Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hum Mol Genet ; 25(14): 2972-2984, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27206984

RESUMO

Mutations in PARK2, encoding the E3 ubiquitin protein ligase Parkin, are a common cause of autosomal recessive Parkinson's disease (PD). Loss of PARK2 function compromises mitochondrial quality by affecting mitochondrial biogenesis, bioenergetics, dynamics, transport and turnover. We investigated the impact of PARK2 dysfunction on the endoplasmic reticulum (ER)-mitochondria interface, which mediates calcium (Ca2+) exchange between the two compartments and is essential for Parkin-dependent mitophagy. Confocal and electron microscopy analyses showed the ER and mitochondria to be in closer proximity in primary fibroblasts from PARK2 knockout (KO) mice and PD patients with PARK2 mutations than in controls. Ca2+ flux to the cytosol was also modified, due to enhanced ER-to-mitochondria Ca2+ transfers, a change that was also observed in neurons derived from induced pluripotent stem cells of a patient with PARK2 mutations. Subcellular fractionation showed the abundance of the Parkin substrate mitofusin 2 (Mfn2), which is known to modulate the ER-mitochondria interface, to be specifically higher in the mitochondrion-associated ER membrane compartment in PARK2 KO tissue. Mfn2 downregulation or the exogenous expression of normal Parkin restored cytosolic Ca2+ transients in fibroblasts from patients with PARK2 mutations. In contrast, a catalytically inactive PD-related Parkin variant had no effect. Overall, our data suggest that Parkin is directly involved in regulating ER-mitochondria contacts and provide new insight into the role of the loss of Parkin function in PD development.


Assuntos
Retículo Endoplasmático/metabolismo , GTP Fosfo-Hidrolases/genética , Mitocôndrias/metabolismo , Doença de Parkinson/genética , Ubiquitina-Proteína Ligases/genética , Animais , Sinalização do Cálcio/genética , Citosol/metabolismo , Retículo Endoplasmático/patologia , Fibroblastos , GTP Fosfo-Hidrolases/biossíntese , Regulação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias/patologia , Mitofagia/genética , Mutação , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia
2.
PLoS One ; 9(6): e99898, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24959870

RESUMO

Loss of Parkin, encoded by PARK2 gene, is a major cause of autosomal recessive Parkinson's disease. In Drosophila and mammalian cell models Parkin has been shown in to play a role in various processes essential to maintenance of mitochondrial quality, including mitochondrial dynamics, biogenesis and degradation. However, the relevance of altered mitochondrial quality control mechanisms to neuronal survival in vivo is still under debate. We addressed this issue in the brain of PARK2-/- mice using an integrated mitochondrial evaluation, including analysis of respiration by polarography or by fluorescence, respiratory complexes activity by spectrophotometric assays, mitochondrial membrane potential by rhodamine 123 fluorescence, mitochondrial DNA content by real time PCR, and oxidative stress by total glutathione measurement, proteasome activity, SOD2 expression and proteins oxidative damage. Respiration rates were lowered in PARK2-/- brain with high resolution but not standard respirometry. This defect was specific to the striatum, where it was prominent in neurons but less severe in astrocytes. It was present in primary embryonic cells and did not worsen in vivo from 9 to 24 months of age. It was not associated with any respiratory complex defect, including complex I. Mitochondrial inner membrane potential in PARK2-/- mice was similar to that of wild-type mice but showed increased sensitivity to uncoupling with ageing in striatum. The presence of oxidative stress was suggested in the striatum by increased mitochondrial glutathione content and oxidative adducts but normal proteasome activity showed efficient compensation. SOD2 expression was increased only in the striatum of PARK2-/- mice at 24 months of age. Altogether our results show a tissue-specific mitochondrial defect, present early in life of PARK2-/- mice, mildly affecting respiration, without prominent impact on mitochondrial membrane potential, whose underlying mechanisms remain to be elucidated, as complex I defect and prominent oxidative damage were ruled out.


Assuntos
Corpo Estriado/embriologia , Mitocôndrias/fisiologia , Superóxido Dismutase/metabolismo , Ubiquitina-Proteína Ligases/deficiência , Animais , Respiração Celular , Corpo Estriado/metabolismo , Potencial da Membrana Mitocondrial , Camundongos , Especificidade de Órgãos , Estresse Oxidativo , Superóxido Dismutase/genética
3.
J Mol Cell Biol ; 5(2): 132-42, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23359614

RESUMO

We previously established that besides its canonical function as E3-ubiquitin ligase, parkin also behaves as a transcriptional repressor of p53. Here we show that parkin differently modulates presenilin-1 and presenilin-2 expression and functions at transcriptional level. Thus, parkin enhances/reduces the protein expression, promoter activity and mRNA levels of presenilin-1 and presenilin-2, respectively, in cells and in vivo. This parkin-associated function is independent of its ubiquitin-ligase activity and remains unrelated to its capacity to repress p53. Accordingly, physical interaction of endogenous or overexpressed parkin with presenilins promoters is demonstrated by chromatin immunoprecipitation assays (ChIP). Furthermore, we identify a consensus sequence, the deletion of which abolishes parkin-dependent modulation of presenilins-1/2 and p53 promoter activities. Interestingly, electrophoretic mobility shift assays (EMSA) revealed a physical interaction between this consensus sequence and wild-type but not mutated parkin. Finally, we demonstrate that the RING1-IBR-RING2 domain of parkin harbors parkin's potential to modulate presenilins promoters. This transcriptional control impacts on presenilins-associated phenotypes, since parkin increases presenilin-1-associated γ-secretase activity and reduces presenilin-2-linked caspase-3 activation. Overall, our data delineate a promoter responsive element targeted by parkin that drives differential regulation of presenilin-1 and presenilin-2 transcription with functional consequences for γ-secretase activity and cell death.


Assuntos
Presenilina-1/metabolismo , Presenilina-2/metabolismo , Regiões Promotoras Genéticas/fisiologia , Transcrição Gênica/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Caspase 3/genética , Caspase 3/metabolismo , Morte Celular/fisiologia , Ativação Enzimática/fisiologia , Camundongos , Camundongos Knockout , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Presenilina-1/genética , Presenilina-2/genética , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética , Ubiquitina-Proteína Ligases/genética
4.
PLoS One ; 7(7): e40501, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22792356

RESUMO

BACKGROUND: Loss of function mutations in the DJ-1 gene have been linked to recessively inherited forms of Parkinsonism. Mitochondrial dysfunction and increased oxidative stress are thought to be key events in the pathogenesis of Parkinson's disease. Although it has been reported that DJ-1 serves as scavenger for reactive oxidative species (ROS) by oxidation on its cysteine residues, how loss of DJ-1 affects mitochondrial function is less clear. METHODOLOGY/PRINCIPAL FINDINGS: Using primary mouse embryonic fibroblasts (MEFs) or brains from DJ-1-/- mice, we found that loss of DJ-1 does not affect mitochondrial respiration. Specifically, endogenous respiratory activity as well as basal and maximal respiration are normal in intact DJ-1-/- MEFs, and substrate-specific state 3 and state 4 mitochondrial respiration are also unaffected in permeabilized DJ-1-/- MEFs and in isolated mitochondria from the cerebral cortex of DJ-1-/- mice at 3 months or 2 years of age. Expression levels and activities of all individual complexes composing the electron transport system are unchanged, but ATP production is reduced in DJ-1-/- MEFs. Mitochondrial transmembrane potential is decreased in the absence of DJ-1. Furthermore, mitochondrial permeability transition pore opening is increased, whereas mitochondrial calcium levels are unchanged in DJ-1-/- cells. Consistent with earlier reports, production of reactive oxygen species (ROS) is increased, though levels of antioxidative enzymes are unaltered. Interestingly, the decreased mitochondrial transmembrane potential and the increased mitochondrial permeability transition pore opening in DJ-1-/- MEFs can be restored by antioxidant treatment, whereas oxidative stress inducers have the opposite effects on mitochondrial transmembrane potential and mitochondrial permeability transition pore opening. CONCLUSIONS/SIGNIFICANCE: Our study shows that loss of DJ-1 does not affect mitochondrial respiration or mitochondrial calcium levels but increases ROS production, leading to elevated mitochondrial permeability transition pore opening and reduced mitochondrial transmembrane potential.


Assuntos
Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Antioxidantes/metabolismo , Cálcio/metabolismo , Córtex Cerebral/metabolismo , Fibroblastos , Glutationa/metabolismo , Potencial da Membrana Mitocondrial/genética , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Poro de Transição de Permeabilidade Mitocondrial , Estresse Oxidativo , Peroxirredoxinas , Proteína Desglicase DJ-1
5.
Mol Neurodegener ; 7: 22, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22630785

RESUMO

BACKGROUND: Loss-of-function mutations in PTEN-induced kinase 1 (PINK1) have been linked to familial Parkinson's disease, but the underlying pathogenic mechanism remains unclear. We previously reported that loss of PINK1 impairs mitochondrial respiratory activity in mouse brains. RESULTS: In this study, we investigate how loss of PINK1 impairs mitochondrial respiration using cultured primary fibroblasts and neurons. We found that intact mitochondria in PINK1-/- cells recapitulate the respiratory defect in isolated mitochondria from PINK1-/- mouse brains, suggesting that these PINK1-/- cells are a valid experimental system to study the underlying mechanisms. Enzymatic activities of the electron transport system complexes are normal in PINK1-/- cells, but mitochondrial transmembrane potential is reduced. Interestingly, the opening of the mitochondrial permeability transition pore (mPTP) is increased in PINK1-/- cells, and this genotypic difference between PINK1-/- and control cells is eliminated by agonists or inhibitors of the mPTP. Furthermore, inhibition of mPTP opening rescues the defects in transmembrane potential and respiration in PINK1-/- cells. Consistent with our earlier findings in mouse brains, mitochondrial morphology is similar between PINK1-/- and wild-type cells, indicating that the observed mitochondrial functional defects are not due to morphological changes. Following FCCP treatment, calcium increases in the cytosol are higher in PINK1-/- compared to wild-type cells, suggesting that intra-mitochondrial calcium concentration is higher in the absence of PINK1. CONCLUSIONS: Our findings show that loss of PINK1 causes selective increases in mPTP opening and mitochondrial calcium, and that the excessive mPTP opening may underlie the mitochondrial functional defects observed in PINK1-/- cells.


Assuntos
Encéfalo/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Proteínas Quinases/metabolismo , Animais , Respiração Celular/fisiologia , Células Cultivadas , Fibroblastos/metabolismo , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Camundongos Knockout , Proteínas de Transporte da Membrana Mitocondrial/genética , Poro de Transição de Permeabilidade Mitocondrial , Neurônios/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Proteínas Quinases/genética
6.
Neurobiol Dis ; 41(1): 111-8, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20817094

RESUMO

Mutations in PTEN-induced putative kinase 1 (PINK1) cause a recessive form of Parkinson's disease (PD). PINK1 is associated with mitochondrial quality control and its partial knock-down induces mitochondrial dysfunction including decreased membrane potential and increased vulnerability against mitochondrial toxins, but the exact function of PINK1 in mitochondria has not been investigated using cells with null expression of PINK1. Here, we show that loss of PINK1 caused mitochondrial dysfunction. In PINK1-deficient (PINK1(-/-)) mouse embryonic fibroblasts (MEFs), mitochondrial membrane potential and cellular ATP levels were decreased compared with those in littermate wild-type MEFs. However, mitochondrial proton leak, which reduces membrane potential in the absence of ATP synthesis, was not altered by loss of PINK1. Instead, activity of the respiratory chain, which produces the membrane potential by oxidizing substrates using oxygen, declined. H(2)O(2) production rate by PINK1(-/-) mitochondria was lower than PINK1(+/+) mitochondria as a consequence of decreased oxygen consumption rate, while the proportion (H(2)O(2) production rate per oxygen consumption rate) was higher. These results suggest that mitochondrial dysfunctions in PD pathogenesis are caused not by proton leak, but by respiratory chain defects.


Assuntos
Respiração Celular , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Neurônios/metabolismo , Doença de Parkinson/metabolismo , Proteínas Quinases/deficiência , Prótons , Animais , Respiração Celular/genética , Células Cultivadas , Fibroblastos/metabolismo , Potencial da Membrana Mitocondrial/genética , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/patologia , Doenças Mitocondriais/genética , Doenças Mitocondriais/patologia , Neurônios/patologia , Doença de Parkinson/genética , Doença de Parkinson/patologia , Proteínas Quinases/genética
7.
J Cell Biol ; 189(2): 211-21, 2010 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-20404107

RESUMO

Parkinson's disease (PD) is a prevalent neurodegenerative disorder. Recent identification of genes linked to familial forms of PD such as Parkin and PINK1 (PTEN-induced putative kinase 1) has revealed that ubiquitylation and mitochondrial integrity are key factors in disease pathogenesis. However, the exact mechanism underlying the functional interplay between Parkin-catalyzed ubiquitylation and PINK1-regulated mitochondrial quality control remains an enigma. In this study, we show that PINK1 is rapidly and constitutively degraded under steady-state conditions in a mitochondrial membrane potential-dependent manner and that a loss in mitochondrial membrane potential stabilizes PINK1 mitochondrial accumulation. Furthermore, PINK1 recruits Parkin from the cytoplasm to mitochondria with low membrane potential to initiate the autophagic degradation of damaged mitochondria. Interestingly, the ubiquitin ligase activity of Parkin is repressed in the cytoplasm under steady-state conditions; however, PINK1-dependent mitochondrial localization liberates the latent enzymatic activity of Parkin. Some pathogenic mutations of PINK1 and Parkin interfere with the aforementioned events, suggesting an etiological importance. These results provide crucial insight into the pathogenic mechanisms of PD.


Assuntos
Potencial da Membrana Mitocondrial/fisiologia , Mitocôndrias/patologia , Mitocôndrias/fisiologia , Proteínas Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Biomarcadores/metabolismo , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Dimetil Sulfóxido/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Corantes Fluorescentes/metabolismo , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Proteínas Quinases/genética , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Solventes/farmacologia , Ubiquitina-Proteína Ligases/genética , Desacopladores/farmacologia
8.
PLoS Biol ; 8(1): e1000298, 2010 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-20126261

RESUMO

Loss-of-function mutations in PINK1 and Parkin cause parkinsonism in humans and mitochondrial dysfunction in model organisms. Parkin is selectively recruited from the cytosol to damaged mitochondria to trigger their autophagy. How Parkin recognizes damaged mitochondria, however, is unknown. Here, we show that expression of PINK1 on individual mitochondria is regulated by voltage-dependent proteolysis to maintain low levels of PINK1 on healthy, polarized mitochondria, while facilitating the rapid accumulation of PINK1 on mitochondria that sustain damage. PINK1 accumulation on mitochondria is both necessary and sufficient for Parkin recruitment to mitochondria, and disease-causing mutations in PINK1 and Parkin disrupt Parkin recruitment and Parkin-induced mitophagy at distinct steps. These findings provide a biochemical explanation for the genetic epistasis between PINK1 and Parkin in Drosophila melanogaster. In addition, they support a novel model for the negative selection of damaged mitochondria, in which PINK1 signals mitochondrial dysfunction to Parkin, and Parkin promotes their elimination.


Assuntos
Mitocôndrias/metabolismo , Proteínas Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Células HeLa , Humanos , Potencial da Membrana Mitocondrial , Metaloproteases/metabolismo , Camundongos , Mitocôndrias/fisiologia , Membranas Mitocondriais/metabolismo , Modelos Biológicos , Doença de Parkinson/genética , Proteínas Quinases/genética , Ratos , Ubiquitina-Proteína Ligases/genética
9.
J Neurochem ; 111(3): 696-702, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19694908

RESUMO

Recessively inherited loss-of-function mutations in the parkin, DJ-1, or PINK1 gene are linked to familial cases of early-onset Parkinson's diseases (PD), and heterozygous mutations are associated with increased incidence of late-onset PD. We previously reported that single knockout mice lacking Parkin, DJ-1, or PINK1 exhibited no nigral degeneration, even though evoked dopamine release from nigrostriatal terminals was reduced and striatal synaptic plasticity was impaired. In this study, we tested whether inactivation of all three recessive PD genes, each of which was required for nigral neuron survival in the aging human brain, resulted in nigral degeneration during the lifespan of mice. Surprisingly, we found that triple knockout mice lacking Parkin, DJ-1, and PINK1 have normal morphology and numbers of dopaminergic and noradrenergic neurons in the substantia nigra and locus coeruleus, respectively, at the ages of 3, 16, and 24 months. Interestingly, levels of striatal dopamine in triple knockout mice were normal at 16 months of age but increased at 24 months. These results demonstrate that inactivation of all three recessive PD genes is insufficient to cause significant nigral degeneration within the lifespan of mice, suggesting that these genes may be protective rather than essential for the survival of dopaminergic neurons during the aging process. These findings also support the notion that mammalian Parkin and PINK1 may function in the same genetic pathway as in Drosophila.


Assuntos
Degeneração Neural/genética , Neurônios/fisiologia , Proteínas Oncogênicas/deficiência , Proteínas Quinases/deficiência , Substância Negra/patologia , Ubiquitina-Proteína Ligases/deficiência , Fatores Etários , Animais , Contagem de Células/métodos , Dopamina/metabolismo , Eletroquímica/métodos , Locus Cerúleo/patologia , Camundongos , Camundongos Knockout , Nordefrin/metabolismo , Peroxirredoxinas , Proteína Desglicase DJ-1 , Tirosina 3-Mono-Oxigenase/metabolismo
10.
Proc Natl Acad Sci U S A ; 105(32): 11364-9, 2008 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-18687901

RESUMO

Parkinson's disease (PD) is a common neurodegenerative disorder thought to be associated with mitochondrial dysfunction. Loss of function mutations in the putative mitochondrial protein PINK1 (PTEN-induced kinase 1) have been linked to familial forms of PD, but the relation of PINK1 to mammalian mitochondrial function remains unclear. Here, we report that germline deletion of the PINK1 gene in mice significantly impairs mitochondrial functions. Quantitative electron microscopic studies of the striatum in PINK1(-/-) mice at 3-4 and 24 months revealed no gross changes in the ultrastructure or the total number of mitochondria, although the number of larger mitochondria is selectively increased. Functional assays showed impaired mitochondrial respiration in the striatum but not in the cerebral cortex at 3-4 months of age, suggesting specificity of this defect for dopaminergic circuitry. Aconitase activity associated with the Krebs cycle is also reduced in the striatum of PINK1(-/-) mice. Interestingly, mitochondrial respiration activities in the cerebral cortex are decreased in PINK1(-/-) mice at 2 years compared with control mice, indicating that aging can exacerbate mitochondrial dysfunction in these mice. Furthermore, mitochondrial respiration defects can be induced in the cerebral cortex of PINK1(-/-) mice by cellular stress, such as exposure to H(2)O(2) or mild heat shock. Together, our findings demonstrate that mammalian PINK1 is important for mitochondrial function and provides critical protection against both intrinsic and environmental stress, suggesting a pathogenic mechanism by which loss of PINK1 may lead to nigrostriatal degeneration in PD.


Assuntos
Deleção de Genes , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Proteínas Mitocondriais/genética , Estresse Oxidativo , Doença de Parkinson/metabolismo , Proteínas Quinases/genética , Animais , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Ciclo do Ácido Cítrico/genética , Corpo Estriado , Mutação em Linhagem Germinativa , Resposta ao Choque Térmico/genética , Peróxido de Hidrogênio/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/patologia , Doenças Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Estresse Oxidativo/genética , Consumo de Oxigênio/genética , Doença de Parkinson/genética , Doença de Parkinson/patologia , Proteínas Quinases/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...