+ melanoblasts cannot respond to WNT1 signaling and do not alter their fate towards NC-M. Instead, WNT1 only increases NC-M numbers through paracrine signaling on melanoblast precursors to increase the numbers of neural crest cells that become NC-Ms.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Melanócitos/fisiologia , Crista Neural/citologia , Proteínas/fisiologia , Animais , Antígenos de Diferenciação/biossíntese , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intercelular/genética , Melanócitos/citologia , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Músculo Liso/citologia , Músculo Liso/fisiologia , Crista Neural/embriologia , Regiões Promotoras Genéticas/genética , Proteínas/genética , Proteínas Proto-Oncogênicas/fisiologia , Transdução de Sinais/fisiologia , Proteínas Wnt , Proteína Wnt-5a , Proteína Wnt1 , Proteína Wnt3 , Proteína Wnt3A
13.
Dev Dyn ; 229(1): 54-62, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14699577

RESUMO

An in vitro gene complementation approach has been developed to dissect gene function and regulation in neural crest (NC) development and disease. The approach uses the avian RCAS virus to express genes in NC cells derived from transgenic mice expressing the RCAS receptor TVA, under the control of defined promoter elements. Constructs for creating TVA transgenic mice were developed using site-specific recombination GATEWAY (GW), compatible vectors that can also be used to facilitate analysis of genomic fragments for transcriptional regulatory elements. By using these GW vectors to facilitate cloning, transgenic mouse lines were generated that express TVA in SOX10-expressing NC stem cells under the control of the Pax3 promoter. The Pax3-tv-a transgene was bred onto a Sox10-deficient background, and the feasibility of complementing genetic NC defects was demonstrated by infecting the Pax3-tv-a cells with an RCAS-Sox10 expression virus, thereby rescuing melanocyte development of Sox10-deficient NC cells. This system will be useful for assessing genetic hierarchies in NC development. Developmental Dynamics 229:54-62, 2004.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Grupo de Alta Mobilidade/genética , Melanócitos/citologia , Melanócitos/metabolismo , Crista Neural/citologia , Crista Neural/metabolismo , Fatores de Transcrição , Animais , Proteínas Aviárias , Sequência de Bases , Diferenciação Celular , DNA/genética , Proteínas de Ligação a DNA/deficiência , Técnicas de Transferência de Genes , Teste de Complementação Genética , Vetores Genéticos , Proteínas de Grupo de Alta Mobilidade/deficiência , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados , Regiões Promotoras Genéticas , Receptores Virais/genética , Fatores de Transcrição SOXE
14.
Hum Mol Genet ; 11(24): 3107-14, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12417532

RESUMO

Niemann-Pick disease type C (NPC) is a neurodegenerative disorder with major visceral complications, including liver disease that can be fatal before onset of neurodegeneration. We have sought to determine the extent to which visceral disease contributes to neurodegeneration by making transgenic mice in which the wild-type NPC1 protein is expressed primarily in the CNS using the prion promoter. When the transgene was introduced into the npc1(-/-) animals neurodegeneration was prevented, a 'normal' lifespan occurred and the sterility of npc1(-/-) mice was corrected. The rescue did not provide complete neurological correction in the CNS as GM2 and GM3 gangliosides were observed to accumulate in some neurons and glia of transgenic animals. Two of three transgenic lines demonstrated some low-level ectopic expression resulting in correction of visceral phenotypes in liver and spleen. Interestingly, the third transgenic line continued to have moderate histocytosis in liver and spleen, yet had no detectable neurodegeneration. Thus, it is primarily the lack of NPC1 in the CNS and not the secondary effects of the visceral involvement that causes the neurological decline in NPC disease. In addition, the expression levels of NPC1 found in the CNS of transgenic animals were much greater than in normal littermates, demonstrating that overexpression of NPC1 is not harmful and allowing possibilities for genetic therapy interventions that utilize overexpression.


Assuntos
Terapia Genética , Doenças de Niemann-Pick/genética , Doenças de Niemann-Pick/terapia , Proteínas/genética , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Emaciação/genética , Imuno-Histoquímica , Infertilidade/genética , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Proteína C1 de Niemann-Pick , Especificidade de Órgãos , Príons/genética , Regiões Promotoras Genéticas , Proteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA