Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 127(4): 502-518, 2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32366200

RESUMEN

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.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Calcineurina/metabolismo , Macroautofagia/fisiología , Complejo de la Endopetidasa Proteasomal/fisiología , ATPasas Asociadas con Actividades Celulares Diversas/genética , Animales , Antineoplásicos/farmacología , Bortezomib/farmacología , Calcineurina/genética , Inhibidores de la Calcineurina , Hipertrofia Ventricular Izquierda/inducido químicamente , Lisosomas/metabolismo , Ratones , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Inhibidores de Proteasoma , Proteostasis , ARN Interferente Pequeño , Ratas , Proteína Sequestosoma-1/metabolismo , Transducción de Señal/fisiología , Canales de Potencial de Receptor Transitorio/metabolismo , Ubiquitina/metabolismo , Regulación hacia Arriba
2.
J Biol Chem ; 289(36): 24944-55, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25049227

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autofagia , Proteínas del Citoesqueleto/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas del Citoesqueleto/genética , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Immunoblotting , Proteína 1 Asociada A ECH Tipo Kelch , Hígado/metabolismo , Hígado/patología , Hígado/ultraestructura , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Microscopía Inmunoelectrónica , Factor 2 Relacionado con NF-E2/genética , Fagosomas/genética , Fagosomas/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal/genética , Proteína Sequestosoma-1 , Factores de Tiempo , Ubiquitina/metabolismo
3.
Biochim Biophys Acta ; 1832(12): 1930-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23851049

RESUMEN

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.


Asunto(s)
Astrocitos/metabolismo , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Estrés Oxidativo , Prosencéfalo/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Astrocitos/patología , Western Blotting , Electroforesis en Gel Bidimensional , Técnicas para Inmunoenzimas , Peroxidación de Lípido , Ratones , Degeneración Nerviosa/patología , Neuronas/patología , Fosfolipasas A2/metabolismo , Prosencéfalo/patología , Complejo de la Endopetidasa Proteasomal/química , Especies Reactivas de Oxígeno/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
4.
Cancer Cell ; 8(1): 75-87, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16023600

RESUMEN

Gankyrin is an ankyrin repeat oncoprotein commonly overexpressed in hepatocellular carcinomas. Gankyrin interacts with the S6 proteasomal ATPase and accelerates the degradation of the tumor suppressor Rb. We show here that gankyrin has an antiapoptotic activity in cells exposed to DNA damaging agents. Downregulation of gankyrin induces apoptosis in cells with wild-type p53. In vitro and in vivo experiments revealed that gankyrin binds to Mdm2, facilitating p53-Mdm2 binding, and increases ubiquitylation and degradation of p53. Gankyrin also enhances Mdm2 autoubiquitylation in the absence of p53. Downregulation of gankyrin reduced amounts of Mdm2 and p53 associated with the 26S proteasome. Thus, gankyrin is a cofactor that increases the activities of Mdm2 on p53 and probably targets polyubiquitylated p53 into the 26S proteasome.


Asunto(s)
Regulación de la Expresión Génica , Proteínas Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Ancirinas/metabolismo , Apoptosis , Células Cultivadas , Humanos , Inmunoprecipitación , Ratones , Datos de Secuencia Molecular , Neoplasias/metabolismo , Neoplasias/patología , Inhibidores de Proteasoma , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2 , ARN Interferente Pequeño/farmacología , Proteína de Retinoblastoma/genética , Proteína p53 Supresora de Tumor/genética , Dedos de Zinc
5.
J Neurochem ; 122(1): 24-37, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22486777

RESUMEN

The metabotropic glutamate receptors (mGluRs) fine-tune the efficacy of synaptic transmission. This unique feature makes mGluRs potential targets for the treatment of various CNS disorders. There is ample evidence to show that the ubiquitin proteasome system mediates changes in synaptic strength leading to multiple forms of synaptic plasticity. The present study describes a novel interaction between post-synaptic adaptors, long Homer-3 proteins, and one of the 26S proteasome regulatory subunits, the S8 ATPase, that influences the degradation of the metabotropic glutamate receptor 1α (mGluR1α). We have shown that the two human long Homer-3 proteins specifically interact with human proteasomal S8 ATPase. We identified that mGluR1α and long Homer-3s immunoprecipitate with the 26S proteasome both in vitro and in vivo. We further found that the mGluR1α receptor can be ubiquitinated and degraded by the 26S proteasome and that Homer-3A facilitates this process. Furthermore, the siRNA mediated silencing of Homer-3 led to increased levels of total and plasma membrane-associated mGluR1α receptors. These results suggest that long Homer-3 proteins control the degradation of mGluR1α receptors by shuttling ubiquitinated mGluR-1α receptors to the 26S proteasome via the S8 ATPase which may modulate synaptic transmission.


Asunto(s)
Proteínas Portadoras/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Transducción de Señal/fisiología , ATPasas Asociadas con Actividades Celulares Diversas , Animales , Cadherinas/metabolismo , Calnexina/metabolismo , Proteínas Portadoras/genética , Células Cultivadas , Corteza Cerebral/citología , Embrión de Mamíferos , Hipocampo/citología , Proteínas de Andamiaje Homer , Humanos , Neuronas/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/genética , Transfección , Ubiquitinación/fisiología
6.
Biochem Soc Trans ; 39(4): 917-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21787323

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Animales , Muerte Celular , Enfermedad Crónica , Humanos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Placa Amiloide/metabolismo
7.
Trends Cell Biol ; 16(5): 229-33, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16581249

RESUMEN

Gankyrin is a new oncoprotein with potent cell cycle and apoptotic properties that is overexpressed early in hepatocarcinogenesis and in hepatocellular carcinomas. Gankyrin regulates the phosphorylation of the retinoblastoma protein (pRb) by CDK4 and enhances the ubiquitylation of p53 by the RING ubiquitin ligase MDM2. Purified preparations of the 26S proteasome contain gankyrin, which specifically interacts with the S6b (Rpt3) ATPase of the 19S regulator. In conclusion, gankyrin is a small versatile cell cycle regulator that illustrates the essential interplay between the ubiquitin proteasome system and gene expression in the cell. Here, we discuss the activities of gankyrin and present a model for its function in the regulation of pRb and p53.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Ancirinas/metabolismo , Humanos , Fosforilación , Complejo de la Endopetidasa Proteasomal/genética , Proteínas Proto-Oncogénicas/genética , Ubiquitina/metabolismo
8.
J Neurosci ; 28(33): 8189-98, 2008 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-18701681

RESUMEN

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.


Asunto(s)
Encéfalo/enzimología , Cuerpos de Inclusión/enzimología , Cuerpos de Lewy/enzimología , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/genética , Neuronas/enzimología , Complejo de la Endopetidasa Proteasomal/deficiencia , Animales , Encéfalo/patología , Femenino , Humanos , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/patología , Cuerpos de Lewy/genética , Cuerpos de Lewy/patología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Degeneración Nerviosa/patología , Neuronas/patología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/fisiología
9.
Biochim Biophys Acta ; 1782(12): 683-90, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18976704

RESUMEN

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.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , alfa-Sinucleína/metabolismo , Animales , Humanos , Cuerpos de Lewy , Enfermedades Neurodegenerativas/genética , alfa-Sinucleína/genética
10.
Essays Biochem ; 41: 187-203, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16250906

RESUMEN

The ubiquitin proteasome system (UPS) has emerged from obscurity to be seen as a major player in all regulatory processes in the cell. The concentrations of key proteins in diverse regulatory pathways are controlled by post-translational ubiquitination and degradation by the 26 S proteasome. These regulatory cascades include growth-factor-controlled signal-transduction pathways and multiple points in the cell cycle. The cell cycle is orchestrated by a combination of cyclin-dependent kinases, kinase inhibitors and protein phosphorylation, together with the timely and specific degradation of cyclins and kinase inhibitors at critical points in the cell cycle by the UPS. These processes provide the irreversibility needed for movement of the cycle through gap 1 (G1), DNA synthesis (S), gap 2 (G2) and mitosis (M). The molecular events include cell-size control, DNA replication, DNA repair, chromosomal rearrangements and cell division. It is doubtful whether these events could be achieved without the temporally and spatially regulated combination of protein phosphorylation and ubiquitin-dependent degradation of key cell-cycle regulatory proteins. The oncogenic transformation of cells is a multistep process that can be triggered by mutation of genes for proteins involved in regulatory processes from the cell surface to the nucleus. Since the UPS has critical functions at all these levels of control, it is to be expected that UPS activities will be central to cell transformation and cancer progression.


Asunto(s)
Neoplasias/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Animales , Ciclo Celular , Reparación del ADN , Sustancias de Crecimiento/metabolismo , Humanos , Neoplasias/enzimología , Neoplasias/patología , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
11.
Methods Enzymol ; 399: 86-119, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16338351

RESUMEN

Ubiquitin immunohistochemistry has changed understanding of the pathophysiology of many diseases, particularly chronic neurodegenerative diseases. Protein aggregates (inclusions) containing ubiquitinated proteins occur in neurones and other cell types in the central nervous system in afflicted cells. The inclusions are present in all the neurological illnesses, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, polyglutamine diseases, and rarer forms of neurodegenerative disease. A new cause of cognitive decline in the elderly, "dementia with Lewy bodies," accounting for some 15-30% of cases, was initially discovered and characterized by ubiquitin immunocytochemistry. The optimal methods for carrying out immunohistochemical analyses of paraffin-embedded tissues are described, and examples of all the types of intracellular inclusions detected by ubiquitin immunohistochemistry in the diseases are illustrated. The role of the ubiquitin proteasome system (UPS) in disease progression is being actively researched globally and increasingly, because it is now realized that the UPS controls most pathways in cellular homeostasis. Many of these regulatory mechanisms will be dysfunctional in diseased cells. The goal is to understand fully the role of the UPS in the disorders and then therapeutically intervene in the ubiquitin pathway to treat these incurable diseases.


Asunto(s)
Diagnóstico , Ubiquitina/metabolismo , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/metabolismo , Anticuerpos/inmunología , Axones , Humanos , Inmunohistoquímica , Enfermedad por Cuerpos de Lewy/diagnóstico , Enfermedad por Cuerpos de Lewy/metabolismo , Neuronas/patología , Repeticiones de Trinucleótidos , Ubiquitina/inmunología
12.
Essays Biochem ; 38: 51-63, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12463161

RESUMEN

Intracellular proteins are targeted for degradation by the covalent attachment of chains of the small protein ubiquitin; a process known as ubiquitylation. Many proteins are phosphorylated prior to ubiquitylation, and therefore ubiquitylation and degradation of these proteins is regulated by kinase activity and signalling cascades. Many ubiquitylated proteins are degraded by the 26 S proteasome complex, which is found in the cytosol and nucleus. The 26 S proteasome consists of a 20 S core with proteolytic activity and 18 S regulatory complexes containing ATPases and ubiquitin-chain-binding proteins. Proteins degraded by the ubiquitin-proteasome pathway include cyclins and other regulators of the cell cycle, and transcription factors. Abnormal polypeptides are also degraded by the ubiquitin pathway, including abnormal polypeptides in the endoplasmic reticulum, which are translocated back out of the endoplasmic reticulum prior to ubiquitylation and degradation by the proteasome. The ubiquitin-proteasome pathway is implicated in numerous diseases including cancer and neurodegenerative diseases.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Complejos Multienzimáticos/metabolismo , Ubiquitina/metabolismo , Animales , Núcleo Celular/metabolismo , Citosol/metabolismo , Humanos , Modelos Biológicos , Modelos Químicos , Modelos Moleculares , Péptido Hidrolasas/metabolismo , Complejo de la Endopetidasa Proteasomal , Proteínas/metabolismo
14.
FEBS Lett ; 569(1-3): 211-6, 2004 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-15225636

RESUMEN

The detailed mechanism of eukaryotic 20S proteasome assembly is currently unknown. In the present study, we demonstrate that the 20S proteasome subunits alpha4 and alpha7 interact with each other as well as all the alpha-subunits in vivo and in vitro. The N-terminal parts of alpha4 and alpha7 are essential for these newly discovered interactions in vitro. Glycerol gradient centrifugation of soluble extracts of HEK293 cells and Western blot analyses show that several alpha-subunits are found in non-proteasomal low-density fractions. The alpha4 and alpha7 subunits co-immunoprecipitate together from these low-density fractions. The unexpected interaction between alpha4 and alpha7 may provide a molecular basis for the formation of previously reported 13S and 16S assembly intermediates.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Complejos Multienzimáticos/metabolismo , Subunidades de Proteína/metabolismo , Sitios de Unión , Fraccionamiento Celular , Línea Celular , Centrifugación por Gradiente de Densidad , Clonación Molecular , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/aislamiento & purificación , Vectores Genéticos , Humanos , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/aislamiento & purificación , Mutagénesis , Complejo de la Endopetidasa Proteasomal , Subunidades de Proteína/genética , Subunidades de Proteína/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Eliminación de Secuencia , Ultracentrifugación
15.
FEBS Lett ; 553(1-2): 200-4, 2003 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-14550573

RESUMEN

The human immunodeficiency virus-1 (HIV-1) Tat protein was previously reported to compete the association of PA28 regulator with the alpha rings of the 20S proteasome and to inhibit its peptidase activity. However, the distinct interaction sites within the proteasome complex remained to be determined. Here we show that HIV-1 Tat binds to alpha4 and alpha7, six beta subunits of the constitutive 20S proteasome and the interferon-gamma-inducible subunits beta2i and beta5i. A Tat-proteasome interaction can also be demonstrated in vivo and leads to inhibition of proteasomal activity. This indicates that Tat can modulate or interfere with cellular proteasome function by specific interaction with distinct proteasomal subunits.


Asunto(s)
Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Productos del Gen tat/metabolismo , Complejos Multienzimáticos/química , Complejos Multienzimáticos/metabolismo , Subunidades de Proteína/metabolismo , Línea Celular , Inhibidores de Cisteína Proteinasa/metabolismo , Células HeLa , Humanos , Pruebas de Precipitina , Complejo de la Endopetidasa Proteasomal , Unión Proteica , Subunidades de Proteína/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Drug News Perspect ; 16(2): 103-8, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12792671

RESUMEN

Several years ago ubiquitin immunocytochemistry first demonstrated that ubiquitin protein conjugates are present in intraneuronal inclusions in all the major human chronic neurodegenerative diseases, as well as in inclusions in cerebellar astrocytomas and in hepatocytes in alcoholic liver disease. Unexpectedly, further studies showed that Lewy bodies are present in the cortex. Lewy bodies were originally described in the brain stem and are pathogonomic in the neuropathological diagnosis of Parkinson's disease. A balanced interpretation of further elegant experimental approaches, including transgenesis, suggests that the formation of intraneuronal inclusions is cytoprotective. Putative oligomeric proaggregates (prefibrillar entities) of cellular proteins inhibit the 26S proteasome and promote apoptosis. In the last few years a clutch of distinct experimental approaches have focused on the roles of ubiquitin-related processes in the development of the nervous system and neurohomeostasis. It is now clear that the ubiquitin/proteasome system (UPP) has a pivotal role in synaptogenesis, the formation of neuromuscular junctions and neurotransmitter receptor function. The inhibitory GABA(A) receptor, alpha1 glycine receptor, beta(2)-adrenergic receptor and arrestin, opiate receptors and the excitatory metabotropic glutamate receptor (mGluR1alpha) are regulated by the UPP. It is also increasingly clear that the regulation of long-term synaptic plasticity, and therefore memory, is dependent on both protein synthesis and protein degradation. Therefore, for the first time we have the opportunity to dissect the substrate of memory and the basis of cognitive decline in aging and in chronic neurodegenerative disease. Clearly, further understanding will provide a platform for novel drug development to treat chronic neurodegenerative diseases, including Alzheimer- and Parkinson-related conditions, and possibly psychiatric disorders.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Complejos Multienzimáticos/metabolismo , Degeneración Nerviosa/enzimología , Enfermedades Neurodegenerativas/metabolismo , Ubiquitinas/metabolismo , Envejecimiento , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Homeostasis , Humanos , Cuerpos de Lewy/patología , Ligasas/antagonistas & inhibidores , Ligasas/metabolismo , Memoria , Complejos Multienzimáticos/antagonistas & inhibidores , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/patología , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Complejo de la Endopetidasa Proteasomal , Ubiquitina-Proteína Ligasas
17.
PLoS One ; 8(1): e54711, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23382946

RESUMEN

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.


Asunto(s)
Encéfalo/metabolismo , Cuerpos de Inclusión , Neuronas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , alfa-Sinucleína/metabolismo , Animales , Autofagia , Encéfalo/patología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Cuerpos de Inclusión/ultraestructura , Cuerpos de Lewy , Lisosomas/metabolismo , Masculino , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Neuronas/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Transducción de Señal , Sustancia Negra/metabolismo , Sustancia Negra/patología , alfa-Sinucleína/genética
18.
Neurosci Lett ; 521(2): 130-5, 2012 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-22677101

RESUMEN

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.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Encéfalo/metabolismo , Fibroblastos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Adenosina Trifosfatasas/genética , Animales , Encéfalo/patología , Células Cultivadas , Fibroblastos/citología , Puntos de Control de la Fase G2 del Ciclo Celular , Heterocigoto , Puntos de Control de la Fase M del Ciclo Celular , Ratones , Ratones Noqueados , Cultivo Primario de Células , Complejo de la Endopetidasa Proteasomal/genética
19.
Neurosci Lett ; 491(1): 44-7, 2011 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-21215295

RESUMEN

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.


Asunto(s)
Neuronas/metabolismo , Poliubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Adaptación Fisiológica/fisiología , Secuencia de Aminoácidos , Animales , Lisina/metabolismo , Espectrometría de Masas/métodos , Ratones , Ratones Noqueados , Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/deficiencia , Estructura Secundaria de Proteína , Transducción de Señal/fisiología , Ubiquitinación/fisiología
20.
Nat Rev Drug Discov ; 10(1): 29-46, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21151032

RESUMEN

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.


Asunto(s)
Sistemas de Liberación de Medicamentos , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Animales , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA