Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 28(4): 548-560, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30329047

RESUMO

Ubiquitin specific proteases (USPs) are de-ubiquitinases (DUBs) that control protein ubiquitination cycle. The role of DUBs is poorly understood in neurodegenerative diseases. We found that USP13 is overexpressed in post-mortem Parkinson's disease (PD) brains. We investigated whether changes in USP13 levels can affect two molecules, parkin and alpha-synuclein, that are implicated in PD pathogenesis. Parkin is an E3 ubiquitin ligase that is regulated by ubiquitination and targets certain proteins for degradation, and alpha-synuclein may be ubiquitinated and recycled in the normal brain. We found that USP13 independently regulates parkin and alpha-synuclein ubiquitination in models of alpha-synucleinopathies. USP13 shRNA knockdown increases alpha-synuclein ubiquitination and clearance, in a parkin-independent manner. Furthermore, USP13 overexpression counteracts the effects of a tyrosine kinase inhibitor, Nilotinib, while USP13 knockdown facilitates Nilotinib effects on alpha-synculein clearance, suggesting that alpha-synuclein ubiquitnation is important for its clearance. These studies provide novel evidence of USP13 effects on parkin and alpha-synuclein metabolism and suggest that USP13 is a potential therapeutic target in the alpha-synucleinopathies.


Assuntos
Endopeptidases/genética , Doença de Parkinson/genética , Ubiquitina-Proteína Ligases/genética , alfa-Sinucleína/genética , Autopsia , Encéfalo/metabolismo , Encéfalo/patologia , Endopeptidases/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Doença de Parkinson/patologia , Pirimidinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Proteases Específicas de Ubiquitina/genética , Ubiquitinação/genética
4.
Mol Cell Neurosci ; 83: 46-54, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28687523

RESUMO

The relationship between RNA-binding proteins, particularly TAR DNA binding protein 43 (TDP-43), and neurodegeneration is an important area of research. TDP-43 is involved in so many cellular processes that perturbation of protein homeostasis can lead to countless downstream effects. Understanding what leads to this disease-related protein imbalance and the resulting cellular and molecular effects will help to develop targets for disease intervention, whether it be prevention of protein accumulation, or addressing a secondary effect of protein accumulation. Here we review the current literature of TDP-43 and TDP-43 pathologies, the effects of TDP-43 overexpression and disruption of synaptic proteins through its binding of messenger RNA, leading to synaptic dysfunction. This review highlights some of the still-limited knowledge of the protein TDP-43 and how it can contribute to disease.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Degeneração Lobar Frontotemporal/metabolismo , Doença dos Neurônios Motores/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Ligação a DNA/genética , Degeneração Lobar Frontotemporal/genética , Humanos , Doença dos Neurônios Motores/genética , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo , Transmissão Sináptica
5.
J Neurochem ; 139(4): 610-623, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27507246

RESUMO

The trans-activating response of DNA/RNA-binding protein (TDP)-43 pathology is associated with many neurodegenerative diseases via unknown mechanisms. Here, we use a transgenic mouse model over-expressing human wild-type neuronal TDP-43 to study the effects of TDP-43 pathology on glutamate metabolism and synaptic function. We found that neuronal TDP-43 over-expression affects synaptic protein expression, including Synapsin I, and alters surrounding astrocytic function. TDP-43 over-expression is associated with an increase in glutamate and γ-amino butyric acid and reduction of glutamine and aspartate levels, indicating impairment of presynaptic terminal. TDP-43 also decreases tricarboxylic acid cycle metabolism and induces oxidative stress via lactate accumulation. Neuronal TDP-43 does not alter microglia activity or significantly changes systemic and brain inflammatory markers compared to control. We previously demonstrated that brain-penetrant tyrosine kinase inhibitors (TKIs), nilotinib and bosutinib, reduce TDP-43-induced cell death in transgenic mice. Here, we show that TKIs reverse the effects of TDP-43 on synaptic proteins, increase astrocytic function and restore glutamate and neurotransmitter balance in TDP-43 mice. Nilotinib, but not bosutinib, reverses mitochondrial impairment and oxidative metabolism. Taken together, these data suggest that TKIs can attenuate TDP-43 toxicity and improve synaptic and astrocytic function, independent of microglial or other inflammatory effects. In conclusion, our data demonstrate novel mechanisms of the effects of neuronal TDP-43 over-expression on synaptic protein expression and alteration of astrocytic function.


Assuntos
Astrócitos/fisiologia , Proteínas de Ligação a DNA/biossíntese , Homeostase/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/metabolismo , Sinapsinas/biossíntese , Animais , Astrócitos/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Expressão Gênica , Homeostase/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas Tirosina Quinases/antagonistas & inibidores , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapsinas/genética
6.
Hum Mol Genet ; 23(18): 4960-9, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24847002

RESUMO

The transactivation DNA-binding protein (TDP)-43 binds to thousands of mRNAs, but the functional outcomes of this binding remain largely unknown. TDP-43 binds to Park2 mRNA, which expresses the E3 ubiquitin ligase parkin. We previously demonstrated that parkin ubiquitinates TDP-43 and facilitates its translocation from the nucleus to the cytoplasm. Here we used brain penetrant tyrosine kinase inhibitors (TKIs), including nilotinib and bosutinib and showed that they reduce the level of nuclear TDP-43, abrogate its effects on neuronal loss, and reverse cognitive and motor decline. Nilotinib decreased soluble and insoluble TDP-43, while bosutinib did not affect the insoluble level. Parkin knockout mice exhibited high levels of endogenous TDP-43, while nilotinib and bosutinib did not alter TDP-43, underscoring an indispensable role for parkin in TDP-43 sub-cellular localization. These data demonstrate a novel functional relationship between parkin and TDP-43 and provide evidence that TKIs are potential therapeutic candidates for TDP-43 pathologies.


Assuntos
Cognição/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Destreza Motora/efeitos dos fármacos , Neurônios/metabolismo , Inibidores de Proteínas Quinases/administração & dosagem , Ubiquitina-Proteína Ligases/metabolismo , Compostos de Anilina/administração & dosagem , Compostos de Anilina/farmacologia , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Neurônios/patologia , Nitrilas/administração & dosagem , Nitrilas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/administração & dosagem , Pirimidinas/farmacologia , Quinolinas/administração & dosagem , Quinolinas/farmacologia , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
7.
Hum Mol Genet ; 23(5): 1365-75, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24154541

RESUMO

Intraneuronal accumulation of ß-amyloid (Aß)42 is one of the earliest pathological events in humans and in animal models of Alzheimer's disease (AD). Apolipoprotein E 4 (APOE4) is the major identified genetic risk factor for late-onset AD, with Aß deposition beginning earlier in apoE4-positive subjects. To directly determine the effects of APOE genotype on intraneuronal accumulation of Aß1-42 at the onset of AD pathogenesis, we introduced lentiviral Aß1-42 into the cortex of APOE targeted replacement (TR) mice at the age of 8-9 months. We demonstrated a significant isoform-dependent effect of human APOE, with dramatically enhanced intracellular Aß1-42 deposits in the cerebral cortex of APOE4-TR mice 2 weeks after injection. Double-immunofluorescent staining showed that intracellular accumulation of lentiviral Aß1-42 was mainly present in neurons, localized to late endosomes/lysosomes. This intraneuronal accumulation of Aß1-42 correlated with increased tau phosphorylation and cell death in the ipsilateral cortex around the injection site. Aß1-42 was also observed in microglia, but not in astrocytes. Quantitative analysis revealed more neurons with Aß1-42 while less microglia with Aß1-42 nearest to the injection site of Aß1-42 lentivirus in APOE4-TR mice. Finally, apoE was present in neurons of the ipsilateral cortex of APOE-TR mice at 2 weeks after lentivirus injection, in addition to astrocytes and microglia in both the ipsilateral and contralateral cerebral cortex. Taken together, these results demonstrate that apoE4 tips the balance of the glial and neuronal Aß toward the intraneuronal accumulation of Aß.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Apolipoproteína E4/genética , Vetores Genéticos/genética , Genótipo , Lentivirus/genética , Neurônios/metabolismo , Transdução Genética , Animais , Apolipoproteína E4/metabolismo , Córtex Cerebral/metabolismo , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Humanos , Espaço Intracelular/metabolismo , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Microinjeções , Ligação Proteica , Transporte Proteico
8.
Hum Mol Genet ; 22(16): 3315-28, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23666528

RESUMO

Parkinson's disease is a movement disorder characterized by death of dopaminergic substantia nigra (SN) neurons and brain accumulation of α-synuclein. The tyrosine kinase Abl is activated in neurodegeneration. Here, we show that lentiviral expression of α-synuclein in the mouse SN leads to Abl activation (phosphorylation) and lentiviral Abl expression increases α-synuclein levels, in agreement with Abl activation in PD brains. Administration of the tyrosine kinase inhibitor nilotinib decreases Abl activity and ameliorates autophagic clearance of α-synuclein in transgenic and lentiviral gene transfer models. Subcellular fractionation shows accumulation of α-synuclein and hyper-phosphorylated Tau (p-Tau) in autophagic vacuoles in α-synuclein expressing brains, but nilotinib enhances protein deposition into the lysosomes. Nilotinib is used for adult leukemia treatment and it enters the brain within US Food and Drug Administration approved doses, leading to autophagic degradation of α-synuclein, protection of SN neurons and amelioration of motor performance. These data suggest that nilotinib may be a therapeutic strategy to degrade α-synuclein in PD and other α-synucleinopathies.


Assuntos
Autofagia , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Transtornos Parkinsonianos/metabolismo , Pirimidinas/farmacologia , alfa-Sinucleína/metabolismo , Adulto , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Neurônios Dopaminérgicos/fisiologia , Genes abl , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/genética , Mutação Puntual , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-abl/metabolismo , Pirimidinas/administração & dosagem , Pirimidinas/uso terapêutico , Substância Negra/metabolismo , alfa-Sinucleína/sangue
9.
Neurodegener Dis ; 15(5): 259-70, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26160424

RESUMO

Parkin biology has emerged as an exciting area of pharmaceutical development for several human diseases, including cancer and neurodegeneration. Parkin's role is multifaceted in human health and disease and its function affecting major cellular quality control mechanisms, including the ubiquitin-proteasome and autophagy-lysosome systems, is critical in the maintenance of cellular homeostasis. Loss of Parkin function due to aging, protein instability and gene mutations is manifest in a number of human diseases, contributing to the validation of this protein as a therapeutic target. Parkin activation to mobilize cellular quality control mechanisms and counteract dyshomeostasis is a highly desirable area for therapeutic development. The elucidation of Parkin's crystal structure and better understanding of possible posttranslational modifications (i.e. phosphorylation, ubiquitination, etc.) that regulate Parkin's enzymatic activity suggest that this protein is a therapeutic drug target in many human diseases. Here we review Parkin's role in health and disease and discuss the effects of self-ubiquitination and deubiquitination on Parkin activity. This review provides further evidence showing the longitudinal effects of Parkin deletion on mitochondrial function, oxidative stress and neurotransmitter balance in vivo using high-frequency (1)H/(13)C NMR spectroscopy.


Assuntos
Autofagia , Neoplasias Encefálicas/enzimologia , Encéfalo/enzimologia , Mitocôndrias/enzimologia , Doenças Neurodegenerativas/enzimologia , Ubiquitina-Proteína Ligases/metabolismo , Aminoácidos/metabolismo , Animais , Apoptose , Homeostase , Humanos , Neurônios/enzimologia , Estresse Oxidativo , Doença de Parkinson/enzimologia , Doença de Parkinson/genética , Ubiquitina-Proteína Ligases/genética
10.
J Biol Chem ; 288(6): 4103-15, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23258539

RESUMO

The importance of E3 ubiquitin ligases, involved in the degradation of misfolded proteins or promotion of protein-protein interaction, is increasingly recognized in neurodegeneration. TDP-43 is a predominantly nuclear protein, which regulates the transcription of thousands of genes and binds to mRNA of the E3 ubiquitin ligase Parkin to regulate its expression. Wild type and mutated TDP-43 are detected in ubiquitinated forms within the cytosol in several neurodegenerative diseases. We elucidated the mechanisms of TDP-43 interaction with Parkin using transgenic A315T mutant TDP-43 (TDP43-Tg) mice, lentiviral wild type TDP-43, and Parkin gene transfer rat models. TDP-43 expression increased Parkin mRNA and protein levels. Lentiviral TDP-43 increased the levels of nuclear and cytosolic protein, whereas Parkin co-expression mediated Lys-48 and Lys-63-linked ubiquitin to TDP-43 and led to cytosolic co-localization of Parkin with ubiquitinated TDP-43. Parkin and TDP-43 formed a multiprotein complex with HDAC6, perhaps to mediate TDP-43 translocation. In conclusion, Parkin ubiquitinates TDP-43 and facilitates its cytosolic accumulation through a multiprotein complex with HDAC6.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Histona Desacetilases/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Substituição de Aminoácidos , Animais , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/genética , Desacetilase 6 de Histona , Histona Desacetilases/genética , Humanos , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Proteínas do Tecido Nervoso/genética , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Transporte Proteico/genética , Proteólise , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Ubiquitina-Proteína Ligases/genética
11.
J Neurochem ; 129(2): 350-61, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24298989

RESUMO

The E3 ubiquitin ligase Parkin plays a central role in the pathogenesis of many neurodegenerative diseases. Parkin promotes specific ubiquitination and affects the localization of transactivation response DNA-binding protein 43 (TDP-43), which controls the translation of thousands of mRNAs. Here we tested the effects of lentiviral Parkin and TDP-43 expression on amino acid metabolism in the rat motor cortex using high frequency ¹³C NMR spectroscopy. TDP-43 expression increased glutamate levels, decreased the levels of other amino acids, including glutamine, aspartate, leucine and isoleucine, and impaired mitochondrial tricarboxylic acid cycle. TDP-43 induced lactate accumulation and altered the balance between excitatory (glutamate) and inhibitory (GABA) neurotransmitters. Parkin restored amino acid levels, neurotransmitter balance and tricarboxylic acid cycle metabolism, rescuing neurons from TDP-43-induced apoptotic death. Furthermore, TDP-43 expression led to an increase in 4E-BP levels, perhaps altering translational control and deregulating amino acid synthesis; while Parkin reversed the effects of TDP-43 on the 4E-BP signaling pathway. Taken together, these data suggest that Parkin may affect TDP-43 localization and mitigate its effects on 4E-BP signaling and loss of amino acid homeostasis.


Assuntos
Aminoácidos/metabolismo , Morte Celular/efeitos dos fármacos , Proteinopatias TDP-43/tratamento farmacológico , Ubiquitina-Proteína Ligases/farmacologia , Animais , Western Blotting , Proteínas de Transporte/metabolismo , Caspase 3/metabolismo , Ciclo do Ácido Cítrico/efeitos dos fármacos , Fluorometria , Vetores Genéticos , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Lentivirus/genética , Espectroscopia de Ressonância Magnética , Masculino , Córtex Motor/efeitos dos fármacos , Córtex Motor/metabolismo , Neurotransmissores/metabolismo , Fosfoproteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Proteinopatias TDP-43/patologia , Serina-Treonina Quinases TOR/metabolismo , Ácido gama-Aminobutírico/metabolismo
12.
J Biol Chem ; 287(24): 20534-44, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22518836

RESUMO

Inclusion body myositis, the most common muscle disorder in the elderly, is partly characterized by abnormal expression of amyloid precursor protein (APP) and intracellular accumulation of its proteolytic fragments collectively known as ß-amyloid. The present study examined the effects of ß-amyloid accumulation on mitochondrial structure and function of skeletal muscle from transgenic mice (MCK-ßAPP) engineered to accumulate intramyofiber ß-amyloid. Electron microscopic analysis revealed that a large fraction of myofibers from 2-3-month-old MCK-ßAPP mice contained numerous, heterogeneous alterations in mitochondria, and other cellular organelles. [(1)H-decoupled](13)C NMR spectroscopy showed a substantial reduction in TCA cycle activity and indicated a switch from aerobic to anaerobic glucose metabolism in the MCK-ßAPP muscle. Isolated muscle fibers from the MCK-ßAPP mice also exhibited a reduction in cytoplasmic pH, an increased rate of ROS production, and a partially depolarized plasmalemma. Treatment of MCK-ßAPP muscle cells with Ru360, a mitochondrial Ca(2+) uniporter antagonist, reversed alterations in the plasmalemmal membrane potential (V(m)) and pH. Consistent with altered redox state of the cells, treatment of MCK-ßAPP muscle cells with glutathione reversed the effects of ß-amyloid accumulation on Ca(2+) transient amplitudes. We conclude that structural and functional alterations in mitochondria precede the reported appearance of histopathological and clinical features in the MCK-ßAPP mice and may represent key early events in the pathogenesis of inclusion body myositis.


Assuntos
Precursor de Proteína beta-Amiloide/biossíntese , Mitocôndrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Miosite de Corpos de Inclusão/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Cálcio/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/patologia , Ciclo do Ácido Cítrico/genética , Citoplasma , Glucose/genética , Glucose/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Espectroscopia de Ressonância Magnética , Camundongos , Camundongos Transgênicos , Mitocôndrias Musculares/genética , Mitocôndrias Musculares/patologia , Músculo Esquelético/patologia , Miosite de Corpos de Inclusão/genética , Miosite de Corpos de Inclusão/patologia , Oxirredução
13.
J Neurochem ; 124(1): 4-14, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22862561

RESUMO

Apolipoprotein E (APOE) genotype affects outcomes of Alzheimer's disease and other conditions of brain damage. Using APOE knock-in mice, we have previously shown that APOE-ε4 Targeted Replacement (TR) mice have fewer dendritic spines and reduced branching in cortical neurons. As dendritic spines are post-synaptic sites of excitatory neurotransmission, we used APOE TR mice to examine whether APOE genotype affected the various elements of the glutamate-glutamine cycle. We found that levels of glutamine synthetase and glutamate uptake transporters were unchanged among the APOE genotypes. However, compared with APOE-ε3 TR mice, APOE-ε4 TR mice had decreased glutaminase levels (18%, p < 0.05), suggesting decreased conversion of glutamine to glutamate. APOE-ε4 TR mice also had increased levels of the vesicular glutamate transporter 1 (20%, p < 0.05), suggesting that APOE genotype affects pre-synaptic terminal composition. To address whether these changes affected normal neurotransmission, we examined the production and metabolism of glutamate and glutamine at 4-5 months and 1 year. Using high-frequency (13)C/(1)H nuclear magnetic resonance spectroscopy, we found that APOE-ε4 TR mice have decreased production of glutamate and increased levels of glutamine. These factors may contribute to the increased risk of neurodegeneration associated with APOE-ε4, and also act as surrogate markers for Alzheimer's disease risk.


Assuntos
Apolipoproteínas E/genética , Encéfalo/citologia , Regulação da Expressão Gênica/genética , Ácido Glutâmico/metabolismo , Terminações Pré-Sinápticas/metabolismo , Animais , Apolipoproteína E2/genética , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Isótopos de Carbono/metabolismo , Glutaminase/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Trítio/metabolismo , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo
14.
Hum Mol Genet ; 20(11): 2091-102, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21378096

RESUMO

Intraneuronal amyloid-ß (Aß) may contribute to extracellular plaque deposition, the characteristic pathology of Alzheimer's disease (AD). The E3-ubiquitin ligase parkin ubiquitinates intracellular proteins and induces mitophagy. We previously demonstrated that parkin reduces Aß levels in lentiviral models of intracellular Aß. Here we used a triple transgenic AD (3xTg-AD) mouse, which over-expresses APP(Swe), Tau(P301L) and harbor the PS1(M146V) knock-in mutation and found that lentiviral parkin ubiquitinated intracellular Aß in vivo, stimulated beclin-dependent molecular cascade of autophagy and facilitated clearance of vesicles containing debris and defective mitochondria. Parkin expression decreased intracellular Aß levels and extracellular plaque deposition. Parkin expression also attenuated caspase activity, prevented mitochondrial dysfunction and oxidative stress and restored neurotransmitter synthesis. Restoration of glutamate synthesis, which was independent of glial-neuronal recycling, depended on mitochondrial activity and led to an increase in γ-amino butyric acid levels. These data indicate that parkin may be used as an alternative strategy to reduce Aß levels and enhance autophagic clearance of Aß-induced defects in AD. Parkin-mediated clearance of ubiquitinated Aß may act in parallel with autophagy to clear molecular debris and defective mitochondria and restore neurotransmitter balance.


Assuntos
Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia , Mitocôndrias/patologia , Ubiquitina-Proteína Ligases/metabolismo , Doença de Alzheimer/metabolismo , Animais , Proteína Beclina-1 , Western Blotting , Caspase 3/metabolismo , Caspase 9/metabolismo , Córtex Cerebral/fisiopatologia , Córtex Cerebral/ultraestrutura , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Técnicas de Introdução de Genes , Vetores Genéticos , Imuno-Histoquímica/métodos , Lentivirus/metabolismo , Espectrometria de Massas , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Mutação , Neurotransmissores/análise , Neurotransmissores/metabolismo , Estresse Oxidativo , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
15.
Mol Cell Neurosci ; 49(1): 44-53, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21945393

RESUMO

Neurodegeneration involves multiple pathogenic proteins, including Tau, Aß, TDP-43 and α-Synuclein, but there is little information how these pathogenic proteins interact. We cloned human wild type 4 repeat Tau (Tau(wt)) and mutant Tau(P301L) into a lentivirus and performed stereotaxic injection into the rat motor cortex to examine Tau modification, neuro-inflammation and changes of other proteins associated with neurodegeneration. Tau(P301L) was associated with more phosphorylation of Tau, including Thr 181 and Ser 262 residues and resulted in more aggregation. Both forms of Tau expression increased glycogen synthase kinase-3 (GSK-3) activity, polo-like kinase-2 (PLK2) levels and decreased protein phosphatase activity, but had no effects on casein kinase-1 (CK1). No changes were observed in glial fibrillary acidic protein (GFAP) staining with either Tau(wt) or Tau(P301L), but both caused microglial changes and higher interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) levels. Tau(wt) and Tau(P301L) increased the levels of endogenous α-Synuclein, but not ß-amyloid precursor protein (ßAPP) or Tar-DNA binding protein (TDP-43). The levels of phosphorylated Ser-129 α-Synuclein (p-Ser129) were also increased with Tau(wt) and Tau(P301L) expressing animals. These data suggest that Tau(wt) and Tau(P301L) alter kinase activities, but they differentially induce inflammation, Tau modification and α-Synuclein phosphorylation. This change of α-Synuclein in Tau gene transfer models suggests that Tau pathology may lead to α-Synuclein modification in neurodegenerative diseases.


Assuntos
Tauopatias/genética , alfa-Sinucleína/metabolismo , Proteínas tau/genética , Animais , Proteínas de Ligação a DNA/metabolismo , Técnicas de Transferência de Genes , Quinase 3 da Glicogênio Sintase/metabolismo , Inflamação/genética , Inflamação/metabolismo , Interleucina-6/metabolismo , Lentivirus/genética , Modelos Animais , Córtex Motor/metabolismo , Fosforilação/fisiologia , Ratos , Ratos Sprague-Dawley , Tauopatias/metabolismo , alfa-Sinucleína/genética , Proteínas tau/metabolismo
16.
J Biol Chem ; 285(10): 7440-6, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20071340

RESUMO

Alzheimer disease is characterized by extracellular beta-amyloid (Abeta) plaques and intracellular inclusions containing neurofibrillary tangles of phospho-Tau and intraneuronal Abeta associated with neuronal cell death. We generated a novel gene transfer animal model using lentiviral Abeta(1-42) that resulted in intracellular but not extracellular Abeta accumulations in the targeted rat primary motor cortex. Expression of intracellular Abeta(1-42) led to pathological changes seen in human Alzheimer disease brains, including cell death, inflammatory signs, activation of two Tau kinases, and Tau hyperphosphorylation. Promoting clearance of lentiviral Abeta(1-42) reversed these effects, demonstrating that intraneuronal Abeta(1-42) is a toxic peptide that lies upstream of Tau modification. These studies reveal the role of intracellular Abeta(1-42) in a novel gene transfer animal model, which is a useful tool to study intraneuronal Abeta(1-42)-induced pathology in the absence of extracellular plaques. Targeted delivery of Abeta will allow speedy delineation of pathological mechanisms associated with specific neurodegenerative lesions.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Amiloide/metabolismo , Morte Celular/fisiologia , Técnicas de Transferência de Genes , Neurônios/metabolismo , Neurônios/patologia , Fragmentos de Peptídeos/metabolismo , Proteínas tau/metabolismo , Amiloide/genética , Peptídeos beta-Amiloides/genética , Animais , Sobrevivência Celular , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Quinase 5 Dependente de Ciclina/metabolismo , Modelos Animais de Doenças , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Inflamação/metabolismo , Camundongos , Neurônios/citologia , Fragmentos de Peptídeos/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Proteínas tau/genética
17.
Hum Mol Genet ; 18(17): 3206-16, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19483198

RESUMO

Alzheimer's disease and Parkinson's disease are common neurodegenerative diseases that may share some underlying mechanisms of pathogenesis. Abeta(1-42) fragments are found intracellularly, and extracellularly as amyloid plaques, in Alzheimer's disease and in dementia with Lewy Bodies. Parkin is an E3-ubiquitin ligase involved in proteasomal degradation of intracellular proteins. Mutations in parkin, which result in loss of parkin function, lead to early onset Parkinsonism. Here we tested whether the ubiquitin ligase activity of parkin could lead to reduction in intracellular human Abeta(1-42). Lentiviral constructs encoding either human parkin or human Abeta(1-42) were used to infect M17 neuroblastoma cells. Parkin expression resulted in reduction of intracellular human Abeta(1-42) levels and protected against its toxicity in M17 cells. Co-injection of lentiviral constructs into control rat primary motor cortex demonstrated that parkin co-expression reduced human Abeta(1-42) levels and Abeta(1-42)-induced neuronal degeneration in vivo. Parkin increased proteasomal activity, and proteasomal inhibition blocked the effects of parkin on reducing Abeta(1-42) levels. Incubation of Abeta(1-42) cell lysates with ubiquitin, in the presence of parkin, demonstrated the generation of Abeta-ubiquitin complexes. These data indicate that parkin promotes ubiquitination and proteasomal degradation of intracellular Abeta(1-42) and demonstrate a protective effect in neurodegenerative diseases with Abeta deposits.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Doenças Neurodegenerativas/metabolismo , Fragmentos de Peptídeos/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Peptídeos beta-Amiloides/genética , Linhagem Celular , Humanos , Mutação , Doenças Neurodegenerativas/genética , Fragmentos de Peptídeos/genética , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
18.
J Neurosci Res ; 88(1): 167-78, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19610108

RESUMO

The significance of intracellular beta-amyloid (Abeta(42)) accumulation is increasingly recognized in Alzheimer's disease (AD) pathogenesis. Abeta removal mechanisms that have attracted attention include IDE/neprilysin degradation and antibody-mediated uptake by immune cells. However, the role of the ubiquitin-proteasome system (UPS) in the disposal of cellular Abeta has not been fully explored. The E3 ubiquitin ligase Parkin targets several proteins for UPS degradation, and Parkin mutations are the major cause of autosomal recessive Parkinson's disease. We tested whether Parkin has cross-function to target misfolded proteins in AD for proteasome-dependent clearance in SH-SY5Y and primary neuronal cells. Wild-type Parkin greatly decreased steady-state levels of intracellular Abeta(42), an action abrogated by proteasome inhibitors. Intracellular Abeta(42) accumulation decreased cell viability and proteasome activity. Accordingly, Parkin reversed both effects. Changes in mitochondrial ATP production from Abeta or Parkin did not account for their effects on the proteasome. Parkin knock-down led to accumulation of Abeta. In AD brain, Parkin was found to interact with Abeta and its levels were reduced. Thus, Parkin is cytoprotective, partially by increasing the removal of cellular Abeta through a proteasome-dependent pathway.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Citoproteção/fisiologia , Fragmentos de Peptídeos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/fisiologia , Trifosfato de Adenosina/metabolismo , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Animais , Western Blotting , Linhagem Celular , Sobrevivência Celular/genética , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Imunofluorescência , Humanos , Imunoprecipitação , Camundongos , Camundongos Transgênicos , Mutação , Neurônios/citologia , Neurônios/metabolismo , Fragmentos de Peptídeos/genética , Complexo de Endopeptidases do Proteassoma/genética , Transfecção , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética
19.
J Mol Neurosci ; 37(1): 25-36, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18561034

RESUMO

Changes in tau (tau) metabolism comprise important pathological landmarks in the tauopathies with parkinsonism as well as Parkinson's disease and Alzheimer's disease. Mutations in the parkin gene are associated with Parkinson's disease. Deposits of amyloid proteins, including Abeta and alpha-synuclein coexist in the brains of patients with dementia with Lewy bodies; however, it is not known how either of them interacts with tau to provoke neurofibrillary tangle formation across the tauopathies. Here, we show a role for parkin against tau pathology in the presence of intracellular Abeta or alpha-synuclein. Parkin attenuates four-repeat human tau, but not mutant P301L, hyperphosphorylation in the presence of intracellular Abeta(1-42), or alpha-synuclein and decreases GSK-3beta activity in amyloid-stressed M17 human neuroblastoma cells. These data suggest that parkin may counteract the alteration of tau metabolism in certain neurodegenerative diseases with tau cytopathy and parkinsonism.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/metabolismo , Tauopatias/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Linhagem Celular Tumoral , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Neuroblastoma , Doença de Parkinson/metabolismo , Fosforilação/fisiologia
20.
Drugs R D ; 19(2): 149-166, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30919310

RESUMO

BACKGROUND AND OBJECTIVES: Inhibition of Abelson (Abl) tyrosine kinase as a therapeutic target has been gaining attention in neurodegeneration. Post-mortem Alzheimer's and Parkinson's disease brains show that the levels of several other tyrosine kinases, including Discoidin Domain Receptors (DDR1/2) are elevated. Knockdown of these tyrosine kinases with shRNA reduces neurotoxic proteins, including alpha-synuclein, beta-amyloid and tau. METHODS: Direct profiling of the pharmacokinetics of multi-kinase inhibitors Nilotinib, Bosutinib, Bafetinib, Radotinib and LCB-03-0110 shows differential levels of brain penetration but the ability of these agents to reduce toxic proteins is independent of brain concentration and selectivity to Abl. RESULTS: Our results indicate that the effective dose of Nilotinib has the lowest plasma:brain ratio (1%) followed by Bosutinib and Radotinib (5%), Bafetinib (12%) and LCB-03-0110 (12%). However, similar doses of multi-kinase Abl/DDR inhibitor Nilotinib, DDR/Src inhibitor LCB-03-0110 and Abl/Src inhibitor Bosutinib were much more effective than the more selective Abl inhibitors Radotinib and Bafetinib. Taken together, these data suggest that a multi-kinase target that includes Abl and other tyrosine kinases (DDRs, and Src) may offer more advantages alleviating neurodegenerative pathologies than the absolute CNS drug concentration and selectivity to Abl. CONCLUSION: DDRs and Src are other potential co-targets with Abl in neurodegeneration.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Parkinson/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Barreira Hematoencefálica/metabolismo , Linhagem Celular Tumoral , Receptores com Domínio Discoidina/antagonistas & inibidores , Receptores com Domínio Discoidina/genética , Receptores com Domínio Discoidina/metabolismo , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Hipocampo/patologia , Humanos , Masculino , Mesencéfalo/patologia , Camundongos , Camundongos Transgênicos , Doença de Parkinson/genética , Doença de Parkinson/patologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-abl/genética , Proteínas Proto-Oncogênicas c-abl/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/genética , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa