Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 109(28): 11172-7, 2012 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-22745165

RESUMO

Some amyloid-forming polypeptides are associated with devastating human diseases and others provide important biological functions. For both, oligomeric intermediates appear during amyloid assembly. Currently we have few tools for characterizing these conformationally labile intermediates and discerning what governs their benign versus toxic states. Here, we examine intermediates in the assembly of a normal, functional amyloid, the prion-determining region of yeast Sup35 (NM). During assembly, NM formed a variety of oligomers with different sizes and conformation-specific antibody reactivities. Earlier oligomers were less compact and reacted with the conformational antibody A11. More mature oligomers were more compact and reacted with conformational antibody OC. We found we could arrest NM in either of these two distinct oligomeric states with small molecules or crosslinking. The A11-reactive oligomers were more hydrophobic (as measured by Nile Red binding) and were highly toxic to neuronal cells, while OC-reactive oligomers were less hydrophobic and were not toxic. The A11 and OC antibodies were originally raised against oligomers of Aß, an amyloidogenic peptide implicated in Alzheimer's disease (AD) that is completely unrelated to NM in sequence. Thus, this natural yeast prion samples two conformational states similar to those sampled by Aß, and when assembly stalls at one of these two states, but not the other, it becomes extremely toxic. Our results have implications for selective pressures operating on the evolution of amyloid folds across a billion years of evolution. Understanding the features that govern such conformational transitions will shed light on human disease and evolution alike.


Assuntos
Amiloide/química , Doença de Alzheimer/metabolismo , Anisotropia , Sequência Conservada , Detergentes/farmacologia , Corantes Fluorescentes/farmacologia , Humanos , Cinética , Modelos Moleculares , Conformação Molecular , Neurônios/metabolismo , Peptídeos/química , Conformação Proteica , Estrutura Terciária de Proteína , Espectrometria de Fluorescência/métodos , Tirosina/química
2.
Hum Mol Genet ; 20(8): 1478-87, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21258085

RESUMO

Although family history is a well-established risk factor for Parkinson's disease (PD), fewer than 5% of PD cases can be attributed to known genetic mutations. The etiology for the remainder of PD cases is unclear; however, neuronal accumulation of the protein α-synuclein is common to nearly all patients, implicating pathways that influence α-synuclein in PD pathogenesis. We report a genome-wide significant association (P = 3.97 × 10(-8)) between a polymorphism, rs1564282, in the cyclin-G-associated kinase (GAK) gene and increased PD risk, with a meta-analysis odds ratio of 1.48. This association result is based on the meta-analysis of three publicly available PD case-control genome-wide association study and genotyping from a new, independent Italian cohort. Microarray expression analysis of post-mortem frontal cortex from PD and control brains demonstrates a significant association between rs1564282 and higher α-synuclein expression, a known cause of early onset PD. Functional knockdown of GAK in cell culture causes a significant increase in toxicity when α-synuclein is over-expressed. Furthermore, knockdown of GAK in rat primary neurons expressing the A53T mutation of α-synuclein, a well-established model for PD, decreases cell viability. These observations provide evidence that GAK is associated with PD risk and suggest that GAK and α-synuclein interact in a pathway involved in PD pathogenesis. The GAK protein, a serine/threonine kinase, belongs to a family of proteins commonly targeted for drug development. This, combined with GAK's observed relationship to the levels of α-synuclein expression and toxicity, suggests that the protein is an attractive therapeutic target for the treatment of PD.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Doença de Parkinson/genética , Proteínas Serina-Treonina Quinases/genética , alfa-Sinucleína/genética , Adenilato Quinase/metabolismo , Animais , Catepsina D/genética , Catepsina D/metabolismo , Sobrevivência Celular , Células Cultivadas , Estudo de Associação Genômica Ampla , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mutação de Sentido Incorreto , Neurônios/citologia , Neurônios/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Polimorfismo de Nucleotídeo Único , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/metabolismo , Transcrição Gênica , alfa-Sinucleína/metabolismo
3.
Hum Mol Genet ; 19(5): 837-47, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20007718

RESUMO

Pathways regulating neuronal vulnerability are poorly understood, yet are central to identifying therapeutic targets for degenerative neurological diseases. Here, we characterize mechanisms underlying neurodegeneration in Niemann-Pick type C (NPC) disease, a lysosomal storage disorder characterized by impaired cholesterol trafficking. To date, the relative contributions of neuronal and glial defects to neuron loss are poorly defined. Using gene targeting, we generate Npc1 conditional null mutant mice. Deletion of Npc1 in mature cerebellar Purkinje cells leads to an age-dependent impairment in motor tasks, including rotarod and balance beam performance. Surprisingly, these mice did not show the early death or weight loss that are characteristic of global Npc1 null mice, suggesting that Purkinje cell degeneration does not underlie these phenotypes. Histological examination revealed the progressive loss of Purkinje cells in an anterior-to-posterior gradient. This cell autonomous neurodegeneration occurs in a spatiotemporal pattern similar to that of global knockout mice. A subpopulation of Purkinje cells in the posterior cerebellum exhibits marked resistance to cell death despite Npc1 deletion. To explore this selective response, we investigated the electrophysiological properties of vulnerable and susceptible Purkinje cell subpopulations. Unexpectedly, Purkinje cells in both subpopulations displayed no electrophysiological abnormalities prior to degeneration. Our data establish that Npc1 deficiency leads to cell autonomous, selective neurodegeneration and suggest that the ataxic symptoms of NPC disease arise from Purkinje cell death rather than cellular dysfunction.


Assuntos
Neurônios/metabolismo , Doença de Niemann-Pick Tipo C/patologia , Proteínas/genética , Células de Purkinje/patologia , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Transgênicos , Degeneração Neural , Proteína C1 de Niemann-Pick , Doença de Niemann-Pick Tipo C/metabolismo , Proteínas/metabolismo
4.
Autophagy ; 5(4): 548-50, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19332999

RESUMO

The tauopathies are a diverse class of devastating neurodegenerative disorders, characterized by the hyperphosphorylation and aggregation of the microtubule binding protein tau. Niemann-Pick type C disease (NPC) is a tauopathy that affects children,and is caused by mutations in intracellular lipid and cholesterol trafficking proteins. Loss-of-function mutations in the NPC1gene are responsible for 95 percent of all NPC cases, and lead to progressive neurodegeneration and early death. To assess the extent to which tau affects NPC pathology, we generated mice that lack both NPC1 and tau. NPC1/tau double-null mutants exhibit an exacerbated NPC phenotype, including severe systemic manifestations,and die significantly earlier than NPC1 single-null mutants.Since autophagy has been previously implicated in NPC pathogenesis,we investigated the impact of tau deletion on this pathway.Acute reductions of tau in NPC1-deficient fibroblasts significantly decrease autophagic induction and flux, while having no effect on the autophagic pathway in control cells. Here we propose a model in which tau's normal function is critical to the induction of autophagy in NPC1 deficiency, and suggest that this novel mechanism contributes to cellular dysfunction in the tauopathies.


Assuntos
Autofagia , Fibroblastos/patologia , Doença de Niemann-Pick Tipo C/metabolismo , Doença de Niemann-Pick Tipo C/patologia , Proteínas/metabolismo , Proteínas tau/metabolismo , Animais , Fibroblastos/metabolismo , Deleção de Genes , Técnicas de Silenciamento de Genes , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Modelos Biológicos , Proteína C1 de Niemann-Pick , Fenótipo
5.
Hum Mol Genet ; 18(5): 956-65, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19074461

RESUMO

Hyperphosphorylation and aggregation of the microtubule-binding protein tau characterize a diverse array of neurodegenerative disorders. Most of these lack mutations in the encoding MAPT gene, and the role of tau in disease pathogenesis remains controversial. Among these tauopathies is Niemann-Pick type C disease (NPC), a lysosomal storage disorder characterized by progressive neurodegeneration and premature death, most often caused by an inherited deficiency in the intracellular lipid trafficking protein NPC1. To determine the extent to which tau affects NPC pathogenesis, we generated Npc1-/- mice deficient in tau. Unexpectedly, NPC1/tau double null mutants are generated in markedly smaller litters, exhibit an enhanced systemic phenotype and die significantly earlier than NPC1 single null mutants. As autophagy is up-regulated in NPC and protein degradation through this pathway depends on movement along microtubules, we knocked down MAPT expression in NPC1-deficient human fibroblasts and examined effects on this pathway. We show that an acute reduction of tau expression in a cellular model of NPC decreases induction and flux through the autophagic pathway. Our data establish that MAPT deletion exacerbates the NPC phenotype through a mechanism independent of tau protein aggregation and identifies a critical role for tau in the regulation of autophagy in NPC1-deficient cells.


Assuntos
Autofagia , Proteínas de Transporte/metabolismo , Deleção de Genes , Glicoproteínas de Membrana/metabolismo , Doença de Niemann-Pick Tipo C/genética , Proteínas/metabolismo , Proteínas tau/genética , Animais , Proteínas de Transporte/genética , Células Cultivadas , Feminino , Fibroblastos/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína C1 de Niemann-Pick , Doença de Niemann-Pick Tipo C/metabolismo , Doença de Niemann-Pick Tipo C/fisiopatologia , Fenótipo , Proteínas/genética , Proteínas tau/metabolismo
6.
Expert Rev Mol Med ; 10: e26, 2008 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-18782459

RESUMO

Niemann-Pick type C disease (NPC) is a sphingolipid-storage disorder that results from inherited deficiencies of intracellular lipid-trafficking proteins, and is characterised by an accumulation of cholesterol and glycosphingolipids in late endosomes and lysosomes. Patients with this disorder develop progressive neurological impairment that often begins in childhood, is ultimately fatal and is currently untreatable. How impaired lipid trafficking leads to neurodegeneration is largely unknown. Here we review NPC clinical features and biochemical defects, and discuss model systems used to study this disorder. Recent studies have established that NPC is associated with an induction of autophagy, a regulated and evolutionarily conserved process by which cytoplasmic proteins are sequestered within autophagosomes and targeted for degradation. This pathway enables recycling of limited or damaged macromolecules to promote cell survival. However, in other instances, robust activation of autophagy leads to cell stress and programmed cell death. We summarise evidence showing that autophagy induction and flux are increased in NPC by signalling through a complex of the class III phosphoinositide 3-kinase and beclin-1. We propose that an imbalance between induction and flux through the autophagic pathway contributes to cell stress and neuronal loss in NPC and related sphingolipid-storage disorders, and discuss potential therapeutic strategies for modulating activity of this pathway.


Assuntos
Autofagia/fisiologia , Doença de Niemann-Pick Tipo C/patologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Transporte Biológico/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Metabolismo dos Lipídeos/fisiologia , Proteínas de Membrana/metabolismo , Camundongos , Proteína C1 de Niemann-Pick , Doença de Niemann-Pick Tipo C/tratamento farmacológico , Doença de Niemann-Pick Tipo C/genética , Doença de Niemann-Pick Tipo C/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas/genética , Proteínas/metabolismo , Transdução de Sinais , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
7.
Hum Mol Genet ; 16(12): 1495-503, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17468177

RESUMO

Niemann-Pick C (NPC) disease is an autosomal recessive lipid storage disorder characterized by a disruption of sphingolipid and cholesterol trafficking that produces cognitive impairment, ataxia and death, often in childhood. Most cases are caused by loss of function mutations in the Npc1 gene, which encodes a protein that localizes to late endosomes and functions in lipid sorting and vesicle trafficking. Here, we demonstrate that NPC1-deficient primary human fibroblasts, like npc1(-/-) mice fibroblasts, showed increased autophagy as evidenced by elevated LC3-II levels, numerous autophagic vacuoles and enhanced degradation of long-lived proteins. Autophagy because of NPC1 deficiency was associated with increased expression of Beclin-1 rather than activation of the Akt-mTOR-p70 S6K signaling pathway, and siRNA knockdown of Beclin-1 decreased long-lived protein degradation. Induction of cholesterol trafficking defects in wild-type fibroblasts by treatment with U18666A increased Beclin-1 and LC3-II expression, whereas treatment of NPC1-deficient fibroblasts with sphingolipid-lowering compound NB-DGJ failed to alter the expression of either Beclin-1 or LC3-II. Primary fibroblasts from patients with two other sphingolipid storage diseases, NPC2 deficiency and Sandhoff disease, characterized by sphingolipid trafficking defects also showed elevation in Beclin-1 and LC3-II levels. In contrast, Gaucher disease fibroblasts, which traffic sphingolipids normally, showed wild-type levels of Beclin-1 and LC3-II. Our data define a critical role for Beclin-1 in the activation of autophagy because of NPC1 deficiency, and reveal an unexpected role for lipid trafficking in the regulation of this pathway in patients with several sphingolipid storage diseases.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Autofagia , Colesterol/metabolismo , Proteínas de Membrana/metabolismo , Doenças de Niemann-Pick/metabolismo , Esfingolipídeos/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Autofagia/genética , Proteína Beclina-1 , Transporte Biológico/genética , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Proteína C1 de Niemann-Pick , Doenças de Niemann-Pick/genética , Proteínas/genética , Proteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA