Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cell ; 146(3): 359-71, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21816272

RESUMO

Directed conversion of mature human cells, as from fibroblasts to neurons, is of potential clinical utility for neurological disease modeling as well as cell therapeutics. Here, we describe the efficient generation of human-induced neuronal (hiN) cells from adult skin fibroblasts of unaffected individuals and Alzheimer's patients, using virally transduced transcription regulators and extrinsic support factors. hiN cells from unaffected individuals display morphological, electrophysiological, and gene expression profiles that typify glutamatergic forebrain neurons and are competent to integrate functionally into the rodent CNS. hiN cells from familial Alzheimer disease (FAD) patients with presenilin-1 or -2 mutations exhibit altered processing and localization of amyloid precursor protein (APP) and increased production of Aß, relative to the source patient fibroblasts or hiN cells from unaffected individuals. Together, our findings demonstrate directed conversion of human fibroblasts to a neuronal phenotype and reveal cell type-selective pathology in hiN cells derived from FAD patients.


Assuntos
Doença de Alzheimer/patologia , Transdiferenciação Celular , Medicina Regenerativa/métodos , Pele/citologia , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/metabolismo , Células Cultivadas , Fibroblastos/citologia , Humanos , Neurônios/metabolismo , Presenilina-1/metabolismo , Presenilina-2/metabolismo
2.
Nature ; 539(7628): 207-216, 2016 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-27830778

RESUMO

Parkinson's disease is a debilitating, age-associated movement disorder. A central aspect of the pathophysiology of Parkinson's disease is the progressive demise of midbrain dopamine neurons and their axonal projections, but the underlying causes of this loss are unclear. Advances in genetics and experimental model systems have illuminated an important role for defects in intracellular transport pathways to lysosomes. The accumulation of altered proteins and damaged mitochondria, particularly at axon terminals, ultimately might overwhelm the capacity of intracellular disposal mechanisms. Cell-extrinsic mechanisms, including inflammation and prion-like spreading, are proposed to have both protective and deleterious functions in Parkinson's disease.


Assuntos
Doença de Parkinson , Animais , Transporte Biológico/genética , Endocitose , Glucosilceramidase/genética , Glucosilceramidase/metabolismo , Humanos , Inflamação/metabolismo , Inflamação/patologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Lisossomos/metabolismo , Modelos Biológicos , Terapia de Alvo Molecular , Neurônios/metabolismo , Neurônios/patologia , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Doença de Parkinson/terapia , Príons/metabolismo , Vesículas Sinápticas/metabolismo , alfa-Sinucleína/metabolismo
3.
Hum Mol Genet ; 27(2): 385-395, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29177506

RESUMO

Human genetic studies implicate LRRK2 and RAB7L1 in susceptibility to Parkinson disease (PD). These two genes function in the same pathway, as knockout of Rab7L1 results in phenotypes similar to LRRK2 knockout, and studies in cells and model organisms demonstrate LRRK2 and Rab7L1 interact in the endolysosomal system. Recently, a subset of Rab proteins have been identified as LRRK2 kinase substrates. Herein, we find that Rab8, Rab10, and Rab7L1 must be membrane and GTP-bound for LRRK2 phosphorylation. LRRK2 mutations that cause PD including R1441C, Y1699C, and G2019S all increase LRRK2 phosphorylation of Rab7L1 four-fold over wild-type LRRK2 in cells, resulting in the phosphorylation of nearly one-third the available Rab7L1 protein in cells. In contrast, the most common pathogenic LRRK2 mutation, G2019S, does not upregulate LRRK2-mediated phosphorylation of Rab8 or Rab10. LRRK2 interaction with membrane and GTP-bound Rab7L1, but not Rab8 or Rab10, results in the activation of LRRK2 autophosphorylation at the serine 1292 position, required for LRRK2 toxicity. Further, Rab7L1 controls the proportion of LRRK2 that is membrane-associated, and LRRK2 mutations enhance Rab7L1-mediated recruitment of LRRK2 to the trans-Golgi network. Interaction studies with the Rab8 and Rab10 GTPase-activating protein TBC1D4/AS160 demonstrate that LRRK2 phosphorylation may block membrane and GTP-bound Rab protein interaction with effectors. These results suggest reciprocal regulation between LRRK2 and Rab protein substrates, where Rab7L1-mediated upregulation of LRRK2 kinase activity results in the stabilization of membrane and GTP-bound Rab proteins that may be unable to interact with Rab effector proteins.


Assuntos
Guanosina Trifosfato/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab1 de Ligação ao GTP/metabolismo , Rede trans-Golgi/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Células HEK293 , Humanos , Proteínas de Membrana/metabolismo , Mutação , Fosforilação , Transporte Proteico
4.
Nature ; 500(7460): 45-50, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23883936

RESUMO

Late-onset Alzheimer's disease (LOAD) risk is strongly influenced by genetic factors such as the presence of the apolipoprotein E ε4 allele (referred to here as APOE4), as well as non-genetic determinants including ageing. To pursue mechanisms by which these affect human brain physiology and modify LOAD risk, we initially analysed whole-transcriptome cerebral cortex gene expression data in unaffected APOE4 carriers and LOAD patients. APOE4 carrier status was associated with a consistent transcriptomic shift that broadly resembled the LOAD profile. Differential co-expression correlation network analysis of the APOE4 and LOAD transcriptomic changes identified a set of candidate core regulatory mediators. Several of these--including APBA2, FYN, RNF219 and SV2A--encode known or novel modulators of LOAD associated amyloid beta A4 precursor protein (APP) endocytosis and metabolism. Furthermore, a genetic variant within RNF219 was found to affect amyloid deposition in human brain and LOAD age-of-onset. These data implicate an APOE4 associated molecular pathway that promotes LOAD.


Assuntos
Doença de Alzheimer/genética , Apolipoproteína E4/genética , Genoma Humano/genética , Genômica , Idade de Início , Idoso , Alelos , Doença de Alzheimer/epidemiologia , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Células Cultivadas , Córtex Cerebral/metabolismo , Endocitose , Epistasia Genética , Feminino , Fibroblastos , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Heterozigoto , Humanos , Levetiracetam , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fenótipo , Piracetam/análogos & derivados , Piracetam/farmacologia , Polimorfismo Genético/genética , Proteólise/efeitos dos fármacos , Transcriptoma/genética
5.
Nature ; 488(7413): 652-5, 2012 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-22902501

RESUMO

Somatic cells can be reprogrammed into induced pluripotent stem cells (iPSCs) by using the pluripotency factors Oct4, Sox2, Klf4 and c-Myc (together referred to as OSKM). iPSC reprogramming erases somatic epigenetic signatures­as typified by DNA methylation or histone modification at silent pluripotency loci­and establishes alternative epigenetic marks of embryonic stem cells (ESCs). Here we describe an early and essential stage of somatic cell reprogramming, preceding the induction of transcription at endogenous pluripotency loci such as Nanog and Esrrb. By day 4 after transduction with OSKM, two epigenetic modification factors necessary for iPSC generation, namely poly(ADP-ribose) polymerase-1 (Parp1) and ten-eleven translocation-2 (Tet2), are recruited to the Nanog and Esrrb loci. These epigenetic modification factors seem to have complementary roles in the establishment of early epigenetic marks during somatic cell reprogramming: Parp1 functions in the regulation of 5-methylcytosine (5mC) modification, whereas Tet2 is essential for the early generation of 5-hydroxymethylcytosine (5hmC) by the oxidation of 5mC (refs 3,4). Although 5hmC has been proposed to serve primarily as an intermediate in 5mC demethylation to cytosine in certain contexts, our data, and also studies of Tet2-mutant human tumour cells, argue in favour of a role for 5hmC as an epigenetic mark distinct from 5mC. Consistent with this, Parp1 and Tet2 are each needed for the early establishment of histone modifications that typify an activated chromatin state at pluripotency loci, whereas Parp1 induction further promotes accessibility to the Oct4 reprogramming factor. These findings suggest that Parp1 and Tet2 contribute to an epigenetic program that directs subsequent transcriptional induction at pluripotency loci during somatic cell reprogramming.


Assuntos
Reprogramação Celular , Proteínas de Ligação a DNA/metabolismo , Epigênese Genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Cromatina/genética , Cromatina/metabolismo , Metilação de DNA , Dioxigenases , Éxons/genética , Fibroblastos/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Íntrons/genética , Fator 4 Semelhante a Kruppel , Camundongos , Proteína Homeobox Nanog , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/genética , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo
7.
PLoS Genet ; 9(10): e1003845, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098148

RESUMO

Macroautophagy is a conserved mechanism for the bulk degradation of proteins and organelles. Pathological studies have implicated defective macroautophagy in neurodegeneration, but physiological functions of macroautophagy in adult neurons remain unclear. Here we show that Atg7, an essential macroautophagy component, regulates dopaminergic axon terminal morphology. Mature Atg7-deficient midbrain dopamine (DA) neurons harbored selectively enlarged axonal terminals. This contrasted with the phenotype of DA neurons deficient in Pten - a key negative regulator of the mTOR kinase signaling pathway and neuron size - that displayed enlarged soma but unaltered axon terminals. Surprisingly, concomitant deficiency of both Atg7 and Pten led to a dramatic enhancement of axon terminal enlargement relative to Atg7 deletion alone. Similar genetic interactions between Atg7 and Pten were observed in the context of DA turnover and DA-dependent locomotor behaviors. These data suggest a model for morphological regulation of mature dopaminergic axon terminals whereby the impact of mTOR pathway is suppressed by macroautophagy.


Assuntos
Proteínas Associadas aos Microtúbulos/genética , PTEN Fosfo-Hidrolase/genética , Terminações Pré-Sinápticas/metabolismo , Serina-Treonina Quinases TOR/genética , Animais , Autofagia/genética , Proteína 7 Relacionada à Autofagia , Corpo Estriado/metabolismo , Neurônios Dopaminérgicos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/metabolismo , Morfogênese/genética , Atividade Motora/genética , Atividade Motora/fisiologia , PTEN Fosfo-Hidrolase/metabolismo , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/metabolismo
9.
Nat Med ; 30(5): 1406-1415, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38745011

RESUMO

GRN mutations cause progranulin haploinsufficiency, which eventually leads to frontotemporal dementia (FTD-GRN). PR006 is an investigational gene therapy delivering the granulin gene (GRN) using an adeno-associated virus serotype 9 (AAV9) vector. In non-clinical studies, PR006 transduced neurons derived from induced pluripotent stem cells of patients with FTD-GRN, resulted in progranulin expression and improvement of lipofuscin, lysosomal and neuroinflammation pathologies in Grn-knockout mice, and was well tolerated except for minimal, asymptomatic dorsal root ganglionopathy in non-human primates. We initiated a first-in-human phase 1/2 open-label trial. Here we report results of a pre-specified interim analysis triggered with the last treated patient of the low-dose cohort (n = 6) reaching the 12-month follow-up timepoint. We also include preliminary data from the mid-dose cohort (n = 7). Primary endpoints were safety, immunogenicity and change in progranulin levels in cerebrospinal fluid (CSF) and blood. Secondary endpoints were Clinical Dementia Rating (CDR) plus National Alzheimer's Disease Coordinating Center (NACC) Frontotemporal Lobar Degeneration (FTLD) rating scale and levels of neurofilament light chain (NfL). One-time administration of PR006 into the cisterna magna was generally safe and well tolerated. All patients developed treatment-emergent anti-AAV9 antibodies in the CSF, but none developed anti-progranulin antibodies. CSF pleocytosis was the most common PR006-related adverse event. Twelve serious adverse events occurred, mostly unrelated to PR006. Deep vein thrombosis developed in three patients. There was one death (unrelated) occurring 18 months after treatment. CSF progranulin increased after PR006 treatment in all patients; blood progranulin increased in most patients but only transiently. NfL levels transiently increased after PR006 treatment, likely reflecting dorsal root ganglia toxicity. Progression rates, based on the CDR scale, were within the broad ranges reported for patients with FTD. These data provide preliminary insights into the safety and bioactivity of PR006. Longer follow-up and additional studies are needed to confirm the safety and potential efficacy of PR006. ClinicalTrials.gov identifier: NCT04408625 .


Assuntos
Dependovirus , Demência Frontotemporal , Terapia Genética , Progranulinas , Humanos , Demência Frontotemporal/genética , Demência Frontotemporal/terapia , Demência Frontotemporal/líquido cefalorraquidiano , Progranulinas/genética , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Dependovirus/genética , Pessoa de Meia-Idade , Feminino , Masculino , Idoso , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/líquido cefalorraquidiano , Vetores Genéticos , Animais , Resultado do Tratamento , Pesquisa Translacional Biomédica , Camundongos , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/líquido cefalorraquidiano , Proteínas de Neurofilamentos/sangue
11.
J Parkinsons Dis ; 11(s2): S183-S188, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34151863

RESUMO

Human genetic studies as well as studies in animal models indicate that lysosomal dysfunction plays a key role in the pathogenesis of Parkinson's disease. Among the lysosomal genes involved, GBA1 has the largest impact on Parkinson's disease risk. Deficiency in the GBA1 encoded enzyme glucocerebrosidase (GCase) leads to the accumulation of the GCase glycolipid substrates glucosylceramide and glucosylsphingosine and ultimately results in toxicity and inflammation and negatively affect many clinical aspects of Parkinson's disease, including disease risk, the severity of presentation, age of onset, and likelihood of progression to dementia. These findings support the view that re-establishing normal levels of GCase enzyme activity may reduce the progression of Parkinson's disease in patients carrying GBA1 mutations. Studies in mouse models indicate that PR001, a AAV9 vector-based gene therapy designed to deliver a functional GBA1 gene to the brain, suggest that this therapeutic approach may slow or stop disease progression. PR001 is currently being evaluated in clinical trials with Parkinson's disease patients carrying GBA1 mutations.


Assuntos
Doença de Gaucher , Doença de Parkinson , Animais , Terapia Genética , Glucosilceramidase/genética , Humanos , Lisossomos , Camundongos , Mutação , Doença de Parkinson/genética , Doença de Parkinson/terapia , alfa-Sinucleína
12.
Neuron ; 52(4): 587-93, 2006 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-17114044

RESUMO

Mutations in LRRK2 underlie an autosomal-dominant, inherited form of Parkinson's disease (PD) that mimics the clinical features of the common "sporadic" form of PD. The LRRK2 protein includes putative GTPase, protein kinase, WD40 repeat, and leucine-rich repeat (LRR) domains of unknown function. Here we show that PD-associated LRRK2 mutations display disinhibited kinase activity and induce a progressive reduction in neurite length and branching both in primary neuronal cultures and in the intact rodent CNS. In contrast, LRRK2 deficiency leads to increased neurite length and branching. Neurons that express PD-associated LRRK2 mutations additionally harbor prominent phospho-tau-positive inclusions with lysosomal characteristics and ultimately undergo apoptosis.


Assuntos
Encéfalo/metabolismo , Predisposição Genética para Doença/genética , Degeneração Neural/metabolismo , Neuritos/metabolismo , Transtornos Parkinsonianos/genética , Proteínas Serina-Treonina Quinases/genética , Animais , Animais Recém-Nascidos , Apoptose/genética , Encéfalo/patologia , Encéfalo/fisiopatologia , Forma Celular/genética , Células Cultivadas , Humanos , Corpos de Inclusão/genética , Corpos de Inclusão/metabolismo , Corpos de Inclusão/patologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Lisossomos/genética , Lisossomos/metabolismo , Lisossomos/patologia , Camundongos , Mutação/genética , Degeneração Neural/genética , Degeneração Neural/fisiopatologia , Neuritos/patologia , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/fisiopatologia , Ratos , Ratos Sprague-Dawley , Substância Negra/metabolismo , Substância Negra/patologia , Substância Negra/fisiopatologia , Proteínas tau/genética , Proteínas tau/metabolismo
13.
Neuron ; 37(5): 735-49, 2003 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-12628165

RESUMO

Mutations in parkin, which encodes a RING domain protein associated with ubiquitin ligase activity, lead to autosomal recessive Parkinson's disease characterized by midbrain dopamine neuron loss. Here we show that parkin functions in a multiprotein ubiquitin ligase complex that includes the F-box/WD repeat protein hSel-10 and Cullin-1. HSel-10 serves to target the parkin ubiquitin ligase activity to cyclin E, an hSel-10-interacting protein previously implicated in the regulation of neuronal apoptosis. Consistent with the notion that cyclin E is a substrate of the parkin ubiquitin ligase complex, parkin deficiency potentiates the accumulation of cyclin E in cultured postmitotic neurons exposed to the glutamatergic excitotoxin kainate and promotes their apoptosis. Furthermore, parkin overexpression attenuates the accumulation of cyclin E in toxin-treated primary neurons, including midbrain dopamine neurons, and protects them from apoptosis.


Assuntos
Ácido Caínico/toxicidade , Ligases/biossíntese , Mitose/efeitos dos fármacos , Mitose/fisiologia , Neurônios/efeitos dos fármacos , Peptídeo Sintases/biossíntese , Ubiquitina-Proteína Ligases , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Células HeLa , Humanos , Ligases/genética , Camundongos , Neurônios/citologia , Neurônios/metabolismo , Peptídeo Sintases/genética , Proteínas Ligases SKP Culina F-Box
15.
PLoS Biol ; 2(11): e362, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15502874

RESUMO

Parkinson's disease (PD) pathology is characterized by the degeneration of midbrain dopamine neurons (DNs) ultimately leading to a progressive movement disorder in patients. The etiology of DN loss in sporadic PD is unknown, although it is hypothesized that aberrant protein aggregation and cellular oxidative stress may promote DN degeneration. Homozygous mutations in DJ-1 were recently described in two families with autosomal recessive inherited PD (Bonifati et al. 2003). In a companion article (Martinat et al. 2004), we show that mutations in DJ-1 alter the cellular response to oxidative stress and proteasomal inhibition. Here we show that DJ-1 functions as a redox-sensitive molecular chaperone that is activated in an oxidative cytoplasmic environment. We further demonstrate that DJ-1 chaperone activity in vivo extends to alpha-synuclein, a protein implicated in PD pathogenesis.


Assuntos
Chaperonas Moleculares/química , Proteínas Oncogênicas/fisiologia , Oxirredução , alfa-Sinucleína/química , Antioxidantes/química , Diferenciação Celular , Linhagem Celular Tumoral , Cisteína/química , Citoplasma/metabolismo , Dimerização , Dopamina/metabolismo , Vetores Genéticos , Células HeLa , Homozigoto , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Chaperonas Moleculares/metabolismo , Mutação , Neurônios/metabolismo , Proteínas Oncogênicas/metabolismo , Estresse Oxidativo , Oxigênio/química , Doença de Parkinson/metabolismo , Proteína Desglicase DJ-1 , Espectrofotometria , Sinucleínas/química
16.
PLoS Biol ; 2(11): e327, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15502868

RESUMO

The hallmark of Parkinson's disease (PD) is the selective loss of dopamine neurons in the ventral midbrain. Although the cause of neurodegeneration in PD is unknown, a Mendelian inheritance pattern is observed in rare cases, indicating a genetic factor. Furthermore, pathological analyses of PD substantia nigra have correlated cellular oxidative stress and altered proteasomal function with PD. Homozygous mutations in DJ-1 were recently described in two families with autosomal recessive Parkinsonism, one of which is a large deletion that is likely to lead to loss of function. Here we show that embryonic stem cells deficient in DJ-1 display increased sensitivity to oxidative stress and proteasomal inhibition. The accumulation of reactive oxygen species in toxin-treated DJ-1-deficient cells initially appears normal, but these cells are unable to cope with the consequent damage that ultimately leads to apoptotic death. Furthermore, we find that dopamine neurons derived from in vitro-differentiated DJ-1-deficient embryonic stem cells display decreased survival and increased sensitivity to oxidative stress. These data are consistent with a protective role for DJ-1, and demonstrate the utility of genetically modified embryonic stem cell-derived neurons as cellular models of neuronal disorders.


Assuntos
Dopamina/metabolismo , Embrião de Mamíferos/citologia , Neurônios/metabolismo , Proteínas Oncogênicas/genética , Estresse Oxidativo , Doença de Parkinson/metabolismo , Células-Tronco/citologia , Animais , Apoptose , Diferenciação Celular , Sobrevivência Celular , DNA Complementar/metabolismo , Modelos Animais de Doenças , Deleção de Genes , Vetores Genéticos , Heterozigoto , Homozigoto , Humanos , Peróxido de Hidrogênio/farmacologia , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Modelos Genéticos , Dados de Sequência Molecular , Mutação , Doenças Neurodegenerativas/metabolismo , Peroxirredoxinas , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína Desglicase DJ-1 , Interferência de RNA , Espécies Reativas de Oxigênio , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Substância Negra/patologia
17.
Cell Syst ; 4(4): 404-415.e5, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28330615

RESUMO

Human age-associated traits, such as cognitive decline, can be highly variable across the population, with some individuals exhibiting traits that are not expected at a given chronological age. Here we present differential aging (Δ-aging), an unbiased method that quantifies individual variability in age-associated phenotypes within a tissue of interest, and apply this approach to the analysis of existing transcriptome-wide cerebral cortex gene expression data from several cohorts totaling 1,904 autopsied human brain samples. We subsequently performed a genome-wide association study and identified the TMEM106B and GRN gene loci, previously associated with frontotemporal dementia, as determinants of Δ-aging in the cerebral cortex with genome-wide significance. TMEM106B risk variants are associated with inflammation, neuronal loss, and cognitive deficits, even in the absence of known brain disease, and their impact is highly selective for the frontal cerebral cortex of older individuals (>65 years). The methodological framework we describe can be broadly applied to the analysis of quantitative traits associated with aging or with other parameters.


Assuntos
Envelhecimento/genética , Córtex Cerebral/metabolismo , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Progranulinas/genética , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/patologia , Envelhecimento/fisiologia , Córtex Cerebral/patologia , Estudos de Coortes , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Imunidade Inata/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Degeneração Neural/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Fenótipo , Progranulinas/metabolismo , Progranulinas/fisiologia
18.
Cell Rep ; 21(7): 1727-1736, 2017 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-29141208

RESUMO

Mutations in presenilin (PSEN) 1 and 2, which encode components of the γ-secretase (GS) complex, cause familial Alzheimer's disease (FAD). It is hypothesized that altered GS-mediated processing of the amyloid precursor protein (APP) to the Aß42 fragment, which is accumulated in diseased brain, may be pathogenic. Here, we describe an in vitro model system that enables the facile analysis of neuronal disease mechanisms in non-neuronal patient cells using CRISPR gene activation of endogenous disease-relevant genes. In FAD patient-derived fibroblast cultures, CRISPR activation of APP or BACE unmasked an occult processivity defect in downstream GS-mediated carboxypeptidase cleavage of APP, ultimately leading to higher Aß42 levels. These data suggest that, selectively in neurons, relatively high levels of BACE1 activity lead to substrate pressure on FAD-mutant GS complexes, promoting CNS Aß42 accumulation. Our results introduce an additional platform for analysis of neurological disease.


Assuntos
Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Fibroblastos/metabolismo , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/genética , Precursor de Proteína beta-Amiloide/genética , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Sistemas CRISPR-Cas , Células Cultivadas , Fibroblastos/citologia , Humanos , Neurônios/citologia , Neurônios/metabolismo , Processamento de Proteína Pós-Traducional , Pele/citologia , Ativação Transcricional
19.
Sci Rep ; 6: 29945, 2016 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-27424887

RESUMO

Leucine-rich repeat kinase 2 (LRRK2) has been linked to several clinical disorders including Parkinson's disease (PD), Crohn's disease, and leprosy. Furthermore in rodents, LRRK2 deficiency or inhibition leads to lysosomal pathology in kidney and lung. Here we provide evidence that LRRK2 functions together with a second PD-associated gene, RAB7L1, within an evolutionarily conserved genetic module in diverse cellular contexts. In C. elegans neurons, orthologues of LRRK2 and RAB7L1 act coordinately in an ordered genetic pathway to regulate axonal elongation. Further genetic studies implicated the AP-3 complex, which is a known regulator of axonal morphology as well as of intracellular protein trafficking to the lysosome compartment, as a physiological downstream effector of LRRK2 and RAB7L1. Additional cell-based studies implicated LRRK2 in the AP-3 complex-related intracellular trafficking of lysosomal membrane proteins. In mice, deficiency of either RAB7L1 or LRRK2 leads to prominent age-associated lysosomal defects in kidney proximal tubule cells, in the absence of frank CNS pathology. We hypothesize that defects in this evolutionarily conserved genetic pathway underlie the diverse pathologies associated with LRRK2 in humans and in animal models.


Assuntos
Axônios/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Lisossomos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Axônios/ultraestrutura , Linhagem Celular , Endossomos/metabolismo , Endossomos/ultraestrutura , Células HEK293 , Humanos , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/deficiência , Lisossomos/ultraestrutura , Proteínas de Membrana/metabolismo , Camundongos , Neurônios Motores/metabolismo , Ligação Proteica , Transporte Proteico , Proteínas rab de Ligação ao GTP/genética
20.
J Mol Neurosci ; 27(2): 175-83, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16186628

RESUMO

The ubiquitin-proteasome pathway (UPP) has been implicated in the regulation of a number of key cellular processes in mammalian cells, including the cell cycle and apoptosis. Furthermore, defects in the UPP have been implicated in neurodegenerative disorders such as Parkinson's disease (PD), as mutations in the ubiquitin ligase Parkin underlie a familial form of parkinsonism and ubiquitinated inclusions are a defining hallmark of PD pathology. To functionally dissect molecular components of the UPP in postmitotic neurons, we used RNA interference to knock down genes that encode genetically characterized components of the UPP. Here, we show that knockdown of two such components, the ubiquitin ligase scaffolding protein Cullin-1 (Cul-1) and the proteasome-associated deubiquitinating protein Pad-1, lead to cell cycle reactivation and apoptosis in subsets of postmitotic neurons. Furthermore, knockdown of Cul-1 appears to specifically affect the dopaminergic population. These data support the hypothesis that the UPP normally functions to regulate cell-cycle reentry in postmitotic neurons and further implicate this pathway in dopamine neuron degeneration.


Assuntos
Ciclo Celular/fisiologia , Neurônios/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Apoptose , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Proteínas Culina/genética , Proteínas Culina/metabolismo , Dopamina/metabolismo , Resistência a Múltiplos Medicamentos/genética , Humanos , Mesencéfalo/citologia , Neurônios/citologia , Complexo de Endopeptidases do Proteassoma/genética , Interferência de RNA , Transativadores/genética , Transativadores/metabolismo , Ubiquitina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA