Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Hum Mol Genet ; 24(19): 5512-23, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26199316

RESUMO

Environmental factors and susceptible genomes interact to determine the risk of neurodevelopmental disorders. Although few genes and environmental factors have been linked, the intervening cellular and molecular mechanisms connecting a disorder susceptibility gene with environmental factors remain mostly unexplored. Here we focus on the schizophrenia susceptibility gene DTNBP1 and its product dysbindin, a subunit of the BLOC-1 complex, and describe a neuronal pathway modulating copper metabolism via ATP7A. Mutations in ATP7A result in Menkes disease, a disorder of copper metabolism. Dysbindin/BLOC-1 and ATP7A genetically and biochemically interact. Furthermore, disruption of this pathway causes alteration in the transcriptional profile of copper-regulatory and dependent factors in the hippocampus of dysbindin/BLOC-1-null mice. Dysbindin/BLOC-1 loss-of-function alleles do not affect cell and tissue copper content, yet they alter the susceptibility to toxic copper challenges in both mammalian cells and Drosophila. Our results demonstrate that perturbations downstream of the schizophrenia susceptibility gene DTNBP1 confer susceptibility to copper, a metal that in excess is a neurotoxin and whose depletion constitutes a micronutrient deficiency.


Assuntos
Cobre/metabolismo , Proteínas de Drosophila/genética , Proteínas Associadas à Distrofina/genética , Esquizofrenia/genética , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Células Cultivadas , ATPases Transportadoras de Cobre , Modelos Animais de Doenças , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Disbindina , Proteínas Associadas à Distrofina/metabolismo , Predisposição Genética para Doença , Hipocampo/metabolismo , Camundongos , Neurônios/metabolismo
2.
J Neurosci ; 35(19): 7643-53, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25972187

RESUMO

Dysbindin is a schizophrenia susceptibility factor and subunit of the biogenesis of lysosome-related organelles complex 1 (BLOC-1) required for lysosome-related organelle biogenesis, and in neurons, synaptic vesicle assembly, neurotransmission, and plasticity. Protein networks, or interactomes, downstream of dysbindin/BLOC-1 remain partially explored despite their potential to illuminate neurodevelopmental disorder mechanisms. Here, we conducted a proteome-wide search for polypeptides whose cellular content is sensitive to dysbindin/BLOC-1 loss of function. We identified components of the vesicle fusion machinery as factors downregulated in dysbindin/BLOC-1 deficiency in neuroectodermal cells and iPSC-derived human neurons, among them the N-ethylmaleimide-sensitive factor (NSF). Human dysbindin/BLOC-1 coprecipitates with NSF and vice versa, and both proteins colocalized in a Drosophila model synapse. To test the hypothesis that NSF and dysbindin/BLOC-1 participate in a pathway-regulating synaptic function, we examined the role for NSF in dysbindin/BLOC-1-dependent synaptic homeostatic plasticity in Drosophila. As previously described, we found that mutations in dysbindin precluded homeostatic synaptic plasticity elicited by acute blockage of postsynaptic receptors. This dysbindin mutant phenotype is fully rescued by presynaptic expression of either dysbindin or Drosophila NSF. However, neither reduction of NSF alone or in combination with dysbindin haploinsufficiency impaired homeostatic synaptic plasticity. Our results demonstrate that dysbindin/BLOC-1 expression defects result in altered cellular content of proteins of the vesicle fusion apparatus and therefore influence synaptic plasticity.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas Associadas à Distrofina/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas Sensíveis a N-Etilmaleimida/metabolismo , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Animais , Animais Geneticamente Modificados , Linhagem Celular Tumoral , Drosophila , Proteínas de Drosophila/genética , Disbindina , Proteínas Associadas à Distrofina/genética , Regulação da Expressão Gênica/genética , Humanos , Melanoma/patologia , Proteínas Sensíveis a N-Etilmaleimida/genética , Rede Nervosa/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroblastoma/patologia , Junção Neuromuscular/genética , Junção Neuromuscular/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/genética , Proteínas SNARE/metabolismo , Sinapses/genética , Vesículas Sinápticas/genética , Vesículas Sinápticas/metabolismo
3.
J Biol Chem ; 289(20): 14291-300, 2014 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-24713699

RESUMO

Post-mortem analysis has revealed reduced levels of the protein dysbindin in the brains of those suffering from the neurodevelopmental disorder schizophrenia. Consequently, mechanisms controlling the cellular levels of dysbindin and its interacting partners may participate in neurodevelopmental processes impaired in that disorder. To address this question, we studied loss of function mutations in the genes encoding dysbindin and its interacting BLOC-1 subunits. We focused on BLOC-1 mutants affecting synapse composition and function in addition to their established systemic pigmentation, hematological, and lung phenotypes. We tested phenotypic homogeneity and gene dosage effects in the mouse null alleles muted (Bloc1s5(mu/mu)) and dysbindin (Bloc1s8(sdy/sdy)). Transcripts of NMDA receptor subunits and GABAergic interneuron markers, as well as expression of BLOC-1 subunit gene products, were affected differently in the brains of Bloc1s5(mu/mu) and Bloc1s8(sdy/sdy) mice. Unlike Bloc1s8(sdy/sdy), elimination of one or two copies of Bloc1s5 generated indistinguishable pallidin transcript phenotypes. We conclude that monogenic mutations abrogating the expression of a protein complex subunit differentially affect the expression of other complex transcripts and polypeptides as well as their downstream effectors. We propose that the genetic disruption of different subunits of protein complexes and combinations thereof diversifies phenotypic presentation of pathway deficiencies, contributing to the wide phenotypic spectrum and complexity of neurodevelopmental disorders.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas Mutantes/metabolismo , Mutação , Fenótipo , Subunidades Proteicas/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Animais , Disbindina , Proteínas Associadas à Distrofina , Hipocampo/metabolismo , Humanos , Camundongos , Proteínas Mutantes/genética , Neurotransmissores/metabolismo , Pigmentação/genética , Subunidades Proteicas/genética , Esquizofrenia/etiologia , Esquizofrenia/genética , Esquizofrenia/metabolismo , Transcrição Gênica/genética , Ácido gama-Aminobutírico/metabolismo
4.
J Neurosci ; 32(11): 3697-711, 2012 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-22423091

RESUMO

The Biogenesis of Lysosome-Related Organelles Complex 1 (BLOC-1) is a protein complex containing the schizophrenia susceptibility factor dysbindin, which is encoded by the gene DTNBP1. However, mechanisms engaged by dysbindin defining schizophrenia susceptibility pathways have not been quantitatively elucidated. Here, we discovered prevalent and novel cellular roles of the BLOC-1 complex in neuronal cells by performing large-scale Stable Isotopic Labeling of Cells in Culture (SILAC) quantitative proteomics combined with genetic analyses in dysbindin-null mice (Mus musculus) and the genome of schizophrenia patients. We identified 24 proteins that associate with the BLOC-1 complex, many of which were altered in content/distribution in cells or tissues deficient in BLOC-1. New findings include BLOC-1 interactions with the COG complex, a Golgi apparatus tether, and antioxidant enzymes peroxiredoxins 1-2. Importantly, loci encoding eight of the 24 proteins are affected by genomic copy number variation in schizophrenia patients. Thus, our quantitative proteomic studies expand the functional repertoire of the BLOC-1 complex and provide insight into putative molecular pathways of schizophrenia susceptibility.


Assuntos
Proteínas de Transporte/genética , Estudos de Associação Genética/métodos , Predisposição Genética para Doença/genética , Proteínas do Tecido Nervoso/genética , Proteômica/métodos , Esquizofrenia/genética , Animais , Proteínas de Transporte/fisiologia , Linhagem Celular Tumoral , Disbindina , Proteínas Associadas à Distrofina , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/fisiologia , Esquizofrenia/metabolismo , Esquizofrenia/patologia
5.
Mol Biol Cell ; 22(24): 4854-67, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21998198

RESUMO

Dysbindin assembles into the biogenesis of lysosome-related organelles complex 1 (BLOC-1), which interacts with the adaptor protein complex 3 (AP-3), mediating a common endosome-trafficking route. Deficiencies in AP-3 and BLOC-1 affect synaptic vesicle composition. However, whether AP-3-BLOC-1-dependent sorting events that control synapse membrane protein content take place in cell bodies upstream of nerve terminals remains unknown. We tested this hypothesis by analyzing the targeting of phosphatidylinositol-4-kinase type II α (PI4KIIα), a membrane protein present in presynaptic and postsynaptic compartments. PI4KIIα copurified with BLOC-1 and AP-3 in neuronal cells. These interactions translated into a decreased PI4KIIα content in the dentate gyrus of dysbindin-null BLOC-1 deficiency and AP-3-null mice. Reduction of PI4KIIα in the dentate reflects a failure to traffic from the cell body. PI4KIIα was targeted to processes in wild-type primary cultured cortical neurons and PC12 cells but failed to reach neurites in cells lacking either AP-3 or BLOC-1. Similarly, disruption of an AP-3-sorting motif in PI4KIIα impaired its sorting into processes of PC12 and primary cultured cortical neuronal cells. Our findings indicate a novel vesicle transport mechanism requiring BLOC-1 and AP-3 complexes for cargo sorting from neuronal cell bodies to neurites and nerve terminals.


Assuntos
Complexo 3 de Proteínas Adaptadoras/metabolismo , Proteínas de Transporte/metabolismo , Lectinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuritos/metabolismo , Membranas Sinápticas/metabolismo , Vesículas Transportadoras/metabolismo , Complexo 3 de Proteínas Adaptadoras/genética , Motivos de Aminoácidos , Animais , Proteínas de Transporte/genética , Disbindina , Proteínas Associadas à Distrofina , Peptídeos e Proteínas de Sinalização Intracelular , Lectinas/genética , Camundongos , Camundongos Mutantes , Proteínas do Tecido Nervoso/genética , Células PC12 , Ratos , Membranas Sinápticas/genética , Vesículas Transportadoras/genética
6.
Neurobiol Dis ; 34(2): 199-211, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19217433

RESUMO

The expression of the methylated DNA-binding protein MeCP2 increases during neuronal development, which suggests that this epigenetic factor is crucial for neuronal terminal differentiation. We evaluated dendritic and axonal development in embryonic day-18 hippocampal neurons in culture by measuring total length and counting branch point numbers at 4 days in vitro, well before synapse formation. Pyramidal neurons transfected with a plasmid encoding a small hairpin RNA (shRNA) to knockdown endogenous Mecp2 had shorter dendrites than control untransfected neurons, without detectable changes in axonal morphology. On the other hand, overexpression of wildtype (wt) human MECP2 increased dendritic branching, in addition to axonal branching and length. Consistent with reduced neuronal growth and complexity in Rett syndrome (RTT) brains, overexpression of human MECP2 carrying missense mutations common in RTT individuals (R106W or T158M) reduced dendritic and axonal length. One of the targets of MeCP2 transcriptional control is the Bdnf gene. Indeed, endogenous Mecp2 knockdown increased the intracellular levels of BDNF protein compared to untransfected neurons, suggesting that MeCP2 represses Bdnf transcription. Surprisingly, overexpression of wt MECP2 also increased BDNF levels, while overexpression of RTT-associated MECP2 mutants failed to affect BDNF levels. The extracellular BDNF scavenger TrkB-Fc prevented dendritic overgrowth in wt MECP2-overexpressing neurons, while overexpression of the Bdnf gene reverted the dendritic atrophy caused by Mecp2-knockdown. However, this effect was only partial, since Bdnf increased dendritic length only to control levels in mutant MECP2-overexpressing neurons, but not as much as in Bdnf-transfected cells. Our results demonstrate that MeCP2 plays varied roles in dendritic and axonal development during neuronal terminal differentiation, and that some of these effects are mediated by autocrine actions of BDNF.


Assuntos
Atrofia/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Dendritos/metabolismo , Hipocampo/anormalidades , Proteína 2 de Ligação a Metil-CpG/metabolismo , Mutação/genética , Animais , Atrofia/genética , Comunicação Autócrina/genética , Fator Neurotrófico Derivado do Encéfalo/genética , Diferenciação Celular/genética , Células Cultivadas , Dendritos/patologia , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Hipocampo/crescimento & desenvolvimento , Hipocampo/patologia , Humanos , Proteína 2 de Ligação a Metil-CpG/genética , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/metabolismo , Malformações do Sistema Nervoso/fisiopatologia , Neurogênese/genética , Células PC12 , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA