Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J. physiol. biochem ; 80(1): 41-51, Feb. 2024. ilus, graf
Artigo em Inglês | IBECS | ID: ibc-EMG-564

RESUMO

Parkin is an ubiquitin‐E3 ligase that is involved in cellular mitophagy and was recently shown to contribute to controlling adipose tissue thermogenic plasticity. We found that Parkin expression is induced in brown (BAT) and white (WAT) adipose tissues of aged mice. We determined the potential role of Parkin in the aging-associated decline in the thermogenic capacity of adipose tissues by analyzing subcutaneous WAT, interscapular BAT, and systemic metabolic and physiological parameters in young (5 month-old) and aged (16 month-old) mice with targeted invalidation of the Parkin (Park2) gene, and their wild-type littermates. Our data indicate that suppression of Parkin prevented adipose accretion, increased energy expenditure and improved the systemic metabolic derangements, such as insulin resistance, seen in aged mice. This was associated with maintenance of browning and reduction of the age-associated induction of inflammation in subcutaneous WAT. BAT in aged mice was much less affected by Parkin gene invalidation. Such protection was associated with a dramatic prevention of the age-associated induction of fibroblast growth factor-21 (FGF21) levels in aged Parkin-invalidated mice. This was associated with a parallel reduction in FGF21 gene expression in adipose tissues and liver in aged Parkin-invalidated mice. Additionally, Parkin invalidation prevented the protein down-regulation of β-Klotho (a key co-receptor mediating FGF21 responsiveness in tissues) in aged adipose tissues. We conclude that Parkin down-regulation leads to improved systemic metabolism in aged mice, in association with maintenance of adipose tissue browning and FGF21 system functionality. (AU)


Assuntos
Animais , Camundongos , Proteínas Associadas à Doença de Parkinson , Peptídeos e Proteínas de Sinalização Intercelular , Envelhecimento , Tecido Adiposo , Metabolismo
2.
J. physiol. biochem ; 80(1): 41-51, Feb. 2024. ilus, graf
Artigo em Inglês | IBECS | ID: ibc-229939

RESUMO

Parkin is an ubiquitin‐E3 ligase that is involved in cellular mitophagy and was recently shown to contribute to controlling adipose tissue thermogenic plasticity. We found that Parkin expression is induced in brown (BAT) and white (WAT) adipose tissues of aged mice. We determined the potential role of Parkin in the aging-associated decline in the thermogenic capacity of adipose tissues by analyzing subcutaneous WAT, interscapular BAT, and systemic metabolic and physiological parameters in young (5 month-old) and aged (16 month-old) mice with targeted invalidation of the Parkin (Park2) gene, and their wild-type littermates. Our data indicate that suppression of Parkin prevented adipose accretion, increased energy expenditure and improved the systemic metabolic derangements, such as insulin resistance, seen in aged mice. This was associated with maintenance of browning and reduction of the age-associated induction of inflammation in subcutaneous WAT. BAT in aged mice was much less affected by Parkin gene invalidation. Such protection was associated with a dramatic prevention of the age-associated induction of fibroblast growth factor-21 (FGF21) levels in aged Parkin-invalidated mice. This was associated with a parallel reduction in FGF21 gene expression in adipose tissues and liver in aged Parkin-invalidated mice. Additionally, Parkin invalidation prevented the protein down-regulation of β-Klotho (a key co-receptor mediating FGF21 responsiveness in tissues) in aged adipose tissues. We conclude that Parkin down-regulation leads to improved systemic metabolism in aged mice, in association with maintenance of adipose tissue browning and FGF21 system functionality. (AU)


Assuntos
Animais , Camundongos , Proteínas Associadas à Doença de Parkinson , Peptídeos e Proteínas de Sinalização Intercelular , Envelhecimento , Tecido Adiposo , Metabolismo
3.
Neurobiol Aging ; 100: 118.e5-118.e13, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33423827

RESUMO

Increased burdens of rare coding variants in genes related to lysosomal storage disease or mitochondrial pathways were reported to be associated with idiopathic Parkinson's disease. Under a hypothesis that the burden of damaging rare coding variants is increased in causative genes for hereditary parkinsonism, we analyzed the burdens of rare coding variants with a case-control design. Two cohorts of whole-exome sequencing data and a cohort of genome-wide genotyping data of clinically validated idiopathic Parkinson's disease cases and controls, which were open to the public, were used. The sequence kernel association test-optimal was used to analyze the burden of rare variants in the hereditary parkinsonism gene set, which was constructed from the Online Mendelian Inheritance in Man database through manual curation. The hereditary parkinsonism gene set consisted of 17 genes with a locus symbol prefix for familial Parkinson's disease and 75 hereditary atypical parkinsonism genes. We detected a significant association of enriched burdens of predicted damaging rare coding variants in hereditary parkinsonism genes in all three datasets. Meta-analyses of the rare variant burden test in a subgroup of gene sets revealed an association between burdens of rare damaging variants with PD in a hereditary atypical parkinsonism gene set, but not in a subgroup gene set with a locus symbol prefix for familial Parkinson's disease. Our results highlight the roles of rare damaging variants in causative genes for hereditary atypical parkinsonian disorders. We propose that Mendelian genes associated with hereditary disorders accompanying parkinsonism are involved in Parkinson's disease-related genetic networks.


Assuntos
Estudos de Associação Genética/métodos , Variação Genética/genética , Doença de Parkinson/genética , Idoso , Estudos de Casos e Controles , Estudos de Coortes , Bases de Dados Genéticas , Conjuntos de Dados como Assunto , Feminino , Genótipo , Humanos , Doenças por Armazenamento dos Lisossomos/genética , Masculino , Pessoa de Meia-Idade , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas Associadas à Doença de Parkinson/genética , Transdução de Sinais/genética , Sequenciamento do Exoma
4.
Neuromolecular Med ; 21(1): 25-32, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30411223

RESUMO

Parkinson's disease (PD) is a prevalent neurodegenerative movement disorder that is characterized pathologically by the progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) of the midbrain. Despite intensive research, the etiology of PD remains poorly understood. Interestingly, recent studies have implicated neuronal energy dysregulation as one of the key perpetrators of the disease. Supporting this, we have recently demonstrated that pharmacological or genetic activation of AMP kinase (AMPK), a master regulator of cellular energy homeostasis, rescues the pathological phenotypes of Drosophila models of PD. However, little is known about the role of AMPK in the mammalian brain. As an initial attempt to clarify this, we examined the expression of AMPK in rodent brains and found that phospho-AMPK (pAMPK) is disproportionately distributed in the adult mouse brain, being high in the ventral midbrain where the SN resides and relatively lower in regions such as the cortex-reflecting perhaps the unique energy demands of midbrain DA neurons. Importantly, the physiologically higher level of midbrain pAMPK is significantly reduced in aged mice and also in Parkin-deficient mice; the loss of function of which in humans causes recessive Parkinsonism. Not surprisingly, the expression of PGC-1α, a downstream target of AMPK activity, and a key regulator of mitochondrial biogenesis, mirrors the expression pattern of pAMPK. Similar observations were made with PINK1-deficient mice. Finally, we showed that metformin administration restores the level of midbrain pAMPK and PGC-1α expression in Parkin-deficient mice. Taken together, our results suggest that the disruption of AMPK-PGC-1α axis in the brains of individuals with Parkin or PINK1 mutations may be a precipitating factor of PD, and that pharmacological AMPK activation may represent a neuroprotective strategy for the disease.


Assuntos
Adenilato Quinase/metabolismo , Mesencéfalo/enzimologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Associadas à Doença de Parkinson/metabolismo , Proteínas Quinases/deficiência , Ubiquitina-Proteína Ligases/deficiência , Envelhecimento/metabolismo , Animais , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Avaliação Pré-Clínica de Medicamentos , Metabolismo Energético , Ativação Enzimática , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Metformina/farmacologia , Metformina/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Especificidade de Órgãos , Proteínas Associadas à Doença de Parkinson/deficiência , Proteínas Associadas à Doença de Parkinson/genética , Parte Compacta da Substância Negra/enzimologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/biossíntese , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Fosforilação , Proteínas Quinases/genética , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Ubiquitina-Proteína Ligases/genética
5.
Neurosci Lett ; 665: 110-116, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29191693

RESUMO

Numerous published case-control studies have investigated a role of PARK16 gene in susceptibility to Parkinson's disease (PD), but the results remain conflicting and under-powered. Herein, we performed this meta-analysis to evaluate the possible association between the polymorphism of the PARK16 rs8231128 (A/G) and PD.A comprehensive search of six databases was conducted to identify all case-control studies involving PARK16rs823128variants and PD risk up to August 2017. The strict inclusion and exclusion criteria were applied. A total of 9 studies including 15 case-control studies with 7277 PD cases and 6188 controls were included in the meta-analysis. And STATA 12.0 statistics software was used to calculate available data from each study. The crude odds ratios (OR) and 95% confidence interval (CI) were calculated to assess the genetic association between PARK16 rs823128 polymorphism and the risk of PD. In the combined analysis, results showed a significant association between rs823128 and PD in allelic model(G vs. A: OR=0.886, 95% CI=0.811-0.969, P=0.008), dominant model (GG+ AG vs. AA: OR=0.886, 95% CI=0.804-0.976, P=0.014), and heterozygote model (AG vs. AA: OR=0.897, 95% CI=0.812-0.991, P=0.032). Further, ethnicity based analysis showed a significant association in Asian and Chilean population, but not in Caucasian samples. Within its limitations, this meta-analysis demonstrated that the rs823128 variants(G allele, GA and GG genotype)in PARK16 might be a potential protective factor for PD. However, these associations vary in different ethnicities.


Assuntos
Predisposição Genética para Doença/genética , Proteínas Associadas à Doença de Parkinson/genética , Doença de Parkinson/genética , Polimorfismo de Nucleotídeo Único/genética , Povo Asiático/genética , Estudos de Associação Genética , Genótipo , Humanos , Risco , Fatores de Risco , População Branca/genética
6.
Med. UIS ; 30(3): 31-38, sep.-dic. 2017. tab, graf
Artigo em Espanhol | LILACS | ID: biblio-894215

RESUMO

Resumen Introducción: La enfermedad de Parkinson se caracteriza por la degeneración y pérdida de las neuronas dopaminérgicas en el cerebro. Existen factores genéticos involucrados en su desarrollo, en su forma de inicio temprano como el gen PARK2, codificante de la parkina, una E3 ubiquitín ligasa, lo que hace que en caso de mutaciones pierda su capacidad reguladora de degradación de proteínas causando estrés y muerte celular. Objetivo: Determinar la presencia de cambios moleculares en los exones 3, 4, y 5 de PARK2 en un grupo de 29 pacientes y 21 controles colombianos con enfermedad de Parkinson de inicio temprano o con antecedentes familiares de ella y la posible correlación con las manifestaciones clínicas de los pacientes. Materiales y métodos: Estudio descriptivo observacional (entre junio de 2013 y noviembre de 2014) en donde se realizó extracción de ADN de sangre total y se utilizó la técnica de PCR para cada uno de los exones. Finalmente se procedió a la secuenciación automática, análisis de las secuencias con el software Sequencher y comparación con información de bases de datos. Resultados: Se identificó una variante en estado homocigoto (Ala46Thr) en el exón 4 en un paciente no reportada anteriormente, posiblemente no patogénica y la variante Ser167Asn en estado heterocigoto en el mismo exón en otro paciente, considerada patogénica y reportada con anterioridad en poblaciones asiática y europea. No se identificaron variantes en los controles. Conclusiones: Los cambios no descritos antes en la población colombiana, -Ala46Thr y Ser167Asn-, fueron identificados en el grupo de pacientes. MÉD.UIS. 2017;30(3):31-8.


Abstract Introduction: Parkinson's disease is characterized by the degeneration and loss of dopaminergic neurons in the brain. There are genetic factors involved in its development in its early onset form, such as the PARK2 gene encoding the parkin, an E3 ubiquitin ligase, which in the case of mutations loses its ability to regulate protein degradation causing stress and cell death. Objective: To determine the presence of molecular changes in PARK2 exons 3, 4 and 5 in a group of 29 patients and 21 colombian controls with early onset Parkinson's disease or a family history of Parkinson's disease and the possible correlation with clinical manifestations from the patients. Materials and methods: Observational descriptive study (between june of 2013 and november of 2014) where DNA extraction of whole blood was performed and the PCR technique was used for each of the exons. Finally, we proceeded to the automatic sequencing, analysis of the sequences with the Sequencher software and comparison with information of databases. Results: A homozygous variant (Ala46Thr) in exon 4 was identified in one patient not previously reported, possibly nonpathogenic and the Ser167Asn variant in heterozygous state in the same exon in another patient, considered pathogenic and previously reported in populations Asian and European. No variants were identified in the controls. Conclusions: changes not previously described in the colombian population, -Ala46Thr and Ser167Asn-, were identified in the group of patients and not in the controls. MÉD.UIS. 2017;30(3):31-8.


Assuntos
Humanos , Masculino , Feminino , Adulto , Doença de Parkinson , Proteínas Associadas à Doença de Parkinson , Reação em Cadeia da Polimerase , Análise de Sequência de DNA , Mutação com Perda de Função , Genes , Homozigoto , Mutação
7.
Ann Neurol ; 80(6): 811-820, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27761938

RESUMO

The mechanisms underlying the high prevalence of cutaneous malignant melanoma (CMM) in Parkinson disease (PD) are unclear, but plausibly involve common pathways. 129Ser-phosphorylated α-synuclein, a pathological PD hallmark, is abundantly expressed in CMM, but not in normal skin. In inherited PD, PARK genes harbor germline mutations; the same genes are somatically mutated in CMM, or their encoded proteins are involved in melanomagenesis. Conversely, genes associated with CMM affect PD risk. PD/CMM-targeted cells share neural crest origin and melanogenesis capability. Pigmentation gene variants may underlie their susceptibility. We review putative genetic intersections that may be suggestive of shared pathways in neurodegeneration/melanomagenesis. Ann Neurol 2016;80:811-820.


Assuntos
Melanoma/complicações , Melanoma/genética , Doença de Parkinson/complicações , Doença de Parkinson/genética , Neoplasias Cutâneas/complicações , Neoplasias Cutâneas/genética , Inibidor p16 de Quinase Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p18/genética , Predisposição Genética para Doença , Genótipo , Humanos , Proteínas Associadas à Doença de Parkinson/genética , Polimorfismo de Nucleotídeo Único , Receptor Tipo 1 de Melanocortina/genética , Receptores de N-Metil-D-Aspartato/genética , alfa-Sinucleína/genética
8.
Biochem Biophys Res Commun ; 476(4): 528-533, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27262440

RESUMO

The Saccharomyces cerevisiae gene VPS35 encodes a component of the retromer complex which is involved in vesicle transport from endosomes to the trans-Golgi network. Yeast and human VPS35 orthologs are highly conserved and mutations in human VPS35 cause an autosomal dominant form of late-onset Parkinson disease (PD). We now show that deletion of VPS35 in yeast (vps35Δ) leads to a dose-dependent growth defect towards copper. This increased sensitivity could be rescued by transformation with yeast wild-type VPS35 but not by the expression of a construct harboring the yeast equivalent (i.e. D686N) of the most commonly identified VPS35-associated PD mutation, p.D620N. In addition, we show that expression of one copy of α-synuclein, which is known to directly interact with copper, leads to a pronounced aggravation of copper toxicity in vps35Δ cells, thereby linking the regulation of copper homeostasis by Vps35p in yeast to one of the key molecules in PD pathophysiology.


Assuntos
Cobre/toxicidade , Doença de Parkinson/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Técnicas de Inativação de Genes , Genes Fúngicos , Teste de Complementação Genética , Humanos , Mutação , Proteínas Associadas à Doença de Parkinson/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Transporte Vesicular/deficiência , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...