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










Base de dados
Intervalo de ano de publicação
1.
Circ Res ; 127(4): 502-518, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32366200

RESUMO

RATIONALE: The ubiquitin-proteasome system (UPS) and the autophagic-lysosomal pathway are pivotal to proteostasis. Targeting these pathways is emerging as an attractive strategy for treating cancer. However, a significant proportion of patients who receive a proteasome inhibitor-containing regime show cardiotoxicity. Moreover, UPS and autophagic-lysosomal pathway defects are implicated in cardiac pathogenesis. Hence, a better understanding of the cross-talk between the 2 catabolic pathways will help advance cardiac pathophysiology and medicine. OBJECTIVE: Systemic proteasome inhibition (PSMI) was shown to increase p62/SQSTM1 expression and induce myocardial macroautophagy. Here we investigate how proteasome malfunction activates cardiac autophagic-lysosomal pathway. METHODS AND RESULTS: Myocardial macroautophagy, TFEB (transcription factor EB) expression and activity, and p62 expression were markedly increased in mice with either cardiomyocyte-restricted ablation of Psmc1 (an essential proteasome subunit gene) or pharmacological PSMI. In cultured cardiomyocytes, PSMI-induced increases in TFEB activation and p62 expression were blunted by pharmacological and genetic calcineurin inhibition and by siRNA-mediated Molcn1 silencing. PSMI induced remarkable increases in myocardial autophagic flux in wild type mice but not p62 null (p62-KO) mice. Bortezomib-induced left ventricular wall thickening and diastolic malfunction was exacerbated by p62 deficiency. In cultured cardiomyocytes from wild type mice but not p62-KO mice, PSMI induced increases in LC3-II flux and the lysosomal removal of ubiquitinated proteins. Myocardial TFEB activation by PSMI as reflected by TFEB nuclear localization and target gene expression was strikingly less in p62-KO mice compared with wild type mice. CONCLUSIONS: (1) The activation of cardiac macroautophagy by proteasomal malfunction is mediated by the Mocln1-calcineurin-TFEB-p62 pathway; (2) p62 unexpectedly exerts a feed-forward effect on TFEB activation by proteasome malfunction; and (3) targeting the Mcoln1 (mucolipin1)-calcineurin-TFEB-p62 pathway may provide new means to intervene cardiac autophagic-lysosomal pathway activation during proteasome malfunction.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Calcineurina/metabolismo , Macroautofagia/fisiologia , Complexo de Endopeptidases do Proteassoma/fisiologia , ATPases Associadas a Diversas Atividades Celulares/genética , Animais , Antineoplásicos/farmacologia , Bortezomib/farmacologia , Calcineurina/genética , Inibidores de Calcineurina , Hipertrofia Ventricular Esquerda/induzido quimicamente , Lisossomos/metabolismo , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Inibidores de Proteassoma , Proteostase , RNA Interferente Pequeno , Ratos , Proteína Sequestossoma-1/metabolismo , Transdução de Sinais/fisiologia , Canais de Potencial de Receptor Transitório/metabolismo , Ubiquitina/metabolismo , Regulação para Cima
2.
Redox Biol ; 20: 167-181, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30336354

RESUMO

Monoamine oxidases (MAOs) are located on the outer mitochondrial membrane and are drug targets for the treatment of neurological disorders. MAOs control the levels of neurotransmitters in the brain via oxidative deamination and contribute to reactive oxygen species (ROS) generation through their catalytic by-product H2O2. Increased ROS levels may modulate mitochondrial function and mitochondrial dysfunction is implicated in a vast array of disorders. However, the downstream effects of MAO-A mediated ROS production in a neuronal model has not been previously investigated. In this study, using MAO-A overexpressing neuroblastoma cells, we demonstrate that higher levels of MAO-A protein/activity results in increased basal ROS levels with associated increase in protein oxidation. Increased MAO-A levels result in increased Lysine-63 linked ubiquitination of mitochondrial proteins and promotes autophagy through Bcl-2 phosphorylation. Furthermore, ROS generated locally on the mitochondrial outer membrane by MAO-A promotes phosphorylation of dynamin-1-like protein, leading to mitochondrial fragmentation and clearance without complete loss of mitochondrial membrane potential. Cellular ATP levels are maintained following MAO-A overexpression and complex IV activity/protein levels increased, revealing a close relationship between MAO-A levels and mitochondrial function. Finally, the downstream effects of increased MAO-A levels are dependent on the availability of amine substrates and in the presence of exogenous substrate, cell viability is dramatically reduced. This study shows for the first time that MAO-A generated ROS is involved in quality control signalling, and increase in MAO-A protein levels leads to a protective cellular response in order to mediate removal of damaged macromolecules/organelles, but substrate availability may ultimately determine cell fate. The latter is particularly important in conditions such as Parkinson's disease, where a dopamine precursor is used to treat disease symptoms and highlights that the fate of MAO-A containing dopaminergic neurons may depend on both MAO-A levels and catecholamine substrate availability.


Assuntos
Autofagia , Monoaminoxidase/metabolismo , Neuroblastoma/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Imunofluorescência , Expressão Gênica , Humanos , Imuno-Histoquímica , Mitocôndrias/genética , Mitocôndrias/metabolismo , Modelos Biológicos , Monoaminoxidase/genética , Neuroblastoma/genética , Oxirredução , Estresse Oxidativo , Fosforilação , Proteoma , Proteômica/métodos , Espécies Reativas de Oxigênio/metabolismo
3.
Sci Rep ; 8(1): 4833, 2018 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-29555943

RESUMO

Metabolite profiling is an important tool that may better capture the multiple features of neurodegeneration. With the considerable parallels between mouse and human metabolism, the use of metabolomics in mouse models with neurodegenerative pathology provides mechanistic insight and ready translation into aspects of human disease. Using 400 MHz nuclear magnetic resonance spectroscopy we have carried out a temporal region-specific investigation of the metabolome of neuron-specific 26S proteasome knockout mice characterised by progressive neurodegeneration and Lewy-like inclusion formation in the forebrain. An early significant decrease in N-acetyl aspartate revealed evidence of neuronal dysfunction before cell death that may be associated with changes in brain neuroenergetics, underpinning the use of this metabolite to track neuronal health. Importantly, we show early and extensive activation of astrocytes and microglia in response to targeted neuronal dysfunction in this context, but only late changes in myo-inositol; the best established glial cell marker in magnetic resonance spectroscopy studies, supporting recent evidence that additional early neuroinflammatory markers are needed. Our results extend the limited understanding of metabolite changes associated with gliosis and provide evidence that changes in glutamate homeostasis and lactate may correlate with astrocyte activation and have biomarker potential for tracking neuroinflammation.


Assuntos
Gliose/metabolismo , Gliose/patologia , Metabolômica , Neurônios/metabolismo , Prosencéfalo/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Camundongos
4.
Cell Death Dis ; 8(1): e2531, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-28055010

RESUMO

The ubiquitin-proteasome system (UPS) and macroautophagy (autophagy) are central to normal proteostasis and interdependent in that autophagy is known to compensate for the UPS to alleviate ensuing proteotoxic stress that impairs cell function. UPS and autophagy dysfunctions are believed to have a major role in the pathomechanisms of neurodegenerative disease. Here we show that continued 26S proteasome dysfunction in mouse brain cortical neurons causes paranuclear accumulation of fragmented dysfunctional mitochondria, associated with earlier recruitment of Parkin and lysine 48-linked ubiquitination of mitochondrial outer membrane (MOM) proteins, including Mitofusin-2. Early events also include phosphorylation of p62/SQSTM1 (p62) and increased optineurin, as well as autophagosomal LC3B and removal of some mitochondria, supporting the induction of selective autophagy. Inhibition of the degradation of ubiquitinated MOM proteins with continued 26S proteasome dysfunction at later stages may impede efficient mitophagy. However, continued 26S proteasome dysfunction also decreases the levels of essential autophagy proteins ATG9 and LC3B, which is characterised by decreases in their gene expression, ultimately leading to impaired autophagy. Intriguingly, serine 351 phosphorylation of p62 did not enhance its binding to Keap1 or stabilise the nuclear factor erythroid 2-related factor 2 (Nrf2) transcription factor in this neuronal context. Nrf2 protein levels were markedly decreased despite transcriptional activation of the Nrf2 gene. Our study reveals novel insights into the interplay between the UPS and autophagy in neurons and is imperative to understanding neurodegenerative disease where long-term proteasome inhibition has been implicated.


Assuntos
Autofagia/genética , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Mitocôndrias/genética , Mitofagia/genética , Fator 2 Relacionado a NF-E2/genética , Proteína Sequestossoma-1/genética , Animais , Proteínas de Ciclo Celular , Proteínas do Olho/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Humanos , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Proteínas de Membrana Transportadoras , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Fator 2 Relacionado a NF-E2/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Fosforilação , Complexo de Endopeptidases do Proteassoma/genética , Proteína Sequestossoma-1/metabolismo , Ubiquitina , Ubiquitina-Proteína Ligases/metabolismo
5.
J Biol Chem ; 289(36): 24944-55, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-25049227

RESUMO

The ubiquitin-proteasome system and autophagy are crucially important for proteostasis in cells. These pathways are interdependent, and dysfunction in either pathway causes accumulation of ubiquitin-positive aggregates, a hallmark of human pathological conditions. To elucidate in vivo compensatory action(s) against proteasomal dysfunction, we developed mice with reduced proteasome activity in their livers. The mutant mice exhibited severe liver damage, accompanied by formation of aggregates positive for ubiquitin and p62/Sqstm1, an adaptor protein for both selective autophagy and the anti-oxidative Keap1-Nrf2 pathway. These aggregates were selectively entrapped by autophagosomes, and pathological features of livers with impaired proteasome activity were exacerbated by simultaneous suppression of autophagy. In contrast, concomitant loss of p62/Sqstm1 had no apparent effect on the liver pathology though p62/Sqstm1 was indispensable for the aggregates formation. Furthermore, defective proteasome function led to transcriptional activation of the Nrf2, which served as a physiological adaptation. Our in vivo data suggest that cells contain networks of cellular defense mechanisms against defective proteostasis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autofagia , Proteínas do Citoesqueleto/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas do Citoesqueleto/genética , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Immunoblotting , Proteína 1 Associada a ECH Semelhante a Kelch , Fígado/metabolismo , Fígado/patologia , Fígado/ultraestrutura , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Microscopia Imunoeletrônica , Fator 2 Relacionado a NF-E2/genética , Fagossomos/genética , Fagossomos/metabolismo , Fosforilação , Complexo de Endopeptidases do Proteassoma/genética , Proteína Sequestossoma-1 , Fatores de Tempo , Ubiquitina/metabolismo
6.
PLoS One ; 9(4): e93586, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24699688

RESUMO

Chronic excessive alcohol intoxications evoke cumulative damage to tissues and organs. We examined prefrontal cortex (Brodmann's area (BA) 9) from 20 human alcoholics and 20 age, gender, and postmortem delay matched control subjects. H & E staining and light microscopy of prefrontal cortex tissue revealed a reduction in the levels of cytoskeleton surrounding the nuclei of cortical and subcortical neurons, and a disruption of subcortical neuron patterning in alcoholic subjects. BA 9 tissue homogenisation and one dimensional polyacrylamide gel electrophoresis (PAGE) proteomics of cytosolic proteins identified dramatic reductions in the protein levels of spectrin ß II, and α- and ß-tubulins in alcoholics, and these were validated and quantitated by Western blotting. We detected a significant increase in α-tubulin acetylation in alcoholics, a non-significant increase in isoaspartate protein damage, but a significant increase in protein isoaspartyl methyltransferase protein levels, the enzyme that triggers isoaspartate damage repair in vivo. There was also a significant reduction in proteasome activity in alcoholics. One dimensional PAGE of membrane-enriched fractions detected a reduction in ß-spectrin protein levels, and a significant increase in transmembranous α3 (catalytic) subunit of the Na+,K+-ATPase in alcoholic subjects. However, control subjects retained stable oligomeric forms of α-subunit that were diminished in alcoholics. In alcoholics, significant loss of cytosolic α- and ß-tubulins were also seen in caudate nucleus, hippocampus and cerebellum, but to different levels, indicative of brain regional susceptibility to alcohol-related damage. Collectively, these protein changes provide a molecular basis for some of the neuronal and behavioural abnormalities attributed to alcoholics.


Assuntos
Etanol/toxicidade , Córtex Pré-Frontal/efeitos dos fármacos , Adulto , Idoso , Western Blotting , Eletroforese em Gel de Poliacrilamida , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/metabolismo , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia
7.
Biochim Biophys Acta ; 1832(12): 1930-8, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23851049

RESUMO

Neurodegenerative diseases are characterized by progressive degeneration of selective neurones in the nervous system, but the underlying mechanisms involved in neuroprotection and neurodegeneration remain unclear. Dysfunction of the ubiquitin proteasome system is one of the proposed hypotheses for the cause and progression of neuronal loss. We have performed quantitative two-dimensional fluorescence difference in-gel electrophoresis combined with peptide mass fingerprinting to reveal proteome changes associated with neurodegeneration following 26S proteasomal depletion in mouse forebrain neurones. Differentially expressed proteins were validated by Western blotting, biochemical assays and immunohistochemistry. Of significance was increased expression of the antioxidant enzyme peroxiredoxin 6 (PRDX6) in astrocytes, associated with oxidative stress. Interestingly, PRDX6 is a bifunctional enzyme with antioxidant peroxidase and phospholipase A2 (PLA2) activities. The PLA2 activity of PRDX6 was also increased following 26S proteasomal depletion and may be involved in neuroprotective or neurodegenerative mechanisms. This is the first in vivo report of oxidative stress caused directly by neuronal proteasome dysfunction in the mammalian brain. The results contribute to understanding neuronal-glial interactions in disease pathogenesis, provide an in vivo link between prominent disease hypotheses and importantly, are of relevance to a heterogeneous spectrum of neurodegenerative diseases.


Assuntos
Astrócitos/metabolismo , Degeneração Neural/metabolismo , Neurônios/metabolismo , Estresse Oxidativo , Prosencéfalo/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Astrócitos/patologia , Western Blotting , Eletroforese em Gel Bidimensional , Técnicas Imunoenzimáticas , Peroxidação de Lipídeos , Camundongos , Degeneração Neural/patologia , Neurônios/patologia , Fosfolipases A2/metabolismo , Prosencéfalo/patologia , Complexo de Endopeptidases do Proteassoma/química , Espécies Reativas de Oxigênio/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
8.
PLoS One ; 8(1): e54711, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23382946

RESUMO

Parkinson's disease (PD) is characterized by the progressive degeneration of substantia nigra pars compacta (SNpc) dopaminergic neurones and the formation of Lewy bodies (LB) in a proportion of the remaining neurones. α-synuclein is the main component of LB, but the pathological mechanisms that lead to neurodegeneration associated with LB formation remain unclear. Three pivotal elements have emerged in the development of PD: α-synuclein, mitochondria and protein degradation systems. We previously reported a unique model, created by conditional genetic depletion of 26S proteasomes in the SNpc of mice, which mechanistically links these three elements with the neuropathology of PD: progressive neurodegeneration and intraneuronal inclusion formation. Using this model, we tested the hypothesis that α-synuclein was essential for the formation of inclusions and neurodegeneration caused by 26S proteasomal depletion. We found that both of these processes were independent of α-synuclein. This provides an important insight into the relationship between the proteasome, α-synuclein, inclusion formation and neurodegeneration. We also show that the autophagy-lysosomal pathway is not activated in 26S proteasome-depleted neurones. This leads us to suggest that the paranuclear accumulation of mitochondria in inclusions in our model may reflect a role for the ubiquitin proteasome system in mitochondrial homeostasis and that neurodegeneration may be mediated through mitochondrial factors linked to inclusion biogenesis.


Assuntos
Encéfalo/metabolismo , Corpos de Inclusão , Neurônios/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , alfa-Sinucleína/metabolismo , Animais , Autofagia , Encéfalo/patologia , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Corpos de Inclusão/ultraestrutura , Corpos de Lewy , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Neurônios/patologia , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Transdução de Sinais , Substância Negra/metabolismo , Substância Negra/patologia , alfa-Sinucleína/genética
9.
Neurosci Lett ; 521(2): 130-5, 2012 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-22677101

RESUMO

The ubiquitin proteasome system (UPS) is a fundamental cellular pathway, degrading most unwanted intracellular soluble proteins. Dysfunction of the UPS has been associated with normal aging as well as various age-related pathological conditions, including chronic human neurodegenerative diseases such as Alzheimer's and Parkinson's diseases, leading to a significant interest in the involvement of this degradative system in neurones. We previously reported that the 26S proteasome was essential for neuronal homeostasis and survival in mouse brains following conditional genetic homozygous knockout of a key subunit of the multi-meric 26S proteasome (19S ATPase Psmc1). Here, we investigated the effects of Psmc1 heterozygosity in the mouse brain and primary mouse embryonic fibroblasts. Neuropathologically and biochemically, Psmc1 heterozygous (Psmc1(+/-)) knockout mice were indistinguishable from wild-type mice. However, we report a novel age-related accumulation of intraneuronal lysine 48-specific polyubiquitin-positive granular staining in both wild-type and heterozygous Psmc1 knockout mouse brain. In Psmc1(+/-) MEFs, we found a significant decrease in PSMC1 levels, altered 26S proteasome assembly and a notable G2/M cell cycle arrest that was not associated with an increase in the cell cycle regulatory protein p21. The disturbance in cell cycle progression may be responsible for the growth inhibitory effects in Psmc1(+/-) MEFs.


Assuntos
Adenosina Trifosfatases/metabolismo , Encéfalo/metabolismo , Fibroblastos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Adenosina Trifosfatases/genética , Animais , Encéfalo/patologia , Células Cultivadas , Fibroblastos/citologia , Pontos de Checagem da Fase G2 do Ciclo Celular , Heterozigoto , Pontos de Checagem da Fase M do Ciclo Celular , Camundongos , Camundongos Knockout , Cultura Primária de Células , Complexo de Endopeptidases do Proteassoma/genética
10.
J Alzheimers Dis ; 27(3): 615-25, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21860090

RESUMO

Collapsin response mediator protein 2 (CRMP2) is an abundant brain-enriched protein that regulates neurite outgrowth. It is phosphorylated by Cdk5 and GSK3, and these modifications are abnormally high in the brains of Alzheimer's disease (AD) patients. Increased phosphorylation of CRMP2 is also apparent in mouse models of AD that express mutated AßPP and PSEN1, but not AßPP or tau alone, where it is detectable before the appearance of amyloid plaques and neurofibrillary tangles, suggesting it is an early event in AD pathogenesis. Here, we have extended these observations by showing that CRMP2 is not hyperphosphorylated in mice overexpressing mutated PSEN1 alone, or in cultured neurons treated with soluble, oligomeric Aß42 peptide. Similarly, CRMP2 phosphorylation was not increased in a mouse model of severe neurodegeneration (PMSC-1 knockout) or in cultured neurons subjected to neurotoxic concentrations of NMDA or staurosporine. Most interestingly, CRMP2 phosphorylation was not increased in frontal cortex from patients with frontotemporal lobar degeneration associated with mutations in MAPT or with Pick bodies. Together, these observations are consistent with the hypothesis that abnormal phosphorylation of CRMP2 is specific to AD and occurs downstream of excessive processing of AßPP, but that neither excessive Aß42 peptide nor neurotoxicity alone are sufficient to promote hyperphosphorylation.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/genética , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Doenças Neurodegenerativas/diagnóstico , Doenças Neurodegenerativas/genética , Fosforilação/genética , Ratos Sprague-Dawley , Ovinos , Tauopatias/diagnóstico , Tauopatias/metabolismo
11.
Biochem Soc Trans ; 39(4): 917-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21787323

RESUMO

Chronic neurodegenerative disease is characterized by extensive regional loss of neurons in the brain and neuropathological hallmarks in surviving neurones. Genetic modelling by overexpression of hallmark proteins does not produce extensive neurodegeneration, whereas genetic deletion of neuronal 26S proteasomes does, as well as some hallmarks of human disease.


Assuntos
Modelos Animais de Doenças , Doenças Neurodegenerativas/patologia , Neurônios/patologia , Animais , Morte Celular , Doença Crônica , Humanos , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Placa Amiloide/metabolismo
12.
Neurosci Lett ; 491(1): 44-7, 2011 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-21215295

RESUMO

A generality has been that polyubiquitin chain linkage can differentially address proteins for various physiological processes. 26S proteasomal degradation is the most established function of ubiquitin signalling, classically linked to Lys48 polyubiquitin chains. The other well-characterised polyubiquitin linkage, via Lys63, mediates nonproteolytic functions. However, there are five other lysine residues and ubiquitin's amino terminus which can participate in polyubiquitination. Our 26S proteasome knockout mouse provides a unique opportunity to comprehensively investigate the ubiquitin signals in their physiological context in neurones following genetic inhibition of the proteasome, using quantitative mass spectrometry of ubiquitin linkage-specific signature peptides. We provide the first evidence for diverse polyubiquitin chains in mammalian neurones in vivo and show that polyubiquitin linked via Lys6, Lys11, Lys29 and Lys48, but not Lys63, accumulates upon 26S proteasome dysfunction. This adaptable nature of ubiquitin signals for proteasomal targeting could reflect the extensive cellular processes which are regulated by proteasome proteolysis and/or may involve specific ubiquitin linkage preferences for subsets of proteins in mammalian neurones. Our molecular pathological findings make a significant contribution to the understanding of ubiquitin signalling in ubiquitin-proteasome function.


Assuntos
Neurônios/metabolismo , Poliubiquitina/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Adaptação Fisiológica/fisiologia , Sequência de Aminoácidos , Animais , Lisina/metabolismo , Espectrometria de Massas/métodos , Camundongos , Camundongos Knockout , Peptídeos/metabolismo , Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/deficiência , Estrutura Secundária de Proteína , Transdução de Sinais/fisiologia , Ubiquitinação/fisiologia
13.
Nat Rev Drug Discov ; 10(1): 29-46, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21151032

RESUMO

The ubiquitin-proteasome system (UPS) and ubiquitin-like protein (UBL) conjugation pathways are integral to cellular protein homeostasis. The growing recognition of the fundamental importance of these pathways to normal cell function and in disease has prompted an in-depth search for small-molecule inhibitors that selectively block the function of these pathways. However, our limited understanding of the molecular mechanisms and biological consequences of UBL conjugation is a significant hurdle to identifying drug-like inhibitors of enzyme targets within these pathways. Here, we highlight recent advances in understanding the role of some of these enzymes and how these new insights may be the key to developing novel therapeutics for diseases including immuno-inflammatory disorders, cancer, infectious diseases, cardiovascular disease and neurodegenerative disorders.


Assuntos
Sistemas de Liberação de Medicamentos , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Animais , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Humanos
14.
Trends Cell Biol ; 20(7): 391-401, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20427185

RESUMO

The 26S proteasome is a large multiprotein complex involved in the regulated degradation of ubiquitinated proteins in the cell. The 26S proteasome has been shown to control an increasing number of essential biochemical mechanisms of the cellular lifecycle including DNA synthesis, repair, transcription, translation, and cell signal transduction. Concurrently, it is increasingly seen that malfunction of the ubiquitin proteasome system contributes to the pathogenesis of disease. The recent identification of four molecular chaperones, in addition to five previously identified chaperones, have provided mechanistic insight into how this cellular megastructure is assembled in the cell. These data, together with new insights into the structure and function of the proteasome, provide a much better understanding of this complex protease.


Assuntos
Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Fenômenos Fisiológicos Celulares , Humanos , Chaperonas Moleculares/análise , Proteínas Ubiquitinadas/metabolismo
15.
Neurosci Lett ; 460(3): 205-8, 2009 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-19500650

RESUMO

The major human neurodegenerative diseases are characterised by ubiquitin-positive intraneuronal inclusions, however the precise nature of the ubiquitin modifications in these structures is unclear. Using a monoclonal antibody specific for Lys63-linked polyubiquitin we have performed the first immunohistochemical analysis of linkage-specific ubiquitination in vivo associated with neurodegeneration. Immunoreactivity was detected within the pathological lesions of Alzheimer's, Huntington's and Parkinson's disease brains, although staining of Lewy bodies in the substantia nigra in Parkinson's disease was rare, indicating a selective involvement of Lys63-linked polyubiquitin in inclusion biogenesis in this disorder. Immunoreactivity was also a feature in neurons of proteasome-depleted mice, suggesting a proteasomal contribution to the degradation of Lys63-linked polyubiquitinated proteins in vivo.


Assuntos
Lisina/metabolismo , Doenças Neurodegenerativas/metabolismo , Poliubiquitina/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Imuno-Histoquímica , Corpos de Lewy/metabolismo , Camundongos , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo
16.
Autophagy ; 5(2): 224-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19077533

RESUMO

In the past twenty years, evidence has accumulated to show that ubiquitinated proteins are a consistent feature of the intraneuronal protein aggregates (inclusions) that characterize chronic neurodegenerative disease. These findings may indicate that age-related dysfunction of the 26S proteasome may be central to disease pathogenesis. The aggregate-prone proteins can also be eliminated by autophagy. We have used the Cre-recombinase/loxP genetic approach to ablate the proteasomal Psmc1 ATPase gene and deplete 26S proteasomes in neurons in different regions of the brain to mimic neurodegeneration. Deletion of the gene in dopaminergic neurons in the substantia nigra generates a new model of Parkinson disease. Ablation of the gene in the forebrain creates the first model of dementia with Lewy bodies. In both neuroanatomical regions, gene ablation causes the formation of Lewy-like inclusions together with extensive neurodegeneration. There is some evidence for neuronal autophagy in areas adjacent to inclusions. The models indicate that neuronal loss in neurodegenerative diseases can be attributed to proteasomal malfunction accompanied by Lewy-like inclusions as seen in dementia with Lewy bodies and Parkinson disease.


Assuntos
Autofagia , Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Animais , Doença Crônica , Humanos , Camundongos , Dobramento de Proteína
17.
Biochim Biophys Acta ; 1782(12): 683-90, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18976704

RESUMO

Neuropathological investigations have identified major hallmarks of chronic neurodegenerative disease. These include protein aggregates called Lewy bodies in dementia with Lewy bodies and Parkinson's disease. Mutations in the alpha-synuclein gene have been found in familial disease and this has led to intense focused research in vitro and in transgenic animals to mimic and understand Parkinson's disease. A decade of transgenesis has lead to overexpression of wild type and mutated alpha-synuclein, but without faithful reproduction of human neuropathology and movement disorder. In particular, widespread regional neuronal cell death in the substantia nigra associated with human disease has not been described. The intraneuronal protein aggregates (inclusions) in all of the human chronic neurodegenerative diseases contain ubiquitylated proteins. There could be several reasons for the accumulation of ubiquitylated proteins, including malfunction of the ubiquitin proteasome system (UPS). This hypothesis has been genetically tested in mice by conditional deletion of a proteasomal regulatory ATPase gene. The consequences of gene ablation in the forebrain include extensive neuronal death and the production of Lewy-like bodies containing ubiquitylated proteins as in dementia with Lewy bodies. Gene deletion in catecholaminergic neurons, including in the substantia nigra, recapitulates the neuropathology of Parkinson's disease.


Assuntos
Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , alfa-Sinucleína/metabolismo , Animais , Humanos , Corpos de Lewy , Doenças Neurodegenerativas/genética , alfa-Sinucleína/genética
18.
J Neurosci ; 28(33): 8189-98, 2008 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-18701681

RESUMO

Ubiquitin-positive intraneuronal inclusions are a consistent feature of the major human neurodegenerative diseases, suggesting that dysfunction of the ubiquitin proteasome system is central to disease etiology. Research using inhibitors of the 20S proteasome to model Parkinson's disease is controversial. We report for the first time that specifically 26S proteasomal dysfunction is sufficient to trigger neurodegenerative disease. Here, we describe novel conditional genetic mouse models using the Cre/loxP system to spatially restrict inactivation of Psmc1 (Rpt2/S4) to neurons of either the substantia nigra or forebrain (e.g., cortex, hippocampus, and striatum). PSMC1 is an essential subunit of the 26S proteasome and Psmc1 conditional knock-out mice display 26S proteasome depletion in targeted neurons, in which the 20S proteasome is not affected. Impairment of specifically ubiquitin-mediated protein degradation caused intraneuronal Lewy-like inclusions and extensive neurodegeneration in the nigrostriatal pathway and forebrain regions. Ubiquitin and alpha-synuclein neuropathology was evident, similar to human Lewy bodies, but interestingly, inclusion bodies contained mitochondria. We support this observation by demonstrating mitochondria in an early form of Lewy body (pale body) from Parkinson's disease patients. The results directly confirm that 26S dysfunction in neurons is involved in the pathology of neurodegenerative disease. The model demonstrates that 26S proteasomes are necessary for normal neuronal homeostasis and that 20S proteasome activity is insufficient for neuronal survival. Finally, we are providing the first reproducible genetic platform for identifying new therapeutic targets to slow or prevent neurodegeneration.


Assuntos
Encéfalo/enzimologia , Corpos de Inclusão/enzimologia , Corpos de Lewy/enzimologia , Degeneração Neural/enzimologia , Degeneração Neural/genética , Neurônios/enzimologia , Complexo de Endopeptidases do Proteassoma/deficiência , Animais , Encéfalo/patologia , Feminino , Humanos , Corpos de Inclusão/genética , Corpos de Inclusão/patologia , Corpos de Lewy/genética , Corpos de Lewy/patologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Degeneração Neural/patologia , Neurônios/patologia , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/fisiologia
19.
Essays Biochem ; 41: 157-71, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16250904

RESUMO

As in all other mammalian tissues, the UPS (ubiquitin-proteasome system) is fundamental to normal brain function. A consistent feature of the major human neurodegenerative disorders is the accumulation of disease-related proteins, in non-native conformations, as protein aggregates within neurons or glial cells. Often the proteins in these aggregates are post-translationally conjugated with ubiquitin, suggesting a possible link between pathological protein-aggregation events in the nervous system and dysfunction of the UPS. Genetic evidence clearly demonstrates that disruption of ubiquitin-mediated processes can lead to neurodegeneration; however, the relationship between the UPS and idiopathic neurodegenerative disorders is less clear. In the latter cases, although a number of different mechanisms could potentially contribute to dysfunction of the UPS and promote the neurodegenerative process, whether UPS dysfunction is causally related to disease pathogenesis, or alternatively arises as a result of the pathological state, and indeed whether ubiquitinated inclusions are harmful or beneficial to cells, remains to be clarified.


Assuntos
Doenças Neurodegenerativas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Humanos , Doença de Huntington/genética , Doença de Huntington/metabolismo , Aprendizagem/fisiologia , Memória/fisiologia , Mutação , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/fisiopatologia
20.
Dev Biol ; 280(2): 386-95, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15882580

RESUMO

Complex intrinsic and extrinsic mechanisms determine neural cell fate during development of the nervous system. Using Id4 deficient mice, we show that Id4 is required for normal development of the central nervous system (CNS), timing neural differentiation in the developing forebrain. In the absence of Id4, the ventricular zone of the neocortex, future hippocampus as well as lateral and medial ganglionic eminences exhibited a 20-30% reduction in mitotic neural precursor cells (NPCs). Although the number of apoptotic cells was significantly increased, the neocortex of Id4(-/-) embryos was consistently thicker due to premature neuronal differentiation, which resulted in an increase in early-born neurons in the adult Id4(-/-) cortex. Late-born cortical neurons and astrocytes in the cortex, septum, hippocampus and caudate putamen of Id4(-/-) adult brains were decreased, however, likely due to the depletion of the NPC pool. Consequently, adult Id4(-/-) brains were smaller and exhibited enlarged ventricles. In vitro analysis of neurosphere cultures revealed that proliferation of Id4-deficient NPCs was impaired and that BMP2-mediated astrocyte differentiation was accelerated in the absence of Id4. Together, these in vivo and in vitro data suggest a crucial role for Id4 in regulating NPC proliferation and differentiation.


Assuntos
Sistema Nervoso Central/embriologia , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/citologia , Fatores de Transcrição/fisiologia , Animais , Apoptose , Astrócitos/metabolismo , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/metabolismo , Encéfalo/embriologia , Encéfalo/metabolismo , Diferenciação Celular , Proliferação de Células , Sistema Nervoso Central/metabolismo , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos/metabolismo , Hipocampo/embriologia , Imuno-Histoquímica , Proteínas Inibidoras de Diferenciação , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Mutação , Neocórtex/metabolismo , Neurônios/metabolismo , Fenótipo , Prosencéfalo/embriologia , Fatores de Tempo , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...