Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Aging Neurosci ; 13: 754956, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34720999

RESUMO

Mutations in leucine-rich repeat kinase 2 gene (LRRK2) are the most frequent genetic factors contributing to Parkinson's disease (PD). G2385R-LRRK2 increases the risk for PD susceptibility in the Chinese population. However, the pathological role of G2385R-LRRK2 is not clear. In this study, we investigate the roles of G2385R-LRRK2 in neurodegeneration underlying PD pathogenesis using cell biology and pharmacology approaches. We demonstrated that expression of G2385R-LRRK2-induced neurotoxicity in human neuroblastoma SH-SY5Y and mouse primary neurons. G2385R-LRRK2 increased mitochondrial ROS, activates caspase-3/7, and increased PARP cleavage, resulting in neurotoxicity. Treatment with curcumin (an antioxidant) significantly protected against G2385R-LRRK2-induced neurodegeneration by reducing mitochondrial ROS, caspase-3/7 activation, and PARP cleavage. We also found that the cellular environmental stressor, H2O2 significantly promotes both WT-LRRK2- and G2385R-LRRK2-induced neurotoxicity by increasing mitochondrial ROS, caspase-3/7 activation, and PARP cleavage, while curcumin attenuated this combined neurotoxicity. These findings not only provide a novel understanding of G2385R roles in neurodegeneration and environment interaction but also provide a pharmacological approach for intervention for G2385R-LRRK2-linked PD.

2.
Mov Disord ; 36(11): 2519-2529, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34390268

RESUMO

BACKGROUND: Spinocerebellar ataxia type 2 (SCA2) is a neurodegenerative disease caused by expansion of a CAG repeat in Ataxin-2 (ATXN2) gene. The mutant ATXN2 protein with a polyglutamine tract is known to be toxic and contributes to the SCA2 pathogenesis. OBJECTIVE: Here, we tested the hypothesis that the mutant ATXN2 transcript with an expanded CAG repeat (expATXN2) is also toxic and contributes to SCA2 pathogenesis. METHODS: The toxic effect of expATXN2 transcripts on SK-N-MC neuroblastoma cells and primary mouse cortical neurons was evaluated by caspase 3/7 activity and nuclear condensation assay, respectively. RNA immunoprecipitation assay was performed to identify RNA binding proteins (RBPs) that bind to expATXN2 RNA. Quantitative PCR was used to examine if ribosomal RNA (rRNA) processing is disrupted in SCA2 and Huntington's disease (HD) human brain tissue. RESULTS: expATXN2 RNA induces neuronal cell death, and aberrantly interacts with RBPs involved in RNA metabolism. One of the RBPs, transducin ß-like protein 3 (TBL3), involved in rRNA processing, binds to both expATXN2 and expanded huntingtin (expHTT) RNA in vitro. rRNA processing is disrupted in both SCA2 and HD human brain tissue. CONCLUSION: These findings provide the first evidence of a contributory role of expATXN2 transcripts in SCA2 pathogenesis, and further support the role of expHTT transcripts in HD pathogenesis. The disruption of rRNA processing, mediated by aberrant interaction of RBPs with expATXN2 and expHTT transcripts, suggest a point of convergence in the pathogeneses of repeat expansion diseases with potential therapeutic implications. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Assuntos
RNA , Ataxias Espinocerebelares , Animais , Ataxinas/metabolismo , Encéfalo/patologia , Camundongos , Neurônios/metabolismo , RNA/metabolismo , Proteínas de Ligação a RNA/genética , Ataxias Espinocerebelares/patologia
3.
Cells ; 10(2)2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33672296

RESUMO

Mutations in the leucine-rich repeat kinase-2 (LRRK2) gene cause autosomal-dominant Parkinson's disease (PD) and contribute to sporadic PD. Common genetic variation in LRRK2 modifies susceptibility to immunological disorders including Crohn's disease and leprosy. Previous studies have reported that LRRK2 is expressed in B lymphocytes and macrophages, suggesting a role for LRRK2 in immunological functions. In this study, we characterized the LRRK2 protein expression and phosphorylation using human lymphoblasts. Lipopolysaccharide (LPS), a proinflammatory agent, induced the increase of LRRK2 expression and kinase activities in human lymphoblasts in a time-dependent manner. Moreover, LPS activated the Toll-like receptor (TLR) signaling pathway, increased TRAF6/LRRK2 interaction, and elevated the phosphorylation levels of MAPK (JNK1/2, p38, and ERK1/2) and IkBα. Treatment with LRRK2 inhibitor 68 reduced LPS-induced TRAF6/LRRK2 interaction and MAPK and IkBα phosphorylation, thereby reducing TNF-α secretion. These results indicate that LRRK2 is actively involved in proinflammatory responses in human lymphoblasts, and inhibition of GTP binding by 68 results in an anti-inflammation effect against proinflammatory stimuli. These findings not only provide novel insights into the mechanisms of LRRK2-linked immune and inflammatory responses in B-cell-like lymphoblasts, but also suggest that 68 may also have potential therapeutic value for LRRK2-linked immunological disorders.


Assuntos
Guanosina Trifosfato/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Lipopolissacarídeos/farmacologia , Linfócitos/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Ativação Enzimática/efeitos dos fármacos , Células HEK293 , Humanos , Linfócitos/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Modelos Biológicos , Inibidor de NF-kappaB alfa/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Fator 6 Associado a Receptor de TNF/metabolismo
5.
Hum Mol Genet ; 29(8): 1340-1352, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32242231

RESUMO

Nemo-like kinase (NLK), an evolutionarily conserved serine/threonine kinase, is highly expressed in the brain, but its function in the adult brain remains not well understood. In this study, we identify NLK as an interactor of huntingtin protein (HTT). We report that NLK levels are significantly decreased in HD human brain and HD models. Importantly, overexpression of NLK in the striatum attenuates brain atrophy, preserves striatal DARPP32 levels and reduces mutant HTT (mHTT) aggregation in HD mice. In contrast, genetic reduction of NLK exacerbates brain atrophy and loss of DARPP32 in HD mice. Moreover, we demonstrate that NLK lowers mHTT levels in a kinase activity-dependent manner, while having no significant effect on normal HTT protein levels in mouse striatal cells, human cells and HD mouse models. The NLK-mediated lowering of mHTT is associated with enhanced phosphorylation of mHTT. Phosphorylation defective mutation of serine at amino acid 120 (S120) abolishes the mHTT-lowering effect of NLK, suggesting that S120 phosphorylation is an important step in the NLK-mediated lowering of mHTT. A further mechanistic study suggests that NLK promotes mHTT ubiquitination and degradation via the proteasome pathway. Taken together, our results indicate a protective role of NLK in HD and reveal a new molecular target to reduce mHTT levels.


Assuntos
Atrofia/genética , Fosfoproteína 32 Regulada por cAMP e Dopamina/genética , Proteína Huntingtina/genética , Doença de Huntington/genética , Proteínas Serina-Treonina Quinases/genética , Animais , Atrofia/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Modelos Animais de Doenças , Humanos , Doença de Huntington/patologia , Camundongos , Neostriado/metabolismo , Neostriado/patologia , Neurônios/metabolismo , Neurônios/patologia , Fosforilação/genética , Complexo de Endopeptidases do Proteassoma/genética
6.
Hum Mol Genet ; 28(23): 3982-3996, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31626293

RESUMO

Mutations in LRRK2 cause autosomal dominant and sporadic Parkinson's disease, but the mechanisms involved in LRRK2 toxicity in PD are yet to be fully understood. We found that LRRK2 translocates to the nucleus by binding to seven in absentia homolog (SIAH-1), and in the nucleus it directly interacts with lamin A/C, independent of its kinase activity. LRRK2 knockdown caused nuclear lamina abnormalities and nuclear disruption. LRRK2 disease mutations mostly abolish the interaction with lamin A/C and, similar to LRRK2 knockdown, cause disorganization of lamin A/C and leakage of nuclear proteins. Dopaminergic neurons of LRRK2 G2019S transgenic and LRRK2 -/- mice display decreased circularity of the nuclear lamina and leakage of the nuclear protein 53BP1 to the cytosol. Dopaminergic nigral and cortical neurons of both LRRK2 G2019S and idiopathic PD patients exhibit abnormalities of the nuclear lamina. Our data indicate that LRRK2 plays an essential role in maintaining nuclear envelope integrity. Disruption of this function by disease mutations suggests a novel phosphorylation-independent loss-of-function mechanism that may synergize with other neurotoxic effects caused by LRRK2 mutations.


Assuntos
Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Membrana Nuclear/metabolismo , Doença de Parkinson/genética , Animais , Células Cultivadas , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Células HEK293 , Humanos , Lamina Tipo A/metabolismo , Mutação com Perda de Função , Camundongos , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Fosforilação , Ratos , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo
7.
J Huntingtons Dis ; 8(2): 129-143, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30856117

RESUMO

BACKGROUND: The potential benefit of cysteamine for Huntington's disease has been demonstrated in HD animal models. Cysteamine and its derivate cystamine were shown to reduce neuropathology and prolong lifespan. Human studies have demonstrated safety, and suggestive results regarding efficacy. Despite all the studies available in vivo, there are only few in vitro studies, and the mechanism of action of cysteamine remains unclear. OBJECTIVE: The objective of this study is to assess the capacity of cysteamine for neuroprotection against mutant Huntingtin in vitro using cellular models of HD, and to provide initial data regarding mechanism of action. METHODS: We tested the neuroprotective properties of cysteamine in vitro in our primary neuron and iPSC models of HD. RESULTS: Cysteamine showed a strong neuroprotective effect (EC50 = 7.1 nM) against mutant Htt-(aa-1-586 82Q) toxicity, in a nuclear condensation cell toxicity assay. Cysteamine also rescued mitochondrial changes induced by mutant Htt. Modulation of the levels of cysteine or glutathione failed to protect neurons, suggesting that cysteamine neuroprotection is not mediated through cysteine metabolism. Taurine and Hypotaurine, which are metabolites of cysteamine can protect neurons against Htt toxicity, but the inhibition of the enzyme converting cysteamine to hypotaurine does not block either protective activity, suggesting independent protective pathways. Cysteamine has been suggested to activate BDNF secretion; however, cysteamine protection was not blocked by BDNF pathway antagonists. CONCLUSIONS: Cysteamine was strongly neuroprotective with relatively high potency. We demonstrated that the main neuroprotective pathways that have been proposed to be the mechanism of protection by cysteamine can all be blocked and still not prevent the neuroprotective effect. The results suggest the involvement of other yet-to-be-determined neuroprotective pathways.


Assuntos
Cisteamina/farmacologia , Proteína Huntingtina/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Animais , Células Cultivadas , Proteína Huntingtina/metabolismo , Proteína Huntingtina/toxicidade , Doença de Huntington , Camundongos , Mutação , Neurônios/patologia
8.
J Neurosci Res ; 97(4): 467-479, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30489648

RESUMO

Altered brain iron content in the striatum of premanifest and manifest Huntington's disease (HD) has been reported. However, its natural history remains unclear. This study aims to investigate altered brain iron content in premanifest and early HD, and the iron deposition rate in these patients through a longitudinal one-year follow-up test, with quantitative magnetic susceptibility as an iron imaging marker. Twenty-four gene mutation carriers divided into three groups (further-from-onset, closer-to-onset and early HD) and 16 age-matched healthy controls were recruited at baseline, and of these, 14 carriers and 7 controls completed the one-year follow-up. Quantitative magnetic susceptibility and effective transverse relaxation rate ( R2∗ ) were measured at 7.0 Tesla and correlated with atrophy and available clinical and cognitive measurements. Higher susceptibility values indicating higher iron content in the striatum and globus pallidus were only observed in closer-to-onset (N = 6, p < 0.05 in caudate and p < 0.01 in putamen) and early HD (N = 9, p < 0.05 in caudate and globus pallidus and p < 0.01 in putamen). Similar results were found by R2∗ measurement. Such increases directly correlated with HD CAG-age product score and brain atrophy, but not with motor or cognitive scores. More importantly, a significantly higher iron deposition rate (11.9%/years in caudate and 6.1%/years in globus pallidus) was firstly observed in closer-to-onset premanifest HD and early HD as compared to the controls. These results suggest that monitoring brain iron may provide further insights into the pathophysiology of HD disease progression, and may provide a biomarker for clinical trials.


Assuntos
Encéfalo/diagnóstico por imagem , Doença de Huntington/diagnóstico por imagem , Doença de Huntington/fisiopatologia , Ferro/metabolismo , Imageamento por Ressonância Magnética/métodos , Adulto , Atrofia/fisiopatologia , Encéfalo/fisiopatologia , Química Encefálica , Mapeamento Encefálico , Transtornos Cognitivos/fisiopatologia , Disfunção Cognitiva/fisiopatologia , Corpo Estriado/fisiopatologia , Estudos Transversais , Progressão da Doença , Feminino , Globo Pálido/fisiopatologia , Substância Cinzenta , Humanos , Doença de Huntington/genética , Processamento de Imagem Assistida por Computador , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Transtornos Motores/fisiopatologia , Fatores de Tempo
9.
Proc Natl Acad Sci U S A ; 114(39): 10479-10484, 2017 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-28894008

RESUMO

Genome-wide association studies have implicated the ANK3 locus in bipolar disorder, a major human psychotic illness. ANK3 encodes ankyrin-G, which organizes the neuronal axon initial segment (AIS). We generated a mouse model with conditional disruption of ANK3 in pyramidal neurons of the adult forebrain (Ank-G cKO). This resulted in the expected loss of pyramidal neuron AIS voltage-gated sodium and potassium channels. There was also dramatic loss of markers of afferent GABAergic cartridge synapses, resembling the cortical microcircuitry changes in brains from psychotic patients, and suggesting disinhibition. Expression of c-fos was increased in cortical pyramidal neurons, consistent with increased neuronal activity due to disinhibition. The mice showed robust behavioral phenotypes reminiscent of aspects of human mania, ameliorated by antimania drugs lithium and valproate. Repeated social defeat stress resulted in repeated episodes of dramatic behavioral changes from hyperactivity to "depression-like" behavior, suggestive of some aspects of human bipolar disorder. Overall, we suggest that this Ank-G cKO mouse model recapitulates some of the core features of human bipolar disorder and indicates that cortical microcircuitry alterations during adulthood may be involved in pathogenesis. The model may be useful for studying disease pathophysiology and for developing experimental therapeutics.


Assuntos
Anquirinas/genética , Transtorno Bipolar/tratamento farmacológico , Transtorno Bipolar/genética , Prosencéfalo/fisiopatologia , Sinapses/patologia , Animais , Transtorno Bipolar/fisiopatologia , Modelos Animais de Doenças , Neurônios GABAérgicos/patologia , Lítio/farmacologia , Metilfenidato/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Proteínas Proto-Oncogênicas c-fyn/biossíntese , Ácido Valproico/farmacologia , Canais de Sódio Disparados por Voltagem/genética
10.
Nat Commun ; 8: 14405, 2017 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-28194040

RESUMO

Huntington's Disease (HD) is a neurodegenerative disease caused by poly-glutamine expansion in the Htt protein, resulting in Htt misfolding and cell death. Expression of the cellular protein folding and pro-survival machinery by heat shock transcription factor 1 (HSF1) ameliorates biochemical and neurobiological defects caused by protein misfolding. We report that HSF1 is degraded in cells and mice expressing mutant Htt, in medium spiny neurons derived from human HD iPSCs and in brain samples from patients with HD. Mutant Htt increases CK2α' kinase and Fbxw7 E3 ligase levels, phosphorylating HSF1 and promoting its proteasomal degradation. An HD mouse model heterozygous for CK2α' shows increased HSF1 and chaperone levels, maintenance of striatal excitatory synapses, clearance of Htt aggregates and preserves body mass compared with HD mice homozygous for CK2α'. These results reveal a pathway that could be modulated to prevent neuronal dysfunction and muscle wasting caused by protein misfolding in HD.


Assuntos
Encéfalo/metabolismo , Fatores de Transcrição de Choque Térmico/metabolismo , Doença de Huntington/metabolismo , Neurônios/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Células HEK293 , Fatores de Transcrição de Choque Térmico/genética , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células PC12 , Ratos
11.
Mol Neuropsychiatry ; 2(3): 133-144, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27867938

RESUMO

Schizophrenia and other major mental illnesses result from a complex interplay of genetic and environmental factors. We previously identified a mutation in NPAS3 that results in a valine to isoleucine (V304I) amino acid substitution segregating with schizophrenia in a small family. The amino acid change occurs in a potentially critical region for protein function. Furthermore, the same amino acid substitution in proteins related to familial Alzheimer's disease and transthyretin amyloidosis has been associated with protein aggregation. In this study, we demonstrate that NPAS3 is prone to aggregation, and that the V304I mutation in NPAS3 increases this propensity in both bacterial and mammalian expression systems. We also show that NPAS3-V304I reduces soluble endogenous NPAS3, and increases insoluble endogenous NPAS3 and leads to alteration of transcriptional activity. These results suggest that protein aggregation, potentially leading to cell dysfunction via a loss of protein function through sequestration, may contribute to the pathogenesis of schizophrenia and other forms of mental illness. Further exploration of the mechanisms leading to abnormal protein quality control could lead to new therapeutic targets.

12.
Hum Mol Genet ; 25(16): 3454-3466, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27378699

RESUMO

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a CAG expansion in the gene-encoding Huntingtin (HTT). Transcriptome dysregulation is a major feature of HD pathogenesis, as revealed by a large body of work on gene expression profiling of tissues from human HD patients and mouse models. These studies were primarily focused on transcriptional changes affecting steady-state overall gene expression levels using microarray based approaches. A major missing component, however, has been the study of transcriptome changes at the post-transcriptional level, such as alternative splicing. Alternative splicing is a critical mechanism for expanding regulatory and functional diversity from a limited number of genes, and is particularly complex in the mammalian brain. Here we carried out a deep RNA-seq analysis of the BA4 (Brodmann area 4) motor cortex from seven human HD brains and seven controls to systematically discover aberrant alternative splicing events and characterize potential associated splicing factors in HD. We identified 593 differential alternative splicing events between HD and control brains. Using two expanded panels with a total of 108 BA4 tissues from patients and controls, we identified four splicing factors exhibiting significantly altered expression levels in HD patient brains. Moreover, follow-up molecular analyses of one splicing factor PTBP1 revealed its impact on disease-associated splicing patterns in HD. Collectively, our data provide genomic evidence for widespread splicing dysregulation in HD brains, and suggest the role of aberrant alternative splicing in the pathogenesis of HD.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas/genética , Doença de Huntington/genética , Córtex Motor/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Transcriptoma/genética , Adulto , Idoso , Processamento Alternativo/genética , Animais , Autopsia , Feminino , Regulação da Expressão Gênica , Predisposição Genética para Doença , Ribonucleoproteínas Nucleares Heterogêneas/biossíntese , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Proteína Huntingtina/genética , Doença de Huntington/fisiopatologia , Masculino , Camundongos , Pessoa de Meia-Idade , Córtex Motor/patologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/biossíntese
13.
Schizophr Res ; 172(1-3): 101-5, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26925800

RESUMO

The extent of age-related changes in glutamate and other neurometabolites in the anterior cingulate cortex (ACC) in individuals with schizophrenia remain unclear. Magnetic resonance spectroscopy (MRS) at 7 T, which yields precise measurements of various metabolites and can distinguish glutamate from glutamine, was used to determine levels of ACC glutamate and other metabolites in 24 individuals with schizophrenia and 24 matched controls. Multiple regression analysis revealed that ACC glutamate decreased with age in patients but not controls. No changes were detected in levels of glutamine, N-acetylaspartate, N-acetylaspartylglutamic acid, myo-inositol, GABA, glutathione, total creatine, and total choline. These results suggest that age may be an important modifier of ACC glutamate in schizophrenia.


Assuntos
Envelhecimento/metabolismo , Ácido Glutâmico/metabolismo , Giro do Cíngulo/metabolismo , Transtornos Psicóticos/metabolismo , Esquizofrenia/metabolismo , Adulto , Antipsicóticos/uso terapêutico , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Colina/metabolismo , Creatina/metabolismo , Dipeptídeos/metabolismo , Feminino , Glutamina/metabolismo , Glutationa/metabolismo , Humanos , Inositol/metabolismo , Masculino , Espectroscopia de Prótons por Ressonância Magnética/instrumentação , Transtornos Psicóticos/tratamento farmacológico , Esquizofrenia/tratamento farmacológico , Ácido gama-Aminobutírico/metabolismo
14.
Hum Mol Genet ; 25(4): 672-80, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26744328

RESUMO

Mutations in leucine-rich repeat kinase 2 (LRRK2) cause autosomal-dominant Parkinsonism with pleomorphic pathology including deposits of aggregated protein and neuronal degeneration. The pathogenesis of LRRK2-linked Parkinson's disease (PD) is not fully understood. Here, using co-immunoprecipitation, we found that LRRK2 interacted with synphilin-1 (SP1), a cytoplasmic protein that interacts with α-synuclein and has implications in PD pathogenesis. LRRK2 interacted with the N-terminus of SP1 whereas SP1 predominantly interacted with the C-terminus of LRRK2, including kinase domain. Co-expression of SP1 with LRRK2 increased LRRK2-induced cytoplasmic aggregation in cultured cells. Moreover, SP1 also attenuated mutant LRRK2-induced toxicity and reduced LRRK2 kinase activity in cultured cells. Knockdown of SP1 by siRNA enhanced LRRK2 neuronal toxicity. In vivo Drosophila studies, co-expression of SP1 and mutant G2019S-LRRK2 in double transgenic Drosophila increased survival and improved locomotor activity. Expression of SP1 protects against G2019S-LRRK2-induced dopamine neuron loss and reduced LRRK2 phosphorylation in double transgenic fly brains. Our findings demonstrate that SP1 attenuates mutant LRRK2-induced PD-like phenotypes and plays a neural protective role.


Assuntos
Proteínas de Transporte/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Doença de Parkinson/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Dopamina/metabolismo , Drosophila , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Camundongos , Mutação , Degeneração Neural/genética , Proteínas do Tecido Nervoso/genética , Neurônios/enzimologia , Neurônios/metabolismo , Neurônios/patologia , Doença de Parkinson/enzimologia , Doença de Parkinson/genética , Doença de Parkinson/patologia , Fosforilação , Domínios e Motivos de Interação entre Proteínas , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
15.
Neuron ; 88(4): 667-77, 2015 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-26590344

RESUMO

Huntington disease (HD) is caused by a CAG ⋅ CTG expansion in the huntingtin (HTT) gene. While most research has focused on the HTT polyGln-expansion protein, we demonstrate that four additional, novel, homopolymeric expansion proteins (polyAla, polySer, polyLeu, and polyCys) accumulate in HD human brains. These sense and antisense repeat-associated non-ATG (RAN) translation proteins accumulate most abundantly in brain regions with neuronal loss, microglial activation and apoptosis, including caudate/putamen, white matter, and, in juvenile-onset cases, also the cerebellum. RAN protein accumulation and aggregation are length dependent, and individual RAN proteins are toxic to neural cells independent of RNA effects. These data suggest RAN proteins contribute to HD and that therapeutic strategies targeting both sense and antisense genes may be required for efficacy in HD patients. This is the first demonstration that RAN proteins are expressed across an expansion located in an open reading frame and suggests RAN translation may also contribute to other polyglutamine diseases.


Assuntos
Encéfalo/metabolismo , Doença de Huntington/genética , Proteínas do Tecido Nervoso/genética , RNA Antissenso/genética , Expansão das Repetições de Trinucleotídeos/genética , Adulto , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Cerebelo/metabolismo , Criança , Feminino , Lobo Frontal/metabolismo , Células HEK293 , Humanos , Proteína Huntingtina , Doença de Huntington/metabolismo , Masculino , Pessoa de Meia-Idade , Neostriado/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Peptídeos/metabolismo , Agregação Patológica de Proteínas/metabolismo , Biossíntese de Proteínas , RNA Antissenso/metabolismo , Adulto Jovem
16.
Sci Rep ; 5: 12521, 2015 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-26218986

RESUMO

Huntington's disease (HD) is caused by a CAG repeat expansion in the huntingtin (HTT) gene. Recent evidence suggests that HD is a consequence of multimodal, non-mutually exclusive mechanisms of pathogenesis that involve both HTT protein- and HTT RNA-triggered mechanisms. Here we provide further evidence for the role of expanded HTT (expHTT) RNA in HD by demonstrating that a fragment of expHTT is cytotoxic in the absence of any translation and that the extent of cytotoxicity is similar to the cytotoxicity of an expHTT protein fragment encoded by a transcript of similar length and with a similar repeat size. In addition, full-length (FL) expHTT is retained in the nucleus. Overexpression of the splicing factor muscleblind-like 1 (MBNL1) increases nuclear retention of expHTT and decreases the expression of expHTT protein in the cytosol. The splicing and nuclear export factor U2AF65 has the opposite effect, decreasing expHTT nuclear retention and increasing expression of expHTT protein. This suggests that MBNL1 and U2AF65 play a role in nuclear export of expHTT RNA.


Assuntos
Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/metabolismo , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo , Transporte Ativo do Núcleo Celular , Linhagem Celular Tumoral , Códon de Iniciação , Regulação da Expressão Gênica , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Biossíntese de Proteínas/genética , RNA Mensageiro/metabolismo , Fator de Processamento U2AF , Expansão das Repetições de Trinucleotídeos
17.
Hum Mol Genet ; 24(9): 2508-27, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25609071

RESUMO

White matter abnormalities have been reported in premanifest Huntington's disease (HD) subjects before overt striatal neuronal loss, but whether the white matter changes represent a necessary step towards further pathology and the underlying mechanism of these changes remains unknown. Here, we characterized a novel knock-in mouse model that expresses mouse HD gene homolog (Hdh) with extended CAG repeat- HdhQ250, which was derived from the selective breeding of HdhQ150 mice. HdhQ250 mice manifest an accelerated and robust phenotype compared with its parent line. HdhQ250 mice exhibit progressive motor deficits, reduction in striatal and cortical volume, accumulation of mutant huntingtin aggregation, decreased levels of DARPP32 and BDNF and altered striatal metabolites. The abnormalities detected in this mouse model are reminiscent of several aspects of human HD. In addition, disturbed myelination was evident in postnatal Day 14 HdhQ250 mouse brain, including reduced levels of myelin regulatory factor and myelin basic protein, and decreased numbers of myelinated axons in the corpus callosum. Thinner myelin sheaths, indicated by increased G-ratio of myelin, were also detected in the corpus callosum of adult HdhQ250 mice. Moreover, proliferation of oligodendrocyte precursor cells is altered by mutant huntingtin both in vitro and in vivo. Our data indicate that this model is suitable for understanding comprehensive pathogenesis of HD in white matter and gray matter as well as developing therapeutics for HD.


Assuntos
Encéfalo/patologia , Doença de Huntington/patologia , Doença de Huntington/fisiopatologia , Atividade Motora , Substância Branca/patologia , Alelos , Animais , Atrofia , Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proliferação de Células , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Espectroscopia de Ressonância Magnética , Camundongos , Camundongos Transgênicos , Mutação , Bainha de Mielina/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Oligodendroglia/metabolismo , Tamanho do Órgão , Agregação Patológica de Proteínas , Substância Branca/metabolismo
18.
Nat Rev Dis Primers ; 1: 15005, 2015 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-27188817

RESUMO

Huntington disease is devastating to patients and their families - with autosomal dominant inheritance, onset typically in the prime of adult life, progressive course, and a combination of motor, cognitive and behavioural features. The disease is caused by an expanded CAG trinucleotide repeat (of variable length) in HTT, the gene that encodes the protein huntingtin. In mutation carriers, huntingtin is produced with abnormally long polyglutamine sequences that confer toxic gains of function and predispose the protein to fragmentation, resulting in neuronal dysfunction and death. In this Primer, we review the epidemiology of Huntington disease, noting that prevalence is higher than previously thought, geographically variable and increasing. We describe the relationship between CAG repeat length and clinical phenotype, as well as the concept of genetic modifiers of the disease. We discuss normal huntingtin protein function, evidence for differential toxicity of mutant huntingtin variants, theories of huntingtin aggregation and the many different mechanisms of Huntington disease pathogenesis. We describe the genetic and clinical diagnosis of the condition, its clinical assessment and the multidisciplinary management of symptoms, given the absence of effective disease-modifying therapies. We review past and present clinical trials and therapeutic strategies under investigation, including impending trials of targeted huntingtin-lowering drugs and the progress in development of biomarkers that will support the next generation of trials. For an illustrated summary of this Primer, visit: http://go.nature.com/hPMENh.


Assuntos
Doença de Huntington/genética , Adulto , Humanos , Proteína Huntingtina/genética , Doença de Huntington/epidemiologia , Doença de Huntington/terapia , Proteínas do Tecido Nervoso/genética , Peptídeos/genética , Fenótipo , Prevalência , Expansão das Repetições de Trinucleotídeos
19.
Hum Mol Genet ; 23(R1): R17-26, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24824217

RESUMO

The cell biology of human neurodegenerative diseases has been difficult to study till recently. The development of human induced pluripotent stem cell (iPSC) models has greatly enhanced our ability to model disease in human cells. Methods have recently been improved, including increasing reprogramming efficiency, introducing non-viral and non-integrating methods of cell reprogramming, and using novel gene editing techniques for generating genetically corrected lines from patient-derived iPSCs, or for generating mutations in control cell lines. In this review, we highlight accomplishments made using iPSC models to study neurodegenerative disorders such as Huntington's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis, Fronto-Temporal Dementia, Alzheimer's disease, Spinomuscular Atrophy and other polyglutamine diseases. We review disease-related phenotypes shown in patient-derived iPSCs differentiated to relevant neural subtypes, often with stressors or cell "aging", to enhance disease-specific phenotypes. We also discuss prospects for the future of using of iPSC models of neurodegenerative disorders, including screening and testing of therapeutic compounds, and possibly of cell transplantation in regenerative medicine. The new iPSC models have the potential to greatly enhance our understanding of pathogenesis and to facilitate the development of novel therapeutics.


Assuntos
Doença de Alzheimer/terapia , Demência Frontotemporal/terapia , Doença de Huntington/terapia , Células-Tronco Pluripotentes Induzidas/transplante , Doença de Parkinson/terapia , Doença de Alzheimer/genética , Animais , Diferenciação Celular , Modelos Animais de Doenças , Demência Frontotemporal/genética , Humanos , Doença de Huntington/genética , Doença de Parkinson/genética , Fenótipo
20.
PLoS One ; 9(2): e88284, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24505464

RESUMO

Phosphorylation has been shown to have a significant impact on expanded huntingtin-mediated cellular toxicity. Several phosphorylation sites have been identified on the huntingtin (Htt) protein. To find new potential therapeutic targets for Huntington's Disease (HD), we used mass spectrometry to identify novel phosphorylation sites on N-terminal Htt, expressed in HEK293 cells. Using site-directed mutagenesis we introduced alterations of phosphorylation sites in a N586 Htt construct containing 82 polyglutamine repeats. The effects of these alterations on expanded Htt toxicity were evaluated in primary neurons using a nuclear condensation assay and a direct time-lapse imaging of neuronal death. As a result of these studies, we identified several novel phosphorylation sites, validated several known sites, and discovered one phospho-null alteration, S116A, that had a protective effect against expanded polyglutamine-mediated cellular toxicity. The results suggest that S116 is a potential therapeutic target, and indicate that our screening method is useful for identifying candidate phosphorylation sites.


Assuntos
Doença de Huntington/genética , Proteínas do Tecido Nervoso/genética , Mutação Puntual , Serina/genética , Sequência de Aminoácidos , Animais , Morte Celular , Células Cultivadas , Células HEK293 , Humanos , Proteína Huntingtina , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Camundongos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Fosforilação , Serina/química , Serina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA